Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 372(1): 11-20, 2020 01.
Article in English | MEDLINE | ID: mdl-31619464

ABSTRACT

The antiepileptic drug (AED) candidate, (4R)-4-(2-chloro-2,2-difluoroethyl)-1-{[2-(methoxymethyl)-6-(trifluoromethyl)imidazo[2,1-b][1,3,4]thiadiazol-5-yl]methyl}pyrrolidin-2-one (padsevonil), is the first in a novel class of drugs that bind to synaptic vesicle protein 2 (SV2) proteins and the GABAA receptor benzodiazepine site, allowing for pre- and postsynaptic activity, respectively. In acute seizure models, padsevonil provided potent, dose-dependent protection against seizures induced by administration of pilocarpine or 11-deoxycortisol, and those induced acoustically or through 6 Hz stimulation; it was less potent in the pentylenetetrazol, bicuculline, and maximal electroshock models. Padsevonil displayed dose-dependent protective effects in chronic epilepsy models, including the intrahippocampal kainate and Genetic Absence Epilepsy Rats from Strasbourg models, which represent human mesial temporal lobe and absence epilepsy, respectively. In the amygdala kindling model, which is predictive of efficacy against focal to bilateral tonic-clonic seizures, padsevonil provided significant protection in kindled rodents; in mice specifically, it was the most potent AED compared with nine others with different mechanisms of action. Its therapeutic index was also the highest, potentially translating into a favorable efficacy and tolerability profile in humans. Importantly, in contrast to diazepam, tolerance to padsevonil's antiseizure effects was not observed in the pentylenetetrazol-induced clonic seizure threshold test. Further results in the 6 Hz model showed that padsevonil provided significantly greater protection than the combination of diazepam with either 2S-(2-oxo-1-pyrrolidinyl)butanamide (levetiracetam) or 2S-2-[(4R)-2-oxo-4-propylpyrrolidin-1-yl] butanamide (brivaracetam), both selective SV2A ligands. This observation suggests that padsevonil's unique mechanism of action confers antiseizure properties beyond the combination of compounds targeting SV2A and the benzodiazepine site. Overall, padsevonil displayed robust efficacy across validated seizure and epilepsy models, including those considered to represent drug-resistant epilepsy. SIGNIFICANCE STATEMENT: Padsevonil, a first-in-class antiepileptic drug candidate, targets SV2 proteins and the benzodiazepine site of GABAA receptors. It demonstrated robust efficacy across a broad range of rodent seizure and epilepsy models, several representing drug-resistant epilepsy. Furthermore, in one rodent model, its efficacy extended beyond the combination of drugs interacting separately with SV2 or the benzodiazepine site. Padsevonil displayed a high therapeutic index, potentially translating into a favorable safety profile in humans; tolerance to antiseizure effects was not observed.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Imidazoles/therapeutic use , Pyrrolidinones/therapeutic use , Seizures/drug therapy , Thiadiazoles/therapeutic use , Amygdala/drug effects , Amygdala/physiopathology , Animals , Anticonvulsants/adverse effects , Anticonvulsants/pharmacology , Drug Evaluation, Preclinical , Female , Imidazoles/adverse effects , Imidazoles/pharmacology , Kindling, Neurologic , Male , Maximum Tolerated Dose , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Pyrrolidinones/adverse effects , Pyrrolidinones/pharmacology , Rats , Rats, Sprague-Dawley , Thiadiazoles/adverse effects , Thiadiazoles/pharmacology
2.
Epilepsia ; 58(11): e157-e161, 2017 11.
Article in English | MEDLINE | ID: mdl-28850675

ABSTRACT

Brivaracetam (BRV) is a selective, high-affinity ligand for synaptic vesicle protein 2A (SV2A), recently approved as adjunctive treatment for drug-refractory partial-onset seizures in adults. BRV binds SV2A with higher affinity than levetiracetam (LEV), and was shown to have a differential interaction with SV2A. Because LEV was reported to interact with multiple excitatory and inhibitory ligand-gated ion channels and that may impact its pharmacological profile, we were interested in determining whether BRV directly modulates inhibitory and excitatory ionotropic receptors in central neurons. Voltage-clamp experiments were performed in primary cultures of mouse hippocampal neurons. At a supratherapeutic concentration of 100 µm, BRV was devoid of any direct effect on currents gated by γ-aminobutyric acidergic type A, glycine, kainate, N-methyl-d-aspartate, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid. Similarly to LEV, BRV reveals a potent ability to oppose the action of negative modulators on the inhibitory receptors. In conclusion, these results show that BRV contrasts with LEV by not displaying any direct action on inhibitory or excitatory postsynaptic ligand-gated receptors at therapeutic concentrations and thereby support BRV's role as a selective SV2A ligand. These findings add further evidence to the validity of SV2A as a relevant antiepileptic drug target and emphasize the potential for exploring further presynaptic mechanisms as a novel approach to antiepileptic drug discovery.


