1
|
Bertram EH, Dudek FE. Addressing the problems of treatment failure in epilepsy: You cannot fix what you do not understand. Epilepsia 2024; 65:2248-2254. [PMID: 38878057 DOI: 10.1111/epi.18044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 08/10/2024]
Affiliation(s)
- Edward H Bertram
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah, 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
|
Mastrocco A, Prittie J, West C, Clark M. A review of the pharmacology and clinical applications of levetiracetam in dogs and cats. J Vet Emerg Crit Care (San Antonio) 2024; 34:9-22. [PMID: 37987141 DOI: 10.1111/vec.13355] [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: 07/17/2022] [Revised: 09/15/2022] [Accepted: 10/18/2022] [Indexed: 11/22/2023]
Abstract
OBJECTIVE To review and summarize the pharmacology of the antiepileptic drug (AED), levetiracetam (LEV), and to discuss its clinical utility in dogs and cats. DATA SOURCES Veterinary and human peer-reviewed medical literature and the authors' clinical experience. SUMMARY LEV is an AED with mechanisms of action distinct from those of other AEDs. In people and small animals, LEV exhibits linear kinetics, excellent oral bioavailability, and minimal drug-drug interactions. Serious side effects are rarely reported in any species. LEV use is gaining favor for treating epilepsy in small animals and may have wider clinical applications in patients with portosystemic shunts, neuroglycopenia, and traumatic brain injury. In people, LEV may improve cognitive function in patients with dementia. CONCLUSION LEV is a well-tolerated AED with well-documented efficacy in human patients. Although its use is becoming more common in veterinary medicine, its role as a first-line monotherapy in small animal epileptics remains to be determined. This review of the human and animal literature regarding LEV describes its role in epileptic people and animals as well as in other disease states and provides recommendations for clinical usage.
Collapse
Affiliation(s)
- Alicia Mastrocco
- Department of Emergency and Critical Care, The Animal Medical Center, New York, New York, USA
| | - Jennifer Prittie
- Department of Emergency and Critical Care, The Animal Medical Center, New York, New York, USA
| | - Chad West
- Department of Neurology, The Animal Medical Center, New York, New York, USA
| | - Melissa Clark
- Department of Internal Medicine, Gulf Coast Veterinary Specialists, Houston, Texas, USA
| |
Collapse
|
4
|
Kusakabe K, Inoue A, Watanabe H, Nakamura Y, Nishikawa M, Ohtsuka Y, Ogura M, Shigekawa S, Taniwaki M, Kitazawa R, Kunieda T. Perioperative perampanel administration for early seizure prophylaxis in brain tumor patients. Surg Neurol Int 2023; 14:287. [PMID: 37680915 PMCID: PMC10481804 DOI: 10.25259/sni_495_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023] Open
Abstract
Background The efficacy of perioperative prophylactic antiepileptic drug therapy in "seizure-naïve" patients with brain tumor, including glioblastoma (GBM), remains controversial. This study investigated whether perampanel (PER) is effective and safe for preventing perioperative onset of epileptic seizures, so-called early seizure, in patients with brain tumors. Methods Forty-five patients underwent tumor resection through craniotomy for a primary supratentorial brain tumor at Ehime University Hospital between April 2021 and July 2022. PER was administered from the 1st to the 6th day after surgery for seizure prophylaxis. Occurrence of early seizure, hematological toxicities, and various side effects were recorded on postoperative days 7 and 14. In addition, the clinical course of these patients was compared with 42 brain tumor patients under the same treatment protocol who received levetiracetam (LEV) for seizure prophylaxis between April 2017 and October 2018. Results In 45 patients with brain tumor, including GBM, who received PER administration, no early seizures were identified within 7 days postoperatively. No adverse drug reactions such as hematological toxicity, liver or kidney dysfunction, or exanthematous drug eruption were observed in any cases. As side effects, somnolence was reported in 14 patients (31.1%), vertigo in 3 patients (6.7%), and headache in 3 patients (6.7%). Although somnolence and vertigo were difficult to assess in the case of intraparenchymal tumors, particularly GBM, these side effects were not identified in patients with extraparenchymal tumors such as meningiomas, epidermoid cysts, and pituitary adenomas. In addition, no significant differences were identified compared to patients who received LEV. Conclusion The efficacy and safety of PER in preventing early seizures among patients with brain tumors were retrospectively evaluated. Perioperative administration of PER to patients with brain tumors may reduce the risk of early seizures without incurring serious side effects, showing no significant differences compared to patients who received LEV.
Collapse
Affiliation(s)
- Kosuke Kusakabe
- Department of Neurosurgery, Ehime University School of Medicine, Toon, Japan
| | - Akihiro Inoue
- Department of Neurosurgery, Ehime University School of Medicine, Toon, Japan
| | - Hideaki Watanabe
- Department of Neurosurgery, Ehime University School of Medicine, Toon, Japan
| | - Yawara Nakamura
- Department of Neurosurgery, Ehime University School of Medicine, Toon, Japan
| | - Masahiro Nishikawa
- Department of Neurosurgery, Ehime University School of Medicine, Toon, Japan
| | - Yoshihiro Ohtsuka
- Department of Neurosurgery, Ehime University School of Medicine, Toon, Japan
| | - Masahiro Ogura
- Department of Rehabilitation, Ehime University Hospital, Toon, Japan
| | - Seiji Shigekawa
- Department of Neurosurgery, Ehime University School of Medicine, Toon, Japan
| | - Mashio Taniwaki
- Division of Diagnostic Pathology, Ehime University Hospital, Toon, Japan
| | - Riko Kitazawa
- Division of Diagnostic Pathology, Ehime University Hospital, Toon, Japan
| | - Takeharu Kunieda
- Department of Neurosurgery, Ehime University School of Medicine, Toon, Japan
| |
Collapse
|
5
|
Coles LD, Saletti PG, Lisgaras CP, Casillas-Espinosa PM, Liu W, Li Q, Jones NC, Shultz S, Ali I, Brady R, Yamakawa G, Hudson M, Silva J, Braine E, Mishra U, Cloyd JC, O'Brien TJ, Moshé SL, Galanopoulou AS. Levetiracetam Pharmacokinetics and Brain Uptake in a Lateral Fluid Percussion Injury Rat Model. J Pharmacol Exp Ther 2023; 386:259-265. [PMID: 37316328 PMCID: PMC10353071 DOI: 10.1124/jpet.122.001377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 04/08/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023] Open
Abstract
Post-traumatic epilepsy (PTE) occurs in some patients after moderate/severe traumatic brain injury (TBI). Although there are no approved therapies to prevent epileptogenesis, levetiracetam (LEV) is commonly given for seizure prophylaxis due to its good safety profile. This led us to study LEV as part of the Epilepsy Bioinformatics Study for Antiepileptogenic Therapy (EpiBioS4Rx) Project. The objective of this work is to characterize the pharmacokinetics (PK) and brain uptake of LEV in naïve control rats and in the lateral fluid percussion injury (LFPI) rat model of TBI after either single intraperitoneal doses or a loading dose followed by a 7-day subcutaneous infusion. Sprague-Dawley rats were used as controls and for the LFPI model induced at the left parietal region using injury parameters optimized for moderate/severe TBI. Naïve and LFPI rats received either a bolus injection (intraperitoneal) or a bolus injection followed by subcutaneous infusion over 7 days. Blood and parietal cortical samples were collected at specified time points throughout the study. LEV concentrations in plasma and brain were measured using validated high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) methods. Noncompartmental analysis and a naive-pooled compartmental PK modeling approach were used. Brain-to-plasma ratios ranged from 0.54 to 1.4 to 1. LEV concentrations were well fit by one-compartment, first-order absorption PK models with a clearance of 112 ml/h per kg and volume of distribution of 293 ml/kg. The single-dose pharmacokinetic data were used to guide dose selection for the longer-term studies, and target drug exposures were confirmed. Obtaining LEV PK information early in the screening phase allowed us to guide optimal treatment protocols in EpiBioS4Rx. SIGNIFICANCE STATEMENT: The characterization of levetiracetam pharmacokinetics and brain uptake in an animal model of post-traumatic epilepsy is essential to identify target concentrations and guide optimal treatment for future studies.
