51
|
Concha J, Sangüesa E, Peña JL, Ribate MP, García CB. Retrospective pharmacogenetic study in a cohort of pediatric tuberous sclerosis complex patients using everolimus. Pharmacogenomics 2023; 24:797-808. [PMID: 37869874 DOI: 10.2217/pgs-2023-0140] [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] [Indexed: 10/24/2023] Open
Abstract
Aim: Tuberous sclerosis complex (TSC) is a rare disease that produces multisystemic disorders. Everolimus (EVR) is the only immunosuppressive drug approved to control the symptoms and progression of the disease. The aim was to evaluate the genotype-phenotype association to improve the pediatric TSC pharmacotherapeutic outcome. Patients & methods: Ten pediatric TSC patients were recruited. Concomitant treatment and main metabolic enzymes and transporter coding gene variants of EVR were analyzed. Results: Significant associations were found between CYP3A4*22 allele and concomitant treatment with valproic acid (CYP3A4-inhibitor) with a poor metabolizer phenotype and the presence of pneumonia. Conclusion: This is the first pharmacogenetic study of EVR in pediatric TSC patients. The authors propose to consider concomitant treatment and pharmacogenetics due to their multifactorial status.
Collapse
Affiliation(s)
- Julia Concha
- Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain
| | - Estela Sangüesa
- Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain
| | - Jose Luis Peña
- Neuropediatrics Area, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | | | | |
Collapse
|
52
|
Curatolo P, Trivisano M, Specchio N. Updated Genotype-Phenotype Correlations in TSC. Semin Pediatr Neurol 2023; 47:101086. [PMID: 37919037 DOI: 10.1016/j.spen.2023.101086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
GENOTYPE/PHENOTYPE CORRELATIONS IN TUBEROUS SCLEROSIS COMPLEX Paolo Curatolo MD, Romina Moavero MD, Denis Roberto, Federica Graziola Seminars in Pediatric Neurology Volume 22, Issue 4, December 2015, Pages 259-273 Tuberous sclerosis complex (TSC) is an autosomal dominant disorder characterized by the development of widespread hamartomatous lesions in various organs, including brain, skin, kidneys, heart, and eyes. Central nervous system is almost invariably involved, with up to 85% of patients presenting with epilepsy, and at least half of patients having intellectual disability or other neuropsychiatric disorders including autism spectrum disorder. TSC is caused by the mutation in one of the 2 genes TSC1, at 9q34, and TSC2, at 16p13.3. They respectively encode for hamartin and tuberin, which form an intracellular complex inhibiting the mammalian target of rapamycin. Mammalian target of rapamycin overactivation following the genetic defect determines the cell growth and proliferation responsible for TSC-related lesions, as well as the alterations in neuronal excitability and synaptogenesis leading to epilepsy and neuropsychiatric disorders. A causative mutation for the disorder is identified in about 85% of patients with a clinical diagnosis of TSC. Mosaicism and technology limits likely explain most of the no mutation identified cases. This review confirms that patients with TSC2 mutations considered as a group usually present a more severe phenotype, characterized by higher number of tubers, earlier age at seizure onset and higher prevalence of intellectual disability. However, the clinical phenotype of the disease presents a high variability, thus making the prediction of the phenotype on an individual basis still challenging. The increasing application of new molecular techniques to subjects with TSC has the potential to significantly reduce the rate of patients with no mutation demonstrated and to identify an increasing higher number of mutations. This would hopefully allow a better characterization of higher risk mutations, which might help clinicians to plan individualized surveillance plans. Furthermore, the increasing availability of disease registries to collect clinical and genetics data of patients help to define more valid and clinically oriented genotype or phenotype correlations.
Collapse
Affiliation(s)
- Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
| | - Marina Trivisano
- Child Neurology, Epilepsy and Movement Disorders, Bambino Gesù, IRCCS Children's Hospital, Full Member of European Reference Network EpiCARE, Rome, Italy
| | - Nicola Specchio
- Child Neurology, Epilepsy and Movement Disorders, Bambino Gesù, IRCCS Children's Hospital, Full Member of European Reference Network EpiCARE, Rome, Italy.
| |
Collapse
|
53
|
Saini L, Mukherjee S, Gunasekaran PK, Malhi P, Saini AG, Sharma R, Sharawat IK, Suthar R, Sahu JK, Sankhyan N. Neurodevelopmental Outcomes of a Cohort of Children with Tuberous Sclerosis Complex with Epileptic Spasms. Neuropediatrics 2023; 54:335-338. [PMID: 37379859 DOI: 10.1055/s-0043-1770937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
The neurodevelopmental outcomes in children with tuberous sclerosis complex (TSC) with epileptic spasms remain underdiagnosed and might be responsible for significant morbidity and mortality burdens, even after spasms abate. The study was a cross-sectional study over 18 months at a tertiary care pediatric hospital, involving 30 children with TSC who had epileptic spasms. They were assessed with Diagnostic and Statistical Manual of Mental Disorders-5 criteria for autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), and intellectual disability (ID), and childhood psychopathology measurement schedule (CPMS) for behavioral disorders. The median age at onset of epileptic spasms was 6.5 (1-12) months, and the age at enrolment was 5 (1-15) years. Of 30 children, 2 (6.7%) had only ADHD, 15 (50%) had only ID/GDD (global developmental delay), 4 (13.3%) had ASD and ID/GDD, 3 (10%) had ADHD and ID/GDD, and 6 (20%) had none. The median intelligence quotient/development quotient (IQ/DQ) score was 60.5 (20-105). CPMS assessment revealed significant behavioral abnormalities in almost half the children. Eight (26.7%) patients were completely seizure-free for at least 2 years, 8 (26.7%) had generalized tonic-clonic seizures, 11 (36.6%) had focal epilepsy, and 3 (10%) had evolved into Lennox-Gastaut syndrome. A high proportion of neurodevelopment disorders, including ASD, ADHD, ID/GDD, and behavioral disorders were seen in this pilot study with a small cohort of children with TSC with epileptic spasms.
Collapse
Affiliation(s)
- Lokesh Saini
- Department of Pediatrics, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Swetlana Mukherjee
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Prahbhjot Malhi
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arushi Gahlot Saini
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajni Sharma
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Indar Kumar Sharawat
- Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Renu Suthar
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jitendra Kumar Sahu
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Naveen Sankhyan
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
54
|
Perucca E, White HS, Bialer M. New GABA-Targeting Therapies for the Treatment of Seizures and Epilepsy: II. Treatments in Clinical Development. CNS Drugs 2023; 37:781-795. [PMID: 37603261 PMCID: PMC10501930 DOI: 10.1007/s40263-023-01025-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 08/22/2023]
Abstract
The inhibitory neurotransmitter γ-aminobutyric acid (GABA) plays an important role in the modulation of neuronal excitability, and a disruption of GABAergic transmission contributes to the pathogenesis of some seizure disorders. Although many currently available antiseizure medications do act at least in part by potentiating GABAergic transmission, there is an opportunity for further research aimed at developing more innovative GABA-targeting therapies. The present article summarises available evidence on a number of such treatments in clinical development. These can be broadly divided into three groups. The first group consists of positive allosteric modulators of GABAA receptors and includes Staccato® alprazolam (an already marketed benzodiazepine being repurposed in epilepsy as a potential rescue inhalation treatment for prolonged and repetitive seizures), the α2/3/5 subtype-selective agents darigabat and ENX-101, and the orally active neurosteroids ETX155 and LPCN 2101. A second group comprises two drugs already marketed for non-neurological indications, which could be repurposed as treatments for seizure disorders. These include bumetanide, a diuretic agent that has undergone clinical trials in phenobarbital-resistant neonatal seizures and for which the rationale for further development in this indication is under debate, and ivermectin, an antiparasitic drug currently investigated in a randomised double-blind trial in focal epilepsy. The last group comprises a series of highly innovative therapies, namely GABAergic interneurons (NRTX-001) delivered via stereotactic cerebral implantation as a treatment for mesial temporal lobe epilepsy, an antisense oligonucleotide (STK-001) aimed at upregulating NaV1.1 currents and restoring the function of GABAergic interneurons, currently tested in a trial in patients with Dravet syndrome, and an adenoviral vector-based gene therapy (ETX-101) scheduled for investigation in Dravet syndrome. Another agent, a subcutaneously administered neuroactive peptide (NRP2945) that reportedly upregulates the expression of GABAA receptor α and β subunits is being investigated, with Lennox-Gastaut syndrome and other epilepsies as proposed indications. The diversity of the current pipeline underscores a strong interest in the GABA system as a target for new treatment development in epilepsy. To date, limited clinical data are available for these investigational treatments and further studies are required to assess their potential value in addressing unmet needs in epilepsy management.
Collapse
Affiliation(s)
- Emilio Perucca
- Department of Medicine (Austin Health), Melbourne Brain Centre, The University of Melbourne, 245 Burgundy Street, Melbourne, VIC, 3084, Australia.
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - H Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Meir Bialer
- Faculty of Medicine, Institute of Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
- David R. Bloom Center for Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
55
|
Di Sapia R, Rizzi M, Moro F, Lisi I, Caccamo A, Ravizza T, Vezzani A, Zanier ER. ECoG spiking activity and signal dimension are early predictive measures of epileptogenesis in a translational mouse model of traumatic brain injury. Neurobiol Dis 2023; 185:106251. [PMID: 37536383 DOI: 10.1016/j.nbd.2023.106251] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/19/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023] Open
Abstract
The latency between traumatic brain injury (TBI) and the onset of epilepsy (PTE) represents an opportunity for counteracting epileptogenesis. Antiepileptogenesis trials are hampered by the lack of sensitive biomarkers that allow to enrich patient's population at-risk for PTE. We aimed to assess whether specific ECoG signals predict PTE in a clinically relevant mouse model with ∼60% epilepsy incidence. TBI was provoked in adult CD1 male mice by controlled cortical impact on the left parieto-temporal cortex, then mice were implanted with two perilesional cortical screw electrodes and two similar electrodes in the hemisphere contralateral to the lesion site. Acute seizures and spikes/sharp waves were ECoG-recorded during 1 week post-TBI. These early ECoG events were analyzed according to PTE incidence as assessed by measuring spontaneous recurrent seizures (SRS) at 5 months post-TBI. We found that incidence, number and duration of acute seizures during 3 days post-TBI were similar in PTE mice and mice not developing epilepsy (No SRS mice). Control mice with cortical electrodes (naïve, n = 5) or with electrodes and craniotomy (sham, n = 5) exhibited acute seizures but did not develop epilepsy. The daily number of spikes/sharp waves at the perilesional electrodes was increased similarly in PTE (n = 15) and No SRS (n = 8) mice vs controls (p < 0.05, n = 10) from day 2 post-injury. Differently, the daily number of spikes/sharp waves at both contralateral electrodes showed a progressive increase in PTE mice vs No SRS and control mice. In particular, spikes number was higher in PTE vs No SRS mice (p < 0.05) at 6 and 7 days post-TBI, and this measure predicted epilepsy development with high accuracy (AUC = 0.77, p = 0.03; CI 0.5830-0.9670). The cut-off value was validated in an independent cohort of TBI mice (n = 12). The daily spike number at the contralateral electrodes showed a circadian distribution in PTE mice which was not observed in No SRS mice. Analysis of non-linear dynamics at each electrode site showed changes in dimensionality during 4 days post-TBI. This measure yielded the best discrimination between PTE and No SRS mice (p < 0.01) at the cortical electrodes contralateral to injury. Data show that epileptiform activity contralateral to the lesion site has the the highest predictive value for PTE in this model reinforcing the hypothesis that the hemisphere contralateral to the lesion core may drive epileptogenic networks after TBI.
Collapse
Affiliation(s)
- Rossella Di Sapia
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Massimo Rizzi
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Federico Moro
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Ilaria Lisi
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Alessia Caccamo
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Teresa Ravizza
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Annamaria Vezzani
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Elisa R Zanier
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| |
Collapse
|
56
|
Bebin EM, Peters JM, Porter BE, McPherson TO, O’Kelley S, Sahin M, Taub KS, Rajaraman R, Randle SC, McClintock WM, Koenig MK, Frost MD, Northrup HA, Werner K, Nolan DA, Wong M, Krefting JL, Biasini F, Peri K, Cutter G, Krueger DA. Early Treatment with Vigabatrin Does Not Decrease Focal Seizures or Improve Cognition in Tuberous Sclerosis Complex: The PREVeNT Trial. Ann Neurol 2023; 95:10.1002/ana.26778. [PMID: 37638552 PMCID: PMC10899525 DOI: 10.1002/ana.26778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/07/2023] [Accepted: 08/20/2023] [Indexed: 08/29/2023]
Abstract
OBJECTIVE This study was undertaken to test the hypothesis that early vigabatrin treatment in tuberous sclerosis complex (TSC) infants improves neurocognitive outcome at 24 months of age. METHODS A phase IIb multicenter randomized double-blind placebo-controlled trial was conducted of vigabatrin at first epileptiform electroencephalogram (EEG) versus vigabatrin at seizure onset in infants with TSC. Primary outcome was Bayley Scales of Infant and Toddler Development, Third Edition (Bayley-III) cognitive assessment score at 24 months. Secondary outcomes were prevalence of drug-resistant epilepsy, additional developmental outcomes, and safety of vigabatrin. RESULTS Of 84 infants enrolled, 12 were screen failures, 4 went straight to open label vigabatrin, and 12 were not randomized (normal EEG throughout). Fifty-six were randomized to early vigabatrin (n = 29) or placebo (n = 27). Nineteen of 27 in the placebo arm transitioned to open label vigabatrin, with a median delay of 44 days after randomization. Bayley-III cognitive composite scores at 24 months were similar for participants randomized to vigabatrin or placebo. Additionally, no significant differences were found between groups in overall epilepsy incidence and drug-resistant epilepsy at 24 months, time to first seizure after randomization, and secondary developmental outcomes. Incidence of infantile spasms was lower and time to spasms after randomization was later in the vigabatrin group. Adverse events were similar across groups. INTERPRETATION Preventative treatment with vigabatrin based on EEG epileptiform activity prior to seizure onset does not improve neurocognitive outcome at 24 months in TSC children, nor does it delay onset or lower the incidence of focal seizures and drug-resistant epilepsy at 24 months. Preventative vigabatrin was associated with later time to onset and lower incidence of infantile spasms. ANN NEUROL 2023.
