Newer Antiepileptic Drugs



Newer Antiepileptic Drugs


Norman Delanty

Jacqueline A. French



Despite advances and optimal current therapy, there remains a continuing need for the development of new agents for patients with epilepsy (1, 2, 3, 4). Among those with epilepsy, 30% to 40% continue to have seizures or experience unacceptable side effects that affect their quality of life (5,6). In a prospective study of 525 patients in a single epilepsy center between 1984 and 1997, only 63% remained seizure free for more than 1 year, with seizure-free rates being similar, regardless of whether a new or an established antiepileptic drug (AED) was used (7). Moreover, the available anticonvulsant agents suppress the symptoms of epilepsy and are not truly antiepileptic or antiepileptogenic in nature. None of these agents have been shown to influence the process of epileptogenesis in humans or to alter the underlying brain dysfunction that expresses itself as epilepsy. An ideal agent would provide complete seizure control without significant side effects or idiosyncratic life-threatening reactions; have simple, predictable pharmacokinetics; be unaffected by other drugs or medical conditions; and be nonteratogenic, affordable, and available in a parenteral formulation. An agent that prevents epilepsy (e.g., after a head injury or stroke) or that alters the underlying mechanisms of a particular epilepsy, or prevents or ameliorates its progression (8), could also be considered to be antiepileptogenic or antiepileptic.

Although nine new agents have been introduced in recent years, with attendant marketing considerations, many novel compounds with promise as useful AEDs are currently in various stages of development (Table 68.1 and Table 68.2). Some of these resulted from the Antiepileptic Drug Development (ADD) Program sponsored by the U. S. National Institutes of Health (9), which has screened more than 24,000 compounds (provided by industry and academia) for potential anticonvulsant efficacy in traditional animal models (9,10). These models have traditionally focused mainly on the maximum electroshock (MES) test and the pentylenetetrazol (PTZ) test, which are believed to predict efficacy against tonic-clonic and absence seizures, respectively (see Chapter 44). Although this approach has identified such agents as topiramate, it does not always recognize potentially useful compounds, predict activity in humans, or test antiepileptogenic potential (11). Newer models, such as pilocarpine, kainate, or electrically induced post-status epilepsy models, are aimed at mimicking human disease and may be better suited to identify useful compounds, but are not effective for high-throughput screening of new chemical entities. Research elucidating the molecular mechanisms underlying some specific epilepsy syndromes, such as benign neonatal convulsions (12) and Unverricht-Lundborg progressive myoclonic epilepsy (13), suggests that targeted therapeutic approaches may prove more successful than mass screening techniques for some of the epilepsies. This may also be true for some of the more common forms of epilepsy, such as juvenile myoclonic epilepsy (14). Despite the limitations of screening methods, promising compounds are in development (15, 16, 17, 18). Some are at a late stage of development (e.g., pregabalin), whereas others are at earlier stages of clinical testing (e.g., safinamide, talampanel). However, not all of these compounds will be approved for use, which is exemplified by the fact that three of the compounds discussed in the prior version of this chapter are no longer in development.

In the previous edition of this textbook, this Chapter discussed 10 anticonvulsant drugs (19). Information on harkoseride, retigabine, rufinamide, and the valproate derivatives has been updated. Zonisamide and levetiracetam, now available, are discussed in Chapters 62 and 63. Pregabalin is discussed along with gabapentin in Chapter 59. Losigamone, remacemide, and soretolide are no longer being developed and are not discussed in this chapter.









TABLE 68.1 CHEMISTRY AND POSSIBLE MECHANISM OF ACTION OF SOME NEW ANTIEPILEPTIC DRUGS









































Agent


Chemistry


Possible Mechanism of Action


Carabersat


Fluorobenzoylamino benzopyran


Unknown; may have unique binding site


Fluorofelbamate


2-fluoro-2-phenyl-1,3-propanediol dicarbamate


Binds to strychnine-insensitive glycine site of NMDA receptor


Harkoseride


Trifunctional amino acid


Unknown; affinity for strychnine-insensitive glycine site of NMDA receptor; may act on novel binding site


Retigabine


Carbamic acid ethyl ester


KCNQ2/Q3 K+ channel opener; GABA potentiation


Rufinamide


Difluorobenzyl-triazole amide


Unknown; Na+ channel blocker; limits frequency of Na+-dependent action potentials in neurons


Safinamide


[(S)-(+)-2-(4-(3-fluorobenzyloxy) benzylamino) propanamide, methanesulfonate]


Na+ and Ca++ channel blocker


Talampanel


[(R)-7-acetyl-5-(4-aminophenyl)-8,9-dihydro-8-methyl-7H-1,3-dioxolo(4,5-h) (2,3) benzodiazepine]


AMPA-receptor blocker


Valrocemide


N-valproyl glycinamide


Valproate derivative


NMDA, N-methyl-D-aspartate; GABA, γ-aminobutyric acid; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid.



