1
|
Sharpe C, Yang DZ, Haas RH, Reiner GE, Lee L, Capparelli EV. Pharmacokinetic and pharmacodynamic data from the NEOLEV1 and NEOLEV2 studies. Arch Dis Child 2024; 109:854-860. [PMID: 38902005 DOI: 10.1136/archdischild-2022-324952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/15/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVES To confirm that levetiracetam (LEV) demonstrates predictable pharmacokinetics(PK) at higher doses and to study the pharmacodynamics(PD) of LEV. DESIGN Pharmacokinetic data from the NEOLEV1 and NEOLEV2 trials were analysed using a non-linear mixed effects modelling approach. A post hoc analysis of the effect of LEV on seizure burden was conducted. SETTING Neonatal intensive care unit. PATIENTS Term neonates with electrographically confirmed seizures. INTERVENTIONS In NEOLEV1, neonates with seizures persisting following phenobarbital (PHB) received LEV 20 or 40 mg/kg bolus followed by 5 or 10 mg/kg maintenance dose(MD) daily. In NEOLEV2, patients received a 40 mg/kg intravenous LEV load, followed by 10 mg/kg doses 8 hourly. If seizures persisted, a further 20 mg/kg intravenous load was given. If seizures persisted, PHB was given. PK data were collected from 16 NEOLEV1 patients and 33 NEOLEV2 patients. cEEG data from 48 NEOLEV2 patients were analysed to investigate onset of action and seizure burden reduction. MAIN OUTCOME MEASURES Clearance (CL) and volume of distribution (Vd) were determined. Covariates that significantly affected LEV disposition were identified. RESULTS Primary outcome: The median initial LEV level was 57 µg/mL (range 19-107) after the first loading dose and at least 12 µg/mL at 48 hours in all infants. CL and Vd were estimated to be 0.0538 L/hour and 0.832 L, respectively. A direct relationship between postnatal age and CL was observed. The final population pharmacokinetic(PopPK) model described the observed data well without significant biases. CL and Vd were described as CL (L/hour)=0.0538×(weight in kg/3.34)0.75×(postnatal age in days/5.5) 0.402 and Vd (L)=0.832×(weight in kg/3.34).Seizure burden reduced within 30 min of LEV administration. 28% of patients were completely seizure free after LEV. In an additional 25% of patients, seizure burden reduced by 50%. CONCLUSIONS LEV pharmacokinetics remained predictable at higher doses. Very high-dose LEV can now be studied in neonates. TRIAL REGISTRATION NUMBER NCT01720667.
Collapse
Affiliation(s)
- Cynthia Sharpe
- Paediatric Neurology, Starship Children's Health, Auckland, New Zealand
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Derek Z Yang
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Richard H Haas
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
- Department of Neurology, Rady Children's Hospital-San Diego, San Diego, California, USA
| | - Gail E Reiner
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Lilly Lee
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Edmund V Capparelli
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
2
|
Mehta P, Soliman A, Rodriguez-Vera L, Schmidt S, Muniz P, Rodriguez M, Forcadell M, Gonzalez-Perez E, Vozmediano V. Interspecies Brain PBPK Modeling Platform to Predict Passive Transport through the Blood-Brain Barrier and Assess Target Site Disposition. Pharmaceutics 2024; 16:226. [PMID: 38399280 PMCID: PMC10892872 DOI: 10.3390/pharmaceutics16020226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The high failure rate of central nervous system (CNS) drugs is partly associated with an insufficient understanding of target site exposure. Blood-brain barrier (BBB) permeability evaluation tools are needed to explore drugs' ability to access the CNS. An outstanding aspect of physiologically based pharmacokinetic (PBPK) models is the integration of knowledge on drug-specific and system-specific characteristics, allowing the identification of the relevant factors involved in target site distribution. We aimed to qualify a PBPK platform model to be used as a tool to predict CNS concentrations when significant transporter activity is absent and human data are sparse or unavailable. Data from the literature on the plasma and CNS of rats and humans regarding acetaminophen, oxycodone, lacosamide, ibuprofen, and levetiracetam were collected. Human BBB permeability values were extrapolated from rats using inter-species differences in BBB surface area. The percentage of predicted AUC and Cmax within the 1.25-fold criterion was 85% and 100% for rats and humans, respectively, with an overall GMFE of <1.25 in all cases. This work demonstrated the successful application of the PBPK platform for predicting human CNS concentrations of drugs passively crossing the BBB. Future applications include the selection of promising CNS drug candidates and the evaluation of new posologies for existing drugs.
Collapse
Affiliation(s)
- Parsshava Mehta
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (P.M.); (A.S.); (S.S.)
| | - Amira Soliman
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (P.M.); (A.S.); (S.S.)
- Department of Pharmacy Practice, Faculty of Pharmacy, Helwan University, Helwan 11795, Egypt
| | - Leyanis Rodriguez-Vera
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA; (L.R.-V.); (P.M.); (M.R.)
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (P.M.); (A.S.); (S.S.)
| | - Paula Muniz
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA; (L.R.-V.); (P.M.); (M.R.)
| | - Monica Rodriguez
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA; (L.R.-V.); (P.M.); (M.R.)
| | - Marta Forcadell
- Neuraxpharm Pharmaceuticals SL, Clinical Research and Evidence-Generation Science, 08970 Barcelona, Spain; (M.F.); (E.G.-P.)
| | - Emili Gonzalez-Perez
- Neuraxpharm Pharmaceuticals SL, Clinical Research and Evidence-Generation Science, 08970 Barcelona, Spain; (M.F.); (E.G.-P.)
| | - Valvanera Vozmediano
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (P.M.); (A.S.); (S.S.)
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA; (L.R.-V.); (P.M.); (M.R.)
| |
Collapse
|
3
|
Guignet M, Vuong J, Martinez A, Ballapapinan T, White HS. Temporal relationship between levetiracetam nonadherence and breakthrough seizures in a preclinical model of temporal lobe epilepsy. Epilepsia 2024; 65:497-510. [PMID: 38031477 DOI: 10.1111/epi.17835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023]
Abstract
OBJECTIVE Poor medication adherence remains a concern for individuals managing their epilepsy with antiseizure medicines (ASMs); however, ethical concerns around withholding medication make it impossible to study the causal relationship between missed doses and seizures in patients. Previous preclinical studies from our group suggest that mechanistically distinct ASMs have varying degrees of forgiveness when a dose is missed. However, with only a few ASMs studied in the context of nonadherence, we sought to expand on previous work to understand the relationship between levetiracetam (LEV) nonadherence and breakthrough seizures. METHODS Chronic oral dosing was initiated in rats with established epilepsy via our automated medication-in-food delivery system coupled to 24/7 video-electroencephalographic recording. Baseline seizure burden was established for 4 weeks before enrolling subjects into a 4-week treatment period with LEV in a 100% fully adherent (75 mg/kg four times daily) or 50% variably adherent paradigm. The temporal relationship between missed doses and breakthrough seizures was correlated with LEV plasma and brain concentrations in separate cohorts of animals. RESULTS Full adherence to LEV significantly improved seizure control by 50% in half of the animals. Poor adherence worsened seizure frequency by 85%, with most rats having more severe seizures that formed in clusters following missed doses. LEV concentrations remained below therapeutic levels (<10 μg/mL) in nonadherent animals, with brain and plasma levels directly correlating with the degree of adherence in a 24-h period. Missed doses of LEV immediately increased the risk of breakthrough seizures; however, this risk was significantly reduced with improved adherence in a 24-h period. SIGNIFICANCE These findings enhance our understanding of ASM nonadherence in preclinical models, highlighting that the timing of missed doses and their impact on seizures may vary between different ASMs. Notably, LEV demonstrates a robust pharmacokinetic reliance on missed doses leading to breakthrough seizures.
Collapse
Affiliation(s)
- Michelle Guignet
- Center for Epilepsy Drug Discovery, Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Jonathan Vuong
- Center for Epilepsy Drug Discovery, Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Alejandra Martinez
- Center for Epilepsy Drug Discovery, Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Ticha Ballapapinan
- Center for Epilepsy Drug Discovery, Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - H Steve White
- Center for Epilepsy Drug Discovery, Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| |
Collapse
|
4
|
Kajevu N, Lipponen A, Andrade P, Bañuelos I, Puhakka N, Hämäläinen E, Natunen T, Hiltunen M, Pitkänen A. Treatment of Status Epilepticus after Traumatic Brain Injury Using an Antiseizure Drug Combined with a Tissue Recovery Enhancer Revealed by Systems Biology. Int J Mol Sci 2023; 24:14049. [PMID: 37762352 PMCID: PMC10531083 DOI: 10.3390/ijms241814049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/30/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
We tested a hypothesis that in silico-discovered compounds targeting traumatic brain injury (TBI)-induced transcriptomics dysregulations will mitigate TBI-induced molecular pathology and augment the effect of co-administered antiseizure treatment, thereby alleviating functional impairment. In silico bioinformatic analysis revealed five compounds substantially affecting TBI-induced transcriptomics regulation, including calpain inhibitor, chlorpromazine, geldanamycin, tranylcypromine, and trichostatin A (TSA). In vitro exposure of neuronal-BV2-microglial co-cultures to compounds revealed that TSA had the best overall neuroprotective, antioxidative, and anti-inflammatory effects. In vivo assessment in a rat TBI model revealed that TSA as a monotherapy (1 mg/kg/d) or in combination with the antiseizure drug levetiracetam (LEV 150 mg/kg/d) mildly mitigated the increase in plasma levels of the neurofilament subunit pNF-H and cortical lesion area. The percentage of rats with seizures during 0-72 h post-injury was reduced in the following order: TBI-vehicle 80%, TBI-TSA (1 mg/kg) 86%, TBI-LEV (54 mg/kg) 50%, TBI-LEV (150 mg/kg) 40% (p < 0.05 vs. TBI-vehicle), and TBI-LEV (150 mg/kg) combined with TSA (1 mg/kg) 30% (p < 0.05). Cumulative seizure duration was reduced in the following order: TBI-vehicle 727 ± 688 s, TBI-TSA 898 ± 937 s, TBI-LEV (54 mg/kg) 358 ± 715 s, TBI-LEV (150 mg/kg) 42 ± 64 (p < 0.05 vs. TBI-vehicle), and TBI-LEV (150 mg/kg) combined with TSA (1 mg/kg) 109 ± 282 s (p < 0.05). This first preclinical intervention study on post-TBI acute seizures shows that a combination therapy with the tissue recovery enhancer TSA and LEV was safe but exhibited no clear benefit over LEV monotherapy on antiseizure efficacy. A longer follow-up is needed to confirm the possible beneficial effects of LEV monotherapy and combination therapy with TSA on chronic post-TBI structural and functional outcomes, including epileptogenesis.