Collapse
Affiliation(s)
| | - Jurriaan M. Peters
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Sarah O’Kelley
- Department of Psychology University of Alabama at Birmingham AL
| | - Mustafa Sahin
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | | | | | | | | | - Mary Kay Koenig
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth) and Children’s Memorial Hermann Hospital, Houston, TX
| | | | - Hope A. Northrup
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth) and Children’s Memorial Hermann Hospital, Houston, TX
| | | | | | - Michael Wong
- Department of Neuroscience Washington University in Saint Louis
| | | | - Fred Biasini
- Department of Psychology University of Alabama at Birmingham AL
| | - Kalyani Peri
- Department of Biostatistics, University of Alabama at Birmingham AL
| | - Gary Cutter
- Department of Biostatistics, University of Alabama at Birmingham AL
| | | |
Collapse
|
57
|
Tipton AE, Del Angel YC, Hixson K, Carlsen J, Strode D, Busquet N, Mesches MH, Gonzalez MI, Napoli E, Russek SJ, Brooks-Kayal AR. Selective Neuronal Knockout of STAT3 Function Inhibits Epilepsy Progression, Improves Cognition, and Restores Dysregulated Gene Networks in a Temporal Lobe Epilepsy Model. Ann Neurol 2023; 94:106-122. [PMID: 36935347 PMCID: PMC10313781 DOI: 10.1002/ana.26644] [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: 12/16/2022] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/21/2023]
Abstract
OBJECTIVE Temporal lobe epilepsy (TLE) is a progressive disorder mediated by pathological changes in molecular cascades and hippocampal neural circuit remodeling that results in spontaneous seizures and cognitive dysfunction. Targeting these cascades may provide disease-modifying treatments for TLE patients. Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) inhibitors have emerged as potential disease-modifying therapies; a more detailed understanding of JAK/STAT participation in epileptogenic responses is required, however, to increase the therapeutic efficacy and reduce adverse effects associated with global inhibition. METHODS We developed a mouse line in which tamoxifen treatment conditionally abolishes STAT3 signaling from forebrain excitatory neurons (nSTAT3KO). Seizure frequency (continuous in vivo electroencephalography) and memory (contextual fear conditioning and motor learning) were analyzed in wild-type and nSTAT3KO mice after intrahippocampal kainate (IHKA) injection as a model of TLE. Hippocampal RNA was obtained 24 h after IHKA and subjected to deep sequencing. RESULTS Selective STAT3 knock-out in excitatory neurons reduced seizure progression and hippocampal memory deficits without reducing the extent of cell death or mossy fiber sprouting induced by IHKA injection. Gene expression was rescued in major networks associated with response to brain injury, neuronal plasticity, and learning and memory. We also provide the first evidence that neuronal STAT3 may directly influence brain inflammation. INTERPRETATION Inhibiting neuronal STAT3 signaling improved outcomes in an animal model of TLE, prevented progression of seizures and cognitive co-morbidities while rescuing pathogenic changes in gene expression of major networks associated with epileptogenesis. Specifically targeting neuronal STAT3 may be an effective disease-modifying strategy for TLE. ANN NEUROL 2023;94:106-122.
Collapse
Affiliation(s)
- Allison E. Tipton
- Graduate Program for Neuroscience, Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Yasmin Cruz Del Angel
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Kathryn Hixson
- Graduate Program for Neuroscience, Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Jessica Carlsen
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Dana Strode
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nicolas Busquet
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael H. Mesches
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marco I. Gonzalez
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Eleonora Napoli
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Shelley J. Russek
- Graduate Program for Neuroscience, Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Amy R. Brooks-Kayal
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
58
|
Prezioso G, Chiarelli F, Matricardi S. Efficacy and safety of vigabatrin in patients with tuberous sclerosis complex and infantile epileptic spasm syndrome: a systematic review. Expert Rev Neurother 2023; 23:661-671. [PMID: 37243682 DOI: 10.1080/14737175.2023.2216385] [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: 02/14/2023] [Accepted: 05/17/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Tuberous sclerosis complex (TSC) is a common genetic cause of epilepsy. Infantile epileptic spasm syndrome (IESS) is often the presenting neurologic feature, progressively evolving into refractory epilepsy. Vigabatrin (VGB) is often used in clinical practice as a first-line therapy in TSC with IESS. This systematic review aims to collect and analyze the efficacy data about VGB in TSC cases with IESS, in order to evaluate the strength of evidence in the literature. METHODS A systematic search of trials, observational studies, and case series involving patients with TSC and IESS treated with VGB was performed using MEDLINE, CENTRAL, and the US NIH Clinical Trials Registry. Single case studies, animal and non-English language studies were excluded. Seventeen studies were selected, of which 3 were RCTs and 14 were observational studies. RESULTS An overall response rate of 67% (231/343 responders) resulted from the analysis, with a spasm-free rate restricted to RCTs of 88% (29/33 subjects). CONCLUSIONS Although all the studies analyzed reported beneficial effects of VGB in TSC patients with IESS, with higher response rates in comparison to non-TSC subjects with IESS, a low level of evidence and high heterogeneity do not guarantee sufficient strength for therapeutic recommendations.
Collapse
Affiliation(s)
- Giovanni Prezioso
- Pediatric Neurologist, Department of Pediatrics, G. D'Annunzio University, Chieti, Italy
| | - Francesco Chiarelli
- Department of Pediatrics, Department of Pediatrics, G. D'Annunzio University, Chieti, Italy
| | - Sara Matricardi
- Pediatric Neurologist, Department of Pediatrics, G. D'Annunzio University, Chieti, Italy
| |
Collapse
|
59
|
Olson HE, Demarest S, Pestana-Knight E, Moosa AN, Zhang X, Pérez-Pérez JR, Weisenberg J, O'Connor Prange E, Marsh ED, Rajaraman RR, Suter B, Katyayan A, Haviland I, Daniels C, Zhang B, Greene C, DeLeo M, Swanson L, Love-Nichols J, Benke T, Harini C, Poduri A. Epileptic spasms in CDKL5 deficiency disorder: Delayed treatment and poor response to first-line therapies. Epilepsia 2023; 64:1821-1832. [PMID: 37114835 PMCID: PMC10524264 DOI: 10.1111/epi.17630] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVE We aimed to assess the treatment response of infantile-onset epileptic spasms (ES) in CDKL5 deficiency disorder (CDD) vs other etiologies. METHODS We evaluated patients with ES from the CDKL5 Centers of Excellence and the National Infantile Spasms Consortium (NISC), with onset from 2 months to 2 years, treated with adrenocorticotropic hormone (ACTH), oral corticosteroids, vigabatrin, and/or the ketogenic diet. We excluded children with tuberous sclerosis complex, trisomy 21, or unknown etiology with normal development because of known differential treatment responses. We compared the two cohorts for time to treatment and ES remission at 14 days and 3 months. RESULTS We evaluated 59 individuals with CDD (79% female, median ES onset 6 months) and 232 individuals from the NISC database (46% female, median onset 7 months). In the CDD cohort, seizures prior to ES were common (88%), and hypsarrhythmia and its variants were present at ES onset in 34%. Initial treatment with ACTH, oral corticosteroids, or vigabatrin started within 1 month of ES onset in 27 of 59 (46%) of the CDD cohort and 182 of 232 (78%) of the NISC cohort (p < .0001). Fourteen-day clinical remission of ES was lower for the CDD group (26%, 7/27) than for the NISC cohort (58%, 106/182, p = .0002). Sustained ES remission at 3 months occurred in 1 of 27 (4%) of CDD patients vs 96 of 182 (53%) of the NISC cohort (p < .0001). Comparable results were observed with longer lead time (≥1 month) or prior treatment. Ketogenic diet, used within 3 months of ES onset, resulted in ES remission at 1 month, sustained at 3 months, in at least 2 of 13 (15%) individuals with CDD. SIGNIFICANCE Compared to the broad group of infants with ES, children with ES in the setting of CDD often experience longer lead time to treatment and respond poorly to standard treatments. Development of alternative treatments for ES in CDD is needed.
Collapse
Affiliation(s)
- Heather E Olson
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Scott Demarest
- Department of Pediatrics, School of Medicine, Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Elia Pestana-Knight
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ahsan N Moosa
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Xiaoming Zhang
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - José R Pérez-Pérez
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Judy Weisenberg
- Department of Pediatric Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Erin O'Connor Prange
- Division of Child Neurology, Children's Hospital of Philadelphia, Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eric D Marsh
- Division of Child Neurology, Children's Hospital of Philadelphia, Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rajsekar R Rajaraman
- Division of Pediatric Neurology, David Geffen School of Medicine and UCLA Mattel Children's Hospital, Los Angeles, California, USA
| | - Bernhard Suter
- Department of Pediatrics and Neurology, Baylor College of Medicine, Texas Children's Hospital, Houston, Houston, Texas, USA
| | - Akshat Katyayan
- Department of Pediatrics and Neurology, Baylor College of Medicine, Texas Children's Hospital, Houston, Houston, Texas, USA
| | - Isabel Haviland
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Carolyn Daniels
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Bo Zhang
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlin Greene
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Michelle DeLeo
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Lindsay Swanson
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jamie Love-Nichols
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Timothy Benke
- Department of Pediatrics, School of Medicine, Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Chellamani Harini
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Annapurna Poduri
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
60
|
Levine A, Davis P, Zhang B, Peters J, Filip-Dhima R, Warfield SK, Prohl A, Capal J, Krueger D, Bebin EM, Northrup H, Wu JY, Sahin M. Epilepsy Severity Is Associated With Head Circumference and Growth Rate in Infants With Tuberous Sclerosis Complex. Pediatr Neurol 2023; 144:26-32. [PMID: 37119787 PMCID: PMC10330061 DOI: 10.1016/j.pediatrneurol.2023.03.015] [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: 08/25/2022] [Revised: 02/05/2023] [Accepted: 03/23/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND Abnormal brain growth in tuberous sclerosis complex (TSC) reflects abnormalities in cellular proliferation and differentiation and results in epilepsy and other neurological manifestations. Head circumference (HC) as a proxy for brain volume may provide an easily tracked clinical measure of brain overgrowth and neurological disease burden. This study investigated the relationship between HC and epilepsy severity in infants with TSC. METHODS Prospective multicenter observational study of children from birth to three years with TSC. Epilepsy data were collected from clinical history, and HC was collected at study visits at age three, six, nine, 12, 18, 24, and 36 months. Epilepsy severity was classified as no epilepsy, low epilepsy severity (one seizure type and one or two antiepileptic drugs [AEDs]), moderate epilepsy severity (either two to three seizure types and one to two AEDs or one seizure type and more than three AEDs), or high epilepsy severity (two to three seizure types and more than three AEDs). RESULTS As a group, children with TSC had HCs approximately 1 S.D. above the mean World Health Organization (WHO) reference by age one year and demonstrated more rapid growth than the normal population reference. Males with epilepsy had larger HCs than those without. Compared with the WHO reference population, infants with TSC and no epilepsy or low or moderate epilepsy had an increased early HC growth rate, whereas those with severe epilepsy had an early larger HC but did not have a faster growth rate. CONCLUSIONS Infants and young children with TSC have larger HCs than typical growth norms and have differing rates of head growth depending on the severity of epilepsy.
Collapse
Affiliation(s)
- Alexis Levine
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts.
| | - Peter Davis
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Bo Zhang
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Jurriaan Peters
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts; Division of Epilepsy and Clinical Neurophysiology, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Rajna Filip-Dhima
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Simon K Warfield
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anna Prohl
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jamie Capal
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Darcy Krueger
- Department of Neurology and Rehabilitation Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - E Martina Bebin
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hope Northrup
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Joyce Y Wu
- Division of Pediatric Neurology, University of California at Los Angeles Mattel Children's Hospital, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts; Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts.
| |
Collapse
|
61
|
Pearsson K, Björk Werner J, Lundgren J, Gränse L, Karlsson E, Källén K, Eklund EA, Bekassy Z. Childhood tuberous sclerosis complex in southern Sweden: a paradigm shift in diagnosis and treatment. BMC Pediatr 2023; 23:329. [PMID: 37386496 PMCID: PMC10308728 DOI: 10.1186/s12887-023-04137-4] [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: 11/08/2022] [Accepted: 06/16/2023] [Indexed: 07/01/2023] Open
Abstract
AIM To investigate the complete clinical spectrum of individuals with paediatric tuberous sclerosis complex in southern Sweden and explore changes over time. METHODS In this retrospective observational study, 52 individuals aged up to 18 years at the study start were followed-up at regional hospitals and centres for habilitation from 2000 to 2020. RESULTS Cardiac rhabdomyoma was detected prenatally/neonatally in 69.2% of the subjects born during the latest ten years of the study period. Epilepsy was diagnosed in 82.7% of subjects, and 10 (19%) were treated with everolimus, mainly (80%) for a neurological indication. Renal cysts were detected in 53%, angiomyolipomas in 47%, astrocytic hamartomas in 28% of the individuals. There was a paucity of standardized follow-up of cardiac, renal, and ophthalmological manifestations and no structured transition to adult care. CONCLUSION Our in-depth analysis shows a clear shift towards an earlier diagnosis of tuberous sclerosis complex in the latter part of the study period, where more than 60% of cases showed evidence of this condition already in utero due to the presence of a cardiac rhabdomyoma. This allows for preventive treatment of epilepsy with vigabatrin and early intervention with everolimus for potential mitigation of other symptoms of tuberous sclerosis complex.
Collapse
Affiliation(s)
- Kevin Pearsson
- Department of Clinical Sciences Lund, Clinical Sciences Helsingborg, Lund University, Lund, Sweden
- Department of Paediatrics, Helsingborg General Hospital, Helsingborg, Sweden
| | | | - Johan Lundgren
- Section for Paediatric Neurology, Skåne University Hospital, Lund, Sweden
| | - Lotta Gränse
- Department of Ophthalmology, Skåne University Hospital, Lund, Sweden
| | - Emma Karlsson
- Department of Paediatrics, Blekingesjukhuset, Karlskrona, Sweden
| | - Kristina Källén
- Department of Clinical Sciences Lund, Clinical Sciences Helsingborg, Lund University, Lund, Sweden
| | - Erik A Eklund
- Section for Paediatric Neurology, Skåne University Hospital, Lund, Sweden
- Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Zivile Bekassy
- Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden.
- Section for Paediatric Nephrology, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
62
|
Perucca E, Perucca P, White HS, Wirrell EC. Drug resistance in epilepsy. Lancet Neurol 2023:S1474-4422(23)00151-5. [PMID: 37352888 DOI: 10.1016/s1474-4422(23)00151-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/23/2023] [Accepted: 04/06/2023] [Indexed: 06/25/2023]
Abstract
Drug resistance is estimated to affect about a third of individuals with epilepsy, but its prevalence differs in relation to the epilepsy syndrome, the cause of epilepsy, and other factors such as age of seizure onset and presence of associated neurological deficits. Although drug-resistant epilepsy is not synonymous with unresponsiveness to any drug treatment, the probability of achieving seizure freedom on a newly tried medication decreases with increasing number of previously failed treatments. After two appropriately used antiseizure medications have failed to control seizures, individuals should be referred whenever possible to a comprehensive epilepsy centre for diagnostic re-evaluation and targeted management. The feasibility of epilepsy surgery and other treatments, including those targeting the cause of epilepsy, should be considered early after diagnosis. Substantial evidence indicates that a delay in identifying an effective treatment can adversely affect ultimate outcome and carry an increased risk of cognitive disability, other comorbidities, and premature mortality. Research on mechanisms of drug resistance and novel therapeutics is progressing rapidly, and potentially improved treatments, including those targeting disease modification, are on the horizon.
