151
|
Kourakis MJ, Borba C, Zhang A, Newman-Smith E, Salas P, Manjunath B, Smith WC. Parallel visual circuitry in a basal chordate. eLife 2019; 8:44753. [PMID: 30998184 PMCID: PMC6499539 DOI: 10.7554/elife.44753] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 04/11/2019] [Indexed: 12/28/2022] Open
Abstract
A common CNS architecture is observed in all chordates, from vertebrates to basal chordates like the ascidian Ciona. Ciona stands apart among chordates in having a complete larval connectome. Starting with visuomotor circuits predicted by the Ciona connectome, we used expression maps of neurotransmitter use with behavioral assays to identify two parallel visuomotor circuits that are responsive to different components of visual stimuli. The first circuit is characterized by glutamatergic photoreceptors and responds to the direction of light. These photoreceptors project to cholinergic motor neurons, via two tiers of cholinergic interneurons. The second circuit responds to changes in ambient light and mediates an escape response. This circuit uses GABAergic photoreceptors which project to GABAergic interneurons, and then to cholinergic interneurons. Our observations on the behavior of larvae either treated with a GABA receptor antagonist or carrying a mutation that eliminates photoreceptors indicate the second circuit is disinhibitory.
Collapse
Affiliation(s)
- Matthew J Kourakis
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States
| | - Cezar Borba
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, United States
| | - Angela Zhang
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, Santa Barbara, United States
| | - Erin Newman-Smith
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States.,Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, United States
| | - Priscilla Salas
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, United States
| | - B Manjunath
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States
| | - William C Smith
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States.,Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, United States
| |
Collapse
|
152
|
Wu T, Ido K, Ohgoh M, Hanada T. Mode of seizure inhibition by sodium channel blockers, an SV2A ligand, and an AMPA receptor antagonist in a rat amygdala kindling model. Epilepsy Res 2019; 154:42-49. [PMID: 31035244 DOI: 10.1016/j.eplepsyres.2019.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/12/2019] [Accepted: 03/20/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE A number of antiepileptic drugs (AEDs) with a variety of modes of action, are effective in treating focal seizures. Several AEDs, such as perampanel (PER), levetiracetam (LEV), lacosamide (LCM), lamotrigine (LTG), and carbamazepine (CBZ), have been shown to elevate the seizure threshold in kindling models. These AEDs are clinically effective, but differences exist in the anti-seizure profiles of drugs with similar modes of action. Therefore, we hypothesized that there are differences in how these AEDs affect seizures. Here, we evaluated the effects of AEDs on various seizure parameters in a rat amygdala kindling model upon stimulation at the after-discharge threshold (ADT) and at three-times the ADT (3xADT) to characterize the differences in the effects of these AEDs. METHODS PER, LEV, LCM, LTG, CBZ, or vehicle was administered intraperitoneally to fully kindled rats. Changes in Racine seizure score, after-discharge duration (ADD), and latency to Racine score 4 generalized seizure (S4L) were measured to assess differences in the modes of seizure inhibition among the AEDs. Stimulation at 3xADT was used to eliminate the influence of any AED-induced elevation of the seizure threshold on these parameters. RESULTS PER, LEV, LCM, LTG, and CBZ significantly reduced the seizure score from Racine score 5 after stimulation at the ADT; this effect was lost with LEV and LTG after stimulation at 3xADT. PER and LEV significantly shortened the ADD when the seizure focus was stimulated at the ADT, whereas LCM, LTG, and CBZ did not. LEV, LCM, LTG, and CBZ failed to shorten the ADD upon stimulation at 3xADT. PER dose-dependently and significantly increased S4L, even at doses that were ineffective for seizure score reduction, after stimulation at both the ADT and 3xADT. LEV and LTG significantly increased S4L after stimulation at the ADT, whereas LCM and CBZ did not significantly increase S4L at any of the doses tested. CONCLUSIONS The sodium channel blockers (LCM, LTG, and CBZ) appeared to act by elevation of the seizure threshold via reduction of neuronal excitability, whereas the AMPA receptor antagonist (PER) and the SV2A ligand (LEV), as well as LTG, exerted their effects through the weakening of synaptic transmission in neuronal networks at the seizure focus. Maintenance of the effect of PER even at 3xADT suggests direct and strong modulation of excitatory synaptic transmission by PER, both at the focus and along the seizure propagation route. These findings may provide further rationale for usage of AEDs beyond their respective modes of action.
Collapse
Affiliation(s)
- Ting Wu
- Neurology Tsukuba Research Department, Discovery, Medicine Creation, Neurology Business Group, Eisai Co., Ltd. Japan
| | - Katsutoshi Ido
- Neurology Tsukuba Research Department, Discovery, Medicine Creation, Neurology Business Group, Eisai Co., Ltd. Japan
| | - Makoto Ohgoh
- Neurology Tsukuba Research Department, Discovery, Medicine Creation, Neurology Business Group, Eisai Co., Ltd. Japan
| | - Takahisa Hanada
- Clinical Science Department, Medical Division, Eisai Co., Ltd. Nishigokencho 13-1, Shinjuku-ku, Tokyo 162-0812, Japan.
| |
Collapse
|
153
|
Hartl E, Seethaler M, Lauseker M, Rémi J, Vollmar C, Noachtar S. Impact of withdrawal of antiepileptic medication on the duration of focal onset seizures. Seizure 2019; 67:40-44. [DOI: 10.1016/j.seizure.2019.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/11/2019] [Accepted: 03/07/2019] [Indexed: 10/27/2022] Open
|
154
|
Kim JE, Choi HC, Song HK, Kang TC. Perampanel Affects Up-Stream Regulatory Signaling Pathways of GluA1 Phosphorylation in Normal and Epileptic Rats. Front Cell Neurosci 2019; 13:80. [PMID: 30881292 PMCID: PMC6405474 DOI: 10.3389/fncel.2019.00080] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 02/18/2019] [Indexed: 01/30/2023] Open
Abstract
To elucidate the pharmacological properties of perampanel [2-(2-oxo-1-phenyl-5-pyridin-2-yl-1,2-dihydropyridin-3-yl)benzonitrile, a novel non-competitive antagonist of AMPA receptor], we investigated its effects on the up-stream regulatory pathways of GluA1 phosphorylation including protein kinase C (PKC), Ca2+-calmodulin-dependent protein kinase II (CAMKII), protein kinase A (PKA), extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), protein phosphatase (PP) 1, PP2A, and PP2B in normal and pilocarpine-induced epileptic rat model using Western blot analysis. In normal animals, perampanel affected GluA1 expression/phosphorylation, PKC, CAMKII, PKA, ERK1/2, JNK, and PPs activities. In epileptic rats, perampanel effectively inhibited spontaneous seizure activities. Perampanel enhanced phospho (p)-GluA1-S831 and -S845 ratios (phosphoprotein/total protein), while it reduced GluA1 expression. Perampanel also increased pCAMKII and pPKA ratios, which phosphorylate GluA1-S831 and -S845 site, respectively. Perampanel elevated pJNK and pPP2B ratios, which phosphorylates and dephosphorylates both GluA1-S831 and -S845 sits. Perampanel also increased pERK1/2 ratio in epileptic animals, while U0126 (an ERK1/2 inhibitor) did not affect pGluA1 ratios. Perampanel did not influence PKC, PP1, and PP2A expression levels and their phosphorylation ratios. In addition, perampanel did not have a detrimental impact on cognitive abilities of epileptic and normal rats in Morris water maze test. These findings suggest that perampanel may regulate AMPA receptor functionality via not only blockade of AMPA receptor but also the regulations of multiple molecules (CAMKII, PKA, JNK, and pPP2B)-mediated GluA1 phosphorylations without negative effects on cognition, although the effects of perampanel on PKC, PP1, and PP2A activities were different between normal and epileptic rats.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,College of Medicine, Institute of Epilepsy Research, Hallym University, Chuncheon, South Korea
| | - Hui-Chul Choi
- College of Medicine, Institute of Epilepsy Research, Hallym University, Chuncheon, South Korea.,Department of Neurology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Hong-Ki Song
- College of Medicine, Institute of Epilepsy Research, Hallym University, Chuncheon, South Korea.,Department of Neurology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,College of Medicine, Institute of Epilepsy Research, Hallym University, Chuncheon, South Korea
| |
Collapse
|
155
|
|
156
|
Lange F, Weßlau K, Porath K, Hörnschemeyer MF, Bergner C, Krause BJ, Mullins CS, Linnebacher M, Köhling R, Kirschstein T. AMPA receptor antagonist perampanel affects glioblastoma cell growth and glutamate release in vitro. PLoS One 2019; 14:e0211644. [PMID: 30716120 PMCID: PMC6361447 DOI: 10.1371/journal.pone.0211644] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/17/2019] [Indexed: 12/17/2022] Open
Abstract
Epileptic seizures are frequent in patients with glioblastoma, and anticonvulsive treatment is often necessary. While clinical guidelines recommend all approved anticonvulsants, so far it is still unclear which of the available drugs is the best therapeutic option for treating glioma-associated seizures, also in view of possible anti-tumorigenic effects. In our study, we employed four patient-derived low-passage cell lines of glioblastoma and three cell lines of brain metastases, and challenged these cultures with four anticonvulsants with different mechanisms of action: levetiracetam, valproic acid, carbamazepine and perampanel. Cell proliferation was determined by bromodeoxyuridine incorporation. To further analyze the effects of perampanel, apoptosis induction was measured by caspase 3/7 activation. Glutamate release was quantified and glucose uptake was determined using 18F-fluorodeoxyglucose. Real-time polymerase chain reaction was employed to assess the expression of genes associated with glutamate release and uptake in brain tumor cells. Of the four anticonvulsants, only perampanel showed systematic inhibitory effects on cell proliferation, whereas all other anticonvulsants failed to inhibit glioma and metastasis cell growth in vitro. Metastasis cells were much more resistant to perampanel than glioblastoma cell lines. Glucose uptake was attenuated in all glioblastoma cells after perampanel exposure, whereas cell death via apoptosis was not induced. Extracellular glutamate levels were found to be significantly higher in glioblastoma cell lines as compared to metastasis cell lines, but could be reduced by perampanel exposure. Incubation with perampanel up-regulated glutamine synthetase expression in glioblastoma cells, whereas treatment with valproic acid and levetiracetam downregulated excitatory amino acid transporter-2 expression. Overall, our data suggest that perampanel acts as an anticonvulsive drug and additionally mediated anti-tumorigenic effects.
Collapse
Affiliation(s)
- Falko Lange
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| | - Konrad Weßlau
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | - Katrin Porath
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | | | - Carina Bergner
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Bernd Joachim Krause
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | | | | | - Rüdiger Köhling
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| |
Collapse
|
157
|
Títoff V, Moury HN, Títoff IB, Kelly KM. Seizures, Antiepileptic Drugs, and CKD. Am J Kidney Dis 2019; 73:90-101. [DOI: 10.1053/j.ajkd.2018.03.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/06/2018] [Indexed: 01/19/2023]
|
158
|
Usefulness of perampanel with concomitant levetiracetam for patients with drug-resistant epilepsy. Eur J Paediatr Neurol 2019; 23:197-203. [PMID: 30424990 DOI: 10.1016/j.ejpn.2018.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/13/2018] [Accepted: 10/26/2018] [Indexed: 12/24/2022]
Abstract
PURPOSE The purpose was to evaluate the efficacy of treatment and the occurrence of aggression-related adverse events among children receiving perampanel (PER) with concomitant levetiracetam (LEV). METHODS Patients were selected according to the following criteria: 1) between 12 and 18 years old; 2) seizures refractory to at least 2 first-line drugs; 3) at least 4 seizures a month before PER administration; and 4) at least 12 months of follow-up. Patients were subdivided into groups with and without LEV as concomitant treatment. PER was administered at a dose of 2 mg/day, increasing by 2 mg/day every 2 weeks up to 12 mg/day if seizures appeared. In comparison with the baseline seizure frequency, response to PER treatment was classified as follows: complete cessation (100% seizure control); response (≥50% reduction in seizures); and exacerbation (≥50% increase in seizures). Responders were identified as patients showing complete cessation or response. RESULTS The study group comprised 39 outpatients with a mean age of 13.7 years at enrollment. Responder status was seen in 13 of the 19 patients with LEV and 4 of the 20 patients without LEV. PER appeared significantly more effective in patients with LEV than in those without LEV (p = 0.0076). Seizure-free status was significantly more frequent among patients with LEV (47.4%) than among those without LEV (15.0% (p = 0.0407)). Aggression was present in 2 patients without LEV, but none with LEV. CONCLUSION The present study suggests the utility of PER with concomitant LEV for children with drug-resistant epilepsy.