Subject(s)
Glutamic Acid/pharmacology , Glycine/pharmacology , Hippocampus/physiology , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/physiology , Pyrrolidinones/pharmacology , gamma-Aminobutyric Acid/pharmacology , Animals , Anticonvulsants/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/drug effects , Membrane Glycoproteins/agonists , Mice , Nerve Tissue Proteins/agonists , Neurons/drug effects , Neurons/physiology
3.
Epilepsia ; 58(7): 1199-1207, 2017 07.
Article in English | MEDLINE | ID: mdl-28597912

ABSTRACT

OBJECTIVE: To evaluate acute and long-term effects of intravenous brivaracetam (BRV) and BRV + diazepam (DZP) combination treatment in a rat model of self-sustaining status epilepticus (SSSE). METHODS: Rats were treated with BRV (10 mg/kg) 10 min after initiation of perforant path stimulation (PPS) as early treatment; or BRV (10-300 mg/kg), DZP (1 mg/kg), or BRV (0.3-10 mg/kg) + DZP (1 mg/kg) 10 min after the end of PPS (established SSSE). Seizure activity was recorded electrographically for 24 h posttreatment (acute effects), and for 1 week at 6-8 weeks or 12 months' posttreatment (long-term effects). All treatments were compared with control rats using one-way analysis of variance (ANOVA) and Bonferroni's test, or Kruskal--Wallis and Dunn's multiple comparison tests, when appropriate. RESULTS: Treatment of established SSSE with BRV (10-300 mg/kg) resulted in dose-dependent reduction in SSSE duration and cumulative seizure time, achieving statistical significance at doses ≥100 mg/kg. Lower doses of BRV (0.3-10 mg/kg) + low-dose DZP (1 mg/kg) significantly reduced SSSE duration and number of seizures. All control rats developed spontaneous recurrent seizures (SRS) 6-8 weeks after SSSE, whereas seizure freedom was noted in 2/10, 5/10, and 6/10 rats treated with BRV 200 mg/kg, 300 mg/kg, and BRV 10 mg/kg + DZP, respectively. BRV (10-300 mg/kg) showed a dose-dependent trend toward reduction of SRS frequency, cumulative seizure time, and spike frequency, achieving statistical significance at 300 mg/kg. Combination of BRV (10 mg/kg) + DZP significantly reduced SRS frequency, cumulative seizure time, and spike frequency. In the 12-month follow-up study, BRV (0.3-10 mg/kg) + low-dose DZP markedly reduced SRS frequency, cumulative seizure time, and spike frequency, achieving statistical significance at some doses. Early treatment of SSSE with BRV 10 mg/kg significantly reduced long-term SRS frequency. SIGNIFICANCE: These findings support clinical evaluation of BRV for treatment of status epilepticus or acute repetitive seizures.


Subject(s)
Anticonvulsants/pharmacology , Diazepam/pharmacology , Disease Models, Animal , Electroencephalography/drug effects , Pyrrolidinones/pharmacology , Signal Processing, Computer-Assisted , Status Epilepticus/drug therapy , Animals , Dentate Gyrus/drug effects , Dentate Gyrus/physiopathology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Electrodes, Implanted , Evoked Potentials/drug effects , Evoked Potentials/physiology , Infusions, Intravenous , Long-Term Care , Male , Perforant Pathway/drug effects , Perforant Pathway/physiopathology , Rats , Rats, Wistar , Status Epilepticus/physiopathology
4.
Epilepsia ; 57(4): 538-48, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26920914