Collapse
Affiliation(s)
- Natallie Kajevu
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Anssi Lipponen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, P.O. Box 95, 70701 Kuopio, Finland
| | - Pedro Andrade
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Ivette Bañuelos
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Noora Puhakka
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Elina Hämäläinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Asla Pitkänen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| |
Collapse
|
5
|
Fukushima K, Higashiyama H, Kazuta Y, Hashimoto K, Watanabe N, Furuya Y, Ito Y, Wu T, Kosasa T, Talos DM, Song Y, Roberts NS, Jensen FE, Hanada T, Ido K. Discovery of E2730, a novel selective uncompetitive GAT1 inhibitor, as a candidate for anti-seizure medication. Epilepsia Open 2023; 8:834-845. [PMID: 37052238 PMCID: PMC10472371 DOI: 10.1002/epi4.12741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/08/2023] [Indexed: 04/14/2023] Open
Abstract
OBJECTIVE As of 2022, 36 anti-seizure medications (ASMs) have been licensed for the treatment of epilepsy, however, adverse effects (AEs) are commonly reported. Therefore, ASMs with a wide margin between therapeutic effects and AEs are preferred over ASMs that are associated with a narrow margin between efficacy and risk of AEs. E2730 was discovered using in vivo phenotypic screening and characterized as an uncompetitive, yet selective, inhibitor of γ-aminobutyric acid (GABA) transporter 1 (GAT1). Here, we describe the preclinical characteristics of E2730. METHODS Anti-seizure effects of E2730 were evaluated in several animal models of epilepsy: corneal kindling, 6 Hz-44 mA psychomotor seizure, amygdala kindling, Fragile X syndrome, and Dravet syndrome models. Effects of E2730 on motor coordination were assessed in accelerating rotarod tests. The mechanism of action of E2730 was explored by [3 H]E2730 binding assay. The GAT1-selectivity over other GABA transporters was examined by GABA uptake assay of GAT1, GAT2, GAT3, or betaine/GABA transporter 1 (BGT-1) stably expressing HEK293 cells. To further investigate the mechanism for E2730-mediated inhibition of GAT1, in vivo microdialysis and in vitro GABA uptake assays were conducted under conditions of different GABA concentrations. RESULTS E2730 showed anti-seizure effects in the assessed animal models with an approximately >20-fold margin between efficacy and motor incoordination. [3 H]E2730 binding on brain synaptosomal membrane was abolished in GAT1-deficient mice, and E2730 selectively inhibited GAT1-mediated GABA uptake over other GABA transporters. In addition, results of GABA uptake assays showed that E2730-mediated inhibition of GAT1 positively correlated to the level of ambient GABA in vitro. E2730 also increased extracellular GABA concentration in hyperactivated conditions but not under basal levels in vivo. SIGNIFICANCE E2730 is a novel, selective, uncompetitive GAT1 inhibitor, which acts selectively under the condition of increasing synaptic activity, contributing to a wide margin between therapeutic effect and motor incoordination.
Collapse
Affiliation(s)
| | | | - Yuji Kazuta
- Deep Human Biology LearningEisai Co., Ltd.TsukubaIbarakiJapan
| | | | - Naoto Watanabe
- Deep Human Biology LearningEisai Co., Ltd.TsukubaIbarakiJapan
| | - Yoshiaki Furuya
- Deep Human Biology LearningEisai Co., Ltd.TsukubaIbarakiJapan
| | - Yoshimasa Ito
- Neurology Business GroupEisai Co., Ltd.TsukubaIbarakiJapan
| | - Ting Wu
- Alzheimer's Disease and Brain HealthEisai Co., Ltd.TsukubaIbarakiJapan
| | - Takashi Kosasa
- Neurology Business GroupEisai Co., Ltd.TsukubaIbarakiJapan
| | - Delia M. Talos
- Department of Neurology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Yeri Song
- Department of Neurology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Nicholas S. Roberts
- Department of Neurology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Frances E. Jensen
- Department of Neurology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Takahisa Hanada
- Deep Human Biology LearningEisai Co., Ltd.TsukubaIbarakiJapan
| | - Katsutoshi Ido
- Neurology Business GroupEisai Co., Ltd.TsukubaIbarakiJapan
| |
Collapse
|
6
|
Elshater AA, Sadek AA, Abdelkreem E. Levetiracetam and Midazolam vs Midazolam Alone for First-Line Treatment of Children With Generalized Convulsive Status Epilepticus (Lev-Mid Study): A Randomized Controlled Trial. Indian Pediatr 2023; 60:630-636. [PMID: 37211889 DOI: 10.1007/s13312-023-2960-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/23/2022] [Accepted: 02/04/2023] [Indexed: 10/31/2024]
Abstract
BACKGROUND Benzodiazepines are the first-line anti-seizure medication (ASM) for generalized convulsive status epilepticus (GCSE), but they fail to end seizures in a third of cases. Combining benzodiazepines with another ASM that acts by a different pathway could be a potential strategy for rapid control of GCSE. OBJECTIVES To evaluate the efficacy of adding levetiracetam to midazolam in the initial treatment of pediatric GCSE. DESIGN Double-blind randomized controlled trial. SETTING Pediatric emergency room at Sohag University Hospital between June, 2021 and August, 2022. PARTICIPANTS Children aged between 1 month and 16 years with GCSE lasting more than 5 min. INTERVENTIONS Intravenous levetiracetam (60 mg/kg over 5 min) and midazolam (Lev-Mid group) or placebo and midazolam (Pla-Mid group) as first-line anticonvulsive therapy. OUTCOME MEASURES Primary: cessation of clinical seizures at 20-min study time point. Secondary: cessation of clinical seizures at 40-min study time point, need for a second midazolam dose, seizure control at 24-hr, need for intubation, and adverse effects. RESULTS Cessation of clinical seizures at 20-min occurred in 55 children (76%) in Lev-Mid group compared with 50 (69%) in the Pla-Mid group [RR (95% CI) 1.1 (0.9-1.34); P=0.35]. No significant difference was found between the two groups regarding the need for a second midazolam dose [44.4% vs 55.6%; RR (95% CI) 0.8 (0.58-1.11); P=0.18] as well as cessation of clinical seizures at 40-min [96% vs 92%; RR (95% CI)1.05 (0.96-1.14); P=0.49] and seizure control at 24-hr [85% vs 76%; RR (95% CI) 1.12 (0.94-1.3); P=0.21]. Intubation was required for three patients in the Lev-Mid group and six patients in the Pla-Mid group [RR (95%CI) 0.5 (0.13- 1.92); P=0.49]. No other adverse effects or mortality were observed during the 24-hour study timeframe. CONCLUSION Combined levetiracetam and midazolam for initial management of pediatric GCSE presents no significant advantage over midazolam alone in cessation of clinical seizures at 20-min.
Collapse
Affiliation(s)
- Ahmed A Elshater
- Department of Pediatrics, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Abdelrahim A Sadek
- Department of Pediatrics, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Elsayed Abdelkreem
- Department of Pediatrics, Faculty of Medicine, Sohag University, Sohag, Egypt. Correspondence to: Dr Elsayed Abdelkreem, Department of Pediatrics, Faculty of Medicine, Sohag University, Sohag 82524, Egypt.
| |
Collapse
|
7
|
Contreras-García IJ, Cárdenas-Rodríguez N, Romo-Mancillas A, Bandala C, Zamudio SR, Gómez-Manzo S, Hernández-Ochoa B, Mendoza-Torreblanca JG, Pichardo-Macías LA. Levetiracetam Mechanisms of Action: From Molecules to Systems. Pharmaceuticals (Basel) 2022; 15:ph15040475. [PMID: 35455472 PMCID: PMC9030752 DOI: 10.3390/ph15040475] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a chronic disease that affects millions of people worldwide. Antiepileptic drugs (AEDs) are used to control seizures. Even though parts of their mechanisms of action are known, there are still components that need to be studied. Therefore, the search for novel drugs, new molecular targets, and a better understanding of the mechanisms of action of existing drugs is still crucial. Levetiracetam (LEV) is an AED that has been shown to be effective in seizure control and is well-tolerable, with a novel mechanism of action through an interaction with the synaptic vesicle protein 2A (SV2A). Moreover, LEV has other molecular targets that involve calcium homeostasis, the GABAergic system, and AMPA receptors among others, that might be integrated into a single mechanism of action that could explain the antiepileptogenic, anti-inflammatory, neuroprotective, and antioxidant properties of LEV. This puts it as a possible multitarget drug with clinical applications other than for epilepsy. According to the above, the objective of this work was to carry out a comprehensive and integrative review of LEV in relation to its clinical uses, structural properties, therapeutical targets, and different molecular, genetic, and systemic action mechanisms in order to consider LEV as a candidate for drug repurposing.
Collapse
Affiliation(s)
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Antonio Romo-Mancillas
- Laboratorio de Diseño Asistido por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico;
| | - Cindy Bandala
- Neurociencia Básica, Instituto Nacional de Rehabilitación LGII, Secretaría de Salud, Ciudad de México 14389, Mexico;
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Sergio R. Zamudio
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México 06720, Mexico;
| | - Julieta Griselda Mendoza-Torreblanca
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
- Correspondence: (J.G.M.-T.); (L.A.P.-M.); Tel.: +52-55-1084-0900 (ext. 1441) (J.G.M.-T.)
| | - Luz Adriana Pichardo-Macías
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
- Correspondence: (J.G.M.-T.); (L.A.P.-M.); Tel.: +52-55-1084-0900 (ext. 1441) (J.G.M.-T.)
| |
Collapse
|
8
|
Alavi MS, Negah SS, Ghorbani A, Hosseini A, Sadeghnia HR. Levetiracetam promoted rat embryonic neurogenesis via NMDA receptor-mediated mechanism in vitro. Life Sci 2021; 284:119923. [PMID: 34481865 DOI: 10.1016/j.lfs.2021.119923] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 10/20/2022]
Abstract
AIMS Levetiracetam (LEV) is a broad-spectrum antiepileptic drug with neuroprotective properties and novel mechanisms of action. Some evidence suggests that LEV may impact adult neurogenesis, but the results are controversial. The present study was aimed to evaluate the effects of LEV on the proliferation and differentiation of rat embryonic neural stem cells (NSCs) and to explore the role of GABAB or NMDA receptors. MAIN METHODS NSCs were isolated from rat fetal ganglionic eminence at embryonic day 14.5. The effects of LEV on viability, proliferation, neurosphere formation, and neuronal or astroglial differentiation of NSCs were assessed using resazurin, BrdU incorporation, immunocytochemistry, quantitative real-time PCR, and western blotting. Additionally, we addressed the relationship between treatment with NMDA and GABAB receptor antagonists (MK801 and saclofen, respectively) in combination with LEV on these parameters. KEY FINDINGS The data showed that LEV (50 μM) significantly increased the number (p < 0.01) and diameter of neurospheres (p < 0.05), enhanced proliferation (p < 0.01), and promoted neuronal differentiation, as revealed by significantly increased expressions of DCX and NeuN. The expressions of astroglial markers, GFAP and Olig2, were markedly reduced. The addition of MK801 (10 μM) significantly diminished neurospheres growth (p < 0.001), decreased the number of proliferating cells (p < 0.01), and reduced the number of new neurons (p < 0.001) but increased the astroglial cells (p < 0.001) induced by LEV. Co-treatment with saclofen (25 μM) did not significantly affect LEV-induced NSCs proliferation and differentiation. SIGNIFICANCE Our findings suggest that LEV may enhance rat embryonic neurogenesis mainly through an NMDA receptor-mediated mechanism.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Ghorbani
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azar Hosseini
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid R Sadeghnia
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Ong CLJ, Goh PSJ, Teo MM, Lim TP, Goh KKK, Ang XY, Lim LJK, Jamaludin NHB, Ang BT, Kwa LHA. Pharmacokinetics of levetiracetam in neurosurgical ICU patients. J Crit Care 2021; 64:255-261. [PMID: 34051579 DOI: 10.1016/j.jcrc.2021.04.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND/OBJECTIVES The pharmacokinetics (PK) of drugs is dramatically altered in critical illness. Augmented renal clearance (ARC), a phenomenon characterized by creatinine clearance (CrCl) greater than 130 ml/min/1.73m2, is commonly described in critically ill patients. Levetiracetam, an antiepileptic drug commonly prescribed for seizure prophylaxis in the neurosurgical ICU, undergoes predominant elimination via the kidneys. Hence, we hypothesize that current dosing practice of intravenous (IV) levetiracetam 500 mg twice daily is inadequate for critically ill patients due to enhanced drug elimination. The objectives of our study were to describe the population PK of levetiractam using a nonparametric approach to design an optimal dosing regimen for critically ill neurosurgical patients. METHODS This was a prospective, observational, population PK study. Serial blood samples were obtained from neurosurgical ICU patients who received at least one dose of IV levetiracetam. We used uHPLC to analyze these samples and Pmetrics™ software to perform PK analysis. RESULTS Twenty subjects were included, with a median age of 54 years and CrCl of 104 ml/min. A two-compartmental model with linear elimination adequately described the profile of levetiracetam. Mean clearance (CL) was 3.55 L/h and volume of distribution (V) was 18.8 L. No covariates were included in the final model. Monte Carlo simulations showed a low probability of target attainment (PTA, trough at steady state of ≥6 mg/L) with a standard dose of 500 mg twice daily. A dose of at least 1000 mg twice daily was required to achieve 80% PTA. Two subjects, both with subtherapeutic trough levels, developed early onset seizures. CONCLUSION Our study examined the population PK of levetiracetam in a critically ill neurosurgical population. We found that this population displayed higher clearance and required higher doses to achieve target levels.