Collapse
Affiliation(s)
- Lisa D Coles
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Patricia G Saletti
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Christos Panagiotis Lisgaras
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Pablo M Casillas-Espinosa
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Wei Liu
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Qianyun Li
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Nigel C Jones
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Sandy Shultz
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Idrish Ali
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Rhys Brady
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Glenn Yamakawa
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Matt Hudson
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Juliana Silva
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Emma Braine
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Usha Mishra
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - James C Cloyd
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Terence J O'Brien
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Solomon L Moshé
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| | - Aristea S Galanopoulou
- University of Minnesota Twin Cities, Minneapolis, Minnesota (L.D.C., U.M., J.C.C.); Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy (P.G.S., C.P.L., W.L., Q.L., S.L.M., A.S.G.), Isabelle Rapin Division of Child Neurology (S.L.M., A.S.G.), Dominick P. Purpura Department of Neuroscience (S.L.M., A.S.G.), and Department of Pediatrics (S.L.M.), Albert Einstein College of Medicine, Bronx, New York; Department of Neuroscience, Monash University, Melbourne, Australia (P.M.C.-E., S.S., R.B., G.Y., M.H., J.S., E.B., T.J.O.); and The University of Melbourne, Parkville, Australia (R.B., G.Y., M.H., J.S., E.B., T.J.O.)
| |
Collapse
|
6
|
Pawlik M, Czarnecka AM, Kołodziej M, Skowrońska K, Węgrzynowicz M, Podgajna M, Czuczwar SJ, Albrecht J. Attenuation of initial pilocarpine-induced electrographic seizures by methionine sulfoximine pretreatment tightly correlates with the reduction of extracellular taurine in the hippocampus. Epilepsia 2023; 64:1390-1402. [PMID: 36808593 DOI: 10.1111/epi.17554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/20/2023]
Abstract
OBJECTIVE Initiation and development of early seizures by chemical stimuli is associated with brain cell swelling resulting in edema of seizure-vulnerable brain regions. We previously reported that pretreatment with a nonconvulsive dose of glutamine (Gln) synthetase inhibitor methionine sulfoximine (MSO) mitigates the intensity of initial pilocarpine (Pilo)-induced seizures in juvenile rats. We hypothesized that MSO exerts its protective effect by preventing the seizure-initiating and seizure-propagating increase of cell volume. Taurine (Tau) is an osmosensitive amino acid, whose release reflects increased cell volume. Therefore, we tested whether the poststimulus rise of amplitude of Pilo-induced electrographic seizures and their attenuation by MSO are correlated with the release of Tau from seizure-affected hippocampus. METHODS Lithium-pretreated animals were administered MSO (75 mg/kg ip) 2.5 h before the induction of convulsions by Pilo (40 mg/kg ip). Electroencephalographic (EEG) power was analyzed during 60 min post-Pilo, at 5-min intervals. Extracellular accumulation of Tau (eTau) served as a marker of cell swelling. eTau, extracellular Gln (eGln), and extracellular glutamate (eGlu) were assayed in the microdialysates of the ventral hippocampal CA1 region collected at 15-min intervals during the whole 3.5-h observation period. RESULTS The first EEG signal became apparent at ~10 min post-Pilo. The EEG amplitude across most frequency bands peaked at ~40 min post-Pilo, and showed strong (r ~ .72-.96) temporal correlation with eTau, but no correlation with eGln or eGlu. MSO pretreatment delayed the first EEG signal in Pilo-treated rats by ~10 min, and depressed the EEG amplitude across most frequency bands, to values that remained strongly correlated with eTau (r > .92) and moderately correlated (r ~ -.59) with eGln, but not with eGlu. SIGNIFICANCE Strong correlation between attenuation of Pilo-induced seizures and Tau release indicates that the beneficial effect of MSO is due to the prevention of cell volume increase concurrent with the onset of seizures.
Collapse
Affiliation(s)
- Marek Pawlik
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Maria Czarnecka
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Marcin Kołodziej
- Institute of Theory of Electrical Engineering, Measurement, and Information Systems, Warsaw University of Technology, Warsaw, Poland
| | - Katarzyna Skowrońska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Michał Węgrzynowicz
- Laboratory of Molecular Basis of Neurodegeneration, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Martyna Podgajna
- Laboratory of Molecular Basis of Neurodegeneration, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | | | - Jan Albrecht
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
7
|
Wang S, Wu X, Xue T, Song Z, Tan X, Sun X, Wang Z. Efficacy and safety of levetiracetam versus valproate in patients with established status epilepticus: A systematic review and meta-analysis. Heliyon 2023; 9:e13380. [PMID: 36816301 PMCID: PMC9932733 DOI: 10.1016/j.heliyon.2023.e13380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 12/13/2022] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Objective Status epilepticus (SE) is a common neurological emergency that is defined as a prolonged seizure or a series of seizures which often leads to irreversible damage. Levetiracetam (LEV) and valproate (VPA) are second-line anti-seizure drugs that are frequently used in patients with established SE (ESE). This meta-analysis compared the efficacy and safety of LEV and VPA for the treatment of ESE. Method MEDLINE, EMBASE, Central Register of Controlled Trials (CENTRAL), and clinicaltrials.gov were searched by two authors, which identified six randomized controlled trials (RCTs) that compared LEV and VPA for ESE. Results The six RCTs included 1213 patients (LEV group, n = 593; VPA group, n = 620). Integrated patient data information display LEV was not superior to VPA in terms of clinical seizure termination (63.55% vs. 64.08%, respectively; relative risk [RR] = 1.03, 95% confidence interval [CI] = 0.94-1.11, p = 0.55), with no significant differences between LEV and VPA in terms of good functional outcome at discharge (Glasgow Outcome Scale [GOS] = 4 or 5), intensive care unit (ICU) admission, adverse events, and mortality. There was no statistically significant difference between the two drugs in different age groups. Previous multicenter studies have demonstrated that VPA was slightly more effective than LEV, whereas single-center studies showed the opposite results. In addition, LEV and VPA had similar rates of clinical seizure termination, ICU admission, and adverse events between the age subgroups (ages <18 and >18 years). Conclusions Levetiracetam (LEV) was not superior to valproate (VPA) in terms of efficacy or safety outcomes. In addition, children (<18 years) and adults (>18 years) might have similar responses to LEV and VPA. Additional RCTs are required to verify our results.
Collapse
Affiliation(s)
- Shixin Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xin Wu
- Department of Neurosurgery, Suzhou Ninth People's Hospital, Suzhou, Jiangsu Province, China
| | - Tao Xue
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhaoming Song
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xin Tan
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu Province, China
| | - Xiaoou Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China,Corresponding author. Department of Neurosurgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China,Corresponding author. Department of Neurosurgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China.
| |
Collapse
|
8
|
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; 15:475. [PMID: 35455472 PMCID: PMC9030752 DOI: 10.3390/ph15040475] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [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;
| | | | - Luz Adriana Pichardo-Macías
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| |
Collapse
|
9
|
Xing H, Xu S, Xie X, Wang Y, Lu C, Han X. Levetiracetam induction of theta frequency oscillations in rodent hippocampus in vitro. Can J Physiol Pharmacol 2020; 98:725-732. [PMID: 32516556 DOI: 10.1139/cjpp-2019-0727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Levetiracetam (LEV) has been demonstrated to improve cognitive function. Hippocampal theta rhythm (4-12 Hz) is associated with a variety of cognitively related behaviors, such as exploration in both humans and animal models. We investigated the effects of LEV on the theta rhythm in the rat hippocampal CA3 in hippocampal slices in vitro. We found that LEV increased the theta power in a dose-dependent manner. The increase in theta power can be blocked by GABAA receptor (GABAAR) or NMDA receptor (NMDAR) antagonists but not by AMPA receptor antagonist, indicating the involvement of GABAAR and NMDAR in the induction of theta activity. Interestingly, LEV enhancement of theta power can be also blocked by taurine or GABA-A agonist THIP, indicating that LEV induction of theta may be related to the indirect boosting of GABA action via reduction of extrasynaptic GABAAR activation. Furthermore, the increased theta power can be partially reduced by the mACh receptor (mAChR) antagonist atropine but not by nACh receptor antagonists, suggesting that mAChR activation provides excitatory input into local network responsible for LEV-induced theta. Our study demonstrated that LEV induced a novel theta oscillation in vitro, which may have implications in the treatment of the neuronal disorders with impaired theta oscillation and cognitive function.