ABSTRACT

Despite availability of effective antiepileptic drugs (AEDs), many patients with epilepsy continue to experience refractory seizures and adverse events. Achievement of better seizure control and fewer side effects is key to improving quality of life. This review describes the rationale for the discovery and preclinical profile of brivaracetam (BRV), currently under regulatory review as adjunctive therapy for adults with partial-onset seizures. The discovery of BRV was triggered by the novel mechanism of action and atypical properties of levetiracetam (LEV) in preclinical seizure and epilepsy models. LEV is associated with several mechanisms that may contribute to its antiepileptic properties and adverse effect profile. Early findings observed a moderate affinity for a unique brain-specific LEV binding site (LBS) that correlated with anticonvulsant effects in animal models of epilepsy. This provided a promising molecular target and rationale for identifying selective, high-affinity ligands for LBS with potential for improved antiepileptic properties. The later discovery that synaptic vesicle protein 2A (SV2A) was the molecular correlate of LBS confirmed the novelty of the target. A drug discovery program resulted in the identification of anticonvulsants, comprising two distinct families of high-affinity SV2A ligands possessing different pharmacologic properties. Among these, BRV differed significantly from LEV by its selective, high affinity and differential interaction with SV2A as well as a higher lipophilicity, correlating with more potent and complete seizure suppression, as well as a more rapid brain penetration in preclinical models. Initial studies in animal models also revealed BRV had a greater antiepileptogenic potential than LEV. These properties of BRV highlight its promising potential as an AED that might provide broad-spectrum efficacy, associated with a promising tolerability profile and a fast onset of action. BRV represents the first selective SV2A ligand for epilepsy treatment and may add a significant contribution to the existing armamentarium of AEDs.


Subject(s)
Anticonvulsants/metabolism , Drug Discovery/trends , Epilepsy/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Pyrrolidinones/metabolism , Animals , Anticonvulsants/therapeutic use , Dose-Response Relationship, Drug , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/trends , Epilepsy/drug therapy , Humans , Ligands , Pyrrolidinones/therapeutic use , Treatment Outcome
5.
Epilepsia ; 57(2): 201-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26663401

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.


Subject(s)
Anticonvulsants/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Pyrrolidinones/pharmacokinetics , Animals , Brain/diagnostic imaging , Caco-2 Cells , Dogs , Epilepsy, Reflex , Humans , In Vitro Techniques , Levetiracetam , Macaca mulatta , Mice , Molecular Targeted Therapy , Permeability , Piracetam/analogs & derivatives , Piracetam/pharmacokinetics , Positron-Emission Tomography , Rats
6.
Epilepsia ; 56(5): 800-5, 2015 May.
Article in English | MEDLINE | ID: mdl-25818358

ABSTRACT

OBJECTIVE: Brivaracetam (BRV) is a new antiepileptic drug candidate rationally designed for high affinity and selectivity for the synaptic vesicle protein 2A. This study explored anti-ictogenic and antiepileptogenic effects of BRV in rats at different stages of development. METHODS: Using a rapid kindling model in P14, P21, P28, and P60 rats, we studied two doses of BRV: 10 and 100 mg/kg injected intraperitoneally 30 min before afterdischarge assessment. We also assessed blood and brain concentrations of BRV 30 min after the injection. RESULTS: BRV 100 mg/kg significantly increased the afterdischarge threshold (ADT) at all ages, whereas BRV at 10 mg/kg increased ADT in P60, P28, and P21 rats. BRV also shortens the afterdischarge duration (ADD), achieving statistical significance with 10 and 100 mg/kg at P60 and with 100 mg/kg at P21. At P60, BRV increases the number of stimulations required to achieve a stage 4-5 seizure in a dose-dependent manner. At P28 and P21, BRV increased the number of stimulations required to develop a stage 4-5 seizure in a dose-dependent manner with almost complete elimination of stage 4-5 seizures. In contrast, at P14, BRV had no effect on the number of stage 4-5 seizures. An age-related decrease in blood and brain concentrations of BRV was observed 30 min after injection of BRV 10 mg/kg, whereas with 100 mg/kg there were no significant age-correlated differences in brain and serum BRV concentrations. SIGNIFICANCE: BRV exerted dose-dependent anti-ictogenic effects from P60 to P14 independent of brain maturation. BRV also exhibited antiepileptogenic effects at P60, whereas this effect need to be further evaluated at P28 and P21. We did not observe any effect on epileptogenesis at P14 at either dose.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Pyrrolidinones/therapeutic use , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Anticonvulsants/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation/adverse effects , Electroencephalography , Epilepsy/etiology , Epilepsy/metabolism , Epilepsy/pathology , Hippocampus/drug effects , Hippocampus/physiopathology , Kindling, Neurologic/drug effects , Kindling, Neurologic/physiology , Male , Pyrrolidinones/metabolism , Rats , Rats, Wistar
7.
Epilepsy Res ; 110: 189-205, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25616473

