SlideShare a Scribd company logo
1 of 16
Download to read offline
336  |  	 Neuropsychopharmacology Reports. 2021;41:336–351.
wileyonlinelibrary.com/journal/nppr
Received: 17 May 2021  |  Revised: 1 July 2021  |  Accepted: 6 July 2021
DOI: 10.1002/npr2.12196
R E V I E W A R T I C L E
Neuropharmacology of Antiseizure Drugs
Tahir Hakami
This is an open access article under the terms of the Creat​
ive Commo​
ns Attri​
butio​
n-­
NonCo​
mmerc​
ial-­
NoDerivs License, which permits use and distribution in
any medium, provided the original work is properly cited, the use is non-­
commercial and no modifications or adaptations are made.
© 2021 The Authors. Neuropsychopharmacology Reports published by John Wiley  Sons Australia, Ltd on behalf of The Japanese Society of
Neuropsychopharmacology.
The Faculty of Medicine, Jazan University,
Jazan, Saudi Arabia
Correspondence
Tahir Hakami, The Faculty of Medicine,
Jazan University, P.O.Box 114, Jazan 45142,
Saudi Arabia.
Email hakamit@jazanu.edu.sa
Funding information
The author received no financial support for
the research, authorship, and/or publication
of this article
Abstract
Background: Antiseizure drugs (ASDs) are the primary therapy for epilepsy, with
more than 20 drugs introduced into clinical practice to date. These drugs are typi-
cally grouped by their mechanisms of action and therapeutic spectrum. This article
aims to educate non-­
neurologists and medical students about the new frontiers in
the pharmacology of ASDs and presents the current state of the literature on the
efficacy and tolerability of these agents.
Methods: Randomized controlled trials, observational studies, and evidence-­
based
meta-­
analyses of ASD efficacy and tolerability as initial monotherapy for epileptic
seizures and syndromes were identified in PubMed, EMBASE, the Cochrane Library,
and Elsevier Clinical Pharmacology.
Results: The choice of ASD varies primarily according to the seizure type. Practical
guidelines for ASD selection in patients with new-­
onset and drug-­
resistant epilepsy
were recently published. The guidelines have shown that the newer-­
generation
drugs, which have unique mechanistic and pharmacokinetic properties, are better
tolerated but have similar efficacy compared with the older drugs. Several ASDs are
effective as first-­
line monotherapy in focal seizures, including lamotrigine, carba-
mazepine, phenytoin, levetiracetam, and zonisamide. Valproate remains the first-­
line
drug for many patients with generalized and unclassified epilepsies. However, val-
proate should be avoided, if possible, in women of childbearing potential because of
teratogenicity. Toxicity profile precludes several drugs from use as first-­
line treat-
ment, for example, vigabatrin, felbamate, and rufinamide.
Conclusions: Antiseizure drugs have different pharmacologic profiles that should be
considered when selecting and prescribing these agents for epilepsy. These include
pharmacokinetic properties, propensity for drug-­
drug interactions, and adverse
effects.
K E Y W O R D S
antiseizure drug, epilepsy, seizure types
    | 337
HAKAMI
1 | INTRODUCTION
Epilepsy is a chronic disorder of brain function characterized by
recurrent seizures.1
At any one time, about 1% of the population
requires medication to treat epilepsy.2
The antiseizure drug (ASD)
is the mainstay of treatment. The choice of ASDs varies with dif-
ferent seizure types and epileptic syndromes.3
The appropriately
chosen ASD provides adequate seizure control in 60%–­
70% of pa-
tients.4
There is no general agreement on when treatment should
start after a “first seizure,” but randomized controlled trials (RCTs)
in adult and pediatric patients have shown that early treatment re-
duces the recurrence for at least two years from the first seizure.5
The ASDs are often administered orally for a long period of time
to prevent seizure recurrence. In designing a therapeutic approach,
the use of a single drug (monotherapy) is preferred because of
fewer adverse effects. However, 30% of the patients will continue
to have uncontrolled seizures and multiple drugs are often used
simultaneously.6
Monotherapy trials of two ASDs that are appro-
priate first-­
line treatment for the patient's seizure type usually be
initiated before combinations are tried. Patients who do not achieve
seizure control following adequate trials with two or more appropri-
ate drugs are considered drug-­
resistant.6
The International League
Against Epilepsy (ILAE), in 2010, has proposed the definition of
drug-­
resistant epilepsy (often used interchangeably with “'medically
refractory”' and “'intractable”) as “failure of adequate trials of two
tolerated, appropriately chosen and used antiseizure drug schedules
(whether as monotherapies or in combination) to achieve sustained
seizure freedom.”7
The older-­
generation ASDs were introduced into clinical
practice more than four decades ago. Research has shown that
30%–­
40% of people treated with an older ASD as monotherapy
(including carbamazepine and valproate) experience adverse ef-
fects that contribute to treatment failure.8
This spurred a continu-
ing search for new ASDs with novel molecular targets that could
provide optimal care for patients with epilepsy.9,10
The past three
decades have seen the licensing of about 20 newer-­(second-­and
third-­
) generation ASDs with unique mechanisms of action and
pharmacokinetics following a period of relative paucity.11
This ad-
vent has expanded the epilepsy therapeutic armamentarium and
allowed the drugs to match the individual patient's characteristics.
The American Academy of Neurology (AAN) subcommittee reports
in 2004 observed that newer ASDs were not different in controlling
seizures but have better tolerability, particularly fewer neurotoxic
adverse effects.12,13
In 2017, the ILAE published a new classifica-
tion for seizure types and epilepsy syndrome to improve our un-
derstanding of epilepsy and include missing seizure types.14-­
17
This
classification replaced the previous versions published in 198118
and 1989,19
and extended in 2010,20
and has been of paramount
importance to accurately classify the patient's seizure type(s). In
2018, the AAN and American Epilepsy Society (AES) subcommit-
tee published updated guidelines for ASD selection in adult and
pediatric patients with new-­
onset and treatment-­
resistant epi-
lepsy.21,22
However, the guidelines primarily relied on studies that
compare the first-­and second-­
generation ASDs and found limited
data for third-­
generation drugs. Given the rapid advance in the
development of ASDs in recent years and the continuous updates
in definitions, classifications and treatment guidelines for seizure
types, and epilepsy syndromes, this article aims to provide non-­
neurologists and medical students with a complete overview of
the new frontiers in the neuropharmacology of antiseizure drugs.
The ASDs classification, pharmacokinetics, mechanisms of action,
clinical use, and adverse effects are reviewed herein based on a
comprehensive assessment of the current literature and recently
published US and UK treatment guidelines.
1.1 | Definition and epidemiology of epilepsy
Epilepsy is ”a neurological disorder that is characterized by an en-
during predisposition to generate epileptic seizures and the asso-
ciated cognitive, psychological, and social consequences”.1
Epilepsy
is the third most common neurologic disorder; almost 10% of peo-
ple will experience a seizure during their lives.2
The prevalence of
epilepsy is 6.4 cases per 1,000 persons, and the annual incidence is
67.8/100,000 person-­
years.23
1.2 | Classification of seizure types and
epilepsy syndromes
The ILAE classification framework, which was revised in 2017,
is the key tool for the diagnosis of individuals presenting with
seizures.14-­
17
According to that classification, epileptic seizures are
classified into “focal,” “generalized,” and “unknown” seizures, while
epilepsy is classified into “focal,” “generalized,” “combined gener-
alized and focal,” and “unknown” epilepsy. Focal-­
onset seizures
can be further described as focal aware (previously called simple
partial), impaired awareness (complex partial), or focal to bilat-
eral (secondarily generalized) tonic-­
clonic seizures. Generalized
seizures are classified into generalized tonic-­
clonic (formerly idi-
opathic) seizures, motor (myoclonic and other motor) seizures, and
nonmotor (absence) seizures. Etiologies of seizures and epilepsy
syndromes have been reintroduced in the 2017 ILAE classifica-
tion of seizure types and epilepsy syndromes, to include “genetic,”
“structural,” “metabolic,” “infectious,” “immune,” and “unknown.”
For a detailed description of seizure types epilepsy syndromes,
see the ILAE Web site.24
1.3 | Drug treatment of epilepsy
Epilepsy can persist for years and often for the patient's lifetime.
ASDs are the primary therapy for epilepsy and are symptomatic
treatments that control seizures.3
The choice of ASDs varies with
different seizure types and epileptic syndromes.3
ASDs have many
different pharmacologic profiles that are relevant when selecting
338  |     HAKAMI
and prescribing these agents in patients with epilepsy. This includes
pharmacokinetic properties, propensity for drug-­
drug interactions,
and adverse-­
effect profiles and toxicities. All older-­
generation ASDs
have acute dose-­
related effects, primarily neurological effects.25,26
This led to treatment failure in 30%–­
40% of people with epilepsy
receiving an older drug as monotherapy.8
Hence, there has been
a great deal of research into formulating better ASDs, primarily
spurred by the fact that the older-­
generation ASDs do not provide
optimal safety, tolerability or seizure control for many patients with
epilepsy. Over the past three decades, 20 newer-­
generation ASDs
have been approved for clinical use,11
and it was hoped that these
drugs would have better efficacy in controlling seizures than older-­
generation drugs. However, the AAN subcommittee reports in 2004
and 2018 observed that the newer-­
generation ASDs were “not dif-
ferent” in controlling seizures, but better tolerated than the older
drugs.12,13,21,22
1.4 | Classifications of ASDs
ASDs are classified as older (first-­
) generation or newer (second-
­and third-­
) generation agents. The older-­
generation ASDs intro-
duced into clinical practice more than four decades ago include
phenobarbital, phenytoin, primidone, ethosuximide, valproate, car-
bamazepine, clonazepam, and clobazam. The “second-­
generation”
ASDs, which have been approved for the treatment of epilepsy
since the late 1980s, include, in chronological order, vigabatrin,
oxcarbazepine, lamotrigine, gabapentin, felbamate, topiramate,
tiagabine, levetiracetam, and zonisamide. The third-­
generation
ASDs include, pregabalin, fosphenytoin, lacosamide, rufinamide,
eslicarbazepine, retigabine (also known as ezogabine), peram-
panel, brivaracetam, cannabidiol, stiripentol, cenobamate, and
fenfluramine.11,12,27,28
The newer ASDs differ substantially in their
mechanisms of action, spectra of activity, pharmacokinetics, and
adverse effects profiles. Figure 1 presents the classification and
year of introduction of ASDs available now. Current information
on the other drugs in the pipeline can be found on the Epilepsy
Foundation Web site.29
1.5 | Pharmacokinetics and drug
interactions of ASDs
ASDs are commonly used for long periods of time, and consideration
of their pharmacokinetic properties is essential for avoiding toxicity
and drug interactions.30
An ideal ASD should demonstrate complete
absorption, linear kinetics, a long elimination half-­
life, and allowing
once or twice-­
daily dosing. Other favorable properties include low
protein binding, lack of active metabolites, and clearance by kidneys.
Generally, all ASDs are well absorbed after oral administration, have
good bioavailability, and readily cross the blood-­
brain barrier. Many
ASDs are medium to long-­
acting drugs (have half-­
lives of 12 hours)
and can be administered twice or three times a day. Phenobarbital,
phenytoin, zonisamide, eslicarbazepine, and perampanel can often
be administered once daily. Extended-­
release preparations of drugs
that have short half-­
lives (eg, carbamazepine, valproate, leveti-
racetam, and lamotrigine) may decrease the incidence of adverse ef-
fects and allow once-­
daily dosing. For optimum therapy, therapeutic
drug concentration should be monitored in individual patients for
some drugs. Examples include carbamazepine, phenytoin, and val-
proate. Drug levels can be helpful (1) to guide dose adjustments, (2)
when breakthrough seizures occur, (3) when an interacting medica-
tion is added, (4) during pregnancy, (5) to assess compliance, and (6)
to determine whether adverse effects are related to drug levels.
Clinically relevant pharmacokinetic profiles of ASDs are summarized
in Table 1.
In general, the ASD should be started at a low dose, with incre-
ments over several weeks to establish an effective and tolerable
regimen. Some agents do not require titration, such as gabapentin
and levetiracetam. Clinicians should be aware of certain factors
that affect dosing. These factors include nonlinear relationships be-
tween dose and drug exposure (eg, phenytoin) and the influence of
hepatic or renal impairment on clearance. Metabolism of phenytoin
is nonlinear; elimination kinetics shift from first order at a low dose
to zero-­
order at moderate to high dose. As a subsequent, any small
increases in the dose may increase phenytoin half-­
life and serum
concentration, and a patient may quickly develop toxicity. A com-
mon clinical error is to increase the dosage directly from 300 mg/d
FI G U R E 1 Classification and year of introduction of antiseizure drugs
    | 339
HAKAMI
to 400 mg/d; toxicity frequently occurs at a variable time thereafter.
Hepatic biotransformation of valproate may lead to hepatotoxicity.
Hence, the liver function test is recommended in individual patients.
Most ASDs are metabolized by hepatic enzymes. Carbamazepine,
oxcarbazepine, eslicarbazepine, phenobarbital, phenytoin, and prim-
idone are inducers of hepatic cytochrome P450 enzyme and may
decrease the effects of other drugs administered concomitantly
(eg, valproate). Valproate and clobazam are inhibitors of hepatic
enzymes and most likely to elevate the plasma concentration of
other drugs administered concomitantly (eg, carbamazepine, etho-
suximide, phenytoin, phenobarbital, and lamotrigine). Drug-­
drug
interactions with ASDs are complex since the drugs are often used
in combination. These interactions may lead to either inadequate
seizure control or drug toxicity. Of the newer drugs, levetiracetam,
gabapentin, pregabalin, and vigabatrin are unique. These drugs are
eliminated unchanged by the kidney and have no drug-­
drug inter-
actions. Lamotrigine, perampanel, tiagabine, topiramate, and zoni-
samide undergo hepatic drug metabolism and have potential drug
interactions. Oxcarbazepine, felbamate, and topiramate selectively
induce the hepatic metabolism of the oral contraceptive pill, failing
birth control.
1.6 | Mechanisms of actions of ASDs
A propensity for seizure generation occurs when there is an imbal-
ance favoring excitation of neurons over inhibition. ASDs actions
can generally be viewed in the context of inhibition of excitation or
strengthening of inhibition, or both. Inhibition of excitation can be
produced by effects on intrinsic excitability mechanisms in excita-
tory neurons (eg, inhibition of sodium and calcium channel), or on
excitatory synaptic transmission (eg, glutamate AMPA and NMDA
receptors and synaptic vesicle protein 2A). Enhancement of inhi-
bition is produced by the increased availability of γ-­
aminobutyric
acid (GABA), increased activation of GABAA receptors, the media-
tors of inhibition in cortical areas relevant to seizures, and modu-
lation of voltage-­
gated potassium channel of the Kv7. For some
drugs, the precise mechanism of action is not known (eg, valproate,
zonisamide, and rufinamide), and some have multiple targets (eg,
topiramate and felbamate).31
The ASD mechanisms of action are
summarized in Table  2 and discussed in detail in the remainder
sections.
1.7 | Voltage-­
gated sodium channels blockade
At therapeutic concentrations, carbamazepine, cenobamate, lacosa-
mide, lamotrigine, phenytoin, and zonisamide block voltage-­
gated
sodium channel in neuronal membranes. These agents act mainly on
action potential firing and do not directly alter excitatory or inhibi-
tory synaptic responses. However, the effect on action potentials
translates into reduced transmitter output at synapses. Topiramate
may also act, in part, by this mechanism. Phenobarbital and valproate
may act on the sodium channel at high doses. This action is rate-­
dependent (ie, the block increases with increased frequency of neu-
ronal discharge).31
1.8 | GABA-­
related targets
Benzodiazepines interact with specific receptors (GABAA receptors)
and increase the frequency of GABA-­
mediated chloride ion chan-
nel opening (facilitate inhibitory effects of GABA). Phenobarbital
and other barbiturates increase the duration of GABA-­
mediated
chloride ion channel opening. Vigabatrin, an analog of GABA, irre-
versibly inactivates GABA transaminase, the enzyme responsible for
the termination of action of GABA. Valproate also can inhibit GABA
transaminase at high doses. Tiagabine selectively inhibits the GAT-­
1
GABA transporter, causing prolongation of GABA-­
mediated inhibi-
tory synaptic responses. Gabapentin and pregabalin are structural
analogs of GABA but do not act through effects on any mechanism
related to GABA-­
mediated neurotransmission. Instead, the drugs
bind to the α2δ subunit of the voltage-­
gated calcium channel and
decrease glutamate release at excitatory synapses. Other drugs that
may facilitate the inhibitory actions of GABA include felbamate and
topiramate.31
1.9 | Calcium channel blockade
Ethosuximide inhibits low voltage-­
gated T type calcium channels
in thalamocortical neurons. Valproate may exert similar action.
Gabapentin and pregabalin bind to the α2δ subunit of voltage-­
gated
calcium channel and may decrease glutamate release at excitatory
synapses (the precise mechanism is not known).31
1.10 | Glutamate synapses and other mechanisms
Levetiracetam binds to the synaptic vesicle protein 2A (SV2A) re-
ceptors on glutamate-­
containing transmitter vesicles, where it is
believed to inhibit excitatory neurotransmitters release (vesicular
exocytosis) and thereby reduce synaptic excitability. Retigabine
opens KCNQ2-­
5 (Kv7.2-­
Kv7.5) voltage-­
gated potassium channel in
presynaptic nerve terminals and thereby inhibits glutamate release.
In addition to its action on calcium and sodium channel, valproate
may enhance voltage-­
gated potassium channel permeability, causing
hyperpolarization. Perampanel is a potent noncompetitive antago-
nist of the AMPA receptors, a subtype of the ionotropic glutamate
receptors that are the primary mediators of synaptic excitation in
the central nervous system. Phenobarbital may block glutamate
AMPA receptors and its action on voltage-­
activated calcium channel
and GABA-­
chloride ion channel. Felbamate blocks glutamate NMDA
receptors, with selectivity for those containing the GluN2B subu-
nit. The drug also produces a barbiturate-­
like potentiation of GABAA
receptor responses. The possible sites for the action of topiramate
340  |     HAKAMI
include voltage-­
gated sodium channel, GABAA receptor subtypes,
and glutamate AMPA receptors.31
1.11 | Principles of ASD selection for specific
seizure type(s)
Treatment should be considered in patients reporting more than
one unprovoked seizure or after a single seizure if the risk of re-
currence is high.1
The initial ASD should be individualized on the
basis of seizure type and/or epilepsy syndrome and slowly titrated
up to a target dosage. Other important selection criteria include pa-
tient characteristics, drug efficacy, adverse-­
effect profile, potential
drug-­
drug interactions, and cost.32
Combination therapy should be
considered after the failure of two monotherapies.33
The therapeu-
tic spectrum of ASDs can be categorized into (1) broad-­
spectrum
drugs used for both focal and generalized seizures, including, in al-
phabetical order, brivaracetam, clobazam, felbamate, lamotrigine,
levetiracetam, perampanel, rufinamide, topiramate, valproate, and
zonisamide; (2) narrow-­
spectrum drugs used primarily for focal and
focal to bilateral (secondarily generalized) tonic-­
clonic seizures in-
cluding carbamazepine, cenobamate, eslicarbazepine, gabapentin,
lacosamide, oxcarbazepine, phenobarbital, phenytoin, pregabalin,
primidone, stiripentol, tiagabine, and vigabatrin; and (3) a narrow-­
spectrum drug used primarily for generalized absence seizures
which is ethosuximide.
Table 3 presents the efficacy of ASDs against common seizure
types and epilepsy syndrome. The data supplemented in the table
are based primarily on the results of RCTs comparing the newer ver-
sus older drugs, including the SANAD studies34-­
37
and others,38-­
62
and recently published meta-­
analyses.63-­
69
Table 4 and the remain-
der sections of this article present guidance for ASD selection for
the specific seizure type according to the 2017 ILAE classification
of seizure types and epilepsy syndromes.14-­
17
The guidance is based
mainly on the current state of the literature on the efficacy and tol-
erability of ASDs and the recently published practical guidelines.
The most popular treatment guidelines applied herein are the guide-
lines published by AAN  AES in 2004, 2015, and 2018,12,13,21,22,70
the ILAE in 2006 and 2013,71,72
the UK National Institute for Health
and Care Excellence in 2012,73,74
and the Scottish Intercollegiate
Guidelines Network in 2018.75,76
1.12 | Drugs effective for focal-­
onset seizures
Focal-­
onset seizures account for 60% of all epilepsies. The RCTs
and meta-­
analyses have demonstrated comparable efficacy of sev-
eral ASDs in controlling new-­
onset focal seizures.22,34,37-­
44,46,58,62-­
64,72,77-­
83
The drug of the first choice for “focal” and “focal to
bilateral” (secondarily generalized) tonic-­
clonic seizures is lamotrig-
ine.22,34,37
Phenytoin is the drug of choice for the urgent treatment
of new-­
onset or recurrent focal epilepsy. The other most widely
used first-­
line drugs for focal seizures include levetiracetam and
zonisamide.22,34
Carbamazepine is considered a second-­
line treat-
ment for new-­
onset focal epilepsy owing to its unfavorable efficacy-­
to-­
tolerability profile.22,34
Oxcarbazepine, topiramate, and valproate
can be used, but they may not be as effective as lamotrigine and
carbamazepine.22
Phenobarbital (and its derivative primidone) is
often regarded as a second-­
line treatment in adults because of seda-
tion and behavioral problems.22
However, phenobarbital is a primary
drug for neonatal seizures.
Given the unique mechanistic and pharmacokinetic profiles
of the third-­
generation ASDs, the United States Food and Drug
Administration (FDA) recently issued a strategy that allows extrap-
olation of these drugs across populations as add-­
on or monother-
apy.22
Accordingly, brivaracetam, eslicarbazepine, lacosamide, and
perampanel received FDA approval as initial monotherapies for
new-­
onset focal epilepsy.22
Individual monotherapy studies showed
similar efficacy of second-­
generation (levetiracetam and zonisamide)
and third-­
generation (eslicarbazepine and lacosamide) ASDs com-
pared with controlled-­
release carbamazepine for the treatment of
new-­
onset focal epilepsy.63
Vigabatrin use appears to be less effica-
cious than carbamazepine use and may not be offered; furthermore,
tolerability profile precludes vigabatrin use as first-­
line therapy.22
The drugs used as add-­
on (adjunctive) therapy in focal seizures in-
clude gabapentin, pregabalin, felbamate, rufinamide, cenobamate,
clobazam, retigabine, and tiagabine. However, the AAN/AES guide-
lines recommend gabapentin and lamotrigine, as first-­
line monother-
apy in patients aged ≥60 years with new-­
onset focal epilepsy.22
A
recent systematic review and network meta-­
analysis showed that
lacosamide, lamotrigine, and levetiracetam had the highest proba-
bility of ranking best for achieving seizure freedom in the elderly
with new-­
onset epilepsy.64
The FDA determined that the efficacy of
ASDs for focal-­
onset seizures in adults can be extrapolated down-
ward to children four years of age and above.22
For drug-­
resistant focal epilepsy in adults, eslicarbazepine can
be used as monotherapy while the immediate-­
release pregabalin,
perampanel, lacosamide, eslicarbazepine, extended-­
release topi-
ramate, rufinamide, clobazam, felbamate, and vigabatrin should be
considered as add-­
on therapy.21
However, vigabatrin, felbamate, and
rufinamide are not first-­
line agents because of the retinopathy risk
with vigabatrin, the modest benefit with rufinamide, and the hepato-
toxicity and hematotoxicity risk with felbamate. In pediatric patients,
levetiracetam, oxcarbazepine, and zonisamide should be considered
as add-­
on therapy.21
1.13 | Drugs effective for generalized tonic-­
clonic
seizures (GTCS)
There has been a limited number of ASDs that can be used as first-­
line agents for GTCS. Valproate remains the first-­
line drug for many
patients with generalized and unclassified epilepsies,22,35,36,72
while
phenobarbital (or primidone) is a primary drug in infants (for neona-
tal seizures) and an alternative in adults. However, valproate should
not be prescribed for women of childbearing potential because of
    | 341
HAKAMI
its dose-­
dependent teratogenic profile; unless other ASDs cannot
control the seizures, in which case the dose should be kept as low as
possible.84
Indeed, some patients with genetic generalized epilepsy
can control their seizures only with valproate. The other most widely
used monotherapies for GTCS include lamotrigine, levetiracetam,
brivaracetam, topiramate, clobazam, and zonisamide.22
However,
there appears to be no evidence to support any second-­
generation
or third-­
generation ASDs to be as efficacious as valproate mono-
therapy for generalized and unclassified epilepsies.35,36
The com-
bination of lamotrigine and valproate is believed to be particularly
efficacious.85
Phenytoin and carbamazepine are effective but may
worsen certain seizure types in generalized epilepsies, including
absence epilepsy, juvenile myoclonic epilepsy, and Dravet's syn-
drome.86
Vigabatrin, tiagabine, oxcarbazepine, and possibly gabap-
entin are other drugs that may worsen these seizure types. Seizures
of most patients with generalized epilepsy are easily controlled with
appropriate medication. However, immediate-­
release and extended-­
release lamotrigine use should be considered add-­
on therapy to
decrease seizure frequency in drug-­
resistant GTCS in adults.21
Levetiracetam should also be effective in both drug-­
resistant GTCS
and drug-­
resistant juvenile myoclonic seizures. Other drugs used as
add-­
on therapy in GTCS include perampanel and lacosamide.
1.14 | Drugs effective for focal seizures and certain
generalized onset seizure types
A variety of drugs are primarily used to treat focal seizures; these
drugs have also been effective in certain generalized onset seizure
types. These drugs are lamotrigine, levetiracetam, brivaracetam,
perampanel, phenobarbital, primidone, and felbamate.
1.15 | Drugs effective for myoclonic seizures
Valproate is widely used for myoclonic seizures, such as in juvenile
myoclonic epilepsy.87
Lamotrigine is possibly effective but may
worsen myoclonic seizures in some cases. Other drugs effective
in treating this seizure type are levetiracetam, brivaracetam, zon-
isamide, topiramate, and clonazepam.
1.16 | Drugs effective for generalized
absence seizures
Ethosuximide is the first-­
line drug. It is often used in uncomplicated
absence seizures if patients can tolerate its gastrointestinal side ef-
fects.22,45
Despite the long half-­
life (~ 40 hours), ethosuximide is gen-
erally administered in two or even three divided doses to minimize
the adverse gastrointestinal effects. Valproate is preferred in pa-
tients with concomitant GTCS or myoclonic seizures (myoclonic ab-
sence seizure). Clonazepam is effective as an alternative drug but has
the disadvantages of causing sedation and tolerance. Lamotrigine,
levetiracetam, and zonisamide are also used in the absence seizures
but not as effective as ethosuximide or valproate. Unless there are
compelling reasons based on adverse events profile, ethosuximide
or valproate use should be considered before lamotrigine in treat-
ing absence seizures.45
Lamotrigine should be considered in women
of childbearing age because of its better tolerability and fewer fetal
risks compared with valproate.
1.17 | Drugs effective for other epilepsy syndromes
In combination with lamotrigine and a benzodiazepine (such as
clobazam), valproate is the most widely used treatment for atonic
seizures (eg, in the Lennox-­
Gastaut syndrome). Other drugs used
for the atonic seizures in Lennox-­
Gastaut syndrome include topira-
mate, felbamate, and rufinamide.85
Because felbamate can cause
aplastic anemia and severe hepatitis, it is used as add-­
on therapy
in patients who respond poorly to other agents. For infantile
spasms (West's syndrome), valproate, topiramate, zonisamide,
or a benzodiazepine (such as clonazepam or nitrazepam) is ef-
fective.85
Vigabatrin and everolimus are used if associated with
tuberous sclerosis complex (a rare genetic disease that causes non-­
cancerous (benign) tumors to grow in the brain and other organs).
However, infantile spasms are primarily treated with intramuscu-
lar adrenocorticotropic hormone (ACTH) or oral corticosteroids
such as prednisone or hydrocortisone (unknown mechanism). For
Dravet's syndrome (the severe myoclonic epilepsy of infancy), val-
proate, topiramate, and clobazam can be used, although none of
these is very effective.88
Stiripentol, a modulator of GABAA recep-
tors, is often used in conjunction with clobazam or valproate.88
Cannabidiol was approved in 2018 to treat Dravet's syndromes66
in addition to Lennox-­
Gastaut syndrome68
and infantile spasms as-
sociated with tuberous sclerosis complex. Recent meta-­
analyses
showed that adjunctive cannabidiol was associated with a greater
reduction in convulsive seizure frequency than placebo in patients
with Lennox-­
Gastaut syndrome68
and children with Dravet syn-
drome.66
Fenfluramine is another new drug approved in 2020 for
Dravet's syndrome. The drug is available only through a restricted
drug distribution program, under a risk evaluation and mitigation
strategy (REMS) because of the risk of valvular heart disease and
pulmonary arterial hypertension. Recent studies, however, showed
that fenfluramine provided a significantly greater reduction in con-
vulsive seizure frequency compared with placebo and was gen-
erally well tolerated, with no observed valvular heart disease or
pulmonary arterial hypertension.65,89
1.18 | Drugs effective for status epilepticus
Status epilepticus (SE) is a life-­
threatening emergency that requires
immediate treatment.90
In clinical practice, it is now generally ac-
cepted that a seizure lasting 5 minutes for GTCS and 10 minutes for
focal seizures with or without impairment of consciousness should
342  |     HAKAMI
be treated as status epilepticus. Figure 2 presents the SE treatment
algorithm. According to the evidence-­
based guideline for the treat-
ment of convulsive SE in children and adults by the AES in 2016,91
the first-­
line treatment for SE is a benzodiazepine (either intravenous
lorazepam or intravenous diazepam or intramuscular midazolam). If
the preferred options are not available, rectal diazepam, intranasal
midazolam, buccal midazolam, or intravenous phenobarbital is ac-
ceptable alternatives.
If the seizure continues, then second-­
line treatment is adminis-
tered.69
Intravenous fosphenytoin is preferred over phenytoin as the
latter may cause cardiotoxicity. With phenytoin intravenous admin-
istration, there is also a risk of the potentially serious “purple glove
syndrome,” in which a purplish-­
black discoloration accompanied by
edema and pain occurs distal to the site of injection. Fosphenytoin
has a lower incidence of purple glove syndrome. The second-­
line
treatment also includes intravenous valproate or intravenous leve-
tiracetam. Intravenous phenobarbital can be alternative (if not given
already) but has a long half-­
life causing persistent adverse effects,
including severe sedation, respiratory depression, and hypotension.
A recent network meta-­
analysis compared efficacy and tolerability
of intravenous valproate, phenytoin, diazepam, phenobarbital, la-
cosamide, and levetiracetam in adults with benzodiazepine-­
resistant
convulsive SE and suggested that phenobarbital had the greatest
probabilities of being best in the achievement of SE control and
seizure freedom, whereas valproate and lacosamide ranked best for
the safety outcomes (respiratory depression and hypotension).69
If the second therapy fails to stop the seizures, another second-­
line agent is often tried. Treatment-­
resistant SE occurs when sei-
zures continue or recur at least 30  minutes after treatment with
first-­and second-­
line agents and should be treated with anesthetic
doses of pentobarbital, propofol, midazolam, or thiopental. Dosing
and frequency are available in the proposed algorithm for convulsive
SE.91
Acute repetitive seizures (seizure clusters), in which there is
complete recovery between seizures, are treated with benzodiaze-
pines. Diazepam rectal gel is the only approved treatment for the
out-­
of-­
hospital treatment of acute repetitive seizures.92
1.19 | Adverse effects of ASDs
The adverse effects of selected ASDs are listed in Table  5. The
first-­
generation ASDs have acute dose-­
related effects, primar-
ily neurological effects such as sedation, dizziness, unsteadiness,
blurred vision, diplopia, and tremor, in addition to neurocognitive
and psychiatric symptoms.25,26,32
These effects are found across the
different ASDs26,62
and often mild and reversible. However, some
drugs are better tolerated than others; for example, lamotrigine and
levetiracetam are better tolerated than carbamazepine in elderly
FI G U R E 2 Status Epilepticus treatment algorithm*
*Adopted from Glauser T, et al Epilepsy Curr. 2016; 16:48-­
6. Refer to publication for complete recommendations.
ABCD, airway, breathing, circulation, disability (Neurologic); B, buccal; ECG, electrocardiogram; ED, emergency department; EEG,
electroencephalogram; IM, intramuscular; IN, intranasal; IV, intravenous; PE, phenytoin sodium equivalents; SE, status epilepticus.
    | 343
HAKAMI
patients.93
Psychiatric adverse effects include depression, anxiety,
irritability, impaired concentration, mood changes, hyperactivity,
and, in rare cases, psychosis. Although the newer ASDs are touted
as better tolerated than older drugs,12
psychiatric adverse effects
are common with levetiracetam, topiramate, zonisamide, vigabatrin,
and perampanel. Lamotrigine, carbamazepine, valproate, gabap-
entin, and pregabalin, in contrast, have mood-­
stabilizing effects in
some patients and less frequently cause behavioral or psychiatric
effects.94
Most women with epilepsy who become pregnant require
continued ASD therapy for seizure control. If possible, valproate,
carbamazepine, phenytoin, phenobarbital, and topiramate should
be avoided in women of childbearing potentials. Recent analysis
from several international pregnancy registers suggests that in
utero exposure to valproate during the first trimester is associ-
ated with a threefold increased risk of congenital malformations,
commonly neural tube defects (spina bifida) and cardiovascular,
orofacial, and digital abnormalities.95
The use of valproate during
the first trimester is also associated with cognitive impairments.
Carbamazepine may cause neural tube defects and craniofacial
anomalies. Fetal hydantoin syndrome is related to the use of phe-
nytoin. Treatment with topiramate during the first trimester of
pregnancy is associated with a 10-­
fold increase in oral clefts risk.
Phenobarbital can cause congenital malformations, most often
cardiac defects. No ASD is known to be entirely safe for the de-
veloping fetus. However, lamotrigine and levetiracetam have the
TA B LE 1 Pharmacokinetic profiles of antiseizure drugs
Antiseizure drug
Bioavailability
%
Peak concentration
(hr)
Plasma protein
binding (%)
Elimination
half-­
life (hr)
Route of
elimination
Therapeutic serum
concentration (mcg/mL)
Brivaracetam ~ 95 1 ≤ 20 7-­
10 ++ 0.2-­
2
Carbamazepine 75-­
85 4-­
5 70-­
80 10-­
17 ++++ 4-­
11
Cannabidiol 10-­
20 2.5-­
5 94 56-­
61 ++++ NE
Cenobamate 88 1-­
4 60 50-­
60 +++ NE
Clobazam 90-­
100 1-­
3 80-­
90 36-­
42 ++++ 0.03-­
3
Clonazepam 80 1-­
4 80-­
90 24-­
48 +++ 10-­
70a
 