Collapse
Affiliation(s)
- Hang Xing
- Key Lab of Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Henan, 453000, P.R. China.,Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - Sihan Xu
- Key Lab of Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Henan, 453000, P.R. China
| | - Xin'e Xie
- Key Lab of Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Henan, 453000, P.R. China
| | - Yuan Wang
- Key Lab of Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Henan, 453000, P.R. China
| | - Chengbiao Lu
- Key Lab of Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Henan, 453000, P.R. China
| | - Xiong Han
- Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| |
Collapse
|
10
|
Gupta M, Bogdanowicz T, Reed MA, Barden CJ, Weaver DF. The Brain Exposure Efficiency (BEE) Score. ACS Chem Neurosci 2020; 11:205-224. [PMID: 31815431 DOI: 10.1021/acschemneuro.9b00650] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The blood-brain barrier (BBB), composed of microvascular tight junctions and glial cell sheathing, selectively controls drug permeation into the central nervous system (CNS) by either passive diffusion or active transport. Computational techniques capable of predicting molecular brain penetration are important to neurological drug design. A novel prediction algorithm, termed the Brain Exposure Efficiency Score (BEE), is presented. BEE addresses the need to incorporate the role of trans-BBB influx and efflux active transporters by considering key brain penetrance parameters, namely, steady state unbound brain to plasma ratio of drug (Kp,uu) and dose normalized unbound concentration of drug in brain (Cu,b). BEE was devised using quantitative structure-activity relationships (QSARs) and molecular modeling studies on known transporter proteins and their ligands. The developed algorithms are provided as a user-friendly open source calculator to assist in optimizing a brain penetrance strategy during the early phases of small molecule molecular therapeutic design.
Collapse
Affiliation(s)
- Mayuri Gupta
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Thomas Bogdanowicz
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Mark A. Reed
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Christopher J. Barden
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - Donald F. Weaver
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario M5S 3M2 Canada
| |
Collapse
|
11
|
Giri TK, Ghosh B, Bose P, Saha S, Sarkar A. Extraction of levetiracetam for therapeutic drug monitoring by transdermal reverse iontophoresis. Eur J Pharm Sci 2019; 128:54-60. [PMID: 30468869 DOI: 10.1016/j.ejps.2018.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/26/2018] [Accepted: 11/19/2018] [Indexed: 10/27/2022]
Abstract
Recently, transdermal reverse iontophoresis across the skin has been investigated as a novel technology for the purpose of diagnosis as well as therapeutic drug monitoring. Accordingly, the objective of this study was to investigate ex vivo and in vivo transdermal extraction of levetiracetam, an antiepileptic drug, across the pig ear skin by reverse iontophoresis. Reverse iontophoresis experiments were performed using three chambered diffusion cells. Extractions profiles were generated in phosphate buffers at different current intensities, pH and ionic strength as well donor drug concentrations. This was followed by ex vivo extraction in gels and in vivo extractions using New Zealand rabbits. Results indicate that levetiracetam was extracted at both anode and cathode. Flux values were unaffected by increase in current intensities (0.5 mA and 0.6 mA) but affected by pH and ionic strength. Neither in cathodal nor in anodal extraction, flux values did show a proportional relationship with the donor drug concentration. At low and medium concentration levels, flux values did not show any major change but the extraction flux at high donor concentration was much higher. In contrast, in vivo experiment with rabbits resulted in wide variation of fluxes with very high fluxes recorded at the cathodal end. Reasons attributed to this difference may include lower current intensity, and species variation. The most significant finding of this study is that measurable amounts of the levetiracetam were extracted at both the ends indicating its feasibility for non-invasive drug monitoring.
Collapse
Affiliation(s)
- Tapan Kumar Giri
- NSHM Knowledge Campus, Kolkata-Group of Institutions, 124 BL Saha Road, Kolkata 700053, West Bengal, India
| | - Bijaya Ghosh
- NSHM Knowledge Campus, Kolkata-Group of Institutions, 124 BL Saha Road, Kolkata 700053, West Bengal, India.
| | - Preeta Bose
- NSHM Knowledge Campus, Kolkata-Group of Institutions, 124 BL Saha Road, Kolkata 700053, West Bengal, India
| | - Sumana Saha
- NSHM Knowledge Campus, Kolkata-Group of Institutions, 124 BL Saha Road, Kolkata 700053, West Bengal, India
| | - Arijit Sarkar
- NSHM Knowledge Campus, Kolkata-Group of Institutions, 124 BL Saha Road, Kolkata 700053, West Bengal, India
| |
Collapse
|
12
|
Reif PS, Männer A, Willems LM, Kay L, Zöllner JP, Klein KM, Rosenow F, Strzelczyk A. Intravenous lacosamide for treatment of absence status epilepticus in genetic generalized epilepsy: A case report and review of literature. Acta Neurol Scand 2018; 138:259-262. [PMID: 29633241 DOI: 10.1111/ane.12935] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Nearly 10 years after its introduction into the market, the significance of lacosamide in genetic generalized epilepsies is still unclear. Its new mode of action may qualify lacosamide as a therapeutic agent in this entity, but only a limited number of cases have been published so far. AIM To describe the efficacy of lacosamide as treatment in a patient with the absence status epilepticus. METHOD We report on a 28-year-old woman with genetic generalized epilepsy who suffered recurrent absence status epilepticus during video-EEG-monitoring. After treatment failure of first- and second-line medication, lacosamide was administered. The outcome in this patient was evaluated, and a systematic literature review was performed for the use of lacosamide in the absence status epilepticus. RESULTS After application of 400 mg lacosamide intravenously, the absence status epilepticus terminated within 30 minutes. No further seizures or epileptiform discharges reoccurred until the end of video-EEG-Monitoring 3 days later. CONCLUSIONS The role of lacosamide as a therapeutic option in patients with the absence status epilepticus is unclear. Only two cases have been reported so far with conflicting results. Further randomized controlled studies are required to validate the relevance of lacosamide as treatment for status epilepticus in genetic generalized and the absence epilepsy.
Collapse
Affiliation(s)
- P S Reif
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University, Frankfurt am Main, Germany
| | - A Männer
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University, Frankfurt am Main, Germany
| | - L M Willems
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University, Frankfurt am Main, Germany
| | - L Kay
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University, Frankfurt am Main, Germany
| | - J P Zöllner
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University, Frankfurt am Main, Germany
| | - K M Klein
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University, Frankfurt am Main, Germany
| | - F Rosenow
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University, Frankfurt am Main, Germany
| | - A Strzelczyk
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
13
|
Effect of levetiracetam on extracellular amino acid levels in the dorsal hippocampus of rats with temporal lobe epilepsy. Epilepsy Res 2018; 140:111-119. [PMID: 29331845 DOI: 10.1016/j.eplepsyres.2018.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/08/2017] [Accepted: 01/03/2018] [Indexed: 12/23/2022]
Abstract
Levetiracetam (LEV) is an anticonvulsant drug with a unique mechanism of action that is not completely understood. However, its activity profile may involve effects on excitatory and/or inhibitory neurotransmission since the primary target of LEV, synaptic vesicle protein 2A, is ubiquitously expressed in all types of synaptic vesicles. Therefore, the objective of the present study was to explore the effect of LEV (300 mg/kg/day for one week, administered via osmotic mini-pumps) on neurotransmitter release and its probable selective effect on extracellular gamma-amino butyric acid (GABA), glutamate (Glu), aspartate (Asp), glutamine (Gln), taurine (Tau) and glycine (Gly) concentrations (using in vivo microdialysis under basal and high-K+ conditions) in the dorsal hippocampus (DH), a region that undergoes major synaptic changes during epilepsy. Epileptic rats developed clear signs of hyperexcitability, i.e., an elevated Glu/GABA ratio in the DH. The LEV concentration in blood after 7 days of treatment was within the therapeutic range. In contrast, LEV was not detected four days after mini-pump removal (washout period). Furthermore, LEV restored the Glu/GABA ratio to approximately the control level and significantly increased the GABA concentration after the initiation of high-K+ conditions. Based on these data, LEV treatment restored the lost balance between the excitatory and inhibitory systems under basal conditions. Moreover, LEV showed a selective effect by preferentially increasing vesicular release of GABA, a mechanism by which LEV could reduce epileptic seizures.