ABSTRACT

The antiepileptic drug lacosamide [(R)-2-acetamido-N-benzyl-3-methoxypropanamide], a chiral functionalized amino acid, was originally identified by virtue of activity in the mouse and rat maximal electroshock (MES) test. Attention was drawn to lacosamide because of its high oral potency and stereoselectivity. Lacosamide is also active in the 6 Hz seizure model but inactive against clonic seizures in rodents induced by subcutaneous pentylenetetrazol, bicuculline and picrotoxin. It is also ineffective in genetic models of absence epilepsy. At doses greater than those required to confer protection in the MES test, lacosamide inhibits behavioral and electrographic seizures in hippocampal kindled rats. It also effectively terminates seizures in the rat perforant path stimulation status epilepticus model when administered early after the onset of seizures. Lacosamide does not exhibit antiepileptogenic effects in kindling or post-status epilepticus models. The profile of lacosamide in animal seizure and epilepsy models is similar to that of sodium channel blocking antiepileptic drugs, such as phenytoin and carbamazepine. However, unlike these agents, lacosamide does not affect sustained repetitive firing (SRF) on a time scale of hundreds of milliseconds or affect fast inactivation of voltage-gated sodium channels; however, it terminates SRF on a time scale of seconds by an apparent effect on sodium channel slow inactivation. Lacosamide shifts the slow inactivation curve to more hyperpolarized potentials and enhances the maximal fraction of channels that are in the slow inactivated state. Currently, lacosamide is the only known antiepileptic drug in clinical practice that exerts its anticonvulsant activity predominantly by selectively enhancing slow sodium channel inactivation.


Subject(s)
Acetamides/pharmacology , Anticonvulsants/pharmacology , Animals , Humans , Lacosamide , Voltage-Gated Sodium Channel Blockers/pharmacology
8.
Epilepsia ; 54(7): 1167-75, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23750855

ABSTRACT

PURPOSE: Lacosamide (LCM, Vimpat) is an anticonvulsant with a unique mode of action. This provides lacosamide with the potential to act additively or even synergistically with other antiepileptic drugs (AEDs). The objective of this study was to determine the presence of such interactions by isobolographic analysis. METHODS: The anticonvulsant effect of LCM in combination with other AEDs including carbamazepine (CBZ), phenytoin (PHT), valproate (VPA), lamotrigine (LTG), topiramate (TPM), gabapentin (GBP), and levetiracetam (LEV) at fixed dose ratios of 1:3, 1:1, and 3:1, was evaluated in the 6-Hz-induced seizure model in mice. In addition, the impact of the combinations of LCM with the other AEDs on motor coordination was assessed in the rotarod test. Finally, AED concentrations were measured in blood and brain to evaluate potential pharmacokinetic drug interactions. KEY FINDINGS: All studied AEDs produced dose-dependent anticonvulsant effects against 6-Hz-induced seizures. Combinations of LCM with CBZ, LTG, TPM, GBP, or LEV were synergistic. All other LCM/AED combinations displayed additive effects with a tendency toward synergism. Furthermore, no enhanced adverse effects were observed in the rotarod test by combining LCM with other AEDs. No pharmacokinetic interactions were seen on brain AED concentrations. Coadministration of LCM and TPM led to an increase in plasma levels of LCM, whereas the plasma concentration of PHT was increased by coadministration of LCM. SIGNIFICANCE: The synergistic anticonvulsant interaction of LCM with various AEDs, without exacerbation of adverse motor effects, highlights promising properties of LCM as add-on therapy for drug refractory epilepsy.


Subject(s)
Acetamides/therapeutic use , Anticonvulsants/therapeutic use , Seizures/drug therapy , Acetamides/pharmacokinetics , Animals , Anticonvulsants/pharmacokinetics , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Electric Stimulation/adverse effects , Lacosamide , Male , Mice , Mice, Inbred CBA , Motor Activity/drug effects , Rotarod Performance Test , Seizures/etiology , Seizures/physiopathology
9.
Epilepsia ; 54(7): 1176-85, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23614482

ABSTRACT

PURPOSE: The antiepileptic drug, lacosamide, exerts its therapeutic activity by enhancing slow inactivation of voltage-gated sodium channels. Because putative preventive or disease-modifying effects of drugs may affect epileptogenesis, intrinsic severity, and comorbidities, it is of particular interest to assess the effect of lacosamide on the development of epilepsy and associated cellular alterations. METHODS: The effect of lacosamide was evaluated in an electrical rat status epilepticus (SE) model with a 24-day treatment phase following induction of SE. The impact of lacosamide on the development of spontaneous seizures based on continuous video-electroencephalography (EEG) monitoring, as well as the impact on neuronal cell loss and alterations in hippocampal neurogenesis, was assessed. KEY FINDINGS: Neither low-dose nor high-dose lacosamide affected the development of spontaneous seizures. A dose-dependent neuroprotective effect of lacosamide with significant reduction of neuronal cell loss was observed in the hippocampal CA1 region, as well as in the piriform cortex. In addition, lacosamide attenuated the impact of SE on the rate of hippocampal cell neurogenesis. Moreover, lacosamide prevented a significant rise in the number of persistent basal dendrites. SIGNIFICANCE: Our data do not support an antiepileptogenic effect of lacosamide. However, because lacosamide reduced SE-associated cellular alterations, it would be of interest to determine whether these effects indicate a putative disease-modifying effect of lacosamide in future studies.