Collapse
|
159
|
Suda S, Kimura K. Therapeutic potential of AMPA receptor antagonist perampanel against cerebral ischemia: beyond epileptic disorder. Neural Regen Res 2019; 14:1525-1526. [PMID: 31089049 PMCID: PMC6557096 DOI: 10.4103/1673-5374.255964] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- Satoshi Suda
- Department of Neurology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Kazumi Kimura
- Department of Neurology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
160
|
Kim S, Kim TE, Kim D, Kim DW. Prolonged Stupor in Perampanel Overdose and Pharmacokinetic Considerations. J Epilepsy Res 2018; 8:87-89. [PMID: 30809502 PMCID: PMC6374537 DOI: 10.14581/jer.18014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/18/2018] [Accepted: 10/01/2018] [Indexed: 12/30/2022] Open
Abstract
Perampanel is a new antiepileptic drug with a long half-life, but there are currently no data on the effects of acute perampanel overdose in epilepsy patients. We report a patient who had such an overdose. A 39-year-old woman was admitted for altered consciousness after taking 10 times the daily dose of perampanel. Her blood level of the drug was predicted, by pharmacokinetic calculation, to decrease to a steady-state level after 7 days, followed by a full recovery 8 days after the overdose. During her hospital stay, the patient experienced pulmonary embolism, which may be associated with the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) antagonistic mechanism of perampanel. The present findings show that perampanel overdose can produce prolonged stupor and that caution should be exercised during transient hypercoagulable states.
Collapse
Affiliation(s)
- Soojeong Kim
- Department of Neurology, Konkuk University School of Medicine, Seoul, Korea
| | - Tae-Eun Kim
- Department of Clinical Pharmacology, Konkuk University School of Medicine, Seoul, Korea
| | - Dayoung Kim
- Department of Neurology, Konkuk University School of Medicine, Seoul, Korea
| | - Dong Wook Kim
- Department of Neurology, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
161
|
Tyrlikova I, Brazdil M, Rektor I, Tyrlik M. Perampanel as monotherapy and adjunctive therapy for focal onset seizures, focal to bilateral tonic-clonic seizures and as adjunctive therapy of generalized onset tonic-clonic seizures. Expert Rev Neurother 2018; 19:5-16. [DOI: 10.1080/14737175.2019.1555474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Ivana Tyrlikova
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
- Brno Epilepsy Center, 1st Department of Neurology, St. Anne’s Univ. Hospital and Medical Faculty of Masaryk University, Brno, Czech Republic
| | - Milan Brazdil
- Brno Epilepsy Center, 1st Department of Neurology, St. Anne’s Univ. Hospital and Medical Faculty of Masaryk University, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Ivan Rektor
- Brno Epilepsy Center, 1st Department of Neurology, St. Anne’s Univ. Hospital and Medical Faculty of Masaryk University, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Michal Tyrlik
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
- University of Maryland, College Park, USA
| |
Collapse
|
162
|
Tringali G, Currò D, Navarra P. Perampanel inhibits calcitonin gene-related peptide release from rat brainstem in vitro. J Headache Pain 2018; 19:107. [PMID: 30419806 PMCID: PMC6755590 DOI: 10.1186/s10194-018-0940-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022] Open
Abstract
Background Perampanel is a novel antiepileptic drug acting via non-competitive antagonism on glutamatergic AMPA receptors, and the subsequent inhibition of ion calcium influx. Since it was recently postulated that the antagonists of glutamate receptors might play a role in the treatment of migraine, in this study we investigated the putative anti-migraine activity of perampanel in an in vitro animal model involving the static incubation of rat brainstem explants and the subsequent measurement of immune-reactive calcitonin gene-related peptide released into the incubation medium. Methods Acute rat brainstem explants were incubated in plain medium or in medium containing graded concentrations of perampanel. The release into the medium was assessed by radioimmunoassay either under baseline conditions or after stimulation by such secretagogues as high K+ concentrations, veratridine or capsaicin. Results We found that: 1) under baseline conditions perampanel, given in the range 0.01–100 μM, inhibited in a concentration-dependent manner calcitonin gene-related peptide’s release compared to controls; the decrease was statistically significant as from 10 μM; 2) a significant and consistent increase in calcitonin gene-related peptide’s secretion was induced by all depolarizing stimuli after 1 h of incubation; 3) under these conditions, calcitonin gene-related peptide’s release stimulated by 56 mM KCl was significantly reduced by perampanel from 0.1 μM onward, whereas secretion stimulated by veratridine was significantly reduced as from 1 μM; 4) on the contrary, perampanel had no effect on capsaicin-induced calcitonin gene-related peptide’s release up to 100 μM. Conclusions Here we provided preliminary in vitro evidence suggesting that perampanel might control pain transmission under conditions of activated trigeminal system, in a preclinical model mimicking the pathophysiology of human migraine.
Collapse
Affiliation(s)
- Giuseppe Tringali
- Institute of Pharmacology, Fondazione Policlinico Universitario A. Gemelli IRCSS, Roma - Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Diego Currò
- Institute of Pharmacology, Fondazione Policlinico Universitario A. Gemelli IRCSS, Roma - Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Pierluigi Navarra
- Institute of Pharmacology, Fondazione Policlinico Universitario A. Gemelli IRCSS, Roma - Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| |
Collapse
|
163
|
Augustin K, Williams S, Cunningham M, Devlin AM, Friedrich M, Jayasekera A, Hussain MA, Holliman D, Mitchell P, Jenkins A, Chen PE, Walker MC, Williams RSB. Perampanel and decanoic acid show synergistic action against AMPA receptors and seizures. Epilepsia 2018; 59:e172-e178. [PMID: 30324610 DOI: 10.1111/epi.14578] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/17/2018] [Accepted: 09/17/2018] [Indexed: 01/27/2023]
Abstract
Perampanel is an adjunctive treatment for epilepsy that works through the direct inhibition of AMPA receptors. The same molecular mechanism has recently been shown for a fatty acid, decanoic acid, prescribed in the medium chain triglyceride ketogenic diet for the treatment of patients with drug-resistant epilepsy. Because each compound has been proposed to act through a distinct AMPA receptor binding site, we predicted that perampanel and decanoic acid would act synergistically against AMPA receptors and, consequently, seizures. Here, we show a synergistic interaction between perampanel and decanoic acid in direct AMPA receptor inhibition, in an ex vivo model of seizure activity, and against seizure-induced activity in human brain slices. These data support a potential role for combination treatment using perampanel and dietary decanoic acid to provide enhanced seizure control.
Collapse
Affiliation(s)
- Katrin Augustin
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Sophie Williams
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK
| | - Mark Cunningham
- Institute of Neuroscience, University of Newcastle upon Tyne, Newcastle, UK
| | - Anita M Devlin
- Department of Paediatric Neurology, Great North Children's Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Maximilian Friedrich
- Department of Neurosurgery, Royal Victoria Infirmary, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Ashan Jayasekera
- Department of Neurosurgery, Royal Victoria Infirmary, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Mohammed A Hussain
- Department of Neurosurgery, Royal Victoria Infirmary, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Damian Holliman
- Department of Neurosurgery, Royal Victoria Infirmary, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Patrick Mitchell
- Department of Neurosurgery, Royal Victoria Infirmary, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Alistair Jenkins
- Department of Neurosurgery, Royal Victoria Infirmary, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Philip E Chen
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK
| | - Robin S B Williams
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| |
Collapse
|
164
|
Pharmacokinetic Considerations with the Use of Antiepileptic Drugs in Patients with HIV and Organ Transplants. Curr Neurol Neurosci Rep 2018; 18:89. [PMID: 30302572 DOI: 10.1007/s11910-018-0897-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW Antiepileptic drugs are frequently administered to patients with HIV infection or in recipients of organ transplants. The potentially serious drug-drug interactions between the "classic" antiepileptic drugs, antiretrovirals, and immunosuppressants have been extensively studied. Evidence-based information on the second and third generation of antiepileptic drugs is almost non-existent. The purpose of this review is to analyze the pharmacokinetic profile of these newer agents to assess their potential for drug interactions with antiretrovirals and immunosuppressants. RECENT FINDINGS As a group, the newer generations of antiepileptic drugs have shown a more favorable drug interaction potential compared to the "classic" ones. A group of moderate enzyme-inducing drugs includes eslicarbazepine acetate, oxcarbazepine, rufinamide, and topiramate. These drugs are not as potent inducers as the "classic" drugs but may potentially decrease the serum concentrations of some antiretrovirals and immunosuppressants. Antiepileptic drugs with no or minimal enzyme-inducing properties include brivaracetam, gabapentin, lacosamide, lamotrigine, levetiracetam, perampanel, pregabalin, and vigabatrin. The newer generations of antiepileptic drugs have expanded the therapeutic options in patients with HIV infection or organ transplants.
Collapse
|
165
|
Perampanel chronic treatment does not induce tolerance and decreases tolerance to clobazam in genetically epilepsy prone rats. Epilepsy Res 2018; 146:94-102. [DOI: 10.1016/j.eplepsyres.2018.07.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 02/08/2018] [Accepted: 07/24/2018] [Indexed: 11/24/2022]
|
166
|
Moavero R, Pisani LR, Pisani F, Curatolo P. Safety and tolerability profile of new antiepileptic drug treatment in children with epilepsy. Expert Opin Drug Saf 2018; 17:1015-1028. [PMID: 30169997 DOI: 10.1080/14740338.2018.1518427] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Treatment of pediatric epilepsy requires a careful evaluation of the safety and tolerability profile of antiepileptic drugs (AEDs) to avoid or minimize as much as possible adverse events (AEs) on various organs, hematological parameters, and growth, pubertal, motor, cognitive and behavioral development. AREAS COVERED Treatment-emergent AEs (TEAEs) reported in the literature 2000-2018 regarding second- and third-generation AEDs used in the pediatric age, with exclusion of the neonatal period that exhibits specific peculiarities, have been described on the basis of their frequency, severity/tolerability, and particular association with a given AED. EXPERT OPINION Somnolence/sedation and behavioral changes, like irritability and nervousness, are among the most commonly observed TEAEs associated with almost all AEDs. Lamotrigine, Gabapentin, Oxcarbazepine, and Levetiracetam appear to be the best-tolerated AEDs with a ≤2% withdrawal rate, while Tiagabine and Everolimus are discontinued in up to >20% of the patients because of intolerable TEAEs. For some AEDs, literature data are scanty to draw a high-level evidence on their safety and tolerability profile. The reasons are: insufficient population size, short duration of treatments, or lack of controlled trials. A future goal is that of identifying clearer, easier, and more homogeneous methodological strategies to facilitate AED testing in pediatric populations.
