Epilepsy & Behavior 22 (2011) 617–640
Contents lists available at ScienceDirect
Epilepsy & Behavior j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / ye b e h
Review
The NMDA receptor complex as a therapeutic target in epilepsy: a review Mehdi Ghasemi a,⁎, Steven C. Schachter b a b
Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Departments of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
a r t i c l e
i n f o
Article history: Received 13 April 2011 Revised 1 July 2011 Accepted 18 July 2011 Available online 4 November 2011 Keywords: Epilepsy Seizure NMDA receptor NMDA receptor antagonist
a b s t r a c t A substantial amount of research has shown that N-methyl-D-aspartate receptors (NMDARs) may play a key role in the pathophysiology of several neurological diseases, including epilepsy. Animal models of epilepsy and clinical studies demonstrate that NMDAR activity and expression can be altered in association with epilepsy and particularly in some specific seizure types. NMDAR antagonists have been shown to have antiepileptic effects in both clinical and preclinical studies. There is some evidence that conventional antiepileptic drugs may also affect NMDAR function. In this review, we describe the evidence for the involvement of NMDARs in the pathophysiology of epilepsy and provide an overview of NMDAR antagonists that have been investigated in clinical trials and animal models of epilepsy. © 2011 Elsevier Inc. All rights reserved.
1. Introduction Epilepsy is one of the most common neurologic disorders encountered in clinical practice, affecting approximately 2 to 4 million people in the United States or 1 in 50 children and 1 in 100 adults [1]. Over the last two decades, a new generation of antiepileptic drugs (AEDs) has emerged for the pharmacological management of seizures. Concurrent with that development, the concept of optimum therapy for seizures has evolved to include complete control of seizures, absence of bothersome side effects, and an emphasis on maximizing quality of life [2]. However, the prospect of freedom from seizures and adverse effects remains elusive for a considerable number of patients with epilepsy despite concerted attempts by their physicians to utilize available pharmacotherapies to their full advantage. Approximately 25 to 30% of patients continue to suffer from seizures despite state-ofthe-art treatment [3]. Epileptic syndromes that are in most cases resistant to therapy with conventional AEDs comprise, for example, Lennox–Gastaut syndrome, West syndrome, early myoclonic encephalopathy, myoclonic astatic seizures and severe myoclonic epilepsy in infancy [4]. The presence of bothersome side effects diminishes quality of life for many patients as well, suggesting the need for better tolerated AEDs. It is well established that alterations in central inhibitory (e.g. γaminobutyric acid or GABA) and excitatory (e.g. glutamate) neuro-
⁎ Corresponding author. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA. E-mail addresses:
[email protected],
[email protected] (M. Ghasemi),
[email protected] (S.C. Schachter). 1525-5050/$ – see front matter © 2011 Elsevier Inc. All rights reserved. doi:10.1016/j.yebeh.2011.07.024
transmission play a pivotal role in the etiology of epilepsy. It has been accepted that overstimulation of glutamatergic transmission and thereby activation of glutamate receptors may be of significant relevance for its clinical manifestations [5]. Among glutamate receptors, N-methyl-D-aspartate receptors (NMDARs) have been the focus of much basic and clinical research over the past two decades, producing an overwhelming body of evidence that blocking or suppressing NMDARs is effective in the prevention of and, in some cases, reversal of pathology in various models of neurological diseases, including epilepsy. The repertoire of NMDAR-targeted drugs in neurology is expected to grow in the near future. In this review we provide a brief overview of the rationale for the development of such drugs and then focus on NMDAR-targeted drugs currently used in the clinical setting and trials for the treatment of epilepsy as well as those evaluated in preclinical studies. 2. The NMDAR complex Glutamate is the major excitatory neurotransmitter in the central nervous system (CNS) and acts on ionotropic and metabotropic glutamate receptors located at the presynaptic terminal and in the postsynaptic membrane at synapses in the brain and spinal cord. NMDARs are tetrameric structures of seven subunits including at least one copy of an obligatory subunit, NR1, and varying expression of a family of NR2 (NR2AD) or NR3 (NR3A-B) subunits, with multiple binding sites including for glutamate, polyamine, Mg2+, and glycine (Fig. 1). Pharmacological regulation of the NMDAR depends on effects on unique combinations of subunit-specific binding sites. Both the NR1 and NR2 subunits contribute to the formation of the NMDAR ion channel. The glutamate-binding site is on the NR2 subunits, and the glycine-binding site is located on the NR1 subunits. The glycine (and/or D-serine) coagonist site must be
618
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
Fig. 1. NMDAR model showing binding sites for agonists and antagonists. The extracellular portions of NR1 and NR2 subunits consist of two domains, the modulatory domain and the agonist binding domain. Glycine and D-serine are agonists for NR1 subunits, whereas glutamate and NMDA act on the NR2 subunit agonistic site. Kynurenic acid is an endogenous ligand for NR1. Zn2+ is an endogenous ligand for NR2A and NR2B modulatory domains. Low concentrations (nanomolar) of Zn2+ have affinity for the NR2A subunit, whereas higher concentrations (micromolar) have affinity for the NR2B subunit. Zn2+ at higher concentrations can also act as an NMDAR pore blocker. Ifenprodil and its derivatives bind at the modulatory domain in an NR2B-selective manner. D-AP5 is an antagonist for the agonistic site of NR2 subunit. Mg2+, MK-801, ketamine, PCP, and memantine act as noncompetitive antagonists whose binding sites are within the ion channel pore region.
occupied before glutamate can activate the ion channel. Although NMDAR channels can conduct Na + and Ca 2+, under basal conditions the channel is blocked by Mg 2+ within the channel pore. The Mg 2+ blockade is relieved by cellular depolarization, which has implications for synaptic plasticity, especially long-term potentiation (LTP). Continuous strong stimulation optimally activates NMDARs and plays an important role in LTP. With neurotoxic insults, disruption of energy metabolism diminishes the driving force for the Na + pump that maintains the resting membrane potential of cells so that neurons become depolarized, relieving the Mg 2+ block of NMDARs. Excess Ca 2+ entry then leads to neuronal excitotoxicity and even cell death. Therefore, NMDAR-mediated responses contribute to the later components in paroxysmal depolarizing shifts and provide for much of the Ca 2+ entry associated with seizure discharges. 3. NMDAR alteration in patients with epilepsy Considering the fact that NMDAR activity plays a major role in neuronal excitation in the CNS, some investigators have evaluated the possible alterations of NMDARs in epilepsy, using a variety of methods such as assessment of subunit gene expression, immunoblotting, and binding affinities. Glutamatergic impulses from the entorhinal cortex constitute the major excitatory input to the hippocampus and a shift in glutamate-mediated excitability may be involved in the pathogenesis of epileptic discharges [6]. Moreover, NMDARs may be responsible for the seizure-induced selective excitotoxic cell death of certain hippocampal neuronal populations inasmuch as NMDAR antagonists provide protection against such damage [7]. Using non-radioactive situ hybridization methods, Bayer et al. in 1995 [8] demonstrated that in situ hippocampal specimens of patients with chronic temporal lobe epilepsy showed a loss of NR1-positive cells that was closely related to the overall neuronal
loss in the resected specimen and to Ammon's horn sclerosis. They suggested that loss of NR1 expression may partly reflect pyramidal cell loss [8]. Further investigation revealed that NR2 subunit mRNA levels were increased in the hippocampus of patients with hippocampal sclerosis (HS) [9]. In the dentate gyrus, there appears to be an increase in NR2 immunoreactivity that is associated with abnormal mossy fiber sprouting in this region [10]. Human hippocampal studies suggest that mossy fiber sprouting is associated with physiologically active NMDARs. It has been consistently demonstrated that inhibition of the glutamate binding site (NR2 subunit) decreases granule cell hyperexcitability in cases showing mossy fiber sprouting in hippocampi, whereas granule cells in non-sprouted hippocampi were not affected by such treatment [11–13]. Mathern et al. [10] demonstrated that, compared with autopsy hippocampi, non-HS and HS patients showed increased NR2A and NR2B hybridization densities per dentate granule cell. Non-HS hippocampi also showed increased NR1 and NR2B mRNA levels per CA2/3 pyramidal neuron compared with autopsy cases. Patients with HS, by contrast, showed decreased NR2A hybridization densities per CA2/3 pyramidal neuron compared with non-HS and autopsy cases. They concluded that chronic temporal lobe seizures are associated with differential changes in hippocampal NR1 and NR2A–D hybridization densities that vary by subfield and physio-pathological category. Therefore, in patients with temporal lobe epilepsy, these findings support the hypothesis that NMDARS are increased in dentate granule cells and further that excitatory postsynaptic potentials should be strongly NMDA-mediated compared with non-epilepsy autopsies [14,15]. Using human focal cortical dysplasia specimens obtained during epilepsy surgery, Crino et al. [16] reported that NR2B and NR2C subunit mRNA was increased, and NR2A subunit mRNA was decreased in dysplastic compared with pyramidal and heterotopic neurons (which were microdissected from postmortem control cortex and from temporal cortex without dysplasia
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
resected during temporal lobectomy). These differential mRNA expressions of NMDAR subunits in dysplastic and heterotopic neurons demonstrate cell specific gene transcription changes in focal cortical dysplasia [16]. More recent studies [17,18] reported an upregulation of NR2B subunit composition and an altered Mg2+ sensitivity in pediatric cortical dysplasia or adult temporal lobe tissues. Some studies have also suggested a role for NMDARs in the genesis of seizures arising from cortical tubers in the tuberous sclerosis complex [19]. White et al. [19] demonstrated an increase in the level of NR2B and 2D subunit mRNAs and functional NR2B-containing receptors (using a ligand-binding method) in tubers. Enhanced expression of NR2B and 2C subunit mRNAs was noted in the dysplastic neurons, whereas only the NR2D mRNA was upregulated in giant cells, indicating that dysplastic neurons and giant cells make differential contributions to epileptogenesis in tuberous sclerosis complex [19]. Considering the results of previous studies, it seems that the type of epilepsy, its anatomical involvement and the underlying diseases that could be responsible for seizure development (such as tuberous sclerosis) could contribute to the variable alterations in different subunits of NMDARs (especially NR1 and NR2 subunits) in different brain regions. Although more investigation is needed to specify the role of each NMDAR subunit in pathophysiologic aspects of epilepsy, targeting specific NMDAR subunits may provide new insights into the control of seizures. For example, because the NR1 subunit is an obligatory subunit of NMDARs, inhibition of this subunit could cause a global inhibition of NMDAR within all areas of the CNS in which NMDARs are present, leading to significant adverse effects on brain function. Accordingly, in patients with antiNMDAR encephalitis, it has been suggested that autoantibodies against NMDARs cause a reversible reduction in NMDAR function on the cell surface of neurons [20–23]. Autoantibodies in the serum or cerebrospinal fluid (CSF) of these patients were also reported to bind specifically to an epitope located in the extracellular domain of NR1 subunits [24]. These patients initially present with a non-specific flulike prodrome followed by a psychosis (bizarre behavior, disorientation, hallucinations, paranoid thoughts, memory deficits and confusion). In the following stage, neurologic symptoms (e.g. decreased
619
consciousness, lethargy and autonomic instability) are prominent and a considerable number of patients develop seizures and status epilepticus. These symptoms can be improved by immediate treatment with immunomodulatory therapy [23]. 4. NMDAR alteration in preclinical studies 4.1. Overview of animal models of epilepsy As summarized in Table 1, a wide variety of animal models of epilepsy and status epilepticus have been used to study potential anticonvulsants, including electrical stimulation models, chemoconvulsant-induced models (e.g. kainic acid (KA), pilocarpine, picrotoxin or bicuculline); physical models (e.g. hyperthermia, or photic or auditory stimulation), genetic models (e.g. mutant, transgenic or knockout) and spontaneous seizure models (e.g. post-kindling or post-chemoconvulsant). The maximal electroshock seizure (MES) test is predictive of activity against generalized tonic-clonic seizures (Table 2). The subcutaneous (s.c.) pentylenetetrazole (PTZ or metrazol) test is thought to be predictive of activity against nonconvulsive (absence or myoclonic) seizures [25]. However, some AEDs (e.g. levetiracetam) that protect against nonconvulsive seizures in patients failed in the s.c. PTZ test (for a review see [26]). Although these two acute animal seizure tests are suitable screening tools for the identification of potential anticonvulsant agents, they are not closely related to human epilepsy, but rather represent models for induction of single epileptic seizures. Moreover, the MES test can fail to identify drugs that are clinically effective in treating partial seizures (e.g. vigabatrin, tiagabine, and levetiracetam) or for identification of drugs for pharmacoresistant epilepsies (for a review see [27]). Chronic models of epilepsy can be divided into models of acquired (symptomatic) epilepsy and models of genetic (idiopathic) epilepsy (Table 2). The first category includes models in which epilepsy or epilepsy-like conditions are induced by electrical or chemical methods in previously healthy (non-epileptic) animals, mostly rats. Models with electrical induction of epilepsy or epilepsy-like conditions include the kindling model and models in
Cobalt-homocystine Lithium-pilocarpine Recurrent stimulation Brain slices Rodent hippocampal slices Isolated cell preparations Human neurosurgical tissue Injection into area tempesta KA Kindling Tetanus toxin
Simple partial (acute)
Bilateral cortical foci General rat models Y-Hydroxy-butyrate Intraventricular opiates Systemic penicillin Thalamic stimulation
Acute electrical stimulation GABA-withdrawal Neocortical brain slices Topical convulsants Anticholinergics Bicuculline Cholinergics Penicillin Picrotoxin Strychnine Other
Simple partial (chronic)
Generalized absence Status epilepticus
Chemical convulsants Bemegride Bicuculline Cocaine Methionine sulfoximide NMDA PTZ Picrotoxin Penicillin Other Genetic Audiogenic seizures in mice Drosophila shakers Genetically prone rats GAERS Mongolian gerbil Photosensitive baboons Totterer and El mice MES test Metabolic derangements Drug withdrawal Hyperbaric oxygen Hypercarbia Hyperthermia Hypoglycemia Hypoxia Uremia
Complex partial
Generalized tonic-clonic or clonic
Table 1 Preclinical animal studies in epilepsy research.
Cortically implanted metals Aluminum hydroxide Cobalt Iron Tungsten Zinc Cryogenic injury Ganglioside antibody injections Systemic focal epileptogenesis
Abbreviations: GAERS, Genetic Absence Epilepsy Rat from Strasbourg; KA, kainite; MES, maximal electroshock; PTZ, pentylenetetrazole.
620
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
Table 2 Overview of acute and chronic models of epilepsy that are currently used widely in epilepsy research.
Acute Models of Seizures (Screening Tools) With electrical stimulation With chemical stimulation
Chronic Models of Epilepsy Models of acquired (symptomatic) epilepsy
With electrical stimulation
With chemical stimulation Models of genetic (idiopathic) epilepsy
Spontaneous mutations in diverse animal species Induced mutations in mice
Test
Related seizure type in humans
MES test in rodents PTZ test
Tonic–clonic seizures Absence Seizures
Kindling Post-status models in which epilepsy develops after an electrically induced status epilepticus Post-status models in which epilepsy develops after a chemically induced status epilepticus (e.g. pilocarpine and KA models of epilepsy) Mutant animals with “reflex epilepsy” (e.g. audiogenic seizuresusceptible rats and DBA/2 mice, photosensitive baboons, or E1 mice) Mutant animals with “spontaneous recurrent” seizures (e.g. GAERS) Transgenic or knock-out mice
Temporal lobe epilepsy (TLE) Status epilepticus Status epilepticus Generalized tonic–clonic seizures in response to sensory stimulation Generalized tonic–clonic seizures Generalized tonic–clonic seizures
Abbreviations: GAERS, Genetic Absence Epilepsy Rat from Strasbourg; KA, kainite; MES, maximal electroshock; PTZ, pentylenetetrazole.
which recurrent spontaneous seizures develop after a self-sustained status epilepticus (SSSE), which is elicited by sustained electrical stimulation of the hippocampus, the lateral or basolateral nucleus of the amygdala, or other limbic brain regions (for a review see [28,29]). The kindling, pilocarpine, and KA models are wellestablished animal models of limbic epileptogenesis. Post-status epilepsy models, in which SSSE is induced by sustained electrical stimulation of the hippocampus or amygdala, are characterized by neuropathological changes reminiscent of mesiotemporal sclerosis (as encountered in many patients with TLE), and recurrent spontaneous seizures develop after the status. SSSE can also be induced by chemical convulsants, such as pilocarpine and KA. Genetic animal models of epilepsy can be subdivided into animals with spontaneous mutations and animals, usually mice, with induced mutations. Models with spontaneous mutations can be further subdivided into two groups: (i) Mutant animals with “reflex epilepsy”. In this group, seizures are elicited by specific sensory stimulation. Audiogenic seizure-susceptible rats and DBA/2 mice, photosensitive baboons, or the E1 mouse (in which seizures are induced by vestibular stimulation) belong to this category; and (ii) animals with “spontaneous recurrent” seizures. For example, the Genetic Absence Epilepsy Rat from Strasbourg (GAERS) has spontaneous spike-wave discharges. Mutant animals, such as transgenic or knockout mice, are rarely used for drug studies (Table 2). 4.2. NMDAR alteration in animal models Using several animal models of epilepsy, investigators have attempted to identify possible alterations in NMDARs. There are variable results among these studies which could be due to differences in the animal models, brain regions, and NMDAR subunits that were examined (Table 3). In 1993, Pratt et al. [30] reported that in partially kindled rats (10 stimulations), while the NR1 subunit mRNA remained unaltered after a period of 2 h, the NR2A and NR2B subunit mRNAs were bilaterally reduced in dentate gyrus granule cells. They also found that in fully kindled animals (40 stimulations), there was a progressive reduction in NR1 subunit mRNA levels in the dentate gyrus, being maximal after 4 h. At the same time point, NR2A and NR2B transcription levels were transiently increased [30]. When studied 28 d after the last evoked seizure, Kraus et al. [31] found that kindling induced a 2.8-fold increase in the number of binding sites for the competitive NMDAR antagonist 3-[(±)-2-(carboxypiperazin-4-yl)][1,2-3H-]propyl-1phosphonic acid ( 3H-CPP). This increase was confined to region CA3 within the hippocampus. However, transcription levels of the
NMDAR genes NR1, NR2A, NR2B, NR2C, and NR2D were not altered by kindling [31]. In another study, in situ hybridization revealed that in amygdaloid kindled rats, there was an increase in the NR2B and decrease in NR2D subunit mRNA levels in the neuroendocrine cells of the supraoptic nucleus one month after kindling, whereas NR1 and NR2C mRNA levels were not altered [32]. Jensen et al. [33] found that seizures result in a rapid significant increase in NR1 mRNA and protein expression in rat cerebral cortex. The NR1 subunit exists in several alternate splice isoforms. Transient reductions in specific isoforms and more sustained changes in splicing at the Cterminal have also been described [34,35]. Northern blots showed a sustained increase in cortical mRNA for NR1 in the amygdaloid kindled rat [36]. Using in situ hybridization histochemistry, GerfinMoser et al. [37] demonstrated that application of picrotoxin (500 μM) for two days led to decreases in the NR2A and NR2B mRNA levels in rat hippocampal slice cultures, whereas NR1 mRNA was not altered. Mathern et al. [38], using both kindled animals and self sustained limbic status epilepticus (SSLSE) rats, reported that the molecular layer of hippocampus in SSLSE rats with spontaneous seizures demonstrated higher levels of NR2A and NR2B immunoreactivity compared to kindled animals and controls. They suggested that hippocampal NR2A and NR2B mRNAs and proteins are differentially increased in association with spontaneous, but not kindled, seizures [38]. Another study indicated that hippocampal NR1 and NR2B mRNA levels change as rats progress from the latent to chronic seizure phase in the pilocarpine model of spontaneous limbic epilepsy and that NR1 subunit alterations correlated with mossy fiber sprouting [39]. In the KA-induced model of limbic seizures in rats, KA-induced seizures decrease NR1 mRNA levels in CA1 and CA3 pyramidal cells, but not dentate gyrus, at 24 and 72 h after drug injection [40]. Moreover, at 3 h after KA, an increase in binding of [ 3H]MK-801 in several apical dendritic fields of pyramidal cells is found [40]. However, pilocarpineinduced seizures cause a significant reduction in both CA1 pyramidal cells and dentate gyrus at 24 and 72 h, and CA3 at only 72 h after drug injection. The binding of [ 3H]MK-801 is not changed at 3 h after pilocarpine injection, whereas it is decreased in stratum lucidum at 3 and 24 h after drug injection [40]. These data point to some differences in hippocampal NMDAR regulation in the pilocarpine and KA models of limbic seizures. In DBA/2 mice, which are prone to audiogenic seizures, NR1 antisense administration resulted in short-term anticonvulsant protection; with the seizure response gradually returning to control levels 12 to 24 h following the termination of antisense administration [41]. Autoradiography ([ 3H]-MK 801 and -CGP 39653 binding) also revealed that NR1 subunit levels were significantly reduced in
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
621
Table 3 Alteration in NMDAR subunit mRNA expression in animal models of epilepsy. NMDAR subunit
Animal model of epilepsy
Brain region
Results
Reference
NR1
Rapid partial hippocampal kindling in rats (10 stimulations) Rapid fully hippocampal kindling in rats (40 stimulations)
Granule cells of the dentate gyrus
[30]
Hippocampal kindling in rats Chronic ethanol exposure in rats Picrotoxin-induced epileptiform discharge Amygdaloid kindling in rats
CA3 pyramidal cells of hippocampus Cortex or hippocampus Rat hippocampal slice cultures (after 2 days) Supraoptic nucleus (after one month) Ipsilateral frontal and temporal cortices at 4 weeks after the last generalized seizure Bilateral piriform cortices at 4 weeks after the last generalized seizure Hippocampal dentate gyrus (after 24 and 72 h) Hippocampal CA1 and CA3 pyramidal cells (after 24 and 72 h) Hippocampal dentate gyrus, CA1 (after 24 and 72 h) and CA3 (only after 72 h) pyramidal cells Fascia dentate, CA1 and CA4 CA1
↔ ↓ Progressive ↔ ↔ ↔ ↔ ↑
Kainate-induced seizure (model of limbic seizures) in rats Pilocarpine model of limbic seizures in rats Pilocarpine model of spontaneous limbic epilepsy in rats
Latent phase (day 11) Early seizure phase (day 25) Chronic seizure phase Subiculum (day 85) Audiogenic seizures in albino Swiss mice acoustically primed at 10 days Central nucleus of the inferior colliculus NR2A
NR2B
Rapid partial hippocampal kindling in rats (10 stimulations) Rapid fully hippocampal kindling in rats (40 stimulations) Hippocampal kindling in rats Picrotoxin-induced epileptiform discharge Self sustained limbic status epilepticus rats Rapid partial hippocampal kindling in rats (10 stimulations) Rapid fully hippocampal kindling in rats (40 stimulations) Hippocampal kindling in rats Picrotoxin-induced epileptiform discharge Amygdaloid kindling in rats
NR2C
NR2D
Self sustained limbic status epilepticus in rats Pilocarpine model of spontaneous limbic Latent phase (day 11) epilepsy in rats Early seizure phase (day 25) Chronic seizure phase (day 85) Hippocampal kindling in rats Amygdaloid kindling in rats Audiogenic seizures in albino Swiss mice acoustically primed at 10 days Hippocampal kindling in rats Amygdaloid kindling in rats
the retrosplenial cortex and the overall cortex after NR1 antisense administration [41]. In albino Swiss mice acoustically primed at 10 days, some subsequently show audiogenic seizures at 20 days. These sound sensitive mice show a marked transient expression of NR2C in the central nucleus of the inferior colliculus (not seen in the non-sensitive mice) and a doubling of the expression of NR1 [42]. It was suggested that the audiogenic susceptibility may be related to the transient expression of the NR2C subunit during a brief neonatal period during which synaptic reorganization happens [42]. Pumain et al. [43] found that in the cortex of rats with absence type seizures, the response to NMDA appears to be enhanced. An evaluation of ethanol withdrawal seizures revealed that chronic ethanol administration results in increased levels of [ 3H]MK-801 recognition sites on NMDARs [44]. Because MK-801 labels all NMDARs, the change presumably involves the number of receptors, not only the recognition sites. However, NR1 subunit mRNAs were not altered following chronic ethanol exposure in rat cortex or hippocampus [44].