Collapse
Affiliation(s)
| | - Pheck Suan June Goh
- Department of Anaesthesiology, Singapore General Hospital, Singapore, Singapore
| | - Miqi Mavis Teo
- Department of Anaesthesiology, Singapore General Hospital, Singapore, Singapore
| | - Tze Peng Lim
- Department of Pharmacy, Singapore General Hospital, Singapore, Singapore
| | | | - Xiu Yun Ang
- Department of Pharmacy, Singapore General Hospital, Singapore, Singapore
| | | | | | - Beng Ti Ang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore; Duke-National University of Singapore Medical School, Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lay Hoon Andrea Kwa
- Department of Pharmacy, Singapore General Hospital, Singapore, Singapore; Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore; Singhealth Duke-NUS Medicine Academic Clinical Programme, Singapore, Singapore
| |
Collapse
|
10
|
Yang L, Afroz S, Valsamis HA, Michelson HB, Goodman JH, Ling DSF. Early intervention with levetiracetam prevents the development of cortical hyperexcitability and spontaneous epileptiform activity in two models of neurotrauma in rats. Exp Neurol 2020; 337:113571. [PMID: 33340499 DOI: 10.1016/j.expneurol.2020.113571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/09/2020] [Accepted: 12/13/2020] [Indexed: 10/22/2022]
Abstract
This study examined the antiepileptogenic potential of the antiseizure drug (ASD) levetiracetam (LEV) using the in vitro traumatized-slice and in vivo controlled cortical impact (CCI) models of traumatic brain injury (TBI) in rats when administered early after the injury. For the in vitro model, acute coronal slices (400-450 μm) of rat neocortex (P21-32) were injured via a surgical cut that separated the superficial layers from the deeper regions. Persistent stimulus-evoked epileptiform activity developed within 1-2 h after trauma. In randomly selected slices, LEV (500 μM) was bath-applied for 1 h starting immediately or delayed by 30-80 min after injury. Treated and untreated slices were examined for epileptiform activity via intracellular and extracellular recordings. For the in vivo model, rats (P24-32) were subjected to a non-penetrating, focal, CCI injury targeting the neocortex (5.0 mm diameter; 2.0 mm depth). Immediately after injury, rats were given either a single dose of LEV (60-150 mg/kg, i.p.) or the saline vehicle. At 2-3 weeks after the injury, ex vivo cortical slices were examined for epileptiform activity. The results from the traumatized-slice experiments showed that in vitro treatment with LEV within 60 min of injury significantly reduced (> 50%) the proportion of slices that exhibited stimulus-evoked epileptiform activity. LEV treatment also increased the stimulus intensity required to trigger epileptiform bursts in injured slices by 2-4 fold. Consistent with these findings, LEV treatment of CCI-injured rats (n = 15) significantly reduced the proportion of animals that exhibited spontaneous and stimulus-evoked epileptiform bursts in ex vivo cortical slices compared to saline-treated controls (n = 15 rats), and also significantly increased the stimulus intensity required to evoke epileptiform bursts. These results suggest that early administration of LEV has the potential to prevent or reduce posttraumatic epileptogenesis and that there may be a narrow therapeutic window for successful prophylactic intervention.
Collapse
Affiliation(s)
- Lie Yang
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA
| | - Sonia Afroz
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Program in Neural and Behavioral Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA
| | - Helen A Valsamis
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Department of Neurology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 1213, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Neurology Service, Kings County Hospital Center, Brooklyn, NY 11203, USA.
| | - Hillary B Michelson
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA.
| | - Jeffrey H Goodman
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Department of Neurology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 1213, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Department of Developmental Neurobiology, The New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA.
| | - Douglas S F Ling
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Program in Neural and Behavioral Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA.
| |
Collapse
|
11
|
Johnson ER, Taylor AR, Boothe DM, Gray-Edwards HL, Winter RL, Martin DR. Pharmacokinetics of a commercially available product and a compounded formulation of extended-release levetiracetam after oral administration of a single dose in cats. Am J Vet Res 2019; 80:950-956. [PMID: 31556719 DOI: 10.2460/ajvr.80.10.950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To compare pharmacokinetics of levetiracetam in serum and CSF of cats after oral administration of extended-release (ER) levetiracetam. ANIMALS 9 healthy cats. PROCEDURES Cats received 1 dose of a commercially available ER levetiracetam product (500 mg, PO). Thirteen blood and 10 CSF samples were collected over a 24-hour period for pharmacokinetic analysis. After 1 week, cats received 1 dose of a compounded ER levetiracetam formulation (500 mg, PO), and samples were obtained at the same times for analysis. RESULTS CSF concentrations of levetiracetam closely paralleled serum concentrations. There were significant differences between the commercially available product and the compounded formulation for mean ± SD serum maximum concentration (Cmax; 126 ± 33 μg/mL and 169 ± 51 μg/mL, respectively), Cmax corrected for dose (0.83 ± 0.10 μg/mL/mg and 1.10 ± 0.28 μg/mL/mg, respectively), and time to Cmax (5.1 ± 1.6 hours and 3.1 ± 1.5 hours, respectively). Half-life for the commercially available product and compounded formulation of ER levetiracetam was 4.3 ± 2.0 hours and 5.0 ± 1.6 hours, respectively. CONCLUSIONS AND CLINICAL RELEVANCE The commercially available product and compounded formulation of ER levetiracetam both maintained concentrations in healthy cats 12 hours after oral administration that have been found to be therapeutic in humans (ie, 5 μg/mL). Results of this study supported dosing intervals of 12 hours, and potentially 24 hours, for oral administration of ER levetiracetam to cats. Monitoring of serum concentrations of levetiracetam can be used as an accurate representation of levetiracetam concentrations in CSF of cats.
Collapse
|
12
|
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: 2.8] [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
|
13
|
Perrenoud M, André P, Buclin T, Decosterd LA, Rossetti AO, Novy J. Levetiracetam circulating concentrations and response in status epilepticus. Epilepsy Behav 2018; 88:61-65. [PMID: 30241055 DOI: 10.1016/j.yebeh.2018.08.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 08/20/2018] [Accepted: 08/20/2018] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Intravenous levetiracetam (LEV) is broadly used in the treatment of status epilepticus (SE). A loading dose is usually infused, aiming to reach quickly the range of plasma concentrations considered as therapeutic (12-46 mg/l). The aim of the study was to evaluate the response to LEV in SE, correlated exposure assessed by plasma concentration monitoring, as well as calculated exposure parameters. MATERIALS & METHODS We retrospectively analyzed a SE registry, including patients since 2015 with at least one available LEV plasma level measured less than 36 h after loading. A Bayesian maximum likelihood approach based on a population pharmacokinetic model was used to estimate LEV exposure parameters. We compared plasma levels and pharmacokinetics parameter estimates between responders and nonresponders. Therapeutic response was defined as SE cessation within 24 h following LEV introduction without a need for additional antiepileptic drug (AED). RESULTS We included 29 patients (45 plasma levels). Variability was salient in LEV loading doses (ranging between 17 and 38 mg/kg) and monitoring practice. There was no difference in median plasma concentrations (19.5 versus 21.5 mg/l; p = 0.71), median estimated LEV exposure (25.8 versus 37.0 mg/l; p = 0.61), peak (30.4 versus 41.5 mg/l; p = 0.36), or residual levels after loading dose (14.4 versus 20.5 mg/l; p = 0.07) between responders and nonresponders. CONCLUSIONS Levetiracetam exposure does not seem to differ significantly between responders and nonresponders; greater exposure was not associated with better outcome. Loading doses of 30 mg/kg seem, however, appropriate to quickly reach the target exposure level. The short LEV half-life makes standardized sampling measurement necessary to obtain directly interpretable LEV levels.
Collapse
Affiliation(s)
- Matthieu Perrenoud
- Neurology Service, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Switzerland
| | - Pascal André
- Division of Clinical Pharmacology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Switzerland
| | - Thierry Buclin
- Division of Clinical Pharmacology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Switzerland
| | - Laurent A Decosterd
- Clinical Pharmacology Laboratory, Laboratory Department, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Switzerland
| | - Andrea O Rossetti
- Neurology Service, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Switzerland
| | - Jan Novy
- Neurology Service, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Switzerland.
| |
Collapse
|
14
|
Marino S, Vitaliti G, Marino SD, Pavone P, Provvidenti S, Romano C, Falsaperla R. Pyridoxine Add-On Treatment for the Control of Behavioral Adverse Effects Induced by Levetiracetam in Children: A Case-Control Prospective Study. Ann Pharmacother 2018; 52:645-649. [PMID: 29442544 DOI: 10.1177/1060028018759637] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Few studies on adult and pediatric patients have shown pyridoxine efficacy as additional therapy for those receiving levetiracetam (LEV) to prevent and mitigate behavioral adverse effects (BAEs). OBJECTIVE The aim of our study was to analyze the safety and efficacy of pyridoxine supplementation in the prevention of LEV adverse effects, including suicidal ideation. METHODS This randomized, case-control trial included patients receiving LEV as monotherapy treatment. Patients were subdivided into 2 groups, according to whether they were treated with LEV only (group 1) or LEV with supplemental pyridoxine (group 2). RESULTS In both cohorts, the most frequent BAEs were irritability/aggression followed by depression and confusion. Those patients (92%) who initiated pyridoxine after 1 month of LEV treatment did not need to change or suspend LEV ( P < 0.001), and BAE improved after 9.06 ± 3.05 days of pyridoxine supplementation. None of the patients complained of symptoms of pyridoxine toxicity, and no new adverse effects of LEV off-label were reported. CONCLUSIONS In our study, we found pyridoxine to be safe and effective in controlling LEV-induced BAEs in children.