Collapse
Affiliation(s)
- Emilio Perucca
- Department of Medicine, Austin Health, University of Melbourne, Melbourne, VIC, Australia; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Piero Perucca
- Department of Medicine, Austin Health, University of Melbourne, Melbourne, VIC, Australia; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Bladin-Berkovic Comprehensive Epilepsy Program, Department of Neurology, Austin Health, Melbourne, VIC, Australia; Department of Neurology, Royal Melbourne Hospital, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - H Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Elaine C Wirrell
- Divisions of Child and Adolescent Neurology and Epilepsy, Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
63
|
Gipson TT, Oller DK, Messinger DS, Perry LK. Understanding speech and language in tuberous sclerosis complex. Front Hum Neurosci 2023; 17:1149071. [PMID: 37323931 PMCID: PMC10267356 DOI: 10.3389/fnhum.2023.1149071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Tuberous Sclerosis Complex (TSC), is a neurocutaneous disorder, associated with a high prevalence of autism spectrum disorder (ASD; ∼50% of individuals). As TSC is a leading cause of syndromic ASD, understanding language development in this population would not only be important for individuals with TSC but may also have implications for those with other causes of syndromic and idiopathic ASD. In this mini review, we consider what is known about language development in this population and how speech and language in TSC are related to ASD. Although up to 70% of individuals with TSC report language difficulties, much of the limited research to date on language in TSC has been based on summary scores from standardized assessments. Missing is a detailed understanding of the mechanisms driving speech and language in TSC and how they relate to ASD. Here, we review recent work suggesting that canonical babbling and volubility-two precursors of language development that predict the emergence of speech and are delayed in infants with idiopathic ASD-are also delayed in infants with TSC. We then look to the broader literature on language development to identify other early precursors of language development that tend to be delayed in children with autism as a guide for future research on speech and language in TSC. We argue that vocal turn-taking, shared attention, and fast mapping are three such skills that can provide important information about how speech and language develop in TSC and where potential delays come from. The overall goal of this line of research is to not only illuminate the trajectory of language in TSC with and without ASD, but to ultimately find strategies for earlier recognition and treatment of the pervasive language difficulties in this population.
Collapse
Affiliation(s)
- Tanjala T Gipson
- Department of Pediatrics, Le Bonheur Children's Hospital, The Boling Center for Developmental Disabilities, University of Tennessee Health Science Center, Memphis, TN, United States
| | - D Kimbrough Oller
- School of Communication Sciences and Disorders, Institute for Intelligent Systems, The University of Memphis, Memphis, TN, United States
| | - Daniel S Messinger
- Department of Psychology, University of Miami, Miami, FL, United States
- Department of Pediatrics, University of Miami, Miami, FL, United States
- Department of Electrical and Computer Engineering, University of Miami, Miami, FL, United States
- Department of Music Engineering, University of Miami, Miami, FL, United States
| | - Lynn K Perry
- Department of Psychology, University of Miami, Miami, FL, United States
| |
Collapse
|
64
|
Bychkova E, Dorofeeva M, Levov A, Kislyakov A, Karandasheva K, Strelnikov V, Anoshkin K. Specific Features of Focal Cortical Dysplasia in Tuberous Sclerosis Complex. Curr Issues Mol Biol 2023; 45:3977-3996. [PMID: 37232723 DOI: 10.3390/cimb45050254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Patients with tuberous sclerosis complex present with cognitive, behavioral, and psychiatric impairments, such as intellectual disabilities, autism spectrum disorders, and drug-resistant epilepsy. It has been shown that these disorders are associated with the presence of cortical tubers. Tuberous sclerosis complex results from inactivating mutations in the TSC1 or TSC2 genes, resulting in hyperactivation of the mTOR signaling pathway, which regulates cell growth, proliferation, survival, and autophagy. TSC1 and TSC2 are classified as tumor suppressor genes and function according to Knudson's two-hit hypothesis, which requires both alleles to be damaged for tumor formation. However, a second-hit mutation is a rare event in cortical tubers. This suggests that the molecular mechanism of cortical tuber formation may be more complicated and requires further research. This review highlights the issues of molecular genetics and genotype-phenotype correlations, considers histopathological characteristics and the mechanism of morphogenesis of cortical tubers, and also presents data on the relationship between these formations and the development of neurological manifestations, as well as treatment options.
Collapse
Affiliation(s)
- Ekaterina Bychkova
- Research Centre for Medical Genetics, Moskvorechye Street 1, 115522 Moscow, Russia
- Faculty of Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova Street 1, 117997 Moscow, Russia
| | - Marina Dorofeeva
- Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery, Pirogov Russian National Research Medical University, Taldomskaya 2, 125412 Moscow, Russia
| | - Aleksandr Levov
- Morozov Children's City Clinical Hospital, 4th Dobryninsky Lane, 1/9, 119049 Moscow, Russia
| | - Alexey Kislyakov
- Morozov Children's City Clinical Hospital, 4th Dobryninsky Lane, 1/9, 119049 Moscow, Russia
| | | | - Vladimir Strelnikov
- Research Centre for Medical Genetics, Moskvorechye Street 1, 115522 Moscow, Russia
| | - Kirill Anoshkin
- Research Centre for Medical Genetics, Moskvorechye Street 1, 115522 Moscow, Russia
| |
Collapse
|
65
|
Abstract
PURPOSE OF REVIEW Epilepsy affects 70 million people worldwide and is a significant cause of morbidity and early mortality. The mainstay of therapy is oral medications. Epilepsy drug development is escalating, driven by continued drug resistance in up to a third of epilepsy patients. Treatment development now focuses on discovery of novel mechanisms of action and syndrome-specific therapies. RECENT FINDINGS Difficult-to-treat epilepsy related to conditions including tuberous sclerosis complex (TSC), Lennox Gastaut syndrome (LGS) and Dravet syndrome (DS) have been the target of recent developments. Disease-modifying therapy for epilepsy related to TSC with vigabatrin at onset of first electroencephalographic epileptiform changes, rather than after first clinical seizure, has demonstrated strongly positive seizure and developmental outcomes. Fenfluramine, approved for DS and, more recently, LGS, has robust data supporting efficacy, safety/tolerability, as well as mortality, quality of life and cognitive function. Rescue therapy has expanded to include better tolerated benzodiazepines in the form of nasal midazolam and valium. Cenobamate, a first-in-class inactivator of the persistent voltage-gated sodium channel and approved for adult partial onset epilepsy, has exceptional efficacy and tolerability and will be expanded to children and to generalized onset epilepsy in adults. SUMMARY The repertoire of available and developmental therapies for epilepsy is rapidly expanding, and now includes disease-modifying vigabatrin in TSC and agents with extraordinary efficacy, fenfluramine and cenobamate.
Collapse
Affiliation(s)
- Amanda W Pong
- Mid-Atlantic Epilepsy and Sleep Center, Mid-Atlantic Neurological Institute, Bethesda, Maryland, USA
| | | | | |
Collapse
|
66
|
Reynolds A, Vranic-Peters M, Lai A, Grayden DB, Cook MJ, Peterson A. Prognostic interictal electroencephalographic biomarkers and models to assess antiseizure medication efficacy for clinical practice: A scoping review. Epilepsia 2023; 64:1125-1174. [PMID: 36790369 DOI: 10.1111/epi.17548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Antiseizure medication (ASM) is the primary treatment for epilepsy. In clinical practice, methods to assess ASM efficacy (predict seizure freedom or seizure reduction), during any phase of the drug treatment lifecycle, are limited. This scoping review identifies and appraises prognostic electroencephalographic (EEG) biomarkers and prognostic models that use EEG features, which are associated with seizure outcomes following ASM initiation, dose adjustment, or withdrawal. We also aim to summarize the population and context in which these biomarkers and models were identified and described, to understand how they could be used in clinical practice. Between January 2021 and October 2022, four databases, references, and citations were systematically searched for ASM studies investigating changes to interictal EEG or prognostic models using EEG features and seizure outcomes. Study bias was appraised using modified Quality in Prognosis Studies criteria. Results were synthesized into a qualitative review. Of 875 studies identified, 93 were included. Biomarkers identified were classed as qualitative (visually identified by wave morphology) or quantitative. Qualitative biomarkers include identifying hypsarrhythmia, centrotemporal spikes, interictal epileptiform discharges (IED), classifying the EEG as normal/abnormal/epileptiform, and photoparoxysmal response. Quantitative biomarkers were statistics applied to IED, high-frequency activity, frequency band power, current source density estimates, pairwise statistical interdependence between EEG channels, and measures of complexity. Prognostic models using EEG features were Cox proportional hazards models and machine learning models. There is promise that some quantitative EEG biomarkers could be used to assess ASM efficacy, but further research is required. There is insufficient evidence to conclude any specific biomarker can be used for a particular population or context to prognosticate ASM efficacy. We identified a potential battery of prognostic EEG biomarkers, which could be combined with prognostic models to assess ASM efficacy. However, many confounders need to be addressed for translation into clinical practice.
Collapse
Affiliation(s)
- Ashley Reynolds
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurosciences, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Michaela Vranic-Peters
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurosciences, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Alan Lai
- Department of Neurosciences, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - David B Grayden
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurosciences, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia.,Graeme Clark Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Mark J Cook
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurosciences, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia.,Graeme Clark Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Andre Peterson
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurosciences, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia.,Graeme Clark Institute, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
67
|
Gettings JV, Shafi S, Boyd J, Snead OC, Rutka J, Drake J, McCoy B, Jain P, Whitney R, Go C. The Epilepsy Surgery Experience in Children With Infantile Epileptic Spasms Syndrome at a Tertiary Care Center in Canada. J Child Neurol 2023; 38:113-120. [PMID: 36788207 DOI: 10.1177/08830738231151993] [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] [Indexed: 02/16/2023]
Abstract
BACKGROUND Infantile epileptic spasms syndrome is an epileptic encephalopathy, characterized by spasms, hypsarrhythmia, and developmental regression. Appropriately selected patients with infantile epileptic spasms syndrome may be candidates for epilepsy surgery. METHODS This is a single-center retrospective case series of children 0-18 years with a current or previous diagnosis of infantile epileptic spasms syndrome with a lesion on magnetic resonance imaging (MRI) and/or positron emission tomography scan who underwent epilepsy surgery at The Hospital for Sick Children (HSC) in Toronto, Canada. The records of 223 patients seen in the infantile epileptic spasms syndrome clinic were reviewed. RESULTS Nineteen patients met inclusion criteria. The etiology of infantile epileptic spasms syndrome was encephalomalacia in 6 patients (32%), malformations of cortical development in 12 patients (63%), and atypical hypoglycemic injury in 1 patient (5%). Nine patients (47%) underwent hemispherectomy, and 10 patients (53%) underwent lobectomy/lesionectomy. Three patients (16%) underwent a second epilepsy surgery. Fifteen patients (79%) were considered ILAE seizure outcome class 1 (completely seizure free; no auras) at their most recent follow-up visit. The percentage of patients who were ILAE class 1 at most recent follow-up decreased with increasing duration of epilepsy prior to surgery. Developmental outcome after surgery was improved in 14 of 19 (74%) and stable in 5 of 19 (26%) patients. CONCLUSIONS Our study found excellent seizure freedom rates and improved developmental outcomes following epilepsy surgery in patients with a history of infantile epileptic spasms syndrome with a structural lesion detected on MRI brain. Patients who undergo surgery earlier have improved seizure freedom rates and improved developmental outcomes.
Collapse
Affiliation(s)
- Jennifer V Gettings
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - Shatha Shafi
- Division of Neurology, Department of Pediatrics, 37853Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Jennifer Boyd
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - O Carter Snead
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - James Rutka
- Division of Neurosurgery, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - James Drake
- Division of Neurosurgery, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - Bláthnaid McCoy
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - Puneet Jain
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - Robyn Whitney
- Division of Neurology, Department of Paediatrics, McMaster Children's Hospital (McMaster University), Hamilton, ON, Canada
| | - Cristina Go
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada.,Division of Neurology, Department of Pediatrics, British Columbia Children's Hospital (University of British Columbia), Toronto, Ontario, Canada
| |
Collapse
|
68
|
Lynch M, Smith K, Riney K. Clinical seizure semiology is subtle and identification of seizures by parents is unreliable in infants with tuberous sclerosis complex. Epilepsia 2023; 64:386-395. [PMID: 36318046 PMCID: PMC10107460 DOI: 10.1111/epi.17454] [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: 08/17/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 12/07/2022]
Abstract
OBJECTIVE The objectives of this study were to assess the accuracy of parental seizure detection in infants with antenatally diagnosed tuberous sclerosis complex (TSC), and to document the total seizure burden (clinical and subclinical) in those patients who met criteria for prolonged electroencephalography (EEG) recording. METHODS Consecutive infants at a single institution with antenatally diagnosed TSC who met criteria for prolonged video-EEG (vEEG) were recruited to this study. The vEEG data were reviewed and when a seizure was identified on EEG, the video and audio recording was assessed for evidence of clinical seizure and, if present, whether there was evidence of parent seizure identification. RESULTS Nine infants were enrolled, for whom 674 focal seizures were identified in eight of nine patients across 24 prolonged vEEG recordings, with vEEG total duration of 634 h 49 min (average seizure frequency of 1 focal seizure/h). Only 220 of 674 (32.6%) were clinical seizures, 395 of 674 (58.6%) were subclinical seizures, and 59 of 674 seizures could not be classified. Only 63 of 220 clinical seizures (28.6%) were identified by parents, with 157 of 220 (71.4%) not identified. Thirty clusters of epileptic spasms were detected in one patient. At least one clinical epileptic spasm occurred in 2 of 30 clusters (6.7%), 24 of 30 clusters of epileptic spasms (80%) were electrographic only, and classification was uncertain for 4 of 30 clusters (13.3%). No clinical epileptic spasms were detected by parents. Clinical seizure frequency was significantly underestimated by parents for all patients. SIGNIFICANCE This study demonstrates that in infants with TSC who met criteria for prolonged vEEG, (1) parents significantly under recognize total clinical seizure count, (2) parents fail to identify epileptic spasms, and (3) seizure frequency is high. This highlights that epilepsy treatment decisions should not be based solely on parental clinical seizure identification. Prolonged vEEG monitoring may have an important role in the routine epilepsy care of infants with TSC, as demonstrating undetected high clinical seizure frequency may allow improved epilepsy management decisions.