Subject(s)
Hippocampus/pathology , Nerve Degeneration/prevention & control , Neurogenesis/drug effects , Neurons/drug effects , Status Epilepticus/drug therapy , Status Epilepticus/pathology , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Death/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Doublecortin Domain Proteins , Electric Stimulation/adverse effects , Female , Hippocampus/drug effects , Hippocampus/physiopathology , Microtubule-Associated Proteins/metabolism , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Neural Stem Cells/drug effects , Neuropeptides/metabolism , Rats , Rats, Sprague-Dawley , Status Epilepticus/complications , Status Epilepticus/etiology
10.
Eur J Pharmacol ; 664(1-3): 36-44, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21575627

ABSTRACT

Brivaracetam is a novel synaptic vesicle protein 2A (SV2A) ligand reported to be 10 fold more potent than levetiracetam in animal models of epilepsy. This study reports the binding profile of brivaracetam in the brain of several species in relation to its anticonvulsant properties. The affinity, kinetics and selectivity of brivaracetam and its tritiated form [(3)H]ucb 34714 have been determined by in vitro binding experiments in rat, human and mouse brain and on recombinant human SV2A. Brivaracetam and levetiracetam ex vivo binding to SV2A and anticonvulsant activities in audiogenic mice were compared in relation to dose and time. Brivaracetam bound selectively with 20 fold higher affinity than levetiracetam to SV2A. [(3)H]ucb 34714 bound reversibly and with high affinity to an homogenous population of binding sites in rat and human brain and to human SV2A expressed in CHO cells. The binding sites labeled by [(3)H]ucb 34714 in brain had the pharmacological characteristics of SV2A and no specific binding could be detected in the brain of SV2A(-/-) knock-out mice. The time- and dose-dependency of brivaracetam and levetiracetam for binding to brain SV2A and for providing seizure protection in audiogenic mice correlated well; brivaracetam being more potent and faster than levetiracetam. Brivaracetam is a potent and selective SV2A ligand. From its affinity and pharmacokinetics, simulations predicted that at therapeutically relevant doses, brivaracetam should occupy more than 80% of SV2A in human brain, in line with levels of occupancy observed in pre-clinical models of epilepsy.


Subject(s)
Anticonvulsants/metabolism , Anticonvulsants/pharmacology , Brain/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Pyrrolidinones/metabolism , Pyrrolidinones/pharmacology , Seizures/prevention & control , Animals , Binding, Competitive , Brain/drug effects , CHO Cells , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Epilepsy, Reflex/metabolism , Epilepsy, Reflex/prevention & control , Female , Humans , Kinetics , Ligands , Male , Mice , Protein Binding , Rats , Seizures/metabolism , Substrate Specificity
11.
Epilepsy Res ; 94(1-2): 10-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21277168

ABSTRACT

The effective management of status epilepticus (SE) continues to be a therapeutic challenge. The aim of this study was to investigate the efficacy of lacosamide treatment in an experimental model of self-sustaining SE. Rats were treated with lacosamide (3, 10, 30 or 50mg/kg) either 10 min (early treatment) or 40 min (late treatment) after the initiation of perforant path stimulation. Early lacosamide treatment significantly and dose-dependently reduced acute SE seizure activity; late treatment showed only a non-significant trend toward reduced seizure activity. Early lacosamide treatment also dose-dependently reduced the number of spontaneous recurrent seizures following a 6-week waiting period, with 70% reduction at the highest dose tested (50mg/kg); there was also a significant reduction in the number of spikes and the cumulative time spent in seizures. Late treatment with high-dose lacosamide (30-50mg/kg) reduced the number of animals that developed spontaneous recurrent seizures (33% vs 100% in controls, P<.05), but did not significantly reduce seizure severity or frequency in rats that developed spontaneous recurrent seizures. The results presented here suggest that lacosamide deserves investigation for the clinical treatment of SE. Potential for disease modification in this rat model of self-sustaining SE will require further studies.


