You are on page 1of 11

Epilepsy & Behavior 21 (2011) 331–341

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

Antiepileptic drug therapy: Does mechanism of action matter?


Martin J. Brodie a,⁎, Athanasios Covanis b, Antonio Gil-Nagel c, Holger Lerche d, Emilio Perucca e, f,
Graeme J. Sills g, H. Steve White h
a
Epilepsy Unit, Western Infirmary, Glasgow, UK
b
Neurology Department, Agia Sophia Children's Hospital, Thivon and Levadias, Goudi, Athens, Greece
c
Epilepsy Programme, Hospital Ruber Internacional, Madrid, Spain
d
Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University Hospital Tübingen, Tübingen, Germany
e
Clinical Pharmacology Unit, Department of Internal Medicine and Therapeutics, University of Pavia
f
National Institute of Neurology IRCCS C Mondino Foundation, Pavia, Italy
g
Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
h
Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA

a r t i c l e i n f o a b s t r a c t

Article history: This article represents a synthesis of presentations made by the authors during a scientific meeting held in
Received 10 March 2011 London on 7 June 2010 and organized by GlaxoSmithKline. Each speaker produced a short précis of his lecture
Revised 19 May 2011 to answer a specific question, resulting in an overview of what we know about the relevance of the
Accepted 24 May 2011
mechanisms of action of antiepileptic drugs in determining appropriate combination therapies for the
Available online 16 July 2011
treatment of drug-resistant epilepsy.
Keywords:
© 2011 Elsevier Inc. All rights reserved.
Antiepileptic drugs
Mechanism of action
Epileptogenesis
Antiepileptogenesis
Disease modification
Epilepsy
Pharmacokinetics
Pharmacodynamics
Interactions
Treatment

1. Introduction potentially useful combinations that require investigation—too many


for a doctor and his or her patient to try in a single lifetime.
The past two decades have witnessed an unprecedented expan- Accordingly, a more focused approach to treating drug-resistant
sion in pharmacological treatment options for epilepsy [1], but there epilepsy is needed [10]. It is logical to suggest that this should be
is limited evidence to suggest that clinical outcomes have substan- based on the mechanisms of action of the available therapeutic
tially improved for people with epilepsy over that period [2–4]. This is armamentarium. Although we have a reasonable idea of how the
perhaps not surprising as the placebo-corrected efficacy of adjunctive majority of AEDs act, little is known about the pathophysiology of
treatment with modern antiepileptic drugs (AEDs) in double-blind, most epilepsies or the neurobiologies underpinning drug response
dose-ranging trials in adults with refractory focal epilepsy has been and pharmacoresistance in the individual patient.
disappointingly small [5], with very few patients becoming seizure A rational approach to combination therapy has been adopted
free, even for the relatively short period of these trials [6]. The in other complex, multifaceted diseases in which a single agent is
situation seems no better for childhood epilepsies [7–9]. With 20 ineffective in a substantial proportion of patients. Examples
currently recognized AEDs, there are, in theory, 190 possible include migraine [11], neuropathic pain [12], hypertension [13],
partnerships of two drugs and 1140 possible combinations of three and human immunodeficiency virus infection [14]. In addition,
drugs. Although a significant proportion of these are not likely to be rational polytherapy is used increasingly in oncology as agents
used in clinical practice, there remains a substantial number of are developed that target specific pathogenic mechanisms
[15,16].
⁎ Corresponding author at: Epilepsy Unit, Western Infirmary, Glasgow G11 6NT,
In this review we consider the theory and practice of combining
Scotland, UK. Fax: + 44 141 211 2072. AEDs in the treatment of epilepsies as approaches to a number of
E-mail address: mjb2k@clinmed.gla.ac.uk (M.J. Brodie). relevant questions.

1525-5050/$ – see front matter © 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.yebeh.2011.05.025
332 M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341

2. Does what we know about mechanisms of action match what inase [31]. However, none of the currently marketed drugs primarily
we know about seizure generation? targets the brain's main excitatory neurotransmitter, glutamate, or
potassium channels, which hyperpolarize neural membranes. The
Ion channels, which form the basis of neuronal excitability and glutamate pathway is targeted by the developmental agent perampanel,
synaptic transmission, are membrane-spanning proteins that form an AMPA-type glutamate receptor antagonist, which has now completed
selective pores with gates that open and close in response to stimuli a phase III program as an add-on treatment in focal epilepsy; full results
such as membrane voltage or ligand binding. Recordings from are awaited [32]. The investigational agent retigabine (also known as
epileptic foci typically show a paroxysmal depolarizing shift, in ezogabine), which was recently approved by the FDA and the CHMP,
which glutamate receptors and calcium channels are implicated in the enhances the activity of KV7.2/KV7.3 potassium channels by binding
depolarizing and plateau phases. These feature recurrent action within the pore region [33,34]. These channels generate the M current, a
potentials, during which GABA receptors and potassium channels non-inactivating potassium conductance that regulates the neuronal
are involved in the hyperpolarizing and terminating phases, respec- firing rate at subthreshold voltages between −60 and −40 mV [35].
tively. Similarly, rhythmic discharges in thalamocortical loops, which Enhancing the M current with retigabine hyperpolarizes the cell
play an important role in generalized seizures, are determined by the membrane toward the potassium equilibrium potential [36]. This drug
interplay of depolarizing and repolarizing ionic conductances. has been shown to be effective in phase III add-on trials in refractory
The most common target among AEDs is the sodium channel, focal epilepsies [37,38].
which is responsible for the upstroke of the action potential in The literature on AED pharmacology is vast, and our current
neurons and other excitable cells [17]. The majority of sodium understanding of the mechanisms of action of drugs used in the treatment
channels expressed in the mammalian brain belong to four subtypes, of epilepsy is undoubtedly incomplete [31,39]. Nevertheless, it is possible
two of which have been found to be mutated in genetic forms of to categorize current AEDs into discrete pharmacological groups based on
epilepsy. NaV1.1 (SCN1A) mutations cause a spectrum of epileptic their most prominent mechanism of action at therapeutic concentrations
disorders ranging from simple febrile seizures to Dravet syndrome (Table 1). This categorization includes a group of drugs known to possess
[18]. Of the more than 200 NaV1.1 mutations that have been multiple prominent cellular effects, giving a total of five broad and nine
described, most cause loss of function, with complete loss of function more specific mechanistic categories of AEDs.
of one allele in Dravet syndrome [18]. At first, it seems paradoxical
that a loss of sodium channel function would produce epileptic 3. Does mechanism of action predict antiepileptic drug efficacy or
seizures. However, knockout mouse models have revealed that NaV1.1 side effects?
seems to be the main sodium channel in inhibitory interneurons
[19,20], which may explain the pathophysiology of neuronal network Animal models of seizures, epilepsy, and drug-related neurotox-
hyperexcitability. Furthermore, the sodium channel-blocking drug icity are widely used in the identification of novel antiseizure agents,
lamotrigine may aggravate epileptic seizures in patients with NaV1.1 and many of them have been in routine laboratory use for decades
mutations [21]. This observation could be explained by a block of the [40]. Together, these models offer an opportunity to characterize the
remaining functional sodium channels which are generated by the efficacy and toxicity of single drugs and combinations of drugs in large
healthy allele in inhibitory interneurons. In contrast, mutations in groups of genetically homogeneous animals and have been instru-
NaV1.2 (SCN2A) cause a benign epilepsy syndrome of neonates and mental in the identification of almost all current AEDs, providing
infants through a gain of function in sodium channels expressed in the valuable surrogates in the search for optimal combination therapies
axon initial segment of excitatory principal neurons in the hippo- [41]. The models most commonly used for AED screening are maximal
campus and cortex [22,23]. The transient high expression of these electroshock (MES), pentylenetetrazol (PTZ), spike–wave models and
channels during development and their subsequent partial replace- kindling models for efficacy purposes, and the rotarod, horizontal
ment with NaV1.6 channels may explain the remission of seizures at screen, and chimney tests for neurotoxicity [42].
later ages in patients with NaV1.2 mutations [22,23].
Mutations in KV7.2 and KV7.3 potassium channels (KCNQ2/KCNQ3)
Table 1
lead to a decrease in functional potassium channels in the axon initial Mechanistic categorization of current antiepileptic drugs based on foremost targets at
segment and cause neonatal seizures [24]. These benign seizures provide therapeutic concentrations.
a useful model of disturbed excitability in epileptogenesis. Other genetic
Target and mechanism Antiepileptic drug
potassium channel defects have been described including KV1.1, related
to episodic ataxia with focal epilepsy [25], and KCa1.1, related to Sodium channel actions
Blockade by stabilizing fast-inactivated state Phenytoin, carbamazepine,
paroxysmal dyskinesia with generalized epilepsy [26]. The involvement lamotrigine, oxcarbazepine,
of potassium channel defects in the generation of focal epilepsies is rufinamide, eslicarbazepine
supported by the observation that KV4.2 channels are reduced in acetate
principal neurons in an animal model of temporal lobe epilepsy [27]. Blockade by stabilizing slow-inactivated state Lacosamide
Calcium channel actions
Traditional sodium channel blockers, such as phenytoin, carbamaz-
Blockade of high voltage-activated Gabapentin, pregabalin
epine and derivatives, and lamotrigine, bind to the fast-inactivated state channel (P/Q type)
of sodium channels and cause a use-dependent blockade following Blockade of low voltage-activated Ethosuximide
opening and inactivation. In contrast, lacosamide binds to the slow- channel (T type)
inactivated state of sodium channels that predominates during GABA-related actions
Activation of GABAA receptor Phenobarbital, benzodiazepines
prolonged depolarization and, in so doing, extends the latency period Blockade of GABA transporter Tiagabine
between action potentials to a greater extent than traditional sodium (GAT1a selective)
channel blockers [28]. However, none of these drugs has only one Inhibition of GABA transaminase Vigabatrin
mechanism of action. For example, some sodium channel blockers also SV2A actions
Modulation of SV2A Levetiracetam
block presynaptic calcium channels [29], and lamotrigine enhances the
Multiple actions
activity of hyperpolarization-activated cyclic nucleotide-gated (HCN) Various actions on multiple targets Valproic acid, felbamate,
cation channels [30]. topiramate, zonisamide
Other AEDs enhance GABA-mediated inhibitory activity, classically Potassium channel activity
benzodiazepines and barbiturates, but also tiagabine, which blocks Opens Kv7 potassium channels Retigabine/ezogabine

synaptic GABA reuptake, and vigabatrin, which inhibits GABA transam- a


GAT1, GABA transporter 1; SV2A, synaptic vesicle protein 2A.
M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341 333