Eslicarbazepine 90 1-­
4 40 13-­
20 ++++ 5-­
35
Ethosuximide 95-­
100 3-­
7 0 30-­
60 ++ 40-­
100
Felbamate 90 3-­
5 22-­
36 16-­
22 ++ 30-­
60
Gabapentin 50 2-­
3 0 5-­
9 -­ 3-­
21
Lacosamide 100 1-­
2 30 12-­
14 + 3-­
10
Lamotrigine ~ 90 1-­
3 55 8-­
35 +++ 3-­
13
Levetiracetam ~ 95 1-­
2 10 6-­
8 -­ 5-­
41
Oxcarbazepine 100 4-­
5 75 10-­
17 ++++ 3-­
36
Perampanel 100 0.5-­
3 95-­
96 70-­
110 +++ 0.1-­
1
Phenobarbital 90 0.5-­
4 55 90 ++ 12-­
30
Phenytoin 85-­
90 5-­
7 90 24 +++b
  10-­
20
Pregabalin ~90 1-­
2 0 4.5-­
7 -­ 2-­
6
Primidone 90 2-­
6 10 8-­
15 ++ 8-­
12
Rufinamide 90 4-­
6 35 6-­
10 ++ 4.5-­
31
Stiripentol Variable 2-­
3 99 4.5-­
13 + 4-­
22
Tiagabine ~90 0.5-­
2 96 2-­
9 +++ 0.02-­
0.2
Topiramate ~80 2-­
4 15 20-­
30 + 2-­
10
Valproate 90 2-­
4 90 15 ++++ 50-­
100
Vigabatrin 100 1 0 5-­
8 -­ 20−160a
 