Collapse
Affiliation(s)
- Romina Moavero
- a Child Neurology and Psychiatry Unit, Systems Medicine Department , Tor Vergata University of Rome , Rome , Italy.,b Child Neurology Unit, Neuroscience and Neurorehabilitation Department , "Bambino Gesù", Children's Hospital, IRCCS , Rome , Italy
| | | | - Francesco Pisani
- d Department of Clinical and Experimental Medicine , University of Messina , Messina , Italy
| | - Paolo Curatolo
- a Child Neurology and Psychiatry Unit, Systems Medicine Department , Tor Vergata University of Rome , Rome , Italy
| |
Collapse
|
167
|
Gil-López FJ, Montoya J, Falip M, Aparicio J, López-González FJ, Toledano R, Gil-Nagel A, Molins A, García I, Serrano P, Domenech G, Torres F, Donaire A, Carreño M. Retrospective study of perampanel efficacy and tolerability in myoclonic seizures. Acta Neurol Scand 2018; 138:122-129. [PMID: 29573400 DOI: 10.1111/ane.12931] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Perampanel is an antiepileptic drug (AED) approved for add-on treatment of focal seizures (with or without generalization) and primary generalized tonic-clonic (GTC) seizures. Our objective was to explore the effectiveness and tolerability of adjunctive perampanel in patients with drug-resistant myoclonic seizures, after failure of other AEDs. MATERIALS AND METHODS Retrospective, multicenter, observational study. Data were collected from individual patient clinical files and analysed using appropriate descriptive statistics and inferential analyses. RESULTS Data are reported for 31 patients with mean age 36.4 years, who had an average epilepsy duration of 18 years, previously taken an average of 5.03 AEDs, and were taking an average of 2.4 AEDs on perampanel initiation. Patients exhibited myoclonic, GTC, absence, tonic and focal seizures, and most had associated cognitive decline and/or ataxia. Median time on perampanel was 6 months, most common dose was 6 mg, and overall retention rate was 84%. The responder rate for myoclonic seizures was defined via reduction of days with myoclonic seizures per month. At 6 months, 15 (48.4%) of the 31 patients were classed as myoclonic seizure responders, 10 (32.3%) were myoclonic seizure free, and 39% saw improvements in functional ability. Of 17 patients with GTC seizures at baseline, 9 (53%) were responders at 6 months, and 8 (47.1%) were seizure free. The most frequent side effects were psychiatric disorders, instability, dizziness and irritability, and mostly resolved with dose reduction. Five patients discontinued perampanel due to side effects. CONCLUSIONS Perampanel caused clinically meaningful improvements in patients with drug-resistant myoclonic seizures. It was generally well tolerated, but psychiatric and neurological side effects sometimes required follow-up and dose reduction.
Collapse
Affiliation(s)
- F. J. Gil-López
- Epilepsy Unit; Department of Neurology; Hospital Clínic de Barcelona; Barcelona Spain
| | - J. Montoya
- Department of Neurology; Hospital Lluis Alcanyís; Xàtiva Spain
| | - M. Falip
- Epilepsy Unit; Department of Neurology; Hospital Universitari de Bellvitge; Hospitalet de Llobregat; Barcelona Spain
| | - J. Aparicio
- Epilepsy Unit; Department of Neurology; Hospital Sant Joan de Déu; Barcelona Spain
| | - F. J. López-González
- Epilepsy Unit; Department of Neurology; Complexo Hospitalario Universitario de Santiago de Compostela; A Coruña Spain
| | - R. Toledano
- Epilepsy Unit; Department of Neurology; Hospital Ruber Internacional; Madrid Spain
| | - A. Gil-Nagel
- Epilepsy Unit; Department of Neurology; Hospital Ruber Internacional; Madrid Spain
| | - A. Molins
- Department of Neurology; Hospital Josep Trueta de Girona; Girona Spain
| | - I. García
- Epilepsy Unit; Hospital Clínico Universitario San Carlos; Madrid Spain
| | - P. Serrano
- Department of Neurology; Hospital Carlos Haya; Málaga Spain
| | - G. Domenech
- Medical Statistics Core Facility; IDIBAPS-Hospital Clínic; Barcelona Spain
| | - F. Torres
- Medical Statistics Core Facility; IDIBAPS-Hospital Clínic; Barcelona Spain
| | - A. Donaire
- Epilepsy Unit; Department of Neurology; Hospital Clínic de Barcelona; Barcelona Spain
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS); Barcelona Spain
| | - M. Carreño
- Epilepsy Unit; Department of Neurology; Hospital Clínic de Barcelona; Barcelona Spain
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS); Barcelona Spain
| |
Collapse
|
168
|
Villanueva V, Montoya J, Castillo A, Mauri-Llerda JÁ, Giner P, López-González FJ, Piera A, Villanueva-Hernández P, Bertol V, Garcia-Escrivá A, Garcia-Peñas JJ, Garamendi I, Esteve-Belloch P, Baiges-Octavio JJ, Miró J, Falip M, Garcés M, Gómez A, Gil-López FJ, Carreño M, Rodriguez-Uranga JJ, Campos D, Bonet M, Querol R, Molins A, Tortosa D, Salas-Puig J. Perampanel in routine clinical use in idiopathic generalized epilepsy: The 12-month GENERAL study. Epilepsia 2018; 59:1740-1752. [PMID: 30062784 DOI: 10.1111/epi.14522] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To analyze the effectiveness and tolerability of perampanel across different seizure types in routine clinical care of patients with idiopathic generalized epilepsy (IGE). METHODS This multicenter, retrospective, 1-year observational study collected data from patient records at 21 specialist epilepsy units in Spain. All patients who were aged ≥12 years, prescribed perampanel before December 2016, and had a confirmed diagnosis of IGE were included. RESULTS The population comprised 149 patients with IGE (60 with juvenile myoclonic epilepsy, 51 generalized tonic-clonic seizures [GTCS] only, 21 juvenile absence epilepsy, 10 childhood absence epilepsy, 6 adulthood absence epilepsy, and one Jeavons syndrome). Mean age was 36 years. The retention rate at 12 months was 83% (124/149), and 4 mg was the most common dose. At 12 months, the seizure-free rate was 59% for all seizures (88/149); 63% for GTCS (72/115), 65% for myoclonic seizures (31/48), and 51% for absence seizures (24/47). Seizure frequency was reduced significantly at 12 months relative to baseline for GTCS (78%), myoclonic (65%), and absence seizures (48%). Increase from baseline seizure frequency was seen in 5.2% of patients with GTCS seizures, 6.3% with myoclonic, and 4.3% with absence seizures. Perampanel was effective regardless of epilepsy syndrome, concomitant antiepileptic drugs (AEDs), and prior AEDs, but retention and seizure freedom were significantly higher when used as early add-on (after ≤2 prior AEDs) than late (≥3 prior AEDs). Adverse events were reported in 50% of patients over 12 months, mostly mild or moderate, and irritability (23%), somnolence (15%), and dizziness (14%) were most frequent. SIGNIFICANCE In routine clinical care of patients with IGE, perampanel improved seizure outcomes for GTCS, myoclonic seizures, and absence seizures, with few discontinuations due to adverse events. This is the first real-world evidence with perampanel across different seizure types in IGE.
Collapse
Affiliation(s)
| | - Javier Montoya
- Lluis Alcanyis Hospital, Xátiva, Spain.,General University Hospital Valencia, Valencia, Spain
| | | | | | - Pau Giner
- University Hospital Dr Peset, Valencia, Spain
| | | | - Anna Piera
- University Hospital Clinic Valencia, Valencia, Spain
| | | | | | | | | | | | | | | | - Júlia Miró
- University Hospital Bellvitge, Barcelona, Spain
| | - Mercè Falip
- University Hospital Bellvitge, Barcelona, Spain
| | | | - Asier Gómez
- University Hospital and Polytechnic La Fe, Valencia, Spain
| | | | | | | | | | | | - Rosa Querol
- University Complex Infanta Cristina, Badajoz, Spain
| | | | | | | |
Collapse
|
169
|
Umanah GKE, Pignatelli M, Yin X, Chen R, Crawford J, Neifert S, Scarffe L, Behensky AA, Guiberson N, Chang M, Ma E, Kim JW, Castro CC, Mao X, Chen L, Andrabi SA, Pletnikov MV, Pulver AE, Avramopoulos D, Bonci A, Valle D, Dawson TM, Dawson VL. Thorase variants are associated with defects in glutamatergic neurotransmission that can be rescued by Perampanel. Sci Transl Med 2018; 9:9/420/eaah4985. [PMID: 29237760 DOI: 10.1126/scitranslmed.aah4985] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 01/20/2017] [Accepted: 06/01/2017] [Indexed: 11/02/2022]
Abstract
The AAA+ adenosine triphosphatase (ATPase) Thorase plays a critical role in controlling synaptic plasticity by regulating the expression of surface α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs). Bidirectional sequencing of exons of ATAD1, the gene encoding Thorase, in a cohort of patients with schizophrenia and healthy controls revealed rare Thorase variants. These variants caused defects in glutamatergic signaling by impairing AMPAR internalization and recycling in mouse primary cortical neurons. This contributed to increased surface expression of the AMPAR subunit GluA2 and enhanced synaptic transmission. Heterozygous Thorase-deficient mice engineered to express these Thorase variants showed altered synaptic transmission and several behavioral deficits compared to heterozygous Thorase-deficient mice expressing wild-type Thorase. These behavioral impairments were rescued by the competitive AMPAR antagonist Perampanel, a U.S. Food and Drug Administration-approved drug. These findings suggest that Perampanel may be useful for treating disorders involving compromised AMPAR-mediated glutamatergic neurotransmission.
Collapse
Affiliation(s)
- George K E Umanah
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Marco Pignatelli
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, USA
| | - Xiling Yin
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rong Chen
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joshua Crawford
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stewart Neifert
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Leslie Scarffe
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Adam A Behensky
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Noah Guiberson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Melissa Chang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Erica Ma
- School of Public Health, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jin Wan Kim
- Department of Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Cibele C Castro
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Biochemistry, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Xiaobo Mao
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Li Chen
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shaida A Andrabi
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ann E Pulver
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dimitrios Avramopoulos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Antonello Bonci
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David Valle
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
170
|
Mohammad H, Sekar S, Wei Z, Moien-Afshari F, Taghibiglou C. Perampanel but Not Amantadine Prevents Behavioral Alterations and Epileptogenesis in Pilocarpine Rat Model of Status Epilepticus. Mol Neurobiol 2018; 56:2508-2523. [PMID: 30039334 DOI: 10.1007/s12035-018-1230-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/08/2018] [Indexed: 12/26/2022]
Abstract
Pilocarpine-induced status epilepticus (SE), which results in the development of spontaneous recurrent seizures (SRSs) activates glutamatergic receptors that contribute to seizure sustenance and neuronal cell death. In the current study, we evaluate whether the exposure to perampanel, an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor blocker, or amantadine, a N-methyl-D-aspartic acid (NMDA) receptor blocker would reduce the SE-induced long-term consequences. SE was induced in adult male Sprague Dawley rats with pilocarpine. Perampanel or amantadine was injected 10 or 60 min after SE onset. The efficacy of either, in overcoming pilocarpine-induced SE was assessed using electroencephalogram (EEG) recordings. In addition, alterations in cognitive function, development of spontaneous recurrent seizures (SRSs), and hippocampal damage that are generally encountered after SE were also assessed at 72 h and 5 weeks after the induction of SE. Our results indicate that both early and late treatment with perampanel but not amantadine significantly reduced seizure activity. Furthermore, perampanel but not amantadine, reversed the memory deficits in Y-maze and novel object recognition (NOR) tests and retarded the appearance of SRSs. Moreover, perampanel treatment led to reduced SE-induced caspase-3 activation in the hippocampal lysates. Taken together, the data obtained from the study reveals that blocking AMPA receptors by perampanel can modify SE-induced long-term consequences. Our results may provide a proof of principle for the potential therapeutic application of perampanel in clinical use for status epilepticus in future.
Collapse
Affiliation(s)
- Hanan Mohammad
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Sathiya Sekar
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Zelan Wei
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Farzad Moien-Afshari
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada.