The kindling model, in which electrical or chemical stimulation To avoid this lack of predictability of efficacy and adverse effects, a
induces a progressive decrease in seizure threshold and a corresponding different approach is needed in AED development, including trials on
increase in seizure severity and afterdischarge duration, is considered a epilepsy syndromes other than focal epilepsies, genetic testing of
valuable model for studying epileptogenesis [43] and the efficacy of large patient populations, and development of novel animal models.
AEDs against focal seizures [44]. Inhibition of hind-leg extension in the Such changes are beginning to take effect: some AEDs are being tested
MES test suggests that an agent is able to prevent seizure propagation in idiopathic and symptomatic generalized epilepsies; most clinical
and, therefore, may be effective against generalized tonic–clonic (GTC) trials now include genetic analysis of the population studied; and the
seizures [42]. Drugs that inhibit voltage-gated sodium channels, such as number of animal models of refractory epilepsy used to evaluate AED
phenytoin, carbamazepine, lamotrigine, and oxcarbazepine, are usually efficacy is expanding. In addition, clinical experience is being applied
effective in this model. In the PTZ test, clonic seizures are blocked by in trial design, providing more specific analyses of efficacy and better
AEDs acting on the GABAA neurotransmitter system, such as barbitu- detection of adverse effects. The regular use of depression scales and
rates, benzodiazepines, valproic acid, and tiagabine, and by ethosux- adverse-event questionnaires in modern clinical trials is encouraging.
imide through its action on T-type calcium channels [45]. Drugs effective However, adverse effects such as depression, anxiety, and psychosis,
in the PTZ test are considered to be likely to suppress myoclonic and/or and effects on cognition probably also need to be better defined. The
absence seizures [46]. Prediction based on these tests correlates with predictability of efficacy and toxicity would be enhanced by improved
clinical results for most AEDs, but there are striking exceptions. testing of larger populations in head-to-head monotherapy trials.
Clonazepam is not effective in the MES test, but has clinical efficacy
against GTC seizures. Similarly, levetiracetam is not effective in the MES 4. What are the pharmacological issues in treating children
and PTZ tests, but is useful in some idiopathic generalized epilepsies, with epilepsy?
including epilepsies with prominent myoclonic features [47–49].
Furthermore, gabapentin, pregabalin, and tiagabine, which demonstrate Almost all of the evidence for the safety and efficacy of AEDs
efficacy in the PTZ test, not only fail to control absence and myoclonic discussed above is based on studies in adults. Children, particularly
seizures, but can exacerbate these seizure types [50]. infants, differ from adults not only in the clinical expression of their
Spike–wave seizure models, such as GAERS (Genetic Absence seizures, but also in the unique age-dependent EEG abnormalities
Epilepsy Rats from Strasbourg), lethargic mouse, and WAG/Rij rat, are characteristic of childhood self-limited epileptic syndromes as well as
sensitive to benzodiazepines, valproic acid, ethosuximide, lamotri- epileptic encephalopathies. Diffuse epileptic activity interferes with
gine, and levetiracetam [51], which correlates with the clinical the development and organization of the immature brain and with
efficacy of these AEDs against absence seizures. However, although cognition [71]. This is most conspicuous in infancy, in which 40% of
clinical trials have shown that some AEDs may be more effective than epilepsies can be classified as epileptic encephalopathies and only a
others in the treatment of childhood absence epilepsy [52], the spike– few respond to pharmacological, nonpharmacological, or neurosur-
wave models so far have not been able to stratify AEDs based on their gical approaches. In this age group, epileptic activity and seizures may
degree of efficacy in the treatment of this syndrome. become persistent, thereby perpetuating hyperexcitability and alter-
For practical reasons, clinical trials are conducted using relatively ing networks in seizure-prone regions. This can affect the outcome by
standard patient selection, which is almost always limited to adults with producing changes in seizure semiology (focal or generalized) and
focal epilepsies [53]. Occasionally, an AED approved for focal epilepsy possibly reduced responsiveness to treatment [72]. To avoid or
will later show efficacy against idiopathic generalized epilepsies, but it is minimize such consequences, the correct diagnosis of seizures and
probable that many compounds potentially effective against general- syndromes is mandatory.
ized epilepsies but not active against focal epilepsies are rejected during The recognition of seizure type and syndrome gives us valuable
development. It is notable that add-on and monotherapy trials often information regarding treatment strategies and prognosis. The
show little differences in efficacy among drugs [54,55], which may be decision to treat a child after the first unprovoked seizure has an
related to the fact that all of these agents went through the same impact on the recurrence risk over a short, but not over a longer,
assessment and development processes. Despite this, some studies have period [73–75]. There are “good epilepsies,” such as “benign” focal
been able to demonstrate the efficacy of AEDs on epilepsies other than epilepsy syndromes, that may not need any treatment. Subsequent
those of focal onset, including levetiracetam in juvenile myoclonic management, however, will depend on the type of seizures,
epilepsy [56], rufinamide in Lennox–Gastaut syndrome [57], vigabatrin recurrence frequency, and the way in which the parents perceive
in infantile spasms [58], and stiripentol in Dravet syndrome [59]. and manage the episodes. There are also “bad epilepsies,” such as
However, it was clinical observation rather than prediction of efficacy Landau–Kleffner syndrome and continuous spike–waves during sleep,
based on mechanism of action that led to these observations. for which children often need aggressive treatment from the outset.
Analysis of adverse-effect profiles based on mechanism of action Parents should be instructed on how to identify these seizures early
shows few differences among approved AEDs. Adverse effects consis- and to act quickly to abort them, for example, with buccal midazolam
tently noted in randomized trials include dizziness, lethargy, diplopia, or rectal diazepam.
vertigo, and unsteadiness. This is probably related to the trial design, in Misdiagnosis is common in children and occurs in 18–30% of
which new compounds are added to one, two, or three background patients, mainly through misinterpretation of clinical and nonepilepto-
AEDs regardless of their dosage or mechanisms of action. Clinical genic paroxysmal interictal EEG activity [76]. An EEG obtained using the
observation, however, indicates that sodium channel blockers are most correct methodology relevant to the clinical event is the most important
often associated with cerebellar adverse effects [60]. In line with this, diagnostic test in evaluating children with suspected epilepsy. Once the
analysis of patient subgroups in some clinical trials has shown that underlying etiology has been identified, the decision will be made
neurotoxic effects can be more prominent when two traditional sodium whether to treat or not to treat, and, if necessary, the first-choice
channel blockers are combined, compared with the addition of a sodium medication selected. The short-term aim is seizure freedom with
channel blocker to AEDs with different mechanisms of action [61–64]. minimal or no adverse effects, and the long-term goal is optimal motor,
Similarly, clinical observations indicate that headache can be social, cognitive, and developmental evolution, coupled with good
associated with lamotrigine [65], depression with levetiracetam [66], quality of life for the child and the family. In choosing the appropriate
anhydrosis with AEDs that inhibit carbonic anhydrase (topiramate AED, the type of seizure or syndrome, pharmacodynamic and
and zonisamide) [67–69], and psychosis with the GABAergic drug pharmacokinetic properties of the drug(s), the patient's age, gender,
vigabatrin [70]. However, the evidence for these associations was not activities, and body weight, and also the preference of parents should, if
always obtained from placebo-controlled, randomized studies. possible, be taken into account.
334 M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341

In patients with primarily generalized seizures (absence, myoclonic, lamotrigine, and other early adverse effects, such as sedation and
tonic–clonic, tonic, atonic), particularly in those with idiopathic aggression. Useful AED combinations in focal seizures include carbamaz-
generalized epilepsies and syndromes with or without a positive epine, or preferably oxcarbazepine, with a broad-spectrum drug such as
response to intermittent photic stimulation, valproic acid can be valproic acid or levetiracetam [93]. Carbamazepine, however, may
regarded as a possible drug of first choice for all types of seizures [77]. aggravate seizures or precipitate continuous spike-and-waves during
Valproic acid is a particularly suitable first choice in syndromes in which sleep in children with Landau–Kleffner syndrome [94] and idiopathic
myoclonic seizures are the only or predominant seizure type and in focal epilepsies [95]. Additionally, in benign childhood epilepsy with
syndromes in which absence and myoclonic seizures are the main centrotemporal spikes and minor atypical features, carbamazepine can
seizure types. aggravate epileptic negative myoclonus and precipitate almost contin-
In cases of obesity, and in girls from the prepubertal phase onward, uous central spike-and-wave activity. Seizure exacerbation has also been
an alternative to valproic acid should be sought. The increased risk of reported in Panayiotopoulos syndrome [96]. Alternative choices for
congenital malformations when this agent is used, compared with treatment of these syndromes are levetiracetam [97–99], valproic acid
other traditional AEDs, particularly when the daily dose exceeds [100], and sulthiame [101,102].
1000 mg, dictates that valproic acid should be avoided, if possible, in Epileptic encephalopathies are severe brain disorders, in which the
girls and women of childbearing potential [78,79]. Cognitive and underlying electrical activity significantly contributes to progressive
behavioral abnormalities have also been observed in children exposed cognitive dysfunction [103]. Treatment is aimed not only at preventing
to valproic acid in utero [80–82]. Serious adverse reactions to valproic seizures, but also at abolishing ictal and interictal EEG abnormalities.
acid, such as hepatic failure and acute pancreatitis, occur mainly in Vigabatrin, a GABA transaminase inhibitor, may be a first-choice agent
children receiving polytherapy and in infants with organic brain for infantile spasms [104]. Adrenocorticotropin can be added if
(metabolic) disease. For some girls and young women with idiopathic vigabatrin fails to prevent seizures and abolish the hypsarrhythmic
generalized epilepsy syndromes, there may be no suitable alternative pattern within 5 days. Additional treatment can include levetiracetam,
to valproic acid, in which case doses under 1000 mg/day should be topiramate, valproic acid, benzodiazepines, zonisamide, pyridoxine (in
employed. pyridoxine dependency cases in which a prompt EEG response is
Potential alternatives to valproic acid include ethosuximide, observed after intravenous infusion of 100–200 mg), or intravenous
lamotrigine, levetiracetam, topiramate, zonisamide, and benzodiaze- immunoglobulin G.
pines [83]. Ethosuximide is effective against absences, and a recent In Dravet syndrome, the main genetic defect is loss of function in
study found it to be less likely to cause cognitive impairment than voltage-gated sodium channels and traditional sodium channel-
valproic acid [52], but it does not protect against GTC seizures. blocking AEDs are not recommended [105]. Stiripentol, a direct
Lamotrigine, primarily a sodium channel blocker, can be effective for allosteric modulator of GABA receptors [106], combined with valproic
primary GTC seizures, but can worsen myoclonic seizures [21,84] and acid and clobazam has given encouraging results [59]. Topiramate
is less effective than ethosuximide or valproic acid for absences [52]. may also be of value in some children [107].
Levetiracetam is particularly effective in juvenile myoclonic epilepsy The majority of AEDs are ineffective in encephalopathies with
[85], whereas topiramate can cause cognitive dysfunction in some continuous spike-and-waves during sleep; some benefit with
children. sulthiame has been reported [108].
Useful combinations that have been identified clinically in patients Specific clinical trials are needed in children and infants with
not responding to monotherapy include valproic acid with ethosux- epileptic encephalopathies because early and successful prevention of
imide for absence seizures [86,87], valproic acid with levetiracetam seizure activity will lead to better outcomes with regard to cognition
for myoclonic seizures, and valproic acid with lamotrigine for a variety and quality of life. In addition, mechanisms underlying drug resistance
of seizure types. in infancy and childhood need to be better understood to develop
Gabapentin and pregabalin can worsen myoclonic and, possibly, novel therapies. The consequences of inadequate seizure control are
absence seizures [88,89]. Vigabatrin and tiagabine are inactive against different for each child, depending on age, type of seizure, diurnal
absence seizures and may exacerbate generalized spike-and-wave variation of seizures, lifestyle, and quality-of life-issues. New genetic
discharges (GSWDs). Tiagabine may also induce nonconvulsive status approaches to mechanisms of epileptogenesis will enhance progress
epilepticus [90]. Classic AEDs that bind to the fast-inactivated state of toward more effective treatment for these challenging epilepsy
sodium channels such as phenytoin, carbamazepine, oxcarbazepine, syndromes.
and eslicarbazepine acetate should be avoided in idiopathic general-
ized epilepsy syndromes associated with absence and myoclonic 5. How can experimental studies on drug combinations inform
seizures [89]. clinical practice?
In focal seizures and syndromes with or without secondary
generalization, drugs with activity in either MES or kindling models are The systematic investigation of all possible AED combinations is
typically effective, including phenobarbital, phenytoin, carbamazepine, scientifically and economically unrealistic. As an alternative, it is
oxcarbazepine, valproic acid, lamotrigine, levetiracetam, lacosamide, reasonable to investigate combinations of mechanisms rather than
topiramate, zonisamide, gabapentin, pregabalin, felbamate, tiagabine, drugs. There are a total of 32 possible combinations from nine
vigabatrin, and benzodiazepines [41]. However, some of these drugs have mechanistic categories (Table 1), a figure that can be substantially
not been adequately tested in children. Carbamazepine, lamotrigine, and reduced as clinical experience dictates that some combinations can be
oxcarbazepine can be regarded as first-choice AEDs for monotherapy for ignored. Even excluding combinations that are not used clinically,
all types of focal seizures in children with or without secondary undertaking a robust clinical evaluation of the remaining mechanistic
generalization. Lamotrigine and oxcarbazepine may be preferable to combinations would be impractical because it would require
carbamazepine in infancy and early childhood because they may be assessment of efficacy and dose-related adverse effects of both
better tolerated inasmuch as they are not broad-spectrum enzyme drugs applied singly and in combination in sufficiently large and
inducers. Lamotrigine, however, carries an increased risk of serious skin homogeneous groups of patients with careful consideration of dose
rashes [91]. Levetiracetam has a favorable pharmacokinetic profile and ratios and drug loads [109]. As an initial and potentially cost-effective
can be used as monotherapy or in polytherapy in all focal or generalized, alternative, animal models can be used to identify the most effective
idiopathic or symptomatic epilepsy syndromes in children, without mechanistic combinations prior to detailed clinical evaluation.
clinically significant pharmacokinetic drug interactions [83,92]. Slow Experimental drug combination studies should incorporate efficacy
titration is advisable to minimize the risk of skin rashes, particularly with and toxicity models in which both drugs are at least minimally effective
M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341 335