Collapse
Affiliation(s)
- Silvia Marino
- 1 University Hospital Policlinico-Vittorio Emanuele, Catania, Italy
| | | | | | - Piero Pavone
- 1 University Hospital Policlinico-Vittorio Emanuele, Catania, Italy
| | | | - Catia Romano
- 1 University Hospital Policlinico-Vittorio Emanuele, Catania, Italy
| | | |
Collapse
|
15
|
Kovačević J, Maroteaux G, Schut D, Loos M, Dubey M, Pitsch J, Remmelink E, Koopmans B, Crowley J, Cornelisse LN, Sullivan PF, Schoch S, Toonen RF, Stiedl O, Verhage M. Protein instability, haploinsufficiency, and cortical hyper-excitability underlie STXBP1 encephalopathy. Brain 2018; 141:1350-1374. [PMID: 29538625 PMCID: PMC5917748 DOI: 10.1093/brain/awy046] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/15/2017] [Accepted: 01/05/2018] [Indexed: 01/14/2023] Open
Abstract
De novo heterozygous mutations in STXBP1/Munc18-1 cause early infantile epileptic encephalopathies (EIEE4, OMIM #612164) characterized by infantile epilepsy, developmental delay, intellectual disability, and can include autistic features. We characterized the cellular deficits for an allelic series of seven STXBP1 mutations and developed four mouse models that recapitulate the abnormal EEG activity and cognitive aspects of human STXBP1-encephalopathy. Disease-causing STXBP1 variants supported synaptic transmission to a variable extent on a null background, but had no effect when overexpressed on a heterozygous background. All disease variants had severely decreased protein levels. Together, these cellular studies suggest that impaired protein stability and STXBP1 haploinsufficiency explain STXBP1-encephalopathy and that, therefore, Stxbp1+/- mice provide a valid mouse model. Simultaneous video and EEG recordings revealed that Stxbp1+/- mice with different genomic backgrounds recapitulate the seizure/spasm phenotype observed in humans, characterized by myoclonic jerks and spike-wave discharges that were suppressed by the antiepileptic drug levetiracetam. Mice heterozygous for Stxbp1 in GABAergic neurons only, showed impaired viability, 50% died within 2-3 weeks, and the rest showed stronger epileptic activity. c-Fos staining implicated neocortical areas, but not other brain regions, as the seizure foci. Stxbp1+/- mice showed impaired cognitive performance, hyperactivity and anxiety-like behaviour, without altered social behaviour. Taken together, these data demonstrate the construct, face and predictive validity of Stxbp1+/- mice and point to protein instability, haploinsufficiency and imbalanced excitation in neocortex, as the underlying mechanism of STXBP1-encephalopathy. The mouse models reported here are valid models for development of therapeutic interventions targeting STXBP1-encephalopathy.
Collapse
Affiliation(s)
- Jovana Kovačević
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
- Sylics (Synaptologics BV), Amsterdam, The Netherlands
| | - Gregoire Maroteaux
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Desiree Schut
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Maarten Loos
- Sylics (Synaptologics BV), Amsterdam, The Netherlands
| | - Mohit Dubey
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Julika Pitsch
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, 53105 Bonn, Germany
| | | | | | - James Crowley
- UNC Center for Psychiatric Genomics, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599-7160, USA
| | - L Niels Cornelisse
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Patrick F Sullivan
- UNC Center for Psychiatric Genomics, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599-7160, USA
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics and Department of (Clinical) Genetics, Nobels väg 12A, 171 77 Stockholm, Sweden
| | - Susanne Schoch
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, 53105 Bonn, Germany
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Oliver Stiedl
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and VU Medical Center, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
16
|
André EA, Forcelli PA, Pak DT. What goes up must come down: homeostatic synaptic plasticity strategies in neurological disease. FUTURE NEUROLOGY 2018; 13:13-21. [PMID: 29379396 DOI: 10.2217/fnl-2017-0028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/07/2017] [Indexed: 11/21/2022]
Abstract
Brain activity levels are tightly regulated to minimize imbalances in activity state. Deviations from the normal range of activity are deleterious and often associated with neurological disorders. To maintain optimal levels of activity, regulatory mechanisms termed homeostatic synaptic plasticity establish desired 'set points' for neural activity, monitor the network for deviations from the set point and initiate compensatory responses to return activity to the appropriate level that permits physiological function [1,2]. We speculate that impaired homeostatic control may contribute to the etiology of various neurological disorders including epilepsy and Alzheimer's disease, two disorders that exhibit hyperexcitability as a key feature during pathogenesis. Here, we will focus on recent progress in developing homeostatic regulation of neural activity as a therapeutic tool.
Collapse
Affiliation(s)
- Emily A André
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Patrick A Forcelli
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Daniel Ts Pak
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington DC 20057, USA
| |
Collapse
|
17
|
MacDonald KD, Hart KA, Davis JL, Berghaus LJ, Giguère S. Pharmacokinetics of the anticonvulsant levetiracetam in neonatal foals. Equine Vet J 2017; 50:532-536. [DOI: 10.1111/evj.12790] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 11/23/2017] [Indexed: 11/27/2022]
Affiliation(s)
- K. D. MacDonald
- Department of Large Animal Medicine; College of Veterinary Medicine; University of Georgia; Athens Georgia USA
| | - K. A. Hart
- Department of Large Animal Medicine; College of Veterinary Medicine; University of Georgia; Athens Georgia USA
| | - J. L. Davis
- Department of Biomedical Sciences and Pathobiology; Virginia-Maryland College of Veterinary Medicine; Blacksburg Virginia USA
| | - L. J. Berghaus
- Department of Large Animal Medicine; College of Veterinary Medicine; University of Georgia; Athens Georgia USA
| | - S. Giguère
- Department of Large Animal Medicine; College of Veterinary Medicine; University of Georgia; Athens Georgia USA
| |
Collapse
|
18
|
Kelly D, Raimondi F, Shihab N. Levetiracetam monotherapy for treatment of structural epilepsy in dogs: 19 cases (2010-2015). Vet Rec 2017; 181:401. [DOI: 10.1136/vr.104190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 06/27/2017] [Accepted: 07/29/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Darren Kelly
- Department of Internal Medicine; Southern Counties Veterinary Specialists LLP; Ringwood UK
| | - Francesca Raimondi
- Department of Neurology and Neurosurgery; Southern Counties Veterinary Specialists LLP; Ringwood UK
| | - Nadia Shihab
- Department of Neurology and Neurosurgery; Southern Counties Veterinary Specialists; Ringwood Hampshire UK
| |
Collapse
|
19
|
Cesar FB, Stewart AJ, Boothe DM, Ravis WR, Duran SH, Wooldridge AA. Disposition of levetiracetam in healthy adult horses. J Vet Pharmacol Ther 2017; 41:92-97. [PMID: 28503770 DOI: 10.1111/jvp.12417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 04/05/2017] [Indexed: 11/29/2022]
Abstract
Nine horses received 20 mg/kg of intravenous (LEVIV ); 30 mg/kg of intragastric, crushed immediate release (LEVCIR ); and 30 mg/kg of intragastric, crushed extended release (LEVCER ) levetiracetam, in a three-way randomized crossover design. Crushed tablets were dissolved in water and administered by nasogastric tube. Serum samples were collected over 48 hr, and levetiracetam concentrations were determined by immunoassay. Mean ± SD peak concentrations for LEVCIR and LEVCER were 50.72 ± 10.60 and 53.58 ± 15.94 μg/ml, respectively. The y-intercept for IV administration was 64.54 ± 24.99 μg/ml. The terminal half-life was 6.38 ± 1.97, 7.07 ± 1.93 and 6.22 ± 1.35 hr for LEVCIR , LEVCER, and LEVIV , respectively. Volume of distribution at steady-state was 630 ± 73.4 ml/kg. Total body clearance after IV administration was 74.40 ± 19.20 ml kg-1 hr-1 . Bioavailability was 96 ± 10, and 98 ± 13% for LEVCIR and LEVCER , respectively. A single dose of Levetiracetam (LEV) was well tolerated. Based on this study, a recommended dosing regimen of intravenous or oral LEV of 32 mg/kg every 12 hr is likely to achieve and maintain plasma concentrations within the therapeutic range suggested for humans, with optimal kinetics throughout the dosing interval in healthy adult horses. Repeated dosing and pharmacodynamic studies are warranted.
Collapse
Affiliation(s)
- F B Cesar
- Department of Clinical Sciences, Auburn University College of Veterinary Medicine, Auburn, AL, USA
| | - A J Stewart
- Department of Clinical Sciences, Auburn University College of Veterinary Medicine, Auburn, AL, USA
| | - D M Boothe
- Department of Veterinary Physiology, Pathology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, USA
| | - W R Ravis
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL, USA
| | - S H Duran
- Department of Clinical Sciences, Auburn University College of Veterinary Medicine, Auburn, AL, USA
| | - A A Wooldridge
- Department of Clinical Sciences, Auburn University College of Veterinary Medicine, Auburn, AL, USA
| |
Collapse
|
20
|
Beuchat I, Novy J, Rossetti AO. Newer Antiepileptic Drugs in Status Epilepticus: Prescription Trends and Outcomes in Comparison with Traditional Agents. CNS Drugs 2017; 31:327-334. [PMID: 28337727 DOI: 10.1007/s40263-017-0424-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Newer antiepileptic drugs (AEDs) are increasingly prescribed; however, relatively limited data are available regarding their use in status epilepticus (SE) and the impact on outcome. OBJECTIVES The aim of this study was to explore the evolution in prescription patterns of newer and traditional AEDs in this clinical setting, and their association with prognosis. METHODS We analyzed our prospective adult SE registry over a 10-year period (2007-2016) and assessed the yearly use of newer and traditional AEDs and their association with mortality, return to baseline conditions at discharge, and SE refractoriness, defined as treatment resistance to two AEDs, including benzodiazepines. RESULTS In 884 SE episodes, corresponding to 719 patients, the prescription of at least one newer AED increased from 0.38 per SE episode in 2007 to 1.24 per SE episode in 2016 (mostly due to the introduction of levetiracetam and lacosamide). Traditional AEDs (excluding benzodiazepines) decreased over time from 0.74 in 2007 to 0.41 in 2016, correlating with the decreasing use of phenytoin. The prescription of newer AEDs was independently associated with a lower chance of return to baseline conditions at discharge (odds ratio [OR] 0.58, 95% confidence interval [CI] 0.40-0.84) and a higher rate of SE refractoriness (OR 19.84, 95% CI 12.76-30.84), but not with changes in mortality (OR 1.08, 95% CI 0.58-2.00). CONCLUSION We observed a growing trend in the prescription of newer AEDs in SE over the last decade; however, our findings might suggest an associated increased risk of SE refractoriness and new disability at hospital discharge. Pending prospective, comparative studies, this may justify some caution in the routine use of newer AEDs in SE.
Collapse
Affiliation(s)
- Isabelle Beuchat
- Service de Neurologie, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois (CHUV), CHUV-BH07, and Lausanne University Hospital, 1011, Lausanne, Switzerland
| | - Jan Novy
- Service de Neurologie, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois (CHUV), CHUV-BH07, and Lausanne University Hospital, 1011, Lausanne, Switzerland
| | - Andrea O Rossetti
- Service de Neurologie, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois (CHUV), CHUV-BH07, and Lausanne University Hospital, 1011, Lausanne, Switzerland.
| |
Collapse
|
21
|
Abbaskhanian A, Shahmohammadi S. Add-on Levetiracetam in Children With Refractory Epilepsy: A Systematic Review. JOURNAL OF PEDIATRICS REVIEW 2016. [DOI: 10.17795/jpr-6276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
22
|
Dose-dependent effects of levetiracetam after hypoxia and hypothermia in the neonatal mouse brain. Brain Res 2016; 1646:116-124. [PMID: 27216570 DOI: 10.1016/j.brainres.2016.05.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/18/2016] [Accepted: 05/19/2016] [Indexed: 10/21/2022]
Abstract
Perinatal asphyxia to the developing brain remains a major cause of morbidity. Hypothermia is currently the only established neuroprotective treatment available for term born infants with hypoxic-ischemic encephalopathy, saving one in seven to eight infants from developing severe neurological deficits. Therefore, additional treatments with clinically applicable drugs are indispensable. This study investigates a potential additive neuroprotective effect of levetiracetam combined with hypothermia after hypoxia-induced brain injury in neonatal mice. 9-day-old C57BL/6-mice (P9) were subjected either to acute hypoxia or room-air. After 90min of systemic hypoxia (6% O2), pups were randomized into six groups: 1) vehicle, 2) low-dose levetiracetam (LEV), 3) high-dose LEV, 4) hypothermia (HT), 5) HT combined with low-dose LEV and 6) HT combined with high-dose LEV. Pro-apoptotic factors, neuronal structures, and myelination were analysed by histology and on protein level at appropriate time points. On P28 to P37 long-term outcome was assessed by neurobehavioral testing. Hypothermia confers acute and long-term neuroprotection by reducing apoptosis and preservation of myelinating oligodendrocytes and neurons in a model of acute hypoxia in the neonatal mouse brain. Low-dose LEV caused no adverse effects after neonatal hypoxic brain damage treated with hypothermia whereas administration of high-dose LEV alone or in combination with hypothermia increased neuronal apoptosis after hypoxic brain injury. LEV in low- dosage had no additive neuroprotective effect following acute hypoxic brain injury.