- Department of Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
- Division of Neurology, Department of Medicine, University of British Colombia, 8247-2775 Laurel St, Vancouver, BC, V5Z 1M9, Canada.
| | - Changiz Taghibiglou
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
171
|
Campos G, Fortuna A, Falcão A, Alves G. In vitro and in vivo experimental models employed in the discovery and development of antiepileptic drugs for pharmacoresistant epilepsy. Epilepsy Res 2018; 146:63-86. [PMID: 30086482 DOI: 10.1016/j.eplepsyres.2018.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/16/2018] [Accepted: 07/20/2018] [Indexed: 12/23/2022]
Abstract
Epilepsy is one of the most common chronic, recurrent and progressive neurological diseases. In spite of the large number of antiepileptic drugs currently available for the suppression of seizures, about one-third of patients develop drug-resistant epilepsy, even when they are administered the most appropriate treatment available. Thus, nonclinical models can be valuable tools for the elucidation of the mechanisms underlying the development of pharmacoresistance and also for the development of new therapeutic agents that may be promising therapeutic approaches for this unmet medical need. Up today, several epilepsy and seizure models have been developed, exhibiting similar physiopathological features of human drug-resistant epilepsy; moreover, pharmacological response to antiepileptic drugs clinically available tends to be similar in animal models and humans. Therefore, they should be more intensively used in the preclinical discovery and development of new candidates to antiepileptic drugs. Although useful, in vitro models cannot completely replicate the complexity of a living being and their potential for a systematic use in antiepileptic drug screening is limited. The whole-animal models are the most commonly employed and they can be classified as per se drug-resistant due to an inherent poor drug response or be based on the selection of subgroups of epileptic animals that respond or not to a specific antiepileptic drug. Although more expensive and time-consuming, the latter are chronic models of epilepsy that better exhibit the disease-associated alterations found in human epilepsy. Several antiepileptic drugs in development or already marketed have been already tested and shown to be effective in these models of drug-resistant epilepsy, constituting a new hope for the treatment of drug-resistant epilepsy. This review will provide epilepsy researchers with detailed information on the in vitro and in vivo nonclinical models of interest in drug-resistant epilepsy, which may enable a refined selection of most relevant models for understanding the mechanisms of the disease and developing novel antiepileptic drugs.
Collapse
Affiliation(s)
- Gonçalo Campos
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Ana Fortuna
- CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Portugal; Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Amílcar Falcão
- CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Portugal; Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Gilberto Alves
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal.
| |
Collapse
|
172
|
Sada N, Inoue T. Electrical Control in Neurons by the Ketogenic Diet. Front Cell Neurosci 2018; 12:208. [PMID: 30061816 PMCID: PMC6054928 DOI: 10.3389/fncel.2018.00208] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/26/2018] [Indexed: 01/01/2023] Open
Abstract
The ketogenic diet is used as a diet treatment for drug-resistant epilepsy, but there are no antiepileptic drugs based on the ketogenic diet. The ketogenic diet changes energy metabolites (ketone bodies, glucose and lactate) in the brain, which consequently changes electrical activities in neurons and ultimately suppresses seizures in epileptic patients. In order to elucidate the antiseizure effects of the ketogenic diet, it is important to clarify the mechanism by which these metabolic changes are converted to electrical changes in neurons. In this review, we summarize electrophysiological studies focusing on electrical control in neurons by the ketogenic diet. Recent studies have identified electrical regulators driven by the ketogenic diet: ion channels (ATP-sensitive K+ channels and voltage-dependent Ca2+ channels), synaptic receptors (AMPA-type glutamate receptors and adenosine A1 receptors), neurotransmitter transporters (vesicular glutamate transporters), and others (BCL-2-associated agonist of cell death and lactate dehydrogenase). Thus, the ketogenic diet presumably elicits neuronal inhibition via the combined actions of these molecules. From the viewpoint of drug development, these molecules are valuable as targets for the development of new antiepileptic drugs. Drug therapy to mimic the ketogenic diet may be feasible in the future, through the combination of multiple antiepileptic drugs targeting these molecules.
Collapse
Affiliation(s)
- Nagisa Sada
- Department of Biophysical Chemistry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Hygiene, Kawasaki Medical School, Kurashiki, Japan
| | - Tsuyoshi Inoue
- Department of Biophysical Chemistry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
173
|
Nakajima M, Suda S, Sowa K, Sakamoto Y, Nito C, Nishiyama Y, Aoki J, Ueda M, Yokobori S, Yamada M, Yokota H, Okada T, Kimura K. AMPA Receptor Antagonist Perampanel Ameliorates Post-Stroke Functional and Cognitive Impairments. Neuroscience 2018; 386:256-264. [PMID: 29981363 DOI: 10.1016/j.neuroscience.2018.06.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 06/23/2018] [Accepted: 06/26/2018] [Indexed: 12/13/2022]
Abstract
Perampanel (PER), a noncompetitive α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor antagonist, clinically used for seizure control, has been reported to exert neuroprotective effects in experimental models of neurodegenerative diseases. However, few studies have investigated the therapeutic effects of PER in brain injury including stroke. Our aim was to investigate the neuroprotective potential of PER using a rat transient middle cerebral artery occlusion (MCAO) model. Sprague-Dawley rats underwent 90-min MCAO followed by intraperitoneal PER administration at a dose of 1.5 mg/kg. Infarct volumes, neurological deficits, and immunological analyses were performed at 7 days after MCAO. PER significantly reduced infarct volumes (p < 0.05) and improved motor function (p < 0.05) compared with vehicle. Immunological analysis showed that PER significantly inhibited microglial activation, pro-inflammatory cytokine expression, and oxidative stress compared with vehicle. Moreover, PER suppressed neurodegeneration in the cortical ischemic boundary zone, via downregulation of Bcl-2-associated x and upregulation of Bcl-extra-large with Akt activation. In addition, post-stroke secondary neuronal damage and cognitive impairments, using the Y-maze test, were assessed 30 days after MCAO. PER significantly improved spatial working memory, which was accompanied by hippocampal CA1 neuronal loss and cortical thinning, compared with vehicle. These results indicate that PER attenuates infarct volumes and motor function deficits possibly through its anti-inflammatory, antioxidant, and anti-apoptotic activities, mediated via activation of phosphatidylinositol 3-kinase (PI3K)/Akt pathways in the acute ischemic phase, and further ameliorates post-stroke cognitive impairments via the suppression of secondary neuronal damage in the chronic ischemic phase.
Collapse
Affiliation(s)
- Masataka Nakajima
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan; Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Satoshi Suda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Kota Sowa
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan; Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Yuki Sakamoto
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan; Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Chikako Nito
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Yasuhiro Nishiyama
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Junya Aoki
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Masayuki Ueda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Shoji Yokobori
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Marina Yamada
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Hiroyuki Yokota
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Kazumi Kimura
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| |
Collapse
|
174
|
Potschka H, Trinka E. Perampanel: Does it have broad-spectrum potential? Epilepsia 2018; 60 Suppl 1:22-36. [PMID: 29953584 DOI: 10.1111/epi.14456] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2017] [Indexed: 12/26/2022]
Abstract
This article reviews the profile of perampanel, a novel noncompetitive α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptor antagonist, and its role as a potential broad-spectrum antiepileptic drug in the treatment of epilepsy. For this narrative review, data were collected using specified search criteria. Articles reporting the evidence for perampanel's efficacy from preclinical models, phase 3 clinical studies, observational studies, and descriptive evidence were included. AMPA receptors play a key role in mediating the action of glutamate at the excitatory synapse. Preclinical research showed the AMPA receptor blockade to constitute a promising target for antiepileptic drug therapy. In animal models, perampanel proved to be protective against seizures and reduce seizure severity and duration. Four phase-3 randomized controlled trials (3 in patients with focal seizures and one in primary generalized tonic-clonic seizures in idiopathic generalized epilepsy) have been completed. In focal (partial) onset seizures, perampanel (4, 8, and 12 mg) significantly reduced seizure frequency per 28 days (23.3%-28.8% vs 12.8%; P < .01) and responder rates (≥50% reduction in seizures) (28.5%-35.3% vs 19.3%; P < .05) compared with placebo. In primary generalized tonic-clonic seizures, perampanel 8 mg resulted in greater reduction in seizure frequency per 28 days (-76.5% vs -38.4%; P < .0001) and responder rate (64.2% vs 39.5%; P = .0019) than placebo. The efficacy, safety, and tolerability of perampanel have been reproduced in real-world clinical practice, and the agent has been shown to be effective in other epilepsy syndromes. Perampanel is a potentially broad-spectrum antiepileptic drug with a novel mechanism of action that may be a useful addition for patients with epilepsy with various seizure types. The availability of novel antiepileptic drugs for epilepsy treatment enables more individualized treatment for these patients.
Collapse
Affiliation(s)
- Heidrun Potschka
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Klinik, University Hospital Paracelsus Medical University, Salzburg, Austria.,Institute of Public Health, Medical Decision Making and Health Technology Assessment, University for Health Sciences, Medical Informatics and Technology, UMIT, Hall in Tyrol, Austria
| |
Collapse
|
175
|
Tabuchi H, Shiba S, Yasuda S, Ohnishi A, Shin JG. Pharmacokinetics of Perampanel in Healthy Korean, White, and Japanese Adult Subjects. Clin Pharmacol Drug Dev 2018; 7:613-620. [DOI: 10.1002/cpdd.581] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/26/2018] [Indexed: 11/06/2022]
|
176
|
Arzimanoglou A, D'Cruz O, Nordli D, Shinnar S, Holmes GL. A Review of the New Antiepileptic Drugs for Focal-Onset Seizures in Pediatrics: Role of Extrapolation. Paediatr Drugs 2018; 20:249-264. [PMID: 29616471 DOI: 10.1007/s40272-018-0286-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Most antiepileptic drugs (AEDs) receive regulatory approval for children years after the drug is available in adults, encouraging off-label use of the drug in children and hindering attempts to obtain quality pediatric data in controlled trials. Extrapolating adult efficacy data to pediatrics can reduce the time between approval in adults and that in children. To extrapolate efficacy from adults to children, several assumptions must be supported, such as (1) a similar disease progression and response to interventions in adults and children, and (2) similar exposure response in adults and children. The Pediatric Epilepsy Academic Consortium for Extrapolation (PEACE) addressed these assumptions in focal-onset seizures (FOS), the most common seizure type in both adults and children. PEACE reviewed the biological and clinical evidence that supported the assumptions that children with FOS have a similar disease progression and response to intervention as adults with FOS. After age 2 years, the pathophysiological underpinnings of FOS and the biological milieu in which seizures are initiated and propagated in children, seizure semiology, electroencephalographic features, etiology and AED response to FOS in children are similar to those in adults with FOS. PEACE concluded that extrapolation of efficacy data in adults to pediatrics in FOS is supported by strong scientific and clinical evidence. However, safety and pharmacokinetic (PK) data cannot be extrapolated from adults to children. Based on extrapolation, eslicarbazepine is now approved for children with FOS, down to age 4 years. Perampanel, lacosamide and brivaracetam are now undergoing PK and safety studies for the purposes of extrapolation down to age 2 or 4 years. When done in conjunction with PK and safety investigations in children, extrapolation of adult data from adults to children can reduce the time delay between approval of effective and safe AEDs in adults and approval in children.