Granule cells of the hippocampal dentate gyrus CA3 pyramidal cells of hippocampus Rat hippocampal slice cultures (after 2 days) Molecular layer of hippocampus Granule cells of the hippocampal dentate gyrus
CA3 pyramidal cells of hippocampus Rat hippocampal slice cultures (after 2 days) Supraoptic nucleus and adjacent piriform cortex, lateral nucleus of the olfactory tract, anterior cortical amygdaloid nucleus and cortex–amygdala transition zone (after one month) Molecular layer of hippocampus Hippocampal fascia dentate Subiculum
↓ ↔ ↓ ↓
[40]
↑
[39]
↓ ↑ Transient ↓ ↑ Transient ↔ ↓ ↑ ↓ ↑ Transient ↔ ↓ ↑
[42]
↑ ↑
[38] [39]
Subiculum
↓
CA3 pyramidal cells of hippocampus Supraoptic nucleus (after one month) Central nucleus of the inferior colliculus
↔ ↔ ↑ Transient ↔ ↓
CA3 pyramidal cells of hippocampus Supraoptic nucleus (after one month)
[31] [44] [37] [32] [36]
[30] [31] [37] [38] [30]
[31] [37] [32]
[31] [32] [42] [31] [32]
5. NMDAR modulators for control of seizures 5.1. Felbamate Felbamate (Felbatol ®) is a propanediol dicarbamate derivative (Fig. 2) which was synthesized in 1955 and submitted to the antiepileptic drug development program within the epilepsy branch of the National Institute of Neurological Disorders and Stroke in 1982 [45]. The mechanisms of action of felbamate have not been completely elucidated yet, though several have been suggested: (a) Inhibition of voltage-sensitive Na + or Ca 2+ channels [46,47]. (b) Potentiation of GABA-induced chloride currents [48], although the direct effect on GABA receptors is unclear [49,50]. (c) Inhibition of the NMDAR. However, the exact site of action is still unclear. Some researchers reported that felbamate binds to the strychnine-insensitive glycine site [51–53]. Others have suggested that felbamate binds to a site within the NMDAR channel
622
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
Gastaut syndrome in children [60,61], a seizure disorder characterized by multiple seizure types (i.e. atypical absence and atonic seizures), slow spike-wave electroencephalographic patterns and mental retardation. Felbamate-treated patients with Lennox–Gastaut syndrome were reported to have a robust decrease in the frequency of atonic seizures (drop attacks) and total seizures compared to placebo groups [61]. Currently, felbamate is not used as a first-line AED and generally is used only for patients with intractable partial seizures, infantile spasms, or Lennox–Gastaut syndrome [62–65]. Two rare but serious adverse effects related to felbamate, aplastic anemia and hepatotoxicity, have limited the use of this AED to patients for whom the potential benefits outweigh the risks. 5.2. Magnesium sulfate
Fig. 2. Chemical structure of some common NMDAR antagonists.
pore and acts as an open channel blocker [48]. There is also evidence that felbamate is a non-competitive, allosteric inhibitor with some modest selectivity for NR2B-containing receptors that might also be associated with the channel pore [54,55]. Felbamate was approved by the U.S. Food and Drug Administration (FDA) in 1993 [56–59]. Felbamate has also been studied in Lennox–
Clinical observations in patients [66–68] and experimental investigations in animals [69] have shown that magnesium (Mg 2+) depletion leads to a marked irritability of the nervous system, eventually resulting in seizures or increased susceptibility to seizureinducing stimuli [70,71]. The possibility that convulsions may occur in the setting of Mg 2+ deficiency led some investigators to study the metabolism of this metal in the epilepsies, and a trend towards low blood concentrations was usually found [72,73]. For example, Mg 2+ levels in serum and CSF were significantly lower in patients with tonic-clonic seizures than in controls [74–76]. Serum and CSF Mg 2+ levels fell with increasing duration of epilepsy and frequency of seizures. Within the brain, Mg 2+ preferentially accumulates within the cortex and hippocampus [77]. Magnesium sulfate enters the cerebrospinal fluid and brain after systemic administration [77]. A rise in brain Mg 2+ concentration is associated with an elevation of the seizure threshold and a marked resistance in animal models to electrically- as well as NMDA-stimulated hippocampal seizures [77]. Seizure activity facilitates blood-brain barrier penetration, leading to elevated magnesium concentrations in the cerebrospinal fluid after systemic injection [78]. Hallak et al. in 1994 [79] provided autoradiographic binding data indicating that Mg 2+ exerts its central action (beside other proposed mechanisms) through an inhibition of the NMDAR. Further experiments demonstrated that Mg 2+ blocks Ca 2+ within the NMDAR channel. When the Mg 2+ blockade is relieved by cellular depolarization, the channel is unblocked and Ca 2+ and Na + enter the postsynaptic neuron as potassium exits [80]. Systemic administration of Mg 2+ exerts an anticonvulsant effect in cats and dogs with experimentally-induced epileptic foci [81], and in models of status epilepticus [82], audiogenic and PTZ-induced clonic seizures [83–85], and NMDA-induced hippocampal seizures [86] in rodents. In humans, parenterally administered Mg 2+ is an effective treatment for the seizures of neonatal tetany [87] and eclampsia [88] and possibly for those associated with ethanol withdrawal and acute intermittent porphyria [89,90]. In spite of its effectiveness in controlling hyperexcitability and seizures associated with toxemia of pregnancy, there is still controversy over magnesium's merits as an anticonvulsant [91,92]. Nonetheless, studies have reported that Mg 2+ could be beneficial to augment the therapeutic effects of adrenocorticotropic hormone on infantile spasms (or West syndrome) [93], which is characterized by early onset (4–7 months), unusual seizure forms (repetitive flexor, extensor or flexor–extensor spasms), EEG hypsarrhythmia, and poor seizure and intellectual prognosis [94]. A recent study also reported that intravenous Mg 2+ therapy improved refractory epilepsy with recurrent status epilepticus and episodes of epilepsia partialis continua in two patients with juvenile-onset Alpers' syndrome [95]. Still, however, there are concerns regarding the possibility of hypermagnesemia toxicity in eclampsia treatment. Total magnesium serum concentrations advocated for the treatment of eclamptic convulsions are 3.5 to 7 mEq/L (4.2 to 8.4 mg/dL) [96,97]. Areflexia, particularly loss of the patellar deep tendon reflex, has been observed
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
at 8 to 10 mEq/L, and respiratory paralysis occurs at N13 mEq/L [98]. Progressively higher serum magnesium levels can ultimately lead to cardiac arrest [99].
623
The adverse reactions associated with remacemide include dizziness, somnolence and gastrointestinal symptoms when given as monotherapy. Adjunctive therapy with the conventional AEDs carbamazepine and/or phenytoin might result in diplopia and fatigue [116].
5.3. Remacemide 5.4. ADCI Remacemide hydrochloride [(±)-2-amino-N-(1-methyl-1,2diphenylethyl)-acetamide monohydrate] is a racemate (Fig. 2). Biotransformation of remacemide yields its desglycinated bioactive metabolite, APL 12495AA, which is about 2-fold more potent than remacemide itself. The two enantiomers of remacemide and those of APL12495AA have been tested for their anticonvulsant properties and their potencies were different depending on the route of administration and type of seizure test [100,101]. Receptor-binding studies have shown that both remacemide hydrochloride and APL 12495AA displace [ 3H]MK-801 binding from the synaptic membrane fractions of rat cerebral cortex and hippocampus [102]. Another binding study using [ 3H]desglycinylremacemide demonstrated a binding site within the NMDAR ion channel, which is thought to be associated with the benzomorphan attachment site of the NMDAR [103]. The density of binding sites for [ 3H]desglycinylremacemide was about 2-fold higher in the brainstem and cerebral cortex than in the hippocampus and cerebellum [103]. Electrophysiological studies have shown that both remacemide and APL 12495AA inhibit NMDA-induced responses in a variety of preparations such as cultured rat hippocampal neurons [104] and cortical wedges prepared from audiogenic seizure-prone DBA/2 mice [105]. Besides acting on NMDARs, both remacemide and its metabolite have been reported to be potent Na + fast-channel blockers [106,107]. It is likely that synergism between these two actions occur. Additionally, because of the relatively low affinity of remacemide and its metabolite to the NMDAR complex, the anticonvulsant effects of these agents may be due to their effects on Na + channels. A possible partial blockade of potassium channels has also been proposed [106]. Remacemide pretreatment decreased pyramidal cell damage in the CA3 and CA1 subregions of the hippocampus in a rat model of status epilepticus [108]. Remacemide and APL 12495AA have anticonvulsant actions in a number of animal models including the MES test [101,109,110]; NMDA, kainic acid, electrical kindling and 4aminopyridine-induced seizures [101,111]; cocaine-induced convulsions [112]; spike-and-wave discharges in the WAG/Rij rat; and in the GAERS rat; (which constitutes a genetic model of absence epilepsy) [113,114]. However, remacemide did not protect against bicorneal kindled seizures and those induced by PTZ, bicuculline, picrotoxin, and strychnine [101,109,110] or by acute heat stress [115]. Remacemide was also investigated as adjunctive therapy in several clinical studies of patients with drug-resistant seizures. In a doubleblind, placebo-controlled, cross-over study with remacemide hydrochloride in 28 patients with drug-resistant seizures, one-third showed a reduction in seizure frequency by 50% or more and four patients were free of seizures during remacemide treatment [116]. In an open study, Owen et al. [117] reported significant improvement. Another placebo-controlled add-on trial in adult patients with drug-resistant seizures demonstrated the percentage of responders (defined as 50% or greater reduction in seizure frequency) was significantly higher in remacemide-treated patients when compared with placebo [118]. Adjunctive remacemide treatment was similarly shown to be associated with a higher, dose-related responder rate than that observed in placebo-administered patients [119]. Another study also suggested that remacemide exhibited therapeutic activity as monotherapy in patients with drug-resistant seizures following presurgical assessment [120]. However, there is evidence that remacemide had no benefit in patients with newly diagnosed epilepsy compared to other AEDs such as carbamazepine [121], putting into question the efficacy of remacemide as monotherapy.
ADCI [(±)-5-aminocarbonyl-10,11-dihydro-5Hdibenzo[a,d]cyclohepten-5,10-imine] possesses features of carbamazepine and MK-801 [116,122] (Fig. 2). The compound is a selective, low-affinity channel blocker of the NMDAR. Sun and Lin [123] reported that ADCI could also block voltage-gated sodium channels in rat superior cervical ganglion and hippocampal neurons as well as cultured human NT2 neurons. Preclinical studies revealed the ADCI has an anticonvulsant effect in the MES test [122], as well as against 4-aminopyridine-, NMDA-, PTZ- and cocaine-induced seizures [122,124,125]; dendrotoxin-induced clonic seizures [126]; and ethanol withdrawal seizures in mice [127]. ADCI possesses an improved toxicity profile, which may be caused by its greater tendency to block NR1A/NR2B subunits (IC50 = 6.6 μM) than NR1A/NR2A subunits (IC50 = 14.3 μM) [116]. 5.5. Ketamine Ketamine hydrochloride (dl2-(o-chlorophenyl)-2-(methylamino) cyclohexanone hydrochloride or Ketalar®) was developed in 1962 (Fig. 2). Ketamine is primarily used for the induction and maintenance of general anesthesia, usually in combination with sedative drugs. The ketamine-induced anesthetic state is believed to result from functional disorganization of the CNS rather than CNS depression [128]. Ketamine has a low affinity for the NMDAR at the phencyclidine site within the ionotropic channel [129,130], with slow open-channel blocking/ unblocking kinetics, and a specific type of channel closure, called “trapping block” [131]. As early as 1965, McCarthy et al. [132] demonstrated in laboratory animals that ketamine is capable of suppressing convulsions induced by electrical stimuli as well as those induced by intravenous infusion of CNS stimulants such as PTZ or caffeine. Since then, many studies reported that ketamine exerted anticonvulsant effects in a variety of animal models of epilepsy including NMDA-, guanidinosuccinate-, p-toluidino-3-propylamino-2-propanol-, mercaptopropionate-, N-(3,5-dimethoxy-4-propoxyphenylethyl)aziridine-, lidocaine-, picrotoxin-, bicuculline-, strychnine- or PTZinduced seizures in rodents [133–148]; young chick model of epilepsy [149]; sound-induced convulsions in epilepsy prone rats [150,151]; seizures kindled by repetitive electrical stimulation of the rat motor cortex [152]; kindled amygdaloid seizures in rats [153] and kindling epileptogenesis and seizure expression in developing rats [154]; soundinduced seizures in DBA/2 mice [155]; generalized tonic–clonic seizures induced by metrazol in rats [156]; morphine-induced hind-limb myoclonic seizures [157]; limbic status epilepticus induced by 90 min continuous electrical stimulation of the hippocampus [158]; prolonged status epilepticus in rats and dogs [159,160] and pilocarpine-induced status epilepticus in rats [161]; rat model of partial status epilepticus [162]; electrically precipitated tonic hind-limb extension [163]; MES test in mice [164,165]; and the lithium–pilocarpine seizure model in rats [166]. Infusions of ketamine into the substantia nigra pars reticulata of adult rats also increase the latency of onset to seizures induced by ether flurothyl [167]. Ketamine can act synergistically with AEDs such as valproate and carbamazepine to decrease MES-induced seizures in mice [168]. Other studies demonstrated the synergistic action of diazepam and ketamine in terminating status epilepticus induced by either lithium-pilocarpine [169] or kainic acid [170]. In a model of fulminant seizures and seizure-related brain damage induced by soman poison in guinea pigs, sub-anesthetic doses of ketamine (10 mg/kg)
624
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
prevented lethality and stopped ongoing seizures in poisoned animals and proved effective in reducing seizure-related brain damage and in increasing survival [171]. Further investigation also revealed that ketamine, either racemic or active ketamine isomer (S(+)-ketamine), in association with atropine sulfate, was highly effective in the delayed treatment of severe soman intoxication [172]. Ketamine consistently suppressed focal seizures arising from neocortical and hippocampal epileptogenic foci in cats but was ineffective in modifying interictal epileptogenic activity both at the primary and mirror focus [173]. In doses producing narcoticcataleptic effects (50–100 mg/kg, i.p.), it reduced the intensity of picrotoxin convulsions and prevented seizures caused by metrazol administration [174]. Bath application of ketamine blocked penicillininduced, synchronized afterdischarges in immature rat CA3 hippocampal neurons and depressed intracellular depolarizations produced by iontophoretic application of NMDA [175]. Ketamine decreased epileptiform activity in both CA1 and CA3 regions of the rat hippocampus [176,177]. Cunha et al. [178] demonstrated that poststatus epilepticus rats exhibited extensive gliosis and cell loss in the hippocampal CA1, CA3 (70% cell loss for both areas) and dentate gyrus (60%) and that ketamine reduced cell loss in the hippocampus [178]. Other studies revealed that ketamine blocked epileptiform activity induced by alkalosis in rat neocortical slices [179], decreased 4aminopyridine- or picrotoxin- or “magnesium free”-induced epileptogenic activity in hippocampal slices of rats [180,181], attenuated epileptogenic responses in a synaptic and a non-synaptic model of epileptogenesis in the CA1 region of the hippocampal slice [182], and had anticonvulsant effects on different patterns of epileptiform activity in rat temporal cortex slices [183]. Cortical epileptiform afterdischarges (spike-and-wave rhythms) induced by low-frequency stimulation of the sensorimotor cortex were dose-dependently shortened by ketamine [184]. Degeneration of thalamic neurons caused by persistent seizure activity in the corticothalamic tract (putative glutamergic transmitter pathway) was prevented by systemic administration of ketamine or MK-801, despite the failure of these agents to eliminate persistent electrographic seizure activity recorded from cortex and thalamus [185]. Ketamine as well as other NMDAR antagonists (MK-801 and phencyclidine) were also reported to protect against kainic acid-induced seizure-related rat brain damage [186]. A clinical study reported a 23-year-old man whose refractory status epilepticus was refractory to standard AEDs and barbiturates, and was successfully terminated only with intravenous ketamine [187]. Although the above mentioned preclinical and anecdotal clinical evidence suggests a robust anticonvulsant effect of ketamine, other studies showed contrary results. Prior studies by Winters et al. in 1972 [188] reported bursts of hippocampal seizure activity without overt behavioral manifestations in cats receiving single cataleptic doses of ketamine. In addition, Miyasaka and Domino in 1968 [189] and Winters et al. [188] demonstrated a progression of EEG changes in both the mesodiencephalon and cortex which were characterized by initial desynchronization, and then intermittent hypersynchrony (low frequency, high amplitude waves) followed by continuous hypersynchrony. Abnormal spike and hypersynchronous bursting activity was also observed during the course of chronic ketamine (30 or 80 mg/kg/day, i.p.) administration in 50% of rats [190]. Further, withdrawal of the drug for 5 days following three months of treatment resulted in the progressive increase of epileptiform activity without gross behavioral manifestations in rats [190]. Subsequent studies [191] concerned with the ketamine-induced alterations of the EEG in human volunteers showed that, under ketamine anesthesia, the EEG changed from an α rhythm to predominantly θ-wave activity, coincidentally with the loss of consciousness and establishment of “dissociative” anesthesia. Kayama and Iwanae [192] studied, also in cats, the effect of ketamine on spontaneous neocortical EEG patterns, cortical evoked potentials, and cortical single-unit activity. They interpreted
their findings as suggesting drug-induced seizure discharges, and went on to propose that the loss of consciousness in ketamine anesthesia is produced in the same manner as absence seizures interfere with consciousness. Ketamine was shown to exacerbate penicillin-induced generalized corticoreticular epilepsy in cats [193] and to increase seizure duration following electrical stimulation in a rat model of electroconvulsive therapy [194]. Observing the activation of primary and projected cortical foci by ketamine at anesthetic and non-anesthetic doses, DeVore et al. [195] speculated that ketamine is epileptogenic in certain patients with a seizure disorder. Thompson in 1972 [196] reported generalized tonic-clonic seizures in a 26-year-old woman with no past neurological history that began 2 min after an induction dose of 130 mg ketamine (i.v.) given for an elective abortion. Another study by Hirshman et al. in 1982 reported the occurrence of extensor-type seizures in 4 asthmatic patients receiving theophylline within minutes following induction of anesthesia with ketamine [197]. Another study [198] in patients who had depth electrodes implanted in the limbic and temporal regions reported that ketamine (2–4 mg/kg, i.v.) led to pronounced abnormal activity in these brain regions that progressed to electrographic seizures in some of the patients. However, Corssen et al. [199] found no evidence that ketamine (1 mg/lb. body weight, i.v.) precipitated generalized convulsions, even in patients with epilepsy and an abnormal EEG, but, on the contrary, they observed that ketamine suppressed or virtually eliminated epileptiform EEG discharges during clinical seizures. Celesia showed that ketamine neither precipitated nor aggravated seizures and was less effective than natural sleep as an activator of epileptic discharges in patients with epilepsy [200]. The anticonvulsant activity of the drug was also observed in 1968 [201]. The benefit of ketamine in severe febrile convulsions was reported by Davis and Tolstoshev in 1976 [202]. Sheth and Gidal [203] described a 13-year-old girl with seizures refractory to standard status epilepticus protocols, alternative therapeutic strategies, and 4 weeks of pentobarbital coma who achieved control of both clinical and electrographic seizures after the administration of ketamine (2 mg/kg bolus; continuous infusion to a maximum rate of 7.5 mg/kg/h for 14 days). Oral ketamine, administered to five children with severe epilepsy (Lennox–Gastaut syndrome, myoclonic-astatic epilepsy, progressive myoclonic epilepsy and pseudo-Lennox syndrome) during an episode of non-convulsive status epilepticus (NCSE) led to the resolution of NCSE in all cases clinically and electroencephalographically within 24–48 h of starting ketamine [204]. A retrospective series of 6 fatal cases of refractory status epilepticus treated with ketamine suggested some modest efficacy, although there was no mention of the dose regimen used or duration of therapy [205]. A series of seven patients treated with ketamine for refractory status epilepticus reported control of electrographic seizures in four patients on 0.3–5.8 mg/kg/h infusion [206]. Ubogu et al. [207] reported that ketamine infusion following low-dose propofol sedation in a 44-year-old man with treated neurosyphilis presenting with subclinical refractory status epilepticus resulted in the gradual control of electrographic seizures over 72 h. However, they noticed diffuse cerebellar and worsened cerebral atrophy 3 months later, consistent with animal models of NMDAR antagonist-mediated neurotoxicity [207]. A recent study reported the development of refractory status epilepticus in a 22-year-old woman with mitochondriopathy and preexisting epilepsy which was treated with administration of continuous ketamine infusion in addition to midazolam [208]. Based on these clinical observations, ketamine may be useful in controlling malignant status epilepticus. However, the response to ketamine and further characterization of adverse effects in patients need further study. 5.6. Amantadine Amantadine (1-aminoadamantane) was the first member of a class of organic molecules called aminoadamantanes to be introduced into
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
clinical use. It was originally licensed to treat influenza A infections [209], and later parkinsonism [210]. Amantadine appears to act through several pharmacological mechanisms. In the early 1990s, it was shown that amantadine could block NMDARs [211,212]. Subsequent studies revealed that when bound in the channel of NMDARs, amantadine increases the rate of channel closure. As a result, the predominant inhibitory mechanism of amantadine is not blockade of current flow through open channels but rather increasing occupancy of channel closed states [213]. In the 1970s, it was reported that amantadine increased seizure susceptibility in the maximal electroshock test (MES) and PTZ-induced seizures in mice [214–216]. Amantadine (25 and 100 mg/kg) decreased the anticonvulsant effect of diphenylhydantoin in the MES [217] and inhibited the anticonvulsant effects of serotonin (5-hydroxytryptamine or 5-HT) on PTZinduced convulsions in mice at 100 mg/kg [214]. At that time, these effects were explained by its possible actions on the central dopaminergic system. However, Kleinrok et al. [218] demonstrated that although amantadine (up to 100 mg/kg) did not affect PTZinduced convulsions in mice, combined treatment of the GABAergic agonist baclofen (5 mg/kg) and amantadine (100 mg/kg) significantly decreased the number of animals with tonic seizures induced by PTZ. Subsequent studies demonstrated that amantadine had anticonvulsant properties against MES and PTZ-induced tonic convulsions and NMDA-induced lethality in mice [125,147]. Reis et al. [219] showed that oral amantadine intake (50 and 100 mg/kg) decreased human motor cortex excitability in healthy subjects. They suggested that the NMDAR antagonism was the most relevant effect on cortical excitability [219]. Rohrbacher et al. [220] found that amantadine (100 μM) decreased the synaptic excitation intrastriatally evoked in rat neostriatal slices. High doses of amantadine at 200 mg/kg were reported to induce convulsions in mice [221]. Twelve- to 16-week treatment with amantadine in 10 children with medically refractory seizures resulted in an improvement in control of myoclonic or atypical absence seizures without alteration (or even worsening) in tonic–clonic and atonic seizures. Tonic seizures were controlled in one patient, but worsened in another [222]. Another study of 10 adolescents and adults with drug-resistant generalized tonic–clonic, myoclonic, or absence seizures showed that adding amantadine to existing AED regimens in weekly increments to 400 mg/day reduced myoclonic or absence seizures in 4 patients, whereas it worsened tonic–clonic seizures in 3 patients [223]. Shahar and Brand [224], reported that add-on amantadine to four children with refractory absence epilepsy resulted in complete resolution of absence seizures within 1 week. All patients remained free of symptoms for 27 to 36 months without adverse effects related to this drug. An attempt to discontinue the medication in three children resulted in a prompt relapse [224]. Matsushige et al. [225] similarly reported a girl with refractory childhood absence epilepsy who improved with add-on amantadine therapy. These reports indicate that amantadine may be beneficial for refractory absence seizures, especially in children. However, it should be noted that high doses of amantadine could induce seizures in patients [226], consistent with animal studies [221].