Collapse
|
23
|
Nicolas JM, Hannestad J, Holden D, Kervyn S, Nabulsi N, Tytgat D, Huang Y, Chanteux H, Staelens L, Matagne A, Mathy FX, Mercier J, Stockis A, Carson RE, Klitgaard H. Brivaracetam, a selective high-affinity synaptic vesicle protein 2A (SV2A) ligand with preclinical evidence of high brain permeability and fast onset of action. Epilepsia 2015; 57:201-9. [PMID: 26663401 DOI: 10.1111/epi.13267] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Rapid distribution to the brain is a prerequisite for antiepileptic drugs used for treatment of acute seizures. The preclinical studies described here investigated the high-affinity synaptic vesicle glycoprotein 2A (SV2A) antiepileptic drug brivara-cetam (BRV) for its rate of brain penetration and its onset of action. BRV was compared with levetiracetam (LEV). METHODS In vitro permeation studies were performed using Caco-2 cells. Plasma and brain levels were measured over time after single oral dosing to audiogenic mice and were correlated with anticonvulsant activity. Tissue distribution was investigated after single dosing to rat (BRV and LEV) and dog (LEV only). Positron emission tomography (PET) displacement studies were performed in rhesus monkeys using the SV2A PET tracer [11C]UCB-J. The time course of PET tracer displacement was measured following single intravenous (IV) dosing with LEV or BRV. Rodent distribution data and physiologically based pharmacokinetic (PBPK) modeling were used to compute blood-brain barrier permeability (permeability surface area product, PS) values and then predict brain kinetics in man. RESULTS In rodents, BRV consistently showed a faster entry into the brain than LEV; this correlated with a faster onset of action against seizures in audiogenic susceptible mice. The higher permeability of BRV was also demonstrated in human cells in vitro. PBPK modeling predicted that, following IV dosing to human subjects, BRV might distribute to the brain within a few minutes compared with approximately 1 h for LEV (PS of 0.315 and 0.015 ml/min/g for BRV and LEV, respectively). These data were supported by a nonhuman primate PET study showing faster SV2A occupancy by BRV compared with LEV. SIGNIFICANCE These preclinical data demonstrate that BRV has rapid brain entry and fast brain SV2A occupancy, consistent with the fast onset of action in the audiogenic seizure mice assay. The potential benefit of BRV for treatment of acute seizures remains to be confirmed in clinical studies.
Collapse
Affiliation(s)
| | | | - Daniel Holden
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, U.S.A
| | | | - Nabeel Nabulsi
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, U.S.A
| | | | - Yiyun Huang
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, U.S.A
| | | | | | | | | | | | | | - Richard E Carson
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, U.S.A
| | | |
Collapse
|
24
|
Abstract
Intravenous and intramuscular antiseizure drugs (ASDs) are essential in the treatment of clinical seizure emergencies as well as in replacement therapy when oral administration is not possible. The parenteral formulations provide rapid delivery and complete (intravenous) or nearly complete (intramuscular) bioavailability. Controlled administration of the ASD is feasible with intravenous but not intramuscular formulations. This article reviews the literature and discusses the chemistry, pharmacology, pharmacokinetics, and clinical use of currently available intravenous and intramuscular ASD formulations as well as the development of new formulations and agents. Intravenous or intramuscular formulations of lorazepam, diazepam, midazolam, and clonazepam are typically used as the initial treatment agents in seizure emergencies. Recent studies also support the use of intramuscular midazolam as easier than the intravenous delivery of lorazepam in the pre-hospital setting. However, benzodiazepines may be associated with hypotension and respiratory depression. Although loading with intravenous phenytoin was an early approach to treatment, it is associated with cardiac arrhythmias, hypotension, and tissue injury at the injection site. This has made it less favored than fosphenytoin, a water-soluble, phosphorylated phenytoin molecule. Other drugs being used for acute seizure emergencies are intravenous formulations of valproic acid, levetiracetam, and lacosamide. However, the comparative effectiveness of these for status epilepticus (SE) has not been evaluated adequately. Consequently, guidelines for the medical management of SE continue to recommend lorazepam followed by fosphenytoin, or phenytoin if fosphenytoin is not available. Intravenous solutions for carbamazepine, lamotrigine, and topiramate have been developed but remain investigational. The current ASDs were not developed for use in emergency situations, but were adapted from ASDs approved for chronic oral use. New approaches for bringing drugs from experimental models to treatment of human SE are needed.
Collapse
|
25
|
Navarro V, Dagron C, Elie C, Lamhaut L, Demeret S, Urien S, An K, Bolgert F, Tréluyer JM, Baulac M, Carli P. Prehospital treatment with levetiracetam plus clonazepam or placebo plus clonazepam in status epilepticus (SAMUKeppra): a randomised, double-blind, phase 3 trial. Lancet Neurol 2015; 15:47-55. [PMID: 26627366 DOI: 10.1016/s1474-4422(15)00296-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/06/2015] [Accepted: 10/09/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND Generalised convulsive status epilepticus (GCSE) should be treated quickly. Benzodiazepines are the only drug treatment available so far that is effective before admission to hospital. We assessed whether addition of the antiepileptic drug levetiracetam to the benzodiazepine clonazepam would improve prehospital treatment of GCSE. METHODS We did a prehospital, randomised, double-blind, phase 3, placebo-controlled, superiority trial to determine the efficacy of adding intravenous levetiracetam (2.5 g) to clonazepam (1 mg) in treatment of GCSE in 13 emergency medical service centres and 26 hospital departments in France. Randomisation was done at the Paris Descartes Clinical Research Unit with a list of random numbers generated by computer. Adults with convulsions lasting longer than 5 min were randomly assigned (1:1) by prehospital physicians to receive levetiracetam or placebo in combination with clonazepam. All physicians and paramedics were masked to group assignments. If the status epilepticus lasted beyond 5 min after drug injection, a second dose of 1 mg clonazepam was given. The primary outcome was cessation of convulsions within 15 min of drug injection. We analysed the modified intention-to-treat population that had received at least one injection of clonazepam and levetiracetam or placebo, excluding patients without valid consent and those randomised more than once. The trial is registered at EudraCT, number 2007-005782-35. FINDINGS Between July 20, 2009, and Dec 15, 2012, 107 patients were randomly assigned to receive placebo and 96 were assigned to receive levetiracetam. The trial was discontinued on Dec 15, 2012 when interim analysis showed no evidence of a treatment difference, and 68 patients in each group were included in the modified intention-to-treat analysis. Convulsions stopped at 15 min of drug injection in 57 of 68 patients (84%) receiving clonazepam and placebo and in 50 of 68 patients (74%) receiving clonazepam and levetiracetam (percentage difference -10.3%, 95% CI -24.0 to 3.4). Three deaths, 19 of 47 (40 %) serious adverse events, and 90 of 197 (46%) non-serious events were reported in the levetiracetam group, and four deaths, 28 of 47 (60%) serious events, and 107 of 197 (54%) non-serious events were reported in the placebo group. INTERPRETATION The addition of levetiracetam to clonazepam treatment presented no advantage over clonazepam treatment alone in the control of GCSE before admission to hospital. Future prehospital trials could assess the efficacy of clonazepam alone as a first-line treatment in status epilepticus and the efficacy of a second injection of clonazepam with another antiepileptic drug as second-line treatment. FUNDING UCB Pharma.
Collapse
Affiliation(s)
- Vincent Navarro
- Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Epilepsy Unit, and Brain and Spine Institute, Pitié-Salpêtrière Hospital and Université Pierre et Marie Curie, Paris, France.
| | - Christelle Dagron
- AP-HP, Necker-Enfants Malades Hospital, SAMU 75, and Université Paris Descartes, Paris, France
| | - Caroline Elie
- AP-HP, Paris Descartes Clinical Research Unit/Clinical Investigation Centre and Université Paris Descartes, France
| | - Lionel Lamhaut
- AP-HP, Necker-Enfants Malades Hospital, SAMU 75, and Université Paris Descartes, Paris, France
| | - Sophie Demeret
- AP-HP, Pitié-Salpêtrière Hospital, Neurological Intensive Care Unit, Paris, France
| | - Saïk Urien
- AP-HP, Paris Descartes Clinical Research Unit/Clinical Investigation Centre and Université Paris Descartes, France
| | - Kim An
- AP-HP, Necker-Enfants Malades Hospital, SAMU 75, and Université Paris Descartes, Paris, France
| | - Francis Bolgert
- AP-HP, Pitié-Salpêtrière Hospital, Neurological Intensive Care Unit, Paris, France
| | - Jean-Marc Tréluyer
- AP-HP, Paris Descartes Clinical Research Unit/Clinical Investigation Centre and Université Paris Descartes, France
| | - Michel Baulac
- Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Epilepsy Unit, and Brain and Spine Institute, Pitié-Salpêtrière Hospital and Université Pierre et Marie Curie, Paris, France
| | - Pierre Carli
- AP-HP, Necker-Enfants Malades Hospital, SAMU 75, and Université Paris Descartes, Paris, France
| | | |
Collapse
|
26
|
Zheng F, Du C, Wang X. Levetiracetam for the treatment of status epilepticus. Expert Rev Neurother 2015; 15:1113-21. [DOI: 10.1586/14737175.2015.1088785] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
27
|
Leppik IE, Patel SI. Intramuscular and rectal therapies of acute seizures. Epilepsy Behav 2015; 49:307-12. [PMID: 26071998 DOI: 10.1016/j.yebeh.2015.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 05/01/2015] [Indexed: 11/16/2022]
Abstract
The intramuscular (IM) and rectal routes are alternative routes of delivery for antiepileptic drugs (AEDs) when the intravenous route is not practical or possible. For treatment of acute seizures, the AED used should have a short time to maximum concentration (Tmax). Some AEDs have preparations that may be given intramuscularly. These include the benzodiazepines (diazepam, lorazepam, and midazolam) and others (fosphenytoin, levetiracetam). Although phenytoin and valproate have parenteral preparations, these should not be given intramuscularly. A recent study of prehospital treatment of status epilepticus evaluated a midazolam (MDZ) autoinjector delivering IM drug compared to IV lorazepam (LZP). Seizures were absent on arrival to the emergency department in 73.4% of the IM MDZ compared to a 63.4% response in LZP-treated subjects (p < 0.001 for superiority). Almost all AEDs have been evaluated for rectal administration as solutions, gels, and suppositories. In a placebo-controlled study, diazepam (DZP) was administered at home by caregivers in doses that ranged from 0.2 to 0.5 mg/kg. Diazepam was superior to placebo in reduced seizure frequency in children (p < 0.001) and in adults (p = 0.02) and time to recurrent seizures after an initial treatment (p < 0.001). Thus, at this time, only MZD given intramuscularly and DZP given rectally appear to have the properties required for rapid enough absorption to be useful when intravenous routes are not possible. Some drugs cannot be administered rectally owing to factors such as poor absorption or poor solubility in aqueous solutions. The relative rectal bioavailability of gabapentin, oxcarbazepine, and phenytoin is so low that the current formulations are not considered to be suitable for administration by this route. When administered as a solution, diazepam is rapidly absorbed rectally, reaching the Tmax within 5-20 min in children. By contrast, rectal administration of lorazepam is relatively slow, with a Tmax of 1-2h. The dependence of gabapentin on an active transport system, and the much-reduced surface area of the rectum compared with the small intestine, may be responsible for its lack of absorption from the rectum. This article is part of a Special Issue entitled "Status Epilepticus".
