Molecular Targets for Novel Antiepileptic Drugs



Molecular Targets for Novel Antiepileptic Drugs


Brian S. Meldrum



Introduction

The majority of antiepileptic drugs (AEDs) introduced since 1938 have been identified as anticonvulsants by means of screening tests using in vivo models of seizures. The most widely applied models have been electrically or chemically induced seizures in normal animals. The maximal electroshock (MES) test in mice and rats and the “threshold” pentylenetetrazol (PTZ) test in mice have been rigorously standardized since 1978.52 This has had several predictable consequences. Drugs functionally similar to phenytoin appear to be preferentially identified by the MES test. Thus, most AEDs introduced since 1978 suppress sustained rapid repetitive firing in cultured neurons, apparently by prolonging inactivation of voltage-dependent sodium channels (e.g., lamotrigine, zonisamide, topiramate, felbamate, and oxcarbazepine).

The other most widely used anticonvulsant screen is the “threshold” pentylenetetrazol test, which is used to identify drugs potentially active against absence seizures52 (see Chapter 139). This is particularly sensitive to benzodiazepines and some other agents acting via the γ-aminobutyric acid (GABA)A/benzodiazepine receptor and to drugs acting on T-type calcium channels.

Thus, a key criticism of the traditional anticonvulsant screens is that they tended to identify drugs with the same mechanism of action as existing drugs because they have been validated in terms of their capacity to select existing AEDs. This applies both in a precise mechanistic sense (i.e., in terms of their molecular targets) but also in the broad sense that most currently used drugs are direct-acting agents with a close temporal correlation between plasma or brain levels and action at the target molecule and antiepileptic effect (with only vigabatrin, gabapentin, and valproate as partial exceptions). Thus, our current acute models may miss compounds that have indirect actions with potential delays in antiepileptic effect, as is possible for compounds acting via metabotropic receptors, protein kinases, neurotrophins, or changes in gene expression.

A further problem is that the epileptic brain differs from the normal brain. Elements thought to play a critical role in acute ictogenesis (i.e., events initiating a spontaneous seizure) include the transmembrane movement of ions (Na+, K+, Ca2+, Cl) and the release, postsynaptic action, and reuptake of excitatory and inhibitory neurotransmitters. The assumption that these processes are abnormal in the epileptic brain is supported by substantial data obtained in kindled animals71 and tentative data from patients with epilepsy.3,4,6,64 It follows that drugs modifying acute ictogenesis need to be studied in the epileptic brain, not in the normal brain. The genuinely epileptic model that has been most extensively used for screening is sound-induced seizures in DBA/2 or Fring’s mice. These are highly sensitive screens that identify a wide range of AEDs; they also identify some agents acting on monoaminergic systems that are relatively effective in reflex epilepsies but are not notably effective against other epileptic syndromes.16

Electrically kindled seizures would appear to be highly appropriate for pharmacologic studies because of their multiple abnormalities in ion channel and neurotransmitter receptor function.71 They are perhaps one of the better models of complex partial seizures, including the drug-resistant cases.58 However, the time-consuming nature of this model means that it is used not as a primary screen, but at a late stage in the evaluation of compounds identified by other anticonvulsant screens. A simplified procedure not involving electrode implantation (corneal kindled seizures) has been used as a tertiary screen14,51 (see also Chapter 139).

There are also genetically determined models of absence seizures in mice and rats that are responsive to antiabsence drugs and could be used to screen novel agents. These include the GAERS (genetic absence epilepsy rat Strasbourg) and several single gene mouse mutants (e.g., lethargic, stargazer, totterer).11

In contrast to the approach of in vivo screening of novel compounds with seizure models or epilepsy models is the mechanism-based approach, in which a molecular target is first defined and compounds are synthesized to optimize the action at this target and tested on a variety of in vitro systems before in vivo evaluation. This approach is totally dependent on our understanding of the causes of epilepsy and the mechanisms of chronic and acute ictogenesis. If attention is focused on mechanisms of action of existing drugs, this approach will have defects similar to in vivo screening. It is clearly important to focus attention on mechanisms of ictogenesis that have not yet been exploited therapeutically.

The following account attempts to describe some of the potential targets for novel therapies, putting them in the context of what is known about the mechanism of action of existing drugs and some novel agents currently in preclinical development whose clinical value is presently unknown.