and use drug ratios that reflect those employed clinically. Drug more rational approach to combination therapy, with potential
concentrations in both plasma and the brain should be measured to advantages in terms of efficacy and minimization of adverse effects.
rule out confounding pharmacokinetic interactions, and an appropriate Interactions whereby an AED affects the gastrointestinal absorp-
method of analysis should be applied, such as isobolography or tion of another AED are uncommon [125]. It has been suggested that
comparison of protective indices [110]. Isobolographic analysis is the fall in plasma phenytoin concentration that occurs in some
preferable as it provides a robust measure of effectiveness and affords patients started on combination therapy with vigabatrin may involve
a definitive determination of infra-additive (antagonistic), additive, or an interaction at the absorption level, but an ad hoc study failed to
supra-additive (synergistic) interactions [111]. The ideal AED combi- provide any evidence for this, and the mechanism underlying this
nation would demonstrate pharmacological synergism, defined as interaction remains unclear [126].
improved efficacy with similar toxicity, similar efficacy with reduced Plasma protein binding interactions are relevant only for highly
toxicity, or, ideally, improved efficacy with reduced toxicity. protein-bound AEDs, such as phenytoin and valproic acid. Contrary to
Experimental investigations of AED combinations have appeared common belief, these interactions are usually of little or no clinical
in the literature since the 1950s [112]. Early studies explored significance, because the displaced drug becomes diluted in a large
combinations of established drugs, such as phenobarbital, phenytoin, volume of distribution and is also eliminated more rapidly as a result of
and carbamazepine, and were performed without any knowledge of an increase in clearance. The usual consequence of these interactions is a
their mechanisms of action. Interestingly, combinations were com- fall in the total plasma concentration of the displaced drug, without any
monly reported to have superior efficacy, reduced toxicity, or both, significant change in the concentration of the unbound, pharmacolog-
compared with either constituent drug alone irrespective of the drugs ically active molecule [125]. For example, when protein-bound
under investigation [113]. However, this was most evident when two phenytoin is displaced by valproic acid, the total plasma phenytoin
drugs with what we now know to have different mechanisms of concentration falls, but the unbound phenytoin concentration is not
action were combined [114]. Combinations of AEDs with similar affected and the phenytoin response is generally unaltered [127]. In
mechanisms were less successful [115]. These studies set the some cases, unbound phenytoin levels may increase moderately, but
benchmark for the concept of “rational polypharmacy” as it evolved this is due to a concurrent inhibitory effect of valproic acid on phenytoin
throughout the 1990s with new knowledge about the pharmacology metabolism rather than displacement from plasma proteins. The
of AEDs and the advent of modern drugs, which are invariably important message for physicians is that, despite the fall in total plasma
introduced as adjunctive therapy for refractory focal epilepsy [116]. phenytoin levels, the phenytoin dosage does not need to be increased
Arguably, the laboratory of Czuczwar and colleagues has contrib- when valproic acid is given concomitantly.
uted more data to the study of AED combinations than all others The most common pharmacokinetic interactions in AED therapy
[117,118]. Some common themes have emerged in the literature, are those resulting in changes in the rate of drug metabolism.
most notably that combining drugs with differing mechanisms of These interactions can be very important clinically [128]. Carba-
action appears to be superior to combining those with the same or mazepine, phenytoin, and phenobarbital, in particular, markedly
similar cellular effects [113]. This observation is exemplified by the stimulate the activity of most cytochrome P450 (CYP) enzymes, as
consistent demonstration of synergism between lamotrigine and well as enzymes involved in glucuronidation (uridine 5'-diphospho-
valproic acid in multiple efficacy models [119,120] and the repeated glucuronosyltransferases [UGTs]). Enzyme-inducing AEDs interact
failure to demonstrate synergism between lamotrigine and either with many other medications that are metabolized in the liver, as
carbamazepine or phenytoin [120,121]. Other combinations produced well as other AEDs (Table 2). Although the reduction in the steady-
less consistent results; for example, gabapentin with carbamazepine state plasma concentration of the affected AED may be compensated
is synergistic in the MES model but not in the DBA/2 audiogenic by the efficacy of the added drug, in many cases the dosage will need
seizure susceptible mouse [122,123]. Overall, however, the message is to be increased to optimize efficacy. This decrease in plasma
relatively clear: reinforcement on a single pharmacological pathway is concentration can be particularly marked (N50%) when the affected
less effective than a combined effect on two distinct pathways. AED depends almost entirely on metabolic clearance for its
Relatively few studies contradict this intuitive hypothesis [124]. elimination, as is the case with lamotrigine, valproic acid, tiagabine,
Some of the most successful two-drug combinations in laboratory and carbamazepine [125]. Caution is also required when an enzyme-
studies appear to involve a single-mechanism drug combined with an inducing AED is withdrawn as the plasma concentration of the
AED known to possess multiple mechanisms of action [113]. previously induced drug may rise, leading to toxicity.
Combinations of drugs that selectively target sodium channels appear Some AEDs have the potential to inhibit drug-metabolizing enzymes,
to offer only additive improvements in efficacy and often supra- thereby increasing the plasma concentration of concomitant medications
additive enhancement of neurotoxicity [118]. Unfortunately, because that are substrates of those enzymes. Among these interactions, those
of variability in experimental methodology and problems comparing
results across multiple studies, it is difficult to conduct an objective
assessment of the available data. Many published investigations are Table 2
also incomplete: neurotoxicity testing is often not reported, which Antiepileptic drugs for which clearance is enhanced by concomitant administration of
undermines any suggestion of synergism in terms of efficacy, and carbamazepine, phenytoin, and barbiturates.

pharmacokinetic analysis is rarely performed, which is necessary to Benzodiazepines


rule out the potential for altered drug absorption, clearance, or brain Carbamazepine
penetration. Ethosuximide
Felbamate
Lamotrigine
Levetiracetam
6. What is the relevance of pharmacokinetics when combining Oxcarbazepinea
antiepileptic drugs? Primidone
Rufinamide
Tiagabine
When two or more AEDs are used together, pharmacokinetic Topiramate
interactions may influence their combined effects and result in Valproic acid
alterations in the absorption, distribution, or elimination of the Zonisamide
affected drug, influencing the concentration of the active molecule(s) a
Interaction leads to decreased plasma concentration of the active monohydroxy-
at the site of action. An understanding of these interactions allows a lated metabolite, for which oxcarbazepine is a prodrug.
336 M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341