Collapse
Affiliation(s)
- Ilo E Leppik
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA; MINCEP Epilepsy Care, University of Minnesota Physicians, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Sima I Patel
- MINCEP Epilepsy Care, University of Minnesota Physicians, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
28
|
Abstract
BACKGROUND Levetiracetam, a second-generation anti-epileptic drug (AED) with a good efficacy and safety profile, is licensed as monotherapy for adults and children older than 16 years with focal seizures with or without secondary generalization. However, it is increasingly being used off-label in younger children. OBJECTIVES We critically reviewed the available evidence and discuss the present status of levetiracetam monotherapy in children 0-16 years old. DATA SOURCES We systematically searched the literature using PubMed, Web of Science and Embase up to August 2014 for articles on levetiracetam monotherapy in children. Keywords were levetiracetam, monotherapy and child*. The titles and abstracts of 532 articles were evaluated by AW, of which 480 were excluded. The full texts of the other 52 articles were assessed for relevance. RESULTS We covered one review, one opinion statement and 32 studies in this review, including four randomized controlled trials, ten open-label prospective studies, eight retrospective studies, and ten case reports. The formal evidence for levetiracetam monotherapy in children is minimal: it is potentially efficacious or effective as initial monotherapy in children with benign epilepsy with centrotemporal spikes. In all of the published studies, however, efficacy and tolerability of levetiracetam seemed to be good and comparable to other AEDs. CONCLUSION The data of 32 studies on levetiracetam monotherapy in children were insufficient to confirm that levetiracetam is effective as initial monotherapy for different types of seizures and/or epilepsy syndromes. There is still an urgent need for well designed trials to justify the widespread use of levetiracetam monotherapy in children of all ages.
Collapse
Affiliation(s)
- Amerins Weijenberg
- Department of Neurology, University Medical Centre Groningen, University of Groningen, AB 51, P.O. Box 30 001, 9700 RB Groningen, The Netherlands
| | - Oebele F. Brouwer
- Department of Neurology, University Medical Centre Groningen, University of Groningen, AB 51, P.O. Box 30 001, 9700 RB Groningen, The Netherlands
| | - Petra M. C. Callenbach
- Department of Neurology, University Medical Centre Groningen, University of Groningen, AB 51, P.O. Box 30 001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
29
|
Schnellbacher R, Beaufrère H, Vet DM, Arnold RD, Tully TN, Mayer J, Divers SJ. Pharmacokinetics of Levetiracetam in Healthy Hispaniolan Amazon Parrots (Amazona ventralis) After Oral Administration of a Single Dose. J Avian Med Surg 2014; 28:193-200. [DOI: 10.1647/2013-057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
30
|
Levetiracetam results in increased and decreased alcohol drinking with different access procedures in C57BL/6J mice. Behav Pharmacol 2014; 25:61-70. [PMID: 24322822 DOI: 10.1097/fbp.0000000000000019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The antiepileptic levetiracetam (LEV) has been investigated for the treatment of alcohol abuse. However, little is known about how LEV alters the behavioral effects of alcohol in laboratory animals. The acute effects of LEV on alcohol drinking by male C57BL/6J mice were investigated using two different drinking procedures, limited access [drinking-in-the-dark (DID)] and intermittent access (IA) drinking. In the first experiment (DID), mice had access to a single bottle containing alcohol or sucrose for 4 h every other day. In the second experiment (IA), mice had IA to two bottles, one containing alcohol or sucrose and one containing water, for 24 h on Monday, Wednesday, and Friday. In both experiments, mice were administered LEV (0.3-100 mg/kg intraperitoneally) or vehicle 30 min before access to the drinking solutions. In the DID mice, LEV increased alcohol intake from 4.3 to 5.4 g/kg, whereas in the IA mice LEV decreased alcohol intake from 4.8 to 3.0 g/kg in the first 4 h of access and decreased 24 h alcohol intake from 20 to ∼15 g/kg. These effects appear specific to alcohol, as LEV did not affect sucrose intake in either experiment. LEV appears to differentially affect drinking in animal models of moderate and heavier alcohol consumption.
Collapse
|
31
|
|
32
|
Cuomo O, Rispoli V, Leo A, Politi GB, Vinciguerra A, di Renzo G, Cataldi M. The antiepileptic drug levetiracetam suppresses non-convulsive seizure activity and reduces ischemic brain damage in rats subjected to permanent middle cerebral artery occlusion. PLoS One 2013; 8:e80852. [PMID: 24236205 PMCID: PMC3827478 DOI: 10.1371/journal.pone.0080852] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/11/2013] [Indexed: 02/06/2023] Open
Abstract
The antiepileptic drug Levetiracetam (Lev) has neuroprotective properties in experimental stroke, cerebral hemorrhage and neurotrauma. In these conditions, non-convulsive seizures (NCSs) propagate from the core of the focal lesion into perilesional tissue, enlarging the damaged area and promoting epileptogenesis. Here, we explore whether Lev neuroprotective effect is accompanied by changes in NCS generation or propagation. In particular, we performed continuous EEG recordings before and after the permanent occlusion of the middle cerebral artery (pMCAO) in rats that received Lev (100 mg/kg) or its vehicle immediately before surgery. Both in Lev-treated and in control rats, EEG activity was suppressed after pMCAO. In control but not in Lev-treated rats, EEG activity reappeared approximately 30-45 min after pMCAO. It initially consisted in single spikes and, then, evolved into spike-and-wave and polyspike-and-wave discharges. In Lev-treated rats, only rare spike events were observed and the EEG power was significantly smaller than in controls. Approximately 24 hours after pMCAO, EEG activity increased in Lev-treated rats because of the appearance of polyspike events whose power was, however, significantly smaller than in controls. In rats sacrificed 24 hours after pMCAO, the ischemic lesion was approximately 50% smaller in Lev-treated than in control rats. A similar neuroprotection was observed in rats sacrificed 72 hours after pMCAO. In conclusion, in rats subjected to pMCAO, a single Lev injection suppresses NCS occurrence for at least 24 hours. This electrophysiological effect could explain the long lasting reduction of ischemic brain damage caused by this drug.
Collapse
Affiliation(s)
- Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatologic Sciences, Federico II University of Naples, Naples, Italy
| | - Vincenzo Rispoli
- Department of Health Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Antonio Leo
- Department of Health Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Giovanni Bosco Politi
- Department of Health Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Antonio Vinciguerra
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatologic Sciences, Federico II University of Naples, Naples, Italy
| | - Gianfranco di Renzo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatologic Sciences, Federico II University of Naples, Naples, Italy
| | - Mauro Cataldi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatologic Sciences, Federico II University of Naples, Naples, Italy
- * E-mail:
| |
Collapse
|
33
|
Barrera-Bailón B, Oliveira JAC, López DE, Muñoz LJ, Garcia-Cairasco N, Sancho C. Pharmacological and neuroethological studies of three antiepileptic drugs in the Genetic Audiogenic Seizure Hamster (GASH:Sal). Epilepsy Behav 2013; 28:413-25. [PMID: 23872084 DOI: 10.1016/j.yebeh.2013.05.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 12/25/2022]
Abstract
Epilepsy modeling is essential for understanding the basic mechanisms of the epileptic process. The Genetic Audiogenic Seizure Hamster (GASH:Sal) exhibits generalized tonic-clonic seizures of genetic origin in response to sound stimulation and is currently being validated as a reliable model of epilepsy. Here, we performed a pharmacological and neuroethological study using well-known and widely used antiepileptic drugs (AEDs), including phenobarbital (PB), valproic acid (VPA), and levetiracetam (LEV). The intraperitoneal administration of PB (5-20mg/kg) and VPA (100-300mg/kg) produced a dose-dependent decrease in GASH:Sal audiogenic seizure severity scores. The administration of LEV (30-100mg/kg) did not produce a clear effect. Phenobarbital showed a short plasmatic life and had a high antiepileptic effect starting at 10mg/kg that was accompanied by ataxia. Valproic acid acted only at high concentrations and was the AED with the most ataxic effects. Levetiracetam at all doses also produced sedation and ataxia side effects. We conclude that the GASH:Sal is a reliable genetic model of epilepsy suitable to evaluate AEDs.
Collapse
Affiliation(s)
- B Barrera-Bailón
- Institute of Neurosciences of Castilla and León/IBSAL, University of Salamanca, Salamanca, Spain
| | | | | | | | | | | |
Collapse
|
34
|
A simple assay for determination of levetiracetam in rat dried blood spots by LC-MS/MS. Bioanalysis 2013; 5:1843-51. [PMID: 23905858 DOI: 10.4155/bio.13.143] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND A simple LC-MS/MS method was developed and validated for the quantification of levetiracetam (LEV, Keppra®), a broad-spectrum antiepileptic drug (AED) in rat dried blood spots (DBS). LEV was simply extracted with methanol spiked with adenosine (ADE) as IS before LC-MS/MS analysis. The correlation between the DBS and plasma concentrations of LEV was also determined. RESULTS Linearity was from 0.067-60 µg/ml for LEV in DBS samples. The intra- and inter-day accuracy and precision of the assay met validation acceptance criteria. The developed assay was applied to monitor levetiracetam DBS levels in Sprague-Dawley rats after intravenous administration. DBS concentrations were well correlated to the plasma concentrations (R² = 0.9399), as fraction of LEV bound to blood cells remains very constant (0.466 ± 0.041) over a wide concentration range. CONCLUSION The study illustrated that DBS could be used as alternative matrix for monitoring LEV in preclinical studies.
Collapse
|
35
|
Levetiracetam has opposite effects on alcohol- and cocaine-related behaviors in C57BL/6J mice. Neuropsychopharmacology 2013; 38:1322-33. [PMID: 23353709 PMCID: PMC3656375 DOI: 10.1038/npp.2013.30] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The antiepileptic drug levetiracetam (LEV) is a potential treatment for alcohol use disorders, yet few preclinical studies exist on its effects in animal models relevant to drug or alcohol abuse. We investigated the effects of LEV on locomotor stimulation following acute and repeated administration of alcohol or cocaine and on alcohol- and cocaine-mediated changes in responding for brain stimulation reward (BSR) in C57BL/6J mice. LEV alone (10.0-100.0 mg/kg intraperitoneally) had no significant effect on locomotor activity or intracranial self-stimulation. Pretreatment with LEV reduced acute locomotor stimulation by 2.0 g/kg alcohol, attenuated the development of locomotor sensitization to alcohol with repeated exposure, and produced a shift in the dose-response curve for alcohol on BSR threshold without affecting maximum operant response rate (MAX). Conversely, LEV pretreatment enhanced both acute locomotor stimulation by 15 mg/kg cocaine and development of locomotor sensitization following repeated exposure and produced a leftward shift in the dose-response curve for cocaine on BSR threshold without affecting MAX. Electrophysiological recordings in vitro showed that LEV reduced excitatory currents in both ventral tegmental area (VTA) dopamine neurons and nucleus accumbens (NAc) medium spiny neurons, consistent with a presynaptic effect. The opposite effects of LEV pretreatment on alcohol- and cocaine-related behaviors may predict its clinical utility in the treatment of patients with alcohol, but not psychostimulant abuse disorders.