Collapse
Affiliation(s)
- Matthew Lynch
- Neurosciences Unit, Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - Kirsty Smith
- Neurosciences Unit, Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - Kate Riney
- Neurosciences Unit, Queensland Children's Hospital, South Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
69
|
Sciaccaluga M, Ruffolo G, Palma E, Costa C. Traditional and Innovative Anti-seizure Medications Targeting Key Physiopathological Mechanisms: Focus on Neurodevelopment and Neurodegeneration. Curr Neuropharmacol 2023; 21:1736-1754. [PMID: 37143270 PMCID: PMC10514539 DOI: 10.2174/1570159x21666230504160948] [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: 09/03/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023] Open
Abstract
Despite the wide range of compounds currently available to treat epilepsy, there is still no drug that directly tackles the physiopathological mechanisms underlying its development. Indeed, antiseizure medications attempt to prevent seizures but are inefficacious in counteracting or rescuing the physiopathological phenomena that underlie their onset and recurrence, and hence do not cure epilepsy. Classically, the altered excitation/inhibition balance is postulated as the mechanism underlying epileptogenesis and seizure generation. This oversimplification, however, does not account for deficits in homeostatic plasticity resulting from either insufficient or excessive compensatory mechanisms in response to a change in network activity. In this respect, both neurodevelopmental epilepsies and those associated with neurodegeneration may share common underlying mechanisms that still need to be fully elucidated. The understanding of these molecular mechanisms shed light on the identification of new classes of drugs able not only to suppress seizures, but also to present potential antiepileptogenic effects or "disease-modifying" properties.
Collapse
Affiliation(s)
- Miriam Sciaccaluga
- Section of Neurology, S.M. della Misericordia Hospital, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06129, Italy
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome, Sapienza, Rome, 00185, Italy
- IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Eleonora Palma
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome, Sapienza, Rome, 00185, Italy
- IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Cinzia Costa
- Section of Neurology, S.M. della Misericordia Hospital, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06129, Italy
| |
Collapse
|
70
|
Singh A, Hadjinicolaou A, Peters JM, Salussolia CL. Treatment-Resistant Epilepsy and Tuberous Sclerosis Complex: Treatment, Maintenance, and Future Directions. Neuropsychiatr Dis Treat 2023; 19:733-748. [PMID: 37041855 PMCID: PMC10083014 DOI: 10.2147/ndt.s347327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/22/2023] [Indexed: 04/13/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a neurogenetic disorder that affects multiple organ systems, including the heart, kidneys, eyes, skin, and central nervous system. The neurologic manifestations have the highest morbidity and mortality, in particular in children. Clinically, patients with TSC often present with new-onset seizures within the first year of life. TSC-associated epilepsy is often difficult to treat and refractory to multiple antiseizure medications. Refractory TSC-associated epilepsy is associated with increased risk of neurodevelopmental comorbidities, including developmental delay, intellectual disability, autism spectrum disorder, and attention hyperactivity disorder. An increasing body of research suggests that early, effective treatment of TSC-associated epilepsy during critical neurodevelopmental periods can potentially improve cognitive outcomes. Therefore, it is important to treat TSC-associated epilepsy aggressively, whether it be with pharmacological therapy, surgical intervention, and/or neuromodulation. This review discusses current and future pharmacological treatments for TSC-associated epilepsy, as well as the importance of early surgical evaluation for refractory epilepsy in children with TSC and consideration of neuromodulatory interventions in young adults.
Collapse
Affiliation(s)
- Avantika Singh
- Division of Epilepsy and Neurophysiology, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Aristides Hadjinicolaou
- Division of Epilepsy and Neurophysiology, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jurriaan M Peters
- Division of Epilepsy and Neurophysiology, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Catherine L Salussolia
- Division of Epilepsy and Neurophysiology, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- Correspondence: Catherine L Salussolia, 3 Blackfan Circle, Center for Life Sciences 14060, Boston, MA, 02115, USA, Tel +617-355-7970, Email
| |
Collapse
|
71
|
Garavatti E, Yamamoto E, Collins K, Selden N, Bushlin I. Surgical Resection of Focal Cortical Dysplasia in a Neonate with Novel TSC1 Mutation Leading to Resolution of Refractory Seizures: Case Report. Child Neurol Open 2023; 10:2329048X231219223. [PMID: 38107744 PMCID: PMC10722926 DOI: 10.1177/2329048x231219223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/20/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
We describe a neonate presenting on first day of life with refractory seizures secondary to a single, large area of focal cortical dysplasia (FCD) who underwent surgical resection at age 3 weeks leading to resolution of seizure activity and dramatic improvement in developmental trajectory. Surgical intervention for epilepsy is infrequently offered for neonates, often reserved only for those with catastrophic presentations. This case demonstrates that surgical intervention can be safe and efficacious in neonates for pharmaco-resistant seizures associated with a focal lesion. Rapid whole exome sequencing in this case yielded a germline novel de novo TSC1 mutation, leading to a genetic diagnosis of tuberous sclerosis complex (TSC). Our patient demonstrates an atypical neonatal presentation of TSC. Limited data is available for those with isolated FCD in TSC; this is the first reported case in a neonate.
Collapse
Affiliation(s)
- Emily Garavatti
- Neurodevelopmental Disabilities, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Erin Yamamoto
- Neurosurgery, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Kelly Collins
- Department of Neurosurgery, Oregon Health & Science University, Portland, OR, USA
| | - Nathan Selden
- Department of Neurosurgery, Oregon Health & Science University, Portland, OR, USA
| | - Ittai Bushlin
- Department of Pediatric Neurology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
72
|
Wan L, He W, Wang YY, Xu Y, Lu Q, Zhang MN, Wang QH, Dun S, Liu LY, Shi XY, Wang J, Hu LY, Zhang B, Yang G, Zou LP. Vigabatrin-associated brain abnormalities on MRI in tuberous sclerosis complex patients with infantile spasms: are they preventable? Ther Adv Neurol Disord 2022; 15:17562864221138148. [PMID: 36601084 PMCID: PMC9806385 DOI: 10.1177/17562864221138148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 10/22/2022] [Indexed: 12/28/2022] Open
Abstract
Background Vigabatrin (VGB) is currently the most widely prescribed first-line medication for individuals with infantile spasms (IS) and especially for those with tuberous sclerosis complex (TSC), with demonstrated efficacy. Meanwhile, its adverse events, such as vigabatrin-associated brain abnormalities on magnetic resonance imaging (MRI; VABAM), have also been widely reported. Objectives The objectives of this study were to observe the occurrences of VABAM in patients with IS caused by TSC (IST) and further explore the associated risk factors. Methods Children with IS receiving VGB were recruited from our institution; clinical, imaging, and medication data were collected. Cerebral MRI was reviewed to determine the occurrence of VABAM. Group comparisons (IS caused by TSC and other etiologies) were performed; subgroup analyses on IST were also performed. Next, a retrospective cohort study of children taking VGB was conducted to explore risk/protective factors associated with VABAM. Results The study enrolled 172 children with IS who received VGB. VABAM was observed in 38 patients (22.1%) with a peak dosage of 103.5 ± 26.7 mg/kg/day. Subsequent analysis found the incidence of VABAM was significantly lower in the 80 patients with IST than in the 92 patients with IS caused by other etiologies (10% versus 32.6%, p-value < 0.001). In subgroup analyses within the IST cohort, VABAM was significantly lower in children who received concomitant rapamycin therapy. Univariate and multivariate logistic regression analysis of the 172 IS children showed that treatment with rapamycin was the independent factor associated with a lower risk of VABAM; similar results were observed in the survival analysis. Conclusion The incidence of VABAM was significantly lower in IST patients. Further research is needed to examine the mechanisms that underlie this phenomenon and to determine if treatment with rapamycin may reduce the risk of VABAM.
Collapse
Affiliation(s)
| | | | | | - Yong Xu
- Department of Pediatrics, PLA General Hospital,
Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Qian Lu
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Meng-Na Zhang
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Qiu-Hong Wang
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Shuo Dun
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Li-Ying Liu
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Xiu-Yu Shi
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China,The Second School of Clinical Medicine,
Southern Medical University, Guangzhou, China
| | - Jing Wang
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Lin-Yan Hu
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China
| | - Bo Zhang
- Department of Neurology and ICCTR
Biostatistics and Research Design Center, Boston Children’s Hospital,
Harvard Medical School, Boston, MA, USA
| | - Guang Yang
- Department of Pediatrics, PLA General
Hospital, Beijing, China,Division of Pediatrics, The First Medical
Center of PLA General Hospital, Beijing, China,Medical School of Chinese People’s Liberation
Army, Beijing, China,The Second School of Clinical Medicine,
Southern Medical University, Guangzhou, China
| | | |
Collapse
|
73
|
Venkatesan C, Countee E, Wong B, Spaeth C, Kline-Fath BM, Nagaraj UD. Imaging Similarities Between Oral-Facial-Digital Syndrome Type 1 and Aicardi Syndrome: Prenatal and Postnatal Magnetic Resonance Imaging (MRI) Findings in 4 Patients. J Child Neurol 2022; 38:31-37. [PMID: 36567511 DOI: 10.1177/08830738221147372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Prenatal identification by magnetic resonance imaging (MRI) of callosal anomalies, particularly with accompanying intracranial abnormalities, poses a challenge for accurate prognostication and fetal counseling as outcome can vary widely depending on underlying etiology. In female patients, Aicardi syndrome is an important consideration, and prompt postnatal ophthalmologic assessment to identify ocular stigmata of Aicardi syndrome can aid with anticipatory guidance and greater vigilance for seizures. We present a case of a female with fetal and postnatal MRI findings of agenesis of corpus callosum and type 2b interhemispheric cysts, characteristically found in Aicardi syndrome, but was found to have oral-facial-digital syndrome type 1 (OFD1). We also present 3 other companion cases with pre- and postnatal imaging of patients with Aicardi syndrome. These cases highlight the importance of widening the differential diagnosis to also include OFD1 for female patients with callosal anomalies.
Collapse
Affiliation(s)
- Charu Venkatesan
- Division of Neurology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Elizabeth Countee
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Beatrix Wong
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Christine Spaeth
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Beth M Kline-Fath
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Radiology and Medical Imaging, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Usha D Nagaraj
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Radiology and Medical Imaging, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| |
Collapse
|
74
|
Pinto ALR. Neurological manifestations of tuberous sclerosis complex: the importance of early diagnosis. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:983-984. [PMID: 36535281 PMCID: PMC9770056 DOI: 10.1055/s-0042-1759689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Anna L. R. Pinto
- Boston Children's Hospital, Department of Neurology, Division of Epilepsy and Clinical Neurophysiology, Boston, United States.,Harvard Medical School, Boston, United States.,Address for correspondence Anna Lecticia Pinto
| |
Collapse
|
75
|
Pereira CCDS, Dantas FDG, Manreza MLGD. Clinical profile of tuberous sclerosis complex patients with and without epilepsy: a need for awareness for early diagnosis. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:1004-1010. [PMID: 36535284 PMCID: PMC9770081 DOI: 10.1055/s-0042-1758456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is a multisystemic disorder. Its clinical features manifest differently in several organs, prompting the need for better knowledge. OBJECTIVE The goal of the present study is to evaluate the neurological findings of TSC, such as cerebral lesions and epilepsy, and to raise awareness of non-neurological findings that could contribute to an earlier diagnosis and treatment. METHODS This was a natural history study of patients with a definitive diagnosis of TSC who were referred to a specialized outpatient clinic and followed-up for 2 years with clinical and radiological exams. RESULTS A total of 130 TSC patients (59 males [45.4%], mean age 20.4 years old [1 to 56 years old]); 107 patients (82.3%) were diagnosed with epilepsy. Seizures predominantly began at < 1 year old (72.8%); focal seizures predominated (86.9%); epileptic spasms occurred in 34.5% of patients, and refractory epilepsy was present in 55.1%. Neuropsychiatric disorders, cortical tubers and cerebellar tubers were significantly more frequent in the epilepsy group. Moreover, rhabdomyomas were significantly more frequent in the epilepsy group (p = 0.044), while lymphangioleiomyomatosis was significantly less frequent in the epilepsy group (p = 0.009). Other non-neurological findings did not differ significantly between the groups with and without epilepsy. CONCLUSIONS The present study of TSC patients demonstrated the predominantly neurological involvement and significantly higher proportion of TSC-associated neuropsychiatric disorders in the epilepsy group. Higher proportions of cortical and cerebellar tubers may be a risk factor for epilepsy and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Conceição Campanario da Silva Pereira
- Universidade de São Paulo, Hospital das Clinicas de São Paulo, Departamento de Neurologia Infantil, São Paulo SP, Brazil.,Address for correspondence Conceição Campanario da Silva Pereira
| | - Felipe Diego Gomes Dantas
- Universidade de São Paulo, Hospital das Clínicas de São Paulo, Departamento de Neurorradiologia, São Paulo SP, Brazil.
| | | |
Collapse
|
76
|
GENE TARGET: A framework for evaluating Mendelian neurodevelopmental disorders for gene therapy. Mol Ther Methods Clin Dev 2022; 27:32-46. [PMID: 36156879 PMCID: PMC9478871 DOI: 10.1016/j.omtm.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interest in gene-based therapies for neurodevelopmental disorders is increasing exponentially, driven by the rise in recognition of underlying genetic etiology, progress in genomic technology, and recent proof of concept in several disorders. The current prioritization of one genetic disorder over another for development of therapies is driven by competing interests of pharmaceutical companies, advocacy groups, and academic scientists. Although these are all valid perspectives, a consolidated framework will facilitate more efficient and rational gene therapy development. Here we outline features of Mendelian neurodevelopmental disorders that warrant consideration when determining suitability for gene therapy. These features fit into four broad domains: genetics, preclinical validation, clinical considerations, and ethics. We propose a simple mnemonic, GENE TARGET, to remember these features and illustrate how they could be scored using a preliminary scoring rubric. In this suggested rubric, for a given disorder, scores for each feature may be added up to a composite GENE TARGET suitability (GTS) score. In addition to proposing a systematic method to evaluate and compare disorders, our framework helps identify gaps in the translational pipeline for a given disorder, which can inform prioritization of future research efforts.