Collapse
|
14
|
Kudlacek J, Chvojka J, Posusta A, Kovacova L, Hong SB, Weiss S, Volna K, Marusic P, Otahal J, Jiruska P. Lacosamide and Levetiracetam Have No Effect on Sharp-Wave Ripple Rate. Front Neurol 2017; 8:687. [PMID: 29312120 PMCID: PMC5742623 DOI: 10.3389/fneur.2017.00687] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 12/01/2017] [Indexed: 01/08/2023] Open
Abstract
Pathological high-frequency oscillations are a novel marker used to improve the delineation of epileptogenic tissue and, hence, the outcome of epilepsy surgery. Their practical clinical utilization is curtailed by the inability to discriminate them from physiological oscillations due to frequency overlap. Although it is well documented that pathological HFOs are suppressed by antiepileptic drugs (AEDs), the effect of AEDs on normal HFOs is not well known. In this experimental study, we have explored whether physiological HFOs (sharp-wave ripples) of hippocampal origin respond to AED treatment. The results show that application of a single dose of levetiracetam or lacosamide does not reduce the rate of sharp-wave ripples. In addition, it seems that these new generation drugs do not negatively affect the cellular and network mechanisms involved in sharp-wave ripple generation, which may provide a plausible explanation for the absence of significant negative effects on cognitive functions of these drugs, particularly on memory.
Collapse
Affiliation(s)
- Jan Kudlacek
- Department of Developmental Epileptology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czechia.,Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czechia
| | - Jan Chvojka
- Department of Developmental Epileptology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czechia.,Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czechia
| | - Antonin Posusta
- Department of Developmental Epileptology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czechia
| | - Lubica Kovacova
- Department of Developmental Epileptology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czechia
| | - Seung Bong Hong
- Department of Neurology, Samsung Medical Center, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University School of Medicine, Seoul, South Korea.,Samsung Biomedical Research Institute, Seoul, South Korea
| | - Shennan Weiss
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Kamila Volna
- Department of Neurology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Petr Marusic
- Department of Neurology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Jakub Otahal
- Department of Developmental Epileptology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czechia
| | - Premysl Jiruska
- Department of Developmental Epileptology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
15
|
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: 5.3] [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
|
16
|
Zou H, Hurwitz M, Fowler L, Wagner AK. Abbreviated levetiracetam treatment effects on behavioural and histological outcomes after experimental TBI. Brain Inj 2016; 29:78-85. [PMID: 25255156 DOI: 10.3109/02699052.2014.955528] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Long-term prophylactic treatment with levetiracetam (LEV) has multiple neuroprotective effects in a traumatic brain injury (TBI) rat model. Although a rational time-frame of seizure prophylactic treatment with LEV for after TBI is not well established, clinical prophylaxis with LEV often includes treatment duration similar to clinical treatment guidelines with Phenytoin. Thus, this study investigated the effects of abbreviated LEV treatment on behavioural function and histological evidence of neuroprotection. RESEARCH DESIGN Pre-clinical trial of abbreviated LEV dosing in an experimental model of TBI Methods: After either controlled cortical impact (CCI) injury or sham surgery, rats received three 50 mg kg(-1) doses over 24 hours or vehicle. After injury/sham surgery, beam performance, spatial learning, contusion volume size and hippocampal neuron survival were assessed. RESULTS Abbreviated LEV did not improve motor or cognitive performance after TBI. Further, abbreviated LEV did not improve hippocampal neuron sparing or contusion volumes compared with vehicle controls. CONCLUSIONS Together with previous work assessing daily LEV treatment, these results suggest that longer-term therapy may be required to confer beneficial effects within these domains. These findings may guide (1) future experimental studies assessing minimal effective dosing for neuroprotection and anti-epileptogenesis and (2) treatment guideline updates for seizure prophylaxis post-TBI.
Collapse
Affiliation(s)
- Huichao Zou
- a Department of Physical Medicine and Rehabilitation and
| | | | | | | |
Collapse
|
17
|
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: 121] [Impact Index Per Article: 12.1] [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
|
18
|
Donovan MD, Boylan GB, Murray DM, Cryan JF, Griffin BT. Treating disorders of the neonatal central nervous system: pharmacokinetic and pharmacodynamic considerations with a focus on antiepileptics. Br J Clin Pharmacol 2015; 81:62-77. [PMID: 26302437 DOI: 10.1111/bcp.12753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 08/05/2015] [Accepted: 08/13/2015] [Indexed: 12/31/2022] Open
Abstract
A major consideration in the treatment of neonatal disorders is that the selected drug, dose and dosage frequency is safe, effective and appropriate for the intended patient population. Thus, a thorough knowledge of the pharmacokinetics and pharmacodynamics of the chosen drug within the patient population is essential. In paediatric and neonatal populations two additional challenges can often complicate drug treatment - the inherently greater physiological variability, and a lack of robust clinical evidence of therapeutic range. There has traditionally been an overreliance in paediatric medicine on extrapolating doses from adult values by adjusting for bodyweight or body surface area, but many other sources of variability exist which complicate the choice of dose in neonates. The lack of reliable drug dosage data in neonates has been highlighted by regulatory authorities, as only ~50% of the most commonly used paediatric medicines have been examined in a paediatric population. Moreover, there is a paucity of information on the pharmacokinetic parameters which affect drug concentrations in different body tissues, and pharmacodynamic responses to drugs in the neonate. Thus, in the present review, we draw attention to the main pharmacokinetic factors that influence the unbound brain concentration of neuroactive drugs. Moreover, the pharmacodynamic differences between neonates and adults that affect the activity of centrally-acting therapeutic agents are briefly examined, with a particular emphasis on antiepileptic drugs.
Collapse
Affiliation(s)
- Maria D Donovan
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Geraldine B Boylan
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,Irish Centre for Fetal and Neonatal Translational Research, University College Cork and Cork University Maternity Hospital, Cork, Ireland
| | - Deirdre M Murray
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Brendan T Griffin
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
19
|
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: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
20
|
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
|
21
|
Deshpande LS, Delorenzo RJ. Mechanisms of levetiracetam in the control of status epilepticus and epilepsy. Front Neurol 2014; 5:11. [PMID: 24550884 PMCID: PMC3907711 DOI: 10.3389/fneur.2014.00011] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/17/2014] [Indexed: 11/27/2022] Open
Abstract
Status epilepticus (SE) is a major clinical emergency that is associated with high mortality and morbidity. SE causes significant neuronal injury and survivors are at a greater risk of developing acquired epilepsy and other neurological morbidities, including depression and cognitive deficits. Benzodiazepines and some anticonvulsant agents are drugs of choice for initial SE management. Despite their effectiveness, over 40% of SE cases are refractory to the initial treatment with two or more medications. Thus, there is an unmet need of developing newer anti-SE drugs. Levetiracetam (LEV) is a widely prescribed anti-epileptic drug that has been reported to be used in SE cases, especially in benzodiazepine-resistant SE or where phenytoin cannot be used due to allergic side-effects. Levetiracetam’s non-classical anti-epileptic mechanisms of action, favorable pharmacokinetic profile, general lack of central depressant effects, and lower incidence of drug interactions contribute to its use in SE management. This review will focus on LEV’s unique mechanism of action that makes it a viable candidate for SE treatment.