Subject(s)
Acetamides/therapeutic use , Anticonvulsants/therapeutic use , Status Epilepticus/drug therapy , Analysis of Variance , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Electroencephalography , Lacosamide , Rats , Self Administration , Status Epilepticus/physiopathology , Time Factors
13.
Epilepsia ; 50(7): 1729-40, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19486357

ABSTRACT

PURPOSE: Synaptic vesicle protein 2A (SV2A) constitutes a distinct binding site for an antiepileptic drug levetiracetam (Keppra). In the present study we characterized SV2A (+/-) heterozygous mice in several seizure models and tested if the anticonvulsant efficacy of levetiracetam is reduced in these mice. METHODS: Seizure thresholds of male SV2A (+/-) mice and their wild-type littermates were assessed in pilocarpine (i.p.), kainic acid (s.c.), pentylenetetrazol (i.v.), 6-Hz and maximal electroshock models. Kindling development was compared in amygdala and corneal kindling models. Ex vivo binding of levetiracetam to SV2A was also performed. RESULTS: Long-term electroencephalography (EEG) monitoring and behavioral observations of SV2A (+/-) mice did not reveal any spontaneous seizure activity. However, a reduced seizure threshold of SV2A (+/-) mice was observed in pilocarpine, kainic acid, pentylenetetrazol, and 6-Hz models, but not in maximal electroshock seizure model. Accelerated epileptogenesis development was also demonstrated in amygdala and corneal kindling models. Anticonvulsant efficacy of levetiracetam, defined as its ability to increase seizure threshold for 6 Hz electrical stimulation, was significantly reduced (approx. 50%) in the SV2A (+/-) mice, consistently with reduced binding to SV2A in these mice. In contrast, valproate produced the same anticonvulsant effect in both SV2A (+/+) and SV2A (+/-) mice. DISCUSSION: The present results evidence that SV2A is involved in mediation of the in vivo anticonvulsant activity of levetiracetam, in accordance with its previously proposed mechanism of action. Furthermore, the present data also indicate that even partial SV2A deficiency may lead to increased seizure vulnerability and accelerated epileptogenesis.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy/genetics , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Piracetam/analogs & derivatives , Amygdala/drug effects , Amygdala/physiology , Animals , Binding Sites/drug effects , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Electroshock/statistics & numerical data , Epilepsy/chemically induced , Epilepsy/drug therapy , Kainic Acid , Kindling, Neurologic/drug effects , Kindling, Neurologic/genetics , Kindling, Neurologic/physiology , Levetiracetam , Male , Membrane Glycoproteins/pharmacology , Membrane Glycoproteins/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins/pharmacology , Nerve Tissue Proteins/physiology , Pentylenetetrazole/pharmacology , Pharmacogenetics , Phenotype , Piracetam/pharmacology
14.
Eur J Pharmacol ; 614(1-3): 30-7, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19383493

ABSTRACT

Seletracetam is a pyrrolidone derivative with a one-log-unit higher affinity for the synaptic vesicle protein 2A (SV2A) than levetiracetam (Keppra). This study explored its anticonvulsant properties in animal models of epilepsy. Seletracetam reduced both the amplitude and repetitive firing of population spikes induced by a high K(+)/low Ca(2+) concentration fluid (HKLCF) in rat hippocampal slices. The reduction of HKLCF-induced increases in population spike amplitude was particularly pronounced, and occurred at approximately 10 times lower seletracetam concentrations than previously observed for levetiracetam. These invitro data suggest that desynchronisation of epileptiform activity may contribute significantly to the antiepileptic properties of seletracetam. Seletracetam also showed a potent anti-seizure activity in animal models mimicking partial-onset (kindled animals) and generalized epilepsy (audiogenic seizure susceptible mice and genetic absence epilepsy rats from Strasbourg (GAERS)). In amygdala-kindled rats, seletracetam increased the generalized seizure threshold current and decreased the duration of the after-discharge and the seizure severity observed at the after-discharge threshold current, and generally had a much more potent effect than previously observed for levetiracetam. Seletracetam showed no psychomimetic effects and a very high central nervous system (CNS) tolerability in both kindled and GAERS rats, markedly superior to that of levetiracetam and other antiepileptic drugs. These results suggest that seletracetam may represent an effective and very well tolerated broad-spectrum agent for the symptomatic treatment of epilepsy.