Collapse
Affiliation(s)
- Alexis Arzimanoglou
- Department of Clinical Epileptology, Sleep Disorders and Functional Pediatric Neurology, University Hospitals of Lyon (HCL), Lyon, France.,Sección Epilepsia, Sueño y Neurofisiología, Servicio Neurología, Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| | - O'Neill D'Cruz
- Consulting and Neurological Services, Chapel Hill, NC, USA
| | - Douglas Nordli
- Division of Pediatric Neurology, Children's Hospital Los Angeles, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Shlomo Shinnar
- Departments of Neurology, Pediatrics and Epidemiology and Population Health, Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY, USA
| | - Gregory L Holmes
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| | | |
Collapse
|
177
|
Tsai JJ, Wu T, Leung H, Desudchit T, Tiamkao S, Lim KS, Dash A. Perampanel, an AMPA receptor antagonist: From clinical research to practice in clinical settings. Acta Neurol Scand 2018; 137:378-391. [PMID: 29214650 DOI: 10.1111/ane.12879] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2017] [Indexed: 12/21/2022]
Abstract
Epileptic seizures are refractory to treatment in approximately one-third of patients despite the recent introduction of many newer antiepileptic drugs (AEDs). Development of novel AEDs therefore remains a high priority. Perampanel is a first-in-class non-competitive selective AMPA receptor antagonist with a unique mechanism of action. Clinical efficacy and safety of perampanel as adjunctive treatment for focal seizures with/without secondary generalization (±SG) and primary generalized tonic-clonic (PGTC) seizures have been established in five phase 3 randomized controlled trials (RCTs), and a long-term extension study, and perampanel is approved as monotherapy for focal seizures ±SG in the USA. In patients with focal seizures ±SG, add-on perampanel resulted in median percent reduction in seizure frequency 23.3%-34.5% and ≥50% responder rate 28.5%-37.6%; in PGTC seizures, these results were 76.5% and 64.2%, respectively. Efficacy among adolescents (reduction in seizure frequency 34.8%-35.6%; ≥50% responder rate 40.9%-45.0%) and elderly people (reduction in seizure frequency 12.5%-16.9%; ≥50% responder rate 22.2%-42.9%) is similar to those in adults, and results remain comparable between Asian (reduction in seizure frequency 17.3%-38.0%) and global populations. Perampanel has been extensively studied in real-world clinical practice, with similar efficacy and safety results to the RCTs (≥50% responder rate 12.8%-75.0%; adverse events of somnolence/sedation, dizziness, ataxia, and behavioral changes). Real-world observational studies suggest that perampanel tolerability can be improved by slow titration (2 mg every 2-4 weeks), and bedtime administration can mitigate somnolence and dizziness. Counseling about the potential for behavioral changes and close monitoring are recommended.
Collapse
Affiliation(s)
- J.-J. Tsai
- Department of Neurology; National Cheng Kung University Hospital and School of Medicine; National Cheng Kung University; Tainan Taiwan
| | - T. Wu
- Department of Neurology; Chang Gung Memorial Hospital; Chang Gung University; Taoyuan City Taiwan
| | - H. Leung
- Department of Medicine and Therapeutics; Faculty of Medicine; Prince of Wales Hospital; Hong Kong Hong Kong
| | - T. Desudchit
- Department of Paediatrics; King Chulalongkorn Memorial Hospital; Bangkok Thailand
| | - S. Tiamkao
- Integrated Epilepsy Research Group; Department of Medicine; Faculty of Medicine; Khon Kaen University; Khon Kaen Thailand
| | - K.-S. Lim
- Division of Neurology; Department of Medicine; Faculty of Medicine; University of Malaya; Kuala Lumpur Malaysia
| | - A. Dash
- Eisai Singapore Pte. Ltd.; Singapore
| |
Collapse
|
178
|
Nishida T, Lee SK, Inoue Y, Saeki K, Ishikawa K, Kaneko S. Adjunctive perampanel in partial-onset seizures: Asia-Pacific, randomized phase III study. Acta Neurol Scand 2018; 137:392-399. [PMID: 29250772 DOI: 10.1111/ane.12883] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2017] [Indexed: 02/01/2023]
Abstract
OBJECTIVES To evaluate the efficacy, safety, and tolerability of perampanel, a selective, non-competitive, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist, as an adjunctive treatment for patients with refractory partial-onset seizures (POS) from Asia-Pacific. MATERIALS & METHODS This multicenter, randomized, double-blind, placebo-controlled trial (ClinicalTrials.gov identifier: NCT01618695) involved patients aged ≥12 years with refractory POS (receiving 1-3 antiepileptic drugs). Patients were randomized (1:1:1:1) to receive once-daily placebo or perampanel 4, 8, or 12 mg over a 6-week titration and 13-week maintenance double-blind period. Enzyme-inducing antiepileptic drugs were equally stratified between groups. The primary efficacy endpoint was percent change in POS frequency per 28 days (double-blind phase vs baseline). Other efficacy endpoints included ≥50% responder rate and seizure freedom. Treatment-emergent adverse events (TEAEs) were also monitored. RESULTS Of 710 randomized patients, seizure frequency data were available for 704 patients. Median percent changes in POS frequency per 28 days indicated dose-proportional reductions in seizure frequency: -10.8% with placebo and -17.3% (P = .2330), -29.0% (P = .0003), and -38.0% (P < .0001) with perampanel 4, 8, and 12 mg, respectively. In total, 108 (15.3%) patients discontinued treatment; 44 (6.2%) due to TEAEs. TEAEs occurring in ≥5% of patients, and reported at least twice as frequently with perampanel vs placebo, included dizziness and irritability. CONCLUSIONS Adjunctive perampanel (8 and 12 mg/d) significantly improved seizure control in patients with refractory POS. Safety and tolerability were acceptable at daily doses of perampanel 4-12 mg.
Collapse
Affiliation(s)
- T. Nishida
- National Epilepsy Center; Shizuoka Institute of Epilepsy and Neurological Disorders; Shizuoka Japan
| | - S. K. Lee
- Department of Neurology; Seoul National University Hospital; Seoul South Korea
| | - Y. Inoue
- National Epilepsy Center; Shizuoka Institute of Epilepsy and Neurological Disorders; Shizuoka Japan
| | | | | | - S. Kaneko
- North Tohoku Epilepsy Center; Minato Hospital; Aomori Japan
| |
Collapse
|
179
|
Niu HX, Wang JZ, Wang DL, Miao JJ, Li H, Liu ZG, Yuan X, Liu W, Zhou JR. The Orally Active Noncompetitive AMPAR Antagonist Perampanel Attenuates Focal Cerebral Ischemia Injury in Rats. Cell Mol Neurobiol 2018; 38:459-466. [PMID: 28401316 PMCID: PMC11481974 DOI: 10.1007/s10571-017-0489-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 03/24/2017] [Indexed: 02/07/2023]
Abstract
Inhibition of ionotropic glutamate receptors (iGluRs) is a potential target of therapy for ischemic stroke. Perampanel is a potent noncompetitive α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor (AMPAR) antagonist with good oral bioavailability and favorable pharmacokinetic properties. Here, we investigated the potential protective effects of perampanel against focal cerebral ischemia in a middle cerebral artery occlusion (MCAO) model in rats. Oral administration with perampanel significantly reduced MCAO-induced brain edema, brain infarct volume, and neuronal apoptosis. These protective effects were associated with improved functional outcomes, as measured by foot-fault test, adhesive removal test, and modified neurological severity score (mNSS) test. Importantly, perampanel was effective even when the administration was delayed to 1 h after reperfusion. The results of enzyme-linked immunosorbent assay (ELISA) showed that perampanel significantly decreased the expression of pro-inflammatory cytokines IL-1β and TNF-α, whereas it increased the levels of anti-inflammatory cytokines IL-10 and TGF-β1 after MCAO. In addition, perampanel treatment markedly decreased the expression of inducible nitric oxide synthase (iNOS) and neuronal nitric oxide synthase (nNOS), and also inhibited nitric oxide (NO) generation in MCAO-injured rats at 24 and 72 h after reperfusion. In conclusion, this study demonstrated that the orally active AMPAR antagonist perampanel protects against experimental ischemic stroke via regulating inflammatory cytokines and NOS pathways.
Collapse
Affiliation(s)
- Hong-Xia Niu
- Emergency Department, Beijing Electric Power Hospital, Beijing, 100071, China
| | - Jun-Zhe Wang
- Basic Medical Sciences Research Center, Shaanxi Fourth People's Hospital, Xi'an, 710043, Shaanxi, China
| | - Dong-Liang Wang
- Department of Neurosurgery, Peking University People's Hospital, Beijing, 100044, China
| | - Jun-Jie Miao
- Department of Neurosurgery, Peking University People's Hospital, Beijing, 100044, China
| | - Hua Li
- Department of Neurosurgery, Shaanxi Fourth People's Hospital, Xi'an, 710043, Shaanxi, China
| | - Zhi-Gang Liu
- Department of Neurosurgery, Shaanxi Fourth People's Hospital, Xi'an, 710043, Shaanxi, China
| | - Xing Yuan
- Department of Neurosurgery, Shaanxi Fourth People's Hospital, Xi'an, 710043, Shaanxi, China
| | - Wei Liu
- Basic Medical Sciences Research Center, Shaanxi Fourth People's Hospital, Xi'an, 710043, Shaanxi, China.
- Department of Neurosurgery, Shaanxi Fourth People's Hospital, Xi'an, 710043, Shaanxi, China.
| | - Jing-Ru Zhou
- Department of Neurosurgery, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
180
|
Chiang HI, Lim SN, Hsieh HY, Cheng MY, Chang CW, Johnny Tseng WE, Li HT, Lin CY, Wu T. Preliminary Asian experience of using perampanel in clinical practice. Biomed J 2018; 40:347-354. [PMID: 29433838 PMCID: PMC6138609 DOI: 10.1016/j.bj.2017.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/21/2017] [Accepted: 09/18/2017] [Indexed: 11/15/2022] Open
Abstract
Background To analyze the efficacy and safety of perampanel over a 3-month period in a sample of Asian people with epilepsy. Methods The efficacy and safety of perampanel as an adjunctive therapy for patients with epilepsy were retrospectively reviewed and analyzed. Patients were categorized according to seizure type, concomitant antiepileptic drug usage, and perampanel dosage. Results A total of 210 patients were included in the study and 131 patients completed 3 months of perampanel treatment. The average dosage of perampanel was 5.31 mg/day, and the 50% responder rate (≥50% seizure frequency reduction) in all patients was 45.8%, with a 27.5% seizure-free rate. For focal seizures, focal to bilateral tonic-clonic seizures, and primary generalized seizures, the 50% responder rates were respectively 29.4%, 49.5%, and 36.4%. In total, 39.5% of patients experienced adverse events within 3 months of observation period, and the rate of drug withdrawal due to adverse events was 8.6%. Dizziness, ataxia, irritability/aggression were the most common adverse events. Conclusions The efficacy and safety of perampanel in a real-world setting with Asian patients is comparable to that in clinical trials that have included fewer Asian patients.
Collapse
Affiliation(s)
- Hsing-I Chiang
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Siew-Na Lim
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Hsiang-Yao Hsieh
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Mei-Yun Cheng
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan; Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Chun-Wei Chang
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Wei-En Johnny Tseng
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Han-Tao Li
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chin-Yin Lin
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Tony Wu
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan.
| |
Collapse
|
181
|
Adotevi NK, Leitch B. Synaptic Changes in AMPA Receptor Subunit Expression in Cortical Parvalbumin Interneurons in the Stargazer Model of Absence Epilepsy. Front Mol Neurosci 2017; 10:434. [PMID: 29311821 PMCID: PMC5744073 DOI: 10.3389/fnmol.2017.00434] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/15/2017] [Indexed: 11/15/2022] Open
Abstract
Feedforward inhibition is essential to prevent run away excitation within the brain. Recent evidence suggests that a loss of feed-forward inhibition in the corticothalamocortical circuitry may underlie some absence seizures. However, it is unclear if this aberration is specifically linked to loss of synaptic excitation onto local fast-spiking parvalbumin-containing (PV+) inhibitory interneurons, which are responsible for mediating feedforward inhibition within cortical networks. We recently reported a global tissue loss of AMPA receptors (AMPARs), and a specific mistrafficking of these AMPARs in PV+ interneurons in the stargazer somatosensory cortex. The current study was aimed at investigating if cellular changes in AMPAR expression were translated into deficits in receptors at specific synapses in the feedforward inhibitory microcircuit. Using western blot immunolabeling on biochemically isolated synaptic fractions, we demonstrate a loss of AMPAR GluA1–4 subunits in the somatosensory cortex of stargazers compared to non-epileptic control mice. Furthermore, using double post-embedding immunogold-cytochemistry, we show a loss of GluA1–4-AMPARs at excitatory synapses onto cortical PV+ interneurons. Altogether, these data indicate a loss of synaptic AMPAR-mediated excitation of cortical PV+ inhibitory neurons. As the cortex is considered the site of initiation of spike wave discharges (SWDs) within the corticothalamocortical circuitry, loss of AMPARs at cortical PV+ interneurons likely impairs feed-forward inhibitory output, and contributes to the generation of SWDs and absence seizures in stargazers.