625
effects in MES-induced seizures with an ED50 of around 5–10 mg/kg [147,227,229–231]; tonic (but not clonic) seizures in mice induced by PTZ, bicuculline, picrotoxin, 3-mercaptopropionic acid and NMDA with an ED50 of 7–12 mg/kg [125,147,232–236]; cocaine-induced convulsions in mice [237]; ethanol withdrawal-associated audiogenic seizures in rats at 5 and 10 mg/kg [238]; soman-induced seizures in rats at 18 mg/kg [239–241]; electrically precipitated tonic hind-limb extension in mice [163]; and audiogenic convulsions in Krushinskii– Molodkina strain rats at 5 and 10 mg/kg [242,243]. Memantine was also shown to augment the anticonvulsant effects of AEDs such as valproate in the MES test [229]. Memantine depressed the spontaneous absence-like paroxysms in the cortical EEG of rats (5–10 mg/kg) [244], the synaptic excitation intrastriatally evoked in rat neostriatal slices [220], and NMDA- or Mg 2+ free-induced epileptiform activity in area CA1 of guinea pig hippocampal slices [245]. It significantly protected hippocampal and cortical neurons in culture against glutamate and NMDA excitotoxicity [240]. Memantine was also reported to protect against diisopropylphosphorofluoridate (an organophosphate compound)-induced seizures and dendritic degeneration of pyramidal neurons in the CA1 hippocampal area in rats [246]. However, Wang and Bausch in 2004 [247] showed that hippocampal cultures chronically treated with memantine showed increased mossy fiber axons, fewer granule cell layer neurons and slightly increased electrographic seizures. Another study revealed that acute memantine (20 mg/kg) significantly retarded the progression of the dystonic attack in a mutant hamster model of paroxysmal dystonia [248]. Although memantine was ineffective against amygdala kindling in rats when given alone (5–10 mg/kg) [249,250], co-administration of memantine with the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid [251] receptor antagonist NBQX (2,3-dihydroxy-6-nitro-7sulfamoylbenzo[f]quinoxaline) had a supra-additive anticonvulsant effect in the kindling model of epilepsy [249]. The favorable sideeffect profile of memantine and the promising results when used together with NBQX suggest that memantine should be further studied for the treatment of epilepsy, possibly in combination with AMPA receptor blockers. Accordingly, Gmiro and Serdyuk [252] have recently reported that although memantine alone decreased the incidence of PTZ-induced generalized tonic–clonic seizures in rats by 4-fold, IEM-1913 (combined blockade of NMDA and AMPA receptors in the brain) decreased PTZ-induced clonic and tonic– clonic seizures in rats by 4- to 8-fold. The therapeutic index of IEM1913 surpassed that of memantine by 200- to 600-fold [252]. However, it is noteworthy that some studies have reported that memantine may cause seizures in patients. For instance, Peltz et al. [253] reported that a 72-year-old Caucasian woman, taking memantine for Alzheimer's disease, was admitted to the hospital with new-onset, generalized tonic–clonic seizures. After memantine was discontinued, the disturbance resolved and increased Δ waves on the patient's EEG were improved. They suggested that the newonset seizures were associated with memantine treatment [253]. The clinical data regarding the anticonvulsant effects of memantine are scarce and more investigations are clearly needed to verify whether memantine can be used as an AED for some seizure types in patients with epilepsy.
5.7. Memantine 5.8. Dextromethorphan Memantine (1-amino-3, 5-dimethyl-adamantane) is a low-affinity, noncompetitive, open-channel NMDAR antagonist that enters the receptor channel preferentially when it is excessively open, and blocks the NMDAR-associated ion channel similar to Mg 2+ by binding to or near the Mg 2+ binding site. Indeed, memantine is able to inhibit the prolonged influx of Ca 2+ ions, which forms the basis of neuronal excitotoxicity. Preliminary preclinical results in the early 1980s suggested that memantine could be beneficial as an anticonvulsant drug alone or in combination with other AEDs [227,228]. Subsequent preclinical studies revealed that memantine exerted anticonvulsant
The FDA approved dextromethorphan as an over-the-counter antitussive in 1958. Its mechanism of action is via multiple effects, including actions as a nonselective serotonin reuptake inhibitor and a sigma-1 receptor agonist [254]. Dextromethorphan can also act as a noncompetitive NMDAR antagonist; however, its metabolite (dextrorphan) is more potent than dextromethorphan in blocking the NMDAR (Fig. 2). At high doses, the pharmacology of dextromethorphan is similar to that of the controlled substances phencyclidine and ketamine, which also antagonize the NMDAR. A double-blind, crossover,
626
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
add-on study in 9 patients with severe drug-resistant complex partial seizures reported that seizure frequency increased 25% among patients randomized to the dextromethorphan (120 mg/day) arm of the study, although this increase was not clinically significant [255]. However, in a study by Schmitt et al. [256], high doses of dextromethorphan (20–42 mg/kg/day) were given to four critically ill children with drugresistant seizures and frequent epileptiform EEG abnormalities. Treatment with dextromethorphan was started between 48 h and 14 days after onset of the illness. In three patients, the EEG improved considerably within 48 h and seizures ceased within 72 h [256]. Another study also demonstrated that adding dextromethorphan to existing AEDs at doses of 40 and 50 mg every 6 h (160 and 200 mg/day) caused a significant improvement in seizure control in sixteen patients with drug-resistant, localization-related epilepsies after 8 weeks. There were mild and transient adverse effects that were well tolerated [257]. Kazis et al. [258] showed that dextromethorphan given in doses up to 50 mg/6 h can produce plasma and brain concentrations similar to the in vitro anticonvulsant levels, without causing significant adverse effects. Dextromethorphan has anticonvulsant properties in several animal models of epilepsy including amygdaloid kindled convulsions in rats [259]; audiogenic seizures in DBA/2 mice and genetically epilepsy-prone rats [151,155]; MES in mice and rats [147,260,261]; NMDA-induced convulsions and lethality in mice [148,262,263]; PTZinduced clonic seizures in mice [147,260]; seizures induced by cocaine, lidocaine, theophylline, kainate and the calcium channel agonist BAY k-8644 in mice and rats [264–266]; ethanol withdrawal seizures in rats [267]; and soman-induced convulsions and electrographic seizure activity in guinea pigs [268]. Dextromethorphan also prevents the adverse effects of neonatal hypoxia on PTZ-induced seizure threshold in rats [269]. By contrast, dextromethorphan has limited anticonvulsant activity against morphine-induced hindlimb myoclonic seizures in mice [157]. 5.9. Riluzole Riluzole (2-amino-6-trifluoromethoxy benzothiazole) was originally synthesized by researchers in France and early studies in the 1980s suggested it had anticonvulsant properties [270]. The discovery that it can disrupt glutamate neurotransmission to prevent NMDARmediated neuronal death in experimental models [271] promoted its further development. Several mechanisms regarding the glutamatergic neurotransmission have been proposed. First, it inhibits Na + channels on glutamate-containing neurons and thereby selectively reduces presynaptic release of glutamate [272]. Second, riluzole might block NMDAR activation, preventing Ca 2+ entry via the channel. Riluzole either acts directly on the NMDAR, although a binding site for riluzole on the receptor has not been identified [273]; or indirectly, possibly via a G-protein-dependent signaling pathway [274]. Third, riluzole facilitates glutamate reuptake by increasing the activity of glutamate transporters expressed on neurons and glia [275], suggesting a modulatory action on glutamate clearance from the synaptic cleft. Preclinical data have shown that riluzole has anticonvulsant properties in a variety of seizure models such as the MES test in rodents; seizures induced by inhibitors of the synthesis of GABA; ouabain, pilocarpine, L-glutamate and kainite in rodents; seizures induced by photic stimulation in the baboon, Papio papio; sound stimuli in DBA/2 mice; postural seizures in E1 mice; seizures in WAG/Rij rats (an animal model of human absence epilepsy); kindled seizures and the development of behavioral seizures in kindling acquisition with relatively mild correlation to afterdischarge duration; and pilocarpine-induced limbic seizures, γ-hydroxybutyrate lactoneinduced absence seizures, and ethanol withdrawal-induced convulsions in mice [270,276–281]. Riluzole has been shown to augment the anticonvulsant effects of conventional AEDs such as carbamazepine, phenobarbital, valproate and diphenylhydantoin in the MES test in
mice [279], and valproate, phenobarbital, or ethosuximide in PTZinduced seizures in mice [282]. Riluzole is the only FDA-approved medication for amyotrophic lateral sclerosis. However, its benefit in controlling seizures in patients with epilepsy has not been shown in any clinical study and this issue warrants further investigation. 5.10. Cycloserine Cycloserine (4-amino-isoxazolidin-3) was historically one of the antituberculosis agents most significantly associated with CNS toxicity. Cycloserine crossreacts with the glycine site at the NMDAR and at maximally stimulating doses produces approximately half of the maximal facilitation of NMDAR function [283–285]. Under conditions where the occupancy of glycine binding sites by agonists was greater than 50%, glycine binding antagonist-like effects emerged [286]. This antagonist-like effect of cycloserine was reflected in an increase in the off-rate for glutamate from the NMDAR [287]. A number of preclinical studies have shown that cycloserine has anticonvulsant properties in tests such as sound-induced epilepsy in DBA/2J epileptic mice; seizures induced by strychnine, metrazol, isonicotinic acid hydrazide, 3-mercaptopropionic acid, cocaine, kainate and NMDA; tonic convulsions induced by 120 mg/kg of PTZ; and focal seizures in amygdala-kindled rats [233,288–301]. DCycloserine in subprotective doses (2.5 and 10 mg/kg) was also shown to potentiate the anticonvulsant action of phenytoin and carbamazepine in the MES test in mice [300]. D-Cycloserine (2.5 mg/kg) also potentiates the anticonvulsant activity of carbamazepine, diazepam, felbamate, lamotrigine, phenytoin, phenobarbital and valproate against sound-induced seizures in DBA/2 mice [302]. Despite the accumulating evidence regarding the anticonvulsant effects of cycloserine in animal studies, some clinical evidence has suggested that cycloserine, especially in patients with tuberculosis, could have adverse effects including exacerbation of psychiatric conditions as well as seizures [303]. Therefore it is generally not used in patients with convulsions and psychiatric states characterized by signification agitation [303]. 5.11. Dizocilpine Dizocilpine or MK-801 {(+)-5-methyl-10,11-dihydro-5H-dibenzo [a,d]cyclohepten-5,10-imine maleate} is a potent noncompetitive NMDAR antagonist. MK-801 binds inside the ion channel of the receptor at the phencyclidine binding site and thus prevents the flow of ions, including an influx of Ca 2+, through the channel. A large number of preclinical investigations demonstrated anticonvulsant properties in several models including seizures induced by NMDA, quinolinic acid, lindane, 4-aminopyridine, caffeine, picrotoxin, bicuculline, cocaine, kainic acid, strychnine, PTZ or hyperbaric oxygen in rodents [125,135,142,147,234,237,263,304–315]; sensory-evoked electromyographic activity induced by catechol in urethane-anesthetized rats [316]; limbic kindling and kindled seizures in rats [317– 319]; limbic status epilepticus induced by 90 min of continuous electrical stimulation of the hippocampus [158]; chemical kindling induced by PTZ in rats [320]; soman-induced seizures in guinea pigs [321]; alcohol withdrawal seizures in rats [322]; febrile seizures in epileptic chicks [323]; hypoxic stress-induced convulsions and death in mice [324]; electrically precipitated tonic hind-limb extension in mice [163]; kindling epileptogenesis and seizure expression in developing rats [154]; focal seizure activity from the feline hippocampus [325]; seizures induced by putrescine in the deep prepiriform cortex [326]; audiogenic seizures in genetically epilepsy-prone rats [151]; sound-induced seizures in DBA/2 mice [155]; generalized tonic–clonic convulsions in genetically epileptic E1 mice [327]; enflurane-induced opisthotonus in mice [328]; hippocampal seizure threshold and afterdischarge in rats implanted with intracranial electrodes [329]; limbic status epilepticus induced by 90 min of
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
continuous electrical stimulation of the hippocampus [158]; pilocarpine-induced status epilepticus in rats [330]; the MES test in mice [147,305,318,331]; and the lithium-pilocarpine seizure model in rats [332]. Combined treatment of MK-801 and nimodipine has anticonvulsant effects against PTZ- and strychnine-induced seizures in mice [306]. Doses of 0.0031 and 0.0125 mg/kg enhanced the protective activity of valproate against MES-induced convulsions in mice [333]. MK-801 (0.0125 and 0.05 mg/kg) potentiated the anticonvulsant action of phenobarbital, totally eliminating phenobarbital-induced motor impairment at 0.05 mg/kg, but did not influence the protection offered by carbamazepine and diphenylhydantoin at 0.05 mg/kg [333]. Sub-effective doses of MK-801 and lithium also exert anticonvulsant effects in the PTZ model of clonic seizures in mice [135]. Moreover, MK-801 improved the antiepileptic activity of phenobarbital, valproate and nicardipine in the rat amygdala kindling models [334]. These findings offer the possibility that a combination of MK-801 with selected conventional AEDs might have clinical benefit. Troupin et al. in 1986 studied the effects of MK-801 and standard AEDs in human epilepsy. They reported considerable improvement in more than 50% of patients in seizure frequency, mood, and alertness. The promising start of this trial faded with time. However, the patients were primarily receiving carbamazepine or diphenylhydantoin therapy [335]. 5.12. Ifenprodil Ifenprodil (4-[2-(4-benzylpiperidin-1-yl)-1-hydroxypropyl]phenol) selectively blocks the polyamine site at the NR2B subunit of the NMDAR. Several preclinical studies have shown that ifenprodil exerts anticonvulsant effects in a variety of animal models of epilepsy including seizures induced by NMDA, spermine, lindane, and PTZ in rodents [336–340]; spinal seizures in mice induced by handling following pretreatment with a sub-convulsive dose of strychnine [341]; the MES test [230,336,342]; audiogenic seizures in genetically epilepsy-prone rats [151,343]; motor and electrocortical seizures induced by unilateral local injection of putrescine into the rat deep prepiriform cortex [326]; ethanol withdrawal seizures in mice [344]; amygdala-kindling development [345]; proconvulsive effects of interleukin(IL)-1β on electrographic seizures induced by intrahippocampal injection of kainic acid in C57BL6 adult mice [346]; PTZinduced seizures in spermidine/spermine N1-acetyltransferase transgenic mice [347]; cocaine-induced convulsions and lethality in mice [237]; prolonged status epilepticus induced by continuous hippocampal stimulation [348]; and activity evoked by intracortical stimulation in brain slices from freeze-lesioned rat neocortex [349]. However, it is noteworthy that ifenprodil had no effect in some animal models. Some of these studies [350,351] demonstrated that ifenprodil was unable to protect against seizures induced by imipenem or pefloxacin in DBA/2 mice [230], suggesting that the polyamine site did not exert a principal role in the genesis of seizures induced by imipenem or pefloxacin. Another study showed that ifenprodil had no effect on DMCM (methyl-6,7-dimethoxy-4-ethyl-beta-carboline-3carboxylate)-induced seizures in mice [352]. Burket et al. [353] demonstrated that ifenprodil was ineffective against electricallyprecipitated tonic hindlimb extension. Although ifenprodil increased the threshold for electroconvulsions when applied at 20 and 40 mg/kg (i.p.), when combined with AEDs (carbamazepine, diphenylhydantoin, phenobarbital and valproate), it did not influence their anticonvulsant actions [342]. Similarly Kleinrok et al. [230] showed that ifenprodil failed to enhance the antielectroshock efficacy of conventional AEDs. In a further study, the combined treatment of ifenprodil with L-701,324, a brain penetrating glycineB antagonist, was examined for anticonvulsive effectiveness in fully amygdala-kindled rats [354]. Both drugs, when given separately up to doses causing severe motor impairment,
627
did not significantly affect seizure parameters comprising focal afterdischarge threshold, seizure severity, duration of seizures and afterdischarges. Coadministration of ifenprodil (10 mg/kg, i.p.) and L-701,324 (5 mg/kg, i.p.) resulted in an increased anticonvulsant effect in afterdischarge threshold, while generalized seizure activity was suppressed [354]. These results indicate that combinations of drugs targeting different sites may offer an advantage compared with a single compound. 5.13. Eliprodil Eliprodil or SL-82.0715 (1-(4-chlorophenyl)-2-[4-[(4-fluorophenyl)methyl]piperidin-1-yl]ethanol) is an NMDAR antagonist that binds to the polyamine modulatory site. It has neuroprotective and anticonvulsant effects in animal studies and does not produce sedation or amnesia [355]. It was evaluated for the treatment of stroke but human trials failed to show clear benefits [356]. Eliprodil has anticonvulsant effects against ethanol withdrawal seizures [357] and amygdala kindling model in rats [358]. It also potentiates anticonvulsant effects of other NMDAR antagonists against electroshock-induced seizures in mice [359] and in the rat amygdala kindling model [358]. Side effects of the drug combination were low-grade ataxia and slight hyperlocomotion. However, no detectable changes in motor coordination (as assessed with the rotarod test) could be observed [358]. These side effect data are encouraging for further clinical development of combined treatment with polyamine site antagonists and glycineB receptor antagonists. 5.14. Other NMDAR antagonists in preclinical studies In addition to the above-mentioned NMDAR antagonists and modulators that were used in clinical settings, a variety of NMDAR antagonists have been evaluated in preclinical studies during the last 30 years. The results of these studies have been summarized in Table 4. These findings suggest that NMDAR antagonism at different sites of the receptor, such as the pore channel, the glycine/D-serine site at the NR1 subunit, and the glutamate/NMDA site at the NR2 subunit could have anticonvulsant activity in several animal models of epilepsy. However, these agents have not been examined in clinical settings yet (Table 4). Side effects occurring with the application of NMDAR antagonists might be an important obstacle to evaluating these agents in the clinical trials. In experimental models of epilepsy, the combinations of conventional AEDs with low doses of NMDAR antagonists have shown a marked benefit with less severe side effects compared with monotherapy [231], suggesting a potential advantage that requires clinical validation. 6. Effects of conventional AEDs on NMDARs Convergent evidence that NMDARs could be a valid target for developing novel targets for epilepsy also comes from reports indicating that a variety of AEDs of different classes (including carbamazepine, ethosuximide, gabapentin, lamotrigine, levetiracetam, oxcarbazepine, pregabalin, topiramate and valproic acid) affect functioning and expression of NMDARs in different brain regions that are involved in several seizure types (Table 5). These results again support the hypothesis that combinations of common AEDs with NMDAR antagonists could be beneficial in controlling epileptic seizures. 7. A role for NMDARs in the bidirectional relationship between epilepsy and depression Psychiatric comorbidities in epilepsy have been considered to reflect a “consequence or complication” [360]. Depression (along with anxiety) is one of the most frequent psychiatric comorbidities in
628
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
Table 4 Anticonvulsant effects of several NMDAR antagonists in a variety of animal behavioral experiments. Substance
Effect
Seizure models
Reference
ACEA-1021 (5-Nitro-6,7-dichloro-1,4dihydro-2,3-quinoxalinedione), ACEA1031, ACEA-1328 (5-Nitro-6,7-dimethyl1,4-dihydro-2, 3-quinoxalinedione) ACPC (1-Aminocyclopropane carboxylic acid)
Competitive strychnineinsensitive glycine site antagonist
Cocaine-induced convulsions and lethality in mice
[237,300,391]
Strychnine-insensitive glycine site partial agonist
Aminocyclobutane carboxylic acid
Strychnine-insensitive glycine site partial agonist Competitive NMDAR antagonist
Audiogenic seizures in genetically epilepsy-prone rats Lithium-pilocarpine status epilepticus in rats MES in rats NMDA-induced seizures in mice and rats NMDA-induced hippocampal seizures in mice NMDA-induced seizures in mice and rats
[392] [393] [299] [233] [394] [233]
Audiogenic seizures in DBA/2 mice
[395]
Aminophylline-induced seizures in mice Febrile seizures in epileptic chicks Aminophylline-induced seizures in mice 4-Aminopyridine-induced seizures in rats Amygdaloid kindled convulsions in rats Audiogenic seizures in DBA/2 mice or genetically epilepsy-prone rats Barbital withdrawal-induced convulsions in rats Cocaine-induced convulsions and lethality in mice Electrically kindled seizures in the rat prepyriform cortex Epileptogenic responses in a synaptic and a non-synaptic model of epileptogenesis in the CA1 region of the hippocampal slice Febrile seizures in epileptic chicks Flurothyl-induced clonic seizures in rats Homocysteine-induced seizures in immature rats Hyperbaric oxygen-induced seizures in rats Insulin-induced convulsions in rats Kindled hippocampal seizures MES in rats Metaphit-induced audiogenic seizures in rats Motor and electrocortical seizures, elicited by administration of the polyamine putrescine into the deep prepiriform cortex Penicillin-induced, synchronized afterdischarges in immature rat CA3 hippocampal neurons Pilocarpine-induced limbic seizures in rats PTZ-induced seizures in mice Seizures in baboons, Papio papio, with photosensitive epilepsy Seizures induced by injection of quinolinic acid into the rat dorsal hippocampus Seizures induced by NMDA, 3MPA, TSC, DMCM, picrotoxin, bicuculline and ibotenate in mice MES in rats and mice
[396] [323] [396] [397] [398] [399–402] [403] [237] [404] [182]
γ-D-glutamylaminomethylphosphonate (Glu-Amp), β-Daspartylaminomethylphosphonate (Asp-Amp) AP-5 (2-Amino-5-phosphonoheptanoic acid) AP-7 (2-Amino-7-phosphonoheptanoic acid)
Competitive NMDAR antagonist Competitive NMDAR antagonist
Biphenyl-analogs of AP-7 (SDZ 220–581) Competitive NMDAR antagonist APV (2-Amino-5-phosphonovalerate) Competitive NMDAR antagonist
CGP 37849 (DL-(E)-2-Amino-4-methyl5-phosphono-3-pentonoic acid)
Competitive strychnineinsensitive glycine site antagonist
CGP 39551 (Carboxyethylester of CGP 37849)
Competitive NMDAR antagonist
CGP 40116 ((R)-Enantiomer of CGP 37849)
Competitive NMDAR antagonist
Amygdaloid kindled convulsions in rats Catechol-induced sensory-evoked electromyographic activity in rats Guanidinosuccinate-induced convulsions in mice Metaphit-induced audiogenic seizures in rats NMDA-induced convulsions and lethality in mice Picrotoxin-induced epileptiform activity in the hippocampal CA1 and CA3 regions Seizures induced by picrotoxin microperfusion in the hippocampus of freely moving rats Spinal seizure model in mice Amygdaloid kindled convulsions in rats Audiogenic seizures in DBA/2 mice and genetically epilepsy-prone rats Electroshock or MES-induced seizures in mice and rats Guanidinosuccinate-induced convulsions in mice NMDA-induced seizures in mice and rats Photically induced myoclonus in Papio papio Amygdaloid kindled convulsions in rats Audiogenic or imipenem-induced seizures in DBA/2 mice or genetically epilepsy-prone rats Electroshock or MES-induced seizures in mice and rats Ethanol withdrawal seizures in mice Photically induced myoclonus in Papio papio PTZ-induced seizures in developing, 7 to 90-day-old rats Bicuculline- or pilocarpine-evoked seizures in mice exposed to transient episode of brain oligemia or ischemia Homocysteine-induced seizures in immature rats Lithium-pilocarpine-induced status epilepticus MES in mice NMDA-induced generalized tonic–clonic seizures in mice
[323] [167,405,406] [407] [315] [408] [409] [410,411] [412–414] [326] [175] [415] [399] [416] [417] [310,418–421] [422] [423] [316] [140] [413] [148,424] [176,181] [425] [341] [426,427] [392,402,428] [359,429–431] [140] [233] [428] [426,427] [351,402,428] [429–431] [432–434] [428] [435] [436,437] [407] [438] [301] [439]
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
629
Table 4 (continued) Substance
Effect
CGP 43487 (Carboxylester of CGP 40116) Competitive NMDAR antagonist CGS 19755 or Selfotel (Cis-4phosphonomethyl-2-piperidine carboxylic acid)
7-CKA (7-chlorokynurenic acid) and Kynurenic acid
Co 101022 CP101,606 (Traxoprodil) CPP (3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid)
Competitive NMDAR antagonist
Strychnine-insensitive glycine site antagonist
Competitive NMDAR antagonist NR2B subunit NMDAR antagonist Competitive NMDAR antagonist
CPPene ((E)-4-(3-phophonoprop-2enyl)-piperazine-2-carboxylic acid)
Competitive NMDAR antagonist
DCQX (6,7-dichloroquinoxaline-2,3dione)
Competitive strychnineinsensitive glycine site antagonist NMDAR antagonist at glycine site
5,7-DCK (5,7-Dichlorokynurenic acid)
Gacyclidine (GK-11) (+,R)-HA-966 ((+,R)-3-amino-1hydroxy-2-pyrrolidine)
Ibogaine [(−)-12-Methoxyibogamine]
L-701,324 (7-Chloro-4-hydroxy-3-(3phenoxy)phenylquinolin-2-[1H]-one)
Non-competitive NMDAR antagonist Strychnine-insensitive glycine site partial agonist
Low-affinity noncompetitive NMDAR antagonist Strychnine-insensitive glycine site antagonist
Seizure models
Reference
Pilocarpine-induced motor seizures in mice PTZ-induced clonic and generalized tonic–clonic seizures Seizure threshold for tonic hindlimb extension Spontaneously recurrent absence seizures in GAERS Wistar rats γ-Hydroxybutyrate model of generalized absence seizures in rats Lithium-pilocarpine-induced status epilepticus in rats Seizure threshold for tonic hindlimb extension in mice Amygdaloid kindled convulsions in rats Audiogenic seizures in DBA/2 mice Focal hippocampal seizures in freely moving rats elicited by low-frequency stimulation Lidocaine-induced seizures in rats MES in rats and mice NMDA-induced seizures in mice Picrotoxin-induced seizures in rats and mice Amygdaloid kindled convulsions in rats Audiogenic seizures in DBA/2 mice Cocaine-induced convulsions and lethality in mice Electroshock-induced seizures or MES in rats Focal hippocampal seizures in freely moving rats elicited by low-frequency stimulation PTZ-induced diazepam-withdrawal seizures in mice Seizures evoked by focal application of bicuculline into the area tempestas of rat deep prepiriform cortex Seizures induced by bicuculline, DMCM and NMDA in mice Cocaine-induced convulsions and lethality in mice
[440] [441] [442] [443] [444,445] [438] [442] [446] [447,448] [449]
Cocaine-induced convulsions and lethality in mice
[237]
4-Aminopyridine-induced seizures in mice and rats Amygdaloid kindled convulsions in rats Audiogenic or imipenem-induced seizures in DBA/2 mice or genetically epilepsy-prone rats Cocaine-induced convulsions and lethality in mice Dendrotoxin-induced clonic seizures in mice Electrically kindled seizures at the rat prepyriform cortex Febrile seizures in epileptic chicks γ-Hydroxybutyrate model of generalized absence seizures in rats MES in mice NMDA-induced seizures in mice PTZ-induced clonic or tonic extensor seizures in mice Status epilepticus induced by continuous electrical stimulation of the hippocampus in rats Audiogenic seizures in genetically epilepsy-prone rats Imipenem-induced seizures in DBA/2 mice MES in mice Spinal seizure model in mice Cocaine-induced convulsions and lethality in mice
[397,461] [446] [351,392,402,455]
[450] [451] [448,452] [451] [297,453,454] [455,456] [237] [457,458] [449] [459] [460] [310,337,352,452] [237]
[237,462] [126] [404] [323] [444,445] [260,299] [262,337,424,452,456] [260,463] [158,348] [151,392] [351] [230] [341] [237,300]
NMDA-induced seizures in mice and rats Self-sustaining status epilepticus induced in rats by brief intermittent electrical stimulation of the perforant path Spontaneously recurrent absence seizures in GAERS Wistar rats Tonic hindlimb extension in MES Soman-induced seizures and EEG activity in primates
[233] [464]
Amygdaloid kindled convulsions in rats Cocaine-induced convulsions and lethality in mice γ-Hydroxybutyrate model of generalized absence seizures in rats Imipenem/cilastatin-induced seizures in rats MES in rats NMDA-induced seizures in mice and rats Pilocarpine-induced motor seizures in mice Seizures evoked by focal application of bicuculline into the area tempestas of rat deep prepiriform cortex Spinal seizure model in mice Tonic extensor seizures from low-intensity electroshock in rats NMDA-induced seizures in mice
[296,297,467] [237,300] [444,445] [468] [299] [233,452] [440] [460]
Amygdaloid kindled convulsions in rats Audiogenic seizures in DBA-2 mice Ethanol withdrawal seizures in rats MES in mice Seizures induced by NMDA, PTZ and electroshock in mice and rats
[354,358,471,472] [473] [357] [301] [473]
[443] [299] [465,466]
[341] [469] [125,470]
(continued on next page)
630
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
Table 4 (continued) Substance
Effect
Seizure models
Reference
MNQX (5,7-dinitroquinoxaline-2,3dione) MRZ 21579 or Neramexane (1-amino1,3,3,5,5-pentamethyl-cyclohexane hydrochloride) Tiletamine
Strychnine-insensitive glycine site antagonist Low-to-moderate affinity non-competitive NMDAR antagonist Non-competitive NMDAR antagonist
Audiogenic seizures in DBA-2 mice NMDA-induced seizures in mice and rats Ethanol withdrawal seizures in rats
[474] [233] [238,475]
Audiogenic seizures in DBA/2 mice NMDA and quinolinate-induced seizure in mice
[155] [476]
Abbreviations: DMCM, methyl-6,7-dimethoxy-4-ethyl-beta-carboline-3-carboxylate; 3MPA, 3-mercaptopropionic acid; TSC, thiosemicarbazide; MES, maximal electroshock; PTZ, pentylenetetrazole.