occurring most commonly in combination AED therapy include Among the 13 patients who did not respond to the consecutive
inhibition of the metabolism of lamotrigine and phenobarbital by addition of valproic acid or lamotrigine to their existing regimen, four
valproic acid [125]. These interactions usually require a reduction in became seizure free and an additional four experienced N50% seizure
the dosage of the affected drug to avoid the risk of toxicity. In the case of reductions when both drugs were given in combination despite lower
lamotrigine, a slower than normal dose titration is also required to doses and lower plasma drug concentrations than occurred during
minimize the risk of idiosyncratic toxicity when treatment is initiated in their separate administration.
patients who are taking valproic acid [129].
Most drug–drug interactions can be predicted from knowledge of 7.2. Other combinations
the effect of the added agent on the CYP and/or UGT enzymes
responsible for the metabolism of the concomitant medications [130]; Other recommended combinations are based largely on anecdotal
such information is now largely retrievable online (http://www. reports in small groups of patients or studies with modest sample
druginteractioninfo.org/, http://www.micromedex.com/products/ sizes. These include valproic acid with ethosuximide for absence
drugdex/, http://www.pdr.net/, http://www.pubmed.gov/). seizures [87], phenobarbital with phenytoin for GTC seizures [133],
carbamazepine with valproic acid or vigabatrin for focal seizures
7. Is there clinical evidence that mechanism of action matters [134], vigabatrin with tiagabine for focal seizures [135], and
when combinations of antiepileptic drugs are used? lamotrigine with topiramate for a range of seizure types [136].
Although none of these reports can be regarded as any more than
It seems reasonable to use a drug possessing a different mechanism observational evidence in support of the mechanistic hypothesis, it is
of action when an AED fails as a result of poor tolerability or, perhaps interesting to note that combinations with different modes of action
more persuasively, lack of efficacy [93]. There are arguments in the were universally employed in these studies [137].
literature for combining a sodium channel blocker with a drug
possessing GABAergic properties [113] or one known to have multiple 7.3. Negative combinations
mechanisms of action [131], but what hard clinical evidence do we have
in support of rational polytherapy based on combining drugs with There are now multiple drugs (phenytoin, carbamazepine,
different mechanisms of action? oxcarbazepine, lamotrigine, lacosamide, eslicarbazepine acetate)
that act primarily by blocking voltage-gated sodium channels. Most
7.1. Valproic acid with lamotrigine of these were licensed following positive adjunctive, dose-ranging,
placebo-controlled regulatory trials, many with patients already
The best evidence for a synergistic interaction between AEDs is established on another sodium blocker. Subanalyses have often failed
with valproic acid and lamotrigine for focal-onset and generalized to show a difference in response between patients already taking or
seizures. Brodie and his European colleagues undertook a pragmatic not taking sodium channel blockers, but the details are not widely
trial during which an attempt was made to substitute lamotrigine as available and such post hoc analyses are not always able to generate
monotherapy in patients suboptimally controlled with carbamaze- useful information about combination strategies.
pine, phenytoin, or valproic acid [63]. Patients were initially stabilized Lacosamide appears to be pharmacologically distinct from the
on combination therapy, and adjustment was made in the lamotrigine other drugs in that it influences the slow- rather than fast-inactivated
dosing schedules for the well-known pharmacokinetic interactions state of sodium channels [138]. This drug can be used successfully in
among these drugs, resulting in similar plasma lamotrigine concen- patients established on traditional sodium channel blockers (carba-
trations in all three treatment groups. When these patients were mazepine, lamotrigine, oxcarbazepine, and phenytoin derivatives)
established on both drugs, it was noted that responder rates were [64]. However, although no formal comparisons were performed
significantly higher for the valproic acid group than for the patients between subgroups of patients taking or not taking traditional sodium
taking lamotrigine with carbamazepine or phenytoin (Fig. 1). Fol- channel blockers, a post hoc analysis of these data suggested that
lowing up on this observation, Pisani and colleagues performed a tolerability profiles and responder rates may be better when
well-designed crossover study in 20 patients with focal seizures [132]. lacosamide is combined with AEDs other than traditional sodium
channel blockers [64].
Another interesting compound is rufinamide, which is licensed for
70 use in Lennox–Gastaut syndrome [57], but not for focal seizures because
64% the trial results for the latter indication were disappointing [139,140]. In
60 a double-blind, placebo-controlled, randomized, parallel-group, multi-
center trial of rufinamide 1600 mg twice daily in adults and adolescents
50 with refractory focal seizures, there was a 12% reduction in monthly
*
seizure frequency in the 96 patients established on carbamazepine
Percentage

41% *
40 38% compared with a 29% seizure reduction (P = 0.05) in the 60 patients
taking a regimen that did not contain carbamazepine [139].
30
There is additional evidence that combining two drugs that block
voltage-dependent sodium channels is more likely to produce neuro-
20
toxic side effects, such as dizziness, diplopia, and ataxia [61–63,141].
Similarly, a dose-dependent increase in dizziness was demonstrated
10
with lacosamide in patients already established on traditional sodium
0 blockers [64].
LTG+VPA LTG+CBZ LTG+PHT
(n=115) (n=129) (n=92) 7.4. Combination strategy
*p<0.001 VPA vs CBZ and PHT

Drug-resistant epilepsy has recently been defined by an Interna-


Fig. 1. Responder rates in patients with uncontrolled epilepsy established on
duotherapy following the addition of lamotrigine (LTG) to monotherapy with valproic
tional League Against Epilepsy Task Force as “failure of adequate trials
acid (VPA), carbamazepine (CBZ), or phenytoin (PHT). Data taken from Brodie and of two tolerated, appropriately chosen and used, antiepileptic drug
Yuen [63]. schedules (whether as monotherapy or in combination) to achieve
M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341 337

sustained seizure freedom” [142]. Thus, drug-resistant epilepsy can quences of epilepsy and designing drugs to manipulate, circumvent,
now be readily recognized, sometimes within 1 year of the diagnosis or overcome those insidious processes should lead to identification of
being made. This should allow earlier referral to specialist centers, and more effective therapies for adults with refractory epilepsy, children
may be used to justify initiation of combination therapy and, most suffering from catastrophic seizures, and perhaps even individuals
importantly, early evaluation of nonpharmacological treatment with a predisposition toward the development of epilepsy. A selection
options and, specifically, feasibility of epilepsy surgery [143]. of emerging targets and strategies for new AED development are
It is increasingly considered appropriate to treat patients estab- briefly described below.
lished on a sodium channel blocker with a drug or drugs that possess
different mechanisms of action, such as levetiracetam and pregabalin, 8.1. Voltage-gated sodium channel subtypes
or drugs that have multiple mechanisms of action, such as valproic
acid, levetiracetam, topiramate, and zonisamide, which may be added Blockade of voltage-gated sodium channels is a mechanism shared
in the hope of producing a beneficial effect in drug-resistant epilepsy by several existing AEDs, but current drugs are relatively nonselective
by focusing on multiple pharmacological targets [93,143]. Drugs with and do not exploit individual characteristics of the channels in the
multiple mechanisms of action also tend to have a broad efficacy brain. These channels are differentially expressed during develop-
spectrum across a range of seizure types [144]. Thus, if a patient ment, at different sites on neuronal membranes, and on different
tolerates the first or second drug well with a useful but suboptimal neuronal populations [155], with distinctive conductance properties
response, combination therapy could be considered, particularly if and multiple biophysical states. In addition, it is increasingly
there is a high seizure density and demonstrable underlying recognized that seizures can influence the expression of sodium
pathology, such as mesial temporal sclerosis or cortical dysplasia channel subtypes and/or alter their structural conformation [156].
[145]. Several duotherapy combinations should be tested before Designing a subtype-selective sodium channel blocker might offer
considering the addition of a third drug. Larger numbers of drugs advantages over existing AEDs. For example, a selective NaV1.6
should be avoided as it is unlikely that this strategy will produce a blocker should inhibit both transient and persistent sodium currents
useful seizure reduction [144]. in excitatory neurons while sparing inhibitory interneurons, which
There are reasons for combining AEDs other than seeking express predominantly the NaV1.1 subtype. Other compounds might
synergistic or adjunctive efficacy in controlling seizures. It has been be designed to specifically target sodium channel subtypes and
suggested that combinations of two drugs at low dosage may be conformations known to be upregulated in epileptic tissue, thus
better tolerated and, therefore, more effective than a high dose of a leaving normal neuronal function unperturbed. A novel sodium
single agent [146], although there is no sound evidence that this is channel blocker with unique characteristics in terms of selectivity
consistently the case. Also, an additional AED may be used not just to for a specific subunit or biophysical state (cf. effects of lacosamide on
treat the epilepsy, but to benefit comorbid conditions such as slow inactivation [28]) might represent a significant improvement on
neuropathic pain (e.g., gabapentin, pregabalin); trigeminal neuralgia an already successful pharmacological strategy.
(e.g., carbamazepine, oxcarbazepine); migraine (e.g., topiramate,
valproic acid); bipolar disorder (e.g., lamotrigine, valproic acid); and 8.2. GABAA receptor subunits
anxiety (e.g., pregabalin, clobazam) [147,148]. There is increasing
interest and knowledge concerning the common mechanisms of The GABAA receptor is a ligand-gated ion channel comprising five
action responsible for the broader range of therapeutic effects independent protein subunits arranged around a central anion pore
possessed by many AEDs [149,150]. that is permeable to chloride and bicarbonate ions. Nineteen GABAA
A personalized treatment plan should be formulated once a patient receptor subunits have been identified to date, and subunit compo-
fulfills the criteria for drug-resistant epilepsy to limit cognitive sition affects the physiology and pharmacology of the receptor [157].
deterioration and psychosocial dysfunction, while taking into consid- Seizures can influence the subunit composition of the GABAA receptor,
eration seizure type(s) and syndrome classification [145]. Special with a consequent effect on the biophysical properties of the
attention should be paid to drug load to avoid adverse effects [151], receptor–channel complex and altered sensitivity to pharmacological
even though a recent large observational study did not provide manipulation [158]. Current GABAA receptor drugs, such as the
evidence for adverse effects being associated with higher drug loads barbiturates, are relatively nonselective. The development of subunit-
[152]. Combinations using modest doses of each AED have a higher specific drugs could potentially avoid undesirable effects associated
chance of being tolerated and, therefore, effective [145]. At this stage, with nonselective GABAA modulation and might allow targeting of
it would also be worthwhile to raise the possibility that seizure drugs to channels in epileptic neurons or at extrasynaptic sites. There
freedom may not be attainable, paving the way for a palliative is already a precedence for subunit-specific AEDs: stiripentol
strategy if this proves to be necessary [153]. preferentially activates α3-β3-γ2-containing receptors [106], and
other such compounds are in development. ELB139, for example, is a
8. What are the emerging targets and novel strategies for future specific α3 agonist that is anxiolytic and anticonvulsant, but not
treatment? sedative [159], and neurosteroid analogs (such as ganaxolone)
selectively target receptors containing δ subunits [160]. Selective
Identifying a new generation of AEDs that better serve patients GABAA compounds, targeted to subunits known to be overexpressed
with uncontrolled seizures may necessitate a paradigm-shift in drug in epileptic brain tissue, may prove effective against seizures, with
development strategies. Indiscriminate screening of compound limited adverse effects.
libraries should be superseded by rational drug design based on
recently acquired knowledge of the pathophysiological mechanisms 8.3. GABA transporters
of ictogenesis and the genetics of the epilepsies. In this way, novel
compounds could be specifically targeted to known defects in the On release from the presynaptic terminal, GABA is actively
epileptic brain, providing direct modulation of causative pathways removed from the synaptic and extrasynaptic space by various
while sparing normal neuronal function. There also needs to be GABA transporters (GATs). Tiagabine is thought to exert its anti-
greater utilization of model systems that display one or more seizure effect by blocking GAT1-mediated reuptake into neurons and
attributes of human pharmacoresistance and an integration of models astrocytes [31]. With the exception of tiagabine, none of the currently
of epileptogenesis to help identify potential disease-modifying available AEDs targets the other GATs, betain-GAT1, GAT2, and GAT3.
therapies [154]. Understanding the molecular causes and conse- Given their broad cellular (neuronal and astrocytic) and anatomical
338 M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341