Collapse
Affiliation(s)
- Nadia K Adotevi
- Department of Anatomy, Brain Health Research Centre, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Beulah Leitch
- Department of Anatomy, Brain Health Research Centre, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
182
|
Rider FK. [Perampanel in the treatment of patients with epilepsy]. Zh Nevrol Psikhiatr Im S S Korsakova 2017; 117:63-69. [PMID: 29213041 DOI: 10.17116/jnevro20171179263-69] [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: 11/17/2022]
Abstract
Development of new antiepileptic drugs (AED) does not stop due to the fact that the number of patients with pharmacoresistant epilepsy remains at about 30%. One of the newest AEDs is perampanel (PER), a selective, non-competitive AMPA receptor antagonist to target post-synaptic glutamate transmission. PER is approved in the Russian Federation as adjunctive treatment for focal seizures with or without secondarily generalized seizures and for primary generalized tonic-clonic (PGTC) seizures in idiopathic generalized epilepsy (IGE) in patients with epilepsy aged ≥12 years. The drug is effective and well-tolerated in the dose of 4-8 mg/day, and most side effects are dose-dependent. The high efficacy of PER combined with the good tolerability, absence of life-threatening adverse reactions and convenient intake allow us to recommend PER as the first choice additional drug in treatment of patients with epilepsy.
Collapse
Affiliation(s)
- F K Rider
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| |
Collapse
|
183
|
Paul D, Allakonda L, Sahu A, Surendran S, Satheeshkumar N. Pharmacokinetics and brain uptake study of novel AMPA receptor antagonist perampanel in SD rats using a validated UHPLC-QTOF-MS method. J Pharm Biomed Anal 2017; 149:234-241. [PMID: 29127904 DOI: 10.1016/j.jpba.2017.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/09/2017] [Accepted: 11/01/2017] [Indexed: 02/06/2023]
Abstract
Perampanel (PER) is a novel AMPA receptor antagonist for antiepileptic therapy and is prospective for the treatment of other neurological disorders. A highly sensitive and rapid UHPLC-QTOF-MS method was developed for the quantification of PER in plasma/brain homogenate of SD rat with alogliptin as an internal standard (IS). Chromatographic separation was carried out on an Acquity UPLC HSS Cyano column (100mm×2.1mm, 1.8μm) using gradient mobile phase consisting of 0.1% formic acid and acetonitrile at a flow rate of 0. 4mL/min. Sample preparation was carried out by a simple protein precipitation method. The mass spectrometric analysis of target ions at [M+H]+m/z 350.1288 for PER and m/z 340.1779 for IS was monitored with extracted ion chromatography. The developed analytical method meets the US-FDA and EMA bioanalytical guidelines and was found to be precise, accurate, selective and rugged. It exhibited good sensitivity (0.4ng/mL) and linearity over a range of 0.4-400ng/mL in both the bio-matrices. The method was successfully applied to pharmacokinetics and brain uptake study of PER after oral administration to SD rats. The study results showed PER has penetrated the blood-brain barrier, brain to plasma ratio (Kp) was found to be 0.62±0.05 and its rapidly eliminated from the brain.
Collapse
Affiliation(s)
- David Paul
- Drug Metabolism and Interactions Research Lab, Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Lingesh Allakonda
- Drug Metabolism and Interactions Research Lab, Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Amit Sahu
- Drug Metabolism and Interactions Research Lab, Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Shruti Surendran
- Drug Metabolism and Interactions Research Lab, Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Nanjappan Satheeshkumar
- Drug Metabolism and Interactions Research Lab, Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| |
Collapse
|
184
|
Ulate-Campos A, Fernández IS. Cognitive and Behavioral Comorbidities: An Unwanted Effect of Antiepileptic Drugs in Children. Semin Pediatr Neurol 2017; 24:320-330. [PMID: 29249512 DOI: 10.1016/j.spen.2017.10.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Epilepsy is one of the most common neurological disorders and, despite optimally chosen and dosed antiepileptic drugs (AEDs), approximately 20%-30% of patients will continue to have seizures. Behavior and cognition are negatively impacted by seizures, but AEDs are also a major contributor to behavioral and cognitive deficits. However, the cognitive and behavioral effect of AEDs in children is insufficiently emphasized in the literature. This review summarizes the cognitive and behavioral effects of AEDs in the pediatric population with the objective of helping pediatricians and pediatric neurologists to select the AEDs with the best profile for their individual patient's needs.
Collapse
Affiliation(s)
- Adriana Ulate-Campos
- Department of Neurology, National Children's Hospital "Dr. Carlos Saenz Herrera", San José, CR.
| | - Ivan Sánchez Fernández
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA; Department of Child Neurology, Hospital Sant Joan de Déu, Universidad de Barcelona, Barcelona, Spain
| |
Collapse
|
185
|
Gil-Nagel A, Burd S, Toledo M, Sander JW, Lebedeva A, Patten A, Laurenza A. A retrospective, multicentre study of perampanel given as monotherapy in routine clinical care in people with epilepsy. Seizure 2017; 54:61-66. [PMID: 29288911 DOI: 10.1016/j.seizure.2017.10.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/03/2017] [Accepted: 10/17/2017] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Perampanel is approved for adjunctive treatment of focal seizures, with or without secondarily generalised seizures, and for primary generalised tonic-clonic seizures in people with epilepsy aged ≥12 years. Perampanel was recently approved for monotherapy use for partial seizures in the United States. This study provides insight into the feasibility of perampanel monotherapy in real-world settings. METHODS This retrospective, non-interventional, multicentre study (NCT02736162) was conducted between January 2013 and March 2016 in specialist epilepsy centres in Europe and Russia. Eligible individuals had a diagnosis of epilepsy and received perampanel primary or secondary monotherapy as routine clinical care. The primary endpoint was proportion of individuals remaining on perampanel monotherapy, after conversion from perampanel adjunctive treatment, at 3, 6, 12, 18 and 24 months (retention rate). RESULTS Sixty individuals were in the safety set (female, 63%; white, 97%; aged 18 to <65 years, 73%). Most (85%) received secondary monotherapy with perampanel. At study cut-off, 68% of individuals were continuing on perampanel monotherapy (secondary monotherapy: 55%). The median duration of retention was not calculable due to the high number of individuals ongoing on monotherapy. Twelve individuals had treatment-emergent adverse events that started during perampanel monotherapy, the most frequent was dizziness (5%). One serious treatment-emergent adverse event was reported (pneumonia during adjunctive perampanel treatment). CONCLUSIONS In this small retrospective study of individuals who received perampanel monotherapy, the majority maintained monotherapy. Perampanel monotherapy may be an achievable option in some people with epilepsy.
Collapse
Affiliation(s)
- Antonio Gil-Nagel
- Hospital Ruber Internacional, Calle de la Masó 38, 28034 Madrid, Spain.
| | - Sergey Burd
- Pirogov Russian National Research Medical University, Ostrovitianov Str. 1, Moscow, 117997, Russia.
| | - Manuel Toledo
- Hospital Universitario Vall d'Hebron, Passeig de la Vall d'Hebron 119-129, 08035 Barcelona, Spain.
| | - Josemir W Sander
- UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK; Chalfont Centre for Epilepsy, Chalfont St Peter, Gerrards Cross, SL9 0RJ, UK; Stichting Epilepsie Instellingen Nederland (SEIN), Achterweg 5, Heemstede, 2103 SW, The Netherlands.
| | - Anna Lebedeva
- Pirogov Russian National Research Medical University, Ostrovitianov Str. 1, Moscow, 117997, Russia.
| | - Anna Patten
- Eisai Ltd., European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire, AL10 9SN, UK.
| | | | | |
Collapse
|
186
|
Di Bonaventura C, Labate A, Maschio M, Meletti S, Russo E. AMPA receptors and perampanel behind selected epilepsies: current evidence and future perspectives. Expert Opin Pharmacother 2017; 18:1751-1764. [PMID: 29023170 DOI: 10.1080/14656566.2017.1392509] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION The alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors are the major mediators of glutamate-mediated excitatory neurotransmission, and are critical for synchronization and spread of epileptic activity. Areas covered: AMPA receptor antagonists have been also developed as antiepileptic drugs and perampanel (PER) is the first highly selective, non-competitive AMPA-type glutamate receptor antagonist that is available on the market. It is approved as adjunctive therapy for the treatment of partial-onset seizures with or without secondary generalization, and for primary generalized tonic-clonic seizures in idiopathic generalized epilepsy, in patients aged ≥ 12 years. This article reviews the role of AMPA receptors in the neuronal hyperexcitability underlying epilepsy, the mechanism of action and clinical experience on the anti-seizure activity of PER. Moreover, the rationale for targeting AMPA receptor in specific epileptic disorders, including brain tumor-related epilepsy, mesial temporal lobe epilepsy with/without hippocampal sclerosis, and status epilepticus is evaluated. Finally, the pharmacological rationale for the development of AMPA receptor antagonists in other neurological disorders beyond epilepsy is considered. Expert opinion: Further research aimed at better understanding the pharmacology and blocking mechanism of PER and other AMPA receptor antagonists will drive future development of therapeutic agents that target epilepsy and other neurological diseases.
Collapse
Affiliation(s)
- Carlo Di Bonaventura
- a Department of Neurology and Psychiatry, Neurology Unit , 'Sapienza' University , Rome , Italy
| | - Angelo Labate
- b Institute of Neurology , University Magna Graecia of Catanzaro , Catanzaro , Italy.,c Institute of Molecular Bioimaging and Physiology of the National Research Council , Catanzaro , Italy
| | - Marta Maschio
- d Center for Tumor-related Epilepsy, UOSD Neurology , Regina Elena National Cancer Institute , Rome , Italy
| | - Stefano Meletti
- e Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology , University of Modena and Reggio Emilia , Modena , Italy
| | - Emilio Russo
- f Department of Science of Health, School of Medicine and Surgery , University 'Magna Graecia' of Catanzaro , Catanzaro , Italy
| |
Collapse
|
187
|
Khangura RK, Bali A, Kaur G, Singh N, Jaggi AS. Neuropathic pain attenuating effects of perampanel in an experimental model of chronic constriction injury in rats. Biomed Pharmacother 2017; 94:557-563. [DOI: 10.1016/j.biopha.2017.07.137] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 07/20/2017] [Accepted: 07/26/2017] [Indexed: 01/31/2023] Open
|
188
|
Effectiveness and tolerability of Perampanel in children, adolescents and young adults with refractory epilepsy: A UK national multicentre study. Seizure 2017; 52:63-70. [PMID: 28992560 DOI: 10.1016/j.seizure.2017.08.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/18/2017] [Accepted: 08/24/2017] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Perampanel is one of the latest antiepileptic drugs (AEDs) approved for the treatment of focal and generalised epilepsy in individuals with epilepsy aged 12 years and older. There is sparse data on the use of Perampanel in children under 12. We conducted a study amongst paediatric neurologists in the United Kingdom to investigate its effectiveness and tolerability as an adjunctive therapy in children of all ages with refractory epilepsy. METHODS Data was collected via an online questionnaire sent to paediatric neurologists in the UK. Data gathered, prospective in 62 (64.5%) and retrospective in 34 (35.5%) patients, included changes in seizure frequency from baseline and unwanted effects at 3, 6 and 12 months follow-up. Only patients with a minimum follow-up of six months were included. RESULTS Ninety six patients (48 females) with refractory epilepsy from 11 of 29 tertiary centres were included. Median [IQR] (range) age was 14 years 11 months [12 years, 16 years 6 months] (11 months-24 years 5 months). Seventy three (76%) had focal epilepsy, sixteen (17%) generalised, and seven (7%) patients both generalised and focal epilepsy. The responder rate, ≥50% seizure reduction from baseline, was 19% for all seizure types at both 6 and 12 months, 19% and 24% for focal seizures, and 25% and 7% for generalised seizures at these time points respectively. The retention rate was 42% at 12 months. Treatment was discontinued due to unwanted effects in 29 (36.7%) of the 79 patients with follow-up data available up to 12 months: 30% due to challenging behaviour, 14% dizziness, and 7.6% somnolence. CONCLUSION Perampanel was fairly effective in a heterogeneous group of 96 children and adolescents with very refractory epilepsy. The rate of adverse events leading to discontinuation was considerable in this group.