Collapse
Affiliation(s)
| | - Robert J Delorenzo
- Department of Neurology, Virginia Commonwealth University , Richmond, VA , USA ; Department of Pharmacology and Toxicology, Virginia Commonwealth University , Richmond, VA , USA ; Department of Biochemistry, Virginia Commonwealth University , Richmond, VA , USA
| |
Collapse
|
22
|
|
23
|
Oja SS, Saransaari P. Taurine and epilepsy. Epilepsy Res 2013; 104:187-94. [DOI: 10.1016/j.eplepsyres.2013.01.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/19/2012] [Accepted: 01/18/2013] [Indexed: 10/27/2022]
|
24
|
Fukuyama K, Tanahashi S, Nakagawa M, Yamamura S, Motomura E, Shiroyama T, Tanii H, Okada M. Levetiracetam inhibits neurotransmitter release associated with CICR. Neurosci Lett 2012; 518:69-74. [DOI: 10.1016/j.neulet.2012.03.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 11/27/2022]
|
25
|
Olah E, Bacsoi G, Fekete J, Sharma VK. Determination of ng/mL Levetiracetam using Ultra-High-Performance Liquid Chromatography-Photodiode Absorbance. J Chromatogr Sci 2012; 50:253-8. [DOI: 10.1093/chromsci/bmr053] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
26
|
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: 4.2] [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
|
27
|
Measurement of the pharmacokinetics and pharmacodynamics of neuroactive compounds. Neurobiol Dis 2010; 37:38-47. [DOI: 10.1016/j.nbd.2009.09.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 09/29/2009] [Accepted: 09/30/2009] [Indexed: 11/24/2022] Open
|
28
|
Ishimaru Y, Chiba S, Serikawa T, Sasa M, Inaba H, Tamura Y, Ishimoto T, Takasaki H, Sakamoto K, Yamaguchi K. Effects of levetiracetam on hippocampal kindling in Noda epileptic rats. Brain Res 2010; 1309:104-9. [DOI: 10.1016/j.brainres.2009.10.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 10/16/2009] [Accepted: 10/22/2009] [Indexed: 10/20/2022]
|
29
|
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: 3.1] [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
|
30
|
Ueda Y, Doi T, Takaki M, Nagatomo K, Nakajima A, Willmore LJ. Levetiracetam enhances endogenous antioxidant in the hippocampus of rats: In vivo evaluation by brain microdialysis combined with ESR spectroscopy. Brain Res 2009; 1266:1-7. [PMID: 19268434 DOI: 10.1016/j.brainres.2009.02.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 01/05/2009] [Accepted: 02/21/2009] [Indexed: 10/21/2022]
|
31
|
Tong X, Ratnaraj N, Patsalos PN. Vigabatrin extracellular pharmacokinetics and concurrent γ-aminobutyric acid neurotransmitter effects in rat frontal cortex and hippocampus using microdialysis. Epilepsia 2009; 50:174-83. [DOI: 10.1111/j.1528-1167.2008.01863.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Alves G, Figueiredo I, Castel-Branco M, Lourenço N, Falcão A, Caramona M, Soares-da-Silva P. Disposition of eslicarbazepine acetate in the mouse after oral administration. Fundam Clin Pharmacol 2008; 22:529-36. [DOI: 10.1111/j.1472-8206.2008.00617.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
33
|
Epstein CM, Girard-Siqueira L, Ehrenberg JA. Prolonged neurophysiologic effects of levetiracetam after oral administration in humans. Epilepsia 2008; 49:1169-73. [DOI: 10.1111/j.1528-1167.2008.01562.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
34
|
Nagarkatti N, Deshpande LS, DeLorenzo RJ. Levetiracetam inhibits both ryanodine and IP3 receptor activated calcium induced calcium release in hippocampal neurons in culture. Neurosci Lett 2008; 436:289-93. [PMID: 18406528 DOI: 10.1016/j.neulet.2008.02.076] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Revised: 02/01/2008] [Accepted: 02/07/2008] [Indexed: 10/22/2022]
Abstract
Epilepsy affects approximately 1% of the population worldwide, and there is a pressing need to develop new anti-epileptic drugs (AEDs) and understand their mechanisms of action. Levetiracetam (LEV) is a novel AED and despite its increasingly widespread clinical use, its mechanism of action is as yet undetermined. Intracellular calcium ([Ca2+]i) regulation by both inositol 1,4,5-triphosphate receptors (IP3R) and ryanodine receptors (RyR) has been implicated in epileptogenesis and the maintenance of epilepsy. To this end, we investigated the effect of LEV on RyR and IP3R activated calcium-induced calcium release (CICR) in hippocampal neuronal cultures. RyR-mediated CICR was stimulated using the well-characterized RyR activator, caffeine. Caffeine (10mM) caused a significant increase in [Ca2+]i in hippocampal neurons. Treatment with LEV (33 microM) prior to stimulation of RyR-mediated CICR by caffeine led to a 61% decrease in the caffeine induced peak height of [Ca2+]i when compared to the control. Bradykinin stimulates IP3R-activated CICR-to test the effect of LEV on IP3R-mediated CICR, bradykinin (1 microM) was used to stimulate cells pre-treated with LEV (100 microM). The data showed that LEV caused a 74% decrease in IP3R-mediated CICR compared to the control. In previous studies we have shown that altered Ca2+ homeostatic mechanisms play a role in seizure activity and the development of spontaneous recurrent epileptiform discharges (SREDs). Elevations in [Ca2+]i mediated by CICR systems have been associated with neurotoxicity, changes in neuronal plasticity, and the development of AE. Thus, the ability of LEV to modulate the two major CICR systems demonstrates an important molecular effect of this agent on a major second messenger system in neurons.
Collapse
Affiliation(s)
- Nisha Nagarkatti
- Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | | | | |
Collapse
|
35
|
Wheless JW. Levetiracetam in the treatment of childhood epilepsy. Neuropsychiatr Dis Treat 2007; 3:409-21. [PMID: 19300570 PMCID: PMC2655081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Epilepsy is a common pediatric neurologic disorder that is difficult to manage in a substantial portion of children. Levetiracetam (LEV) is a novel antiepileptic drug (AED) that has recently been approved as add-on treatment for various seizure types in epilepsy populations that include children: for refractory partial seizures in epilepsy patients >/=4 years old, for myoclonic seizures in juvenile myoclonic epilepsy patients >/=12 years old, and for primary generalized tonic-clonic seizures in idiopathic generalized epilepsy patients (>/=6 years old with FDA approval; >/=12 years old with EMEA approval). A review of published pediatric studies indicates that the efficacy of LEV is best established for partial seizures; however, results from recent double-blind and open-label trials indicate that adjunctive LEV also controls generalized seizures - particularly myoclonic and generalized tonic-clonic - in children and adolescents with primary generalized epilepsy. LEV was well-tolerated in pediatric studies. The most common adverse events (AEs) reported were sedation related. Behavioral AEs were among the most commonly reported events in some trials; conversely, improvements in behavior and cognition were also frequently reported. LEV appears to be a safe and effective AED with unique characteristics that benefit the treatment of children with epilepsy.
Collapse
Affiliation(s)
- James W Wheless
- Pediatric Neurology, University of Tennessee Health Science Center; Neuroscience Institute, LeBonheur Children's Medical Center, Memphis, TN, USA.
| |
Collapse
|
36
|
De Smedt T, Raedt R, Vonck K, Boon P. Levetiracetam: the profile of a novel anticonvulsant drug-part I: preclinical data. CNS DRUG REVIEWS 2007; 13:43-56. [PMID: 17461889 PMCID: PMC6494143 DOI: 10.1111/j.1527-3458.2007.00004.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The objective of this article was to review and summarize the available reports on the preclinical profile of the novel anticonvulsant drug levetiracetam (LEV). Therefore, a careful search was conducted in the MEDLINE database and combined with guidelines from regulatory agencies, proceedings of professional scientific meetings, and information provided by the manufacturers. This article provides detailed information on the anticonvulsant effects of LEV in various animal models of epilepsy and on its pharmacology in laboratory animals. The mechanism of action of LEV is reviewed, with special regard to its recently discovered binding site, the synaptic vesicle protein 2A. In general, LEV is shown to be a safe, broad-spectrum anticonvulsant drug with highly beneficial pharmacokinetic properties and a distinct mechanism of action. The clinical studies with LEV will be discussed in the second part of this review article to be published subsequently.
Collapse
Affiliation(s)
- Tim De Smedt
- Laboratory for Clinical and Experimental Neurophysiology - Reference Center for Refractory Epilepsy, University Hospital Ghent, Ghent, Belgium.
| | | | | | | |
Collapse
|
37
|
Reif R, Wang M, Joshi S, A'Amar O, Bigio IJ. Optical method for real-time monitoring of drug concentrations facilitates the development of novel methods for drug delivery to brain tissue. JOURNAL OF BIOMEDICAL OPTICS 2007; 12:034036. [PMID: 17614744 DOI: 10.1117/1.2744025] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The understanding of drug delivery to organs, such as the brain, has been hampered by the inability to measure tissue drug concentrations in real time. We report an application of an optical spectroscopy technique that monitors in vivo the real-time drug concentrations in small volumes of brain tissue. This method will facilitate development of new protocols for delivery of drugs to treat brain cancers. The delivery of many anticancer drugs to the brain is limited by the presence of the blood-brain barrier (BBB). Mitoxantrone (MTX) is a water-soluble anticancer drug that poorly penetrates the BBB. It is preliminarily determined in an animal model that the brain tissue uptake of chemotherapy agents-in this demonstration, MTX-delivered intra-arterially is enhanced when the BBB is disrupted.