epilepsy [361]. The prevalence of mood disorders in patients with epilepsy is 20–50%; the higher prevalence rates have been typically identified in patients with poorly controlled seizures [362]. Several studies have shown that lifetime risks of major depression in patients with chronic and intractable epilepsy are considerably higher than the general population [360–362]. A lifetime history of depression may predict persistent auras in the absence of disabling seizures and a failure to achieve freedom from disabling seizures [361,363]. Recently, a bidirectional relationship between depressive disorders and epilepsy has been suggested, as not only are people with epilepsy at greater risk of developing depressive disorders, but patients with depression have a three- to seven-fold higher risk of developing epilepsy [364–366]. These results may suggest the existence of common pathogenic mechanisms underlying both conditions. In recent decades, glutamate and glutamate-mediated activation of NMDARs have become potential therapeutic targets in many neuropsychiatric diseases including mood disorders. Using a variety of methods such as NMDAR subunit gene expression, immunoblotting, binding affinities and autoradiography, several laboratories have demonstrated a significant alteration in functioning and expression of NMDARs in both patients and suicidal victims with major depressive disorder [367]. Various NMDAR antagonists exert antidepressant-like effects in several animal models of depression [368,369]. Moreover, clinical studies revealed that NMDAR antagonists including ketamine [370,371], memantine [372], amantadine [373,374] and traxoprodil [375] are able to significantly improve depressive symptoms in patients with either major depression or bipolar disorder. Therefore, further studies should explore whether using NMDAR antagonists may be beneficial in improving both seizures and depression in comorbid patients whose symptoms are resistant to conventional AEDs. 8. Adverse effects of NMDAR antagonists in clinical settings Because glutamate is a major excitatory transmitter in the CNS, generalized inhibition of a glutamate receptor subtype such as the NMDAR causes adverse effects that could limit the potential for clinical applications. Therefore, while using NMDAR antagonists could be a potential therapeutic approach for controlling seizures, NMDAR antagonism is associated with an array of adverse effects. Both competitive NMDA and glycine antagonists, even although effective in preventing glutamate-mediated neurotoxicity, cause generalized inhibition of NMDAR function. While open-channel block with the noncompetitive antagonists is the most appealing strategy for therapeutic intervention during excessive NMDAR activation, as this action of blockade requires prior activation of the receptor [376], progressive increase in the severity of NMDAR hypofunction can lead to an increasing range of effects on brain function. Underexcitation of the NMDAR, induced by even relatively low doses of NMDAR antagonists, can cause memory dysfunction and learning deficits without clinically evident psychosis. More severe NMDAR hypofunction can produce psychosis [377] characterized by hallucinations,
paranoid delusions, confusion, difficulty in concentration, alterations in mood, agitation, nightmares, catatonia, ataxia, and loss of sensation [378,379]. Therefore, there should be considerable caution regarding the use of NMDAR antagonists, especially at high doses in clinical settings and particularly in children. This issue stems from the fact that NMDAR function plays a critical role in brain development, longterm potentiation, and neuronal migration and synaptic pruning [380]. Accordingly, NMDAR blockers could potentially result in neurodegeneration in the cingulate and retrosplenial cortex [381]. Several lines of evidence have demonstrated that strychnineinsensitive glycine site NMDAR antagonists (glycineB antagonists) lack neuronal vacuolization and a psychotomimetic potential, indicating a more acceptable adverse effect profile over NMDAR antagonists at other sites (for a review see [382]). The route of administration may also influence the appearance of side effects. For example, NMDAR antagonists could result in spinal cord pathology when given intrathecally. Brock-Utne et al. [383] showed that intrathecal administration of ketamine to monkeys caused focal degeneration with loss of myelin and axoplasm within a solitary nerve root. Another study by Errando et al. [384] demonstrated that subarachnoid ketamine without preservative did not show significant neurotoxic effects, whereas ketamine with preservative produced minimal changes. Vranken et al. reported that intrathecal administration of preservative-free ketamine in a clinically relevant concentration and dosage had a toxic effect on the CNS of rabbits [385]. Other investigators indicated that chronic intrathecal administration of pharmacologically active doses of NMDAR antagonists including CPP (3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid), kynurenic acid and ketamine in rats or rabbits did not produce significant neurotoxic effects in the spinal cord [386–388]. However, a blood– brain barrier study showed evidence of neurotoxicity for ketamine after intrathecal administration in rabbits [389]. Moreover, Karpinski et al. [390] reported post-mortem CNS histopathological changes of subpial spinal cord vacuolation in a terminally ill cancer patient who received continuous infusion of intrathecal ketamine at a rate of 5 mg/day for a duration of 3 weeks. 9. Conclusion During the last three decades, a large number of studies have indicated that the NMDAR complex is important in seizure phenomena, although more studies are clearly needed to demonstrate that pathology of NMDARs underlies epilepsy. Considering animal seizure models, NMDAR antagonists may be effective anticonvulsants. However, there is no compelling evidence to date that they are useful in the chronic treatment of human epilepsy. Some antagonists such as ketamine and magnesium sulfate should be further studied as possible treatments for controlling status epilepticus and eclampsia-related seizures, respectively, whereas a vast majority of agents have not been evaluated in clinical trials yet. Moreover, some NMDAR antagonists (e.g. D-cycloserine) have proconvulsant activity in animal models, in vitro models or even in patients with epilepsy. Several conventional AEDs have been shown to affect
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
631
Table 5 Effect of common AEDs on NMDARs. Drug
Species
Carbamazepine Rat fetus Fetal mouse Rat pup Genetically epilepsyprone mouse Guinea pig Rabbit Rat
Ethosuximide
Mouse Rat
Gabapentin
Rat
Lamotrigine
– Rats with inflammation induced by injection of CFA to hindpaw Rat
[477] [478] [479,480] [481]
NMDA-mediated incease in the frequency of occurrence of low Mg2+ induced EFP NMDA-mediated induction of long-term potentiation NMDA-evoked excitatory postsynaptic potential NMDA-evoked responses NMDA-, glutamate- or kainate-induced increase in [Ca2+] NMDA component of fPSP response NMDA-induced cytoplasmic vacuolization NMDA-induced regional arachidonic acid incorporation coefficients k* Release of preloaded D-[3H]aspartate NMDAR hypofunction neurotoxicity
Hippocampal CA3
↓ ↓ ↓ ↓ ↑ ↔
Perforant path-dentate gyrus pathway Hippocampal and amygdaloid slices Cortical wedge Cultured hippocampal neurons Hippocampal slices Primary hippocampal neurons Brain
↓ ↓ ↓ ↓ ↓ ↓ ↓
[483] [484,485] [486] [487] [488] [489] [490]
Cerebral cortical slices ↓ Layer IV–Va pyramidal neurons of retrosplenial ↓ cortex Epileptiform field potential discharges evoked by removing Hippocampal slices ↓ 2+ Mg from perfusion fluid NMDA-evoked depolarizations Cultured cortical neurons ↔ NMDA-induced presynaptic glutamate release Entorhinal cortex ↔ NMDA-evoked excitatory postsynaptic currents Superficial horn neurons of spinal cord slices ↓ Presynaptically NMDA-induced axon excitability Presynaptic fiber volley in the hippocampal ↓ CA1 slices NMDA-induced excitotoxicity Hippocampal CA1 neurons ↓ NMDAR currents Xenopus laevis oocytes ↓ NMDA-evoked currents Single dorsal horn neurons ↑
Kainate-induced neurotoxicity Quinolinic acid- or ibotenic acid-induced neurotoxicity Veratrine-evoked release of glutamate and aspartate NMDA-evoked responses Evoked NMDA-mediated excitatory postsynaptic currents Release of preloaded D-[3H]aspartate NMDA-induced epileptiform activity NMDA-evoked EPSC Specific binding of [3H]-MK-801 and [3H]-glycine
Striatum
↓ ↔ ↓
[482]
[491] [492] [493] [494] [495] [496] [497]
[498] [499]
[500]
↔ ↓ ↓ ↓ ↓ ↔ ↓
[508]
Caudate–putamen, hippocampal CA1 and CA3 Striatal synapses Primary hippocampal neurons 3 week old primary hippocampal cultures Entorhinal cortex Hippocampal CA1 region
↓ ↔ ↓ ↔ ↔ ↔
[509] [510] [511] [512] [495] [513]
Hippocampus Forebrain membranes Cortical wedge Layer IV–Va pyramidal neurons of retrosplenial cortex Epileptiform field potential discharges evoked by removing Hippocampal slices Mg2+ from perfusion fluid Rat NMDAR-mediated trafficking and release of synaptic Cultured hippocampal neurons vesicles Rat L-Glutamate- or NMDA-induced paroxysms Planarians (Dugesia dorotocephala) Rat NMDA-induced cytoplasmic vacuolization Primary hippocampal neurons NMDA-induced regional arachidonic acid incorporation Brain coefficients k* 2-week-old rats prenatally NR2A and NR2B mRNA expression Neocortex exposed to the drug Somatosensory cortex
↔ ↔ ↓ ↓
[514] [515] [486] [492]
↓
[493]
↓
[516,517]
↓ ↓ ↓
[518,519] [489] [520]
↑ ↔
[521] [522]
Olanzapine
Rat
Topiramate Valproic acid
Result Reference
Cultured hippocampal pyramidal neurons Cultured spinal neurons Primary cultures of cerebellar granule cells Cortical wedges
[501] [502] [486] [503] [491,504] [505] [506] [507]
Mouse Rat
Pregabalin
Region/cells
Glutamate-induced neurotoxicity NMDA-activated membrane currents Kainate- and NMDA-induced intracellular Ca2+ levels NMDA-induced depolarizations
Cortical slices Forebrain slices Cortical wedge Granule cells in the dentate gyrus Cerebral cortical slices CA3 subfield of hippocampal slices Hippocampal dentate gyrus Synaptic plasma membranes prepared from medial prefrontal cortex Pyramidal cell of the medial prefrontal cortex
Levetiracetam
Oxcarbazepine Phenytoin
Measurement
Rat 19 day embryonic rat Rat
Phencyclidine-evoked increase in excitatory postsynaptic currents NMDAR binding Protein levels of NR1, NR2A and NR2B subunits kainic acid-induced apoptosis NMDA-induced neurotoxicity NMDA-induced presynaptic glutamate release NMDA receptor-mediated epileptiform population response NMDAR-dependent LTP [3H]glycine binding NMDA-evoked responses NMDAR hypofunction neurotoxicity
Abbreviations: CFA, complete Freund adjuvant; EFP, extracellular field potentials; EPSC, excitatory postsynaptic currents; fPSP, field excitatory postsynaptic potentials; I/R, ischemia/reperfusion; LTP, long-term potentiation; PKC, protein kinase C.
NMDAR function and some studies indicate that combinations of AEDs with NMDAR antagonists may be a promising strategy to increase clinical benefit. However, establishing the safety of NMDARs alone or in combination with AEDs requires further investigation.
References [1] Schachter SC. Seizure disorders. Med Clin North Am 2009;93:343–51 (viii). [2] Schachter SC. Currently available antiepileptic drugs. Neurotherapeutics 2007;4: 4–11. [3] Dichter MA, Brodie MJ. New antiepileptic drugs. N Engl J Med 1996;334:1583–90.
632
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
[4] Loscher W, Schmidt D. Which animal models should be used in the search for new antiepileptic drugs? A proposal based on experimental and clinical considerations. Epilepsy Res 1988;2:145–81. [5] Urbanska EM, Czuczwar SJ, Kleinrok Z, Turski WA. Excitatory amino acids in epilepsy. Restor Neurol Neurosci 1998;13:25–39. [6] Carter DS, Deshpande LS, Rafiq A, Sombati S, Delorenzo RJ. Characterization of spontaneous recurrent epileptiform discharges in hippocampal-entorhinal cortical slices prepared from chronic epileptic animals. Seizure 2010;20:218–24. [7] Meldrum BS. Excitotoxicity and selective neuronal loss in epilepsy. Brain Pathol 1993;3:405–12. [8] Bayer TA, Wiestler OD, Wolf HK. Hippocampal loss of N-methyl-D-aspartate receptor subunit 1 mRNA in chronic temporal lobe epilepsy. Acta Neuropathol 1995;89:446–50. [9] Mathern GW, Pretorius JK, Kornblum HI, et al. Human hippocampal AMPA and NMDA mRNA levels in temporal lobe epilepsy patients. Brain 1997;120(Pt 11): 1937–59. [10] Mathern GW, Leite JP, Babb TL, et al. Aberrant hippocampal mossy fiber sprouting correlates with greater NMDAR2 receptor staining. Neuroreport 1996;7: 1029–35. [11] Franck JE, Pokorny J, Kunkel DD, Schwartzkroin PA. Physiologic and morphologic characteristics of granule cell circuitry in human epileptic hippocampus. Epilepsia 1995;36:543–58. [12] Isokawa M, Levesque MF. Increased NMDA responses and dendritic degeneration in human epileptic hippocampal neurons in slices. Neurosci Lett 1991;132: 212–6. [13] Masukawa LM, Higashima M, Hart GJ, Spencer DD, O'Connor MJ. NMDA receptor activation during epileptiform responses in the dentate gyrus of epileptic patients. Brain Res 1991;562:176–80. [14] Mathern GW, Pretorius JK, Mendoza D, et al. Hippocampal N-methyl-D-aspartate receptor subunit mRNA levels in temporal lobe epilepsy patients. Ann Neurol 1999;46:343–58. [15] Neder L, Valente V, Carlotti Jr CG, et al. Glutamate NMDA receptor subunit R1 and GAD mRNA expression in human temporal lobe epilepsy. Cell Mol Neurobiol 2002;22:689–98. [16] Crino PB, Duhaime AC, Baltuch G, White R. Differential expression of glutamate and GABA-A receptor subunit mRNA in cortical dysplasia. Neurology 2001;56: 906–13. [17] Andre VM, Flores-Hernandez J, Cepeda C, et al. NMDA receptor alterations in neurons from pediatric cortical dysplasia tissue. Cereb Cortex 2004;14:634–46. [18] Liu FY, Wang XF, et al. Upregulated expression of postsynaptic density-93 and Nmethyl-D-aspartate receptors subunits 2B mRNA in temporal lobe tissue of epilepsy. Biochem Biophys Res Commun 2007;358:825–30. [19] White R, Hua Y, Scheithauer B, Lynch DR, Henske EP, Crino PB. Selective alterations in glutamate and GABA receptor subunit mRNA expression in dysplastic neurons and giant cells of cortical tubers. Ann Neurol 2001;49:67–78. [20] Dalmau J, Lancaster E, Martinez-Hernandez E, Rosenfeld MR, Balice-Gordon R. Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol 2011;10:63–74. [21] Florance-Ryan N, Dalmau J. Update on anti-N-methyl-D-aspartate receptor encephalitis in children and adolescents. Curr Opin Pediatr 2010;22:739–44. [22] Johnson N, Henry C, Fessler AJ, Dalmau J. Anti-NMDA receptor encephalitis causing prolonged nonconvulsive status epilepticus. Neurology 2010;75:1480–2. [23] Wandinger KP, Saschenbrecker S, Stoecker W, Dalmau J. Anti-NMDA-receptor encephalitis: a severe, multistage, treatable disorder presenting with psychosis. J Neuroimmunol 2011;231:86–91. [24] Dalmau J, Gleichman AJ, Hughes EG, et al. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 2008;7:1091–8. [25] Krall RL, Penry JK, White BG, Kupferberg HJ, Swinyard EA. Antiepileptic drug development: II. Anticonvulsant drug screening. Epilepsia 1978;19:409–28. [26] Loscher W. Critical review of current animal models of seizures and epilepsy used in the discovery and development of new antiepileptic drugs. Seizure 2011;20: 359–68. [27] Castel-Branco MM, Alves GL, Figueiredo IV, Falcao AC, Caramona MM. The maximal electroshock seizure (MES) model in the preclinical assessment of potential new antiepileptic drugs. Methods Find Exp Clin Pharmacol 2009;31: 101–6. [28] Fisher RS. Animal models of the epilepsies. Brain Res Brain Res Rev 1989;14: 245–78. [29] Loscher W. Animal models of epilepsy for the development of antiepileptogenic and disease-modifying drugs. A comparison of the pharmacology of kindling and post-status epilepticus models of temporal lobe epilepsy. Epilepsy Res 2002;50: 105–23. [30] Pratt GD, Kokaia M, Bengzon J, et al. Differential regulation of N-methyl-Daspartate receptor subunit messenger RNAs in kindling-induced epileptogenesis. Neuroscience 1993;57:307–18. [31] Kraus JE, Yeh GC, Bonhaus DW, Nadler JV, McNamara JO. Kindling induces the long-lasting expression of a novel population of NMDA receptors in hippocampal region CA3. J Neurosci 1994;14:4196–205. [32] Al-Ghoul WM, Meeker RB, Greenwood RS. Amygdala kindling alters N-methyl-Daspartate receptor subunit messenger RNA expression in the rat supraoptic nucleus. Neuroscience 1997;77:985–92. [33] Jensen PJ, Millan N, Mack KJ. Cortical NMDAR-1 gene expression is rapidly upregulated after seizure. Brain Res Mol Brain Res 1997;44:157–62. [34] Vezzani A, Speciale C, Della Vedova F, Tamburin M, Benatti L. Alternative splicing at the C-terminal but not at the N-terminal domain of the NMDA receptor NR1 is altered in the kindled hippocampus. Eur J Neurosci 1995;7:2513–7.
[35] Kraus JE, Nadler JV, McNamara JO. Regulation of alternative splicing of NMDAR1 in the kindling model. Brain Res Mol Brain Res 1996;41:97–104. [36] Kikuchi S, Iwasa H, Sato T. Lasting changes in NMDAR1 mRNA level in various regions of cerebral cortex in epileptogenesis of amygdaloid-kindled rat. Psychiatry Clin Neurosci 2000;54:573–7. [37] Gerfin-Moser A, Grogg F, Rietschin L, Thompson SM, Streit P. Alterations in glutamate but not GABAA receptor subunit expression as a consequence of epileptiform activity in vitro. Neuroscience 1995;67:849–65. [38] Mathern GW, Pretorius JK, Leite JP, et al. Hippocampal AMPA and NMDA mRNA levels and subunit immunoreactivity in human temporal lobe epilepsy patients and a rodent model of chronic mesial limbic epilepsy. Epilepsy Res 1998;32: 154–71. [39] Mathern GW, Pretorius JK, Mendoza D, Lozada A, Kornblum HI. Hippocampal AMPA and NMDA mRNA levels correlate with aberrant fascia dentata mossy fiber sprouting in the pilocarpine model of spontaneous limbic epilepsy. J Neurosci Res 1998;54:734–53. [40] Lason W, Turchan J, Przewlocki R, Machelska H, Labuz D, Przewlocka B. Effects of pilocarpine and kainate-induced seizures on N-methyl-D-aspartate receptor gene expression in the rat hippocampus. Neuroscience 1997;78:997–1004. [41] Chapman AG, Woodburn VL, Woodruff GN, Meldrum BS. Anticonvulsant effect of reduced NMDA receptor expression in audiogenic DBA/2 mice. Epilepsy Res 1996;26:25–35. [42] Marianowski R, Pollard H, Moreau J, et al. N-Methyl-D-aspartate receptor subunits NR1 and NR2C are overexpressed in the inferior colliculus of audiogenic mice. Neurosci Lett 1995;189:190–4. [43] Pumain R, Louvel J, Gastard M, Kurcewicz I, Vergnes M. Responses to N-methylD-aspartate are enhanced in rats with petit mal-like seizures. J Neural Transm Suppl 1992;35:97–108. [44] Morrow AL, Devaud LL, Bucci D, Smith FD. GABAA and NMDA receptor subunit mRNA expression in ethanol dependent rats. Alcohol Alcohol Suppl 1994;2: 89–95. [45] Graves NM. Felbamate. Ann Pharmacother 1993;27:1073–81. [46] Taglialatela M, Ongini E, Brown AM, Di Renzo G, Annunziato L. Felbamate inhibits cloned voltage-dependent Na+ channels from human and rat brain. Eur J Pharmacol 1996;316:373–7. [47] Stefani A, Calabresi P, Pisani A, Mercuri NB, Siniscalchi A, Bernardi G. Felbamate inhibits dihydropyridine-sensitive calcium channels in central neurons. J Pharmacol Exp Ther 1996;277:121–7. [48] Rho JM, Donevan SD, Rogawski MA. Mechanism of action of the anticonvulsant felbamate: opposing effects on N-methyl-D-aspartate and gamma-aminobutyric acid A receptors. Ann Neurol 1994;35:229–34. [49] Ticku MK, Kamatchi GL, Sofia RD. Effect of anticonvulsant felbamate on GABAA receptor system. Epilepsia 1991;32:389–91. [50] Kume A, Greenfield Jr LJ, Macdonald RL, Albin RL. Felbamate inhibits [3H]tbutylbicycloorthobenzoate (TBOB) binding and enhances Cl− current at the gamma-aminobutyric Acid A (GABAA) receptor. J Pharmacol Exp Ther 1996;277: 1784–92. [51] McCabe RT, Wasterlain CG, Kucharczyk N, Sofia RD, Vogel JR. Evidence for anticonvulsant and neuroprotectant action of felbamate mediated by strychnineinsensitive glycine receptors. J Pharmacol Exp Ther 1993;264:1248–52. [52] White HS, Harmsworth WL, Sofia RD, Wolf HH. Felbamate modulates the strychnine-insensitive glycine receptor. Epilepsy Res 1995;20:41–8. [53] Subramaniam S, Rho JM, Penix L, Donevan SD, Fielding RP, Rogawski MA. Felbamate block of the N-methyl-D-aspartate receptor. J Pharmacol Exp Ther 1995;273:878–86. [54] Kleckner NW, Glazewski JC, Chen CC, Moscrip TD. Subtype-selective antagonism of N-methyl-D-aspartate receptors by felbamate: insights into the mechanism of action. J Pharmacol Exp Ther 1999;289:886–94. [55] Harty TP, Rogawski MA. Felbamate block of recombinant N-methyl-D-aspartate receptors: selectivity for the NR2B subunit. Epilepsy Res 2000;39:47–55. [56] Sachdeo R, Kramer LD, Rosenberg A, Sachdeo S. Felbamate monotherapy: controlled trial in patients with partial onset seizures. Ann Neurol 1992;32: 386–92. [57] Theodore WH, Raubertas RF, Porter RJ, et al. Felbamate: a clinical trial for complex partial seizures. Epilepsia 1991;32:392–7. [58] Leppik IE, Dreifuss FE, Pledger GW, et al. Felbamate for partial seizures: results of a controlled clinical trial. Neurology 1991;41:1785–9. [59] Faught E, Sachdeo RC, Remler MP, et al. Felbamate monotherapy for partial-onset seizures: an active-control trial. Neurology 1993;43:688–92. [60] Kalviainen R, Keranen T, Riekkinen Sr PJ. Place of newer antiepileptic drugs in the treatment of epilepsy. Drugs 1993;46:1009–24. [61] Dodson WE. Felbamate in the treatment of Lennox–Gastaut syndrome: results of a 12-month open-label study following a randomized clinical trial. Epilepsia 1993;34(Suppl 7):S18–24. [62] Hosain S, Nagarajan L, Carson D, Solomon G, Mast J, Labar D. Felbamate for refractory infantile spasms. J Child Neurol 1997;12:466–8. [63] French J, Smith M, Faught E, Brown L. Practice advisory: The use of felbamate in the treatment of patients with intractable epilepsy: report of the Quality Standards Subcommittee of the American Academy of Neurology and the American Epilepsy Society. Neurology 1999;52:1540–5. [64] Zupanc ML, Roell Werner R, Schwabe MS, et al. Efficacy of felbamate in the treatment of intractable pediatric epilepsy. Pediatr Neurol 2010;42:396–403. [65] Hurst DL, Rolan TD. The use of felbamate to treat infantile spasms. J Child Neurol 1995;10:134–6. [66] Randall Jr RE, Rossmeisl EC, Bleifer KH. Magnesium depletion in man. Ann Intern Med 1959;50:257–87.