Collapse
|
77
|
Le JT, Ballester-Rosado CJ, Frost JD, Swann JW. Neurobehavioral deficits and a progressive ictogenesis in the tetrodotoxin model of epileptic spasms. Epilepsia 2022; 63:3078-3089. [PMID: 36179064 PMCID: PMC9742150 DOI: 10.1111/epi.17428] [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: 06/29/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Our goal was to determine whether animals with a history of epileptic spasms have learning and memory deficits. We also used continuous (24/7) long-term electroencephalographic (EEG) recordings to evaluate the evolution of epileptiform activity in the same animals over time. METHODS Object recognition memory and object location memory tests were undertaken, as well as a matching to place water maze test that evaluated working memory. A retrospective analysis was undertaken of long-term video/EEG recordings from rats with epileptic spasms. The frequency and duration of the ictal events of spasms were quantified. RESULTS Rats with a history of epileptic spasms showed impairment on the three behavioral tests, and their scores on the object recognition memory and matching to place water maze tests indicated neocortical involvement in the observed impaired cognition. Analysis of EEG recordings unexpectedly showed that the ictal events of spasms and their accompanying behaviors progressively increased in duration over a 2-week period soon after onset, after which spasm duration plateaued. At the same time, spasm frequency remained unchanged. Soon after spasm onset, ictal events were variable in wave form but became more stereotyped as the syndrome evolved. SIGNIFICANCE Our EEG findings are the first to demonstrate progressive ictogenesis for epileptic spasms. Furthermore, in demonstrating cognitive deficits in the tetrodotoxin model, we have met a criterion for an animal model of West syndrome. Animal models will allow in-depth studies of spasm progression's potential role in cognitive regression and may elucidate why early treatment is considered essential for improved neurodevelopmental outcomes in children.
Collapse
Affiliation(s)
- John T. Le
- The Cain Foundation Laboratories, the Jan and Dan Neurological Research Institute, Texas Children’s Hospital, Houston Texas
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
| | - Carlos J. Ballester-Rosado
- The Cain Foundation Laboratories, the Jan and Dan Neurological Research Institute, Texas Children’s Hospital, Houston Texas
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
| | - James D. Frost
- Department of Neurology, Baylor College of Medicine, Houston Texas
| | - John W. Swann
- The Cain Foundation Laboratories, the Jan and Dan Neurological Research Institute, Texas Children’s Hospital, Houston Texas
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
- Department of Neuroscience, Baylor College of Medicine, Houston Texas
| |
Collapse
|
78
|
Van Bogaert P, Nabbout R. Updating Tuberous sclerosis complex care for pediatric neurologists. Arch Pediatr 2022; 29:5S1-5S2. [PMID: 36585065 DOI: 10.1016/s0929-693x(22)00282-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- P Van Bogaert
- Service de neuropédiatrie et neurochirurgie de l'enfant, CHU d'Angers, France; Laboratoire Angevin de Recherche en Ingénierie des Systèmes (LARIS), Université d'Angers, France
| | - R Nabbout
- Service de neuropédiatrie, Hôpital Necker-Enfants Malades, APHP Paris, Université Paris cité, Paris, France; INSERM U1163, Chaire GEEN-DS, institute Imagine, Paris, France
| |
Collapse
|
79
|
Abstract
Numerous studies showed that epilepsy represents a high burden in Tuberous Sclerosis Complex (TSC), affecting 63 to 78% of the patients. Epilepsy will be refractory to medication in over 60% of cases in early presentations, and accompanied by intellectual disabilities and/or autism spectrum disorders. The emerging experimental and clinical data suggest that the molecular and cellular changes triggered by seizures, particularly during the first weeks of life, can be limited by early action. Making any effort to avoid or delay epilepsy onset is a promising pathway to improve global outcome for TSC patients, although it is not possible to tidy up the specific roles of seizures, interictal abnormalities, and cortical abnormalities upon neurodevelopment. Early diagnosis of epilepsy can be made during a "symptomatic phase," shortly after the onset of seizures (focal seizures or spasms), revealing the TSC in a young infant. As soon as the diagnosis is made, a treatment with Vigabatrin is now recommended. The diagnosis of epilepsy can also be performed during a "presymptomatic phase", with the improvement of fetal and neonatal diagnosis of TSC. Recent studies demonstrated a significant delay of more than 3 months between the detection of EEG abnormalities and the first clinical seizures, which allows to consider a preventive treatment. Beside vigabatrin, mTOR inhibitors may have a place in this early management. The last recommendations about early detection and treatment of epilepsy in TSC will be detailed in this review. © 2022 French Society of Pediatrics. Published by Elsevier Masson SAS. All rights reserved.
Collapse
|
80
|
Kuchenbuch M, Chiron C, Milh M. Overview of therapeutic options for epilepsy. Arch Pediatr 2022; 29:5S14-5S19. [PMID: 36585066 DOI: 10.1016/s0929-693x(22)00285-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tuberous sclerosis (TSC) epilepsy includes infantile spasms and focal seizures before the age of 2 years, whereas focal seizures are predominant over 2 years and generalized seizures may occasionally be part of Lennox-Gastaut syndrome. The better and earlier the seizure control, the better the child's subsequent cognitive and behavioral prognosis. As for epilepsy of other causes, therapeutic options depend on the type of seizure/epilepsy, age and drug resistance, but there are significant specificities for TSC. (1) As first-line treatment, vigabatrin is unanimously recommended for infantile spasms and focal seizures before 2 years and is also widely used for seizures over 2 years, as are levetiracetam and carbamazepine. (2) If seizures persist (about 40% of children and adolescents), cannabidiol and everolimus, an inhibitor of the mTOR pathway, have recently been approved as adjunctive therapy to the arsenal of antiseizure medications authorized for this age group and to the ketogenic diet. (3) Surgery is an essential treatment option in cases of drug resistance and should be discussed as soon as two treatments have failed. Presurgical investigations and operating techniques have recently progressed spectacularly, for example laser thermocoagulation with stereotactic location. A particularity of TSC is the possibility of sequential interventions on several epileptogenic tubers. (4) Finally, the innovative principle of initiating "pre-seizure" treatment with vigabatrin from the first months of life has just proven effective on the subsequent development of epilepsy in TSC. © 2022 French Society of Pediatrics. Published by Elsevier Masson SAS. All rights reserved.
Collapse
Affiliation(s)
- M Kuchenbuch
- Département de Pédiatrie et Génétique, CHU de Nancy, France.
| | - C Chiron
- Inserm U1141 et APHP, Service de Neurologie pédiatrique, Hôpital Necker-Enfants Malades, Paris, France; Service de Neurologie Pédiatrique, Necker Enfants Malades, APHP, centre de référence épilepsies rares, Paris, France
| | - M Milh
- Service de Neurologie pédiatrique, Hôpital de la Timone, CHU de Marseille, France
| |
Collapse
|
81
|
Yuskaitis CJ, Mytinger JR, Baumer FM, Zhang B, Liu S, Samanta D, Hussain SA, Yozawitz EG, Keator CG, Joshi C, Singh RK, Bhatia S, Bhalla S, Shellhaas R, Harini C. Association of Time to Clinical Remission With Sustained Resolution in Children With New-Onset Infantile Spasms. Neurology 2022; 99:e2494-e2503. [PMID: 36038267 PMCID: PMC9728034 DOI: 10.1212/wnl.0000000000201232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Standard therapies (adrenocorticotropic hormone [ACTH], oral steroids, or vigabatrin) fail to control infantile spasms in almost half of children. Early identification of nonresponders could enable rapid initiation of sequential therapy. We aimed to determine the time to clinical remission after appropriate infantile spasms treatment initiation and identify predictors of the time to infantile spasms treatment response. METHODS The National Infantile Spasms Consortium prospectively followed children aged 2-24 months with new-onset infantile spasms at 23 US centers (2012-2018). We included children treated with standard therapy (ACTH, oral steroids, or vigabatrin). Sustained treatment response was defined as having the last clinically recognized infantile spasms on or before treatment day 14, absence of hypsarrhythmia on EEG 2-4 weeks after treatment, and persistence of remission to day 30. We analyzed the time to treatment response and assessed clinical characteristics to predict sustained treatment response. RESULTS Among 395 infants, clinical infantile spasms remission occurred in 43% (n = 171) within the first 2 weeks of treatment, of which 81% (138/171) responded within the first week of treatment. There was no difference in the median time to response across standard therapies (ACTH: median 4 days, interquartile range [IQR] 3-7; oral steroids: median 3 days, IQR 2-5; vigabatrin: median 3 days, IQR 1-6). Individuals without hypsarrhythmia on the pretreatment EEG (i.e., abnormal but not hypsarrhythmia) were more likely to have early treatment response than infants with hypsarrhythmia at infantile spasms onset (hazard ratio 2.23, 95% CI 1.39-3.57). No other clinical factors predicted early responders to therapy. DISCUSSION Remission after first infantile spasms treatment can be identified by treatment day 7 in most children. Given the importance of early and effective treatment, these data suggest that children who do not respond to standard infantile spasms therapy within 1 week should be reassessed immediately for additional standard treatment. This approach could optimize outcomes by facilitating early sequential therapy for children with infantile spasms.
Collapse
Affiliation(s)
- Christopher J Yuskaitis
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI.
| | - John R Mytinger
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Fiona M Baumer
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Bo Zhang
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Shanshan Liu
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Debopam Samanta
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Shaun A Hussain
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Elissa G Yozawitz
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Cynthia G Keator
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Charuta Joshi
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Rani K Singh
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Sonal Bhatia
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Sonam Bhalla
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Renée Shellhaas
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Chellamani Harini
- From the Division of Epilepsy and Clinical Neurophysiology (C.J.Y., C.H.), Department of Neurology, Boston Children's Hospital, MA; Department of Pediatrics (J.R.M.), Division of Pediatric Neurology, Nationwide Children's Hospital, The Ohio State University, Columbus; Division of Child Neurology (F.M.B.), Department of Neurology, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology and ICCTR Biostatistics and Research Design Center (B.Z., S.L.), Boston Children's Hospital and Harvard Medical School, MA; Division of Child Neurology (D.S.), Department of Pediatrics, University of Arkansas for Medical Sciences, AR; Department of Pediatrics (S.A.H.), Division of Neurology, University of California, Los Angeles; Department of Neurology (E.G.Y.), Montefiore Medical Center, Bronx, NY; Jane and John Justin Neurosciences (C.G.K.), Cook Children's Hospital, Fort Worth, TX; Departments of Pediatrics and Neurology (C.J.), University of Colorado School of Medicine and Children's Hospital Colorado, Aurora; Department of Pediatrics (R.K.S.), Division of Neurology, Atrium Health/Levine Children's, Charlotte, NC; Division of Pediatric Neurology (S. Bhatia), Department of Pediatrics, Medical University of South Carolina, Charleston; Department of Pediatrics (S. Bhalla), Division of Child Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, GA; and Department of Pediatrics (R.S.), Michigan Medicine, University of Michigan, Ann Arbor, MI
| | | |
Collapse
|
82
|
Whitney R, Zak M, Haile D, Nabavi Nouri M. The state of pediatric tuberous sclerosis complex epilepsy care: Results from a national survey. Epilepsia Open 2022; 7:718-728. [PMID: 36161285 PMCID: PMC9712483 DOI: 10.1002/epi4.12652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/21/2022] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Epilepsy associated with tuberous sclerosis complex (TSC) can be challenging to treat and is associated with significant disease burden. Our objective was to better understand the state of epilepsy care of TSC amongst pediatric neurologists in Canada, identify gaps in care and determine whether access to a dedicated TSC clinic has an impact on epilepsy management. METHODS A survey was developed after a literature review and discussion amongst two pediatric epileptologists and one nurse practitioner with expertise in TSC about the state of epilepsy care of TSC patients in Canada. Canadian pediatric neurologists were asked to participate in sharing their experiences via an anonymous web-based survey through the Canadian League Against Epilepsy (CLAE) and the Canadian Neurological Sciences Federation (CNSF). RESULTS Fifty-seven responses were received. Access to a dedicated TSC clinic was reported by 25% (n = 14). Sixty percent (n = 34) reported performing serial EEG monitoring in infants with TSC and 57% (n = 33) started prophylactic antiseizure therapy when EEG abnormalities were detected, regardless of whether there was access to a TSC clinic (P = .06 and P = .29, respectively). While 52% (n = 29) did not feel comfortable prescribing mTORi for epilepsy, 65% (n = 36) indicated they would consider it with additional training. Epilepsy surgery was offered in 93% (n = 13) of centers with a dedicated TSC clinic but only 45% of centers without a TSC clinic (n = 19) (P = .002). SIGNIFICANCE Our findings demonstrate the variability in neurological care of pediatric patients with TSC as it pertains to epilepsy management. There is a need for the establishment of epilepsy practice guidelines and a national network to support clinical practice, research, and education.