(synaptic and extrasynaptic) distribution in the central nervous controversial. Lamotrigine activates Ih in the dendrites of hippocam-
system, coupled with the diverse stoichiometry of extrasynaptic pal pyramidal cells [30], whereas ZD-7288, a putative HCN blocker,
versus synaptic GABAA receptors, non-GAT1 GATs have the potential has been shown to inhibit epileptiform activity in hippocampal slices
to exert control over neuronal excitability and could potentially [176]. Dissecting the anatomical, cellular, and subcellular localizations
offer some unique advantages in efficacy and tolerability over and the functions of individual HCN channels and their relative
tiagabine [161]. contributions to overall neuronal excitability is required before their
full potential as AED targets can be realized.
8.4. Potassium channels
8.7. Overcoming pharmacoresistance
Potassium channels are the most heterogeneous of all brain-
expressed ion channel families and arguably represent one of the last Two major hypotheses to explain pharmacoresistance in epilepsy
major unexplored targets in neuropharmacology [162]. Voltage-gated have arisen in the past decade; the so-called “transporter hypothesis”
potassium channels (of which more than 40 are known) display a and the “target hypothesis.” The latter proposes that traditional AED
multiplicity of functions associated with membrane hyperpolarization targets are disrupted in epileptic brain tissue to the extent that their
including: (1) regulation of the resting potential; (2) repolariation of sensitivity to pharmacological manipulation is lost [177]. Overcoming
the membrane after action potential generation; and (3) determina- this situation would require the development of compounds that
tion of action potential duration [163]. Mutations in the genes selectively target those proteins (or subunits thereof) that are
encoding KV7.2 and KV7.3 channels have been observed in inherited differentially expressed or disrupted in the epileptic brain. The
forms of human epilepsy, confirming the role of voltage-gated “transporter hypothesis” offers a more immediate opportunity for
potassium channels in neuronal excitability and their potential as therapeutic intervention. It is based on the premise that drug efflux
targets for AEDs [164]. Indeed, the successful development of transporters are upregulated in the blood–brain barrier at epileptic
retigabine, an activator of KV7.2 and KV7.3 channels [165], un- foci, thereby preventing AEDs from reaching their intended site of action
derscores the importance of the KV7 family as a molecular target [177]. In theory, this could be circumvented by co-administration of
worthy of pharmacological manipulation. Efforts to develop further efflux transport inhibitors (currently being developed in oncology),
potassium channel activators (such as those targeting KV1–KV4 which would restore the brain penetration capability of AEDs. An
subunits, which carry the delayed rectifier current) are justified on alternative approach might be the development of focal drug delivery
the basis of current experimental and clinical data [166]. systems that bypass the blood–brain barrier.

8.5. Gap junctions 8.8. Disease modification and antiepileptogenesis

Gap junctions provide a mechanism for rapid, nonsynaptic Available evidence suggests that current AEDs are antiseizure and
communication between adjacent neurons and astrocytes [167]. not antiepileptogenic; they control the symptoms but not the initial
They are believed to underlie the synchronization of epileptiform development or subsequent progression of the epilepsy [178]. The
activity and the generation of high-frequency oscillations which ultimate aim for the treatment of epilepsy should be the identification
precede the onset of ictal discharges in temporal lobe epilepsies [168]. of a therapeutic agent that will halt or reverse the neurological and
Gap junctions are formed from two hemichannels (one in each cell), neuropsychological decline associated with chronic seizures or,
each comprising six connexin proteins with a central pore permeable perhaps, even prevent epilepsy in “at risk” individuals. This concept
to ions and small molecules [169]. Eleven connexins have been represents an enormous challenge to both preclinical and clinical
identified in brain tissue, with differential expression depending on researchers, but recent advances in our understanding of epilepsy at
the cell type and stage of development. Connexin 43 is the major the genetic, molecular, cellular, and network levels suggest that this
isoform in astrocytes, whereas connexin 36 and connexin 45 aspiration may eventually become a reality. Some of the therapeutic
predominate in hippocampal and neocortical neurons [170]. The targets and strategies that have already attracted attention as
11β-hydroxysteroid dehydrogenase inhibitor carbenoxolone blocks potential disease modifiers are listed in Table 3. In the postgenomic
gap junctions at high concentrations. It reduces epileptiform era and with new integrative systems biology approaches to
discharges in hippocampal slices, and is effective in the tetanus biomedical science, tackling the causes of epilepsy and not simply
toxin model of temporal lobe epilepsy and against experimental one of its consequences (i.e., seizures) may be within reach.
absence seizures in WAG/Rij rats and lethargic mice [171,172].
Tonabersat is a gap junction blocker currently in early-stage Conflict of interest statement
development for epilepsy. Identification of further gap junction
blockers with selectivity for individual brain-expressed connexins is M.J. Brodie serves on the scientific advisory boards of Pfizer, UCB
warranted. Pharma, Eisai Pharmaceuticals, GlaxoSmithKline, Novartis, Valeant
Pharmaceuticals, ICVRx, and Medtronic. He has accepted honoraria for
8.6. HCN channels

HCN channels contribute to neuronal pacemaker currents, such as Table 3


Ih, and comprise four subunits (HCN1–4) arranged in homomeric or Novel targets and emerging strategies to modify disease progression and prevent
heteromeric tetramers around a central pore that is permeable to epileptogenesis.
sodium and potassium [173]. Differential expression of HCN subunits Mammalian target of rapamycin (mTOR)
in the brain (HCN1 in hippocampus and cortex, HCN2 in thalamus) Inflammatory modulators (cf. interleukins, high-mobility group protein
makes them an attractive target, with the potential for HCN1 agents to 1 [HMGB1])
Neuropeptide modulators (cf. galanin, neuropeptide Y)
be effective in limbic and neocortical epilepsies and HCN2 agents in
Gene therapy (cf. galanin, neuropeptide Y)
absence seizures [166]. Changes in HCN subunit expression and in Ih Gene silencing techniques (i.e., small interfering RNA [siRNA])
currents have been observed in models of absence seizures and Stem cell therapy (cf. medial ganglionic eminence [MGE] precursors in KV1.1
epileptogenesis, and HCN2 knockout mice have an absence-like knockout mice)
phenotype [174,175]. Evidence to support the pharmacological Modulators of growth factors (i.e., brain-derived neurotrophic factor
[BDNF]) or transcription factors (i.e., neuron-restrictive silencer factor [NRSF])
manipulation of HCN channels as an antiseizure mechanism remains
M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341 339