Collapse
Affiliation(s)
- Roberto Reif
- Boston University, Department of Biomedical Engineering, 44 Cummington Street, Boston, Massachusetts 02215, USA.
| | | | | | | | | |
Collapse
|
38
|
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: 4.2] [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
|
39
|
Kinirons P, McCarthy M, Doherty CP, Delanty N. Predicting drug-resistant patients who respond to add-on therapy with levetiracetam. Seizure 2006; 15:387-92. [PMID: 16766211 DOI: 10.1016/j.seizure.2006.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Revised: 04/04/2006] [Accepted: 05/01/2006] [Indexed: 10/24/2022] Open
Abstract
INTRODUCTION Levetiracetam (LEV) is approved for use as add-on therapy in adult patients with partial epilepsy. It is apparent from clinical trials that up to 8% of previously drug-resistant patients may be rendered seizure-free by adding-on levetiracetam. As yet there is no way of predicting these unexpectedly responsive patients. We set out to identify our previously refractory patients who had demonstrated unexpected responsiveness to add-on therapy with levetiracetam, and compared these to patients who had not responded to the drug. We then attempted to characterise any clinical features that differentiated these groups of patients. METHODS We included all patients with a history of present or previous exposure to levetiracetam who had been unresponsive to at least two other prior anti-epileptic drugs (AEDs) and recorded their demographic and clinical data. We divided response into (a) 'seizure-free' (seizure-free for a minimum of 6 months after commencing LEV); (b) 'partial > 50%' (greater than 50% reduction in seizures for a minimum of 6 months after commencing LEV); (c) 'honeymoon' (seizure-free for less than 6 months after commencing LEV and then returned towards baseline frequency); and (d) 'no-response'. For the purpose of analysis we considered the 'seizure-free' and 'partial > 50%' groups as 'responders', and the 'no response' group as 'non responders'. RESULTS 344 patients were included in the analysis. Fifty-six patients (16.3%) were rendered seizure-free on levetiracetam. Idiopathic generalised epilepsy and post-traumatic partial epilepsy were more common in the responder than the non-responder group (p = 0.005 and 0.05 respectively). Lamotrigine was used significantly more often in combination with levetiracetam in responders than non-responders (p = 0.003). The mean daily dose of levetiracetam was lower in responders than non-responders. DISCUSSION A higher than expected number of previously drug resistant patients was rendered seizure-free by add-on therapy with levetiracetam. Those who respond best appear to do so at relatively low doses and our data suggest the possibility of a beneficial pharmacodynamic interaction between levetiracetam and lamotrigine. We were unable to identify any clinical factors that clearly predicted which patients would become seizure-free and we hypothesise that response may be determined by genetic or molecular factors. All drug-resistant patients, including those being assessed for surgery, should be considered for a trial of levetiracetam, regardless of their epilepsy classification.
Collapse
Affiliation(s)
- P Kinirons
- Division of Epilepsy, Department of Clinical Neurological Sciences, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | | | | | | |
Collapse
|
40
|
Abstract
Epilepsy is a hetergenous syndrome characterized by recurrently and repeatedly occurring seizures. Although able to inhibit the epileptic seizures, the currently available antiepileptic drugs (AEDs) have no effects on epileptogenesis. Such AEDs should be classified as drugs against ictogenesis, which are transient events in ion and/or receptor-gated channels related with triggering to evoke seizures. Epileptogenesis involves long-term and histological/biochemical/physiological alterations formed in brain structures over a long period, ranging from months to years. This review focuses on the effects of AEDs on epileptogenesis and novel candidates of antiepileptogenic drugs using a genetically defined epilepsy model animal, the spontaneous epileptic rat (SER).
Collapse
|
41
|
Gibbs JE, Walker MC, Cock HR. Levetiracetam: antiepileptic properties and protective effects on mitochondrial dysfunction in experimental status epilepticus. Epilepsia 2006; 47:469-78. [PMID: 16529608 DOI: 10.1111/j.1528-1167.2006.00454.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE To assess the anticonvulsant activity of the novel antiepileptic drug, levetiracetam (LEV) in a model of self-sustaining limbic status epilepticus, and to measure the consequence of LEV treatment on the pattern of mitochondrial dysfunction known to occur after status epilepticus (SE). METHODS The rat perforant pathway was stimulated for 2 h to induce self-sustaining status epilepticus (SSSE). Stimulated rats were assigned to one of three treatment groups, receiving intraperitoneal injections of saline, 200 mg/kg LEV, or 1,000 mg/kg LEV, 15 min into SSSE and at 3 times over the next 44-h period. All animals received diazepam after 3-h SSSE to terminate seizures. Forty-four hours later, the hippocampi were extracted and prepared for electrochemical high-performance liquid chromatography (HPLC), to measure reduced glutathione levels, and for spectrophotometric assays to measure activities of mitochondrial enzymes (aconitase, alpha-ketoglutarate dehydrogenase, citrate synthase, complex I, and complex II/III). These parameters were compared between treatment groups and with sham-operated rats. RESULTS LEV administration did not terminate seizures or have any significant effect on spike frequency, although rats that received 1,000 mg/kg LEV did exhibit improved behavioral seizure parameters. Significant biochemical changes occurred in saline-treated stimulated rats compared with shams: with reductions in glutathione, alpha-ketoglutarate dehydrogenase, aconitase, citrate synthase, and complex I activities. Complex II/III activities were unchanged throughout. Rats that received 1,000 mg/kg LEV had significantly improved biochemical parameters, in many instances, comparable to sham control levels. CONCLUSIONS Despite continuing seizures, administration of LEV (1,000 mg/kg) protects against mitochondrial dysfunction, indicating that in addition to its antiepileptic actions, LEV may have neuroprotective effects.
Collapse
Affiliation(s)
- Julie E Gibbs
- Centre for Clinical Neurosciences, St George's, University of London, Tooting, London, England
| | | | | |
Collapse
|
42
|
Ji-qun C, Ishihara K, Nagayama T, Serikawa T, Sasa M. Long-lasting antiepileptic effects of levetiracetam against epileptic seizures in the spontaneously epileptic rat (SER): differentiation of levetiracetam from conventional antiepileptic drugs. Epilepsia 2005; 46:1362-70. [PMID: 16146430 DOI: 10.1111/j.1528-1167.2005.29604.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE Some evidence suggests that levetiracetam (LEV) possesses antiepileptogenic characteristics. The purpose of this study was to investigate the time course of seizure protection by LEV compared with that of phenytoin (PHT), phenobarbital (PB), valproate (VPA), and carbamazepine (CBZ) in the spontaneously epileptic rat (SER). The SER is a double mutant (tm/tm, zi/zi) showing both tonic convulsions and absence-like seizures. METHODS The effect of single (40, 80, and 160 mg/kg, i.p.) and 5-day (80 mg/kg/day, i.p.) administration of LEV on tonic convulsions and absence-like seizures in SERs were studied. Tonic convulsions induced by blowing air onto the animal's head at 5-min intervals for 30 min and spontaneous absence-like seizures characterized by 5- to 7-Hz spike-wave-like complexes in the cortical and hippocampal EEG were recorded for 30 min. In the single-administration study, observations for seizure activity were performed once before and 3 times (45, 75, and 135 min) after drug administration. In the 5-day administration study, seizure observation was performed 4 times for 30 min (once before and 3 times after drug administration) during the 5-day drug-administration period, and continued once a day until 8 days after the final administration. The antiepileptic effects of 5-day administration of conventional AEDs (PHT, PB, VPA, and CBZ) were examined by using similar methods. RESULTS Tonic convulsions and absence-like seizures were inhibited by a single administration of LEV at 80 and 160 mg/kg, i.p., but not significantly at 40 mg/kg, i.p. When LEV was repeatedly administered at 80 mg/kg/day, i.p., for 5 days to SERs, the inhibitory effects on seizures increased with administration time. The number of tonic convulsions and absence-like seizures were significantly reduced to 39.1% and 38.4% compared with previous values, respectively, after 5-day LEV administration. Furthermore, significant inhibition of tonic convulsions was detected <or=3 days after the final administration, and significant inhibition of absence-like seizures was still observed 8 days after the final injection of LEV. This demonstrates long-lasting seizure protection by LEV after cessation of treatment. PHT, PB, VPA, and CBZ inhibited tonic convulsions more potently compared with LEV in SERs. The maximal antiseizure effects of these drugs were reached after the initial administration, with almost the same antiseizure effects observed through day 5, despite continued drug administration. Moreover, a long-lasting treatment effect was not observed with any of these drugs except for PHT and CBZ, both of which showed moderately prolonged antiseizure effects. CONCLUSIONS These results show that LEV is effective in the treatment of both convulsive and absence-like seizures in SERs after single- and multiple-dose administration. Interestingly, in the 5-day administration study, it was found that the antiepileptic effects for tonic convulsions and absence-like seizures were observed both during the drug-administration period and <or=8 days after the final administration of LEV. This long-lasting effect suggests that LEV may possess an antiepileptogenic effect that it does not share with PHT, PB, VPA, and CBZ.