Subject(s)
Epilepsy/drug therapy , Pyrrolidinones/pharmacology , Amygdala/drug effects , Amygdala/physiopathology , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Epilepsy/physiopathology , Female , Hippocampus/drug effects , Hippocampus/physiopathology , In Vitro Techniques , Male , Mice , Motor Activity/drug effects , Pyrrolidinones/administration & dosage , Pyrrolidinones/adverse effects , Pyrrolidinones/therapeutic use , Rats , Rats, Sprague-Dawley , Rotarod Performance Test , Seizures/drug therapy , Seizures/physiopathology
15.
Epilepsia ; 50(3): 387-97, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18627416

ABSTRACT

Levetiracetam (Keppra) is an antiepileptic drug (AED) characterized by a novel mechanism of action, unique profile of activity in seizure models, and broad-spectrum clinical efficacy. The present report critically reviews several preclinical studies focused on combination therapy with levetiracetam and other anticonvulsants in various seizure and epilepsy models. Administration of levetiracetam together with many different clinically used AEDs or other anticonvulsants generally enhances their protective activity and, among existing AEDs, this was particularly prevalent with valproate. The protective activity of other AEDs was also enhanced by levetiracetam, which seems to be a universal finding that is independent of seizure model or drug combination studied. However, particularly strong enhancement was observed when levetiracetam was combined with agents either enhancing GABAergic or reducing glutamatergic neurotransmission. Importantly, these combinations were not associated with exacerbation of side effects or pharmacokinetic interactions. Based on the available preclinical data, it appears that combination treatment with levetiracetam and other anticonvulsants provides additional therapeutic benefit that may be attributed to its novel and distinct mechanism of action. Moreover, combinations of levetiracetam with clinically used AEDs that enhance GABAergic inhibition may be considered for rational polytherapy, which is often necessary in drug-resistant patients.


Subject(s)
Anticonvulsants/therapeutic use , Drug Evaluation, Preclinical , Epilepsy/drug therapy , Piracetam/analogs & derivatives , Animals , Anticonvulsants/pharmacology , Anticonvulsants/toxicity , Brain/drug effects , Disease Models, Animal , Drug Interactions , Drug Therapy, Combination , Epilepsy/blood , Glutamic Acid/metabolism , Humans , Levetiracetam , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Piracetam/pharmacokinetics , Piracetam/therapeutic use , Piracetam/toxicity , gamma-Aminobutyric Acid/metabolism
16.
Brain Res Bull ; 77(5): 282-5, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-18722515

ABSTRACT

Levetiracetam (Keppra) is a new generation antiepileptic drug characterized by a unique profile of activity in experimental models of epilepsy. It also has a distinct binding site in the brain, i.e. the synaptic vesicle protein type 2 (SV2A). Levetiracetam has been reported to have antiepileptogenic and disease-modifying properties. In the present study the effects of chronic treatment with levetiracetam were assessed in rats that sustained pilocarpine-induced status epilepticus (SE). Hippocampal field potentials were recorded in vivo in anesthetized animals after 3-day washout period that followed 21-day treatment with different doses of levetiracetam (50, 150 or 300 mg/kg/day) administered via ALZET osmotic mini-pumps. Vehicle treated rats together with naive animals (not subjected to SE) were used as control groups. Chronic treatment with levetiracetam yielded clinically relevant plasma concentrations throughout the experiment with complete washout of the drug 3 days after treatment cessation. At this point in time post-SE rats chronically treated with vehicle developed clear signs of hippocampal hyperexcitability, i.e. increased amplitude of population spike (PS) recorded in the dentate gyrus and reduced paired-pulse inhibition in the CA1 area. Levetiracetam treatment dose-dependently counteracted these long-term effects of pilocarpine-induced SE. Furthermore, at the dose of 300 mg/kg/day levetiracetam restored these parameters back to control level. The present results indicate that chronic treatment with levetiracetam completely inhibits the development of hippocampal hyperexcitability following pilocarpine-induced SE.


Subject(s)
Anticonvulsants/pharmacology , Hippocampus , Membrane Potentials , Muscarinic Agonists/pharmacology , Pilocarpine/pharmacology , Piracetam/analogs & derivatives , Status Epilepticus , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/blood , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/physiology , Humans , Levetiracetam , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Piracetam/administration & dosage , Piracetam/blood , Piracetam/pharmacology , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology
17.
Biochem Biophys Res Commun ; 375(4): 491-5, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18692481

ABSTRACT

The synaptic vesicle protein 2A (SV2A), the brain-binding site of the anti-epileptic drug levetiracetam (LEV), has been characterized by Protein Tomography. We identified two major conformations of SV2A in mouse brain tissue: first, a compact, funnel-structure with a pore-like opening towards the cytoplasm; second, a more open, V-shaped structure with a cleft-like opening towards the intravesicular space. The large differences between these conformations suggest a high degree of flexibility and support a valve-like mechanism consistent with the postulated transporter role of SV2A. These two conformations are represented both in samples treated with LEV, and in saline-treated samples, which indicates that LEV binding does not cause a large-scale conformational change of SV2A, or lock a specific conformational state of the protein. This study provides the first direct structural data on SV2A, and supports a transporter function suggested by sequence homology to MFS class of transporter proteins.