Collapse
Affiliation(s)
- Robyn Whitney
- Division of Neurology, Department of PaediatricsMcMaster UniversityHamiltonOntarioCanada
| | - Maria Zak
- Division of Neurology, Department of PaediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Denait Haile
- Department of Paediatrics, Schulich School of Dentistry and MedicineWestern UniversityLondonOntarioCanada
| | - Maryam Nabavi Nouri
- Department of Paediatrics, Schulich School of Dentistry and MedicineWestern UniversityLondonOntarioCanada
- Children's Health Research InstituteLawson Health Research InstituteLondonOntarioCanada
| |
Collapse
|
83
|
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: 44] [Impact Index Per Article: 22.0] [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
|
84
|
Patil P, Pencheva BB, Patil VM, Fangusaro J. Nervous system (NS) Tumors in Cancer Predisposition Syndromes. Neurotherapeutics 2022; 19:1752-1771. [PMID: 36056180 PMCID: PMC9723057 DOI: 10.1007/s13311-022-01277-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
Genetic syndromes which develop one or more nervous system (NS) tumors as one of the manifestations can be grouped under the umbrella term of NS tumor predisposition syndromes. Understanding the underlying pathological pathways at the molecular level has led us to many radical discoveries, in understanding the mechanisms of tumorigenesis, tumor progression, interactions with the tumor microenvironment, and development of targeted therapies. Currently, at least 7-10% of all pediatric cancers are now recognized to occur in the setting of genetic predisposition to cancer or cancer predisposition syndromes. Specifically, the cancer predisposition rate in pediatric patients with NS tumors has been reported to be as high as 15%, though it can approach 50% in certain tumor types (i.e., choroid plexus carcinoma associated with Li Fraumeni Syndrome). Cancer predisposition syndromes are caused by pathogenic variation in genes that primarily function as tumor suppressors and proto-oncogenes. These variants are found in the germline or constitutional DNA. Mosaicism, however, can affect only certain tissues, resulting in varied manifestations. Increased understanding of the genetic underpinnings of cancer predisposition syndromes and the ability of clinical laboratories to offer molecular genetic testing allows for improvement in the identification of these patients. The identification of a cancer predisposition syndrome in a CNS tumor patient allows for changes to medical management to be made, including the initiation of cancer surveillance protocols. Finally, the identification of at-risk biologic relatives becomes feasible through cascade (genetic) testing. These fundamental discoveries have also broadened the horizon of novel therapeutic possibilities and have helped to be better predictors of prognosis and survival. The treatment paradigm of specific NS tumors may also vary based on the patient's cancer predisposition syndrome and may be used to guide therapy (i.e., immune checkpoint inhibitors in constitutional mismatch repair deficiency [CMMRD] predisposition syndrome) [8]. Early diagnosis of these cancer predisposition syndromes is therefore critical, in both unaffected and affected patients. Genetic counselors are uniquely trained master's level healthcare providers with a focus on the identification of hereditary disorders, including hereditary cancer, or cancer predisposition syndromes. Genetic counseling, defined as "the process of helping people understand and adapt to the medical, psychological and familial implications of genetic contributions to disease" plays a vital role in the adaptation to a genetic diagnosis and the overall management of these diseases. Cancer predisposition syndromes that increase risks for NS tumor development in childhood include classic neurocutaneous disorders like neurofibromatosis type 1 and type 2 (NF1, NF2) and tuberous sclerosis complex (TSC) type 1 and 2 (TSC1, TSC2). Li Fraumeni Syndrome, Constitutional Mismatch Repair Deficiency, Gorlin syndrome (Nevoid Basal Cell Carcinoma), Rhabdoid Tumor Predisposition syndrome, and Von Hippel-Lindau disease. Ataxia Telangiectasia will also be discussed given the profound neurological manifestations of this syndrome. In addition, there are other cancer predisposition syndromes like Cowden/PTEN Hamartoma Tumor Syndrome, DICER1 syndrome, among many others which also increase the risk of NS neoplasia and are briefly described. Herein, we discuss the NS tumor spectrum seen in the abovementioned cancer predisposition syndromes as with their respective germline genetic abnormalities and recommended surveillance guidelines when applicable. We conclude with a discussion of the importance and rationale for genetic counseling in these patients and their families.
Collapse
Affiliation(s)
- Prabhumallikarjun Patil
- Children's Healthcare of Atlanta, Aflac Cancer Center, Atlanta, GA, USA.
- Emory University School of Medicine, Atlanta, GA, USA.
| | - Bojana Borislavova Pencheva
- Children's Healthcare of Atlanta, Aflac Cancer Center, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - Vinayak Mahesh Patil
- Intensive Care Unit Medical Officer, District Hospital Vijayapura, Karnataka, India
| | - Jason Fangusaro
- Children's Healthcare of Atlanta, Aflac Cancer Center, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
85
|
Syrbe S. Developmental and epileptic encephalopathies - therapeutic consequences of genetic testing. MED GENET-BERLIN 2022; 34:215-224. [PMID: 38835873 PMCID: PMC11006352 DOI: 10.1515/medgen-2022-2145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Developmental and epileptic encephalopathies comprise a heterogeneous group of monogenic neurodevelopmental disorders characterized by early-onset seizures, marked epileptic activity and abnormal neurocognitive development. The identification of an increasing number of underlying genetic alterations and their pathophysiological roles in cellular signaling drives the way toward novel precision therapies. The implementation of novel treatments that target the underlying mechanisms gives hope for disease modification that will improve not only the seizure burden but also the neurodevelopmental outcome of affected children. So far, beneficial effects are mostly reported in individual trials and small numbers of patients. There is a need for international collaborative studies to define the natural history and relevant outcome measures and to test novel pharmacological approaches.
Collapse
Affiliation(s)
- Steffen Syrbe
- Division of Paediatric Epileptology, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| |
Collapse
|
86
|
Franz DN. Changing the outcome in genetic brain disorders. Dev Med Child Neurol 2022; 64:1184-1185. [PMID: 35866540 PMCID: PMC9543475 DOI: 10.1111/dmcn.15350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022]
Abstract
This commentary is on the original articles by Wang et al. and Zhang et al. on pages 1230–1236 and 1237–1245 of this issue.
Collapse
Affiliation(s)
- David Neal Franz
- Cincinnati Children's HospitalUniversity of Cincinnati College of Medicine – NeurologyCincinnatiOHUSA
| |
Collapse
|
87
|
Ramantani G, Bölsterli BK, Alber M, Klepper J, Korinthenberg R, Kurlemann G, Tibussek D, Wolff M, Schmitt B. Treatment of Infantile Spasm Syndrome: Update from the Interdisciplinary Guideline Committee Coordinated by the German-Speaking Society of Neuropediatrics. Neuropediatrics 2022; 53:389-401. [PMID: 35882373 PMCID: PMC9643068 DOI: 10.1055/a-1909-2977] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
OBJECTIVES The manuscript serves as an update on the current management practices for infantile spasm syndrome (ISS). It includes a detailed summary of the level of current evidence of different treatment options for ISS and gives recommendations for the treatment and care of patients with ISS. METHODS A literature search was performed using the Cochrane and Medline Databases (2014 to July 2020). All studies were objectively rated using the Scottish Intercollegiate Guidelines Network. For recommendations, the evidence from these studies was combined with the evidence from studies used in the 2014 guideline. RECOMMENDATIONS If ISS is suspected, electroencephalography (EEG) should be performed within a few days and, if confirmed, treatment should be initiated immediately. Response to first-line treatment should be evaluated clinically and electroencephalographically after 14 days. The preferred first-line treatment for ISS consists of either hormone-based monotherapy (AdrenoCorticoTropic Hormone [ACTH] or prednisolone) or a combination of hormone and vigabatrin. Children with tuberous sclerosis complex and those with contraindications against hormone treatment should be treated with vigabatrin. If first-line drugs are ineffective, second-line treatment options such as ketogenic dietary therapies, sulthiame, topiramate, valproate, zonisamide, or benzodiazepines should be considered. Children refractory to drug therapy should be evaluated early for epilepsy surgery, especially if focal brain lesions are present. Parents should be informed about the disease, the efficacy and adverse effects of the medication, and support options for the family. Regular follow-up controls are recommended.
Collapse
Affiliation(s)
- Georgia Ramantani
- Division of Clinical Neurophysiology and Epilepsy, University Children's Hospital, Zurich, Switzerland,Address for correspondence Georgia Ramantani, MD, PhD Department of Neuropediatrics, Steinwiesstrasse 758032 ZurichSwitzerland
| | - Bigna K. Bölsterli
- Division of Clinical Neurophysiology and Epilepsy, University Children's Hospital, Zurich, Switzerland
| | - Michael Alber
- Department of Pediatric Neurology and Developmental Medicine, University Children's Hospital, Tubingen, Germany
| | - Joerg Klepper
- Department of Pediatrics, Klinikum Aschaffenburg-Alzenau, Aschaffenburg, Germany
| | - Rudolf Korinthenberg
- Department of Neuropediatrics and Muscular Diseases, Centre of Pediatrics and Adolescent Medicine, University Medical Centre, Freiburg, Germany
| | - Gerhard Kurlemann
- St. Bonifatius Hospital Lingen, Children's Hospital, Lingen, Germany
| | - Daniel Tibussek
- Center for Pediatric and Teenage Health Care, Child Neurology, Sankt Augustin, Germany
| | - Markus Wolff
- Department of Pediatric Neurology, Vivantes Hospital Neukölln, Berlin, Germany
| | - Bernhard Schmitt
- Division of Clinical Neurophysiology and Epilepsy, University Children's Hospital, Zurich, Switzerland
| |
Collapse
|
88
|
The longitudinal evolution of cerebral blood flow in children with tuberous sclerosis assessed by arterial spin labeling magnetic resonance imaging may be related to cognitive performance. Eur Radiol 2022; 33:196-206. [PMID: 36066730 DOI: 10.1007/s00330-022-09036-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To study longitudinal changes in tuber and whole-brain perfusion in children with tuberous sclerosis complex (TSC) using arterial spin labeling (ASL) perfusion MRI and correlate them with pathological EEG slow wave activity and neurodevelopmental outcomes. METHODS Retrospective longitudinal cohort study of 13 children with TSC, 3 to 6 serial ASL-MRI scans between 2 months and 7 years of age (53 scans in total), and an EEG examination performed within 2 months of the last MRI. Tuber cerebral blood flow (CBF) values were calculated in tuber segmentation masks, and tuber:cortical CBF ratios were used to study tuber perfusion. Logistic regression analysis was performed to identify which initial tuber characteristics (CBF value, volume, location) in the first MRI predicted tubers subsequently associated with EEG slow waves. Whole-brain and lobar CBF values were extracted for all patient scans and age-matched controls. CBF ratios were compared in patients and controls to study longitudinal changes in whole-brain CBF. RESULTS Perfusion was reduced in tubers associated with EEG slow waves compared with other tubers. Low tuber CBF values around 6 months of age and large tuber volumes were predictive of tubers subsequently associated with EEG slow waves. Patients with severe developmental delay had more severe whole-brain hypoperfusion than those with no/mild delay, which became apparent after 2 years of age and were not associated with a higher tuber load. CONCLUSIONS Dynamic changes in tuber and brain perfusion occur over time. Perfusion is significantly reduced in tubers associated with EEG slow waves. Whole-brain perfusion is significantly reduced in patients with severe delay. KEY POINTS • Tubers associated with EEG slow wave activity were significantly more hypoperfused than other tubers, especially after 1 year of age. • Larger and more hypoperfused tubers at 6 months of age were more likely to subsequently be associated with pathological EEG slow wave activity. • Patients with severe developmental delay had more extensive and severe global hypoperfusion than those without developmental delay.
Collapse
|
89
|
Buchhalter J, Neuray C, Cheng JY, D’Cruz O, Datta AN, Dlugos D, French J, Haubenberger D, Hulihan J, Klein P, Komorowski RW, Kramer L, Lothe A, Nabbout R, Perucca E, der Ark PV. EEG Parameters as Endpoints in Epilepsy Clinical Trials- An Expert Panel Opinion Paper. Epilepsy Res 2022; 187:107028. [DOI: 10.1016/j.eplepsyres.2022.107028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/29/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022]
|
90
|
Neal A, Bouet R, Lagarde S, Ostrowsky‐Coste K, Maillard L, Kahane P, Touraine R, Catenoix H, Montavont A, Isnard J, Arzimanoglou A, Hermier M, Guenot M, Bartolomei F, Rheims S, Jung J. Epileptic spasms are associated with increased stereo-electroencephalography derived functional connectivity in tuberous sclerosis complex. Epilepsia 2022; 63:2359-2370. [PMID: 35775943 PMCID: PMC9796462 DOI: 10.1111/epi.17353] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Epileptic spasms (ES) are common in tuberous sclerosis complex (TSC). However, the underlying network alterations and relationship with epileptogenic tubers are poorly understood. We examined interictal functional connectivity (FC) using stereo-electroencephalography (SEEG) in patients with TSC to investigate the relationship between tubers, epileptogenicity, and ES. METHODS We analyzed 18 patients with TSC who underwent SEEG (mean age = 11.5 years). The dominant tuber (DT) was defined as the most epileptogenic tuber using the epileptogenicity index. Epileptogenic zone (EZ) organization was quantitatively separated into focal (isolated DT) and complex (all other patterns). Using a 20-min interictal recording, FC was estimated with nonlinear regression, h2 . We calculated (1) intrazone FC within all sampled tubers and normal-appearing cortical zones, respectively; and (2) interzone FC involving connections between DT, other tubers, and normal cortex. The relationship between FC and (1) presence of ES as a current seizure type at the time of SEEG, (2) EZ organization, and (3) epileptogenicity was analyzed using a mixed generalized linear model. Spike rate and distance between zones were considered in the model as covariates. RESULTS Six patients had ES as a current seizure type at time of SEEG. ES patients had a greater number of tubers with a fluid-attenuated inversion recovery hypointense center (p < .001), and none had TSC1 mutations. The presence of ES was independently associated with increased FC within both intrazone (p = .033) and interzone (p = .011) networks. Post hoc analyses identified that increased FC was associated with ES across tuber and nontuber networks. EZ organization and epileptogenicity biomarkers were not associated with FC. SIGNIFICANCE Increased cortical synchrony among both tuber and nontuber networks is characteristic of patients with ES and independent of both EZ organization and tuber epileptogenicity. This further supports the prospect of FC biomarkers aiding treatment paradigms in TSC.