Collapse
|
36
|
Hardy B, Patterson EE, Cloyd J, Hardy R, Leppik I. Double-Masked, Placebo-Controlled Study of Intravenous Levetiracetam for the Treatment of Status Epilepticus and Acute Repetitive Seizures in Dogs. J Vet Intern Med 2012; 26:334-40. [DOI: 10.1111/j.1939-1676.2011.00868.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 11/03/2011] [Accepted: 12/01/2011] [Indexed: 12/01/2022] Open
Affiliation(s)
- B.T. Hardy
- Veterinary Clinical Sciences; University of Minnesota; Saint Paul; MN
| | - E. E. Patterson
- Veterinary Clinical Sciences; University of Minnesota; Saint Paul; MN
| | - J.M. Cloyd
- Experimental and Clinical Pharmacology; University of Minnesota; Minneapolis; MN
| | - R.M. Hardy
- Veterinary Clinical Sciences; University of Minnesota; Saint Paul; MN
| | | |
Collapse
|
37
|
Meehan AL, Yang X, McAdams BD, Yuan L, Rothman SM. A new mechanism for antiepileptic drug action: vesicular entry may mediate the effects of levetiracetam. J Neurophysiol 2011; 106:1227-39. [PMID: 21653714 PMCID: PMC3174821 DOI: 10.1152/jn.00279.2011] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/03/2011] [Indexed: 11/22/2022] Open
Abstract
Levetiracetam (LEV) is one of the most commonly prescribed antiepileptic drugs, but its mechanism of action is uncertain. Based on prior information that LEV binds to the vesicular protein synaptic vesicle protein 2A and reduces presynaptic neurotransmitter release, we wanted to more rigorously characterize its effect on transmitter release and explain the requirement for a prolonged incubation period for its full effect to manifest. During whole cell patch recordings from rat hippocampal pyramidal neurons in vitro, we found that LEV decreased synaptic currents in a frequency-dependent manner and reduced the readily releasable pool of vesicles. When we manipulated spontaneous activity and stimulation paradigms, we found that synaptic activity during LEV incubation alters the time at which LEV's effect appears, as well as its magnitude. We believe that synaptic activity and concomitant vesicular release allow LEV to enter recycling vesicles to reach its binding site, synaptic vesicle protein 2A. In support of this hypothesis, a vesicular "load-unload" protocol using hypertonic sucrose in the presence of LEV quickly induced LEV's effect. The effect rapidly disappeared after unloading in the absence of LEV. These findings are compatible with LEV acting at an intravesicular binding site to modulate the release of transmitter and with its most marked effect on rapidly discharging neurons. Our results identify a unique neurobiological explanation for LEV's highly selective antiepileptic effect and suggest that synaptic vesicle proteins might be appropriate targets for the development of other neuroactive drugs.
Collapse
Affiliation(s)
- Anna L Meehan
- Department of Neurology, University of Minnesota Medical School, 420 Delaware St., MMC 295, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
38
|
Carnes MB, Axlund TW, Boothe DM. Pharmacokinetics of levetiracetam after oral and intravenous administration of a single dose to clinically normal cats. Am J Vet Res 2011; 72:1247-52. [DOI: 10.2460/ajvr.72.9.1247] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
39
|
Tokuda S, Sofue N, Ohno Y, Sasa M, Serikawa T. Inhibitory effects of levetiracetam on absence seizures in a novel absence-like epilepsy animal model, Groggy rat. Brain Res 2010; 1359:298-303. [DOI: 10.1016/j.brainres.2010.08.084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 08/24/2010] [Accepted: 08/25/2010] [Indexed: 10/19/2022]
|
40
|
Ahishali B, Kaya M, Orhan N, Arican N, Ekizoglu O, Elmas I, Kucuk M, Kemikler G, Kalayci R, Gurses C. Effects of levetiracetam on blood-brain barrier disturbances following hyperthermia-induced seizures in rats with cortical dysplasia. Life Sci 2010; 87:609-19. [DOI: 10.1016/j.lfs.2010.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 08/12/2010] [Accepted: 09/18/2010] [Indexed: 02/02/2023]
|
41
|
Coppola G, Arcieri S, D'Aniello A, Messana T, Verrotti A, Signoriello G, Pascotto A. Levetiracetam in submaximal subcutaneous pentylentetrazol-induced seizures in rats. Seizure 2010; 19:296-9. [PMID: 20399683 DOI: 10.1016/j.seizure.2010.03.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Revised: 02/28/2010] [Accepted: 03/18/2010] [Indexed: 11/25/2022] Open
Abstract
Despite anticonvulsant efficacy in animal models of generalized epilepsy, levetiracetam was not effective in the maximal subcutaneous PTZ model in mice and rats. Aim of this study was to assess the efficacy of levetiracetam (LEV) against submaximal, s.c. MET test (PTZ at the dose of 70 mg/kg) acute seizures in Wistar rats, in comparison to valproic acid (VPA). Thirty male Wistar rats (P42) were divided in three drug-treatment groups (10 rats in each group) as follows: valproic acid, levetiracetam, and controls. All animals were tested for seizure threshold at age P50. VPA (110 mg/kg) and LEV (108 mg/kg) were freshly dissolved in saline and injected i.p. in 2-3 ml/kg, 15 and 30 min, respectively, before pentylenetetrazol (PTZ) injection at the dose of 70 mg/kg. The average latency of the seizure type 3 (generalized clonic seizure with loss of righting reflexes) significantly differed between controls and the drug-treated animal groups (p < or = 0.02). The average duration of the seizure type 2 (threshold seizure) was significantly longer in both groups compared to controls (<0.02). In conclusion, LEV plays a role against seizures triggered by subcutaneous PTZ injection given at submaximal doses in rats, as demonstrated by a significant increase in duration of the seizure type 2 (threshold seizure).
Collapse
Affiliation(s)
- Giangennaro Coppola
- Clinic of Child Neuropsychiatry, Second University of Naples, Via Pansini 5, Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
42
|
Bobustuc GC, Baker CH, Limaye A, Jenkins WD, Pearl G, Avgeropoulos NG, Konduri SD. Levetiracetam enhances p53-mediated MGMT inhibition and sensitizes glioblastoma cells to temozolomide. Neuro Oncol 2010; 12:917-27. [PMID: 20525765 DOI: 10.1093/neuonc/noq044] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Antiepileptic drugs (AEDs) are frequently used to treat seizures in glioma patients. AEDs may have an unrecognized impact in modulating O(6)-methylguanine-DNA methyltransferase (MGMT), a DNA repair protein that has an important role in tumor cell resistance to alkylating agents. We report that levetiracetam (LEV) is the most potent MGMT inhibitor among several AEDs with diverse MGMT regulatory actions. In vitro, when used at concentrations within the human therapeutic range for seizure prophylaxis, LEV decreases MGMT protein and mRNA expression levels. Chromatin immunoprecipitation analysis reveals that LEV enhances p53 binding on the MGMT promoter by recruiting the mSin3A/histone deacetylase 1 (HDAC1) corepressor complex. However, LEV does not exert any MGMT inhibitory activity when the expression of either p53, mSin3A, or HDAC1 is abrogated. LEV inhibits malignant glioma cell proliferation and increases glioma cell sensitivity to the monofunctional alkylating agent temozolomide. In 4 newly diagnosed patients who had 2 craniotomies 7-14 days apart, prior to the initiation of any tumor-specific treatment, samples obtained before and after LEV treatment showed the inhibition of MGMT expression. Our results suggest that the choice of AED in patients with malignant gliomas may have an unrecognized impact in clinical practice and research trial design.
Collapse
Affiliation(s)
- George C Bobustuc
- Cancer Research Institute, M. D. Anderson Cancer Center Orlando, Orlando, Florida 32806, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Zheng Y, Moussally J, Cash SS, Karnam HB, Cole AJ. Intravenous levetiracetam in the rat pilocarpine-induced status epilepticus model: behavioral, physiological and histological studies. Neuropharmacology 2009; 58:793-8. [PMID: 20026136 DOI: 10.1016/j.neuropharm.2009.12.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Revised: 11/05/2009] [Accepted: 12/11/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE Status epilepticus is a neurological emergency associated with neuronal injury, lasting behavioral disturbance, and a high rate of mortality. Intravenous levetiracetam (LEV), an anti-epileptic drug approved to treat partial seizures, has recently been introduced. We sought to determine the effect of LEV administered intravenously in a chemoconvulsant model of status epilepticus. METHODS We examined the effect of intravenous LEV in the rat lithium-pilocarpine model of status epilepticus. Ten or 30 min after the onset of behavioral status epilepticus, animals were treated with LEV (200-1200 mg/kg i.v.) administered in a single bolus. Behavioral responses were recorded. Selected animals had continuous EEG recording before, during and after the administration of LEV. Some animals were sacrificed 24 h after the experiment and processed for histochemical assessment of neuronal injury. RESULTS When administered 30 min after the onset of behavioral epileptic seizures, transient attenuation of ictal behavior was observed in animals treated with 800 mg/kg or more of LEV. The duration of behavioral attenuation increased sharply as the dose rose to 1000 mg/kg or higher, from a mean of 4-23.6 min. When administered 10 min after seizure onset, 400 mg/kg of LEV resulted in transient ictal behavioral attenuation, and higher doses caused relatively longer periods of attenuation. Pretreatment with LEV prior to pilocarpine also delayed the onset of seizures. EEG recordings, however, showed no significant attenuation of ictal discharge. By contrast, TUNEL staining demonstrated less neuronal injury in hippocampii and other limbic structures in animals that responded behaviorally to LEV. CONCLUSIONS Intravenous administration of LEV in a chemoconvulsant model of status epilepticus results in attenuation of behavioral manifestations of seizure discharge and in reduction of neuronal injury but does not significantly alter ictal discharge recorded by EEG.
Collapse
Affiliation(s)
- Yi Zheng
- Epilepsy Research Laboratory, Epilepsy Service and Neurology Service, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
44
|
Eue S, Grumbt M, Müller M, Schulze A. Two years of experience in the treatment of status epilepticus with intravenous levetiracetam. Epilepsy Behav 2009; 15:467-9. [PMID: 19616482 DOI: 10.1016/j.yebeh.2009.05.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 05/22/2009] [Accepted: 05/23/2009] [Indexed: 11/24/2022]
Abstract
Since its introduction in 2006, 43 patients with various forms of status epilepticus (SE) have been treated with the intravenous formulation of levetiracetam (LEV) in our clinic. After ineffective treatment with benzodiazepines, intravenous LEV was administered as a short infusion (nonconvulsive and subtle SE) at a dose of 1000 or 2000 mg. In cases of convulsive SE, a fractionated injection of 1000 or 2000 mg was used. When the results for both are combined, SE could be terminated in 19 of 43 patients. Intravenous LEV was more effective in simple focal SE (3/5), complex focal SE (11/18) and myoclonic status (2/2) than in nonconvulsive (2/8) and subtle (1/2) SE. In no case was (secondarily) generalized convulsive status epilepticus (0/8) terminated. Intravenous LEV was also well-tolerated when injected in fractionated form. No severe adverse reactions were observed. As a result of this investigation, intravenous LEV in moderate doses may represent an efficacious and well-tolerated alternative for the treatment of focal (simple and complex focal) and myoclonic SE. Further investigations are needed to confirm this assumption as the patient numbers are quite low.