Collapse
|
189
|
Auvin S, Dozieres B, Ilea A, Delanoë C. Use of perampanel in children and adolescents with Lennox-Gastaut Syndrome. Epilepsy Behav 2017; 74:59-63. [PMID: 28715780 DOI: 10.1016/j.yebeh.2017.05.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 05/23/2017] [Accepted: 05/27/2017] [Indexed: 10/19/2022]
Abstract
AIM Report the use of perampanel treatment in children with Lennox-Gastaut syndrome (LGS). METHOD We conducted a prospective study of 13 LGS patients (seven male; mean age, 12.8years) treated with adjunctive perampanel therapy. Perampanel was initiated at 2mg/day and titrated to a median maximum dose of 6mg/day. RESULTS After a mean follow-up duration of 10.8months (range, 1-24months), nine patients (69.2%) were responders (≥50% reduction in total seizure frequency) and nine (69.2%) were rated by their physician as "much improved" or "very much improved". Four patients (30.8%) discontinued perampanel due to the lack of efficacy (n=2) and seizure aggravation (n=2). No patients discontinued due to other adverse events (AEs). AEs were reported for six patients (46.2%) and comprised decreased activity/social interaction (n=3), behavior disturbance with agitation (n=2), and/or fatigue (n=2). All AEs became manageable after perampanel dosing was decreased. Improvements in cognitive function and/or behavior were reported for seven patients (53.8%). Introduction of perampanel allowed the dose reduction and/or discontinuation of other treatments in seven patients (53.8%). INTERPRETATION Perampanel was efficacious and generally well tolerated as an adjunctive treatment for seizures associated with LGS, supporting further research in this area.
Collapse
Affiliation(s)
- Stéphane Auvin
- Université Paris Diderot, Sorbonne Paris Cité, INSERM UMR1141, 75019 Paris, France; AP-HP, Hôpital Robert Debré, Service de Neurologie Pédiatrique et des Maladies Métaboliques, 75019 Paris, France.
| | - Blandine Dozieres
- AP-HP, Hôpital Robert Debré, Service de Neurologie Pédiatrique et des Maladies Métaboliques, 75019 Paris, France
| | - Adina Ilea
- AP-HP, Hôpital Robert Debré, Service de Neurologie Pédiatrique et des Maladies Métaboliques, 75019 Paris, France
| | - Catherine Delanoë
- AP-HP, Hôpital Robert Debré, Service des Explorations Fonctionnelles, 75019 Paris, France
| |
Collapse
|
190
|
Dupuis N, Enderlin J, Thomas J, Desnous B, Dournaud P, Allorge D, Auvin S. Anti-ictogenic and antiepileptogenic properties of perampanel in mature and immature rats. Epilepsia 2017; 58:1985-1992. [DOI: 10.1111/epi.13894] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Nina Dupuis
- National Institute of Health and Medical Research, U1141; Paris France
- Sorbonne Paris Cité; Paris Diderot University; National Institute of Health and Medical Research UMR1141; Paris France
| | - Julie Enderlin
- National Institute of Health and Medical Research, U1141; Paris France
- Sorbonne Paris Cité; Paris Diderot University; National Institute of Health and Medical Research UMR1141; Paris France
| | - Jeremy Thomas
- Functional Toxicology Unit, Lille University Hospital Center; Lille France
| | - Béatrice Desnous
- National Institute of Health and Medical Research, U1141; Paris France
- Sorbonne Paris Cité; Paris Diderot University; National Institute of Health and Medical Research UMR1141; Paris France
- Pediatric Neurology Department; Robert Debré Hospital; Public Hospital Network of Paris; Paris France
| | - Pascal Dournaud
- National Institute of Health and Medical Research, U1141; Paris France
- Sorbonne Paris Cité; Paris Diderot University; National Institute of Health and Medical Research UMR1141; Paris France
| | - Delphine Allorge
- Functional Toxicology Unit, Lille University Hospital Center; Lille France
| | - Stéphane Auvin
- National Institute of Health and Medical Research, U1141; Paris France
- Sorbonne Paris Cité; Paris Diderot University; National Institute of Health and Medical Research UMR1141; Paris France
- Pediatric Neurology Department; Robert Debré Hospital; Public Hospital Network of Paris; Paris France
| |
Collapse
|
191
|
Calpain-Dependent Degradation of Nucleoporins Contributes to Motor Neuron Death in a Mouse Model of Chronic Excitotoxicity. J Neurosci 2017; 37:8830-8844. [PMID: 28821644 DOI: 10.1523/jneurosci.0730-17.2017] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/02/2017] [Accepted: 08/09/2017] [Indexed: 01/07/2023] Open
Abstract
Glutamate-mediated excitotoxicity induces neuronal death by altering various intracellular signaling pathways and is implicated as a common pathogenic pathway in many neurodegenerative diseases. In the case of motor neuron disease, there is significant evidence to suggest that the overactivation of AMPA receptors due to deficiencies in the expression and function of glial glutamate transporters GLT1 and GLAST plays an important role in the mechanisms of neuronal death. However, a causal role for glial glutamate transporter dysfunction in motor neuron death remains unknown. Here, we developed a new animal model of excitotoxicity by conditionally deleting astroglial glutamate transporters GLT1 and GLAST in the spinal cords of mice (GLAST+/-/GLT1-cKO). GLAST+/-/GLT1-cKO mice (both sexes) exhibited nuclear irregularity and calpain-mediated degradation of nuclear pore complexes (NPCs), which are responsible for nucleocytoplasmic transport. These abnormalities were associated with progressive motor neuron loss, severe paralysis, and shortened lifespan. The nuclear export inhibitor KPT-350 slowed but did not prevent motor neuron death, whereas long-term treatment of the AMPA receptor antagonist perampanel and the calpain inhibitor SNJ-1945 had more persistent beneficial effects. Thus, NPC degradation contributes to AMPA receptor-mediated excitotoxic motor neuronal death, and preventing NPC degradation has robust protective effects. Normalization of NPC function could be a novel therapeutic strategy for neurodegenerative disorders in which AMPA receptor-mediated excitotoxicity is a contributory factor.SIGNIFICANCE STATEMENT Despite glial glutamate transporter dysfunction leading to excitotoxicity has been documented in many neurological diseases, it remains unclear whether its dysfunction is a primary cause or secondary outcome of neuronal death at disease state. Here we show the combined loss of glial glutamate transporters GLT1 and GLAST in spinal cord caused motor neuronal death and hindlimb paralysis. Further, our novel mutant exhibits the nuclear irregularities and calpain-mediated progressive nuclear pore complex degradation. Our study reveals that glial glutamate transporter dysfunction is sufficient to cause motor neuronal death in vivo.
Collapse
|
192
|
Datta AN, Xu Q, Sachedina S, Boelman C, Huh L, Connolly MB. Clinical Experience With Perampanel for Refractory Pediatric Epilepsy in One Canadian Center. J Child Neurol 2017; 32:834-839. [PMID: 28511603 DOI: 10.1177/0883073817709195] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Perampanel (PER) is a new antiseizure medication that inhibits the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) class of glutamate receptors. It is important for physicians to be aware of the efficacy and tolerability of new drugs in the postmarketing phase. We performed a retrospective review of our experience with perampanel at BC Children's Hospital. Twenty-four pediatric patients prescribed perampanel from 2014 to 2016 were identified. Fifteen (63%) discontinued perampanel, and 10 (42%) had greater than 50% reduction in seizures. Twelve (50%) had behavioral and 8 (33%) had nonbehavioral adverse events. One-third experienced serious adverse events. One patient experienced oculogyric crisis, which is not previously reported with perampanel. Adverse events were not dose related and were reversible. Possible risk factors for behavioral adverse events include a history of behavioral problems with other antiseizure medications and preexisting behavioral comorbidities. It is important to counsel patients about the potential for serious adverse events, particularly behavioral, when prescribing perampanel.
Collapse
Affiliation(s)
- Anita N Datta
- Department of Pediatrics, Division of Neurology, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Qi Xu
- Department of Pediatrics, Division of Neurology, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Shafina Sachedina
- Department of Pediatrics, Division of Neurology, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Cyrus Boelman
- Department of Pediatrics, Division of Neurology, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Linda Huh
- Department of Pediatrics, Division of Neurology, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Mary B Connolly
- Department of Pediatrics, Division of Neurology, BC Children's Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
193
|
Epilepsy and synaptic proteins. Curr Opin Neurobiol 2017; 45:1-8. [DOI: 10.1016/j.conb.2017.02.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/25/2017] [Accepted: 02/01/2017] [Indexed: 12/22/2022]
|
194
|
Witkin JM, Li J, Gilmour G, Mitchell SN, Carter G, Gleason SD, Seidel WF, Eastwood BJ, McCarthy A, Porter WJ, Reel J, Gardinier KM, Kato AS, Wafford KA. Electroencephalographic, cognitive, and neurochemical effects of LY3130481 (CERC-611), a selective antagonist of TARP-γ8-associated AMPA receptors. Neuropharmacology 2017; 126:257-270. [PMID: 28757050 DOI: 10.1016/j.neuropharm.2017.07.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/27/2017] [Accepted: 07/26/2017] [Indexed: 11/18/2022]
Abstract
6-[(1S)-1-[1-[5-(2-hydroxyethoxy)-2-pyridyl]pyrazol-3-yl]ethyl]-3H-1,3-benzothiazol-2-one (LY3130481 or CERC-611) is a selective antagonist of AMPA receptors containing transmembrane AMPA receptor regulatory protein (TARP) γ-8. This molecule has been characterized as a potent and efficacious anticonvulsant in an array of acute and chronic epilepsy models in rodents. The present set of experiments was designed to assess the effects of LY3130481 on the electroencephelogram (EEG), cognitive function, and neurochemical outflow. LY3130481 disrupted food-maintained responding in rats and spontaneous alternation in a Y-maze in mice. In rat fear conditioning, LY3130481 caused a deficit in trace (hippocampal-dependent), but not in delay fear conditioning. Although these effects on cognitive performances were observed, the known cognitive-impairing anticonvulsant, topiramate, did not always produce deficits under these assay conditions. LY3130481 produced modest increases in wake times in rats. In addition, LY3130481 was able to attenuate some impairing effects of standard antiepileptic drugs. The motor-impairing effects of the lacosamide were attenuated by LY3130481 as was the decrease in non-rapid-eye movement sleep induced by carbamazepine. Evaluation of the effect of LY3130481 on neurotransmitter and metabolite efflux in the rat medial prefrontal cortex, using in vivo microdialysis, revealed significant increases in the pro-cognitive and wake-promoting neurotransmitters, histamine and acetylcholine, as well as in serotonin, telemethylhistamine, 5-HIAA, HVA and MHPG. LY3130481 thus presents a novel behavioral profile that will have to be evaluated in patients to fully appreciate its implications for therapeutics. LY3130481 is currently under clinical development as CERC-611 as an antiepileptic.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA.
| | - Jennifer Li
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Gary Gilmour
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Stephen N Mitchell
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Guy Carter
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Scott D Gleason
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Wesley F Seidel
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Brian J Eastwood
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Andrew McCarthy
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Warren J Porter
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jon Reel
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Kevin M Gardinier
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Akihiko S Kato
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Keith A Wafford
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK.
| |
Collapse
|
195
|
De Liso P, Moavero R, Coppola G, Curatolo P, Cusmai R, De Sarro G, Franzoni E, Vigevano F, Verrotti A. Current role of perampanel in pediatric epilepsy. Ital J Pediatr 2017; 43:51. [PMID: 28577562 PMCID: PMC5457730 DOI: 10.1186/s13052-017-0368-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/24/2017] [Indexed: 02/07/2023] Open
Abstract
Perampanel is among the latest AEDs approved, indicated for the treatment of partial-onset seizures with or without secondary generalization, and for primary generalized tonic-clonic seizures, in patients aged 12 years and older. This paper summarizes the clinical recommendations on the current role of perampanel in the treatment of pediatric epilepsies and future directions for research. The optimal dosage should be comprised between 4 and 12 mg/day, with 8 mg/day being the most common dosage used. The rate and severity of adverse events, including psychiatric symptoms, can be decreased by starting at low doses, and titrating slowly. Overall, perampanel presents an acceptable risk/benefit ratio, but special caution should be made to the risk of seizure aggravation and behavioral problems. The favorable cognitive profile, the ease of use of the titration scheme and the once-daily formulation offer advantage over other AEDs and make this drug particularly suitable for adolescent population. Perampanel is a welcome addition to the armamentarium of the existing AEDs, as it represents a new approach in the management of epilepsy, with a novel mechanism of action and a potential to have a considerable impact on the treatment of adolescents with epilepsy.