Collapse
Affiliation(s)
- Andrew Neal
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance,Department of Functional Neurology and EpileptologyLyon Civil Hospices, member of the ERN EpiCARE, and Lyon 1 UniversityLyonFrance,Department of Neuroscience, Faculty of Medicine, Nursing, and Health SciencesCentral Clinical School, Monash UniversityMelbourneVictoriaAustralia
| | - Romain Bouet
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance
| | - Stanislas Lagarde
- Epileptology Department, Timone HospitalPublic Assistance Hospitals of Marseille, member of the ERN EpiCAREMarseilleFrance,Institute of Systems Neurosciences, National Institute of Health and Medical ResearchAix‐Marseille UniversityMarseilleFrance
| | - Karine Ostrowsky‐Coste
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance,Department of Pediatric Clinical Epileptology, Sleep Disorders, and Functional NeurologyLyon Civil Hospices, member of the ERN EpiCARELyonFrance
| | - Louis Maillard
- Neurology DepartmentUniversity Hospital of Nancy, member of the ERN EpiCARENancyFrance
| | - Philippe Kahane
- Grenoble‐Alpes University Hospital Center, collaborating partner of the ERN EpiCAREGrenoble‐Alpes University, Grenoble Institute of Neuroscience, National Institute of Health and Medical ResearchGrenobleFrance
| | - Renaud Touraine
- Department of GeneticsSaint Etienne University Hospital Center–North HospitalSaint‐Priest‐en‐JarezFrance
| | - Helene Catenoix
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance,Department of Functional Neurology and EpileptologyLyon Civil Hospices, member of the ERN EpiCARE, and Lyon 1 UniversityLyonFrance
| | - Alexandra Montavont
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance,Department of Functional Neurology and EpileptologyLyon Civil Hospices, member of the ERN EpiCARE, and Lyon 1 UniversityLyonFrance
| | - Jean Isnard
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance,Department of Functional Neurology and EpileptologyLyon Civil Hospices, member of the ERN EpiCARE, and Lyon 1 UniversityLyonFrance
| | - Alexis Arzimanoglou
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance,Department of Pediatric Clinical Epileptology, Sleep Disorders, and Functional NeurologyLyon Civil Hospices, member of the ERN EpiCARELyonFrance
| | - Marc Hermier
- Department of NeuroradiologyLyon Civil HospicesLyonFrance
| | - Marc Guenot
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance,Department of Functional NeurosurgeryLyon Civil Hospices, member of the ERN EpiCARE, and Lyon 1 UniversityLyonFrance
| | - Fabrice Bartolomei
- Epileptology Department, Timone HospitalPublic Assistance Hospitals of Marseille, member of the ERN EpiCAREMarseilleFrance,Institute of Systems Neurosciences, National Institute of Health and Medical ResearchAix‐Marseille UniversityMarseilleFrance
| | - Sylvain Rheims
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance,Department of Functional Neurology and EpileptologyLyon Civil Hospices, member of the ERN EpiCARE, and Lyon 1 UniversityLyonFrance,Epilepsy InstituteLyonFrance
| | - Julien Jung
- Eduwell team, Inserm U1028, CNRS UMR5292, UCBL1, UJMLyon Neuroscience Research CenterLyonFrance,Department of Functional Neurology and EpileptologyLyon Civil Hospices, member of the ERN EpiCARE, and Lyon 1 UniversityLyonFrance
| |
Collapse
|
91
|
Advances in the genetics and neuropathology of tuberous sclerosis complex: edging closer to targeted therapy. Lancet Neurol 2022; 21:843-856. [DOI: 10.1016/s1474-4422(22)00213-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 03/09/2022] [Accepted: 05/11/2022] [Indexed: 12/23/2022]
|
92
|
Foryś-Basiejko M, Kotulska K, Maryniak A, Siłuszyk A, Szkop M, Borkowska J, Sugalska M, Głowacka-Walas J, Jóźwiak S. Epilepsy and Language Development in 8–36-Month-Old Toddlers with Tuberous Sclerosis Complex. J Clin Med 2022; 11:jcm11154564. [PMID: 35956179 PMCID: PMC9369686 DOI: 10.3390/jcm11154564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
This paper aimed to assess language development in infants and toddlers with tuberous sclerosis complex (TSC) and epilepsy, which increase the risk of autism spectrum disorder. We assessed language development in 61 patients with TSC at 8–36 months using a standardized Speech Development and Communication Inventory tool. The results showed differences in outcomes due to the duration of the seizures and the number of drugs (pFDR = 0.007 **—pFDR = 0.037 *). Children with TSC with longer epilepsy duration and receiving more antiepileptic drugs have a greater risk of language development delay.
Collapse
Affiliation(s)
- Małgorzata Foryś-Basiejko
- Department of Child Clinical Psychology and Family, Faculty of Psychology, University of Warsaw, 00-183 Warszawa, Poland
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, 04-736 Warszawa, Poland
- Department of Pediatric Neurology, Medical University of Warsaw, 02-091 Warszawa, Poland
- Correspondence:
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, 04-736 Warszawa, Poland
| | - Agnieszka Maryniak
- Department of Child Clinical Psychology and Family, Faculty of Psychology, University of Warsaw, 00-183 Warszawa, Poland
| | - Agata Siłuszyk
- Department of Pediatric Neurology, Medical University of Warsaw, 02-091 Warszawa, Poland
| | - Monika Szkop
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, 04-736 Warszawa, Poland
| | - Julita Borkowska
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, 04-736 Warszawa, Poland
| | - Monika Sugalska
- Department of Pediatric Neurology, Medical University of Warsaw, 02-091 Warszawa, Poland
| | - Jagoda Głowacka-Walas
- Institute of Computer Science, Warsaw University of Technology, 00-665 Warszawa, Poland
- Transition Technologies Science, 01-030 Warsaw, Poland
| | - Sergiusz Jóźwiak
- Department of Pediatric Neurology, Medical University of Warsaw, 02-091 Warszawa, Poland
| |
Collapse
|
93
|
Scheper M, Romagnolo A, Besharat ZM, Iyer AM, Moavero R, Hertzberg C, Weschke B, Riney K, Feucht M, Scholl T, Petrak B, Maulisova A, Nabbout R, Jansen AC, Jansen FE, Lagae L, Urbanska M, Ferretti E, Tempes A, Blazejczyk M, Jaworski J, Kwiatkowski DJ, Jozwiak S, Kotulska K, Sadowski K, Borkowska J, Curatolo P, Mills JD, Aronica E. miRNAs and isomiRs: Serum-Based Biomarkers for the Development of Intellectual Disability and Autism Spectrum Disorder in Tuberous Sclerosis Complex. Biomedicines 2022; 10:biomedicines10081838. [PMID: 36009385 PMCID: PMC9405248 DOI: 10.3390/biomedicines10081838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare multi-system genetic disorder characterized by a high incidence of epilepsy and neuropsychiatric manifestations known as tuberous-sclerosis-associated neuropsychiatric disorders (TANDs), including autism spectrum disorder (ASD) and intellectual disability (ID). MicroRNAs (miRNAs) are small regulatory non-coding RNAs that regulate the expression of more than 60% of all protein-coding genes in humans and have been reported to be dysregulated in several diseases, including TSC. In the current study, RNA sequencing analysis was performed to define the miRNA and isoform (isomiR) expression patterns in serum. A Receiver Operating Characteristic (ROC) curve analysis was used to identify circulating molecular biomarkers, miRNAs, and isomiRs, able to discriminate the development of neuropsychiatric comorbidity, either ASD, ID, or ASD + ID, in patients with TSC. Part of our bioinformatics predictions was verified with RT-qPCR performed on RNA isolated from patients’ serum. Our results support the notion that circulating miRNAs and isomiRs have the potential to aid standard clinical testing in the early risk assessment of ASD and ID development in TSC patients.
Collapse
Affiliation(s)
- Mirte Scheper
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.S.); (A.R.); (A.M.I.)
| | - Alessia Romagnolo
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.S.); (A.R.); (A.M.I.)
| | - Zein Mersini Besharat
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (Z.M.B.); (E.F.)
| | - Anand M. Iyer
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.S.); (A.R.); (A.M.I.)
- Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Romina Moavero
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, 00133 Rome, Italy; (R.M.); (P.C.)
- Child Neurology Unit, Neuroscience Department, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Christoph Hertzberg
- Diagnose-und Behandlungszentrum für Kinder, Vivantes-Klinikum Neukölln, 12351 Berlin, Germany;
| | - Bernhard Weschke
- Department of Neuropediatrics, Charité University Medicine Berlin, 13353 Berlin, Germany;
| | - Kate Riney
- Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia;
- Neurosciences Unit, Queensland Children’s Hospital, South Brisbane, QLD 4101, Australia
| | - Martha Feucht
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, “Member of ERN EpiCARE”, 1090 Vienna, Austria; (M.F.); (T.S.)
| | - Theresa Scholl
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, “Member of ERN EpiCARE”, 1090 Vienna, Austria; (M.F.); (T.S.)
| | - Borivoj Petrak
- Motol University Hospital, Charles University, 15000 Prague, Czech Republic; (B.P.); (A.M.)
| | - Alice Maulisova
- Motol University Hospital, Charles University, 15000 Prague, Czech Republic; (B.P.); (A.M.)
| | - Rima Nabbout
- Reference Centre for Rare Epilepsies, Department of Pediatric Neurology, Necker Enfants Malades University Hospital, APHP, Member of ERN EpiCARE, Université de Paris, 149 Rue de Sèvres, 75015 Paris, France;
| | - Anna C. Jansen
- Department of Translational Neurosciences, University of Antwerp, 2000 Antwerp, Belgium;
| | - Floor E. Jansen
- Department of Child Neurology, Brain Center University Medical Center, Member of ERN EpiCare, 3584 BA Utrecht, The Netherlands;
| | - Lieven Lagae
- Department of Development and Regeneration Section Pediatric Neurology, University Hospitals KU Leuven, 3000 Leuven, Belgium;
| | - Malgorzata Urbanska
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland; (M.U.); (S.J.); (K.K.); (K.S.); (J.B.)
| | - Elisabetta Ferretti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (Z.M.B.); (E.F.)
| | - Aleksandra Tempes
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland; (A.T.); (M.B.); (J.J.)
| | - Magdalena Blazejczyk
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland; (A.T.); (M.B.); (J.J.)
| | - Jacek Jaworski
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland; (A.T.); (M.B.); (J.J.)
| | | | - Sergiusz Jozwiak
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland; (M.U.); (S.J.); (K.K.); (K.S.); (J.B.)
- Department of Child Neurology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland; (M.U.); (S.J.); (K.K.); (K.S.); (J.B.)
| | - Krzysztof Sadowski
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland; (M.U.); (S.J.); (K.K.); (K.S.); (J.B.)
| | - Julita Borkowska
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland; (M.U.); (S.J.); (K.K.); (K.S.); (J.B.)
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, 00133 Rome, Italy; (R.M.); (P.C.)
| | - James D. Mills
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.S.); (A.R.); (A.M.I.)
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1E 6BT, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter SL9 0RJ, UK
- Correspondence: (J.D.M.); (E.A.)
| | - Eleonora Aronica
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.S.); (A.R.); (A.M.I.)
- Correspondence: (J.D.M.); (E.A.)
| | | |
Collapse
|
94
|
Patel AA, Birbeck GL, Mazumdar M, Mwanza S, Nyirongo R, Berejena D, Kasolo J, Mwale T, Nambeye V, Nkole KL, Kawatu N, Zhang B, Rotenberg A. Identifying biomarkers for epilepsy after cerebral malaria in Zambian children: rationale and design of a prospective observational study. BMJ Open 2022; 12:e062948. [PMID: 35851014 PMCID: PMC9297226 DOI: 10.1136/bmjopen-2022-062948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Malaria affecting the central nervous system (CM) is a major contributor to paediatric epilepsy in resource-poor settings, with 10%-16% of survivors developing epilepsy within 2 years of infection. Despite high risk for post-malaria epilepsy (PME), biomarkers indicating which CM survivors will develop epilepsy are absent. Such biomarkers are essential to identify those at highest risk who might benefit most from close surveillance and/or preventive treatments. Electroencephalography (EEG) contains signals (specifically gamma frequency activity), which are correlated with higher risk of PME and provide a biomarker for the development of epilepsy. We propose to study the sensitivity of quantitative and qualitative EEG metrics in predicting PME, and the potential increased sensitivity of this measure with additional clinical metrics. Our goal is to develop a predictive PME index composed of EEG and clinical history metrics that are highly feasible to obtain in low-resourced regions. METHODS AND ANALYSES This prospective observational study being conducted in Eastern Zambia will recruit 250 children aged 6 months to 11 years presenting with acute CM and follow them for two years. Children with pre-existing epilepsy diagnoses will be excluded. Outcome measures will include qualitative and quantitative analysis of routine EEG recordings, as well as clinical metrics in the acute and subacute period, including histidine-rich protein 2 levels of parasite burden, depth and length of coma, presence and severity of acute seizures, presence of hypoglycaemia, maximum temperature and 1-month post-CM neurodevelopmental assessment scores. We will test the performance of these EEG and clinical metrics in predicting development of epilepsy through multivariate logistic regression analyses. ETHICS AND DISSEMINATION This study has been approved by the Boston Children's Hospital Institutional Review Board, University of Zambia Biomedical Research Ethics Committee, and National Health Research Authority of Zambia. Results will be disseminated locally in Zambia followed by publication in international, open access, peer-reviewed journals when feasible.
Collapse
Affiliation(s)
- Archana A Patel
- Neurology, Division of Epilepsy & Clinical Neurophysiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Paediatrics and Child Health, University of Zambia School of Medicine, Lusaka, Zambia
| | - Gretchen L Birbeck
- Department of Paediatrics and Child Health, University of Zambia School of Medicine, Lusaka, Zambia
- Epilepsy Division, University of Rochester Department of Neurology, Rochester, New York, USA
| | - Maitreyi Mazumdar
- Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Environmental Health, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | | | | | | | - Joseph Kasolo
- Paediatrics, Chipata Central Hospital, Chipata, Zambia
| | - Tina Mwale
- Paediatrics, Chipata Central Hospital, Chipata, Zambia
| | | | | | - Nfwama Kawatu
- University Teaching Hospitals- Children's Hospital, Lusaka, Zambia
| | - Bo Zhang
- Department of Neurology and ICCTR Biostatistics and Research Design Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander Rotenberg
- Neurology, Division of Epilepsy & Clinical Neurophysiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
95
|
Specchio N, Pietrafusa N, Perucca E, Cross JH. New paradigms for the treatment of pediatric monogenic epilepsies: Progressing toward precision medicine. Epilepsy Behav 2022; 131:107961. [PMID: 33867301 DOI: 10.1016/j.yebeh.2021.107961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/25/2022]
Abstract
Despite the availability of 28 antiseizure medications (ASMs), one-third of people with epilepsy fail to achieve sustained freedom from seizures. Clinical outcome is even poorer for children with developmental and epileptic encephalopathies (DEEs), many of which are due to single-gene mutations. Discovery of causative genes, however, has paved the way to understanding the molecular mechanism underlying these epilepsies, and to the rational application, or development, of precision treatments aimed at correcting the specific functional defects or their consequences. This article provides an overview of current progress toward precision medicine (PM) in the management of monogenic pediatric epilepsies, by focusing on four different scenarios, namely (a) rational selection of ASMs targeting specifically the underlying pathogenetic mechanisms; (b) development of targeted therapies based on novel molecules; (c) use of dietary treatments or food constituents aimed at correcting specific metabolic defects; and (d) repurposing of medications originally approved for other indications. This article is part of the Special Issue "Severe Infantile Epilepsies".
Collapse
Affiliation(s)
- Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Rome, Italy.
| | - Nicola Pietrafusa
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Rome, Italy
| | - Emilio Perucca
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - J Helen Cross
- UCL NIHR BRC Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London, UK
| |
Collapse
|
96
|
Riney K, Bogacz A, Somerville E, Hirsch E, Nabbout R, Scheffer IE, Zuberi SM, Alsaadi T, Jain S, French J, Specchio N, Trinka E, Wiebe S, Auvin S, Cabral-Lim L, Naidoo A, Perucca E, Moshé SL, Wirrell EC, Tinuper P. International League Against Epilepsy classification and definition of epilepsy syndromes with onset at a variable age: position statement by the ILAE Task Force on Nosology and Definitions. Epilepsia 2022; 63:1443-1474. [PMID: 35503725 DOI: 10.1111/epi.17240] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 01/15/2023]
Abstract
The goal of this paper is to provide updated diagnostic criteria for the epilepsy syndromes that have a variable age of onset, based on expert consensus of the International League Against Epilepsy Nosology and Definitions Taskforce (2017-2021). We use language consistent with current accepted epilepsy and seizure classifications and incorporate knowledge from advances in genetics, electroencephalography, and imaging. Our aim in delineating the epilepsy syndromes that present at a variable age is to aid diagnosis and to guide investigations for etiology and treatments for these patients.