Zonisamide 90 2-­
6 40-­
60 50-­
68 ++ 10-­
38
Note: NE, not established
++++Extensive hepatic metabolism and active metabolite(s)
+++Extensive hepatic metabolism but no active metabolite(s)
++Hepatic metabolism (with or without active metabolites) and renal excretion.
+Variable (or moderate) hepatic metabolism (with or without active metabolites)
-­
Renal excretion (unchanged). No hepatic metabolism
a
ng/mL
b
Saturable
344  |     HAKAMI
lowest risks of major congenital malformations and may be safer,
particularly for cognition compared with valproate.85
Valproate causes hepatotoxicity in children less than two years
of age. Carbamazepine and lamotrigine may cause life-­
threatening
Steven-­
Johnson syndrome and toxic epidermal necrolysis, which
is strongly associated with the HLA-­
B*1502 allele. Asians, who
have a 10-­
fold increased risk of the drug-­
induced Stevens-­
Johnson
syndrome compared with other ethnic groups, should be tested
before starting the drug. The use of zonisamide is also associated
with severe skin reactions. Aplastic anemia and acute hepatic fail-
ure have limited the use of felbamate to severe and drug-­
resistant
epilepsy. Overdose toxicity with benzodiazepines and barbiturates
may cause respiratory depression. Management is primarily sup-
portive (airway management, mechanical ventilation) and fluma-
zenil in benzodiazepine overdose. Withdrawal from ASDs should
be accomplished gradually (over a 1-­to 3-­
month period or longer)
to avoid the occurrence of severe seizures or status epilepticus.
Physical dependence occurs with barbiturates and benzodiaze-
pines, and there is a well-­
recognized risk of rebound seizures with
abrupt withdrawal. However, withdrawal is less likely to be a prob-
lem with ethosuximide. Withdrawal is believed to be successful in
patients with generalized epilepsies who exhibit a single seizure
type, whereas the longer duration of epilepsy, an abnormal neu-
rologic examination, an abnormal EEG, and certain epilepsy syn-
dromes, including juvenile myoclonic epilepsy, are associated with
increased risk of recurrence.85
Clinical studies suggest a possible association of lamotrigine, le-
vetiracetam, and topiramate with suicidality. In 2008, the FDA issued
an alert that ASDs, as a class, may be associated with an increased risk
of suicidality based on an analysis of data from placebo-­
controlled
add-­
on clinical trials of ASDs in patients with drug-­
resistant epilepsy,
although this is still highly controversial.96,97
Long-­
term treatment
with ASDs is associated with a twofold to threefold increased risk
of osteoporosis and bone fractures.98
In addition, increased body
weight and fat are common in patients using valproate, carbamaz-
epine, gabapentin, pregabalin, vigabatrin, and perampanel and can
lead to serious health consequences associated with obesity and in-
creased cardiovascular disease risk.99
In March 2021, the FDA issued
an alert that lamotrigine may be associated with an increased risk
of cardiac arrhythmias in people with underlying cardiac disease.100
However, this risk is not apparent in healthy individuals.101
1.20 | The new frontiers in epilepsy pharmacology
Since 1989, nearly 20  second-­
generation and third-­
generation an-
tiseizure drugs (ASDs) with different mechanisms of action have
reached the market, resulting in a greatly increased range of treat-
ment options for patients and prescribers.102,103
Some second-­
generation ASDs have shown advantages in drug tolerability,
drug-­
drug interactions, and teratogenicity and thus offer valuable
individualized options in treating epilepsy. Disappointingly, however,
Target and mechanisma
  Antiseizure drug
Inhibition of voltage-­
gated sodium channels Phenytoin, fosphenytoin, carbamazepine,
cenobamate, lamotrigine, oxcarbazepine,
eslicarbazepine, lacosamide, and possibly
topiramate, zonisamide, rufinamide, and
phenobarbital
Inhibition of α2δ subunit of voltage-­
gated
calcium channels
Gabapentin, pregabalin
Inhibition of T type voltage-­
gated calcium
channels
Ethosuximide
Activation of GABAA receptor Phenobarbital, benzodiazepines, and possibly
topiramate, felbamate, retigabine, and
stiripentol.
Inhibition of GABA transporter (selective) Tiagabine
Inhibition of GABA transaminase enzyme Vigabatrin
Modulation of synaptic vesicle protein 2A Levetiracetam, brivaracetam
Various actions on multiple targets Valproate, felbamate, topiramate, zonisamide,
and cannabidiol
Opening KCNQ2-­
5 (Kv7.2-­
Kv7.5) voltage-­
gated potassium channels
Retigabine (ezogabine)b
 
Inhibition of NMDA-­
type glutamate receptors Felbamate, topiramate and phenobarbital
Inhibition of AMPA-­
type glutamate receptors Perampanel
a
Fenfluramine's mechanism of action for the treatment of seizures associated with Dravet
syndrome is unknown.
b
Production of the drug retigabine has been discontinued by the manufacturer, and it is no longer
available.
TA B LE 2 Mechanistic categorization
of current antiseizure drugs based
on foremost targets at therapeutic
concentrations
    | 345
HAKAMI
none of these medications appear to be more efficacious than first-­
generation ASDs, and none have substantially reduced the propor-
tion of patients with drug-­
resistant epilepsy, highlighting the need for
novel strategies in epilepsy drug development. Given the favorable
pharmacokinetic characteristics and adverse-­
effect profiles for the
third-­
generation medications, additional evidence to further define
their efficacy is crucial to the future treatment of epilepsy. The high
incidence of drug-­
resistance and unwanted adverse effects caused
by taking long-­
term ASDs raise substantial concern. In particular,
there is an urgent need for developing novel drugs that act beyond
the membrane ion channels and neural transmissions. Deciphering
the genetics that underpins the mechanisms that generate seizures
is one of the promising areas in the field.104
Many epilepsy genes
have been identified, including genes that increase the risk of dif-
ferent types of epilepsy, such as generalized and focal epilepsy and
developmental and epileptic encephalopathies. These genes have
TA B LE 3 Efficacy of antiseizure drugs against common seizure types and epilepsy syndromes
Antiseizure drug/
seizure type
Focal
seizures GTCS Absence Myoclonic
Lennox–­Gastaut
syndrome
Infantile
spasm
Dravet's
syndrome
Brivaracetam + + +
Cannabidiol + +a
  +
Carbamazepine + + –­ –­ –­
Cenobamate +
Clobazam + + + +e
 
Clonazepam + + +
Eslicarbazepine + –­ –­
Ethosuximide +
Felbamate +b
  +
Fenfluramine +
Gabapentin + ?+ –­ –­ –­
Lacosamide + ?+
Lamotrigine + + + ?+c
  +
Levetiracetam + + ?+ +
Oxcarbazepine + + –­ –­ –­
Perampanel + +
Phenobarbital + + –­ ?+
Phenytoin + + –­ –­ –­
Pregabalin + –­
Primidone + + –­
Retigabineg
  +
Rufinamide + +
Stiripentol +f
 
Tiagabine + –­ –­
Topiramate + + + + + +e
 
Valproate + + + +d
  + + +e
 
Vigabatrin + ?+ –­ –­ +a
 
Zonisamide + + ?+ + +
Note: GTCS; generalized tonic-­
clonic seizure
Note that although there is evidence to support the use of these drugs for these seizure types, the drugs may not be indicated for this use by the US
Food and Drug Administration.
+Effective;?+ possibly effective; –­worsen seizure.
a
Especially when associated with tuberous sclerosis complex.
b
Can cause aplastic anemia and severe hepatitis, used only for patients who respond poorly to other agents.
c
Possibly effective but may worsen myoclonic seizures in some cases.
d
Preferred in patients with concomitant GTCS or myoclonic seizures (myoclonic absence seizure).
e
None of these is very effective in Dravet's syndrome.
f
In combination with clobazam and valproate.
g
Has been discontinued by the manufacturer, and it is no longer available.
346  |     HAKAMI
a role in ion channel and synaptic dysfunction in addition to tran-
scriptional regulation. Most mutations occur in the protein-­
coding
exons, the sodium and potassium channel, and glutamate receptors.32
Ganaxolone, sirolimus, everolimus, cannabidiol, fenfluramine, stirip-
entol, and memantine are new selective disease-­
modifying therapies
recently approved for severe forms of genetic epilepsies.32
Data to
support gene-­
therapy treatments need to be established in future re-
search, and there are often other disorders that these therapies can
improve, including acquired epilepsies.104
Furthermore, with a deeper
understanding of the cellular and molecular mechanisms of epilep-
togenesis, non-­
classical novel antiseizure agents, such as neuroster-
oid and allopregnanolone, with their effect on both GABAergic and
TA B LE 4 Recommendations for add-­
on and monotherapy in adults and pediatric patients 4 years of age with new-­
onset epilepsy based
on an assessment of current literature and published guidelines
Seizure type
First-­
line
(Monotherapy or add-­
on)
Second-­
line
(Monotherapy or add-­
on)
Third-­
line
(Add-­
on)
Focal-­
onset seizures, including focal to
bilateral tonic-­
clonic seizure
Lamotriginea
  Carbamazepine
Levetiracetam
Zonisamide
Phenytoin
Valproate
Topiramateb
 
Oxcarbazepineb
 
Gabapentinb
 
Phenobarbitalc
 
Brivaracetamd
 
Eslicarbazepined
 
Lacosamided
 
Perampaneld
 
Cenobamatee
 
Clobazam
Retigabine
Felbamate
Rufinamide
Pregabalin
Tiagabine
Vigabatrin
Generalized tonic-­
clonic seizures
(GTCS)
Valproatef
  Carbamazepine
Phenytoin
Lamotrigine
Topiramate
Levetiracetam
Brivaracetam
Perampanel
Zonisamide
Clobazam
Phenobarbital
Myoclonic seizure Valproate Lamotrigineg
 