The molecular targets for AEDs have recently been reviewed by Meldrum and Rogawski.69


Drugs Acting on Ion Channels








Table 1 Genetic Mutations Involving Voltage-gated and Ligand-gated Ion Channels and Epileptic Syndromes
























































































Gene Gene product (Channel) Epilepsy syndromes
SCN1A α subunit of Nav1.1 GEFS+ (type 2), SMEI, ICEGTCS
SCN2A α subunit of Nav1.2 GEFS+ (type 2), BFNIS
SCN1B β subunit of Nav1.1 to 1.4 GEFS+ (type 1)
CACNA1A α1 subunit of Cav2.1 P/Q type EAT2 + E, AEA
EFHC1 Coassembles with Cav2.3 R type JME
CACNA1H α1 subunit of Cav3.2 T type CAE
CACNB4 β4 subunit of P/Q and other channels IGE + EA
KCNA1 α subunit of Kv1.1 EAT1, MK, PS
LGI1 Coassembles with Kv1.1, 1.4, and Kvβ1 ADTLE
KCNAB2 β subunit (Kvβ2) combines with α subunit of Kv1.1 and Kv1.4 Epilepsy
KCNMA1 α subunit of Kca1.1 (BK) GEPD
KCNQ2
KCNQ3
α subunit of Kv7.2
α subunit of Kv7.3
BFNC
BFNC
KCNJ3 α subunit of Kir3.1 (Girk1) Absence
CLCN2 CLC2 Cl VG channel JME, CAE, JAE, EGMA
CHRNB2 β2 subunit nicotinic ACh receptor ADNFLE
CHRNA4 α4 subunit of nicotinic Ach receptor ADNFLE
GABRA1 α1 subunit, GABAA receptor JME
GABRBD β subunit GABAA receptor β3 subunit GABAA receptor JME Angelman syndrome
GABRG2 γ subunit GABAA receptor GEFS+, CAE, SME1
GABRD δ subunit GABAA receptor GEFS+
ATLE, autosomal dominant temporal lobe epilepsy; AEA, absence epilepsy with ataxia; BFNC, benign familial neonatal convulsions; BFNIS, benign familial neonatal-infantile seizures; CAE, childhood absence epilepsy; EAT1, MK, PS, episodic ataxia type 1 with myokymia; EAT2 + E, episodic ataxia type 2 with epilepsy; EGMA, epilepsy with grand mal seizures on awakening; GEFS+, generalized epilepsy with febrile seizures plus syndrome; GEPD, generalized epilepsy with paroxysmal dyskinesia; ICEGTCS, intractable childhood epilepsy with generalized tonic–clonic seizures, IGE + EA, idiopathic generalized epilepsy with episodic ataxia (Escayg et al. 2000); JAE, juvenile absence epilepsy; JME, juvenile myoclonic epilepsy; SMEI, severe myoclonic epilepsy of infancy.

Voltage-gated or ligand-gated ion channels in the neuronal membrane generate the electrical currents associated with the normal and abnormal functioning of the nervous system. The abnormal pattern of neuronal firing associated with epileptic seizures thus depends on abnormal functioning of these ion channels. They are thus the primary target for AEDs. This is confirmed by our present understanding of the mechanism of action of the established AEDs (see Chapter 136 and references 69 and 84). It is also broadly confirmed if we look at genetically determined syndromes of epilepsy, either those occurring spontaneously in man and rodents or those induced
by genetic manipulations in mice.2,35,99 These studies (Table 1) clearly identify the voltage-gated Na+, Ca++, and K+ ion channels as playing a critical role in causing idiopathic generalized seizures (and possibly some partial seizures in the case of K+). They do suggest that the VG K+ channels are of equivalent significance to the Na+ and Ca++ channels. They also identify the GABAA receptors as crucial contributors to generalized epilepsies.


Voltage-Gated Sodium Channels+

The majority of AEDs introduced since 1938 have properties that suggest they act on voltage-dependent sodium channels (Table 2). As suggested above, this emphasis on Na+ channel inactivation may be a consequence of the screening procedures employed.

Advances in the molecular biology of voltage-dependent Na+ channels now permit a precise definition of potential target sites for anticonvulsant drug action.24,55

The principal element of a voltage-dependent Na+ channel is the α subunit (260 kDa), which has four domains, each with six transmembrane helices, four of which form the voltage sensor and two (S5 and S6 connected by the P-loop that confers the ion-selectivity) forming the “pore domain.” The α subunit is assembled with subsidiary β1 to β4 subunits. Different cells express different α subunits; in the brain, I, II, IIA, and NaCh6 α subunits are found. Following an action potential, there is a transient process of inactivation during which the Na+ channel is unresponsive to voltage. This inactivation has fast and slow components that have been shown by site-directed mutagenesis to have distinct structural correlates. The loop linking domains III and IV act as a mechanical gate—“hinged lid”—producing fast inactivation. It can be rendered inoperative by proteolysis or site-directed mutagenesis.