Collapse
Affiliation(s)
- S Eue
- Department of Neurology, Klinikum Bernburg gGmbH, Kustrenaer Strasse 98, Bernburg, Germany.
| | | | | | | |
Collapse
|
45
|
Matveeva EA, Vanaman TC, Whiteheart SW, Slevin JT. Levetiracetam prevents kindling-induced asymmetric accumulation of hippocampal 7S SNARE complexes. Epilepsia 2008; 49:1749-58. [PMID: 18513349 DOI: 10.1111/j.1528-1167.2008.01687.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE Understanding the molecular mechanisms underlying epilepsy is crucial to designing novel therapeutic regimens. This report focuses on alterations in the secretory machinery responsible for neurotransmitter (NT) release. Soluble N-ethylmaleimide sensitive factor (NSF) attachment protein receptor (SNARE) complexes mediate the fusion of synaptic vesicle and active zone membranes, thus mediating NT secretion. SNARE regulators control where and when SNARE complexes are formed. Previous studies showed an asymmetric accumulation of 7S SNARE complexes (7SC) in the ipsilateral hippocampus of kindled animals. The present studies probe the persistence of 7SC accumulation and the effect of the anticonvulsant, levetiracetam (LEV), on 7SC and SNARE regulators. METHOD Quantitative Western blotting was used to monitor levels of 7SC and SNARE regulators in hippocampal synaptosomes from kindled animals both before and after LEV treatment. RESULTS The asymmetric accumulation of 7SC is present 1-year postamygdalar kindling. The synaptic vesicle protein, synaptic vesicle protein 2 (SV2), a primary LEV-binding protein, and the SNARE regulator Tomosyn increase, whereas NSF decreases in association with this accumulation. Treatment with LEV prevented kindling-induced accumulation of SV2, but did not affect the transient increase of Tomosyn or the long-term decrease NSF. LEV treatment retarded the electrical and behavioral concomitants of amygdalar kindling coincident with a decrease in accumulation of 7SC. CONCLUSIONS The ipsilateral hippocampal accumulation of SNARE complexes is an altered molecular process associated with kindling that appears permanent. Kindling epileptogenesis alters synaptosomal levels of the SNARE regulators: NSF, SV2, and Tomosyn. Concomitant treatment with LEV reverses the kindling-induced 7SC accumulation and increase of SV2.
Collapse
Affiliation(s)
- Elena A Matveeva
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Medical Center, Lexington, Kentucky, USA
| | | | | | | |
Collapse
|
46
|
Trollmann R, Schneider J, Keller S, Strasser K, Wenzel D, Rascher W, Ogunshola OO, Gassmann M. HIF-1-regulated vasoactive systems are differentially involved in acute hypoxic stress responses of the developing brain of newborn mice and are not affected by levetiracetam. Brain Res 2008; 1199:27-36. [DOI: 10.1016/j.brainres.2007.12.069] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2007] [Revised: 12/18/2007] [Accepted: 12/20/2007] [Indexed: 01/19/2023]
|
47
|
Brandt C, Glien M, Gastens AM, Fedrowitz M, Bethmann K, Volk HA, Potschka H, Löscher W. Prophylactic treatment with levetiracetam after status epilepticus: lack of effect on epileptogenesis, neuronal damage, and behavioral alterations in rats. Neuropharmacology 2007; 53:207-21. [PMID: 17585956 DOI: 10.1016/j.neuropharm.2007.05.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Revised: 02/28/2007] [Accepted: 05/03/2007] [Indexed: 11/25/2022]
Abstract
Levetiracetam (LEV) is a structurally novel antiepileptic drug (AED) which has demonstrated a broad spectrum of anticonvulsant activities both in experimental and clinical studies. Previous experiments in the kindling model suggested that LEV, in addition to its seizure-suppressing activity, may possess antiepileptogenic or disease-modifying activity. In the present study, we evaluated this possibility by using a rat model in which epilepsy with spontaneous recurrent seizures (SRS), behavioral alterations, and hippocampal damages develop after a status epilepticus (SE) induced by sustained electrical stimulation of the basal amygdala. Two experimental protocols were used. In the first protocol, LEV treatment was started 24h after onset of electrical amygdala stimulation without prior termination of the SE. In the second protocol, the SE was interrupted after 4h by diazepam, immediately followed by onset of treatment with LEV. Treatment with LEV was continued for 8 weeks (experiment #1) or 5 weeks (experiment #2) after SE, using continuous drug administration via osmotic minipumps. The occurrence of SRS was recorded during and after treatment. In addition, the rats were tested in a battery of behavioral tests, including the elevated-plus maze and the Morris water maze. Finally, the brains of the animals were analyzed for histological lesions in the hippocampal formation. With the experimental protocols chosen for these experiments, LEV did not exert antiepileptogenic or neuroprotective activity. Furthermore, the behavioral alterations, e.g., behavioral hyperexcitability and learning deficits, in epileptic rats were not affected by treatment with LEV after SE. These data do not support the idea that administration of LEV after SE prevents or reduces the long-term alterations developing after such brain insult in rats.
Collapse
Affiliation(s)
- Claudia Brandt
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Bünteweg 17, D-30559 Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Volk HA, Matiasek LA, Luján Feliu-Pascual A, Platt SR, Chandler KE. The efficacy and tolerability of levetiracetam in pharmacoresistant epileptic dogs. Vet J 2007; 176:310-9. [PMID: 17468024 DOI: 10.1016/j.tvjl.2007.03.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2007] [Revised: 02/21/2007] [Accepted: 03/08/2007] [Indexed: 11/18/2022]
Abstract
Twenty-two dogs with idiopathic epilepsy which were pharmacoresistant to phenobarbitone and bromide were treated with levetiracetam as an add-on medication. Records of eight dogs were used retrospectively to determine a safe, efficient levetiracetam dosage. Fourteen dogs were entered into a prospective, open label, non-comparative study. After 2 months of levetiracetam oral treatment (10 mg/kg TID), 8/14 dogs responded significantly to the treatment and seizure frequency was reduced by 50%. In dogs that remained refractory, the dosage was increased to 20 mg/kg TID for 2 months. One further dog responded to levetiracetam treatment. Levetiracetam responders had a significant decrease in seizure frequency of 77% (7.9+/-5.2 to 1.8+/-1.7 seizures/month) and a decrease in seizure days per month of 68% (3.8+/-1.7 to 1.2+/-1.1 seizure days/month). However, 6/9 responders experienced an increase in seizure frequency and seizure days after 4-8 months continuing with the levetiracetam treatment at the last effective dosage. Levetiracetam was well tolerated by all dogs and sedation was the only side-effect reported in just one of the 14 dogs.
Collapse
Affiliation(s)
- Holger A Volk
- Department of Veterinary Clinical Sciences, Neurology, Royal Veterinary College, Hatfield, UK.
| | | | | | | | | |
Collapse
|
49
|
Löscher W. The pharmacokinetics of antiepileptic drugs in rats: consequences for maintaining effective drug levels during prolonged drug administration in rat models of epilepsy. Epilepsia 2007; 48:1245-58. [PMID: 17441999 DOI: 10.1111/j.1528-1167.2007.01093.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rodent models of chronic epilepsy with spontaneous recurrent seizures likely represent the closest parallel to the human condition. Such models may be best suited for therapy discovery for pharmacoresistant epilepsy and for antiepileptogenic or disease-modifying therapeutics. However, the use of such rodent models for therapy discovery creates problems with regard to maintaining effective drug levels throughout a prolonged testing period. This is particularly due to the fact that rodents such as rats and mice eliminate most drugs much more rapidly than humans. Thus, knowledge about elimination rate of a test drug in a laboratory species is essential for development of a treatment paradigm that allows maintaining adequate drug levels in the system over the period of treatment. Currently, the most popular models of epilepsy with spontaneous seizures are poststatus epilepticus models of temporal lobe epilepsy in rats. Such models are both used for studies on antiepileptogenesis and drug resistance. For validation of these models, current antiepileptic drugs (AEDs) have to be used. In this article, the elimination rates of these AEDs and their effective plasma levels in rats are reviewed as a guide for developing treatment protocols for chronic drug testing. The advantages and disadvantages of several technologies for drug delivery are discussed, and some examples for calculation of adequate treatment protocols are given. As shown in this review, because of the rapid elimination of most AEDs in rats, it is no trivial task to maintain effective steady-state AED levels in the plasma throughout the day over multiple days to ensure that there will be adequate levels in the system for the purpose of the experiment. However, the use of an adequate dosing regimen that is based on elimination rate is an absolute prerequisite when using rat models for discovery of new antiepileptogenic therapies or therapies for pharmacoresistant epilepsy, because otherwise such models may lead to erroneous conclusions about drug efficacy.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, and Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
50
|
Zhao Q, Jiang J, Li X, Lu ZS, Hu P. Single-dose pharmacokinetics of levetiracetam in healthy Chinese male subjects. Br J Clin Pharmacol 2007; 63:614-7. [PMID: 17324224 PMCID: PMC2000749 DOI: 10.1111/j.1365-2125.2006.02782.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Levetiracetam has been evaluated for epilepsy since 1992. * Pharmacokinetic studies of levetiracetam have been conducted in healthy volunteers, in adults, children and elderly patients with epilepsy, and in patients with renal and hepatic impairment. * Although this antiepileptic has been well studied in Western countries, this paper describes the first such trial of the drug in a Chinese population. WHAT THIS STUDY ADDS * Information is given on the pharmacokinetics, dose proportionality, safety and tolerability profile of levetiracetam in healthy male Chinese volunteers, and the results are compared with published data obtained in White subjects. * The pharmacokinetics and the pattern of adverse events of levetiracetam in Chinese subjects are similar to the data reported in White subjects. AIMS The main aims of this study were to evaluate the pharmacokinetics of levetiracetam in healthy male Chinese volunteers and to assess the dose proportionality between the 500-mg and 1500-mg single doses. METHODS This was a randomized, single-centre, single-dose, two-way crossover study. Twenty-six healthy male Chinese subjects were enrolled. All subjects received a single dose of 500 mg or 1500 mg levetiracetam tablet(s) on the dosing day, and the wash-out period was 7 days. Blood was obtained for a 36-h pharmacokinetic evaluation. RESULTS Following single-dose administration of 500 mg and 1500 mg of levetiracetam, the median t(max) was 0.5 and 0.5 h; t(1/2) was 7.3 +/- 0.8 and 7.3 +/- 0.7 h; C(max) was 13.6 +/- 3.2 and 47.1 +/- 12.1 microg ml(-1); AUC(0-infinity) was 109.3 +/- 14.1 and 340.4 +/- 50.6 microg h(-1) ml(-1); and AUC(0-t) was 105.7 +/- 13.3 and 329.0 +/- 47.9 microg h(-1) ml(-1), respectively. CONCLUSIONS Both C(max) and AUCs were dose-proportional over the range of 500-1500 mg. The pharmacokinetic data obtained in these Chinese subjects were similar to the historical data from a matched group of White subjects.
Collapse
Affiliation(s)
- Qian Zhao
- Clinical Pharmacology Research Centre, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | | | | | | |
Collapse
|