speaking on behalf of Pfizer, UCB Pharma, Eisai Pharmaceuticals, [7] Berg AT, Shinnar S, Levy SR, Testa FM. Status epilepticus in children with newly
diagnosed epilepsy. Ann Neurol 1999;45:618–23.
GlaxoSmithKline, and Valeant Pharmaceuticals, and has received [8] Geerts A, Arts WF, Stroink H, et al. Course and outcome of childhood epilepsy: a 15-
research grants from Pfizer, UCB Pharma, and Eisai Pharmaceuticals year follow-up of the Dutch Study of Epilepsy in Childhood. Epilepsia 2010;51:
within the past 3 years. 1189–97.
[9] Sillanpaa M, Schmidt D. Early seizure frequency and aetiology predict long-term
A. Covanis has received speaker/consultancy fees from medical outcome in childhood-onset epilepsy. Brain 2009;132:989–98.
GlaxoSmithKline, Actelion, and Biocodex. He does not have any [10] Kwan P, Schachter SC, Brodie MJ. Drug-resistant epilepsy. N Engl J Med, in press.
other professional or personal relationship with pharmaceutical [11] Krymchantowski AV, Bigal ME. Polytherapy in the preventive and acute
treatment of migraine: fundamentals for changing the approach. Expert Rev
companies.
Neurother 2006;6:283–9.
A. Gil-Nagel serves on the scientific advisory boards of Bial, Eisai [12] Dworkin RH, O'Connor AB, Audette J, et al. Recommendations for the
Pharmaceuticals, GlaxoSmithKline, and UCB Pharma, and has received pharmacological management of neuropathic pain: an overview and literature
update. Mayo Clin Proc 2010;85:S3–S14.
honoraria for Grand Rounds and Symposia from Bial, Eisai Pharma-
[13] Epstein BJ. Improving blood pressure control rates by optimizing combination
ceuticals, GlaxoSmithKline, Sepracor, and UCB Pharma. He serves as a antihypertensive therapy. Expert Opin Pharmacother 2010;11:2011–26.
consultant to Bial, Eisai Pharmaceuticals, GlaxoSmithKline, Medtronic, [14] Thompson MA, Aberg JA, Cahn P, et al. Antiretroviral treatment of adult HIV
UCB Pharma, and Valeant Pharmaceuticals, and has received research infection: 2010 recommendations of the International AIDS Society—USA panel.
JAMA 2010;304:321–33.
support from UCB Pharma. [15] Cheung MC, Haynes AE, Meyer RM, Stevens A, Imrie KR. Rituximab in lymphoma:
H. Lerche has received speaker/consultancy fees, and/or travel a systematic review and consensus practice guideline from Cancer Care Ontario.
funding from Desitin Pharmaceuticals, GlaxoSmithKline, Eisai Inc., Cancer Treat Rev 2007;33:161–76.
[16] Engstrom PF. Systemic therapy for advanced or metastatic colorectal cancer:
Pfizer, UCB Pharma, and Valeant Pharmaceuticals. He has received National Comprehensive Cancer Network guidelines for combining anti-vascular
research support from Sanofi–Aventis and UCB Pharma. endothelial growth factor and anti-epidermal growth factor receptor monoclonal
E. Perucca has received speaker/consultancy fees and/or research antibodies with chemotherapy. Pharmacotherapy 2008;28:18-22S.
[17] Meisler MH, Kearney JA. Sodium channel mutations in epilepsy and other
grants from Bial, Eisai Pharmaceuticals, GlaxoSmithKline, Johnson and neurological disorders. J Clin Invest 2005;115:2010–7.
Johnson, Novartis, Pfizer Inc., Sanofi–Aventis, Sepracor, SK Holdings, [18] Catterall WA, Kalume F, Oakley JC. NaV1.1 channels and epilepsy. J Physiol
UCB Pharma, and Valeant Pharmaceuticals. He has also received 2010;588:1849–59.
[19] Ogiwara I, Miyamoto H, Morita N, et al. Na(v)1.1 localizes to axons of
research grants from the European Union, the Italian Medicines
parvalbumin-positive inhibitory interneurons: a circuit basis for epileptic
Agency (AIFA), the Italian Ministry of Health, and the Italian Ministry seizures in mice carrying an Scn1a gene mutation. J Neurosci 2007;27:5903–14.
for University and Scientific Research. [20] Yu FH, Mantegazza M, Westenbroek RE, et al. Reduced sodium current in
GABAergic interneurons in a mouse model of severe myoclonic epilepsy in
G.J. Sills has received speaker/consultancy fees and support
infancy. Nat Neurosci 2006;9:1142–9.
for conference attendance from Alapis, Eisai Pharmaceuticals, [21] Guerrini R, Dravet C, Genton P, Belmonte A, Kaminska A, Dulac O. Lamotrigine
GlaxoSmithKline, Janssen–Cilag Ltd, Pfizer, and UCB Pharma. and seizure aggravation in severe myoclonic epilepsy. Epilepsia 1998;39:508–12.
H. Steve White has served as a paid consultant to Johnson and [22] Liao Y, Deprez L, Maljevic S, et al. Molecular correlates of age-dependent seizures
in an inherited neonatal-infantile epilepsy. Brain 2010;133:1403–14.
Johnson Pharmaceutical Research and Development, GlaxoSmithKline, [23] Vacher H, Mohapatra DP, Trimmer JS. Localization and targeting of voltage-
Valeant Pharmaceuticals, Eli Lilly & Company, and Upsher–Smith dependent ion channels in mammalian central neurons. Physiol Rev 2008;88:
Laboratories, Inc. He is a member of the UCB Pharma Speakers Bureau 1407–47.
[24] Maljevic S, Wuttke TV, Seebohm G, Lerche H. KV7 channelopathies. Pfluegers
and a member of the NeuroTherapeutics Scientific Advisory Board, and Arch 2010;460:277–88.
has received research funding from NeuroAdjuvants, Inc. He is also one [25] Zuberi SM, Eunson LH, Spauschus A, et al. A novel mutation in the human
of two scientific co-founders of NeuroAdjuvants, Inc., Salt Lake City, UT, voltage-gated potassium channel gene (Kv1.1) associates with episodic ataxia
type 1 and sometimes with partial epilepsy. Brain 1999;122(Pt. 5):817–25.
USA. [26] Du W, Bautista JF, Yang H, et al. Calcium-sensitive potassium channelopathy in
human epilepsy and paroxysmal movement disorder. Nat Genet 2005;37:733–8.
Acknowledgments [27] Bernard C, Anderson A, Becker A, Poolos NP, Beck H, Johnston D. Acquired
dendritic channelopathy in temporal lobe epilepsy. Science 2004;305:532–5.
[28] Errington AC, Stohr T, Heers C, Lees G. The investigational anticonvulsant
The scientific meeting on which this article is based (Antiepileptic lacosamide selectively enhances slow inactivation of voltage-gated sodium
Drug Therapy: Does Mechanism of Action Matter? 7 June 2010, channels. Mol Pharmacol 2008;73:157–69.
[29] Kito M, Maehara M, Watanabe K. Antiepileptic drugs: calcium current interaction
London, UK) was initiated and supported by GlaxoSmithKline (GSK). in cultured human neuroblastoma cells. Seizure 1994;3:141–9.
Members of the Scientific Committee who developed the meeting [30] Poolos NP, Migliore M, Johnston D. Pharmacological upregulation of H-channels
program were selected by the Chairman, Professor Martin Brodie, in reduces the excitability of pyramidal neuron dendrites. Nat Neurosci 2002;5:767–74.
[31] Kwan P, Sills GJ, Brodie MJ. The mechanisms of action of commonly used
consultation with GSK. Scientific Committee members and presenters
antiepileptic drugs. Pharmacol Ther 2001;90:21–34.
who were not members of the Scientific Committee were paid an [32] Stephen LJ, Brodie MJ. Pharmacotherapy of epilepsy: newly approved and
honorarium for their participation in the meeting. Editorial support in developmental agents. CNS Drugs 2011;25:89–107.
the form of editing and formatting of the article, collation of author [33] Rundfeldt C, Netzer R. The novel anticonvulsant retigabine activates M-currents
in Chinese hamster ovary-cells tranfected with human KCNQ2/3 subunits.
comments, and coordination of submission requirements was Neurosci Lett 2000;282:73–6.
provided by Janet M. Brandon of Caudex Medical Ltd. and was funded [34] Wuttke TV, Seebohm G, Bail S, Maljevic S, Lerche H. The new anticonvulsant
by GSK. A draft of the article was reviewed by GSK for factual accuracy. retigabine favors voltage-dependent opening of the Kv7.2 (KCNQ2) channel by
binding to its activation gate. Mol Pharmacol 2005;67:1009–17.
[35] Delmas P, Brown DA. Pathways modulating neural KCNQ/M (Kv7) potassium
References channels. Nat Rev Neurosci 2005;6:850–62.
[36] Main MJ, Cryan JE, Dupere JR, Cox B, Clare JJ, Burbidge SA. Modulation of KCNQ2/3
[1] Chong DJ, Bazil CW. Update on anticonvulsant drugs. Curr Neurol Neurosci Rep potassium channels by the novel anticonvulsant retigabine. Mol Pharmacol
2010;10:308–18. 2000;58:253–62.
[2] Brodie MJ, Bamagous G, Kwan P. Improved outcomes in newly diagnosed [37] Brodie MJ, Lerche H, Gil-Nagel A, et al. Efficacy and safety of adjunctive ezogabine
epilepsy. Epilepsia 2009;50:411–2. (retigabine) in refractory partial epilepsy. Neurology 2010;75:1817–24.
[3] Callaghan BC, Anand K, Hesdorffer D, Hauser WA, French JA. Likelihood of seizure [38] French JA, Abou-Khalil BW, Leroy R, on behalf of the RESTORE 1/Study 301
remission in an adult population with refractory epilepsy. Ann Neurol 2007;62: investigators. Randomized double-blind placebo-controlled trial of ezogabine
382–9. (retigabine) in partial epilepsy. Neurology 2011;76:1555–63.
[4] Luciano AL, Shorvon SD. Results of treatment changes in patients with apparently [39] Rogawski MA, Loscher W. The neurobiology of antiepileptic drugs. Nat Rev
drug-resistant chronic epilepsy. Ann Neurol 2007;62:375–81. Neurosci 2004;5:553–64.
[5] Beyenburg S, Stavem K, Schmidt D. Placebo-corrected efficacy of modern [40] Löscher W, Schmidt D. Which animal models should be used in the search for
antiepileptic drugs for refractory epilepsy: systematic review and meta-analysis. new antiepileptic drugs? A proposal based on experimental and clinical
Epilepsia 2010;51:7–26. considerations. Epilepsy Res 1988;2:145–81.
[6] Gazzolla DM, Balcer LJ, French JA. Seizure-free outcome in randomized add-on [41] Bialer M, White HS. Key factors in the discovery and development of new
trials of the new antiepileptic drugs. Epilepsia 2007;48:1303–7. antiepileptic drugs. Nat Rev Drug Discov 2010;9:68–82.
340 M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341