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640 [67] Griffiths Jr WJ. Audiogenic fits produced by magnesium deficiency in tame domestic Norway rats and in wild Norway and Alexandrine rats. Am J Physiol 1947;149:135–41. [68] Hanna S, Harrison M, Macintyre I, Fraser R. The syndrome of magnesium deficiency in man. Lancet 1960;2:172–6. [69] Kruse HD, Orent ER, McCollum EV. Studies on magnesium deficiency in animals. I. Symptomatology resulting from magnesium deprivation. J Biol Chem 1932;96: 519–39. [70] Greenberg DM, Tufts EV. Effect of a diet low in magnesium on the rat. Proc Soc Exp Biol Med 1934;5:674–5. [71] Buck DR, Mahoney AW, Hendricks DG. Effect of magnesium deficiency on nonspecific excitability level (NEL) and audiogenic seizure susceptibility. Pharmacol Biochem Behav 1976;5:529–34. [72] Suter C, Klingman WO. Neurologic manifestations of magnesium depletion states. Neurology 1955;5:691–9. [73] Canelas HM, De Assis LM, De Jorge FB. Disorders of magnesium metabolism in epilepsy. J Neurol Neurosurg Psychiatry 1965;28:378–81. [74] Babel CS, Bedi HK, Devpura JC, Pandey SK. Serum and cerebrospinal fluid magnesium and calcium contents in idiopathic grand mal epilepsy. J Assoc Physicians India 1973;21:481–7. [75] Govil MK, Mangal BD, Alam SM, Mahendru RK, Srivastava DK, Mudgal JC. Serum and cerebrospinal fluid calcium and magnesium levels in cases of idiopathic grand mal epilepsy and induced convulsions. J Assoc Physicians India 1981;29: 695–9. [76] Sinert R, Zehtabchi S, Desai S, Peacock P, Altura BT, Altura BM. Serum ionized magnesium and calcium levels in adult patients with seizures. Scand J Clin Lab Invest 2007;67:317–26. [77] Hallak M. Effect of parenteral magnesium sulfate administration on excitatory amino acid receptors in the rat brain. Magnes Res 1998;11:117–31. [78] Hallak M, Berman RF, Irtenkauf SM, Evans MI, Cotton DB. Peripheral magnesium sulfate enters the brain and increases the threshold for hippocampal seizures in rats. Am J Obstet Gynecol 1992;167:1605–10. [79] Hallak M, Berman RF, Irtenkauf SM, Janusz CA, Cotton DB. Magnesium sulfate treatment decreases N-methyl-D-aspartate receptor binding in the rat brain: an autoradiographic study. J Soc Gynecol Investig 1994;1:25–30. [80] Stephenson FA. Structure and trafficking of NMDA and GABAA receptors. Biochem Soc Trans 2006;34:877–81. [81] Borges LF, Gucer G. Effect of magnesium on epileptic foci. Epilepsia 1978;19: 81–91. [82] Mikati MA, Injibar H, Kurdi RM, et al. Effects of magnesium sulfate in kainic acidinduced status epilepticus. J Med Liban 2006;54:200–4. [83] Ghasemi A, Saberi M, Ghasemi M, et al. Administration of lithium and magnesium chloride inhibited tolerance to the anticonvulsant effect of morphine on pentylenetetrazole-induced seizures in mice. Epilepsy Behav 2010;19:568–74. [84] Bac P, Herrenknecht C, Binet P, Durlach J. Audiogenic seizures in magnesiumdeficient mice: effects of magnesium pyrrolidone-2-carboxylate, magnesium acetyltaurinate, magnesium chloride and vitamin B-6. Magnes Res 1993;6:11–9. [85] Safar MM, Abdallah DM, Arafa NM, Abdel-Aziz MT. Magnesium supplementation enhances the anticonvulsant potential of valproate in pentylenetetrazol-treated rats. Brain Res 2010;1334:58–64. [86] Cotton DB, Hallak M, Janusz C, Irtenkauf SM, Berman RF. Central anticonvulsant effects of magnesium sulfate on N-methyl-D-aspartate-induced seizures. Am J Obstet Gynecol 1993;168:974–8. [87] Turner TL, Cockburn F, Forfar JO. Magnesium therapy in neonatal tetany. Lancet 1977;1:283–4. [88] Euser AG, Cipolla MJ. Magnesium sulfate for the treatment of eclampsia: a brief review. Stroke 2009;40:1169–75. [89] Sadeh M, Blatt I, Martonovits G, Karni A, Goldhammer Y. Treatment of porphyric convulsions with magnesium sulfate. Epilepsia 1991;32:712–5. [90] Taylor RL. Magnesium sulfate for AIP seizures. Neurology 1981;31:1371–2. [91] Kaplan PW, Lesser RP, Fisher RS, Repke JT, Hanley DF. A continuing controversy: magnesium sulfate in the treatment of eclamptic seizures. Arch Neurol 1990;47: 1031–2. [92] Tso EL, Barish RA. Magnesium: clinical considerations. J Emerg Med 1992;10: 735–45. [93] Zou LP, Wang X, Dong CH, Chen CH, Zhao W, Zhao RY. Three-week combination treatment with ACTH + magnesium sulfate versus ACTH monotherapy for infantile spasms: a 24-week, randomized, open-label, follow-up study in China. Clin Ther 2010;32:692–700. [94] Sakakihara Y. Treatment of West syndrome. Brain Dev 2011;33:202–6. [95] Visser NA, Braun KP, Leijten FS, van Nieuwenhuizen O, Wokke JH, van den Bergh WM. Magnesium treatment for patients with refractory status epilepticus due to POLG1-mutations. J Neurol 2011;258:218–22. [96] Pritchard JA. The use of the magnesium ion in the management of eclamptogenic toxemias. Surg Gynecol Obstet 1955;100:131–40. [97] Sibai BM, Graham JM, McCubbin JH. A comparison of intravenous and intramuscular magnesium sulfate regimens in preeclampsia. Am J Obstet Gynecol 1984;150:728–33. [98] Pritchard JA, Cunningham FG, Pritchard SA. The Parkland Memorial Hospital protocol for treatment of eclampsia: evaluation of 245 cases. Am J Obstet Gynecol 1984;148:951–63. [99] McCubbin JM, Sibai BM, Ardella TN, Anderson GD. Cardiopulmonary arrest due to acute maternal hypermagnesaemia. Lancet 1981;1:1058. [100] Palmer GC, Stagnitto ML, Ordy JM, et al. Preclinical profile of stereoisomers of the anticonvulsant remacemide in mice. Epilepsy Res 1991;8:36–48.
633
[101] Garske GE, Palmer GC, Napier JJ, et al. Preclinical profile of the anticonvulsant remacemide and its enantiomers in the rat. Epilepsy Res 1991;9:161–74. [102] Ray R, Julien R, Gordon J, Blosser J. FPL 12495, a metabolite of remacemide, is a noncompetitive NMDA antagonist. Soc Neurosci Abstr 1992;17:391. [103] Ahmed MS, Mather A, Enna SJ. Binding of [3H]desglycinyl remacemide to rat brain membranes: association with the benzomorphan attachment site of the Nmethyl-D-aspartic acid receptor channel. Brain Res 1999;827:46–50. [104] Subramaniam S, Donevan SD, Rogawski MA. 1.2-Diphenyl-2-propylamine. A major metabolite of the anti-convulsant remacemide, produces a stereoselective block of NMDA receptor currents. Soc Neurosci Abstr 1993;19:717. [105] Hu RQ, Davies JA. The effect of the desglycinyl metabolite of remacemide on cortical wedges prepared from DBA/2 mice. Eur J Pharmacol 1995;287: 251–6. [106] Norris SK, King AE. Electrophysiological effects of the anticonvulsant remacemide hydrochloride and its metabolite ARL 12495AA on rat CA1 hippocampal neurons in vitro. Neuropharmacology 1997;36:951–9. [107] Santangeli S, Sills GJ, Thompson GG, Brodie MJ. Na+ channel effects of remacemide and desglycinyl-remacemide in rat cortical synaptosomes. Eur J Pharmacol 2002;438:63–8. [108] Halonen T, Nissinen J, Pitkanen A. Neuroprotective effect of remacemide hydrochloride in a perforant pathway stimulation model of status epilepticus in the rat. Epilepsy Res 1999;34:251–69. [109] Palmer GC, Murray RJ, Wilson TC, et al. Biological profile of the metabolites and potential metabolites of the anticonvulsant remacemide. Epilepsy Res 1992;12: 9–20. [110] Stagnitto ML, Palmer GC, Ordy JM, et al. Preclinical profile of remacemide: a novel anticonvulsant effective against maximal electroshock seizures in mice. Epilepsy Res 1990;7:11–28. [111] Cramer CL, Stagnitto ML, Knowles MA, Palmer GC. Kainic acid and 4aminopyridine seizure models in mice: evaluation of efficacy of anti-epileptic agents and calcium antagonists. Life Sci 1994;54:PL271–5. [112] Gasior M, Ungard JT, Witkin JM. Preclinical evaluation of newly approved and potential antiepileptic drugs against cocaine-induced seizures. J Pharmacol Exp Ther 1999;290:1148–56. [113] Nehlig A, Boehrer A. Effects of remacemide in two models of genetically determined generalized epilepsy, the GAERS and the audiogenic Wistar AS. Epilepsy Res 2003;52:253–61. [114] van Luijtelaar EL, Coenen AM. Effects of remacemide and its metabolite FPL 12495 on spike-wave discharges, electroencephalogram and behaviour in rats with absence epilepsy. Neuropharmacology 1995;34:419–25. [115] Palmer GC, Borrelli AR, Hudzik TJ, Sparber S. Acute heat stress model of seizures in weanling rats: influence of prototypic anti-seizure compounds. Epilepsy Res 1998;30:203–17. [116] Bialer M, Johannessen SI, Kupferberg HJ, Levy RH, Loiseau P, Perucca E. Progress report on new antiepileptic drugs: a summary of the fourth Eilat conference (EILAT IV). Epilepsy Res 1999;34:1–41. [117] Owen L, Cresswell P, Gifford C, McDade G, Mawer G. Influence of remacemide on EEG in chronic epilepsy. Epilepsy 1992;1:7. [118] Chadwick DW, Betts TA, Boddie HG, et al. Remacemide hydrochloride as an addon therapy in epilepsy: a randomized, placebo-controlled trial of three dose levels (300, 600 and 1200 mg/day) in a Q.I.D. regimen. Seizure 2002;11:114–23. [119] Jones MW, Blume WT, Guberman A, et al. Remacemide hydrochloride as an addon therapy in epilepsy: a randomized, placebo-controlled trial of three dose levels (300, 600 and 800 mg/day) in a B.I.D. regimen. Seizure 2002;11:104–13. [120] Devinsky O, Vazquez B, Faught E, et al. A double-blind, placebo-controlled study of remacemide hydrochloride in patients with refractory epilepsy following presurgical assessment. Seizure 2002;11:371–6. [121] Brodie MJ, Wroe SJ, Dean AD, Holdich TA, Whitehead J, Stevens JW. Efficacy and safety of remacemide versus carbamazepine in newly diagnosed epilepsy: comparison by sequential analysis. Epilepsy Behav 2002;3:140–6. [122] Rogawski MA, Yamaguchi S, Jones SM, Rice KC, Thurkauf A, Monn JA. Anticonvulsant activity of the low-affinity uncompetitive N-methyl-D- aspartate antagonist (+−)5-aminocarbonyl-10,11-dihydro-5H-dibenzo[a, d]cyclohepten-5,10-imine (ADCI): comparison with the structural analogs dizocilpine (MK-801) and carbamazepine. J Pharmacol Exp Ther 1991;259:30–7. [123] Sun L, Lin SS. The anticonvulsant SGB-017 (ADCI) blocks voltage-gated sodium channels in rat and human neurons: comparison with carbamazepine. Epilepsia 2000;41:263–70. [124] Seidleck BK, Thurkauf A, Witkin JM. Evaluation of ADCI against convulsant and locomotor stimulant effects of cocaine: comparison with the structural analogs dizocilpine and carbamazepine. Pharmacol Biochem Behav 1994;47:839–44. [125] Geter-Douglass B, Witkin JM. Behavioral effects and anticonvulsant efficacies of low-affinity, uncompetitive NMDA antagonists in mice. Psychopharmacology (Berl) 1999;146:280–9. [126] Coleman MH, Yamaguchi S, Rogawski MA. Protection against dendrotoxininduced clonic seizures in mice by anticonvulsant drugs. Brain Res 1992;575: 138–42. [127] Grant KA, Snell LD, Rogawski MA, Thurkauf A, Tabakoff B. Comparison of the effects of the uncompetitive N-methyl-D-aspartate antagonist (+−)-5aminocarbonyl-10,11-dihydro-5H-dibenzo[a, d] cyclohepten-5,10-imine (ADCI) with its structural analogs dizocilpine (MK-801) and carbamazepine on ethanol withdrawal seizures. J Pharmacol Exp Ther 1992;260:1017–22. [128] Mori K, Kawamata M, Mitani H, Yamazaki Y, Fujita M. A neurophysiologic study of ketamine anesthesia in the cat. Anesthesiology 1971;35:373–83. [129] Mealing GA, Lanthorn TH, Murray CL, Small DL, Morley P. Differences in degree of trapping of low-affinity uncompetitive N-methyl-D-aspartic acid receptor
634
[130]
[131]
[132]
[133]
[134]
[135]
[136]
[137]
[138]
[139]
[140] [141]
[142]
[143] [144]
[145]
[146] [147]
[148]
[149]
[150]
[151]
[152] [153] [154]
[155]
[156]
[157]
[158]
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640 antagonists with similar kinetics of block. J Pharmacol Exp Ther 1999;288: 204–10. Harrison NL, Simmonds MA. Quantitative studies on some antagonists of Nmethyl D-aspartate in slices of rat cerebral cortex. Br J Pharmacol 1985;84: 381–91. Machado-Vieira R, Salvadore G, Diazgranados N, Zarate Jr CA. Ketamine and the next generation of antidepressants with a rapid onset of action. Pharmacol Ther 2009;123:143–50. McCarthy DA, Cheng G, Kaump DH, Ensor C. General anesthetic and other pharmacological properties of 2-(O-chlorophenyl)-2-methylamino-cyclohexanone HCl (CI-581). J New Drugs 1965;5:21–33. Chen G, Ensor CR, Bohner B. The neuropharmacology of 2-(omicron-chlorophenyl)2-methylaminocyclohexanoe hydrochloride. J Pharmacol Exp Ther 1966;152: 332–9. Ghasemi M, Shafaroodi H, Nazarbeiki S, et al. Inhibition of NMDA receptor/NO signaling blocked tolerance to the anticonvulsant effect of morphine on pentylenetetrazole-induced seizures in mice. Epilepsy Res 2010;91:39–48. Ghasemi M, Shafaroodi H, Nazarbeiki S, et al. Voltage-dependent calcium channel and NMDA receptor antagonists augment anticonvulsant effects of lithium chloride on pentylenetetrazole-induced clonic seizures in mice. Epilepsy Behav 2010;18:171–8. Taberner PV. The anticonvulsant activity of ketamine against siezures induced by pentylenetetrazol and mercaptopropionic acid. Eur J Pharmacol 1976;39: 305–11. Wardley-Smith B, Little HJ, Halsey MJ. Lack of correlation between the anaesthetic and anti-convulsant potencies of althesin, ketamine and methohexitone. Br J Anaesth 1988;60:140–5. Tricklebank MD, Singh L, Oles RJ, Preston C, Iversen SD. The behavioural effects of MK-801: a comparison with antagonists acting non-competitively and competitively at the NMDA receptor. Eur J Pharmacol 1989;167:127–35. Veliskova J, Velisek L, Mares P, Rokyta R. Ketamine suppresses both bicucullineand picrotoxin-induced generalized tonic-clonic seizures during ontogenesis. Pharmacol Biochem Behav 1990;37:667–74. D'Hooge R, Pei YQ, De Deyn PP. N-methyl-D-aspartate receptors contribute to guanidinosuccinate-induced convulsions in mice. Neurosci Lett 1993;157:123–6. Velisek L, Vondrickova R, Mares P. Models of simple partial and absence seizures in freely moving rats: action of ketamine. Pharmacol Biochem Behav 1993;45: 889–96. Kubova H, Mares P. Effects of MK-801 (dizocilpine) and ketamine on strychnineinduced convulsions in rats: comparison with benzodiazepines and standard anticonvulsants. Physiol Res 1994;43:313–20. Herink J. Effect of alprazolam and ketamine on seizures induced by two different convulsants. Acta Medica (Hradec Kralove) 1997;40:9–11. Irifune M, Sato T, Kamata Y, Nishikawa T, Dohi T, Kawahara M. Evidence for GABA (A) receptor agonistic properties of ketamine: convulsive and anesthetic behavioral models in mice. Anesth Analg 2000;91:230–6. Stafstrom CE, Sasaki-Adams DM. NMDA-induced seizures in developing rats cause long-term learning impairment and increased seizure susceptibility. Epilepsy Res 2003;53:129–37. Guler G, Erdogan F, Golgeli A, Akin A, Boyaci A. Ketamine reduces lidocaineinduced seizures in mice. Int J Neurosci 2005;115:1239–44. Parsons CG, Quack G, Bresink I, et al. Comparison of the potency, kinetics and voltage-dependency of a series of uncompetitive NMDA receptor antagonists in vitro with anticonvulsive and motor impairment activity in vivo. Neuropharmacology 1995;34:1239–58. Sofia RD, Gordon R, Gels M, Diamantis W. Comparative effects of felbamate and other compounds on N-methyl-D-aspartic acid-induced convulsions and lethality in mice. Pharmacol Res 1994;29:139–44. Reder BS, Trapp LD, Troutman KC. Ketamine suppression of chemically induced convulsions in the two-day-old white leghorn cockerel. Anesth Analg 1980;59: 406–9. Bourn WM, Yang DJ, Davisson JN. Effect of ketamine enantiomers on soundinduced convulsions in epilepsy prone rats. Pharmacol Res Commun 1983;15: 815–24. De Sarro GB, De Sarro A. Anticonvulsant properties of non-competitive antagonists of the N-methyl-D-aspartate receptor in genetically epilepsyprone rats: comparison with CPPene. Neuropharmacology 1993;32:51–8. Bowyer JF, Albertson TE, Winters WD. Cortical kindled seizures: modification by excitant and depressant drugs. Epilepsia 1983;24:356–67. Bowyer JF, Albertson TE, Winters WD, Baselt RC. Ketamine-induced changes in kindled amygdaloid seizures. Neuropharmacology 1983;22:887–94. Trommer BL, Pasternak JF. NMDA receptor antagonists inhibit kindling epileptogenesis and seizure expression in developing rats. Brain Res Dev Brain Res 1990;53:248–52. Chapman AG, Meldrum BS. Non-competitive N-methyl-D-aspartate antagonists protect against sound-induced seizures in DBA/2 mice. Eur J Pharmacol 1989;166:201–11. Velisek L, Mikolasova R, Blankova-Vankova S, Mares P. Effects of ketamine on metrazol-induced seizures during ontogenesis in rats. Pharmacol Biochem Behav 1989;32:405–10. Kolesnikov Y, Jain S, Wilson R, Pasternak GW. Blockade of morphine-induced hindlimb myoclonic seizures in mice by ketamine. Pharmacol Biochem Behav 1997;56:423–5. Bertram EH, Lothman EW. NMDA receptor antagonists and limbic status epilepticus: a comparison with standard anticonvulsants. Epilepsy Res 1990;5: 177–84.