Collapse
Affiliation(s)
- Kate Riney
- Neurosciences Unit, Queensland Children's Hospital, South Brisbane, Queensland, Australia.,Faculty of Medicine, University of Queensland, South Brisbane, Queensland, Australia
| | - Alicia Bogacz
- Institute of Neurology, University of the Republic, Montevideo, Uruguay
| | - Ernest Somerville
- Prince of Wales Hospital, Sydney, New South Wales, Australia.,University of New South Wales, Sydney, New South Wales, Australia
| | - Edouard Hirsch
- Francis Rohmer Epilepsy Unit, Hautepierre Hospital, Strasbourg, France.,National Institute of Health and Medical Research 1258, Strasbourg, France.,Federation of Translational Medicine of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Rima Nabbout
- Reference Centre for Rare Epilepsies, Assistance Publique - Hôpitaux de Paris, Department of Pediatric Neurology, Necker-Enfants Malades Hospital, Member of Epicare, Paris, France.,Imagine Institute, National Institute of Health and Medical Research Mixed Unit of Research 1163, Paris, France.,University City University, Paris, France
| | - Ingrid E Scheffer
- Austin Health, Royal Children's Hospital, Florey Institute and Murdoch Children's Research Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Sameer M Zuberi
- University City University, Paris, France.,Paediatric Neurosciences Research Group, Royal Hospital for Children, Glasgow, UK.,Institute of Health & Wellbeing, University of Glasgow, Glasgow, UK
| | - Taoufik Alsaadi
- Department of Neurology, American Center for Psychiatry and Neurology, Abu Dhabi, United Arab Emirates
| | | | - Jacqueline French
- New York University Grossman School of Medicine and NYU Langone Health, New York, New York, USA
| | - Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, Scientific Institute for Research and Health Care, member of EpiCARE, Rome, Italy
| | - Eugen Trinka
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University, Center for Cognitive Neuroscience, member of EpiCARE, Salzburg, Austria.,Neuroscience Institute, Christian Doppler University Hospital, Center for Cognitive Neuroscience, Salzburg, Austria.,Department of Public Health, Health Services Research and Health Technology Assessment, University for Health Sciences, Medical Informatics, and Technology, Hall in Tirol, Austria
| | - Samuel Wiebe
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Stéphane Auvin
- Institut Universitaire de France, Paris, France.,Paediatric Neurology, Assistance Publique - Hôpitaux de Paris, Robert-Debré Hospital, Paris, France.,University of Paris, Paris, France
| | - Leonor Cabral-Lim
- Department of Neurosciences, College of Medicine and Philippine General Hospital, Health Sciences Center, University of the Philippines Manila, Manila, the Philippines
| | - Ansuya Naidoo
- Neurology Unit, Greys Hospital, Pietermaritzburg, South Africa.,Department of Neurology, University of KwaZulu Natal, KwaZulu Natal, South Africa
| | - Emilio Perucca
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia.,Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Solomon L Moshé
- Isabelle Rapin Division of Child Neurology, Saul R. Korey Department of Neurology and Departments of Neuroscience and Pediatrics, Albert Einstein College of Medicine, New York, New York, USA.,Montefiore Medical Center, Bronx, New York, USA
| | - Elaine C Wirrell
- Divisions of Child and Adolescent Neurology and Epilepsy, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Paolo Tinuper
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Reference Centre for Rare and Complex Epilepsies, IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| |
Collapse
|
97
|
Jonsson H, Lehto M, Vanhatalo S, Gaily E, Linnankivi T. Visual field defects after vigabatrin treatment during infancy: retrospective population-based study. Dev Med Child Neurol 2022; 64:641-648. [PMID: 34716587 DOI: 10.1111/dmcn.15099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 02/02/2023]
Abstract
AIM To investigate the prevalence of vigabatrin-attributed visual field defect (VAVFD) in infantile spasms and the utility of optical coherence tomography (OCT) in detecting vigabatrin-related damage. METHOD We examined visual fields by Goldmann or Octopus perimetry and the thickness of peripapillary retinal nerve fiber layer (RNFL) with spectral-domain OCT at school age or adolescence. RESULTS Out of 88 patients (38 females, mean age at study 15y, SD 4y 3mo, range 6y 4mo-23y 3mo [n=65] or deceased [n=21] or moved abroad [n=2]) exposed to vigabatrin in infancy, 28 were able to perform formal visual field testing. Two had visual field defect from structural causes. We found mild VAVFD in four patients and severe VAVFD in one patient. Median vigabatrin treatment duration for those with normal visual field was 11 months compared to 19 months for those with VAVFD (p=0.04). OCT showed concomitant attenuated RNFL in three children with VAVFD, and was normal in one. The temporal half of the peripapillary RNFL was significantly thinner in the VAVFD group compared to the normal visual field group. INTERPRETATION The overall prevalence of VAVFD is lower after exposure in infancy compared to 52% which has been reported after exposure in adulthood. The risk increases with longer treatment duration. Further studies should identify infants particularly susceptible to VAVFD and clarify the role of OCT.
Collapse
Affiliation(s)
- Henna Jonsson
- Department of Pediatric Neurology, New Children's Hospital and Pediatric Research Center, Epilepsia Helsinki, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mikko Lehto
- Department of Ophthalmology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Sampsa Vanhatalo
- Department of Children's Clinical Neurophysiology, BABA Center, Department of Clinical Neurophysiology, Children's Hospital, Helsinki University Hospital, Helsinki, Finland.,Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Eija Gaily
- Department of Pediatric Neurology, New Children's Hospital and Pediatric Research Center, Epilepsia Helsinki, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tarja Linnankivi
- Department of Pediatric Neurology, New Children's Hospital and Pediatric Research Center, Epilepsia Helsinki, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
98
|
De Ridder J, Kotulska K, Curatolo P, Jansen AC, Aronica E, Kwiatkowski DJ, Jansen FE, Jóźwiak S, Lagae L. Evolution of electroencephalogram in infants with tuberous sclerosis complex and neurodevelopmental outcome: a prospective cohort study. Dev Med Child Neurol 2022; 64:495-501. [PMID: 34601720 DOI: 10.1111/dmcn.15073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 01/20/2023]
Abstract
AIM To describe the evolution of electroencephalogram (EEG) characteristics in infants with tuberous sclerosis complex (TSC) and the relationship with neurodevelopmental outcome at 24 months. METHOD Eighty-three infants were enrolled in the EPISTOP trial and underwent serial EEG follow-up until the age of 24 months (males n=45, females n=37, median age at enrolment 28d, interquartile range 14-54d). Maturation of the EEG background and epileptiform discharges were compared between the TSC1 and TSC2 variants and between preventive and conventional groups respectively. RESULTS Children with TSC2 more frequently had a slower posterior dominant rhythm (PDR) at 24 months (51% vs 11%, p=0.002), a higher number of epileptiform foci (median=8 vs 4, p=0.003), and a lower fraction of EEGs without epileptiform discharges (18% vs 61%, p=0.001) at follow-up. A slower PDR at 24 months was significantly associated with lower cognitive (median=70 vs 80, p=0.028) and motor developmental quotients (median=70 vs 79, p=0.008). A higher fraction of EEGs without epileptiform discharges was associated with a lower probability of autism spectrum disorder symptoms (odds ratio=0.092, 95% confidence interval=0.009-0.912, p=0.042) and higher cognitive (p=0.004), language (p=0.002), and motor (p=0.001) developmental quotients at 24 months. INTERPRETATION TSC2 is associated with more abnormal EEG characteristics compared to TSC1, which are predictive for neurodevelopmental outcome.
Collapse
Affiliation(s)
- Jessie De Ridder
- Department of Development and Regeneration, Section Pediatric Neurology, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
| | - Anna C Jansen
- Pediatric Neurology Unit, University Hospital Brussel, Brussels, Belgium
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.,Stichting Epilepsie Instellingen Nederland, Heemstede, the Netherlands
| | | | - Floor E Jansen
- Department of Child Neurology, Brain Centre, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Sergiusz Jóźwiak
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland.,Department of Child Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Lieven Lagae
- Department of Development and Regeneration, Section Pediatric Neurology, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | | |
Collapse
|
99
|
Pong AW, Ross J, Tyrlikova I, Giermek AJ, Kohli MP, Khan YA, Salgado RD, Klein P. Epilepsy: Expert opinion on emerging drugs in phase 2/3 clinical trials. Expert Opin Emerg Drugs 2022; 27:75-90. [PMID: 35341431 DOI: 10.1080/14728214.2022.2059464] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Despite the existence of over 30 anti-seizure medications (ASM), including 20 over the last 30 years, a third of patients with epilepsy remain refractory to treatment, with no disease-modifying or preventive therapies until very recently. The development of new ASMs with new mechanisms of action is therefore critical. Recent clinical trials of new treatments have shifted focus from the traditional common epilepsies to rare, genetic epilepsies with known mechanistic targets for treatment and disease-specific animal models. AREAS COVERED ASMs in phase 2a/b and 3 clinical trials target cholesterol, serotonin, sigma-1 receptors, potassium channels and metabotrobic glutamate receptors. Neuroinflammation, protein misfolding, abnormal thalamocortical firing, and molecular deficiencies are among the targeted pathways. Clinically, the current phase 2a/b-3 agents hold promise for variety of epilepsy conditions, from developmental epileptic encephalopathies (Dravet Syndrome, Lennox-Gastaut syndrome, CDKL5 and PCDH19, Rett's Syndrome), Infantile Spasms, Tuberous Sclerosis as well as focal and idiopathic generalized epilepsies and acute rescue therapy for cluster seizures. EXPERT OPINION New delivery mechanisms increase potency and site-specificity of existing drugs. Novel mechanisms of action involve cholesterol degradation, mitochondrial pathways, anti-inflammation and neuro-regeneration. Earlier identification of genetic conditions through genetic testing will allow for earlier use of disease specific and disease-modifying therapies.
Collapse
Affiliation(s)
- Amanda W Pong
- Comprehensive Neurology Clinics of Bethesda, Mid-Atlantic Epilepsy and Sleep Center,Bethesda, MD, USA
| | - Jonathan Ross
- Comprehensive Neurology Clinics of Bethesda, Mid-Atlantic Epilepsy and Sleep Center,Bethesda, MD, USA
| | - Ivana Tyrlikova
- Comprehensive Neurology Clinics of Bethesda, Mid-Atlantic Epilepsy and Sleep Center,Bethesda, MD, USA
| | - Alexander J Giermek
- Comprehensive Neurology Clinics of Bethesda, Mid-Atlantic Epilepsy and Sleep Center,Bethesda, MD, USA
| | - Maya P Kohli
- Comprehensive Neurology Clinics of Bethesda, Mid-Atlantic Epilepsy and Sleep Center,Bethesda, MD, USA
| | - Yousef A Khan
- Comprehensive Neurology Clinics of Bethesda, Mid-Atlantic Epilepsy and Sleep Center,Bethesda, MD, USA
| | - Roger D Salgado
- Comprehensive Neurology Clinics of Bethesda, Mid-Atlantic Epilepsy and Sleep Center,Bethesda, MD, USA
| | - Pavel Klein
- Comprehensive Neurology Clinics of Bethesda, Mid-Atlantic Epilepsy and Sleep Center,Bethesda, MD, USA
| |
Collapse
|
100
|
Sato A, Tominaga K, Iwatani Y, Kato Y, Wataya-Kaneda M, Makita K, Nemoto K, Taniike M, Kagitani-Shimono K. Abnormal White Matter Microstructure in the Limbic System Is Associated With Tuberous Sclerosis Complex-Associated Neuropsychiatric Disorders. Front Neurol 2022; 13:782479. [PMID: 35359647 PMCID: PMC8963953 DOI: 10.3389/fneur.2022.782479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTuberous sclerosis complex (TSC) is a genetic disease that arises from TSC1 or TSC2 abnormalities and induces the overactivation of the mammalian/mechanistic target of rapamycin pathways. The neurological symptoms of TSC include epilepsy and tuberous sclerosis complex-associated neuropsychiatric disorders (TAND). Although TAND affects TSC patients' quality of life, the specific region in the brain associated with TAND remains unknown. We examined the association between white matter microstructural abnormalities and TAND, using diffusion tensor imaging (DTI).MethodsA total of 19 subjects with TSC and 24 age-matched control subjects were enrolled. Tract-based spatial statistics (TBSS) were performed to assess group differences in fractional anisotropy (FA) between the TSC and control groups. Atlas-based association analysis was performed to reveal TAND-related white matter in subjects with TSC. Multiple linear regression was performed to evaluate the association between TAND and the DTI parameters; FA and mean diffusivity in seven target regions and projection fibers.ResultsThe TBSS showed significantly reduced FA in the right hemisphere and particularly in the inferior frontal occipital fasciculus (IFOF), inferior longitudinal fasciculus (ILF), superior longitudinal fasciculus (SLF), uncinate fasciculus (UF), and genu of corpus callosum (CC) in the TSC group relative to the control group. In the association analysis, intellectual disability was widely associated with all target regions. In contrast, behavioral problems and autistic features were associated with the limbic system white matter and anterior limb of the internal capsule (ALIC) and CC.ConclusionThe disruption of white matter integrity may induce underconnectivity between cortical and subcortical regions. These findings suggest that TANDs are not the result of an abnormality in a specific brain region, but rather caused by connectivity dysfunction as a network disorder. This study indicates that abnormal white matter connectivity including the limbic system is relevant to TAND. The analysis of brain and behavior relationship is a feasible approach to reveal TAND related white matter and neural networks. TAND should be carefully assessed and treated at an early stage.
Collapse
Affiliation(s)
- Akemi Sato
- United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Koji Tominaga
- United Graduate School of Child Development, Osaka University, Osaka, Japan
- Molecular Research Center for Children's Mental Development, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiko Iwatani
- United Graduate School of Child Development, Osaka University, Osaka, Japan
- Molecular Research Center for Children's Mental Development, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoko Kato
- Molecular Research Center for Children's Mental Development, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Mari Wataya-Kaneda
- Division of Health Science, Department of Neurocutaneous Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kai Makita
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan
| | - Kiyotaka Nemoto
- Division of Clinical Medicine, Department of Psychiatry, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masako Taniike
- United Graduate School of Child Development, Osaka University, Osaka, Japan
- Molecular Research Center for Children's Mental Development, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kuriko Kagitani-Shimono
- United Graduate School of Child Development, Osaka University, Osaka, Japan
- Molecular Research Center for Children's Mental Development, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
- *Correspondence: Kuriko Kagitani-Shimono
| |
Collapse
|