[42] White HS, Woodhead JH, Wilcox KS, Stables JP, Kupferberg HJ, Wolf HH. [77] Covanis A, Gupta AK, Jeavons PM. Sodium valproate: monotherapy and
Discovery and preclinical development of antiepileptic drugs. In: Levy RH, polytherapy. Epilepsia 1982;23:693–720.
Mattson RH, Meldrum BS, Perucca E, editors. Antiepileptic drugs. Philadelphia: [78] Jentink J, Loane MA, Dolk H, et al. Valproic acid monotherapy in pregnancy and
Lippincott Williams & Williams; 2002. p. 36–48. major congenital malformations. N Engl J Med 2010;362:2185–93.
[43] Stables JP, Bertram E, Dudek FE, et al. Therapy discovery for pharmacoresistant [79] Tomson T, Battino D. Teratogenic effects of antiepileptic medications. Neurol Clin
epilepsy and for disease-modifying therapeutics: summary of the NIH/NINDS/ 2009;27:993–1002.
AES Models II Workshop. Epilepsia 2003;44:1472–8. [80] Banach R, Boskovic R, Einarson T, Koren G. Long-term developmental outcome of
[44] McNamara JO. Development of new pharmacological agents for epilepsy: lessons children of women with epilepsy, unexposed or exposed prenatally to
from the kindling model. Epilepsia 1989;30(Suppl. 1):S13–8. antiepileptic drugs: a meta-analysis of cohort studies. Drug Saf 2010;33:73–9.
[45] Sills GJ, Brodie MJ. Preclinical drug development in epilepsy. In: Schwartzkroin [81] Bromley RL, Mawer G, Love J, et al. Early cognitive development in children born
PA, editor. Encyclopedia of basic epilepsy research. London: Elsevier; 2009. to women with epilepsy: a prospective report. Epilepsia 2010;51:2058–65.
p. 97–103. [82] Meador KJ, Baker GA, Browning N, et al. Cognitive function at 3 years of age after
[46] Kupferberg HJ, Schmutz M. Screening of new compounds and the role of the fetal exposure to antiepileptic drugs. N Engl J Med 2009;360:1597–605.
pharmaceutical industry. In: Engel J, Pedley TA, editors. Epilepsy: a comprehen- [83] Chung AM, Eiland LS. Use of second-generation antiepileptic drugs in the
sive textbook. Philadelphia: Lippincott–Raven; 1997. p. 1417–34. pediatric population. Paediatr Drugs 2008;10:217–54.
[47] Berkovic SF, Knowlton RC, Leroy RF, Schiemann J, Falter U. Placebo-controlled study [84] Crespel A, Genton P, Berramdane M, et al. Lamotrigine associated with
of levetiracetam in idiopathic generalized epilepsy. Neurology 2007;69:1751–60. exacerbation or de novo myoclonus in idiopathic generalized epilepsies.
[48] Krauss GL, Bergin A, Kramer RE, Cho YW, Reich SG. Suppression of post-hypoxic Neurology 2005;65:762–4.
and post-encephalitic myoclonus with levetiracetam. Neurology 2001;56:411–2. [85] Sharpe DV, Patel AD, Abou-Khalil B, Fenichel GM. Levetiracetam monotherapy in
[49] Noachtar S, Andermann E, Meyvisch P, Andermann F, Gough WB, Schiemann-Delgado juvenile myoclonic epilepsy. Seizure 2008;17:64–8.
J. Levetiracetam for the treatment of idiopathic generalized epilepsy with myoclonic [86] Covanis A, Skiadas K, Loli N, Lada C, Theodorou V. Absence epilepsy: early
seizures. Neurology 2008;70:607–16. prognostic signs. Seizure 1992;1:281–9.
[50] Perucca E, Gram L, Avanzini G, Dulac O. Antiepileptic drugs as a cause of [87] Rowan AJ, Meijer JW, de Beer-Pawlikowski N, van der Geest P, Meinardi H.
worsening seizures. Epilepsia 1998;39:5–17. Valproate–ethosuximide combination therapy for refractory absence seizures.
[51] Smith M, Wilcox KS, White HS. Discovery of antiepileptic drugs. Neurotherapeutics Arch Neurol 1983;40:797–802.
2007;4:12–7. [88] Modur PN, Milteer WE. Adjunctive pregabalin therapy in mentally retarded,
[52] Glauser TA, Cnaan A, Shinnar S, et al. Ethosuximide, valproic acid, and developmentally delayed patients with epilepsy. Epilepsy Behav 2008;13:554–6.
lamotrigine in childhood absence epilepsy. N Engl J Med 2010;362:790–9. [89] Sazgar M, Bourgeois BF. Aggravation of epilepsy by antiepileptic drugs. Pediatr
[53] Schmidt D, Holmes GL. Commentary: novel delivery approaches for antiepileptic Neurol 2005;33:227–34.
drugs: hope and hurdles. Neurotherapeutics 2009;6:381–2. [90] Kellinghaus C, Dziewas R, Ludemann P. Tiagabine-related non-convulsive status
[54] Brodie MJ, Perucca E, Ryvlin P, Ben-Menachem E, Meencke HJ. Comparison of epilepticus in partial epilepsy: three case reports and a review of the literature.
levetiracetam and controlled-release carbamazepine in newly diagnosed Seizure 2002;11:243–9.
epilepsy. Neurology 2007;68:402–8. [91] Guberman AH, Besag FM, Brodie MJ, et al. Lamotrigine-associated rash: risk/
[55] Marson AG, Al-Kharusi AM, Alwaidh M, et al. The SANAD study of effectiveness of benefit considerations in adults and children. Epilepsia 1999;40:985–91.
carbamazepine, gabapentin, lamotrigine, oxcarbazepine, or topiramate for [92] Kaminski RM, Matagne A, Patsalos PN, Klitgaard H. Benefit of combination
treatment of partial epilepsy: an unblinded randomised controlled trial. Lancet therapy in epilepsy: a review of the preclinical evidence with levetiracetam.
2007;369:1000–15. Epilepsia 2009;50:387–97.
[56] Specchio LM, Gambardella A, Giallonardo AT, et al. Open label, long-term, [93] Kwan P, Brodie MJ. Combination therapy in epilepsy: when and what to use.
pragmatic study on levetiracetam in the treatment of juvenile myoclonic Drugs 2006;66:1817–29.
epilepsy. Epilepsy Res 2006;71:32–9. [94] Marescaux C, Hirsch E, Finck S, et al. Landau–Kleffner syndrome: a pharmacologic
[57] Glauser T, Kluger G, Sachdeo R, Krauss G, Perdomo C, Arroyo S. Rufinamide for study of five cases. Epilepsia 1990;31:768–77.
generalized seizures associated with Lennox–Gastaut syndrome. Neurology [95] Fejerman N, Di Blasi AM. Status epilepticus of benign partial epilepsies in
2008;70:1950–8. children: report of two cases. Epilepsia 1987;28:351–5.
[58] Elterman RD, Shields WD, Mansfield KA, Nakagawa J. Randomized trial of [96] Kikumoto K, Yoshinaga H, Oka M, et al. EEG and seizure exacerbation induced by
vigabatrin in patients with infantile spasms. Neurology 2001;57:1416–21. carbamazepine in Panayiotopoulos syndrome. Epileptic Disord 2006;8:53–6.
[59] Chiron C, Marchand MC, Tran A, et al, for the STICLO Study Group. Stiripentol in [97] Bello-Espinosa LE, Roberts SL. Levetiracetam for benign epilepsy of childhood
severe myoclonic epilepsy in infancy: a randomised placebo-controlled syndrome- with centrotemporal spikes: three cases. Seizure 2003;12:157–9.
dedicated trial. Lancet 2000;356:1638–42. [98] Coppola G, Franzoni E, Verrotti A, et al. Levetiracetam or oxcarbazepine as
[60] Toledano R, Gil-Nagel A. Adverse effects of antiepileptic drugs. Semin Neurol monotherapy in newly diagnosed benign epilepsy of childhood with centrotemporal
2008;28:317–27. spikes (BECTS): an open-label, parallel group trial. Brain Dev 2007;29:281–4.
[61] Barcs G, Walker EB, Elger CE, et al. Oxcarbazepine placebo-controlled, dose- [99] Garcia C, Rubio G. Efficacy and safety of levetiracetam in the treatment of
ranging trial in refractory partial epilepsy. Epilepsia 2000;41:1597–607. Panayiotopoulos syndrome. Epilepsy Res 2009;85:318–20.
[62] Besag FM, Berry DJ, Pool F, Newbery JE, Subel B. Carbamazepine toxicity with [100] Wheless JW, Clarke DF, Arzimanoglou A, Carpenter D. Treatment of pediatric
lamotrigine: pharmacokinetic or pharmacodynamic interaction? Epilepsia epilepsy: European expert opinion, 2007. Epileptic Disord 2007;9:353–412.
1998;39:183–7. [101] Bast T, Volp A, Wolf C, Rating D. The influence of sulthiame on EEG in children
[63] Brodie MJ, Yuen AW, for the 105 Study Group. Lamotrigine substitution study: with benign childhood epilepsy with centrotemporal spikes (BECTS). Epilepsia
evidence for synergism with sodium valproate? Epilepsy Res 1997;26:423–32. 2003;44:215–20.
[64] Saké J-K, Herbert D, Isojarvi J, et al. A pooled analysis of lacosamide clinical trial [102] Engler F, Maeder-Ingvar M, Roulet E, Deonna T. Treatment with sulthiame
data grouped by mechanisms of action of concomitant antiepileptic drugs. CNS (Ospolot) in benign partial epilepsy of childhood and related syndromes: an
Drugs 2010;24:1055–68. open clinical and EEG study. Neuropediatrics 2003;34:105–9.
[65] Holmes GL, Frank LM, Sheth RD, et al. Lamotrigine monotherapy for newly [103] Engel J. Report of the ILAE classification core group. Epilepsia 2006;47:1558–68.
diagnosed typical absence seizures in children. Epilepsy Res 2008;82:124–32. [104] Wohlrab G, Boltshauser E, Schmitt B. Vigabatrin as a first-line drug in West syndrome:
[66] Labiner DM, Ettinger AB, Fakhoury TA, et al. Effects of lamotrigine compared with clinical and electroencephalographic outcome. Neuropediatrics 1998;29:133–6.
levetiracetam on anger, hostility, and total mood in patients with partial [105] Escayg A, Goldin AL. Sodium channel SCN1A and epilepsy: mutations and
epilepsy. Epilepsia 2009;50:434–42. mechanisms. Epilepsia 2010;51:1650–8.
[67] Arcas J, Ferrer T, Roche MC, Martinez-Bermejo A, Lopez-Martin V. Hypohidrosis [106] Fisher JL. The anti-convulsant stiripentol acts directly on the GABA(A) receptor as
related to the administration of topiramate to children. Epilepsia 2001;42:1363–5. a positive allosteric modulator. Neuropharmacology 2009;56:190–7.
[68] Ohtahara S, Yamatogi Y. Safety of zonisamide therapy: prospective follow-up [107] Grosso S, Franzoni E, Iannetti P, et al. Efficacy and safety of topiramate in
survey. Seizure 2004;13(Suppl. 1):S50–5. refractory epilepsy of childhood: long-term follow-up study. J Child Neurol
[69] Ohtahara S, Yamatogi Y. Erratum to "Safety of zonisamide therapy: prospective 2005;20:893–7.
follow-up survey.". Seizure 2007;16:87–93. [108] Wirrell E, Ho AW, Hamiwka L. Sulthiame therapy for continuous spike and wave
[70] Sander JW, Hart YM. Vigabatrin and behaviour disturbances. Lancet 1990;335:57. in slow-wave sleep. Pediatr Neurol 2006;35:204–8.
[71] Marsh ED, Brooks-Kayal AR, Porter BE. Seizures and antiepileptic drugs: does [109] Reife R. Assessing pharmacokinetic and pharmacodynamic interactions in clinical
exposure alter normal brain development? Epilepsia 2006;47:1999–2010. trials of antiepileptic drugs. In: French J, Leppik I, Dichter MA, editors. Antiepileptic
[72] Cohen I, Navarro V, Clemenceau S, Baulac M, Miles R. On the origin of interictal drug development. Advances in Neurology. Philadelphia: Lippincott–Raven; 1998.
activity in human temporal lobe epilepsy in vitro. Science 2002;298:1418–21. p. 95–103.
[73] First Seizure Trial Group. Randomized clinical trial on the efficacy of antiepileptic [110] Jonker DM, Voskuyl RA, Danhof M. Synergistic combinations of anticonvulsant
drugs in reducing the risk of relapse after a first unprovoked tonic–clonic seizure. agents: what is the evidence from animal experiments? Epilepsia 2007;48:412–34.
Neurology 1993;43:478–83. [111] Tallarida RJ. An overview of drug combination analysis with isobolograms.
[74] Leone MA, Solari A, Beghi E. Treatment of the first tonic–clonic seizure does not J Pharmacol Exp Ther 2006;319:1–7.
affect long-term remission of epilepsy. Neurology 2006;67:2227–9. [112] Weaver LC, Swinyard EA, Woodbury LA, Goodman LS. Studies on anticonvulsant
[75] Musicco M, Beghi E, Solari A, Viani F, for the First Seizure Trial Group. Treatment of drug combinations: phenobarbital and diphenylhydantoin. J Pharmacol Exp Ther
first tonic-clonic seizure does not improve the prognosis of epilepsy. Neurology 1955;113:359–70.
1997;49:991–8. [113] Deckers CL, Czuczwar SJ, Hekster YA, et al. Selection of antiepileptic drug
[76] Schachter SC, LaFrance WC, eds. Gates and Rowan's: Nonepileptic seizures. Third polytherapy based on mechanisms of action: the evidence reviewed. Epilepsia
Edition. Cambridge: Cambridge University Press; 2010. 2000;41:1364–74.
M.J. Brodie et al. / Epilepsy & Behavior 21 (2011) 331–341 341