Collapse
Affiliation(s)
- Cai Ji-qun
- Department of Pharmacology, Hiroshima University School of Medicine, Hiroshima, Japan
| | | | | | | | | |
Collapse
|
43
|
Yan HD, Ji-qun C, Ishihara K, Nagayama T, Serikawa T, Sasa M. Separation of Antiepileptogenic and Antiseizure Effects of Levetiracetam in the Spontaneously Epileptic Rat (SER). Epilepsia 2005; 46:1170-7. [PMID: 16060925 DOI: 10.1111/j.1528-1167.2005.35204.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE The long-lasting antiseizure effects of levetiracetam (LEV) have been observed in the spontaneously epileptic rat (SER) that expresses both tonic and absence-like seizures. Furthermore, the antiepileptogenic effects of LEV in addition to antiseizure effects have been reported in the amygdala-kindling model in rats. This suggests that the long-lasting seizure protection of LEV may be at least partly due to its antiepileptogenic effects. Therefore this study aimed to differentiate the antiseizure and potential antiepileptogenic effects of LEV by administering LEV continuously to SERs before the appearance of any seizure expression. METHODS LEV was administered to the SERs at 80 mg/kg/day (i.p.) from postnatal weeks 5 to 8. The period of observation for tonic convulsions was from postnatal week 5 to 13. Absence-like seizures were recorded by using conventional EEG in weeks 12 and 13. RESULTS After age 7-8 weeks, SERs exhibit spontaneous tonic convulsions. Development of tonic convulsions was significantly inhibited in the LEV group, compared with the control group, by the middle of week 9. A significant reduction of tonic convulsions also was observed in the LEV group until week 13 (5 weeks after termination of the administration). In week 12, the absence-like seizures were significantly lower in the LEV group, compared with the control group. CONCLUSIONS This study demonstrates a significant inhibition of seizures after prolonged treatment with LEV before the developmental expression of seizure activity in SERs. This effect is suggested to be due to an antiepileptogenic effect and not an antiseizure effect of LEV, because the half-life of the drug in plasma is short (2-3 h in rats) after single and long-term administration. Furthermore, the inhibition of seizure expression in SERs was still apparent 5 weeks after termination of LEV treatment. These results further suggest that LEV possesses not only antiseizure effects but also antiepileptogenic properties.
Collapse
Affiliation(s)
- Hai-Dun Yan
- Department of Pharmacology, Hiroshima University School of Medicine, Hiroshima, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
Effects of the novel anti-epileptic drug levetiracetam (50 and 100 mg/kg) on spike and wave discharges (SWDs) of WAG/Rij rats were studied. Levetiracetam decreased the incidence, average duration, total duration and peak frequency of the SWDs. There was no difference between the two doses. These results agree with results obtained in Genetic Absence Epilepsy Rat from Strasbourg (GAERS). Furthermore, the decrease of the SWD peak frequency might support the suggestions that levetiracetam might have a GABAergic mechanism of action.
Collapse
Affiliation(s)
- Brigitte M Bouwman
- NICI/Department of Biological Psychology, University of Nijmegen, P.O. Box 9104, 6500 HE Nijmegen, The Netherlands.
| | | |
Collapse
|
45
|
Wang X, Ratnaraj N, Patsalos PN. The pharmacokinetic inter-relationship of tiagabine in blood, cerebrospinal fluid and brain extracellular fluid (frontal cortex and hippocampus). Seizure 2005; 13:574-81. [PMID: 15519917 DOI: 10.1016/j.seizure.2004.01.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Tiagabine is a unique antiepileptic drug with a novel mechanism of action. Whilst some limited data are available as to the peripheral blood pharmacokinetics of tiagabine, data regarding the kinetics of tiagabine in the central brain compartment are very limited. We therefore sought to investigate serum, cerebrospinal fluid (CSF) and frontal cortex and hippocampal extracellular fluid (ECF) kinetic inter-relationship of tiagabine in a freely moving rat model. METHODS Adult male rats were implanted with either a jugular vein catheter and a cisterna magna catheter for blood and CSF sampling, respectively, or a blood catheter and a microdialysis probe in the hippocampus and frontal cortex (for ECF sampling). Tiagabine was administered intraperitoneal (i.p.) at 20 or 40 mg/kg and blood, CSF and ECF were collected at timed intervals for the measurement of tiagabine concentrations by high performance liquid chromatography. RESULTS Tiagabine concentrations in blood and CSF rose linearly and dose-dependently and time to maximum concentration (Tmax) was 15 and 29 min, respectively. Mean CSF/serum tiagabine concentration ratios (range, 0.008-0.01) were much smaller than the mean free/total tiagabine concentration ratios in serum (0.045 +/- 0.003). Entry of tiagabine into brain ECF (frontal cortex and hippocampus) was rapid with Tmax values of 31-46 min. Distribution of tiagabine in brain was not brain region specific with values in the frontal cortex and hippocampus being indistinguishable. Whilst elimination from CSF was comparable to that of serum, half-life (t(1/2)) values in ECF were three times longer. CONCLUSIONS Tiagabine is associated with linear kinetic characteristics and with rapid brain penetration. However, CSF concentrations are not reflective of free non-protein-bound concentrations in serum. The observation that tiagabine elimination from the brain is threefold slower than that seen in blood, may explain as to the relatively long duration of action of tiagabine.
Collapse
Affiliation(s)
- Xiaolan Wang
- Pharmacology and Therapeutics Unit, Department of Clinical and Experimental Epilepsy, Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | | | | |
Collapse
|
46
|
Potschka H, Baltes S, Löscher W. Inhibition of multidrug transporters by verapamil or probenecid does not alter blood-brain barrier penetration of levetiracetam in rats. Epilepsy Res 2004; 58:85-91. [PMID: 15120740 DOI: 10.1016/j.eplepsyres.2003.12.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2003] [Accepted: 12/26/2003] [Indexed: 10/26/2022]
Abstract
Overexpression of multidrug efflux transporters such as P-glycoprotein (Pgp; ABCB1) or multidrug resistance proteins (MRPs; ABCC) in the blood-brain barrier has recently been suggested to explain, at least in part, pharmacoresistance in epilepsy, which affects about 30% of all patients with this common brain disorder. The novel antiepileptic drug (AED) levetiracetam (LEV) is an effective and well tolerated drug in many patients with otherwise AED-refractory epilepsy. One explanation for the favorable efficacy of LEV in pharmacoresistant patients would be that LEV is not a substrate for Pgp or MRPs in the BBB. In the present study, we used in vivo microdialysis in rats to study whether the concentration of LEV in the extracellular fluid of the cerebral cortex can be modulated by inhibition of Pgp or MRPs, using the Pgp inhibitor verapamil and the MRP1/2 inhibitor probenecid. Local perfusion with verapamil or probenecid via the microdialysis probe did not increase the extracellular brain concentration of LEV, which is in contrast to various other AEDs which have been studied previously by the same experimental protocol in this model. The data indicate that brain uptake of LEV is not affected by Pgp or MRP1/2 which may be an important reason for its antiepileptic efficacy in patients whose seizures are poorly controlled by other AEDs.