Topiramate
Levetiracetam
Brivaracetam
Clonazepam
Zonisamide
Absence seizures Ethosuximide
Valproate
Lamotrigine
Clonazepam
Levetiracetam
Unclassified seizuresh
  Valproate Lamotrigine
Levetiracetam
Topiramate
Zonisamide
b
Evidence is insufficient to consider gabapentin, oxcarbazepine, or topiramate instead of lamotrigine in patients with new-­
onset focal epilepsy.
Gabapentin may be considered first-­
line monotherapy in patients aged ≥60 years.
c
Often regarded as second-­
line treatment in adults because of sedation and behavioral problems.
d
Received FDA approval for extrapolation of efficacy as monotherapy across individuals with focal seizures (Kanner AM, et al Neurology 2018;
91:82-­
90).
e
Evidence is insufficient to consider the use of clobazam, felbamate, tiagabine, vigabatrin, or third-­
generation antiseizure drugs as monotherapies in
treating new-­
onset focal epilepsy.
f
Valproate should be avoided, if possible, in women of childbearing potential.
g
May worsen myoclonic seizures in some cases.
h
Evidence is insufficient to support efficacy of newer antiseizure drugs in unclassified generalized tonic-­
clonic seizures.
a
Lamotrigine was superior to levetiracetam and zonisamide for time to 12-­
month remission and should remain a first-­
line treatment for new-­
onset
focal epilepsy.
    | 347
HAKAMI
TA B LE 5 Common and serious adverse effects of selected antiseizure drugs
Antiseizure
drug Systemic adverse effects Neurologic adverse effects Rare idiosyncratic reactions
Brivaracetam Nausea, vomiting, constipation,
fatigue
Headache, somnolence, dizziness, abnormal
coordination, nystagmus, mood changes
Carbamazepine Nausea, vomiting, diarrhea, a
plastic anemia, leukopenia,
hyponatremia (common
reason for discontinuation),
hepatotoxicity, rash, pruritus
Ataxia, dizziness, blurred vision, diplopia,
headache
Erythematous maculopapular
rash (Steven-­
Johnson syndrome
and toxic epidermal necrolysis),
teratogenicity
Cenobamate Nausea, vomiting, fatigue,
hyperkalemia, QT shortening
Somnolence, dizziness, headache, balance
disorder, diplopia
Drug reaction with eosinophilia
and systemic symptoms (DRESS)/
multiorgan hypersensitivity (at
high doses)
Eslicarbazepine Nausea, vomiting, diarrhea,
hyponatremia, rash
Dizziness, drowsiness, headache, somnolence,
diplopia, ataxia, blurred vision, tremor
Ethosuximide Nausea, vomiting Sleep disturbance, drowsiness, hyperactivity
Felbamate Nausea, vomiting, anorexia,
weight loss
Insomnia, dizziness, headache, ataxia Aplastic anemia, severe hepatitis/
hepatic failure
Gabapentin Infrequent Somnolence, dizziness, ataxia, headache, tremor,
and fatigue
Lacosamide Nausea, vomiting, increased
cardiac conduction (PR
interval)
Dizziness, ataxia, diplopia, headache
Lamotrigine Nausea, rash, cardiac
arrhythmias
Dizziness, tremor, diplopia Steven-­
Johnson syndrome
Levetiracetam Fatigue, infection, anemia,
leukopenia
Somnolence, dizziness, agitation, anxiety,
irritability, depression, psychosis
Oxcarbazepine Nausea, rash, hyponatremia
(more common)
Somnolence, headache, dizziness, vertigo, ataxia,
diplopia
Perampanel Weight gain, fatigue, nausea Dizziness, somnolence, irritability, gait
disturbance, falls (with high dose), aggression,
mood alteration
Phenobarbital Nausea, rash Somnolence, ataxia, dizziness, confusion,
cognitive dysfunction, tolerance, dependence
Phenytoin Gingival hyperplasia, hirsutism,
megaloblastic anemia,
peripheral neuropathy,
osteoporosis, rash
Nystagmus (early sign of phenytoin
administration), diplopia, ataxia, somnolence
Pregabalin Weight gain, peripheral edema,
dry mouth
Somnolence, dizziness, ataxia, headache, and
tremor
Rufinamide Nausea, vomiting, leukopenia,
cardiac conduction (QT
interval shortening)
Somnolence, fatigue, dizziness, ataxia, headache,
diplopia
Tiagabine Abdominal pain, nausea, lack of
energy
Dizziness, difficulty concentrating, somnolence,
nervousness, tremor, language problems
Topiramate Anorexia, weight loss,
paresthesia, fatigue
Nervousness, psychomotor slowing, language
problems, depression, anxiety, mood problems,
tremor
Acute glaucoma (may require
prompt drug withdrawal).
Valproate Gastrointestinal irritation,
weight gain, hair loss, easy
bruising
Ataxia, somnolence, tremor Hepatotoxicity, teratogenicity, and
thrombocytopenia
Vigabatrin Fatigue Somnolence, headache, dizziness, agitation,
confusion, psychosis.
Irreversible bilateral concentric
visual field defect
Zonisamide Weight loss, nausea, anorexia Somnolence, dizziness, confusion, headache,
psychosis
Potentially serious skin rashes
348  |     HAKAMI
glutaminergic systems, may shed light on finding a new approach to
treat epilepsy, which is not only controlling seizures but also retarding
epileptogenesis.105
Novel pharmacological approaches built on gene
therapies and epileptogenesis may overcome many of the adverse ef-
fects of currently available treatment options.
2 | CONCLUSIONS
Antiseizure drugs (ASDs) have many different pharmacologic profiles
that are relevant when selecting and prescribing these medications
in patients with epilepsy. These include pharmacokinetic properties,
propensity for drug-­
drug interactions, and adverse-­
effect profile and
toxicities. This article reviewed the current state of the literature
about the clinical pharmacology of ASDs, based primarily on the most
recent evidence for the efficacy and tolerability of the currently avail-
able drugs and the recent US and UK treatment guidelines. It is antici-
pated that Table 4 in this review (recommendations for ASD selection)
will require updating as future studies yield more detailed results. It
is hoped that this review remains a succinct and practical guide to as-
sist non-­
neurologists, particularly the primary healthcare practition-
ers, in patient care decisions. The medical students may also wish to
read this review to improve their understanding of epilepsy and its
pharmacological treatment. Finally, the content of this review is not
intended to include all legitimate criteria for choosing to use or ex-
clude a specific drug, nor recommended as a substitute for current sci-
entific and clinical information. Physicians are encouraged to carefully
review the literature in addition to the full guidelines to understand all
recommendations associated with patient care and be confident that
the information contained in this work is accurate, particularly for new
or infrequently used drugs. Physicians are also encouraged to obtain
information about drug dosing and frequency from the product infor-
mation sheet included in each drug package. Complete information on
US Food and Drug Administration (FDA) labeling for each drug can be
accessed using the FDA searchable database (FDALabel).
CONFLICT OF INTEREST
The author declares no conflict of interest.
AUTHOR CONTRIBUTIONS
The author has made substantial contributions to the conception
and design of the work, and acquisition, analysis, and interpretation
of data for the work; drafting the work and revising it critically for
important intellectual content; final approval of the version to be
published; and agreed to be accountable for all aspects of the work
in ensuring that questions related to the accuracy or integrity of any
part of the work are appropriately investigated and resolved.
DATA AVAILABILITY STATEMENT
Data sharing not applicable to this article.
ORCID
Tahir Hakami  https://orcid.org/0000-0002-1933-6044
REFERENCES
	 1.	 Fisher RS, Acevedo C, Arzimanoglou A, Bogacz A, Cross JH, Elger
CE, et al. ILAE official report: a practical clinical definition of epi-
lepsy. Epilepsia. 2014;55:475-­
82.
	 2.	 Hauser WA, Beghi E. First seizure definitions and worldwide inci-
dence and mortality. Epilepsia. 2008;49(Suppl 1):8-­
12.
	 3.	 Pitkanen A. Therapeutic approaches to epileptogenesis–­
hope on
the horizon. Epilepsia. 2010;51(Suppl 3):2-­
17.
	 4.	 Kwan P, Brodie MJ. Early identification of refractory epilepsy. N
Engl J Med. 2000;342:314-­
9.
	 5.	 Marson A, Jacoby A, Johnson A, Kim L, Gamble C, Chadwick D,
et al. Immediate versus deferred antiepileptic drug treatment for
early epilepsy and single seizures: a randomised controlled trial.
Lancet. 2005;365:2007-­
13.
	 6.	 Kwan P, Brodie MJ. Drug treatment of epilepsy: when does it fail
and how to optimize its use? CNS Spectr. 2004;9:110-­
9.
	 7.	 Kwan P, Arzimanoglou A, Berg AT, Brodie MJ, Allen Hauser W,
Mathern G, et al. Definition of drug resistant epilepsy: consen-
sus proposal by the ad hoc Task Force of the ILAE Commission on
Therapeutic Strategies. Epilepsia. 2010;51:1069-­
77.
	 8.	 Mattson RH, Cramer JA, Collins JF, Smith DB, Delgado-­
Escueta
AV, Browne TR, et al. Comparison of carbamazepine, phenobar-
bital, phenytoin, and primidone in partial and secondarily general-
ized tonic-­
clonic seizures. N Engl J Med. 1985;313:145-­
51.
	 9.	 French JA, White HS, Klitgaard H, Holmes GL, Privitera MD, Cole
AJ, et al. Development of new treatment approaches for epilepsy:
unmet needs and opportunities. Epilepsia. 2013;54(Suppl 4):3-­
12.
	 10.	 Wilcox KS, Dixon-­
Salazar T, Sills GJ, Ben-­
Menachem E, Steve
White H, Porter RJ, et al. Issues related to development of new
antiseizure treatments. Epilepsia. 2013;54(Suppl 4):24-­
34.
	 11.	 Stephen LJ, Brodie MJ. Pharmacotherapy of epilepsy: newly ap-
proved and developmental agents. CNS Drugs. 2011;25:89-­
107.
	 12.	 French JA, Kanner AM, Bautista J, Abou-­
Khalil B, Browne T, Harden
CL, et al. Efficacy and tolerability of the new antiepileptic drugs I:
treatment of new onset epilepsy: report of the Therapeutics and
Technology Assessment Subcommittee and Quality Standards
Subcommittee of the American Academy of Neurology and the
American Epilepsy Society. Neurology. 2004;62:1252-­
60.
	 13.	 French JA, Kanner AM, Bautista J, Abou-­
Khalil B, Browne T, Harden
CL, et al. Efficacy and tolerability of the new antiepileptic drugs II:
treatment of refractory epilepsy: report of the Therapeutics and
Technology Assessment Subcommittee and Quality Standards
Subcommittee of the American Academy of Neurology and the
American Epilepsy Society. Neurology. 2004;62:1261-­
73.
	 14.	 Fisher RS. An overview of the 2017 ILAE operational classification
of seizure types. Epilepsy Behav. 2017;70:271-­
3.
	 15.	 Fisher RS, Cross JH, D'Souza C, et al. Instruction manual for the
ILAE 2017 operational classification of seizure types. Epilepsia.
2017;58:531-­
42.
	 16.	 Fisher RS, Cross JH, French JA, Higurashi N, Hirsch E, Jansen FE,
et al. Operational classification of seizure types by the International
League Against Epilepsy: Position Paper of the ILAE Commission
for Classification and Terminology. Epilepsia. 2017;58:522-­
30.
	 17.	 Scheffer IE, Berkovic S, Capovilla G, et al. ILAE classification of the
epilepsies: position paper of the ILAE commission for classification
and terminology. Epilepsia. 2017;58:512-­
21.
	 18.	 Proposal for revised clinical and electroencephalographic classi-
fication of epileptic seizures. From the commission on classifica-
tion and terminology of the international league against epilepsy.
Epilepsia. 1981;22:489-­
501.
	 19.	 Proposal for revised classification of epilepsies and epileptic syn-
dromes. Commission on classification and terminology of the in-
ternational league against epilepsy. Epilepsia. 1989;30:389-­
99.
	 20.	 Berg AT, Berkovic SF, Brodie MJ, Buchhalter J, Cross JH, van Emde
Boas W, et al. Revised terminology and concepts for organization
    | 349
HAKAMI
of seizures and epilepsies: report of the ILAE Commission on
Classification and Terminology, 2005–­
2009. Epilepsia.
2010;51:676-­
85.
	 21.	 Kanner AM, Ashman E, Gloss D, Harden C, Bourgeois B, Bautista
JF, et al. Practice guideline update summary: efficacy and tolerabil-
ity of the new antiepileptic drugs II: treatment-­
resistant epilepsy:
report of the guideline development, dissemination, and imple-
mentation subcommittee of the American Academy of Neurology
and the American Epilepsy Society. Neurology. 2018;91:82-­
90.
	 22.	 Kanner AM, Ashman E, Gloss D, Harden C, Bourgeois B, Bautista
JF, et al. Practice guideline update summary: efficacy and toler-
ability of the new antiepileptic drugs I: treatment of new-­
onset
epilepsy: report of the guideline development, dissemination,
and implementation subcommittee of the American Academy
of Neurology and the American Epilepsy Society. Neurology.
2018;91:74-­
81.
	 23.	 Fiest KM, Sauro KM, Wiebe S, Patten SB, Kwon C-­
S, Dykeman
J, et al. Prevalence and incidence of epilepsy: a systematic re-
view and meta-­
analysis of international studies. Neurology.
2017;88:296-­
303.
	 24.	 Epilepsy Foundation. Epilepsy Pipeline Tracker, https://www.epile​
psy.com/pipel​
ine-­
listi​
ng-­
page (accessed April 25, 2021)
	 25.	 Baker GA, Jacoby A, Buck D, Stalgis C, Monnet D, et al. Quality
of life of people with epilepsy: a European study. Epilepsia.
1997;38:353-­
62.
	 26.	 Loscher W, Schmidt D. Modern antiepileptic drug development
has failed to deliver: ways out of the current dilemma. Epilepsia.
2011;52:657-­
78.
	 27.	 Marson AG, Kadir ZA, Hutton JL, Chadwick DW, et al. The new
antiepileptic drugs: a systematic review of their efficacy and toler-
ability. Epilepsia. 1997;38:859-­
80.
	 28.	 Vajda FJ. Pharmacotherapy of epilepsy: new armamentar-
ium, new issues. J Clin Neurosci Off J Neurosurg Soci Australa.
2007;14:813-­
23.
	 29.	 International League Against Epilepsy. Epilepsy Syndromes,
https://www.epile​
psydi​
agnos​
is.org/syndr​
ome/epile​
psy-­
syndr​
ome-­
group​
overv​
iew.html (accessed April 25, 2021)
	 30.	 Zaccara G, Perucca E. Interactions between antiepileptic drugs,
and between antiepileptic drugs and other drugs. Epileptic Disord.
2014;16:409-­
31.
	 31.	 Sills GJ, Rogawski MA. Mechanisms of action of currently used an-
tiseizure drugs. Neuropharmacology. 2020;168:107966.
	 32.	 Devinsky O, Vezzani A, O'Brien TJ, Jette N, Scheffer IE, de Curtis
M, et al. Epilepsy. Nat Rev Dis Primers. 2018;4:18024.
	 33.	 Karceski S, Morrell M, Carpenter D. The expert consensus guide-
line series treatment of epilepsy. Epilepsy Behav. 2001;2:A1-­
A50.
	 34.	 Marson AG, Al-­
Kharusi AM, Alwaidh M, Appleton R, Baker GA,
Chadwick DW, et al. The SANAD study of effectiveness of carba-
mazepine, gabapentin, lamotrigine, oxcarbazepine, or topiramate
for treatment of partial epilepsy: an unblinded randomised con-
trolled trial. Lancet. 2007;369:1000-­
15.
	 35.	 Marson AG, Al-­
Kharusi AM, Alwaidh M, Appleton R, Baker GA,
Chadwick DW, et al. The SANAD study of effectiveness of val-
proate, lamotrigine, or topiramate for generalised and unclassifi-
able epilepsy: an unblinded randomised controlled trial. Lancet.
2007;369:1016-­
26.
	 36.	 Marson A, Burnside G, Appleton R, Smith D, Leach JP, Sills G, et al.
The SANAD II study of the effectiveness and cost-­
effectiveness
of valproate versus levetiracetam for newly diagnosed gener-
alised and unclassifiable epilepsy: an open-­
label, non-­
inferiority,
multicentre, phase 4, randomised controlled trial. Lancet.
2021;397:1375-­
86.
	 37.	 Marson A, Burnside G, Appleton R, Smith D, Leach JP, Sills G, et al.
The SANAD II study of the effectiveness and cost-­
effectiveness
of levetiracetam, zonisamide, or lamotrigine for newly diagnosed
focal epilepsy: an open-­
label, non-­
inferiority, multicentre, phase 4,
randomised controlled trial. Lancet. 2021;397:1363-­
74.
	 38.	 Privitera MD, Brodie MJ, Mattson RH, Chadwick DW, Neto W,
Wang S, et al. Topiramate, carbamazepine and valproate mono-
therapy: double-­
blind comparison in newly diagnosed epilepsy.
Acta Neurol Scand. 2003;107:165-­
75.
	 39.	 Rowan AJ, Ramsay RE, Collins JF, Pryor F, Boardman KD, Uthman
BM, et al. New onset geriatric epilepsy: a randomized study
of gabapentin, lamotrigine, and carbamazepine. Neurology.
2005;64:1868-­
73.
	 40.	 Brodie MJ, Perucca E, Ryvlin P, Ben-­
Menachem E, Meencke H-­
J,
et al. Comparison of levetiracetam and controlled-­
release carba-
mazepine in newly diagnosed epilepsy. Neurology. 2007;68:402-­
8.
	 41.	 Brodie MJ, Overstall PW, Multicentre GL. Multicentre, double-­
blind, randomised comparison between lamotrigine and
carbamazepine in elderly patients with newly diagnosed epi-
lepsy. The UK Lamotrigine Elderly Study Group. Epilepsy Res.
1999;37:81-­
7.
	 42.	 Baulac M, Brodie MJ, Patten A, Segieth J, Giorgi L, et al. Efficacy
and tolerability of zonisamide versus controlled-­
release carbamaz-
epine for newly diagnosed partial epilepsy: a phase 3, randomised,
double-­
blind, non-­
inferiority trial. Lancet Neurol. 2012;11:579-­
88.
	 43.	 Chadwick D. Safety and efficacy of vigabatrin and carbamazepine
in newly diagnosed epilepsy: a multicentre randomised double-­
blind study. Vigabatrin European Monotherapy Study Group.
Lancet. 1999;354:13-­
9.
	 44.	 Kwan P, Brodie MJ, Kälviäinen R, Yurkewicz L, Weaver J, Knapp
LE, et al. Efficacy and safety of pregabalin versus lamotrigine
in patients with newly diagnosed partial seizures: a phase 3,
double-­
blind, randomised, parallel-­
group trial. Lancet Neurol.
2011;10:881-­
90.
	 45.	 Glauser TA, Cnaan A, Shinnar S, et al. Ethosuximide, valproic acid,
and lamotrigine in childhood absence epilepsy: initial monother-
apy outcomes at 12 months. Epilepsia. 2013;54:141-­
55.
	 46.	 Saetre E, Perucca E, Isojärvi J, Gjerstad L, et al. An international
multicenter randomized double-­
blind controlled trial of lamotrig-
ine and sustained-­
release carbamazepine in the treatment of newly
diagnosed epilepsy in the elderly. Epilepsia. 2007;48:1292-­
302.
	 47.	 French JA, Krauss GL, Wechsler RT, Wang X-­
F, DiVentura B, Brandt
C, et al. Perampanel for tonic-­
clonic seizures in idiopathic general-
ized epilepsy A randomized trial. Neurology. 2015;85:950-­
7.
	 48.	 French J, Brandt C, Friedman D, Biton V, Knapp L, Pitman V,
et al. Adjunctive use of controlled-­
release pregabalin in adults with
treatment-­
resistant partial seizures: a double-­
blind, randomized,
placebo-­
controlled trial. Epilepsia. 2014;55:1220-­
8.
	 49.	 Brodie MJ, Rosenfeld WE, Vazquez B, Sachdeo R, Perdomo C,
Mann A, et al. Rufinamide for the adjunctive treatment of par-
tial seizures in adults and adolescents: a randomized placebo-­
controlled trial. Epilepsia. 2009;50:1899-­
909.
	 50.	 Brodie MJ, Lerche H, Gil-­
Nagel A, Elger C, Hall S, Shin P, et al.
Efficacy and safety of adjunctive ezogabine (retigabine) in refrac-
tory partial epilepsy. Neurology. 2010;75:1817-­
24.
	 51.	 French JA, Mosier M, Walker S, Sommerville K, Sussman N, et al.
A double-­
blind, placebo-­
controlled study of vigabatrin three g/day
in patients with uncontrolled complex partial seizures. Vigabatrin
Protocol 024 Investigative Cohort. Neurology. 1996;46:54-­
61.
	 52.	 Elger C, Halász P, Maia J, Almeida L, Soares-­
da-­
Silva P, et al.
Efficacy and safety of eslicarbazepine acetate as adjunctive treat-
ment in adults with refractory partial-­
onset seizures: a random-
ized, double-­
blind, placebo-­
controlled, parallel-­
group phase III
study. Epilepsia. 2009;50:454-­
63.
	 53.	 French JA, Baroldi P, Brittain ST, Johnson JK, et al. Efficacy and
safety of extended-­
release oxcarbazepine (Oxtellar XR) as adjunc-
tive therapy in patients with refractory partial-­
onset seizures: a
randomized controlled trial. Acta Neurol Scand. 2014;129:143-­
53.
350  |     HAKAMI
	 54.	 French JA, Costantini C, Brodsky A, von Rosenstiel P, et al.
Adjunctive brivaracetam for refractory partial-­
onset seizures: a
randomized, controlled trial. Neurology. 2010;75:519-­
25.
	 55.	 Baulac M, Rosenow F, Toledo M, Terada K, Li T, De Backer M,
et al. Efficacy, safety, and tolerability of lacosamide monotherapy
versus controlled-­
release carbamazepine in patients with newly
diagnosed epilepsy: a phase 3, randomised, double-­
blind, non-­
inferiority trial. Lancet Neurol. 2017;16:43-­
54.
	 56.	 French JA, Abou-­
Khalil BW, Leroy RF, Yacubian EMT, Shin P, Hall S,
et al. Randomized, double-­
blind, placebo-­
controlled trial of ezoga-
bine (retigabine) in partial epilepsy. Neurology. 2011;76:1555-­
63.
	 57.	 Dam M, Ekberg R, Loyning Y, et al. A double-­
blind study compar-
ing oxcarbazepine and carbamazepine in patients with newly diag-
nosed, previously untreated epilepsy. Epilepsy Res. 1989;3:70-­
6.
	 58.	 Kalviainen R, Aikia M, Saukkonen AM, et al. Vigabatrin vs carba-
mazepine monotherapy in patients with newly diagnosed epilepsy.
A randomized, controlled study. Arch Neurol. 1995;52:989-­
96.
	 59.	 Ramsay E, Faught E, Krumholz A, Naritoku D, Privitera M,
Schwarzman L, et al. Efficacy, tolerability, and safety of rapid
initiation of topiramate versus phenytoin in patients with new-­
onset epilepsy: a randomized double-­
blind clinical trial. Epilepsia.
2010;51:1970-­
7.
	 60.	 Trinka E, Marson AG, Van Paesschen W, Kälviäinen R, Marovac J,
Duncan B, et al. KOMET: an unblinded, randomised, two parallel-­
group, stratified trial comparing the effectiveness of levetiracetam
with controlled-­
release carbamazepine and extended-­
release so-
dium valproate as monotherapy in patients with newly diagnosed
epilepsy. J Neurol Neurosurg Psychiatry. 2013;84:1138-­
47.
	 61.	 Trinka E, Ben-­
Menachem E, Kowacs PA, Elger C, Keller B, Löffler
K, et al. Efficacy and safety of eslicarbazepine acetate versus
controlled-­
release carbamazepine monotherapy in newly diag-
nosed epilepsy: a phase III double-­
blind, randomized, parallel-­
group, multicenter study. Epilepsia. 2018;59:479-­
91.
	 62.	 Hakami T, Todaro M, Petrovski S, MacGregor L, Velakoulis D, Tan
M, et al. Substitution monotherapy with levetiracetam vs older
antiepileptic drugs: a randomized comparative trial. Arch Neurol.
2012;69:1563-­
71.
	 63.	 Lattanzi S, Zaccara G, Giovannelli F, Grillo E, Nardone R,
Silvestrini M, et al. Antiepileptic monotherapy in newly diagnosed
focal epilepsy. A network meta-­
analysis. Acta Neurol Scand.
2019;139:33-­
41.
	 64.	 Lattanzi S, Trinka E, Del Giovane C, Nardone R, Silvestrini M,
Brigo F, et al. Antiepileptic drug monotherapy for epilepsy in the
elderly: A systematic review and network meta-­
analysis. Epilepsia.
2019;60:2245-­
54.
	 65.	 Sharawat IK, Panda PK, Kasinathan A, Panda P, Dawman L, Joshi
K, et al. Efficacy and tolerability of fenfluramine in patients with
Dravet syndrome: A systematic review and meta-­
analysis. Seizure.
2021;85:119-­
26.
	 66.	 Lattanzi S, Brigo F, Trinka E, Zaccara G, Striano P, Del Giovane C,
et al. Adjunctive cannabidiol in patients with dravet syndrome: a
systematic review and meta-­
analysis of efficacy and safety. CNS
Drugs. 2020;34:229-­
41.
	 67.	 Veroniki AA, Cogo E, Rios P, Straus SE, Finkelstein Y, Kealey R,
et al. Comparative safety of anti-­
epileptic drugs during pregnancy:
a systematic review and network meta-­
analysis of congenital mal-
formations and prenatal outcomes. BMC Med. 2017;15:95.
	 68.	 Lattanzi S, Brigo F, Cagnetti C, Trinka E, Silvestrini M, et al. Efficacy
and safety of adjunctive cannabidiol in patients with Lennox-­
Gastaut syndrome: a systematic review and meta-­
analysis. CNS
Drugs. 2018;32:905-­
16.
	 69.	 Brigo F, Del Giovane C, Nardone R, Trinka E, Lattanzi S, et al.
Intravenous antiepileptic drugs in adults with benzodiazepine-­
resistant convulsive status epilepticus: a systematic review and
network meta-­
analysis. Epilepsy Behav. 2019;101:106466.
	 70.	 Krumholz A, Shinnar S, French J, et al. Evidence-­
based guideline:
management of an unprovoked first seizure in adults: report of the
guideline development subcommittee of the American Academy
of Neurology and the American Epilepsy Society. Neurology.
2015;85:1526-­
7.
	 71.	 Glauser T, Ben-­
Menachem E, Bourgeois B, Cnaan A, Chadwick
D, Guerreiro C, et al. ILAE treatment guidelines: evidence-­
based
analysis of antiepileptic drug efficacy and effectiveness as initial
monotherapy for epileptic seizures and syndromes. Epilepsia.
2006;47:1094-­
120.
	 72.	 Glauser T, Ben-­
Menachem E, Bourgeois B, Cnaan A, Guerreiro C,
Kälviäinen R, et al. Updated ILAE evidence review of antiepileptic
drug efficacy and effectiveness as initial monotherapy for epilep-
tic seizures and syndromes. Epilepsia. 2013;54:551-­
63.
	 73.	 Appleton RE, Freeman A, Cross JH. Diagnosis and management of
the epilepsies in children: a summary of the partial update of the
2012 NICE epilepsy guideline. Arch Dis Child. 2012;97:1073-­
6.
	 74.	 Nunes VD, Sawyer L, Neilson J, Sarri G, Cross JH, et al. Diagnosis
and management of the epilepsies in adults and children: summary
of updated NICE guidance. BMJ. 2012;344:e281.
	 75.	 Network SIG. SIGN 143-­
Diagnosis and management of epilepsy in
adults. 2018.
	 76.	 Network SIG. Diagnosis and Management of Epilepsy in adults.
SIGN national clinical guideline 143. Edinburgh: SIGN; 2015.
	 77.	 Stephen LJ, Sills GJ, Leach JP, Butler E, Parker P, Hitiris N, et al.
Sodium valproate versus lamotrigine: a randomised comparison
of efficacy, tolerability and effects on circulating androgenic hor-
mones in newly diagnosed epilepsy. Epilepsy Res. 2007;75:122-­
9.
	 78.	 Brodie MJ, Richens A, Yuen AW. Double-­
blind comparison of la-
motrigine and carbamazepine in newly diagnosed epilepsy. UK
Lamotrigine/Carbamazepine Monotherapy Trial Group. Lancet.
1995;345:476-­
9.
	 79.	 Reunanen M, Dam M, Yuen AW. A randomised open multicentre
comparative trial of lamotrigine and carbamazepine as mono-
therapy in patients with newly diagnosed or recurrent epilepsy.
Epilepsy Res. 1996;23:149-­
55.
	 80.	 Nieto-­
Barrera M, Brozmanova M, Capovilla G, Christe W, Pedersen
B, Kane K, et al. A comparison of monotherapy with lamotrigine or
carbamazepine in patients with newly diagnosed partial epilepsy.
Epilepsy Res. 2001;46:145-­
55.
	 81.	 Tanganelli P, Regesta G. Vigabatrin vs. carbamazepine monother-
apy in newly diagnosed focal epilepsy: a randomized response
conditional cross-­
over study. Epilepsy Res. 1996;25:257-­
62.
	 82.	 Aikia M, Jutila L, Salmenpera T, Mervaala E, Kalviainen R, et al.
Comparison of the cognitive effects of tiagabine and carbamaz-
epine as monotherapy in newly diagnosed adult patients with
partial epilepsy: pooled analysis of two long-­
term, randomized,
follow-­
up studies. Epilepsia. 2006;47:1121-­
7.
	 83.	 Christe W, Krämer G, Vigonius U, Pohlmann H, Steinhoff BJ,
Brodie MJ, et al. A double-­
blind controlled clinical trial: oxcarba-
zepine versus sodium valproate in adults with newly diagnosed
epilepsy. Epilepsy Res. 1997;26:451-­
60.
	 84.	 Tomson T, Battino D, Perucca E. Teratogenicity of antiepileptic
drugs. Curr Opin Neurol. 2019;32:246-­
52.
	 85.	 Roger JP, Michael AR. Antiseizure drugs. In: Katzung BG, editor.
Basic  clinical pharmacology. McGraw-­
Hill; 2018, p. 409-­
39.
	 86.	 French JA. Seizure exacerbation by antiepileptic drugs. Epilepsy
Curr. 2005;5:192-­
3.
	 87.	 Silvennoinen K, Lange N, Zagaglia S, Balestrini S, Androsova G,
Wassenaar M, et al. Comparative effectiveness of antiepileptic drugs
in juvenile myoclonic epilepsy. Epilepsia Open. 2019;4:420-­
30.
	 88.	 Wallace A, Wirrell E, Kenney-­
Jung DL. Pharmacotherapy for
dravet syndrome. Paediatr Drugs. 2016;18:197-­
208.
	 89.	 Lagae L, Sullivan J, Knupp K, Laux L, Polster T, Nikanorova M,
et al. Fenfluramine hydrochloride for the treatment of seizures in
    | 351
HAKAMI
Dravet syndrome: a randomised, double-­
blind, placebo-­
controlled
trial. Lancet. 2019;394:2243-­
54.
	 90.	 Trinka E, Cock H, Hesdorffer D, Rossetti AO, Scheffer IE, Shinnar
S, et al. A definition and classification of status epilepticus–­
report
of the ILAE task force on classification of status epilepticus.
Epilepsia. 2015;56:1515-­
23.
	 91.	 Glauser T, Shinnar S, Gloss D, Alldredge B, Arya R, Bainbridge J,
et al. Evidence-­
based guideline: treatment of convulsive status ep-
ilepticus in children and adults: report of the guideline committee
of the American Epilepsy Society. Epilepsy Curr. 2016;16:48-­
61.
	 92.	 Haut SR. Seizure clusters: characteristics and treatment. Curr
Opin Neurol. 2015;28:143-­
50.
	 93.	 Werhahn KJ, Trinka E, Dobesberger J, Unterberger I, Baum P,
Deckert-­
Schmitz M, et al. A randomized, double-­
blind comparison
of antiepileptic drug treatment in the elderly with new-­
onset focal
epilepsy. Epilepsia. 2015;56:450-­
9.
	 94.	 Ettinger AB. Psychotropic effects of antiepileptic drugs.
Neurology. 2006;67:1916-­
25.
	 95.	 Tomson T, Battino D, Bonizzoni E, Craig J, Lindhout D, Sabers A,
et al. Dose-­
dependent risk of malformations with antiepileptic
drugs: an analysis of data from the EURAP epilepsy and pregnancy
registry. Lancet Neurol. 2011;10:609-­
17.
	 96.	 Patorno E, Bohn RL, Wahl PM, et al. Anticonvulsant medications
and the risk of suicide, attempted suicide, or violent death. JAMA.
2010;303(14):1401-­
9.
	 97.	 Gibbons RD, Hur K, Brown CH, Mann JJ, et al. Relationship be-
tween antiepileptic drugs and suicide attempts in patients with
bipolar disorder. Arch Gen Psychiatry. 2009;66:1354-­
60.
	 98.	 Shiek Ahmad B, Hill KD, O'Brien TJ, Gorelik A, Habib N, Wark JD,
et al. Falls and fractures in patients chronically treated with antie-
pileptic drugs. Neurology. 2012;79:145-­
51.
	 99.	 Chukwu J, Delanty N, Webb D, Cavalleri GL, et al. Weight change,
genetics and antiepileptic drugs. Expert Rev Clin Pharmacol.
2014;7:43-­
51.
	