[114] Bourgeois BF. Antiepileptic drug combinations and experimental background: the [145] Brodie MJ, Kwan P. Staged approach to epilepsy management. Neurology
case of phenobarbital and phenytoin. Naunyn Schmiedebergs Arch Pharmacol 2002;58:S2–8.
1986;333:406–11. [146] Beghi E, Gatti G, Tonini C, et al. Adjunctive therapy versus alternative monotherapy
[115] Morris JC, Dodson WE, Hatlelid JM, Ferrendelli JA. Phenytoin and carbamazepine, in patients with partial epilepsy failing on a single drug: a multicentre, randomised,
alone and in combination: anticonvulsant and neurotoxic effects. Neurology pragmatic controlled trial. Epilepsy Res 2003;57:1–13.
1987;37:1111–8. [147] Ettinger AB, Argoff CE. Use of antiepileptic drugs for nonepileptic conditions:
[116] Stafstrom CE. Mechanisms of action of antiepileptic drugs: the search for psychiatric disorders and chronic pain. Neurotherapeutics 2007;4:75–83.
synergy. Curr Opin Neurol 2010;23:157–63. [148] Spina E, Perugi G. Antiepileptic drugs: indications other than epilepsy. Epileptic
[117] Czuczwar SJ, Borowicz KK. Polytherapy in epilepsy: the experimental evidence. Disord 2004;6:57–75.
Epilepsy Res 2002;52:15–23. [149] Johannessen LC, Johannessen SI. Pharmacological management of epilepsy:
[118] Luszczki JJ, Czuczwar SJ. Preclinical profile of combinations of some second- recent advances and future prospects. Drugs 2008;68:1925–39.
generation antiepileptic drugs: an isobolographic analysis. Epilepsia 2004;45: [150] Rogawski MA, Loscher W. The neurobiology of antiepileptic drugs for the
895–907. treatment of nonepileptic conditions. Nat Med 2004;10:685–92.
[119] Cuadrado A, de las Cuevas I, Valdizan EM, Armijo JA. Synergistic interaction [151] Deckers CL, Hekster YA, Keyser A, Meinardi H, Renier WO. Reappraisal of
between valproate and lamotrigine against seizures induced by 4-aminopyridine polytherapy in epilepsy: a critical review of drug load and adverse effects.
and pentylenetetrazole in mice. Eur J Pharmacol 2002;453:43–52. Epilepsia 1997;38:570–5.
[120] De Sarro G, Nava F, Aguglia U, De Sarro A. Lamotrigine potentiates the antiseizure [152] Canevini MP, De SG, Galimberti CA, et al. Relationship between adverse effects of
activity of some anticonvulsants in DBA/2 mice. Neuropharmacology 1996;35: antiepileptic drugs, number of coprescribed drugs, and drug load in a large
153–8. cohort of consecutive patients with drug-refractory epilepsy. Epilepsia 2010;51:
[121] Luszczki JJ, Czuczwar M, Kis J, et al. Interactions of lamotrigine with topiramate 797–804.
and first-generation antiepileptic drugs in the maximal electroshock test in mice: [153] Devinsky O. Patients with refractory seizures. N Engl J Med 1999;340:1565–70.
an isobolographic analysis. Epilepsia 2003;44:1003–13. [154] Stables JP, Bertram EH, White HS, et al. Models for epilepsy and epileptogenesis:
[122] Borowicz KK, Swiader M, Luszczki J, Czuczwar SJ. Effect of gabapentin on the report from the NIH workshop, Bethesda, Maryland. Epilepsia 2002;43:1410–20.
anticonvulsant activity of antiepileptic drugs against electroconvulsions in mice: [155] Mantegazza M, Curia G, Biagini G, Ragsdale DS, Avoli M. Voltage-gated sodium
an isobolographic analysis. Epilepsia 2002;43:956–63. channels as therapeutic targets in epilepsy and other neurological disorders.
[123] De Sarro G, Spagnolo C, Gareri P, Gallelli L, De Sarro A. Gabapentin potentiates the Lancet Neurol 2010;9:413–24.
antiseizure activity of certain anticonvulsants in DBA/2 mice. Eur J Pharmacol [156] Remy S, Gabriel S, Urban BW, et al. A novel mechanism underlying drug
1998;349:179–85. resistance in chronic epilepsy. Ann Neurol 2003;53:469–79.
[124] Klitgaard H, Knudsen ML, Jackson HC. Synergism between drugs with different [157] Johnston GA. GABA(A) receptor channel pharmacology. Curr Pharm Des 2005;11:
mechanisms of action against audiogenic seizures in DBA/2 mice. Epilepsia 1867–85.
1993;34:93–4. [158] Brooks-Kayal AR, Shumate MD, Jin H, Rikhter TY, Coulter DA. Selective changes in
[125] Patsalos PN, Perucca E. Clinically important drug interactions in epilepsy: general single cell GABA(A) receptor subunit expression and function in temporal lobe
features and interactions between antiepileptic drugs. Lancet Neurol 2003;2:347–56. epilepsy. Nat Med 1998;4:1166–72.
[126] Gatti G, Bartoli A, Marchiselli R, et al. Vigabatrin-induced decrease in serum [159] Langen B, Egerland U, Bernoster K, Dost R, Unverferth K, Rundfeldt C. Character-
phenytoin concentration does not involve a change in phenytoin bioavailability. ization in rats of the anxiolytic potential of ELB139 [1-(4-chlorophenyl)-4-
Br J Clin Pharmacol 1993;36:603–6. piperidin-1-yl-1,5-dihydro-imidazol-2-on], a new agonist at the benzodiazepine
[127] Mattson RH, Cramer JA, Williamson PD, Novelly RA. Valproic acid in epilepsy: binding site of the GABAA receptor. J Pharmacol Exp Ther 2005;314:717–24.
clinical and pharmacological effects. Ann Neurol 1978;3:20–5. [160] Stell BM, Brickley SG, Tang CY, Farrant M, Mody I. Neuroactive steroids reduce
[128] Perucca E. Clinically relevant drug interactions with antiepileptic drugs. Br J Clin neuronal excitability by selectively enhancing tonic inhibition mediated by delta
Pharmacol 2006;61:246–55. subunit-containing GABAA receptors. Proc Natl Acad Sci USA 2003;100:14439–44.
[129] Messenheimer J, Mullens EL, Giorgi L, Young F. Safety review of adult clinical trial [161] Madsen KK, Clausen RP, Larsson OM, Krogsgaard-Larsen P, Schousboe A, White
experience with lamotrigine. Drug Saf 1998;18:281–96. HS. Synaptic and extrasynaptic GABA transporters as targets for anti-epileptic
[130] Obach RS, Walsky RL, Venkatakrishnan K, Houston JB, Tremaine LM. In vitro drugs. J Neurochem 2009;109(Suppl. 1):139–44.
cytochrome P450 inhibition data and the prediction of drug–drug interactions: [162] Wulff H, Castle NA, Pardo LA. Voltage-gated potassium channels as therapeutic
qualitative relationships, quantitative predictions, and the rank-order approach. targets. Nat Rev Drug Discov 2009;8:982–1001.
Clin Pharmacol Ther 2005;78:582–92. [163] Benarroch EE. Potassium channels: brief overview and implications in epilepsy.
[131] Kwan P, Brodie MJ. Epilepsy after the first drug fails: substitution or add-on? Neurology 2009;72:664–9.
Seizure 2000;9:464–8. [164] Wickenden AD, Roeloffs R, McNaughton-Smith G, Rigdon GC. KCNQ potassium
[132] Pisani F, Oteri G, Russo MF, Di Perri R, Perucca E, Richens A. The efficacy of channels: drug targets for the treatment of epilepsy and pain. Expert Opin Ther
valproate–lamotrigine comedication in refractory complex partial seizures: Patents 2004;14:457–69.
evidence for a pharmacodynamic interaction. Epilepsia 1999;40:1141–6. [165] Wickenden AD, Yu W, Zou A, Jegla T, Wagoner PK. Retigabine, a novel anti-
[133] Cereghino JJ, Brock JT, Van Meter JC, Penry JK, Smith LD, White BG. The efficacy of convulsant, enhances activation of KCNQ2/Q3 potassium channels. Mol
carbamazepine combinations in epilepsy. Clin Pharmacol Ther 1975;18:733–41. Pharmacol 2000;58:591–600.
[134] Brodie MJ, Mumford JP, for the 012 Study Group. Double-blind substitution of [166] Meldrum BS, Rogawski MA. Molecular targets for antiepileptic drug develop-
vigabatrin and valproate in carbamazepine-resistant partial epilepsy. Epilepsy ment. Neurotherapeutics 2007;4:18–61.
Res 1999;34:199–205. [167] Nemani VM, Binder DK. Emerging role of gap junctions in epilepsy. Histol
[135] Leach JP, Brodie MJ. Synergism with GABAergic drugs in refractory epilepsy. Histopathol 2005;20:253–9.
Lancet 1994;343:1650. [168] Timofeev I, Steriade M. Neocortical seizures: initiation, development and
[136] Stephen LJ, Sills GJ, Brodie MJ. Lamotrigine and topiramate may be a useful cessation. Neuroscience 2004;123:299–336.
combination. Lancet 1998;351:958–9. [169] Nakase T, Naus CC. Gap junctions and neurological disorders of the central
[137] Sills GJ, Brodie MJ. Update on the mechanisms of action of antiepileptic drugs. nervous system. Biochim Biophys Acta 2004;1662:149–58.
Epileptic Disord 2001;3:165–72. [170] Venance L, Rozov A, Blatow M, Burnashev N, Feldmeyer D, Monyer H. Connexin
[138] Beydoun A, D'Souza J, Hebert D, Doty P. Lacosamide: pharmacology, mechanisms expression in electrically coupled postnatal rat brain neurons. Proc Natl Acad Sci
of action and pooled efficacy and safety data in partial-onset seizures. Expert Rev USA 2000;97:10260–5.
Neurother 2009;9:33–42. [171] Gareri P, Condorelli D, Belluardo N, et al. Antiabsence effects of carbenoxolone in
[139] Brodie MJ, Rosenfeld WE, Vazquez B, et al. Rufinamide for the adjunctive two genetic animal models of absence epilepsy (WAG/Rij rats and lh/lh mice).
treatment of partial seizures in adults and adolescents: a randomized placebo- Neuropharmacology 2005;49:551–63.
controlled trial. Epilepsia 2009;50:1899–909. [172] Nilsen KE, Kelso AR, Cock HR. Antiepileptic effect of gap-junction blockers in a rat
[140] Elger CE, Stefan H, Mann A, Narurkar M, Sun Y, Perdomo C. A 24-week model of refractory focal cortical epilepsy. Epilepsia 2006;47:1169–75.
multicenter, randomized, double-blind, parallel-group, dose-ranging study of [173] Herrmann S, Stieber J, Ludwig A. Pathophysiology of HCN channels. Pfluegers
rufinamide in adults and adolescents with inadequately controlled partial Arch 2007;454:517–22.
seizures. Epilepsy Res 2010;88:255–63. [174] Ludwig A, Budde T, Stieber J, et al. Absence epilepsy and sinus dysrhythmia in
[141] Wong IC, Mawer GE, Sander JW, Lhatoo SD. A pharmacoepidemiologic study of mice lacking the pacemaker channel HCN2. EMBO J 2003;22:216–24.
factors influencing the outcome of treatment with lamotrigine in chronic [175] Shah MM, Anderson AE, Leung V, Lin X, Johnston D. Seizure-induced plasticity of h
epilepsy. Epilepsia 2001;42:1354–8. channels in entorhinal cortical layer III pyramidal neurons. Neuron 2004;44:495–508.
[142] Kwan P, Arzimanoglou A, Berg AT, et al. Definition of drug resistant epilepsy: [176] Arias RL, Bowlby MR. Pharmacological characterization of antiepileptic drugs and
consensus proposal by the ad hoc Task Force of the ILAE Commission on experimental analgesics on low magnesium-induced hyperexcitability in rat
Therapeutic Strategies. Epilepsia 2010;51:1069–77. hippocampal slices. Brain Res 2005;1047:233–44.
[143] Kwan P, Brodie MJ. Definition of refractory epilepsy: defining the indefinable? [177] Schmidt D, Löscher W. Drug resistance in epilepsy: putative neurobiologic and
Lancet Neurol 2010;9:27–9. clinical mechanisms. Epilepsia 2005;46:858–77.
[144] Stephen LJ, Brodie MJ. Seizure freedom with more than one antiepileptic drug. [178] Walker MC, White HS, Sander JW. Disease modification in partial epilepsy. Brain
Seizure 2002;11:349–51. 2002;125:1937–50.

You might also like