Collapse
Affiliation(s)
- Heidrun Potschka
- Department of Pharmacology, Toxicology and Pharmacy, School of Veterinary Medicine, Bünteweg 17, D-30559 Hannover, Germany
| | | | | |
Collapse
|
47
|
Lynch BA, Lambeng N, Nocka K, Kensel-Hammes P, Bajjalieh SM, Matagne A, Fuks B. The synaptic vesicle protein SV2A is the binding site for the antiepileptic drug levetiracetam. Proc Natl Acad Sci U S A 2004; 101:9861-6. [PMID: 15210974 PMCID: PMC470764 DOI: 10.1073/pnas.0308208101] [Citation(s) in RCA: 1038] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Here, we show that the synaptic vesicle protein SV2A is the brain binding site of levetiracetam (LEV), a new antiepileptic drug with a unique activity profile in animal models of seizure and epilepsy. The LEV-binding site is enriched in synaptic vesicles, and photoaffinity labeling of purified synaptic vesicles confirms that it has an apparent molecular mass of approximately 90 kDa. Brain membranes and purified synaptic vesicles from mice lacking SV2A do not bind a tritiated LEV derivative, indicating that SV2A is necessary for LEV binding. LEV and related compounds bind to SV2A expressed in fibroblasts, indicating that SV2A is sufficient for LEV binding. No binding was observed to the related isoforms SV2B and SV2C. Furthermore, there is a high degree of correlation between binding affinities of a series of LEV derivatives to SV2A in fibroblasts and to the LEV-binding site in brain. Finally, there is a strong correlation between the affinity of a compound for SV2A and its ability to protect against seizures in an audiogenic mouse animal model of epilepsy. These experimental results suggest that SV2A is the binding site of LEV in the brain and that LEV acts by modulating the function of SV2A, supporting previous indications that LEV possesses a mechanism of action distinct from that of other antiepileptic drugs. Further, these results indicate that proteins involved in vesicle exocytosis, and SV2 in particular, are promising targets for the development of new CNS drug therapies.
Collapse
Affiliation(s)
- Berkley A Lynch
- Department of Molecular and Cellular Biology, UCB Research Inc., 840 Memorial Drive, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Handforth A, Martin FC. Pilot efficacy and tolerability: A randomized, placebo-controlled trial of levetiracetam for essential tremor. Mov Disord 2004; 19:1215-21. [PMID: 15390011 DOI: 10.1002/mds.20147] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The purpose of this pilot single-site study was to assess efficacy and safety of levetiracetam for essential tremor, using a placebo-controlled, double-blind, randomized crossover design with an interim analysis planned after completion of the first 10 to 15 subjects. The study was designed to detect a mean 30% reduction in composite tremor score, comparable to that of primidone or propranolol, which can be demonstrated with 30 or fewer subjects. Each treatment arm included baseline tremor assessments, a 4-week medication titration, 2 weeks of stable dose, and treatment tremor assessments. Levetiracetam was titrated to 3,000 mg/day or to a lower maximal tolerated dose. The median age was 72 years, with 28 years median tremor duration. There was no statistically significant difference in response between placebo and levetiracetam on any tremor rating scale or accelerometry measure. The 95% confidence interval for the true mean difference between placebo and levetiracetam treatments was +18.5 to -22.5%, which excludes the minimum 30% drop required to consider levetiracetam clinically effective to a degree comparable to primidone or propranolol. Whether levetiracetam has lesser-degree antitremor efficacy was not addressed in this pilot study.
Collapse
Affiliation(s)
- Adrian Handforth
- Neurology Services, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073, USA
| | | |
Collapse
|
49
|
Abstract
Since 1989, eight new antiepileptic drugs (AEDs) have been licensed for clinical use. Levetiracetam is the latest to be licensed and is used as adjunctive therapy for the treatment of adult patients with partial seizures with or without secondary generalisation that are refractory to other established first-line AEDs. 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. After oral ingestion, levetiracetam is rapidly absorbed, with peak concentration occurring after 1.3 hours, and its bioavailability is >95%. Co-ingestion of food slows the rate but not the extent of absorption. Levetiracetam is not bound to plasma proteins and has a volume of distribution of 0.5-0.7 L/kg. Plasma concentrations increase in proportion to dose over the clinically relevant dose range (500-5000 mg) and there is no evidence of accumulation during multiple administration. Steady-state blood concentrations are achieved within 24-48 hours. The elimination half-life in adult volunteers, adults with epilepsy, children with epilepsy and elderly volunteers is 6-8, 6-8, 5-7 and 10-11 hours, respectively. Approximately 34% of a levetiracetam dose is metabolised and 66% is excreted in urine unmetabolised; however, the metabolism is not hepatic but occurs primarily in blood by hydrolysis. Autoinduction is not a feature. As clearance is renal in nature it is directly dependent on creatinine clearance. Consequently, dosage adjustments are necessary for patients with moderate to severe renal impairment. To date, no clinically relevant pharmacokinetic interactions between AEDs and levetiracetam have been identified. Similarly, levetiracetam does not interact with digoxin, warfarin and the low-dose contraceptive pill; however, adverse pharmacodynamic interactions with carbamazepine and topiramate have been demonstrated. Overall, the pharmacokinetic characteristics of levetiracetam are highly favourable and make its clinical use simple and straightforward.
Collapse
Affiliation(s)
- Philip N Patsalos
- Pharmacology and Therapeutics Unit, Department of Clinical and Experimental Epilepsy, Institute of Neurology/The National Hospital for Neurology and Neurosurgery, London, UK.
| |
Collapse
|
50
|
Zhang ZJ, Xing GQ, Russell S, Obeng K, Post RM. Unidirectional Cross-tolerance from Levetiracetam to Carbamazepine in Amygdala-kindled Seizures. Epilepsia 2003; 44:1487-93. [PMID: 14636317 DOI: 10.1111/j.0013-9580.2003.34803.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Tolerance is a potential problem in long-term anticonvulsant therapy of epilepsy, bipolar disorder, and neuropathic pain. The present study was designed to determine whether cross-tolerance occurs between levetiracetam (LEV) and carbamazepine (CBZ) in amygdala-kindled rats. METHODS Male Sprague-Dawley rats were implanted with an electrode into the left amygdala. While kindling stimulation was started, animals received repeated treatment (i.p.) with saline (n = 7) or LEV (150 mg/kg, n = 8). Saline-injected rats were subsequently challenged with a single dose of 150 mg/kg LEV when full kindling developed (stage > or =4). Both groups of rats were then administered long-term CBZ (5 mg/kg) until rats developed complete tolerance. All CBZ-tolerant rats were subsequently re-exposed to LEV (150 mg/kg) for an additional 10 consecutive days. RESULTS Repeated LEV treatment significantly suppressed the increase in seizure stage, seizure duration, and afterdischarge duration induced by amygdala stimulation, markedly increasing the number of stimulations to achieve a kindling major motor seizure. The LEV challenge produced a more robust suppression of seizure stage in saline-injected rats compared with LEV-treated animals. CBZ treatment markedly suppressed fully kindled seizures in rats initially injected with saline, and then anticonvulsant tolerance rapidly developed after 3-4 days of repeated treatment. In contrast, rats that had initially received repeated LEV treatment did not show a response to treatment with CBZ (5 mg/kg). When CBZ-tolerant rats were subsequently exposed to LEV (150 mg/kg), noticeable anticonvulsant effects were observed; but these were gradually lost with increasing numbers of LEV exposures. CONCLUSIONS Whereas LEV shows potent antiepileptogenic and anticonvulsant effects in amygdala-kindled rats, its repeated treatment induces anticonvulsant tolerance and unidirectional cross-tolerance to CBZ. In contrast, anticonvulsant tolerance to CBZ does not transfer to LEV. The mechanistic implications of the present results for clinical therapeutics remain to be evaluated.
Collapse
Affiliation(s)
- Zhang-Jin Zhang
- Department of Psychiatry, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, U.S.A.
| | | | | | | | | |
Collapse
|