100.	 French JA, Perucca E, Sander JW, Bergfeldt L, Baulac M, Auerbach
DS, et al. FDA safety warning on the cardiac effects of lamotrigine:
an advisory from the Ad Hoc ILAE/AES task force. Epilepsy Curr.
2021: 1535759721996344.
	
101.	 Dixon R, Alexander S, Brickel N. Effect of lamotrigine on the PR
interval in healthy subjects. Br J Clin Pharmacol. 2011;71:961-­
2.
	
102.	 Perucca E, Brodie MJ, Kwan P, Tomson T, et al. 30 years of second-­
generation antiseizure medications: impact and future perspec-
tives. Lancet Neurol. 2020;19:544-­
56.
	
103.	 Chen Z, Brodie MJ, Kwan P. What has been the impact of new drug
treatments on epilepsy? Curr Opin Neurol. 2020;33:185-­
90.
	
104.	 Drew L. Gene therapy targets epilepsy. Nature. 2018;564:S10-­
1.
	
105.	 Rogawski MA, Zolkowska D. Anticonvulsant activity of steroids.
Google Patents. 2021.
How to cite this article: Hakami T. Neuropharmacology of
Antiseizure Drugs. Neuropsychopharmacol Rep. 2021;41:​
336–­351. https://doi.org/10.1002/npr2.12196

More Related Content

What's hot

Missed Diagnoses association in Rear end collisions
Missed Diagnoses association in Rear end collisions  Missed Diagnoses association in Rear end collisions
Missed Diagnoses association in Rear end collisions Nelson Hendler
 
Whistler Anesthesia Symposium Regional Anesthesia Outcomes 2017
Whistler Anesthesia Symposium Regional Anesthesia Outcomes 2017Whistler Anesthesia Symposium Regional Anesthesia Outcomes 2017
Whistler Anesthesia Symposium Regional Anesthesia Outcomes 2017Colin McCartney
 
Balancing Risks and Benefits of Regional Anaesthesia
Balancing Risks and Benefits of Regional AnaesthesiaBalancing Risks and Benefits of Regional Anaesthesia
Balancing Risks and Benefits of Regional AnaesthesiaColin McCartney
 
Journal club 1 jan 2020
Journal club 1 jan 2020Journal club 1 jan 2020
Journal club 1 jan 2020NeurologyKota
 
Evaluating Chronic Pain Patients Using Methods from Johns Hopkins Hospital Ph...
Evaluating Chronic Pain Patients Using Methods from Johns Hopkins Hospital Ph...Evaluating Chronic Pain Patients Using Methods from Johns Hopkins Hospital Ph...
Evaluating Chronic Pain Patients Using Methods from Johns Hopkins Hospital Ph...Nelson Hendler
 
SISTEMA NERVIOSO
SISTEMA NERVIOSOSISTEMA NERVIOSO
SISTEMA NERVIOSOeglimar00
 
Pemf on early knee osteoarthritis
Pemf on early knee osteoarthritisPemf on early knee osteoarthritis
Pemf on early knee osteoarthritisPainezee Specialist
 
Does medicine need academic anesthesia
Does medicine need academic anesthesiaDoes medicine need academic anesthesia
Does medicine need academic anesthesiaColin McCartney
 
Depression Involved in the Chemotherapy Induced Event-based Prospective Memor...
Depression Involved in the Chemotherapy Induced Event-based Prospective Memor...Depression Involved in the Chemotherapy Induced Event-based Prospective Memor...
Depression Involved in the Chemotherapy Induced Event-based Prospective Memor...IJEAB
 
ZMPCZM016000.11.04
ZMPCZM016000.11.04 ZMPCZM016000.11.04
ZMPCZM016000.11.04 painezeeman
 
Why I do (some) blocks under general anaesthesia?
Why I do (some) blocks under general anaesthesia?Why I do (some) blocks under general anaesthesia?
Why I do (some) blocks under general anaesthesia?Colin McCartney
 
ZMPCZM016000.11.08
ZMPCZM016000.11.08ZMPCZM016000.11.08
ZMPCZM016000.11.08painezeeman
 

What's hot (19)

Missed Diagnoses association in Rear end collisions
Missed Diagnoses association in Rear end collisions  Missed Diagnoses association in Rear end collisions
Missed Diagnoses association in Rear end collisions
 
Whistler Anesthesia Symposium Regional Anesthesia Outcomes 2017
Whistler Anesthesia Symposium Regional Anesthesia Outcomes 2017Whistler Anesthesia Symposium Regional Anesthesia Outcomes 2017
Whistler Anesthesia Symposium Regional Anesthesia Outcomes 2017
 
Balancing Risks and Benefits of Regional Anaesthesia
Balancing Risks and Benefits of Regional AnaesthesiaBalancing Risks and Benefits of Regional Anaesthesia
Balancing Risks and Benefits of Regional Anaesthesia
 
A Retrospective Study about Autonomy of the Patients with Neurological Disord...
A Retrospective Study about Autonomy of the Patients with Neurological Disord...A Retrospective Study about Autonomy of the Patients with Neurological Disord...
A Retrospective Study about Autonomy of the Patients with Neurological Disord...
 
International Journal of Clinical Cardiology & Research
International Journal of Clinical Cardiology & ResearchInternational Journal of Clinical Cardiology & Research
International Journal of Clinical Cardiology & Research
 
Chiro Cervical Adjustment
Chiro Cervical AdjustmentChiro Cervical Adjustment
Chiro Cervical Adjustment
 
Journal club 1 jan 2020
Journal club 1 jan 2020Journal club 1 jan 2020
Journal club 1 jan 2020
 
Inmunos Y Spr
Inmunos Y SprInmunos Y Spr
Inmunos Y Spr
 
Evaluating Chronic Pain Patients Using Methods from Johns Hopkins Hospital Ph...
Evaluating Chronic Pain Patients Using Methods from Johns Hopkins Hospital Ph...Evaluating Chronic Pain Patients Using Methods from Johns Hopkins Hospital Ph...
Evaluating Chronic Pain Patients Using Methods from Johns Hopkins Hospital Ph...
 