Site-directed mutagenesis was also used to establish that three amino acids in the sixth transmembrane segment in domain 4 play a critical role in the binding of local anesthetics, phenytoin, and carbamazepine.12,82 Recently it has been shown that binding of lamotrigine and some related compounds is dependent on the same three amino acid residues in IV6 and also two in III6.12,82,119








Table 2 Voltage-gated Sodium Channels: Genes, Currents and AED Actions




















Gene Channel current AED action
SCN1A NaV 1.1 transient INa persistent INa Phenytoin, carbamazepine, and lamotrigine prolong inactivation of transient INa
SCN2A NaV 1.2 transient INa persistent INa Phenytoin, carbamazepine, and lamotrigine prolong inactivation of transient INa
SCN8A NaV 1.6 transient INa persistent INa Phenytoin, topiramate, and valproate decrease persistent INa
AED, antiepileptic drug.








Table 3 Voltage-gated Calcium Channels: Genes, Channels, and AED Actions
































Gene Channel current AEDs decreasing current
CACN A1S
CACN A1C
CACN A1D
CACN A1F
CaV 1.1 L-type HVA
CaV 1.2 L-type HVA
CaV 1.3 L-type HVA
CaV 1.4 L-type HVA
(Barbiturates, felbamate)
(Dihydropyridines block)
CACN A1A CaV 2.1 P/Q type HVA Lamotrigine (gabapentin)
CACN A1B CaV 2.2 N-type HVA Lamotrigine (gabapentin)
CACN A1E CaV 2.3 R-type HVA Topiramate (gabapentin)
CACN A1G
CACN A1H
CACN A1I
CaV 3.1 T-type LVA
CaV 3.2 T-type LVA
CaV 3.3 T-type LVA
Ethosuximide, zonisamide (modest reduction in ICa)
ACNA 2D1
CACN A2D2
α2δ1 subunit in Cav X.X
α2δ2 subunit in CavX.X
Gabapentin, pregabalin
Gabapentin, pregabalin
AED, antiepileptic drug.

Thus, there are specific sites within the α subunit that provide targets for AEDs, prolonging inactivation of voltage-sensitive Na+ channels. The four different α subunits expressed in the brain also may be seen as distinct targets.

There is a further function of Na+ channels that is a target for AEDs. They provide persistent, noninactivating Na+ currents that contribute to certain patterns of burst firing.103 Phenytoin diminishes these currents,15 as do topiramate and valproate.

Thus, Na+ channels provide a multiplicity of targets. Acting on two families of sodium channels may be advantageous for anticonvulsant efficacy. There are homologies of sequence and morphology between voltage-sensitive Na+, Ca2+, and K+channels, and some known Ca2+ channel blockers also act on Na+ channels (e.g., flunarizine). Thus, designing compounds that block both classes of ion channel may lead to improved AED efficacy.


Voltage-gated Calcium Channels

These Ca2+ channels were originally classified in terms of biophysical measurements across neuronal membranes, determining voltage activation threshold, conductance per channel, and inactivation kinetics, giving T-, N-, and L-type Ca2+ channels. Other channels (P/Q, N, and R) were later identified (Table 3).13,105 The role of voltage-gated calcium channels in immunoglobin (IG) E has recently been reviewed.43,50

As explained in Chapter 136, there is an apparent link between action against 2- to 3-Hz absence attacks and a partial suppression of Ca2+ T-channel conductance.22 The T-type calcium current makes a specific contribution to oscillatory responses in thalamic neurons, and these oscillations play a critical role in 2- to 3-Hz discharges in the cortex and thalamus.122 Thus, the suppression of absence attacks by ethosuximide and dimethadione has been attributed to the reduction in T Ca2+ currents they produce, although a variety of other effects on ion channels may also be involved. High concentrations of valproate reduce T currents in primary afferent neurons.46 Zonisamide also reduces T Ca2+ currents. Perhaps T-type Ca2+ channels are best regarded as an auxiliary or supplementary target to broaden the spectrum of activity of a compound intended to affect more than one molecular mechanism.


The greatest interest in terms of targets for novel drugs are the high-voltage-activated channels designated as P/Q, N, and R type (Cav 2.1 to 2.3), because of their presynaptic role controlling release of neurotransmitters. N-type channels appear to be important for synaptic release of monoamines and some peptides.109 The P/Q-type Ca2+ channels are important for the release of monoamines, glutamate, and GABA.37 There is evidence suggesting that some established and novel AEDs act on presynaptic Ca2+ channels.30,53,98,111 Thus, P/Q-, N-, and R-type calcium channels appear to be key targets for the development of novel AEDs. It should be remembered, however, that mutations leading to impaired function of P/Q-type channels produce ataxia and absence seizures in man and mouse.








Table 4 Potassium Channels: Genes, Channels, and AED Actions




































Gene Channel current AED action
KCNA1 K 1.1 delayed rectifier (A-type)
KCNQ2 K 7.2 M-type Retigabine
KCNQ3 K 7.3 M-type
KCNQ5 K 7.5 M-type
KCNJ3 Kir 3.1 Girk1
KCNJ6 Kir 3.2 Girk2
KCNJ10 Kir 4.1
KCNJ11 Kir 6.2
KCNMA1 KCa 1.1 BK or slo1
AED, antiepileptic drug.