CARABERSAT

Carabersat is a structurally novel fluorobenzoylamino benzopyran with anticonvulsant efficacy in in vitro (elevated K+ rat hippocampal slice) and in vivo (MES and PTZ) models, with a greater potency than that of either carbamazepine or lamotrigine (20). The agent also has effects on the development of amygdala-kindled seizures in rats and thus may have antiepileptogenic effects. The mechanism of action of carabersat appears to be novel, depends on stereoselective activity at a unique binding site (18), and does not depend on ion channel, γ-aminobutyric acid (GABA) receptor, or glutamate modulation. In normal volunteers (136 individuals), the agent is well tolerated, reaches peak serum levels within a few hours, and has a half-life of 24 hours. Bioavailability is increased by the addition of food. In a phase 2, add-on, placebo-controlled study of 305 patients with refractory partial epilepsy, a 20% to 30% decrease in seizure frequency was reported in the high-dose carabersat (1200 mg per day) group compared with the placebo group, and the agent was well tolerated. Further studies are planned (20).








TABLE 68.2 IMPORTANT PHARMACOKINETIC PARAMETERS OF SELECTED NEW ANTIEPILEPTIC DRUGS





















































Agent


Tmax (h)


Protein Binding (%)


Half-Life (h)


Metabolism


Carabersat


2-4


NA


24


Hepatic


Harkoseride


2-3


<10


12


80% renal


Retigabine


1-2


NA


8-10


Hepatic


Rufinamide


5-6


30-40


7-10


85% renal


Safinamide


1-2


NA


24


NA


Talampanel


2


65-85


7


Hepatic


Valrocemide


NA


25


6-10


5% renal to VPA


Tmax, time to maximum serum concentration; NA, not available; VPA, valproic acid.




FLUOROFELBAMATE

The potential idiosyncratic life-threatening toxicity of felbamate has relegated this drug to use as a treatment of last resort in occasional refractory patients. Its adverse hematologic and hepatic effects are believed to be caused by formation of a reactive aldehyde intermediate metabolite (atropaldehyde or 2-phenylpropanol). To circumvent this, fluorofelbamate (2-fluoro-2-phenyl-1,3-propanediol dicarbamate), which does not have a reactive intermediate, has been developed and is currently undergoing investigation (18). A glutathione-aldehyde adduct found in the urine of rats and humans treated with felbamate has not been found in rats treated with fluorofelbamate. The mechanism of action of fluorofelbamate is likely to be similar to that of its analogue—i.e., modulation of the N-methyl-D-aspartate (NMDA) receptor by binding to its strychnine-insensitive glycine binding site. The anticonvulsant efficacy of fluorofelbamate in animal screening models appears to be at least as potent as that of felbamate. In the rat model of self-sustaining status epilepticus (SSSE) induced by 30-minute perforant path stimulation, intravenous (IV) fluorofelbamate reduced the frequency and severity of seizures, did not cause any behavioral toxicity, and protected against the evolution to chronic epilepsy (21). Fluorofelbamate is currently in phase 2 development. If the agent fulfills its promise as a safe derivative of felbamate, an IV formulation would be a welcome addition to the AED armamentarium.


HARKOSERIDE (SPM 927)

Harkoseride (SPM 927) is a novel trifunctional amino acid derivative, [R]-2-acetamido-N-benzyl-3-methoxypropionamide. The agent has potent anticonvulsant activity in several animal models of seizures, including MES and kindling paradigms (22). Harkoseride is very effective in validated models of status epilepticus, such as the cobalt homocysteine-induced generalized tonic-clonic seizure model, in which it appears to be synergistic with benzodiazepines; it also inhibits seizures in the perforant pathway model of SSSE. In addition, harkoseride is neuroprotective in several animal models of global and focal cerebral ischemia. The agent is being developed as an oral and IV treatment for patients with epilepsy and for those with neuropathic pain. Its precise mechanism of action is unclear, although harkoseride does show affinity for the strychnine-insensitive glycine site of the NMDA receptor complex, and it may also bind to an unidentified unique receptor in the brain. Radioligand binding studies of more than 100 central nervous system (CNS) proteins have revealed no significant displacement. Harkoseride is pH neutral and has good lipid solubility.

In phase 1 human studies (250 individuals), oral and IV administration caused only transient headache, fatigue, and light-headedness. Following oral administration, harkoseride exhibits near-complete absorption, which is not affected by food. The half-life of harkoseride is approximately 12 hours. The agent has linear pharmacokinetics and minimal protein binding; approximately 95% of the drug is renally excreted. No significant hemodynamic changes were observed after single IV doses of up to 300 mg in healthy volunteers. There are no significant interactions with carbamazepine, valproate, or oral contraceptives. In an initial multicenter study of 13 patients with refractory epilepsy, harkoseride was well tolerated in escalating doses of up to 600 mg per day (23). Recently, a similar study in 100 refractory patients showed a median reduction in seizures of 32% over a 12-week treatment phase, with a 50% responder rate of 33% (24). In this study, with the most common maximum tolerated dose being 600 mg per day, reported adverse effects included mild to moderate dizziness, fatigue, and somnolence. Additional studies are on-going.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Oct 17, 2016 | Posted by in NEUROLOGY | Comments Off on Newer Antiepileptic Drugs

Full access? Get Clinical Tree

Get Clinical Tree app for offline access