[159] Borris DJ, Bertram EH, Kapur J. Ketamine controls prolonged status epilepticus. Epilepsy Res 2000;42:117–22. [160] Serrano S, Hughes D, Chandler K. Use of ketamine for the management of refractory status epilepticus in a dog. J Vet Intern Med 2006;20:194–7. [161] Freitas RM, Sousa FC, Viana GS, Fonteles MM. Effect of gabaergic, glutamatergic, antipsychotic and antidepressant drugs on pilocarpine-induced seizures and status epilepticus. Neurosci Lett 2006;408:79–83. [162] Navarro V, Le Van Quyen M, Martinerie J, Rudrauf D, Baulac M, Menini C. Loss of phase synchrony in an animal model of partial status epilepticus. Neuroscience 2007;148:304–13. [163] Deutsch SI, Mastropaolo J, Riggs RL, Rosse RB. The antiseizure efficacies of MK801, phencyclidine, ketamine, and memantine are altered selectively by stress. Pharmacol Biochem Behav 1997;58:709–12. [164] Khanna N, Bhalla S. Role of ketamine in convulsions. Indian J Med Sci 1999;53: 475–80. [165] Manocha A, Sharma KK, Mediratta PK. Possible mechanism of anticonvulsant effect of ketamine in mice. Indian J Exp Biol 2001;39:1002–8. [166] Cook LL. Ventricular enlargement and the lithium/pilocarpine seizure model: possible explanation for agonistic behaviour. Int J Neurosci 2000;100:117–23. [167] Wurpel JN, Sperber EF, Moshe SL. Age-dependent differences in the anticonvulsant effects of 2-amino-7-phosphono-heptanoic acid or ketamine infusions into the substantia nigra of rats. Epilepsia 1992;33:439–43. [168] Borowicz KK, Luszczki J, Czuczwar SJ. Interactions between non-barbiturate injectable anesthetics and conventional antiepileptic drugs in the maximal electroshock test in mice—an isobolographic analysis. Eur Neuropsychopharmacol 2004;14:163–72. [169] Martin BS, Kapur J. A combination of ketamine and diazepam synergistically controls refractory status epilepticus induced by cholinergic stimulation. Epilepsia 2008;49:248–55. [170] Vermoesen K, Smolders I, Massie A, Michotte Y, Clinckers R. The control of kainic acid-induced status epilepticus. Epilepsy Res 2010;90:164–6. [171] Dorandeu F, Carpentier P, Baubichon D, et al. Efficacy of the ketamine–atropine combination in the delayed treatment of soman-induced status epilepticus. Brain Res 2005;1051:164–75. [172] Dorandeu F, Baille V, Mikler J, et al. Protective effects of S+ ketamine and atropine against lethality and brain damage during soman-induced status epilepticus in guinea-pigs. Toxicology 2007;234:185–93. [173] Celesia GG, Chen RC. Effects of ketamine on EEG activity in cats and monkeys. Electroencephalogr Clin Neurophysiol 1974;37:354-53. [174] Myslobodsky MS, Golovchinsky V, Mintz M. Ketamine: convulsant or anticonvulsant? Pharmacol Biochem Behav 1981;14:27–33. [175] Brady RJ, Swann JW. Ketamine selectively suppresses synchronized afterdischarges in immature hippocampus. Neurosci Lett 1986;69:143–9. [176] Lee WL, Hablitz JJ. Effect of APV and ketamine on epileptiform activity in the CA1 and CA3 regions of the hippocampus. Epilepsy Res 1990;6:87–94. [177] Mikolasova R, Velisek L, Vorlicek J, Mares P. Developmental changes of ketamine action against epileptic afterdischarges induced by hippocampal stimulation in rats. Brain Res Dev Brain Res 1994;81:105–12. [178] Cunha AO, Mortari MR, Liberato JL, dos Santos WF. Neuroprotective effects of diazepam, carbamazepine, phenytoin and ketamine after pilocarpine-induced status epilepticus. Basic Clin Pharmacol Toxicol 2009;104:470–7. [179] Aram JA, Lodge D. Epileptiform activity induced by alkalosis in rat neocortical slices: block by antagonists of N-methyl-D-aspartate. Neurosci Lett 1987;83: 345–50. [180] Sagratella S, Frank C, de Carolis AS. Effects of ketamine and (+)cyclazocine on 4aminopyridine and “magnesium free” epileptogenic activity in hippocampal slices of rats. Neuropharmacology 1987;26:1181–4. [181] Kohr G, Heinemann U. Effects of NMDA antagonists on picrotoxin-, low Mg2+and low Ca2+-induced epileptogenesis and on evoked changes in extracellular Na+ and Ca2+ concentrations in rat hippocampal slices. Epilepsy Res 1989;4: 187–200. [182] Ashton D, Willems R, De Prins E, Wauquier A. Selective inhibition of synaptic versus non-synaptic epileptogenesis by NMDA antagonists in the in vitro hippocampus. Epilepsy Res 1988;2:219–22. [183] Zhang CL, Gloveli T, Heinemann U. Effects of NMDA- and AMPA-receptor antagonists on different forms of epileptiform activity in rat temporal cortex slices. Epilepsia 1994;35(Suppl 5):S68–73. [184] Mares P, Lanstiakova M, Vankova S, Kubova H, Velisek L. Ketamine blocks cortical epileptic afterdischarges but not paired-pulse and frequency potentiation. Neuroscience 1992;50:339–44. [185] Clifford DB, Zorumski CF, Olney JW. Ketamine and MK-801 prevent degeneration of thalamic neurons induced by focal cortical seizures. Exp Neurol 1989;105:272–9. [186] Clifford DB, Olney JW, Benz AM, Fuller TA, Zorumski CF. Ketamine, phencyclidine, and MK-801 protect against kainic acid-induced seizure-related brain damage. Epilepsia 1990;31:382–90. [187] Hsieh CY, Sung PS, Tsai JJ, Huang CW. Terminating prolonged refractory status epilepticus using ketamine. Clin Neuropharmacol 2010;33:165–7. [188] Winters WD. Epilepsy or anesthesia with ketamine. Anesthesiology 1972;36: 309–12. [189] Miyasaka M, Domino EF. Neural mechanisms of ketamine-induced anesthesia. Int J Neuropharmacol 1968;7:557–73. [190] Manohar S, Maxwell D, Winters WD. Development of EEG seizure activity during and after chronic ketamine administration in the rat. Neuropharmacology 1972;11:819–26. [191] Corssen G, Domino EF, Bree RL. Electroencephalographic effects of ketamine anesthesia in children. Anesth Analg 1969;48:141–7.
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640 [192] Kayama Y, Iwama K. The EEG, evoked potentials, and single-unit activity during ketamine anesthesia in cats. Anesthesiology 1972;36:316–28. [193] Black JA, Golden GT, Fariello RG. Ketamine activation of experimental corticoreticular epilepsy. Neurology 1980;30:315–8. [194] Lunn RJ, Savageau MM, Beatty WW, Gerst JW, Staton RD, Brumback RA. Anesthetics and electroconvulsive therapy seizure duration: implications for therapy from a rat model. Biol Psychiatry 1981;16:1163–75. [195] DeVore GR, McQueen JK, Woodbury DM. Ketamine hydrochloride and its effect on a chronic cobalt epileptic cortical focus. Epilepsia 1976;17:111–7. [196] Thompson GE. Ketamine-induced convulsions. Anesthesiology 1972;37:662–3. [197] Hirshman CA, Krieger W, Littlejohn G, Lee R, Julien R. Ketamine-aminophyllineinduced decrease in seizure threshold. Anesthesiology 1982;56:464–7. [198] Ferrer-Allado T, Brechner VL, Dymond A, Cozen H, Crandall P. Ketamine-induced electroconvulsive phenomena in the human limbic and thalamic regions. Anesthesiology 1973;38:333–44. [199] Corssen G, Little SC, Tavakoli M. Ketamine and epilepsy. Anesth Analg 1974;53: 319–35. [200] Celesia GG, Chen RC, Bamforth BJ. Effects of ketamine in epilepsy. Neurology 1975;25:169–72. [201] Corssen G, Miyasaka M, Domino EF. Changing concepts in pain control during surgery: dissociative anesthesia with CI-581. A progress report. Anesth Analg 1968;47:746–59. [202] Davis RW, Tolstoshev GC. Ketamine—use in severe febrile convulsions. Med J Aust 1976;2:465–6. [203] Sheth RD, Gidal BE. Refractory status epilepticus: response to ketamine. Neurology 1998;51:1765–6. [204] Mewasingh LD, Sekhara T, Aeby A, Christiaens FJ, Dan B. Oral ketamine in paediatric non-convulsive status epilepticus. Seizure 2003;12:483–9. [205] Nathan BR, Smith TL, Bleck TP. The use of ketamine in the treatment of refractory status epilepticus. Neurology 2002(Suppl 3):A197. [206] Bleck TP, Quigg M, Nathan BR, Smith TL, Kapur J. Electroencephalographic effects of ketamine treatment for refractory status epilepticus. Epilepsia 2002;43:282. [207] Ubogu EE, Sagar SM, Lerner AJ, Maddux BN, Suarez JI, Werz MA. Ketamine for refractory status epilepticus: a case of possible ketamine-induced neurotoxicity. Epilepsy Behav 2003;4:70–5. [208] Pruss H, Holtkamp M. Ketamine successfully terminates malignant status epilepticus. Epilepsy Res 2008;82:219–22. [209] Davies WL, Grunert RR, Haff RF, et al. Antiviral activity of 1-adamantanamine (amantadine). Science 1964;144:862–3. [210] Schwab RS, England Jr AC. Amantadine HCL (Symmetrel) and its relation to LevoDopa in the treatment of Parkinson's disease. Trans Am Neurol Assoc 1969;94: 85–90. [211] Kornhuber J, Bormann J, Hubers M, Rusche K, Riederer P. Effects of the 1amino-adamantanes at the MK-801-binding site of the NMDA-receptor-gated ion channel: a human postmortem brain study. Eur J Pharmacol 1991;206: 297–300. [212] Lupp A, Lucking CH, Koch R, Jackisch R, Feuerstein TJ. Inhibitory effects of the antiparkinsonian drugs memantine and amantadine on N-methyl-D-aspartateevoked acetylcholine release in the rabbit caudate nucleus in vitro. J Pharmacol Exp Ther 1992;263:717–24. [213] Blanpied TA, Clarke RJ, Johnson JW. Amantadine inhibits NMDA receptors by accelerating channel closure during channel block. J Neurosci 2005;25:3312–22. [214] Lazarova M, Roussinov K. On some relationships between dopaminergic and serotoninergic mechanisms in pentylenetetrazol convulsions in albino mice. Acta Physiol Pharmacol Bulg 1979;5:67–74. [215] Kleinrok Z, Czuczwar S, Wojcik A, Przegalinski E. Brain dopamine and seizure susceptibility in mice. Pol J Pharmacol Pharm 1978;30:513–9. [216] Lazarova MB, Roussinov KS. On certain effects of dopaminergic agents in pentylenetetrazol convulsions. Acta Physiol Pharmacol Bulg 1978;4:50–5. [217] Kleinrok Z, Czuczwar SJ, Kozicka M. Effect of dopaminergic and GABA-ergic drugs given alone or in combination on the anticonvulsant action of phenobarbital and diphenylhydantoin in the electroshock test in mice. Epilepsia 1980;21:519–29. [218] Kleinrok Z, Czuczwar SJ, Kozicka M, Zarkowski A. Effect of combined GABA-ergic and dopaminergic stimulation on the action of some antiepileptic drugs in pentylenetetrazol-induced convulsions. Pol J Pharmacol Pharm 1981;33:13–23. [219] Reis J, John D, Heimeroth A, Mueller HH, Oertel WH, Arndt T, et al. Modulation of human motor cortex excitability by single doses of amantadine. Neuropsychopharmacology 2006;31:2758–66. [220] Rohrbacher J, Bijak M, Misgeld U. Suppression by memantine and amantadine of synaptic excitation intrastriatally evoked in rat neostriatal slices. Neurosci Lett 1994;182:95–8. [221] Vamvakides A. Are there glutamatergic, anti-GABAergic or antiglycinergic components in the action mechanism of adamantamines? Study of convulsions induced by amantadine or 1-adamantylcyclopentanamine in mice. Ann Pharm Fr 1990;48:312–20. [222] Shields WD, Lake JL, Chugani HT. Amantadine in the treatment of refractory epilepsy in childhood: an open trial in 10 patients. Neurology 1985;35:579–81. [223] Drake Jr ME, Pakalnis A, Denio LS, Phillips B. Amantadine hydrochloride for refractory generalized epilepsy in adults. Acta Neurol Belg 1991;91:159–64. [224] Shahar EM, Brand N. Effect of add-on amantadine therapy for refractory absence epilepsy. J Pediatr 1992;121:819–21. [225] Matsushige T, Matsufuji H, Ichiyama T, Furukawa S. A girl with refractory childhood absence epilepsy improved by add-on amantadine therapy. No To Hattatsu 2007;39:309–10.
635
[226] Claudet I, Marechal C. Status epilepticus in a pediatric patient with amantadine overdose. Pediatr Neurol 2009;40:120–2. [227] Chojnacka-Wojcik E, Tatarczynska E, Maj J. The influence of memantine on the anticonvulsant effects of the antiepileptic drugs. Pol J Pharmacol Pharm 1983;35: 511–5. [228] McLean MJ. In vitro electrophysiological evidence predicting anticonvulsant efficacy of memantine and flunarizine. Pol J Pharmacol Pharm 1987;39:513–25. [229] Urbanska E, Dziki M, Czuczwar SJ, Kleinrok Z, Turski WA. Antiparkinsonian drugs memantine and trihexyphenidyl potentiate the anticonvulsant activity of valproate against maximal electroshock-induced seizures. Neuropharmacology 1992;31:1021–6. [230] Kleinrok Z, Turski WA, Czuczwar SJ. Excitatory amino acid antagonists and the anticonvulsive activity of conventional antiepileptic drugs. Pol J Pharmacol 1995;47:247–52. [231] Czuczwar SJ, Turski WA, Kleinrok Z. Interactions of excitatory amino acid antagonists with conventional antiepileptic drugs. Metab Brain Dis 1996;11: 143–52. [232] Meldrum BS, Turski L, Schwarz M, Czuczwar SJ, Sontag KH. Anticonvulsant action of 1,3-dimethyl-5-aminoadamantane. Pharmacological studies in rodents and baboon, Papio papio. Naunyn Schmiedebergs Arch Pharmacol 1986;332:93–7. [233] Bisaga A, Krzascik P, Jankowska E, Palejko W, Kostowski W, Danysz W. Effect of glutamate receptor antagonists on N-methyl-D-aspartate- and (S)-alpha-amino3-hydroxy-5-methyl-4-isoxazolepropionic acid-induced convulsant effects in mice and rats. Eur J Pharmacol 1993;242:213–20. [234] Lukomskaia N, Rukoiatkina NI, Gorbunova LV, Gmiro VE, Magazanik LG. Role of NMDA and AMPA glutamate receptors in the mechanism of korazol-induced convulsions in mice. Ross Fiziol Zh Im I M Sechenova 2003;89:292–301. [235] Lukomskaya NY, Rukoyatkina NI, Gorbunova LV, Gmiro VE, Magazanik LG. Studies of the roles of NMDA and AMPA glutamate receptors in the mechanism of corasole convulsions in mice. Neurosci Behav Physiol 2004;34:783–9. [236] Mares P, Mikulecka A. Different effects of two N-methyl-D-aspartate receptor antagonists on seizures, spontaneous behavior, and motor performance in immature rats. Epilepsy Behav 2009;14:32–9. [237] Brackett RL, Pouw B, Blyden JF, Nour M, Matsumoto RR. Prevention of cocaineinduced convulsions and lethality in mice: effectiveness of targeting different sites on the NMDA receptor complex. Neuropharmacology 2000;39:407–18. [238] Bienkowski P, Krzascik P, Koros E, Kostowski W, Scinska A, Danysz W. Effects of a novel uncompetitive NMDA receptor antagonist, MRZ 2/579 on ethanol selfadministration and ethanol withdrawal seizures in the rat. Eur J Pharmacol 2001;413:81–9. [239] McLean MJ, Gupta RC, Dettbarn WD, Wamil AW. Prophylactic and therapeutic efficacy of memantine against seizures produced by soman in the rat. Toxicol Appl Pharmacol 1992;112:95–103. [240] Deshpande SS, Smith CD, Filbert MG. Assessment of primary neuronal culture as a model for soman-induced neurotoxicity and effectiveness of memantine as a neuroprotective drug. Arch Toxicol 1995;69:384–90. [241] Shih T, McDonough Jr JH, Koplovitz I. Anticonvulsants for soman-induced seizure activity. J Biomed Sci 1999;6:86–96. [242] Vataev SI, Zhabko EP, Lukomskaia N, Oganesian GA, Magazanik LG. Effects of memantine on convulsive reactions and sleep–waking cycle in KrushinskiiMolodkina strain rats with the inherited predisposition to audiogenic convulsions. Ross Fiziol Zh Im I M Sechenova 2009;95:802–12. [243] Vataev SI, Zhabko EP, Lukomskaya NY, Oganesyan GA, Magazanik LG. Effects of memantine on convulsive reactions and the organization of sleep in KrushinskiiMolodkina rats with an inherited predisposition to audiogenic convulsions. Neurosci Behav Physiol 2010;40:913–9. [244] Frey HH, Voits M. Effect of psychotropic agents on a model of absence epilepsy in rats. Neuropharmacology 1991;30:651–6. [245] Apland JP, Cann FJ. Anticonvulsant effects of memantine and MK-801 in guinea pig hippocampal slices. Brain Res Bull 1995;37:311–6. [246] Zaja-Milatovic S, Gupta RC, Aschner M, Milatovic D. Protection of DFP-induced oxidative damage and neurodegeneration by antioxidants and NMDA receptor antagonist. Toxicol Appl Pharmacol 2009;240:124–31. [247] Wang XM, Bausch SB. Effects of distinct classes of N-methyl-D-aspartate receptor antagonists on seizures, axonal sprouting and neuronal loss in vitro: suppression by NR2B-selective antagonists. Neuropharmacology 2004;47:1008–20. [248] Richter A, Fredow G, Loscher W. Antidystonic effects of the NMDA receptor antagonists memantine, MK-801 and CGP 37849 in a mutant hamster model of paroxysmal dystonia. Neurosci Lett 1991;133:57–60. [249] Loscher W, Honack D. Over-additive anticonvulsant effect of memantine and NBQX in kindled rats. Eur J Pharmacol 1994;259:R3–5. [250] Loscher W, Honack D. High doses of memantine (1-amino-3,5-dimethyladamantane) induce seizures in kindled but not in non-kindled rats. Naunyn Schmiedebergs Arch Pharmacol 1990;341:476–81. [251] Shoji K, Mariotto S, Ciampa AR, Suzuki H. Regulation of serine racemace activity by d-serine and nitric oxide in human glioblastoma cells. Neurosci Lett 2006;392: 75–8. [252] Gmiro VE, Serdyuk SE. Combined blockade of AMPA and NMDA receptors in the brain of rats prevents pentylenetetrazole-induced clonic and tonic–clonic seizures without ataxia. Bull Exp Biol Med 2008;145:728–30. [253] Peltz G, Pacific DM, Noviasky JA, Shatla A, Mehalic T. Seizures associated with memantine use. Am J Health Syst Pharm 2005;62:420–1. [254] Weinbroum AA, Ben-Abraham R. Dextromethorphan and dexmedetomidine: new agents for the control of perioperative pain. Eur J Surg 2001;167:563–9. [255] Fisher RS, Cysyk BJ, Lesser RP, et al. Dextromethorphan for treatment of complex partial seizures. Neurology 1990;40:547–9.
636
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
[256] Schmitt B, Netzer R, Fanconi S, Baumann P, Boltshauser E. Drug refractory epilepsy in brain damage: effect of dextromethorphan on EEG in four patients. J Neurol Neurosurg Psychiatry 1994;57:333–9. [257] Kimiskidis VK, Mirtsou-Fidani V, Papaioannidou PG, et al. A phase I clinical trial of dextromethorphan in intractable partial epilepsy. Methods Find Exp Clin Pharmacol 1999;21:673–8. [258] Kazis A, Kimiskidis V, Niopas I. Pharmacokinetics of dextromethorphan and dextrorphan in epileptic patients. Acta Neurol Scand 1996;93:94–8. [259] Feeser HR, Kadis JL, Prince DA. Dextromethorphan, a common antitussive, reduces kindled amygdala seizures in the rat. Neurosci Lett 1988;86:340–5. [260] Palmer GC, Harris EW, Ray R, Stagnitto ML, Schmiesing RJ. Classification of compounds for prevention of NMDA-induced seizures/mortality, or maximal electroshock and pentylenetetrazol seizures in mice and antagonism of MK801 binding in vitro. Arch Int Pharmacodyn Ther 1992;317:16–34. [261] Tortella FC, Musacchio JM. Dextromethorphan and carbetapentane: centrally acting non-opioid antitussive agents with novel anticonvulsant properties. Brain Res 1986;383:314–8. [262] Moreau JL, Pieri L, Prud'hon B. Convulsions induced by centrally administered NMDA in mice: effects of NMDA antagonists, benzodiazepines, minor tranquilizers and anticonvulsants. Br J Pharmacol 1989;98:1050–4. [263] Ferkany JW, Borosky SA, Clissold DB, Pontecorvo MJ. Dextromethorphan inhibits NMDA-induced convulsions. Eur J Pharmacol 1988;151:151–4. [264] Zhi JG, Levy G. Effect of orally administered dextromethorphan on theophyllineand pentylenetetrazol-induced seizures in rats. J Pharm Sci 1990;79:1053–5. [265] Kim HC, Ko KH, Kim WK, et al. Effects of dextromethorphan on the seizures induced by kainate and the calcium channel agonist BAY k-8644: comparison with the effects of dextrorphan. Behav Brain Res 2001;120:169–75. [266] Barat SA, Abdel-Rahman MS. Decreased cocaine- and lidocaine-induced seizure response by dextromethorphan and DNQX in rat. Brain Res 1997;756:179–83. [267] Erden BF, Ozdemirci S, Yildiran G, Utkan T, Gacar N, Ulak G. Dextromethorphan attenuates ethanol withdrawal syndrome in rats. Pharmacol Biochem Behav 1999;62:537–41. [268] Sparenborg S, Brennecke LH, Braitman DJ. Prevention of soman neurotoxicity by non-opioid antitussives. Neurotoxicology 1990;11:509–20. [269] Laroia N, McBride L, Baggs R, Guillet R. Dextromethorphan ameliorates effects of neonatal hypoxia on brain morphology and seizure threshold in rats. Brain Res Dev Brain Res 1997;100:29–34. [270] Mizoule J, Meldrum B, Mazadier M, et al. 2-Amino-6-trifluoromethoxy benzothiazole, a possible antagonist of excitatory amino acid neurotransmission—I. Anticonvulsant properties. Neuropharmacology 1985;24:767–73. [271] Malgouris C, Daniel M, Doble A. Neuroprotective effects of riluzole on N-methylD-aspartate- or veratridine-induced neurotoxicity in rat hippocampal slices. Neurosci Lett 1994;177:95–9. [272] Prakriya M, Mennerick S. Selective depression of low-release probability excitatory synapses by sodium channel blockers. Neuron 2000;26:671–82. [273] Debono MW, Le Guern J, Canton T, Doble A, Pradier L. Inhibition by riluzole of electrophysiological responses mediated by rat kainate and NMDA receptors expressed in Xenopus oocytes. Eur J Pharmacol 1993;235:283–9. [274] Hubert JP, Delumeau JC, Glowinski J, Premont J, Doble A. Antagonism by riluzole of entry of calcium evoked by NMDA and veratridine in rat cultured granule cells: evidence for a dual mechanism of action. Br J Pharmacol 1994;113:261–7. [275] Fumagalli E, Funicello M, Rauen T, Gobbi M, Mennini T. Riluzole enhances the activity of glutamate transporters GLAST, GLT1 and EAAC1. Eur J Pharmacol 2008;578:171–6. [276] Romettino S, Lazdunski M, Gottesmann C. Anticonvulsant and sleep-waking influences of riluzole in a rat model of absence epilepsy. Eur J Pharmacol 1991;199:371–3. [277] De Sarro G, Siniscalchi A, Ferreri G, Gallelli L, De Sarro A. NMDA and AMPA/kainate receptors are involved in the anticonvulsant activity of riluzole in DBA/2 mice. Eur J Pharmacol 2000;408:25–34. [278] Yoshida M, Noguchi E, Tsuru N, Ohkoshi N. Effect of riluzole on the acquisition and expression of amygdala kindling. Epilepsy Res 2001;46:101–9. [279] Borowicz KK, Rwiader M, Drelewska E, Czuczwar SJ. Interactions between riluzole and conventional antiepileptic drugs — a comparison of results obtained in the subthreshold method and isobolographic analysis. J Neural Transm 2004;111:1511–22. [280] Kim JE, Kim DS, Kwak SE, et al. Anti-glutamatergic effect of riluzole: comparison with valproic acid. Neuroscience 2007;147:136–45. [281] Besheer J, Lepoutre V, Hodge CW. Preclinical evaluation of riluzole: assessments of ethanol self-administration and ethanol withdrawal symptoms. Alcohol Clin Exp Res 2009;33:1460–8. [282] Borowicz KK, Sekowski A, Drelewska E, Czuczwar SJ. Riluzole enhances the antiseizure action of conventional antiepileptic drugs against pentylenetetrazolinduced convulsions in mice. Pol J Pharmacol 2004;56:187–93. [283] Monahan JB, Corpus VM, Hood WF, Thomas JW, Compton RP. Characterization of a [3H]glycine recognition site as a modulatory site of the N-methyl-D-aspartate receptor complex. J Neurochem 1989;53:370–5. [284] Emmett MR, Mick SJ, Cler JA, Rao TS, Iyengar S, Wood PL. Actions of D-cycloserine at the N-methyl-D-aspartate-associated glycine receptor site in vivo. Neuropharmacology 1991;30:1167–71. [285] Hood WF, Compton RP, Monahan JB. D-cycloserine: a ligand for the N-methyl-Daspartate coupled glycine receptor has partial agonist characteristics. Neurosci Lett 1989;98:91–5. [286] Henderson G, Johnson JW, Ascher P. Competitive antagonists and partial agonists at the glycine modulatory site of the mouse N-methyl-D-aspartate receptor. J Physiol 1990;430:189–212.