Voltage-gated and Other Potassium Channels

Potassium selective ion channels were originally differentiated in biophysical studies and are found in all mammalian cells. Today they are classified partially on the basis of their functional properties and partially on the basis of their evolutionary origin and molecular morphology120,121 (see Tables 1 and 4). The 75 or more distinct α subunits identified in genetic studies provide four main families: Voltage gated (Kv1 to Kv10); calcium activated (Kca2.1 to 2.3, Kca1.1); inwardly rectifying (kir); and the two-pore K+ channels that provide the “leak” currents (K2P). The voltage-gated and calcium-activated K+ channels are part of the S4 superfamily and have an α subunit with a core formed by six putative transmembrane helices. The S1 to S4 segment provides the voltage sensor domain homologous with that of the Na+ and Ca++ion channels. A linker helix connects this to the S5 and S6 segments, which, with their P-loop, form the ion-selective channel.21,56,57 Opening K+ channels has the effect of hyperpolarizing neurons or reversing depolarizing actions. At least two different calcium-activated K+ channels underlie afterhyperpolarizations.89 One, SKCa, with a fast time course, is mediated by small conductance channels; the other has a slow time course and is modulated by numerous G-protein–coupled neurotransmitters. Studies of dentate granule cells isolated from patients with temporal lobe epilepsy show a prominent delayed rectifier outward current (IK) whose steady-state voltage dependence differs according to whether the specimen shows Ammon horn sclerosis or not.3

Many compounds that act as potassium channel blockers are convulsant, for example, 4-aminopyridine and various scorpion and sea anemone toxins such as dendrotoxin I and pandinus toxin.44,108 In in vitro systems, 4-aminopyridine–induced discharges are commonly used as a model for analyzing seizure activity. Several drugs thought to act as potassium channel openers such as chromakalim, minoxidil, diazoxide, and pinacidil show antiepileptic effects when injected intracerebroventricularly in rodent models of epilepsy.32,80 The most definitive data concern a drug retigabine, developed as a GABA modulator but shown in 2000 to activate Kv7.2 and Kv7.3 channels responsible for the M-type current on soma and dendrites and the slow K+ current (Iks) on axons.88,115 Mutations involving the genes encoding these channels (KCNQ2/KCNQ3) are responsible for benign familial neonatal convulsions.19,96 Retigabine shifts the voltage dependence of the M current to hyperpolarized potentials, speeds the rate of activation, and slows inactivation. It has a similar effect on Kv7.4 and Kv7.5, which are also expressed in the brain, but is inactive on Kv7.1, the cardiac homolog. Experiments with chimeric channels and point mutations90,118 have shown that the pore domain is the site of action of retigabine and that its action is dependent on a tryptophan residue on S5 found in KCNQ2 to 5 but not in KCNQ1. Further compounds have been identified with a similar channel-opening effect and activity in rodent models of epilepsy.117 Thus, the potassium channels responsible for the M current are clearly identified as a target for AEDs. Genetic and pharmacologic data suggest that the voltage-gated K channels responsible for the A-type K+ currents, which play an important role in controlling the excitatory threshold in synaptic terminals, are also an important potential target for novel AEDs,21,69 but drugs potentiating these currents have yet to be identified. The calcium-activated channels and the inwardly rectifying and leak currents may also be targets, but evidence is less clear.

Many reports describe effects of established AEDs (such as phenytoin, carbamazepine, lamotrigine, and levetiracetam) on various K+ currents,69 but the significance of these for antiepileptic effect remains uncertain.


Carbonic Anhydrase Inhibition

A possible mechanism for the antiepileptic action of carbonic anhydrase inhibition involves GABA-mediated inhibitory potentials.97 GABAAreceptors open ion channels that are permeable to HCO3 as well as to Cl. HCO3 moves outward, producing a depolarizing effect that is normally smaller than the hyperpolarizing action of the inward flux of Cl. With strong GABAA receptor activation, the Cl gradient in dendrites collapses sooner than does the HCO3 gradient (which is preserved by the inward flux of CO2 and carbonic anhydrase activity). GABA thus develops a depolarizing effect that can be blocked or diminished by carbonic anhydrase inhibitors. The conversion of GABA’s effect to depolarization may contribute to several epileptic manifestations, and carbonic anhydrase inhibitors may be beneficial.






Table 5 AEDs and Related Compounds Potentiating GABA-Mediated Inhibition

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

Stay updated, free articles. Join our Telegram channel

Aug 1, 2016 | Posted by in NEUROLOGY | Comments Off on Molecular Targets for Novel Antiepileptic Drugs

Full access? Get Clinical Tree

Get Clinical Tree app for offline access