Subject(s)
Membrane Glycoproteins/chemistry , Nerve Tissue Proteins/chemistry , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Brain Chemistry , Immunohistochemistry/methods , Levetiracetam , Membrane Glycoproteins/metabolism , Mice , Microscopy, Electron, Transmission/methods , Microscopy, Immunoelectron/methods , Nerve Tissue Proteins/metabolism , Piracetam/analogs & derivatives , Piracetam/chemistry , Piracetam/pharmacology , Protein Conformation
18.
Neuropharmacology ; 54(4): 715-20, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18207204

ABSTRACT

SV2A, a synaptic vesicle protein, has been recently identified as a binding target for levetiracetam (Keppra). The specific mechanism by which SV2A binding leads to seizure protection has not yet been fully elucidated. However, a functional correlation between SV2A binding affinity and anticonvulsant potency has been observed in the mouse audiogenic seizure model. The present study was undertaken to test whether similar correlations exist in rodent models of partial and generalized epilepsies. As expected, there was a high degree of correlation between anticonvulsant potency and SV2A binding affinity in the mouse audiogenic seizure model (r(2)=0.77; p<0.001). A similar correlation was also observed in the mouse corneal kindling (r(2)=0.80; p<0.01) and in the rat model of generalized absence epilepsy (GAERS) (r(2)=0.72; p<0.01). Moreover, there were no significant differences between the slopes and intercepts of regression lines in these models. Interestingly, the protective potencies in these three epilepsy models were also well correlated with each other. As such, protective doses of a given SV2A ligand in one model could be easily predicted based on the data obtained in another model. Taken together, these results support the concept that SV2A protein is an important target for both partial and generalized epilepsies and thereby relevant for the generation of new antiepileptic drugs with potential broad-spectrum efficacy.


Subject(s)
Epilepsies, Partial/metabolism , Epilepsy, Generalized/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Acoustic Stimulation/adverse effects , Animals , Anticonvulsants/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Epilepsies, Partial/diet therapy , Epilepsies, Partial/etiology , Epilepsy, Generalized/drug therapy , Epilepsy, Generalized/etiology , Female , Inhibitory Concentration 50 , Ligands , Male , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Binding/drug effects
19.
Neurotherapeutics ; 4(1): 117-22, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17199025

ABSTRACT

Better pharmacotherapies for epilepsy are needed for patients who are refractory to or have tolerability difficulties with current treatments. Seletracetam, a new drug in epilepsy development, is a pyrrolidone derivative structurally related to levetiracetam (trade name Keppra). It was discovered because of its high binding affinity to the synaptic vesicle 2A (SV2A) protein, which is now known to be the binding site for this family of compounds. Seletracetam shows very potent seizure suppression in models of acquired or genetic epilepsy, as well as high CNS tolerability in various animal models. Pharmacokinetic studies in animals suggest that seletracetam is rapidly and highly absorbed, with linear and time-independent pharmacokinetics. Seletracetam appears neither to inhibit nor to induce the major human drug metabolizing enzymes, and it demonstrates low plasma protein binding (<10%), which suggests a low potential for drug-drug interactions. Initial studies in humans demonstrated first-order monocompartmental kinetics with a half-life of 8 h and an oral bioavailability of >90%. Studies in healthy volunteers showed that the treatment emergent adverse events were of mild to moderate severity, were mostly of CNS origin and were resolved within 24 h. Altogether, these results suggest that seletracetam represents a promising new antiepileptic drug candidate, one that demonstrates a potent, broad spectrum of seizure protection and a high CNS tolerability in animal models, with initial clinical findings suggestive of straightforward pharmacokinetics and good tolerability.


Subject(s)
Anticonvulsants , Brain/drug effects , Epilepsy/drug therapy , Pyrrolidinones/pharmacology , Animals , Clinical Trials as Topic , Humans
20.
Proc Natl Acad Sci U S A ; 101(26): 9861-6, 2004 Jun 29.
Article in English | MEDLINE | ID: mdl-15210974

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.


Subject(s)
Anticonvulsants/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Piracetam/metabolism , Animals , Binding Sites , Brain/cytology , Brain/metabolism , Fibroblasts , Gene Deletion , Humans , Inhibitory Concentration 50 , Intracellular Membranes/metabolism , Levetiracetam , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Molecular Weight , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Photoaffinity Labels , Piracetam/analogs & derivatives , Precipitin Tests , Protein Binding , Rats , Seizures , Synaptic Vesicles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...