[287] Priestley T, Kemp JA. Kinetic study of the interactions between the glutamate and glycine recognition sites on the N-methyl-D-aspartic acid receptor complex. Mol Pharmacol 1994;46:1191–6. [288] Peterson SL. Localization of an anatomic substrate for the anticonvulsant activity induced by D-cycloserine. Epilepsia 1994;35:933–8. [289] Peterson SL, Schwade ND. The anticonvulsant activity of D-cycloserine is specific for tonic convulsions. Epilepsy Res 1993;15:141–8. [290] Monaco P. Studies on pharmacological activity of 1,4-amino-3-isoxazolidone. Boll Soc Ital Biol Sper 1958;34:1464–6. [291] Wood JD, Russell MP, Kurylo E. The gamma-aminobutyrate content of nerve endings (synaptosomes) in mice after the intramuscular injection of gammaaminobutyrate-elevating agents: a possible role in anticonvulsant activity. J Neurochem 1980;35:125–30. [292] Chung SH, Johnson MS, Gronenborn AM. L-cycloserine: a potent anticonvulsant. Epilepsia 1984;25:353–62. [293] Scotto P, Monaco P, Scardi V, Bonavita V. Neurochemical studies with Lcycloserine, a central depressant agent. J Neurochem 1963;10:831–9. [294] Polc P, Pieri L, Bonetti EP, et al. L-cycloserine: behavioural and biochemical effects after single and repeated administration to mice, rats and cats. Neuropharmacology 1986;25:411–8. [295] Baran H, Loscher W, Mevissen M. The glycine/NMDA receptor partial agonist Dcycloserine blocks kainate-induced seizures in rats. Comparison with MK-801 and diazepam. Brain Res 1994;652:195–200. [296] Loscher W, Wlaz P, Rundfeldt C, Baran H, Honack D. Anticonvulsant effects of the glycine/NMDA receptor ligands D-cycloserine and D-serine but not R-(+)-HA966 in amygdala-kindled rats. Br J Pharmacol 1994;112:97–106. [297] Rundfeldt C, Wlaz P, Loscher W. Anticonvulsant activity of antagonists and partial agonists for the NMDA receptor-associated glycine site in the kindling model of epilepsy. Brain Res 1994;653:125–30. [298] Wlaz P, Baran H, Loscher W. Effect of the glycine/NMDA receptor partial agonist, D-cycloserine, on seizure threshold and some pharmacodynamic effects of MK801 in mice. Eur J Pharmacol 1994;257:217–25. [299] Peterson SL. Infusion of NMDA antagonists into the nucleus reticularis pontis oralis inhibits the maximal electroshock seizure response. Brain Res 1995;702:101–9. [300] Matsumoto RR, Brackett RL, Kanthasamy AG. Novel NMDA/glycine site antagonists attenuate cocaine-induced behavioral toxicity. Eur J Pharmacol 1997;338:233–42. [301] Wlaz P. Anti-convulsant and adverse effects of the glycine B receptor ligands, Dcycloserine and L-701,324: comparison with competitive and non-competitive N-methyl-D-aspartate receptor antagonists. Brain Res Bull 1998;46:535–40. [302] De Sarro G, Gratteri S, Naccari F, Pasculli MP, De Sarro A. Influence of Dcycloserine on the anticonvulsant activity of some antiepileptic drugs against audiogenic seizures in DBA/2 mice. Epilepsy Res 2000;40:109–21. [303] Kass JS, Shandera WX. Nervous system effects of antituberculosis therapy. CNS Drugs 2010;24:655–67. [304] Karler R, Calder LD, Chaudhry IA, Turkanis SA. Blockade of “reverse tolerance” to cocaine and amphetamine by MK-801. Life Sci 1989;45:599–606. [305] Kulkarni SK, Ticku MK. Interaction between GABAergic anticonvulsants and the NMDA receptor antagonist MK 801 against MES- and picrotoxin-induced convulsions in rats. Life Sci 1989;44:1317–23. [306] O'Neill SK, Bolger GT. Anticonvulsant activity of MK-801 and nimodipine alone and in combination against pentylenetetrazole and strychnine. Pharmacol Biochem Behav 1989;32:595–600. [307] Vezzani A, Serafini R, Stasi MA, et al. Kinetics of MK-801 and its effect on quinolinic acid-induced seizures and neurotoxicity in rats. J Pharmacol Exp Ther 1989;249:278–83. [308] Itzhak Y, Stein I. Sensitization to the toxic effects of cocaine in mice is associated with the regulation of N-methyl-D-aspartate receptors in the cortex. J Pharmacol Exp Ther 1992;262:464–70. [309] Kulkarni SK, Mehta AK, Ticku MK. Comparison of anticonvulsant effect of ethanol against NMDA-, kainic acid- and picrotoxin-induced convulsions in rats. Life Sci 1990;46:481–7. [310] Turski WA, Urbanska E, Dziki M, Parada-Turska J, Ikonomidou C. Excitatory amino acid antagonists protect mice against seizures induced by bicuculline. Brain Res 1990;514:131–4. [311] Thorat SN, Kulkarni SK. Antagonism of caffeine-induced convulsions by ethanol and dizocilpine (MK-801) in mice. Methods Find Exp Clin Pharmacol 1991;13:413–7. [312] Wardley-Smith B, Wann KT. Effects of four drugs on 4-aminopyridine seizures: a comparison with their effects on HPNS. Undersea Biomed Res 1991;18:413–9. [313] Tusell JM, Vendrell M, Serratosa J, Trullas R. Lindane-induced convulsions in NMRI and OF1 mice: antagonism with (+)MK-801 and voltage-dependent calcium channel blockers. Brain Res 1992;593:209–14. [314] Lukomskaya NY, Rukoyatkina NI, Gorbunova LV, Gmiro VE, Bol'shakov KV, Magazanik LG. Comparison of the anticonvulsive activities of organic mono- and di-cations with their abilities to inhibit NMDA and AMPA glutamate receptors. Neurosci Behav Physiol 2004;34:181–7. [315] Chavko M, Braisted JC, Harabin AL. Effect of MK-801 on seizures induced by exposure to hyperbaric oxygen: comparison with AP-7. Toxicol Appl Pharmacol 1998;151:222–8. [316] Dewhurst DG. Possible role of excitatory amino acids in the convulsant action of catechol. Eur J Pharmacol 1988;145:209–12. [317] Gilbert ME. The NMDA-receptor antagonist, MK-801, suppresses limbic kindling and kindled seizures. Brain Res 1988;463:90–9. [318] McNamara JO, Russell RD, Rigsbee L, Bonhaus DW. Anticonvulsant and antiepileptogenic actions of MK-801 in the kindling and electroshock models. Neuropharmacology 1988;27:563–8.
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640 [319] Sato K, Morimoto K, Okamoto M. Anticonvulsant action of a non-competitive antagonist of NMDA receptors (MK-801) in the kindling model of epilepsy. Brain Res 1988;463:12–20. [320] Giorgi O, Orlandi M, Lecca D, Corda MG. MK-801 prevents chemical kindling induced by pentylenetetrazol in rats. Eur J Pharmacol 1991;193:363–5. [321] Braitman DJ, Sparenborg S. MK-801 protects against seizures induced by the cholinesterase inhibitor soman. Brain Res Bull 1989;23:145–8. [322] Morrisett RA, Rezvani AH, Overstreet D, Janowsky DS, Wilson WA, Swartzwelder HS. MK-801 potently inhibits alcohol withdrawal seizures in rats. Eur J Pharmacol 1990;176:103–5. [323] Pedder SC, Wilcox RI, Tuchek JM, Johnson DD, Crawford RD. Attenuation of febrile seizures in epileptic chicks by N-methyl-D-aspartate receptor antagonists. Can J Physiol Pharmacol 1990;68:84–8. [324] Thorat SN, Kulkarni SK. Effect of MK-801 and its interaction with adenosinergic agents and carbamazepine against hypoxic stress-induced convulsions and death in mice. Methods Find Exp Clin Pharmacol 1990;12:595–600. [325] Wada Y, Hasegawa H, Nakamura M, Yamaguchi N. Action of the N-methyl-Daspartate receptor antagonist MK-801 against focal seizure activity from the feline hippocampus. Neuropsychobiology 1992;26:205–9. [326] De Sarro GB, Bagetta G, Spagnolo C, Nistico G. Antagonists of N-methyl-Daspartate receptors block seizures induced by putrescine in the deep prepiriform cortex. Neuropharmacology 1993;32:43–50. [327] Sato K, Morimoto K, Hiramatsu M, Mori A, Otsuki S. Effect of a noncompetitive antagonist (MK-801) of NMDA receptors on convulsions and brain amino acid level in E1 mice. Neurochem Res 1989;14:741–4. [328] Komatsu H, Nogaya J, Anabuki D, Ogli K. The N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine (MK-801) suppresses enflurane-induced opisthotonus in mice. J Anesth 1993;7:520–3. [329] Peterson SL, Boehnke LE. Anticonvulsant effects of MK-801 and glycine on hippocampal afterdischarge. Exp Neurol 1989;104:113–7. [330] Hughes P, Young D, Dragunow M. MK-801 sensitizes rats to pilocarpine induced limbic seizures and status epilepticus. Neuroreport 1993;4:314–6. [331] Leander JD, Rathbun RC, Zimmerman DM. Anticonvulsant effects of phencyclidinelike drugs: relation to N-methyl-D-aspartic acid antagonism. Brain Res 1988;454: 368–72. [332] Ormandy GC, Jope RS, Snead III OC. Anticonvulsant actions of MK-801 on the lithium-pilocarpine model of status epilepticus in rats. Exp Neurol 1989;106: 172–80. [333] Urbanska E, Dziki M, Kleinrok Z, Czuczwar SJ, Turski WA. Influence of MK-801 on the anticonvulsant activity of antiepileptics. Eur J Pharmacol 1991;200:277–82. [334] Zhong WZ, Zhao YJ, Gao GM, Yang ZH, Yue W. Effect of combination of dizocilpine with general antiepileptic drugs on amygdala kindling models in rats. Yao Xue Xue Bao 2004;39:89–92. [335] Troupin AS, Mendius JR, Cheng F, Risinger MW. MK-801. In: Meldrum BS, Porter RJ, editors. New anticonvulsant drugs. London, Paris: John Libbey; 1986. p. 191–202. [336] Pontecorvo MJ, Karbon EW, Goode S, et al. Possible cerebroprotective and in vivo NMDA antagonist activities of sigma agents. Brain Res Bull 1991;26: 461–5. [337] Singh L, Oles RJ, Vass CA, Woodruff GN. A slow intravenous infusion of N-methylDL-aspartate as a seizure model in the mouse. J Neurosci Methods 1991;37: 227–32. [338] Doyle KM, Shaw GG. Investigation of the involvement of the N-methyl-Daspartate receptor macrocomplex in the development of spermine-induced CNS excitation in vivo. Br J Pharmacol 1996;117:1803–8. [339] Tsuda M, Suzuki T, Misawa M. Age-related decrease in the antiseizure effect of ifenprodil against pentylenetetrazole in mice. Brain Res Dev Brain Res 1997;104: 201–4. [340] Maroso M, Balosso S, Ravizza T, Liu J, Aronica E, Iyer AM, et al. Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures. Nat Med 2010;16:413–9. [341] McAllister KH. N-methyl-D-aspartate receptor antagonists and channel blockers have different effects upon a spinal seizure model in mice. Eur J Pharmacol 1992;211:105–8. [342] Zarnowski T, Kleinrok Z, Turski WA, Czuczwar SJ. The NMDA antagonist procyclidine, but not ifenprodil, enhances the protective efficacy of common antiepileptics against maximal electroshock-induced seizures in mice. J Neural Transm Gen Sect 1994;97:1–12. [343] Hironaka N, Niki H. Effects of N-methyl-D-aspartate receptor subunit antagonists on regulation of susceptibility to audiogenic seizures in rats. Neurosci Lett 2000;288:139–42. [344] Malinowska B, Napiorkowska-Pawlak D, Pawlak R, Buczko W, Gothert M. Ifenprodil influences changes in mouse behaviour related to acute and chronic ethanol administration. Eur J Pharmacol 1999;377:13–9. [345] Yourick DL, Repasi RT, Rittase WB, Staten LD, Meyerhoff JL. Ifenprodil and arcaine alter amygdala-kindling development. Eur J Pharmacol 1999;371:147–52. [346] Balosso S, Maroso M, Sanchez-Alavez M, et al. A novel non-transcriptional pathway mediates the proconvulsive effects of interleukin-1beta. Brain 2008;131:3256–65. [347] Kaasinen SK, Grohn OH, Keinanen TA, Alhonen L, Janne J. Overexpression of spermidine/spermine N1-acetyltransferase elevates the threshold to pentylenetetrazol-induced seizure activity in transgenic mice. Exp Neurol 2003;183:645–52. [348] Yen W, Williamson J, Bertram EH, Kapur J. A comparison of three NMDA receptor antagonists in the treatment of prolonged status epilepticus. Epilepsy Res 2004;59:43–50.
637
[349] Bandyopadhyay S, Hablitz JJ. NR2B antagonists restrict spatiotemporal spread of activity in a rat model of cortical dysplasia. Epilepsy Res 2006;72:127–39. [350] De Sarro G, Nava F, Calapai G, De Sarro A. Effects of some excitatory amino acid antagonists and drugs enhancing gamma-aminobutyric acid neurotransmission on pefloxacin-induced seizures in DBA/2 mice. Antimicrob Agents Chemother 1997;41:427–34. [351] De Sarro G, Ammendola D, Nava F, De Sarro A. Effects of some excitatory amino acid antagonists on imipenem-induced seizures in DBA/2 mice. Brain Res 1995;671:131–40. [352] Tsuda M, Suzuki T, Misawa M. Role of the NMDA receptor complex in DMCMinduced seizure in mice. Neuroreport 1997;8:603–6. [353] Burket JA, Mastropaolo J, Rosse RB, Katz EU, Deutsch SI. NMDA NR2B subtypeselective receptor antagonists fail to antagonize electrically-precipitated seizures and elicit popping in mice. Eur Neuropsychopharmacol 2010;20: 207–10. [354] Ebert U, Wlaz P, Loscher W. Anticonvulsant effects by combined treatment with a glycine B receptor antagonist and a polyamine site antagonist in amygdalakindled rats. Eur J Pharmacol 1997;322:179–84. [355] Patat A, Molinier P, Hergueta T, et al. Lack of amnestic, psychotomimetic or impairing effect on psychomotor performance of eliprodil, a new NMDA antagonist. Int Clin Psychopharmacol 1994;9:155–62. [356] Ikonomidou C, Turski L. Why did NMDA receptor antagonists fail clinical trials for stroke and traumatic brain injury? Lancet Neurol 2002;1:383–6. [357] Kotlinska J, Liljequist S. Oral administration of glycine and polyamine receptor antagonists blocks ethanol withdrawal seizures. Psychopharmacology (Berl) 1996;127:238–44. [358] Wlaz P, Ebert U, Loscher W. Anticonvulsant effects of eliprodil alone or combined with the glycine B receptor antagonist L-701,324 or the competitive NMDA antagonist CGP 40116 in the amygdala kindling model in rats. Neuropharmacology 1999;38:243–51. [359] Deren-Wesolek A, Maj J. Central effects of SL 82.0715, an antagonist of polyamine site of the NMDA receptor complex. Pol J Pharmacol 1993;45: 467–80. [360] Kanner AM. Depression and epilepsy: A bidirectional relation? Epilepsia 2011;52 (Suppl 1):21–7. [361] Kanner AM. Mood disorders in epilepsy: two different disorders with common pathogenic mechanisms? In: Schachter SC, Holmes GL, Trenité DGAK, editors. Behavioral aspects of epilepsy: principles and practice. New York: Demos Medical Publishing; 2008. p. 189–94. [362] Kanner AM. Depression in epilepsy: prevalence, clinical semiology, pathogenic mechanisms, and treatment. Biol Psychiatry 2003;54:388–98. [363] Kanner AM, Byrne R, Chicharro AV, et al. A lifetime history of depression is the sole predictor of post-surgical aura in seizure-free patients that underwent an antero-temporal lobectomy. Epilepsia 2006;44:198. [364] Forsgren L, Nystrom L. An incident case-referent study of epileptic seizures in adults. Epilepsy Res 1990;6:66–81. [365] Hesdorffer DC, Hauser WA, Annegers JF, Cascino G. Major depression is a risk factor for seizures in older adults. Ann Neurol 2000;47:246–9. [366] Hesdorffer DC, Hauser WA, Olafsson E, Ludvigsson P, Kjartansson O. Depression and suicide attempt as risk factors for incident unprovoked seizures. Ann Neurol 2006;59:35–41. [367] Hashimoto K. Emerging role of glutamate in the pathophysiology of major depressive disorder. Brain Res Rev 2009;61:105–23. [368] Skolnick P. Beyond monoamine-based therapies: clues to new approaches. J Clin Psychiatry 2002;63(Suppl 2):19–23. [369] Skolnick P, Popik P, Trullas R. Glutamate-based antidepressants: 20 years on. Trends Pharmacol Sci 2009;30:563–9. [370] Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 2000;47:351–4. [371] Zarate Jr CA, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-Daspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 2006;63:856–64. [372] Ferguson JM, Shingleton RN. An open-label, flexible-dose study of memantine in major depressive disorder. Clin Neuropharmacol 2007;30:136–44. [373] Dietrich DE, Bode L, Spannhuth CW, et al. Amantadine in depressive patients with Borna disease virus (BDV) infection: an open trial. Bipolar Disord 2000;2: 65–70. [374] Rogoz Z, Skuza G, Daniel WA, Wojcikowski J, Dudek D, Wrobel A. Amantadine as an additive treatment in patients suffering from drug-resistant unipolar depression. Pharmacol Rep 2007;59:778–84. [375] Preskorn SH, Baker B, Kolluri S, Menniti FS, Krams M, Landen JW. An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-D-aspartate antagonist, CP-101,606, in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol 2008;28:631–7. [376] Chen HS, Lipton SA. The chemical biology of clinically tolerated NMDA receptor antagonists. J Neurochem 2006;97:1611–26. [377] Farber NB. The NMDA receptor hypofunction model of psychosis. Ann N Y Acad Sci 2003;1003:119–30. [378] Muir KW, Lees KR. Clinical experience with excitatory amino acid antagonist drugs. Stroke 1995;26:503–13. [379] Gouzoulis-Mayfrank E, Heekeren K, Neukirch A, et al. Psychological effects of (S)ketamine and N, N-dimethyltryptamine (DMT): a double-blind, cross-over study in healthy volunteers. Pharmacopsychiatry 2005;38:301–11. [380] Johnston MV. Neurotransmitters and vulnerability of the developing brain. Brain Dev 1995;17:301–6.
638
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
[381] Berger P, Farrel K, Sharp F, Skolnick P. Drugs acting at the strychnine-insensitive glycine receptor do not induce HSP-70 protein in the cingulate cortex. Neurosci Lett 1994;168:147–50. [382] Danysz W, Parsons CG, Karcz-Kubicha M, et al. Glycine B antagonists as potential therapeutic agents. Previous hopes and present reality. Amino Acids 1998;14: 235–9. [383] Brock-Utne JG, Mankowitz E, Kallichurum S, Downing JW. Effects of intrathecal saline and ketamine with and without preservative on the spinal nerve roots of monkeys. S Afr Med J 1982;61:360–1. [384] Errando CL, Sifre C, Moliner S, et al. Subarachnoid ketamine in swine— pathological findings after repeated doses: acute toxicity study. Reg Anesth Pain Med 1999;24:146–52. [385] Vranken JH, Troost D, de Haan P, et al. Severe toxic damage to the rabbit spinal cord after intrathecal administration of preservative-free S(+)-ketamine. Anesthesiology 2006;105:813–8. [386] Kristensen JD, Post C, Gordh Jr T, Svensson BA. Spinal cord morphology and antinociception after chronic intrathecal administration of excitatory amino acid antagonists in the rat. Pain 1993;54:309–16. [387] Borgbjerg FM, Svensson BA, Frigast C, Gordh Jr T. Histopathology after repeated intrathecal injections of preservative-free ketamine in the rabbit: a light and electron microscopic examination. Anesth Analg 1994;79:105–11. [388] Malinovsky JM, Lepage JY, Cozian A, Mussini JM, Pinaudt M, Souron R. Is ketamine or its preservative responsible for neurotoxicity in the rabbit? Anesthesiology 1993;78:109–15. [389] Malinovsky JM, Cozian A, Lepage JY, Mussini JM, Pinaud M, Souron R. Ketamine and midazolam neurotoxicity in the rabbit. Anesthesiology 1991;75:91–7. [390] Karpinski N, Dunn J, Hansen L, Masliah E. Subpial vacuolar myelopathy after intrathecal ketamine: report of a case. Pain 1997;73:103–5. [391] Pouw B, Nour M, Matsumoto RR. Effects of AMPA/kainate glutamate receptor antagonists on cocaine-induced convulsions and lethality in mice. Eur J Pharmacol 1999;386:181–6. [392] Smith SE, al-Zubaidy ZA, Chapman AG, Meldrum BS. Excitatory amino acid antagonists, lamotrigine and BW 1003C87 as anticonvulsants in the genetically epilepsy-prone rat. Epilepsy Res 1993;15:101–11. [393] Peterson SL, Purvis RS, Griffith JW. Differential neuroprotective effects of the NMDA receptor-associated glycine site partial agonists 1-aminocyclopropanecarboxylic acid (ACPC) and D-cycloserine in lithium-pilocarpine status epilepticus. Neurotoxicology 2004;25:835–47. [394] Zapata A, Capdevila JL, Viu E, Trullas R. 1-Aminocyclopropane-carboxylic acid reduces NMDA-induced hippocampal neurodegeneration in vivo. Neuroreport 1996;7:397–400. [395] Jones AW, Croucher MJ, Meldrum BS, Watkins JC. Suppression of audiogenic seizures in DBA/2 mice by two new dipeptide NMDA receptor antagonists. Neurosci Lett 1984;45:157–61. [396] Czuczwar SJ, Janusz W, Wamil A, Kleinrok Z. Inhibition of aminophylline-induced convulsions in mice by antiepileptic drugs and other agents. Eur J Pharmacol 1987;144:309–15. [397] Morales-Villagran A, Urena-Guerrero ME, Tapia R. Protection by NMDA receptor antagonists against seizures induced by intracerebral administration of 4aminopyridine. Eur J Pharmacol 1996;305:87–93. [398] Mori N, Wada JA. Suppression of amygdaloid kindled convulsion following unilateral injection of 2-amino-7-phosphonoheptanoic acid (2-APH) into the substantia innominata of rats. Brain Res 1989;486:141–6. [399] Croucher MJ, Collins JF, Meldrum BS. Anticonvulsant action of excitatory amino acid antagonists. Science 1982;216:899–901. [400] Croucher MJ, Meldrum BS, Collins JF. Anticonvulsant and proconvulsant properties of a series of structural isomers of piperidine dicarboxylic acid. Neuropharmacology 1984;23:467–72. [401] Faingold CL, Millan MH, Boersma CA, Meldrum BS. Excitant amino acids and audiogenic seizures in the genetically epilepsy-prone rat. I. Afferent seizure initiation pathway. Exp Neurol 1988;99:678–86. [402] De Sarro G, De Sarro A. Anticonvulsant activity of competitive antagonists of NMDA receptor in genetically epilepsy-prone rats. Eur J Pharmacol 1992;215: 221–9. [403] McCaslin PP, Morgan WW. 2-Amino-7-phosphonoheptanoic acid, a selective antagonist of N-methyl-D-aspartate, prevents barbital withdrawal-induced convulsions and the elevation of cerebellar cyclic GMP in dependent rats. Neuropharmacology 1987;26:731–5. [404] Croucher MJ, Bradford HF, Sunter DC, Watkins JC. Inhibition of the development of electrical kindling of the prepyriform cortex by daily focal injections of excitatory amino acid antagonists. Eur J Pharmacol 1988;152:29–38. [405] Veliskova J, Liptakova S, Hussain S. The effects of N-methyl-D-aspartate antagonist 2-amino-7-phosphonoheptanoic acid microinfusions into the adult male rat substantia nigra pars reticulata are site-specific. Neurosci Lett 2001;316: 108–10. [406] Holmes GL, Thompson JL, Carl GF, Gallagher BS, Hoy J, McLaughlin M. Effect of 2amino-7-phosphonoheptanoic acid (APH) on seizure susceptibility in the prepubescent and mature rat. Epilepsy Res 1990;5:125–30. [407] Folbergrova J. Anticonvulsant action of both NMDA and non-NMDA receptor antagonists against seizures induced by homocysteine in immature rats. Exp Neurol 1997;145:442–50. [408] Chapman AG, Engelsen B, Meldrum BS. 2-Amino-7-phosphonoheptanoic acid inhibits insulin-induced convulsions and striatal aspartate accumulation in rats with frontal cortical ablation. J Neurochem 1987;49:121–7. [409] Peterson DW, Collins JF, Bradford HF. Anticonvulsant action of amino acid antagonists against kindled hippocampal seizures. Brain Res 1984;311:176–80.