Collapse
Affiliation(s)
- Paola De Liso
- Child Neurology Unit, Department of Neuroscience and Neurorehabilitation, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Romina Moavero
- Child Neurology Unit, Department of Neuroscience and Neurorehabilitation, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Piazza S. Onofrio 4, 00165, Rome, Italy. .,Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University of Rome, Via Montpellier, 1, 00137, Rome, Italy.
| | - Giangennaro Coppola
- Child and Adolescent Neuropsychiatry, Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University of Rome, Via Montpellier, 1, 00137, Rome, Italy
| | - Raffaella Cusmai
- Child Neurology Unit, Department of Neuroscience and Neurorehabilitation, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Giovambattista De Sarro
- Department of Science of Health, Clinical Pharmacological Unit, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Emilio Franzoni
- Child Neurology and Psychiatry Unit, University of Bologna, Bologna, Italy
| | - Federico Vigevano
- Child Neurology Unit, Department of Neuroscience and Neurorehabilitation, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Alberto Verrotti
- Department of Pediatrics, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
196
|
Das D, Dey S, Brewster RC, Choubey S. Effect of transcription factor resource sharing on gene expression noise. PLoS Comput Biol 2017; 13:e1005491. [PMID: 28414750 PMCID: PMC5411101 DOI: 10.1371/journal.pcbi.1005491] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/01/2017] [Accepted: 03/31/2017] [Indexed: 12/31/2022] Open
Abstract
Gene expression is intrinsically a stochastic (noisy) process with important implications for cellular functions. Deciphering the underlying mechanisms of gene expression noise remains one of the key challenges of regulatory biology. Theoretical models of transcription often incorporate the kinetics of how transcription factors (TFs) interact with a single promoter to impact gene expression noise. However, inside single cells multiple identical gene copies as well as additional binding sites can compete for a limiting pool of TFs. Here we develop a simple kinetic model of transcription, which explicitly incorporates this interplay between TF copy number and its binding sites. We show that TF sharing enhances noise in mRNA distribution across an isogenic population of cells. Moreover, when a single gene copy shares it’s TFs with multiple competitor sites, the mRNA variance as a function of the mean remains unaltered by their presence. Hence, all the data for variance as a function of mean expression collapse onto a single master curve independent of the strength and number of competitor sites. However, this result does not hold true when the competition stems from multiple copies of the same gene. Therefore, although previous studies showed that the mean expression follows a universal master curve, our findings suggest that different scenarios of competition bear distinct signatures at the level of variance. Intriguingly, the introduction of competitor sites can transform a unimodal mRNA distribution into a multimodal distribution. These results demonstrate the impact of limited availability of TF resource on the regulation of noise in gene expression. Genetically identical cells, even when they are exposed to the same environmental conditions, display incredible diversity. Gene expression noise is attributed to be a key source of this phenotypic diversity. Transcriptional dynamics is a dominant source of expression noise. Although scores of theoretical and experimental studies have explored how noise is regulated at the level of transcription, most of them focus on the gene specific, cis regulatory elements, such as the number of transcription factor (TF) binding sites, their binding strength, etc. However, how the global properties of transcription, such as the limited availability of TFs impact noise in gene expression remains rather elusive. Here we build a theoretical model that incorporates the effect of limiting TF pool on gene expression noise. We find that competition between genes for TFs leads to enhanced variability in mRNA copy number across an isogenic population. Moreover, for gene copies sharing TFs with other competitor sites, mRNA variance as a function of the mean shows distinct imprints for one gene copy and multiple gene copies respectively. This stands in sharp contrast to the universal behavior found in mean expression irrespective of the different scenarios of competition. An interesting feature of competition is that introduction of competitor sites can transform a unimodal mRNA distribution into a multimodal distribution, which could lead to phenotypic variability.
Collapse
Affiliation(s)
- Dipjyoti Das
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, United States of America
| | - Supravat Dey
- Laboratoire Charles Coulomb, Université de Montpellier and CNRS, Montpellier, France
| | - Robert C. Brewster
- Program in Systems Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (RCB); (SC)
| | - Sandeep Choubey
- FAS Center for Systems Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, United States of America
- * E-mail: (RCB); (SC)
| |
Collapse
|
197
|
NMDA receptor antagonism with novel indolyl, 2-(1,1-Dimethyl-1,3-dihydro-benzo[e]indol-2-ylidene)-malonaldehyde, reduces seizures duration in a rat model of epilepsy. Sci Rep 2017; 7:45540. [PMID: 28358047 PMCID: PMC5371989 DOI: 10.1038/srep45540] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/27/2017] [Indexed: 12/15/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDAR) play a central role in epileptogensis and NMDAR antagonists have been shown to have antiepileptic effects in animals and humans. Despite significant progress in the development of antiepileptic therapies over the previous 3 decades, a need still exists for novel therapies. We screened an in-house library of small molecules targeting the NMDA receptor. A novel indolyl compound, 2-(1,1-Dimethyl-1,3-dihydro-benzo[e]indol-2-ylidene)-malonaldehyde, (DDBM) showed the best binding with the NMDA receptor and computational docking data showed that DDBM antagonised the binding sites of the NMDA receptor at lower docking energies compared to other molecules. Using a rat electroconvulsive shock (ECS) model of epilepsy we showed that DDBM decreased seizure duration and improved the histological outcomes. Our data show for the first time that indolyls like DDBM have robust anticonvulsive activity and have the potential to be developed as novel anticonvulsants.
Collapse
|
198
|
Ahrens-Nicklas RC, Umanah GKE, Sondheimer N, Deardorff MA, Wilkens AB, Conlin LK, Santani AB, Nesbitt A, Juulsola J, Ma E, Dawson TM, Dawson VL, Marsh ED. Precision therapy for a new disorder of AMPA receptor recycling due to mutations in ATAD1. NEUROLOGY-GENETICS 2017; 3:e130. [PMID: 28180185 PMCID: PMC5289017 DOI: 10.1212/nxg.0000000000000130] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/13/2016] [Indexed: 11/15/2022]
Abstract
OBJECTIVE ATAD1 encodes Thorase, a mediator of α-amino-3-hydroxy-5-methylisoxazole-4-proprionate (AMPA) receptor recycling; in this work, we characterized the phenotype resulting from ATAD1 mutations and developed a targeted therapy in both mice and humans. METHODS Using exome sequencing, we identified a novel ATAD1 mutation (p.E276X) as the etiology of a devastating neurologic disorder characterized by hypertonia, seizures, and death in a consanguineous family. We postulated that pathogenesis was a result of excessive AMPA receptor activity and designed a targeted therapeutic approach using perampanel, an AMPA-receptor antagonist. RESULTS Perampanel therapy in ATAD1 knockout mice reversed behavioral defects, normalized brain MRI abnormalities, prevented seizures, and prolonged survival. The ATAD1 patients treated with perampanel showed improvement in hypertonicity and resolution of seizures. CONCLUSIONS This work demonstrates that identification of novel monogenic neurologic disorders and observation of response to targeted therapeutics can provide important insights into human nervous system functioning.
Collapse
Affiliation(s)
- Rebecca C Ahrens-Nicklas
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - George K E Umanah
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Neal Sondheimer
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Matthew A Deardorff
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Alisha B Wilkens
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Laura K Conlin
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Avni B Santani
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Addie Nesbitt
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Jane Juulsola
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Erica Ma
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Ted M Dawson
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Valina L Dawson
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| | - Eric D Marsh
- Section of Biochemical Genetics (R.C.A.-N., N.S.), Division of Human Genetics (R.C.A.-N., M.A.D., A.B.W.), Department of Pathology and Laboratory Medicine (L.K.C., A.B.S., A.N.), Division of Child Neurology (E.D.M.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.C.A.-N., N.S., M.A.D., E.D.M.) and Department of Neurology (E.D.M.), Perelman School of Medicine, and Department of Clinical Pathology (L.K.C., A.B.S.), University of Pennsylvania, Philadelphia; GeneDx (J.J.), Gaithersburg, MD; Neuroregeneration and Stem Cell Programs (G.K.E.U., T.M.D., V.L.D.), Institute for Cell Engineering; Departments of Neurology (G.K.E.U., T.M.D.), Solomon H. Snyder Department of Neuroscience (T.M.D., V.L.D.), Pharmacology and Molecular Sciences (T.M.D., V.L.D.), Physiology (V.L.D.), and Public Health (E.M.), Johns Hopkins University, Baltimore, MD
| |
Collapse
|
199
|
Guzeva VI, Okhrim IV, Guzeva OV, Guzeva VV, Maksimova NE. New opportunities in the optimization of epilepsy treatment in adolescents. Zh Nevrol Psikhiatr Im S S Korsakova 2017; 117:70-74. [DOI: 10.17116/jnevro20171179270-74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
200
|
Chen T, Dai SH, Jiang ZQ, Luo P, Jiang XF, Fei Z, Gui SB, Qi YL. The AMPAR Antagonist Perampanel Attenuates Traumatic Brain Injury Through Anti-Oxidative and Anti-Inflammatory Activity. Cell Mol Neurobiol 2017; 37:43-52. [PMID: 26883519 PMCID: PMC11482069 DOI: 10.1007/s10571-016-0341-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/01/2016] [Indexed: 11/25/2022]
Abstract
Perampanel is a novel α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor (AMPAR) antagonist, approved in over 35 countries as an adjunctive therapy for the treatment of seizures. Recently, it was found to exert protective effects against ischemic neuronal injury in vitro. In the present study, we investigated the potential protective effects of perampanel in a traumatic brain injury (TBI) model in rats. Oral administration with perampanel at a dose of 5 mg/kg exerted no major organ-related toxicities. We found that perampanel significantly attenuated TBI-induced brain edema, brain contusion volume, and gross motor dysfunction. The results of Morris water maze test demonstrated that perampanel treatment also improved cognitive function after TBI. These neuroprotective effects were accompanied by reduced neuronal apoptosis, as evidenced by decreased TUNEL-positive cells in brain sections. Moreover, perampanel markedly inhibited lipid peroxidation and obviously preserved the endogenous antioxidant system after TBI. In addition, enzyme-linked immunosorbent assay (ELISA) was performed at 4 and 24 h after TBI to evaluate the expression of inflammatory cytokines. The results showed that perampanel suppressed the expression of pro-inflammatory cytokines TNF-α and IL-1β, whereas increased the levels of anti-inflammatory cytokines IL-10 and TGF-β1. These data show that the orally active AMPAR antagonist perampanel affords protection against TBI-induced neuronal damage and neurological dysfunction through anti-oxidative and anti-inflammatory activity.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, The 123th Hospital of PLA, Bengbu, 233010, Anhui, China
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Shu-Hui Dai
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhi-Quan Jiang
- Department of Neurosurgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xiao-Fan Jiang
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Song-Bai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China.
| | - Yi-Long Qi
- Department of Neurosurgery, The 123th Hospital of PLA, Bengbu, 233010, Anhui, China.
| |
Collapse
|