SISTEMA NERVIOSO
SISTEMA NERVIOSOSISTEMA NERVIOSO
SISTEMA NERVIOSO
 
Articulo linea
Articulo lineaArticulo linea
Articulo linea
 
Pemf on early knee osteoarthritis
Pemf on early knee osteoarthritisPemf on early knee osteoarthritis
Pemf on early knee osteoarthritis
 
Austin Oncology
Austin OncologyAustin Oncology
Austin Oncology
 
Austin Oncology
Austin OncologyAustin Oncology
Austin Oncology
 
Does medicine need academic anesthesia
Does medicine need academic anesthesiaDoes medicine need academic anesthesia
Does medicine need academic anesthesia
 
Depression Involved in the Chemotherapy Induced Event-based Prospective Memor...
Depression Involved in the Chemotherapy Induced Event-based Prospective Memor...Depression Involved in the Chemotherapy Induced Event-based Prospective Memor...
Depression Involved in the Chemotherapy Induced Event-based Prospective Memor...
 
ZMPCZM016000.11.04
ZMPCZM016000.11.04 ZMPCZM016000.11.04
ZMPCZM016000.11.04
 
Why I do (some) blocks under general anaesthesia?
Why I do (some) blocks under general anaesthesia?Why I do (some) blocks under general anaesthesia?
Why I do (some) blocks under general anaesthesia?
 
ZMPCZM016000.11.08
ZMPCZM016000.11.08ZMPCZM016000.11.08
ZMPCZM016000.11.08
 

Similar to fármacos antiepilépticos

Advances in current medication and new therapeutic approaches in epilepsy
Advances in current medication and new therapeutic approaches in epilepsyAdvances in current medication and new therapeutic approaches in epilepsy
Advances in current medication and new therapeutic approaches in epilepsySelf-employed researcher
 
Navigating anti epileptic medications in difficult to treat epilepsies
Navigating anti epileptic medications in difficult to treat epilepsiesNavigating anti epileptic medications in difficult to treat epilepsies
Navigating anti epileptic medications in difficult to treat epilepsiesPramod Krishnan
 
Paper id 71201931
Paper id 71201931Paper id 71201931
Paper id 71201931IJRAT
 
Anestesia pacientes criticos - urgencias
Anestesia   pacientes criticos - urgenciasAnestesia   pacientes criticos - urgencias
Anestesia pacientes criticos - urgenciasJOSE CARLOS VARGAS
 
Prospective Observational Study of Sodium Valproate in Seizure Control and As...
Prospective Observational Study of Sodium Valproate in Seizure Control and As...Prospective Observational Study of Sodium Valproate in Seizure Control and As...
Prospective Observational Study of Sodium Valproate in Seizure Control and As...iosrjce
 
Neurological DisordersNeurological disorders, such as headaches, s.docx
Neurological DisordersNeurological disorders, such as headaches, s.docxNeurological DisordersNeurological disorders, such as headaches, s.docx
Neurological DisordersNeurological disorders, such as headaches, s.docxTanaMaeskm
 
recent seminar topic for m.pharm
recent seminar topic for m.pharmrecent seminar topic for m.pharm
recent seminar topic for m.pharmnilesh1208
 
Anticholinergic Drugs And Risk Of Dementia Case-Control Study
Anticholinergic Drugs And Risk Of Dementia  Case-Control StudyAnticholinergic Drugs And Risk Of Dementia  Case-Control Study
Anticholinergic Drugs And Risk Of Dementia Case-Control StudySandra Long
 
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...Dr. Rafael Higashi
 
SEMINAR ON categories of patients of personalized medicine.pptx
SEMINAR ON categories of patients of personalized medicine.pptxSEMINAR ON categories of patients of personalized medicine.pptx
SEMINAR ON categories of patients of personalized medicine.pptxPawanDhamala1
 
Choosing the right antiseizure medication for epilepsy
Choosing the right antiseizure medication for epilepsy  Choosing the right antiseizure medication for epilepsy
Choosing the right antiseizure medication for epilepsy Ersifa Fatimah
 
Management of Chronic Insomnia In Adults
Management of Chronic Insomnia In AdultsManagement of Chronic Insomnia In Adults
Management of Chronic Insomnia In AdultsPaul Coelho, MD
 
Adverse Drug Reactions - Basics and Classification
Adverse Drug Reactions - Basics and ClassificationAdverse Drug Reactions - Basics and Classification
Adverse Drug Reactions - Basics and ClassificationVenkateswaramurthy N
 
Quick Clinical Review of Antipsychotics
Quick Clinical Review of AntipsychoticsQuick Clinical Review of Antipsychotics
Quick Clinical Review of AntipsychoticsShah Parind
 
pharmaco pharmaco-epidemiology
pharmaco pharmaco-epidemiologypharmaco pharmaco-epidemiology
pharmaco pharmaco-epidemiologyfarranajwa
 
BIOSIMILARS AND ITS FUTUE
BIOSIMILARS AND ITS FUTUEBIOSIMILARS AND ITS FUTUE
BIOSIMILARS AND ITS FUTUEAsmitaGpt
 

Similar to fármacos antiepilépticos (20)

Advances in current medication and new therapeutic approaches in epilepsy
Advances in current medication and new therapeutic approaches in epilepsyAdvances in current medication and new therapeutic approaches in epilepsy
Advances in current medication and new therapeutic approaches in epilepsy
 
Navigating anti epileptic medications in difficult to treat epilepsies
Navigating anti epileptic medications in difficult to treat epilepsiesNavigating anti epileptic medications in difficult to treat epilepsies
Navigating anti epileptic medications in difficult to treat epilepsies
 
Refractory epilepsy
Refractory epilepsyRefractory epilepsy
Refractory epilepsy
 
Paper id 71201931
Paper id 71201931Paper id 71201931
Paper id 71201931
 
Anestesia pacientes criticos - urgencias
Anestesia   pacientes criticos - urgenciasAnestesia   pacientes criticos - urgencias
Anestesia pacientes criticos - urgencias
 
Prospective Observational Study of Sodium Valproate in Seizure Control and As...
Prospective Observational Study of Sodium Valproate in Seizure Control and As...Prospective Observational Study of Sodium Valproate in Seizure Control and As...
Prospective Observational Study of Sodium Valproate in Seizure Control and As...
 
Guia NICE epilepsia
Guia NICE epilepsiaGuia NICE epilepsia
Guia NICE epilepsia
 
Neurological DisordersNeurological disorders, such as headaches, s.docx
Neurological DisordersNeurological disorders, such as headaches, s.docxNeurological DisordersNeurological disorders, such as headaches, s.docx
Neurological DisordersNeurological disorders, such as headaches, s.docx
 
Drug resistant epilepsy
Drug resistant epilepsyDrug resistant epilepsy
Drug resistant epilepsy
 
recent seminar topic for m.pharm
recent seminar topic for m.pharmrecent seminar topic for m.pharm
recent seminar topic for m.pharm
 
Anticholinergic Drugs And Risk Of Dementia Case-Control Study
Anticholinergic Drugs And Risk Of Dementia  Case-Control StudyAnticholinergic Drugs And Risk Of Dementia  Case-Control Study
Anticholinergic Drugs And Risk Of Dementia Case-Control Study
 
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
 
SEMINAR ON categories of patients of personalized medicine.pptx
SEMINAR ON categories of patients of personalized medicine.pptxSEMINAR ON categories of patients of personalized medicine.pptx
SEMINAR ON categories of patients of personalized medicine.pptx
 
Choosing the right antiseizure medication for epilepsy
Choosing the right antiseizure medication for epilepsy  Choosing the right antiseizure medication for epilepsy
Choosing the right antiseizure medication for epilepsy
 
Management of Chronic Insomnia In Adults
Management of Chronic Insomnia In AdultsManagement of Chronic Insomnia In Adults
Management of Chronic Insomnia In Adults
 
Adverse Drug Reactions - Basics and Classification
Adverse Drug Reactions - Basics and ClassificationAdverse Drug Reactions - Basics and Classification
Adverse Drug Reactions - Basics and Classification
 
Quick Clinical Review of Antipsychotics
Quick Clinical Review of AntipsychoticsQuick Clinical Review of Antipsychotics
Quick Clinical Review of Antipsychotics
 
pharmaco pharmaco-epidemiology
pharmaco pharmaco-epidemiologypharmaco pharmaco-epidemiology
pharmaco pharmaco-epidemiology
 
BIOSIMILARS AND ITS FUTUE
BIOSIMILARS AND ITS FUTUEBIOSIMILARS AND ITS FUTUE
BIOSIMILARS AND ITS FUTUE
 
Medical management of epilepsy
Medical management of epilepsyMedical management of epilepsy
Medical management of epilepsy
 

Recently uploaded

Boyles law module in the grade 10 science
Boyles law module in the grade 10 scienceBoyles law module in the grade 10 science
Boyles law module in the grade 10 sciencefloriejanemacaya1
 
TOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physicsTOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physicsssuserddc89b
 
Analytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptxAnalytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptxSwapnil Therkar
 
Genomic DNA And Complementary DNA Libraries construction.
Genomic DNA And Complementary DNA Libraries construction.Genomic DNA And Complementary DNA Libraries construction.
Genomic DNA And Complementary DNA Libraries construction.k64182334
 
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...Sérgio Sacani
 
Call Girls in Munirka Delhi 💯Call Us 🔝9953322196🔝 💯Escort.
Call Girls in Munirka Delhi 💯Call Us 🔝9953322196🔝 💯Escort.Call Girls in Munirka Delhi 💯Call Us 🔝9953322196🔝 💯Escort.
Call Girls in Munirka Delhi 💯Call Us 🔝9953322196🔝 💯Escort.aasikanpl
 
Natural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsNatural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsAArockiyaNisha
 
Recombinant DNA technology( Transgenic plant and animal)
Recombinant DNA technology( Transgenic plant and animal)Recombinant DNA technology( Transgenic plant and animal)
Recombinant DNA technology( Transgenic plant and animal)DHURKADEVIBASKAR
 
Disentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOSTDisentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOSTSérgio Sacani
 
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...anilsa9823
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real timeSatoshi NAKAHIRA
 
A relative description on Sonoporation.pdf
A relative description on Sonoporation.pdfA relative description on Sonoporation.pdf
A relative description on Sonoporation.pdfnehabiju2046
 
Recombination DNA Technology (Microinjection)
Recombination DNA Technology (Microinjection)Recombination DNA Technology (Microinjection)
Recombination DNA Technology (Microinjection)Jshifa
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfSwapnil Therkar
 
Isotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on IoIsotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on IoSérgio Sacani
 
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroidsHubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroidsSérgio Sacani
 
Module 4: Mendelian Genetics and Punnett Square
Module 4:  Mendelian Genetics and Punnett SquareModule 4:  Mendelian Genetics and Punnett Square
Module 4: Mendelian Genetics and Punnett SquareIsiahStephanRadaza
 
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |aasikanpl
 

Recently uploaded (20)

Boyles law module in the grade 10 science
Boyles law module in the grade 10 scienceBoyles law module in the grade 10 science
Boyles law module in the grade 10 science
 
TOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physicsTOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physics
 
Analytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptxAnalytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptx
 
Genomic DNA And Complementary DNA Libraries construction.
Genomic DNA And Complementary DNA Libraries construction.Genomic DNA And Complementary DNA Libraries construction.
Genomic DNA And Complementary DNA Libraries construction.
 
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
 
Call Girls in Munirka Delhi 💯Call Us 🔝9953322196🔝 💯Escort.
Call Girls in Munirka Delhi 💯Call Us 🔝9953322196🔝 💯Escort.Call Girls in Munirka Delhi 💯Call Us 🔝9953322196🔝 💯Escort.
Call Girls in Munirka Delhi 💯Call Us 🔝9953322196🔝 💯Escort.
 
Natural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsNatural Polymer Based Nanomaterials
Natural Polymer Based Nanomaterials
 
Recombinant DNA technology( Transgenic plant and animal)
Recombinant DNA technology( Transgenic plant and animal)Recombinant DNA technology( Transgenic plant and animal)
Recombinant DNA technology( Transgenic plant and animal)
 
Disentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOSTDisentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOST
 
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real time
 
Engler and Prantl system of classification in plant taxonomy
Engler and Prantl system of classification in plant taxonomyEngler and Prantl system of classification in plant taxonomy
Engler and Prantl system of classification in plant taxonomy
 
A relative description on Sonoporation.pdf
A relative description on Sonoporation.pdfA relative description on Sonoporation.pdf
A relative description on Sonoporation.pdf
 
Recombination DNA Technology (Microinjection)
Recombination DNA Technology (Microinjection)Recombination DNA Technology (Microinjection)
Recombination DNA Technology (Microinjection)
 
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
 
Isotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on IoIsotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on Io
 
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroidsHubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
 
Module 4: Mendelian Genetics and Punnett Square
Module 4:  Mendelian Genetics and Punnett SquareModule 4:  Mendelian Genetics and Punnett Square
Module 4: Mendelian Genetics and Punnett Square
 