[410] De Sarro G, Meldrum BS, Reavill C. Anticonvulsant action of 2-amino-7phosphonoheptanoic acid in the substantia nigra. Eur J Pharmacol 1984;106: 175–9. [411] Czuczwar SJ, Cavalheiro EA, Turski L, Turski WA, Kleinrok Z. Phosphonic analogues of excitatory amino acids raise the threshold for maximal electroconvulsions in mice. Neurosci Res 1985;3:86–90. [412] Susic V, Hivanovic D, Stanojlovic O. Inhibition of metaphit-induced audiogenic seizures by 2-amino-7-phosphonoheptanoic acid. Glas Srp Akad Nauka Med 2002:103–10. [413] Zivanovic D, Susic V, Stanojlovic O. The anticonvulsant potency of (+/−)-2Amino-7-phosphonoheptanoic acid and (+/−)-2-amino-5-phosphonovaleric acid against metaphit-induced audiogenic seizures in rats. Pharmacol Res 1998;38:347–51. [414] Zivanovic D, Susic V, Stanojlovic O. APH, an N-methyl-D-aspartate receptor antagonist, blocks the metaphit-induced audiogenic seizures in rats. J Physiol Paris 1997;91:317–23. [415] Millan MH, Patel S, Mello LM, Meldrum BS. Focal injection of 2-amino-7phosphonoheptanoic acid into prepiriform cortex protects against pilocarpineinduced limbic seizures in rats. Neurosci Lett 1986;70:69–74. [416] Meldrum BS, Croucher MJ, Badman G, Collins JF. Antiepileptic action of excitatory amino acid antagonists in the photosensitive baboon, Papio papio. Neurosci Lett 1983;39:101–4. [417] Schwarcz R, Brush GS, Foster AC, French ED. Seizure activity and lesions after intrahippocampal quinolinic acid injection. Exp Neurol 1984;84:1–17. [418] Czuczwar SJ, Meldrum B. Protection against chemically induced seizures by 2amino-7-phosphonoheptanoic acid. Eur J Pharmacol 1982;83:335–8. [419] Aldinio C, French ED, Schwarcz R. The effects of intrahippocampal ibotenic acid and their blockade by (−) 2-amino-7-phosphonoheptanoic acid: morphological and electroencephalographical analysis. Exp Brain Res 1983;51:36–44. [420] Czuczwar SJ, Frey HH, Loscher W. Antagonism of N-methyl-D, L-aspartic acidinduced convulsions by antiepileptic drugs and other agents. Eur J Pharmacol 1985;108:273–80. [421] Piredda S, Gale K. Anticonvulsant action of 2-amino-7-phosphonoheptanoic acid and muscimol in the deep prepiriform cortex. Eur J Pharmacol 1986;120: 115–8. [422] Urwyler S, Campbell E, Fricker G, et al. Biphenyl-derivatives of 2-amino-7phosphono-heptanoic acid, a novel class of potent competitive N-methyl-Daspartate receptor antagonists—II. Pharmacological characterization in vivo. Neuropharmacology 1996;35:655–69. [423] Holmes KH, Bilkey DK, Laverty R. The infusion of an NMDA antagonist into perirhinal cortex suppresses amygdala-kindled seizures. Brain Res 1992;587: 285–90. [424] Chiamulera C, Costa S, Reggiani A. Effect of NMDA- and strychnine-insensitive glycine site antagonists on NMDA-mediated convulsions and learning. Psychopharmacology (Berl) 1990;102:551–2. [425] Sierra-Paredes G, Sierra-Marcuno G. Effects of NMDA antagonists on seizure thresholds induced by intrahippocampal microdialysis of picrotoxin in freely moving rats. Neurosci Lett 1996;218:62–6. [426] Loscher W, Honack D. Anticonvulsant and behavioral effects of two novel competitive N-methyl-D-aspartic acid receptor antagonists, CGP 37849 and CGP 39551, in the kindling model of epilepsy. Comparison with MK-801 and carbamazepine. J Pharmacol Exp Ther 1991;256:432–40. [427] Cotterell KL, Croucher MJ, Bradford HF. Weak anticonvulsant activity of CGP 37849 and CGP 39551 against kindled seizures following systemic administration. Eur J Pharmacol 1992;214:285–7. [428] Chapman AG, Graham JL, Patel S, Meldrum BS. Anticonvulsant activity of two orally active competitive N-methyl-D-aspartate antagonists, CGP 37849 and CGP 39551, against sound-induced seizures in DBA/2 mice and photically induced myoclonus in Papio papio. Epilepsia 1991;32:578–87. [429] Schmutz M, Portet C, Jeker A, et al. The competitive NMDA receptor antagonists CGP 37849 and CGP 39551 are potent, orally-active anticonvulsants in rodents. Naunyn Schmiedebergs Arch Pharmacol 1990;342:61–6. [430] Czechowska G, Dziki M, Pietrasiewicz T, Kleinrok Z, Turski WA, Czuczwar SJ. Competitive antagonists of NMDA receptors, CGP 37849 and CGP 39551, enhance the anticonvulsant activity of valproate against electroconvulsions in mice. Eur J Pharmacol 1993;232:59–64. [431] Fagg GE, Olpe HR, Pozza MF, et al. CGP 37849 and CGP 39551: novel and potent competitive N-methyl-D-aspartate receptor antagonists with oral activity. Br J Pharmacol 1990;99:791–7. [432] Liljequist S. The competitive NMDA receptor antagonist, CGP 39551, inhibits ethanol withdrawal seizures. Eur J Pharmacol 1991;192:197–8. [433] Ripley TL, Dunworth SJ, Stephens DN. Effect of CGP39551 administration on the kindling of ethanol-withdrawal seizures. Psychopharmacology (Berl) 2002;163: 157–65. [434] Ripley TL, Little HJ. Effects on ethanol withdrawal hyperexcitability of chronic treatment with a competitive N-methyl-D-aspartate receptor antagonist. J Pharmacol Exp Ther 1995;272:112–8. [435] Velisek L, Vachova D, Mares P. Excitatory amino acid antagonists and pentylenetetrazol-induced seizures during ontogenesis. IV. Effects of CGP 39551. Pharmacol Biochem Behav 1997;56:493–8. [436] Rejdak K, Rejdak R, Czuczwar SJ, Kleinrok Z, Sieklucka-Dziuba M. The influence of CGP-40116 on bicuculline evoked seizures in mice exposed to transient episode of brain oligemia. J Neural Transm 2000;107:1263–72. [437] Rejdak K, Rejdak R, Stelmasiak Z, Czuczwar SJ, Kleinrok Z, Sieklucka-Dziuba M. The effect of CGP-40116 on pilocarpine evoked seizures in mice exposed to transient episode of brain ischemia. Epilepsy Res 2000;41:213–22.
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640 [438] Fujikawa DG, Daniels AH, Kim JS. The competitive NMDA receptor antagonist CGP 40116 protects against status epilepticus-induced neuronal damage. Epilepsy Res 1994;17:207–19. [439] Urbanska EM, Tomczyk T, Haberek G, et al. NMDA- but not kainate-mediated events reduce efficacy of some antiepileptic drugs against generalized tonic–clonic seizures in mice. Epilepsia 1999;40:1507–11. [440] Starr MS, Starr BS. Glutamate–dopamine interactions in the production of pilocarpine motor seizures in the mouse. J Neural Transm Park Dis Dement Sect 1993;6:109–17. [441] Haugvicova R, Mares P. Anticonvulsant action of a NMDA receptor antagonist CGP 40116 varies only quantitatively during ontogeny in rats. Fundam Clin Pharmacol 1998;12:521–5. [442] Gasior M, Borowicz K, Starownik R, Kleinrok Z, Czuczwar SJ. Ca2+ channel blockade and the antielectroshock activity of NMDA receptor antagonists, CGP 40116 and CGP 43487, in mice. Eur J Pharmacol 1996;312:27–33. [443] Koerner C, Danober L, Boehrer A, Marescaux C, Vergnes M. Thalamic NMDA transmission in a genetic model of absence epilepsy in rats. Epilepsy Res 1996;25:11–9. [444] Banerjee PK, Snead III OC. Involvement of excitatory amino acid mechanisms in gamma-hydroxybutyrate model of generalized absence seizures in rats. Neuropharmacology 1992;31:1009–19. [445] Banerjee PK, Snead III OC. Thalamic NMDA receptors in the gammahydroxybutyrate model of absence seizures: a cerebral microinjection study in rats. Neuropharmacology 1995;34:43–53. [446] Morimoto K, Katayama K, Inoue K, Sato K. Effects of competitive and noncompetitive NMDA receptor antagonists on kindling and LTP. Pharmacol Biochem Behav 1991;40:893–9. [447] Lehmann J, Chapman AG, Meldrum BS, Hutchison A, Tsai C, Wood PL. CGS 19755 is a potent and competitive antagonist at NMDA-type receptors. Eur J Pharmacol 1988;154:89–93. [448] Bennett DA, Bernard PS, Amrick CL, Wilson DE, Liebman JM, Hutchison AJ. Behavioral pharmacological profile of CGS 19755, a competitive antagonist at Nmethyl-D-aspartate receptors. J Pharmacol Exp Ther 1989;250:454–60. [449] Katsumori H, Minabe Y, Osawa M, Ashby Jr CR. Acute effects of various GABA receptor agonists and glutamate antagonists on focal hippocampal seizures in freely moving rats elicited by low-frequency stimulation. Synapse 1998;28: 103–9. [450] McFarlane C, Warner DS, Dexter F, Todd MM. Glutamatergic antagonism: effects on lidocaine-induced seizures in the rat. Anesth Analg 1994;79:701–5. [451] Lehmann J, Hutchison AJ, McPherson SE, et al. CGS 19755, a selective and competitive N-methyl-D-aspartate-type excitatory amino acid receptor antagonist. J Pharmacol Exp Ther 1988;246:65–75. [452] Koek W, Colpaert FC. Selective blockade of N-methyl-D-aspartate (NMDA)induced convulsions by NMDA antagonists and putative glycine antagonists: relationship with phencyclidine-like behavioral effects. J Pharmacol Exp Ther 1990;252:349–57. [453] Croucher MJ, Bradford HF. 7-Chlorokynurenic acid, a strychnine-insensitive glycine receptor antagonist, inhibits limbic seizure kindling. Neurosci Lett 1990;118:29–32. [454] Namba T, Morimoto K, Yamada N, Otsuki S. Antiepileptogenic action of 7chlorokynurenic acid on amygdala kindling of rats. Pharmacol Biochem Behav 1993;46:275–81. [455] Hu RQ, Davies JA. Glutamate receptor antagonists reduce spontaneous epileptiform activity in cortical wedges prepared from DBA/2 mice. Exp Brain Res 1997;115:311–8. [456] Singh L, Oles RJ, Tricklebank MD. Modulation of seizure susceptibility in the mouse by the strychnine-insensitive glycine recognition site of the NMDA receptor/ion channel complex. Br J Pharmacol 1990;99:285–8. [457] Maj J, Rogoz Z, Skuza G, Kolodziejczyk K. Some central effects of kynurenic acid, 7-chlorokynurenic acid and 5,7-dichloro-kynurenic acid, glycine site antagonists. Pol J Pharmacol 1994;46:115–24. [458] Peterson SL. Anticonvulsant drug potentiation by glycine in maximal electroshock seizures is mimicked by D-serine and antagonized by 7-chlorokynurenic acid. Eur J Pharmacol 1991;199:341–8. [459] Tsuda M, Suzuki T, Misawa M. Recovery of decreased seizure threshold for pentylenetetrazole during diazepam withdrawal by NMDA receptor antagonists. Eur J Pharmacol 1997;324:63–6. [460] Wardas J, Graham J, Gale K. Evidence for a role of glycine in area tempestas for triggering convulsive seizures. Eur J Pharmacol 1990;187:59–66. [461] Yamaguchi S, Rogawski MA. Effects of anticonvulsant drugs on 4-aminopyridineinduced seizures in mice. Epilepsy Res 1992;11:9–16. [462] Rockhold RW, Oden G, Ho IK, Andrew M, Farley JM. Glutamate receptor antagonists block cocaine-induced convulsions and death. Brain Res Bull 1991;27:721–3. [463] Nevins ME, Arnolde SM. A comparison of the anticonvulsant effects of competitive and non-competitive antagonists of the N-methyl-D-aspartate receptor. Brain Res 1989;503:1–4. [464] Mazarati AM, Wasterlain CG. N-methyl-D-asparate receptor antagonists abolish the maintenance phase of self-sustaining status epilepticus in rat. Neurosci Lett 1999;265:187–90. [465] Lallement G, Clarencon D, Galonnier M, Baubichon D, Burckhart MF, Peoc'h M. Acute soman poisoning in primates neither pretreated nor receiving immediate therapy: value of gacyclidine (GK-11) in delayed medical support. Arch Toxicol 1999;73:115–22. [466] Lallement G, Clarencon D, Masqueliez C, et al. Nerve agent poisoning in primates: antilethal, anti-epileptic and neuroprotective effects of GK-11. Arch Toxicol 1998;72:84–92.
639
[467] Croucher MJ, Bradford HF. The influence of strychnine-insensitive glycine receptor agonists and antagonists on generalized seizure thresholds. Brain Res 1991;543:91–6. [468] Zivanovic D, Lovic OS, Susic V. Effects of manipulation of N-methyl-D-aspartate receptors on imipenem/cilastatin-induced seizures in rats. Indian J Med Res 2004;119:79–85. [469] Vartanian MG, Taylor CP. Different stereoselectivity of the enantiomers of HA966 (3-amino-1-hydroxy-2-pyrrolidinone) for neuroprotective and anticonvulsant actions in vivo. Neurosci Lett 1991;133:109–12. [470] Leal MB, de Souza DO, Elisabetsky E. Long-lasting ibogaine protection against NMDA-induced convulsions in mice. Neurochem Res 2000;25:1083–7. [471] Wlaz P, Loscher W. Weak anticonvulsant effects of two novel glycine B receptor antagonists in the amygdala-kindling model in rats. Eur J Pharmacol 1998;342: 39–46. [472] Potschka H, Loscher W, Wlaz P, et al. LU 73068, a new non-NMDA and glycine/ NMDA receptor antagonist: pharmacological characterization and comparison with NBQX and L-701,324 in the kindling model of epilepsy. Br J Pharmacol 1998;125: 1258–66. [473] Bristow LJ, Hutson PH, Kulagowski JJ, et al. Anticonvulsant and behavioral profile of L-701,324, a potent, orally active antagonist at the glycine modulatory site on the N-methyl-D-aspartate receptor complex. J Pharmacol Exp Ther 1996;279: 492–501. [474] Sheardown MJ, Drejer J, Jensen LH, Stidsen CE, Honore T. A potent antagonist of the strychnine insensitive glycine receptor has anticonvulsant properties. Eur J Pharmacol 1989;174:197–204. [475] Kotlinska J, Biala G, Rafalski P, Bochenski M, Danysz W. Effect of neramexane on ethanol dependence and reinforcement. Eur J Pharmacol 2004;503:95–8. [476] Klockgether T, Turski L, Schwarz M, Sontag KH, Lehmann J. Paradoxical convulsant action of a novel non-competitive N-methyl-D-aspartate (NMDA) antagonist, tiletamine. Brain Res 1988;461:343–8. [477] Mattson MP, Kater SB. Excitatory and inhibitory neurotransmitters in the generation and degeneration of hippocampal neuroarchitecture. Brain Res 1989;478:337–48. [478] Lampe H, Bigalke H. Carbamazepine blocks NMDA-activated currents in cultured spinal cord neurons. Neuroreport 1990;1:26–8. [479] Cai Z, McCaslin PP. Amitriptyline, desipramine, cyproheptadine and carbamazepine, in concentrations used therapeutically, reduce kainate- and N-methyl-D-aspartateinduced intracellular Ca2+ levels in neuronal culture. Eur J Pharmacol 1992;219: 53–7. [480] Hough CJ, Irwin RP, Gao XM, Rogawski MA, Chuang DM. Carbamazepine inhibition of N-methyl-D-aspartate-evoked calcium influx in rat cerebellar granule cells. J Pharmacol Exp Ther 1996;276:143–9. [481] Lancaster JM, Davies JA. Carbamazepine inhibits NMDA-induced depolarizations in cortical wedges prepared from DBA/2 mice. Experientia 1992;48:751–3. [482] Walden J, Grunze H, Bingmann D, Liu Z, Dusing R. Calcium antagonistic effects of carbamazepine as a mechanism of action in neuropsychiatric disorders: studies in calcium dependent model epilepsies. Eur Neuropsychopharmacol 1992;2:455–62. [483] Kubota T, Jibiki I, Fukushima T, Kurokawa K, Yamaguchi N. Carbamazepine-induced blockade of induction of long-term potentiation in the perforant path-dentate gyrus pathway in chronically prepared rabbits. Neurosci Lett 1994;170:171–4. [484] Gean PW, Huang CC, Kuo JR, Lin JH, Yi PL, Tsai JJ. Analysis of carbamazepine's anticonvulsant actions in hippocampal and amygdaloid slices of the rat. Chin J Physiol 1993;36:199–204. [485] Wang SJ, Huang CC, Hsu KS, Tsai JJ, Gean PW. Presynaptic inhibition of excitatory neurotransmission by lamotrigine in the rat amygdalar neurons. Synapse 1996;24:248–55. [486] Phillips I, Martin KF, Thompson KS, Heal DJ. Weak blockade of AMPA receptormediated depolarisations in the rat cortical wedge by phenytoin but not lamotrigine or carbamazepine. Eur J Pharmacol 1997;337:189–95. [487] Ambrosio AF, Silva AP, Malva JO, Soares-da-Silva P, Carvalho AP, Carvalho CM. Carbamazepine inhibits L-type Ca2+ channels in cultured rat hippocampal neurons stimulated with glutamate receptor agonists. Neuropharmacology 1999;38:1349–59. [488] Cunha RA, Coelho JE, Costenla AR, et al. Effects of carbamazepine and novel 10,11-dihydro-5H-dibenz[b, f]azepine-5-carboxamide derivatives on synaptic transmission in rat hippocampal slices. Pharmacol Toxicol 2002;90:208–13. [489] Bown CD, Wang JF, Young LT. Attenuation of N-methyl-D-aspartate-mediated cytoplasmic vacuolization in primary rat hippocampal neurons by mood stabilizers. Neuroscience 2003;117:949–55. [490] Basselin M, Villacreses NE, Chen M, Bell JM, Rapoport SI. Chronic carbamazepine administration reduces N-methyl-D-aspartate receptor-initiated signaling via arachidonic acid in rat brain. Biol Psychiatry 2007;62:934–43. [491] Afanas'ev I, Kudrin V, Rayevsky KS, Varga V, Saransaari P, Oja SS. Lamotrigine and carbamazepine affect differently the release of D-[3H]aspartate from mouse cerebral cortex slices: involvement of NO. Neurochem Res 1999;24: 1153–9. [492] Farber NB, Jiang XP, Heinkel C, Nemmers B. Antiepileptic drugs and agents that inhibit voltage-gated sodium channels prevent NMDA antagonist neurotoxicity. Mol Psychiatry 2002;7:726–33. [493] Psarropoulou C, Haas HL. Action of anticonvulsants on hippocampal slices in Mgfree medium. Naunyn Schmiedebergs Arch Pharmacol 1989;339:613–6. [494] Rock DM, Kelly KM, Macdonald RL. Gabapentin actions on ligand- and voltagegated responses in cultured rodent neurons. Epilepsy Res 1993;16:89–98. [495] Yang J, Wetterstrand C, Jones RS. Felbamate but not phenytoin or gabapentin reduces glutamate release by blocking presynaptic NMDA receptors in the entorhinal cortex. Epilepsy Res 2007;77:157–64.
640
M. Ghasemi, S.C. Schachter / Epilepsy & Behavior 22 (2011) 617–640
[496] Shimoyama M, Shimoyama N, Hori Y. Gabapentin affects glutamatergic excitatory neurotransmission in the rat dorsal horn. Pain 2000;85:405–14. [497] Suarez LM, Suarez F, Del Olmo N, Ruiz M, Gonzalez-Escalada JR, Solis JM. Presynaptic NMDA autoreceptors facilitate axon excitability: a new molecular target for the anticonvulsant gabapentin. Eur J Neurosci 2005;21:197–209. [498] Hara K, Sata T. Inhibitory effect of gabapentin on N-methyl-D-aspartate receptors expressed in Xenopus oocytes. Acta Anaesthesiol Scand 2007;51:122–8. [499] Gu Y, Huang LY. Gabapentin actions on N-methyl-D-aspartate receptor channels are protein kinase C-dependent. Pain 2001;93:85–92. [500] McGeer EG, Zhu SG. Lamotrigine protects against kainate but not ibotenate lesions in rat striatum. Neurosci Lett 1990;112:348–51. [501] Leach MJ, Baxter MG, Critchley MAE. Neurochemical and behavioral aspects of lamotrigine. Epilepsia 1991;32:S4–8. [502] Lizasoain I, Knowles RG, Moncada S. Inhibition by lamotrigine of the generation of nitric oxide in rat forebrain slices. J Neurochem 1995;64:636–42. [503] Lee CY, Fu WM, Chen CC, Su MJ, Liou HH. Lamotrigine inhibits postsynaptic AMPA receptor and glutamate release in the dentate gyrus. Epilepsia 2008;49: 888–97. [504] Afanas'ev II, Kudrin VS, Varga V, Saransaari P, Oja SS, Raevskii KS. The effect of sodium nitroprusside and lamotrigine on D-[3H]aspartate release from mouse cortex slices. Eksp Klin Farmakol 1998;61:9–12. [505] Birnstiel S, Wulfert E, Beck SG. Levetiracetam (ucb LO59) affects in vitro models of epilepsy in CA3 pyramidal neurons without altering normal synaptic transmission. Naunyn Schmiedebergs Arch Pharmacol 1997;356:611–8. [506] Lee CY, Chen CC, Liou HH. Levetiracetam inhibits glutamate transmission through presynaptic P/Q-type calcium channels on the granule cells of the dentate gyrus. Br J Pharmacol 2009;158:1753–62. [507] Gandolfi O, Voltattorni M, Gaggi R, Dall'Olio R. Repeated administration of the novel antipsychotic olanzapine does not modulate NMDA-sensitive glutamate and 5HT2 serotonin receptors in rats. Naunyn Schmiedebergs Arch Pharmacol 2003;367:615–20. [508] Ninan I, Jardemark KE, Wang RY. Olanzapine and clozapine but not haloperidol reverse subchronic phencyclidine-induced functional hyperactivity of N-methylD-aspartate receptors in pyramidal cells of the rat medial prefrontal cortex. Neuropharmacology 2003;44:462–72. [509] Tarazi FI, Baldessarini RJ, Kula NS, Zhang K. Long-term effects of olanzapine, risperidone, and quetiapine on ionotropic glutamate receptor types: implications for antipsychotic drug treatment. J Pharmacol Exp Ther 2003;306:1145–51.
[510] Gardoni F, Frasca A, Zianni E, Riva MA, Di Luca M, Fumagalli F. Repeated treatment with haloperidol, but not olanzapine, alters synaptic NMDA receptor composition in rat striatum. Eur Neuropsychopharmacol 2008;18:531–4. [511] Das A, McDowell M, O'Dell CM, et al. Post-treatment with voltage-gated Na(+) channel blocker attenuates kainic acid-induced apoptosis in rat primary hippocampal neurons. Neurochem Res 2010;35:2175–83. [512] Boehm FH, Liem LK, Stanton PK, Potter PE, Moskal JR. Phenytoin protects against hypoxia-induced death of cultured hippocampal neurons. Neurosci Lett 1994;175:171–4. [513] Laffling AJ, Scherr P, McGivern JG, Patmore L, Sheridan RD. Electrophysiological actions of phenytoin on N-methyl-D-aspartate receptor-mediated responses in rat hippocampus in vitro. Br J Pharmacol 1995;115:67–72. [514] Lee GY, Brown LM, Teyler TJ. The effects of anticonvulsant drugs on long-term potentiation (LTP) in the rat hippocampus. Brain Res Bull 1996;39:39–42. [515] McCabe RT, Sofia RD, Layer RT, et al. Felbamate increases [3H]glycine binding in rat brain and sections of human postmortem brain. J Pharmacol Exp Ther 1998;286:991–9. [516] Holz RW. Pharmacology meets vesicular trafficking at a central nervous system synapse: pregabalin effects on synaptic vesicle cycling in hippocampal neurons. Mol Pharmacol 2006;70:444–6. [517] Micheva KD, Taylor CP, Smith SJ. Pregabalin reduces the release of synaptic vesicles from cultured hippocampal neurons. Mol Pharmacol 2006;70: 467–76. [518] Raffa RB, Finno KE, Tallarida CS, Rawls SM. Topiramate-antagonism of Lglutamate-induced paroxysms in planarians. Eur J Pharmacol 2010;649: 150–3. [519] Rawls SM, Thomas T, Adeola M, et al. Topiramate antagonizes NMDA- and AMPAinduced seizure-like activity in planarians. Pharmacol Biochem Behav 2009;93: 363–7. [520] Basselin M, Chang L, Chen M, Bell JM, Rapoport SI. Chronic administration of valproic acid reduces brain NMDA signaling via arachidonic acid in unanesthetized rats. Neurochem Res 2008;33:2229–40. [521] Rinaldi T, Kulangara K, Antoniello K, Markram H. Elevated NMDA receptor levels and enhanced postsynaptic long-term potentiation induced by prenatal exposure to valproic acid. Proc Natl Acad Sci U S A 2007;104:13501–6. [522] Roullet FI, Wollaston L, Decatanzaro D, Foster JA. Behavioral and molecular changes in the mouse in response to prenatal exposure to the anti-epileptic drug valproic acid. Neuroscience 2010;170:514–22.