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
 

fármacos antiepilépticos

  • 1. 336  |   Neuropsychopharmacology Reports. 2021;41:336–351. wileyonlinelibrary.com/journal/nppr Received: 17 May 2021  |  Revised: 1 July 2021  |  Accepted: 6 July 2021 DOI: 10.1002/npr2.12196 R E V I E W A R T I C L E Neuropharmacology of Antiseizure Drugs Tahir Hakami This is an open access article under the terms of the Creat​ ive Commo​ ns Attri​ butio​ n-­ NonCo​ mmerc​ ial-­ NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-­ commercial and no modifications or adaptations are made. © 2021 The Authors. Neuropsychopharmacology Reports published by John Wiley Sons Australia, Ltd on behalf of The Japanese Society of Neuropsychopharmacology. The Faculty of Medicine, Jazan University, Jazan, Saudi Arabia Correspondence Tahir Hakami, The Faculty of Medicine, Jazan University, P.O.Box 114, Jazan 45142, Saudi Arabia. Email hakamit@jazanu.edu.sa Funding information The author received no financial support for the research, authorship, and/or publication of this article Abstract Background: Antiseizure drugs (ASDs) are the primary therapy for epilepsy, with more than 20 drugs introduced into clinical practice to date. These drugs are typi- cally grouped by their mechanisms of action and therapeutic spectrum. This article aims to educate non-­ neurologists and medical students about the new frontiers in the pharmacology of ASDs and presents the current state of the literature on the efficacy and tolerability of these agents. Methods: Randomized controlled trials, observational studies, and evidence-­ based meta-­ analyses of ASD efficacy and tolerability as initial monotherapy for epileptic seizures and syndromes were identified in PubMed, EMBASE, the Cochrane Library, and Elsevier Clinical Pharmacology. Results: The choice of ASD varies primarily according to the seizure type. Practical guidelines for ASD selection in patients with new-­ onset and drug-­ resistant epilepsy were recently published. The guidelines have shown that the newer-­ generation drugs, which have unique mechanistic and pharmacokinetic properties, are better tolerated but have similar efficacy compared with the older drugs. Several ASDs are effective as first-­ line monotherapy in focal seizures, including lamotrigine, carba- mazepine, phenytoin, levetiracetam, and zonisamide. Valproate remains the first-­ line drug for many patients with generalized and unclassified epilepsies. However, val- proate should be avoided, if possible, in women of childbearing potential because of teratogenicity. Toxicity profile precludes several drugs from use as first-­ line treat- ment, for example, vigabatrin, felbamate, and rufinamide. Conclusions: Antiseizure drugs have different pharmacologic profiles that should be considered when selecting and prescribing these agents for epilepsy. These include pharmacokinetic properties, propensity for drug-­ drug interactions, and adverse effects. K E Y W O R D S antiseizure drug, epilepsy, seizure types
  • 2.     | 337 HAKAMI 1 | INTRODUCTION Epilepsy is a chronic disorder of brain function characterized by recurrent seizures.1 At any one time, about 1% of the population requires medication to treat epilepsy.2 The antiseizure drug (ASD) is the mainstay of treatment. The choice of ASDs varies with dif- ferent seizure types and epileptic syndromes.3 The appropriately chosen ASD provides adequate seizure control in 60%–­ 70% of pa- tients.4 There is no general agreement on when treatment should start after a “first seizure,” but randomized controlled trials (RCTs) in adult and pediatric patients have shown that early treatment re- duces the recurrence for at least two years from the first seizure.5 The ASDs are often administered orally for a long period of time to prevent seizure recurrence. In designing a therapeutic approach, the use of a single drug (monotherapy) is preferred because of fewer adverse effects. However, 30% of the patients will continue to have uncontrolled seizures and multiple drugs are often used simultaneously.6 Monotherapy trials of two ASDs that are appro- priate first-­ line treatment for the patient's seizure type usually be initiated before combinations are tried. Patients who do not achieve seizure control following adequate trials with two or more appropri- ate drugs are considered drug-­ resistant.6 The International League Against Epilepsy (ILAE), in 2010, has proposed the definition of drug-­ resistant epilepsy (often used interchangeably with “'medically refractory”' and “'intractable”) as “failure of adequate trials of two tolerated, appropriately chosen and used antiseizure drug schedules (whether as monotherapies or in combination) to achieve sustained seizure freedom.”7 The older-­ generation ASDs were introduced into clinical practice more than four decades ago. Research has shown that 30%–­ 40% of people treated with an older ASD as monotherapy (including carbamazepine and valproate) experience adverse ef- fects that contribute to treatment failure.8 This spurred a continu- ing search for new ASDs with novel molecular targets that could provide optimal care for patients with epilepsy.9,10 The past three decades have seen the licensing of about 20 newer-­(second-­and third-­ ) generation ASDs with unique mechanisms of action and pharmacokinetics following a period of relative paucity.11 This ad- vent has expanded the epilepsy therapeutic armamentarium and allowed the drugs to match the individual patient's characteristics. The American Academy of Neurology (AAN) subcommittee reports in 2004 observed that newer ASDs were not different in controlling seizures but have better tolerability, particularly fewer neurotoxic adverse effects.12,13 In 2017, the ILAE published a new classifica- tion for seizure types and epilepsy syndrome to improve our un- derstanding of epilepsy and include missing seizure types.14-­ 17 This classification replaced the previous versions published in 198118 and 1989,19 and extended in 2010,20 and has been of paramount importance to accurately classify the patient's seizure type(s). In 2018, the AAN and American Epilepsy Society (AES) subcommit- tee published updated guidelines for ASD selection in adult and pediatric patients with new-­ onset and treatment-­ resistant epi- lepsy.21,22 However, the guidelines primarily relied on studies that compare the first-­and second-­ generation ASDs and found limited data for third-­ generation drugs. Given the rapid advance in the development of ASDs in recent years and the continuous updates in definitions, classifications and treatment guidelines for seizure types, and epilepsy syndromes, this article aims to provide non-­ neurologists and medical students with a complete overview of the new frontiers in the neuropharmacology of antiseizure drugs. The ASDs classification, pharmacokinetics, mechanisms of action, clinical use, and adverse effects are reviewed herein based on a comprehensive assessment of the current literature and recently published US and UK treatment guidelines. 1.1 | Definition and epidemiology of epilepsy Epilepsy is ”a neurological disorder that is characterized by an en- during predisposition to generate epileptic seizures and the asso- ciated cognitive, psychological, and social consequences”.1 Epilepsy is the third most common neurologic disorder; almost 10% of peo- ple will experience a seizure during their lives.2 The prevalence of epilepsy is 6.4 cases per 1,000 persons, and the annual incidence is 67.8/100,000 person-­ years.23 1.2 | Classification of seizure types and epilepsy syndromes The ILAE classification framework, which was revised in 2017, is the key tool for the diagnosis of individuals presenting with seizures.14-­ 17 According to that classification, epileptic seizures are classified into “focal,” “generalized,” and “unknown” seizures, while epilepsy is classified into “focal,” “generalized,” “combined gener- alized and focal,” and “unknown” epilepsy. Focal-­ onset seizures can be further described as focal aware (previously called simple partial), impaired awareness (complex partial), or focal to bilat- eral (secondarily generalized) tonic-­ clonic seizures. Generalized seizures are classified into generalized tonic-­ clonic (formerly idi- opathic) seizures, motor (myoclonic and other motor) seizures, and nonmotor (absence) seizures. Etiologies of seizures and epilepsy syndromes have been reintroduced in the 2017 ILAE classifica- tion of seizure types and epilepsy syndromes, to include “genetic,” “structural,” “metabolic,” “infectious,” “immune,” and “unknown.” For a detailed description of seizure types epilepsy syndromes, see the ILAE Web site.24 1.3 | Drug treatment of epilepsy Epilepsy can persist for years and often for the patient's lifetime. ASDs are the primary therapy for epilepsy and are symptomatic treatments that control seizures.3 The choice of ASDs varies with different seizure types and epileptic syndromes.3 ASDs have many different pharmacologic profiles that are relevant when selecting
  • 3. 338  |     HAKAMI and prescribing these agents in patients with epilepsy. This includes pharmacokinetic properties, propensity for drug-­ drug interactions, and adverse-­ effect profiles and toxicities. All older-­ generation ASDs have acute dose-­ related effects, primarily neurological effects.25,26 This led to treatment failure in 30%–­ 40% of people with epilepsy receiving an older drug as monotherapy.8 Hence, there has been a great deal of research into formulating better ASDs, primarily spurred by the fact that the older-­ generation ASDs do not provide optimal safety, tolerability or seizure control for many patients with epilepsy. Over the past three decades, 20 newer-­ generation ASDs have been approved for clinical use,11 and it was hoped that these drugs would have better efficacy in controlling seizures than older-­ generation drugs. However, the AAN subcommittee reports in 2004 and 2018 observed that the newer-­ generation ASDs were “not dif- ferent” in controlling seizures, but better tolerated than the older drugs.12,13,21,22 1.4 | Classifications of ASDs ASDs are classified as older (first-­ ) generation or newer (second- ­and third-­ ) generation agents. The older-­ generation ASDs intro- duced into clinical practice more than four decades ago include phenobarbital, phenytoin, primidone, ethosuximide, valproate, car- bamazepine, clonazepam, and clobazam. The “second-­ generation” ASDs, which have been approved for the treatment of epilepsy since the late 1980s, include, in chronological order, vigabatrin, oxcarbazepine, lamotrigine, gabapentin, felbamate, topiramate, tiagabine, levetiracetam, and zonisamide. The third-­ generation ASDs include, pregabalin, fosphenytoin, lacosamide, rufinamide, eslicarbazepine, retigabine (also known as ezogabine), peram- panel, brivaracetam, cannabidiol, stiripentol, cenobamate, and fenfluramine.11,12,27,28 The newer ASDs differ substantially in their mechanisms of action, spectra of activity, pharmacokinetics, and adverse effects profiles. Figure 1 presents the classification and year of introduction of ASDs available now. Current information on the other drugs in the pipeline can be found on the Epilepsy Foundation Web site.29 1.5 | Pharmacokinetics and drug interactions of ASDs ASDs are commonly used for long periods of time, and consideration of their pharmacokinetic properties is essential for avoiding toxicity and drug interactions.30 An ideal ASD should demonstrate complete absorption, linear kinetics, a long elimination half-­ life, and allowing once or twice-­ daily dosing. Other favorable properties include low protein binding, lack of active metabolites, and clearance by kidneys. Generally, all ASDs are well absorbed after oral administration, have good bioavailability, and readily cross the blood-­ brain barrier. Many ASDs are medium to long-­ acting drugs (have half-­ lives of 12 hours) and can be administered twice or three times a day. Phenobarbital, phenytoin, zonisamide, eslicarbazepine, and perampanel can often be administered once daily. Extended-­ release preparations of drugs that have short half-­ lives (eg, carbamazepine, valproate, leveti- racetam, and lamotrigine) may decrease the incidence of adverse ef- fects and allow once-­ daily dosing. For optimum therapy, therapeutic drug concentration should be monitored in individual patients for some drugs. Examples include carbamazepine, phenytoin, and val- proate. Drug levels can be helpful (1) to guide dose adjustments, (2) when breakthrough seizures occur, (3) when an interacting medica- tion is added, (4) during pregnancy, (5) to assess compliance, and (6) to determine whether adverse effects are related to drug levels. Clinically relevant pharmacokinetic profiles of ASDs are summarized in Table 1. In general, the ASD should be started at a low dose, with incre- ments over several weeks to establish an effective and tolerable regimen. Some agents do not require titration, such as gabapentin and levetiracetam. Clinicians should be aware of certain factors that affect dosing. These factors include nonlinear relationships be- tween dose and drug exposure (eg, phenytoin) and the influence of hepatic or renal impairment on clearance. Metabolism of phenytoin is nonlinear; elimination kinetics shift from first order at a low dose to zero-­ order at moderate to high dose. As a subsequent, any small increases in the dose may increase phenytoin half-­ life and serum concentration, and a patient may quickly develop toxicity. A com- mon clinical error is to increase the dosage directly from 300 mg/d FI G U R E 1 Classification and year of introduction of antiseizure drugs
  • 4.     | 339 HAKAMI to 400 mg/d; toxicity frequently occurs at a variable time thereafter. Hepatic biotransformation of valproate may lead to hepatotoxicity. Hence, the liver function test is recommended in individual patients. Most ASDs are metabolized by hepatic enzymes. Carbamazepine, oxcarbazepine, eslicarbazepine, phenobarbital, phenytoin, and prim- idone are inducers of hepatic cytochrome P450 enzyme and may decrease the effects of other drugs administered concomitantly (eg, valproate). Valproate and clobazam are inhibitors of hepatic enzymes and most likely to elevate the plasma concentration of other drugs administered concomitantly (eg, carbamazepine, etho- suximide, phenytoin, phenobarbital, and lamotrigine). Drug-­ drug interactions with ASDs are complex since the drugs are often used in combination. These interactions may lead to either inadequate seizure control or drug toxicity. Of the newer drugs, levetiracetam, gabapentin, pregabalin, and vigabatrin are unique. These drugs are eliminated unchanged by the kidney and have no drug-­ drug inter- actions. Lamotrigine, perampanel, tiagabine, topiramate, and zoni- samide undergo hepatic drug metabolism and have potential drug interactions. Oxcarbazepine, felbamate, and topiramate selectively induce the hepatic metabolism of the oral contraceptive pill, failing birth control. 1.6 | Mechanisms of actions of ASDs A propensity for seizure generation occurs when there is an imbal- ance favoring excitation of neurons over inhibition. ASDs actions can generally be viewed in the context of inhibition of excitation or strengthening of inhibition, or both. Inhibition of excitation can be produced by effects on intrinsic excitability mechanisms in excita- tory neurons (eg, inhibition of sodium and calcium channel), or on excitatory synaptic transmission (eg, glutamate AMPA and NMDA receptors and synaptic vesicle protein 2A). Enhancement of inhi- bition is produced by the increased availability of γ-­ aminobutyric acid (GABA), increased activation of GABAA receptors, the media- tors of inhibition in cortical areas relevant to seizures, and modu- lation of voltage-­ gated potassium channel of the Kv7. For some drugs, the precise mechanism of action is not known (eg, valproate, zonisamide, and rufinamide), and some have multiple targets (eg, topiramate and felbamate).31 The ASD mechanisms of action are summarized in Table  2 and discussed in detail in the remainder sections. 1.7 | Voltage-­ gated sodium channels blockade At therapeutic concentrations, carbamazepine, cenobamate, lacosa- mide, lamotrigine, phenytoin, and zonisamide block voltage-­ gated sodium channel in neuronal membranes. These agents act mainly on action potential firing and do not directly alter excitatory or inhibi- tory synaptic responses. However, the effect on action potentials translates into reduced transmitter output at synapses. Topiramate may also act, in part, by this mechanism. Phenobarbital and valproate may act on the sodium channel at high doses. This action is rate-­ dependent (ie, the block increases with increased frequency of neu- ronal discharge).31 1.8 | GABA-­ related targets Benzodiazepines interact with specific receptors (GABAA receptors) and increase the frequency of GABA-­ mediated chloride ion chan- nel opening (facilitate inhibitory effects of GABA). Phenobarbital and other barbiturates increase the duration of GABA-­ mediated chloride ion channel opening. Vigabatrin, an analog of GABA, irre- versibly inactivates GABA transaminase, the enzyme responsible for the termination of action of GABA. Valproate also can inhibit GABA transaminase at high doses. Tiagabine selectively inhibits the GAT-­ 1 GABA transporter, causing prolongation of GABA-­ mediated inhibi- tory synaptic responses. Gabapentin and pregabalin are structural analogs of GABA but do not act through effects on any mechanism related to GABA-­ mediated neurotransmission. Instead, the drugs bind to the α2δ subunit of the voltage-­ gated calcium channel and decrease glutamate release at excitatory synapses. Other drugs that may facilitate the inhibitory actions of GABA include felbamate and topiramate.31 1.9 | Calcium channel blockade Ethosuximide inhibits low voltage-­ gated T type calcium channels in thalamocortical neurons. Valproate may exert similar action. Gabapentin and pregabalin bind to the α2δ subunit of voltage-­ gated calcium channel and may decrease glutamate release at excitatory synapses (the precise mechanism is not known).31 1.10 | Glutamate synapses and other mechanisms Levetiracetam binds to the synaptic vesicle protein 2A (SV2A) re- ceptors on glutamate-­ containing transmitter vesicles, where it is believed to inhibit excitatory neurotransmitters release (vesicular exocytosis) and thereby reduce synaptic excitability. Retigabine opens KCNQ2-­ 5 (Kv7.2-­ Kv7.5) voltage-­ gated potassium channel in presynaptic nerve terminals and thereby inhibits glutamate release. In addition to its action on calcium and sodium channel, valproate may enhance voltage-­ gated potassium channel permeability, causing hyperpolarization. Perampanel is a potent noncompetitive antago- nist of the AMPA receptors, a subtype of the ionotropic glutamate receptors that are the primary mediators of synaptic excitation in the central nervous system. Phenobarbital may block glutamate AMPA receptors and its action on voltage-­ activated calcium channel and GABA-­ chloride ion channel. Felbamate blocks glutamate NMDA receptors, with selectivity for those containing the GluN2B subu- nit. The drug also produces a barbiturate-­ like potentiation of GABAA receptor responses. The possible sites for the action of topiramate
  • 5. 340  |     HAKAMI include voltage-­ gated sodium channel, GABAA receptor subtypes, and glutamate AMPA receptors.31 1.11 | Principles of ASD selection for specific seizure type(s) Treatment should be considered in patients reporting more than one unprovoked seizure or after a single seizure if the risk of re- currence is high.1 The initial ASD should be individualized on the basis of seizure type and/or epilepsy syndrome and slowly titrated up to a target dosage. Other important selection criteria include pa- tient characteristics, drug efficacy, adverse-­ effect profile, potential drug-­ drug interactions, and cost.32 Combination therapy should be considered after the failure of two monotherapies.33 The therapeu- tic spectrum of ASDs can be categorized into (1) broad-­ spectrum drugs used for both focal and generalized seizures, including, in al- phabetical order, brivaracetam, clobazam, felbamate, lamotrigine, levetiracetam, perampanel, rufinamide, topiramate, valproate, and zonisamide; (2) narrow-­ spectrum drugs used primarily for focal and focal to bilateral (secondarily generalized) tonic-­ clonic seizures in- cluding carbamazepine, cenobamate, eslicarbazepine, gabapentin, lacosamide, oxcarbazepine, phenobarbital, phenytoin, pregabalin, primidone, stiripentol, tiagabine, and vigabatrin; and (3) a narrow-­ spectrum drug used primarily for generalized absence seizures which is ethosuximide. Table 3 presents the efficacy of ASDs against common seizure types and epilepsy syndrome. The data supplemented in the table are based primarily on the results of RCTs comparing the newer ver- sus older drugs, including the SANAD studies34-­ 37 and others,38-­ 62 and recently published meta-­ analyses.63-­ 69 Table 4 and the remain- der sections of this article present guidance for ASD selection for the specific seizure type according to the 2017 ILAE classification of seizure types and epilepsy syndromes.14-­ 17 The guidance is based mainly on the current state of the literature on the efficacy and tol- erability of ASDs and the recently published practical guidelines. The most popular treatment guidelines applied herein are the guide- lines published by AAN AES in 2004, 2015, and 2018,12,13,21,22,70 the ILAE in 2006 and 2013,71,72 the UK National Institute for Health and Care Excellence in 2012,73,74 and the Scottish Intercollegiate Guidelines Network in 2018.75,76 1.12 | Drugs effective for focal-­ onset seizures Focal-­ onset seizures account for 60% of all epilepsies. The RCTs and meta-­ analyses have demonstrated comparable efficacy of sev- eral ASDs in controlling new-­ onset focal seizures.22,34,37-­ 44,46,58,62-­ 64,72,77-­ 83 The drug of the first choice for “focal” and “focal to bilateral” (secondarily generalized) tonic-­ clonic seizures is lamotrig- ine.22,34,37 Phenytoin is the drug of choice for the urgent treatment of new-­ onset or recurrent focal epilepsy. The other most widely used first-­ line drugs for focal seizures include levetiracetam and zonisamide.22,34 Carbamazepine is considered a second-­ line treat- ment for new-­ onset focal epilepsy owing to its unfavorable efficacy-­ to-­ tolerability profile.22,34 Oxcarbazepine, topiramate, and valproate can be used, but they may not be as effective as lamotrigine and carbamazepine.22 Phenobarbital (and its derivative primidone) is often regarded as a second-­ line treatment in adults because of seda- tion and behavioral problems.22 However, phenobarbital is a primary drug for neonatal seizures. Given the unique mechanistic and pharmacokinetic profiles of the third-­ generation ASDs, the United States Food and Drug Administration (FDA) recently issued a strategy that allows extrap- olation of these drugs across populations as add-­ on or monother- apy.22 Accordingly, brivaracetam, eslicarbazepine, lacosamide, and perampanel received FDA approval as initial monotherapies for new-­ onset focal epilepsy.22 Individual monotherapy studies showed similar efficacy of second-­ generation (levetiracetam and zonisamide) and third-­ generation (eslicarbazepine and lacosamide) ASDs com- pared with controlled-­ release carbamazepine for the treatment of new-­ onset focal epilepsy.63 Vigabatrin use appears to be less effica- cious than carbamazepine use and may not be offered; furthermore, tolerability profile precludes vigabatrin use as first-­ line therapy.22 The drugs used as add-­ on (adjunctive) therapy in focal seizures in- clude gabapentin, pregabalin, felbamate, rufinamide, cenobamate, clobazam, retigabine, and tiagabine. However, the AAN/AES guide- lines recommend gabapentin and lamotrigine, as first-­ line monother- apy in patients aged ≥60 years with new-­ onset focal epilepsy.22 A recent systematic review and network meta-­ analysis showed that lacosamide, lamotrigine, and levetiracetam had the highest proba- bility of ranking best for achieving seizure freedom in the elderly with new-­ onset epilepsy.64 The FDA determined that the efficacy of ASDs for focal-­ onset seizures in adults can be extrapolated down- ward to children four years of age and above.22 For drug-­ resistant focal epilepsy in adults, eslicarbazepine can be used as monotherapy while the immediate-­ release pregabalin, perampanel, lacosamide, eslicarbazepine, extended-­ release topi- ramate, rufinamide, clobazam, felbamate, and vigabatrin should be considered as add-­ on therapy.21 However, vigabatrin, felbamate, and rufinamide are not first-­ line agents because of the retinopathy risk with vigabatrin, the modest benefit with rufinamide, and the hepato- toxicity and hematotoxicity risk with felbamate. In pediatric patients, levetiracetam, oxcarbazepine, and zonisamide should be considered as add-­ on therapy.21 1.13 | Drugs effective for generalized tonic-­ clonic seizures (GTCS) There has been a limited number of ASDs that can be used as first-­ line agents for GTCS. Valproate remains the first-­ line drug for many patients with generalized and unclassified epilepsies,22,35,36,72 while phenobarbital (or primidone) is a primary drug in infants (for neona- tal seizures) and an alternative in adults. However, valproate should not be prescribed for women of childbearing potential because of
  • 6.     | 341 HAKAMI its dose-­ dependent teratogenic profile; unless other ASDs cannot control the seizures, in which case the dose should be kept as low as possible.84 Indeed, some patients with genetic generalized epilepsy can control their seizures only with valproate. The other most widely used monotherapies for GTCS include lamotrigine, levetiracetam, brivaracetam, topiramate, clobazam, and zonisamide.22 However, there appears to be no evidence to support any second-­ generation or third-­ generation ASDs to be as efficacious as valproate mono- therapy for generalized and unclassified epilepsies.35,36 The com- bination of lamotrigine and valproate is believed to be particularly efficacious.85 Phenytoin and carbamazepine are effective but may worsen certain seizure types in generalized epilepsies, including absence epilepsy, juvenile myoclonic epilepsy, and Dravet's syn- drome.86 Vigabatrin, tiagabine, oxcarbazepine, and possibly gabap- entin are other drugs that may worsen these seizure types. Seizures of most patients with generalized epilepsy are easily controlled with appropriate medication. However, immediate-­ release and extended-­ release lamotrigine use should be considered add-­ on therapy to decrease seizure frequency in drug-­ resistant GTCS in adults.21 Levetiracetam should also be effective in both drug-­ resistant GTCS and drug-­ resistant juvenile myoclonic seizures. Other drugs used as add-­ on therapy in GTCS include perampanel and lacosamide. 1.14 | Drugs effective for focal seizures and certain generalized onset seizure types A variety of drugs are primarily used to treat focal seizures; these drugs have also been effective in certain generalized onset seizure types. These drugs are lamotrigine, levetiracetam, brivaracetam, perampanel, phenobarbital, primidone, and felbamate. 1.15 | Drugs effective for myoclonic seizures Valproate is widely used for myoclonic seizures, such as in juvenile myoclonic epilepsy.87 Lamotrigine is possibly effective but may worsen myoclonic seizures in some cases. Other drugs effective in treating this seizure type are levetiracetam, brivaracetam, zon- isamide, topiramate, and clonazepam. 1.16 | Drugs effective for generalized absence seizures Ethosuximide is the first-­ line drug. It is often used in uncomplicated absence seizures if patients can tolerate its gastrointestinal side ef- fects.22,45 Despite the long half-­ life (~ 40 hours), ethosuximide is gen- erally administered in two or even three divided doses to minimize the adverse gastrointestinal effects. Valproate is preferred in pa- tients with concomitant GTCS or myoclonic seizures (myoclonic ab- sence seizure). Clonazepam is effective as an alternative drug but has the disadvantages of causing sedation and tolerance. Lamotrigine, levetiracetam, and zonisamide are also used in the absence seizures but not as effective as ethosuximide or valproate. Unless there are compelling reasons based on adverse events profile, ethosuximide or valproate use should be considered before lamotrigine in treat- ing absence seizures.45 Lamotrigine should be considered in women of childbearing age because of its better tolerability and fewer fetal risks compared with valproate. 1.17 | Drugs effective for other epilepsy syndromes In combination with lamotrigine and a benzodiazepine (such as clobazam), valproate is the most widely used treatment for atonic seizures (eg, in the Lennox-­ Gastaut syndrome). Other drugs used for the atonic seizures in Lennox-­ Gastaut syndrome include topira- mate, felbamate, and rufinamide.85 Because felbamate can cause aplastic anemia and severe hepatitis, it is used as add-­ on therapy in patients who respond poorly to other agents. For infantile spasms (West's syndrome), valproate, topiramate, zonisamide, or a benzodiazepine (such as clonazepam or nitrazepam) is ef- fective.85 Vigabatrin and everolimus are used if associated with tuberous sclerosis complex (a rare genetic disease that causes non-­ cancerous (benign) tumors to grow in the brain and other organs). However, infantile spasms are primarily treated with intramuscu- lar adrenocorticotropic hormone (ACTH) or oral corticosteroids such as prednisone or hydrocortisone (unknown mechanism). For Dravet's syndrome (the severe myoclonic epilepsy of infancy), val- proate, topiramate, and clobazam can be used, although none of these is very effective.88 Stiripentol, a modulator of GABAA recep- tors, is often used in conjunction with clobazam or valproate.88 Cannabidiol was approved in 2018 to treat Dravet's syndromes66 in addition to Lennox-­ Gastaut syndrome68 and infantile spasms as- sociated with tuberous sclerosis complex. Recent meta-­ analyses showed that adjunctive cannabidiol was associated with a greater reduction in convulsive seizure frequency than placebo in patients with Lennox-­ Gastaut syndrome68 and children with Dravet syn- drome.66 Fenfluramine is another new drug approved in 2020 for Dravet's syndrome. The drug is available only through a restricted drug distribution program, under a risk evaluation and mitigation strategy (REMS) because of the risk of valvular heart disease and pulmonary arterial hypertension. Recent studies, however, showed that fenfluramine provided a significantly greater reduction in con- vulsive seizure frequency compared with placebo and was gen- erally well tolerated, with no observed valvular heart disease or pulmonary arterial hypertension.65,89 1.18 | Drugs effective for status epilepticus Status epilepticus (SE) is a life-­ threatening emergency that requires immediate treatment.90 In clinical practice, it is now generally ac- cepted that a seizure lasting 5 minutes for GTCS and 10 minutes for focal seizures with or without impairment of consciousness should
  • 7. 342  |     HAKAMI be treated as status epilepticus. Figure 2 presents the SE treatment algorithm. According to the evidence-­ based guideline for the treat- ment of convulsive SE in children and adults by the AES in 2016,91 the first-­ line treatment for SE is a benzodiazepine (either intravenous lorazepam or intravenous diazepam or intramuscular midazolam). If the preferred options are not available, rectal diazepam, intranasal midazolam, buccal midazolam, or intravenous phenobarbital is ac- ceptable alternatives. If the seizure continues, then second-­ line treatment is adminis- tered.69 Intravenous fosphenytoin is preferred over phenytoin as the latter may cause cardiotoxicity. With phenytoin intravenous admin- istration, there is also a risk of the potentially serious “purple glove syndrome,” in which a purplish-­ black discoloration accompanied by edema and pain occurs distal to the site of injection. Fosphenytoin has a lower incidence of purple glove syndrome. The second-­ line treatment also includes intravenous valproate or intravenous leve- tiracetam. Intravenous phenobarbital can be alternative (if not given already) but has a long half-­ life causing persistent adverse effects, including severe sedation, respiratory depression, and hypotension. A recent network meta-­ analysis compared efficacy and tolerability of intravenous valproate, phenytoin, diazepam, phenobarbital, la- cosamide, and levetiracetam in adults with benzodiazepine-­ resistant convulsive SE and suggested that phenobarbital had the greatest probabilities of being best in the achievement of SE control and seizure freedom, whereas valproate and lacosamide ranked best for the safety outcomes (respiratory depression and hypotension).69 If the second therapy fails to stop the seizures, another second-­ line agent is often tried. Treatment-­ resistant SE occurs when sei- zures continue or recur at least 30  minutes after treatment with first-­and second-­ line agents and should be treated with anesthetic doses of pentobarbital, propofol, midazolam, or thiopental. Dosing and frequency are available in the proposed algorithm for convulsive SE.91 Acute repetitive seizures (seizure clusters), in which there is complete recovery between seizures, are treated with benzodiaze- pines. Diazepam rectal gel is the only approved treatment for the out-­ of-­ hospital treatment of acute repetitive seizures.92 1.19 | Adverse effects of ASDs The adverse effects of selected ASDs are listed in Table  5. The first-­ generation ASDs have acute dose-­ related effects, primar- ily neurological effects such as sedation, dizziness, unsteadiness, blurred vision, diplopia, and tremor, in addition to neurocognitive and psychiatric symptoms.25,26,32 These effects are found across the different ASDs26,62 and often mild and reversible. However, some drugs are better tolerated than others; for example, lamotrigine and levetiracetam are better tolerated than carbamazepine in elderly FI G U R E 2 Status Epilepticus treatment algorithm* *Adopted from Glauser T, et al Epilepsy Curr. 2016; 16:48-­ 6. Refer to publication for complete recommendations. ABCD, airway, breathing, circulation, disability (Neurologic); B, buccal; ECG, electrocardiogram; ED, emergency department; EEG, electroencephalogram; IM, intramuscular; IN, intranasal; IV, intravenous; PE, phenytoin sodium equivalents; SE, status epilepticus.
  • 8.     | 343 HAKAMI patients.93 Psychiatric adverse effects include depression, anxiety, irritability, impaired concentration, mood changes, hyperactivity, and, in rare cases, psychosis. Although the newer ASDs are touted as better tolerated than older drugs,12 psychiatric adverse effects are common with levetiracetam, topiramate, zonisamide, vigabatrin, and perampanel. Lamotrigine, carbamazepine, valproate, gabap- entin, and pregabalin, in contrast, have mood-­ stabilizing effects in some patients and less frequently cause behavioral or psychiatric effects.94 Most women with epilepsy who become pregnant require continued ASD therapy for seizure control. If possible, valproate, carbamazepine, phenytoin, phenobarbital, and topiramate should be avoided in women of childbearing potentials. Recent analysis from several international pregnancy registers suggests that in utero exposure to valproate during the first trimester is associ- ated with a threefold increased risk of congenital malformations, commonly neural tube defects (spina bifida) and cardiovascular, orofacial, and digital abnormalities.95 The use of valproate during the first trimester is also associated with cognitive impairments. Carbamazepine may cause neural tube defects and craniofacial anomalies. Fetal hydantoin syndrome is related to the use of phe- nytoin. Treatment with topiramate during the first trimester of pregnancy is associated with a 10-­ fold increase in oral clefts risk. Phenobarbital can cause congenital malformations, most often cardiac defects. No ASD is known to be entirely safe for the de- veloping fetus. However, lamotrigine and levetiracetam have the TA B LE 1 Pharmacokinetic profiles of antiseizure drugs Antiseizure drug Bioavailability % Peak concentration (hr) Plasma protein binding (%) Elimination half-­ life (hr) Route of elimination Therapeutic serum concentration (mcg/mL) Brivaracetam ~ 95 1 ≤ 20 7-­ 10 ++ 0.2-­ 2 Carbamazepine 75-­ 85 4-­ 5 70-­ 80 10-­ 17 ++++ 4-­ 11 Cannabidiol 10-­ 20 2.5-­ 5 94 56-­ 61 ++++ NE Cenobamate 88 1-­ 4 60 50-­ 60 +++ NE Clobazam 90-­ 100 1-­ 3 80-­ 90 36-­ 42 ++++ 0.03-­ 3 Clonazepam 80 1-­ 4 80-­ 90 24-­ 48 +++ 10-­ 70a   Eslicarbazepine 90 1-­ 4 40 13-­ 20 ++++ 5-­ 35 Ethosuximide 95-­ 100 3-­ 7 0 30-­ 60 ++ 40-­ 100 Felbamate 90 3-­ 5 22-­ 36 16-­ 22 ++ 30-­ 60 Gabapentin 50 2-­ 3 0 5-­ 9 -­ 3-­ 21 Lacosamide 100 1-­ 2 30 12-­ 14 + 3-­ 10 Lamotrigine ~ 90 1-­ 3 55 8-­ 35 +++ 3-­ 13 Levetiracetam ~ 95 1-­ 2 10 6-­ 8 -­ 5-­ 41 Oxcarbazepine 100 4-­ 5 75 10-­ 17 ++++ 3-­ 36 Perampanel 100 0.5-­ 3 95-­ 96 70-­ 110 +++ 0.1-­ 1 Phenobarbital 90 0.5-­ 4 55 90 ++ 12-­ 30 Phenytoin 85-­ 90 5-­ 7 90 24 +++b   10-­ 20 Pregabalin ~90 1-­ 2 0 4.5-­ 7 -­ 2-­ 6 Primidone 90 2-­ 6 10 8-­ 15 ++ 8-­ 12 Rufinamide 90 4-­ 6 35 6-­ 10 ++ 4.5-­ 31 Stiripentol Variable 2-­ 3 99 4.5-­ 13 + 4-­ 22 Tiagabine ~90 0.5-­ 2 96 2-­ 9 +++ 0.02-­ 0.2 Topiramate ~80 2-­ 4 15 20-­ 30 + 2-­ 10 Valproate 90 2-­ 4 90 15 ++++ 50-­ 100 Vigabatrin 100 1 0 5-­ 8 -­ 20−160a   Zonisamide 90 2-­ 6 40-­ 60 50-­ 68 ++ 10-­ 38 Note: NE, not established ++++Extensive hepatic metabolism and active metabolite(s) +++Extensive hepatic metabolism but no active metabolite(s) ++Hepatic metabolism (with or without active metabolites) and renal excretion. +Variable (or moderate) hepatic metabolism (with or without active metabolites) -­ Renal excretion (unchanged). No hepatic metabolism a ng/mL b Saturable
  • 9. 344  |     HAKAMI lowest risks of major congenital malformations and may be safer, particularly for cognition compared with valproate.85 Valproate causes hepatotoxicity in children less than two years of age. Carbamazepine and lamotrigine may cause life-­ threatening Steven-­ Johnson syndrome and toxic epidermal necrolysis, which is strongly associated with the HLA-­ B*1502 allele. Asians, who have a 10-­ fold increased risk of the drug-­ induced Stevens-­ Johnson syndrome compared with other ethnic groups, should be tested before starting the drug. The use of zonisamide is also associated with severe skin reactions. Aplastic anemia and acute hepatic fail- ure have limited the use of felbamate to severe and drug-­ resistant epilepsy. Overdose toxicity with benzodiazepines and barbiturates may cause respiratory depression. Management is primarily sup- portive (airway management, mechanical ventilation) and fluma- zenil in benzodiazepine overdose. Withdrawal from ASDs should be accomplished gradually (over a 1-­to 3-­ month period or longer) to avoid the occurrence of severe seizures or status epilepticus. Physical dependence occurs with barbiturates and benzodiaze- pines, and there is a well-­ recognized risk of rebound seizures with abrupt withdrawal. However, withdrawal is less likely to be a prob- lem with ethosuximide. Withdrawal is believed to be successful in patients with generalized epilepsies who exhibit a single seizure type, whereas the longer duration of epilepsy, an abnormal neu- rologic examination, an abnormal EEG, and certain epilepsy syn- dromes, including juvenile myoclonic epilepsy, are associated with increased risk of recurrence.85 Clinical studies suggest a possible association of lamotrigine, le- vetiracetam, and topiramate with suicidality. In 2008, the FDA issued an alert that ASDs, as a class, may be associated with an increased risk of suicidality based on an analysis of data from placebo-­ controlled add-­ on clinical trials of ASDs in patients with drug-­ resistant epilepsy, although this is still highly controversial.96,97 Long-­ term treatment with ASDs is associated with a twofold to threefold increased risk of osteoporosis and bone fractures.98 In addition, increased body weight and fat are common in patients using valproate, carbamaz- epine, gabapentin, pregabalin, vigabatrin, and perampanel and can lead to serious health consequences associated with obesity and in- creased cardiovascular disease risk.99 In March 2021, the FDA issued an alert that lamotrigine may be associated with an increased risk of cardiac arrhythmias in people with underlying cardiac disease.100 However, this risk is not apparent in healthy individuals.101 1.20 | The new frontiers in epilepsy pharmacology Since 1989, nearly 20  second-­ generation and third-­ generation an- tiseizure drugs (ASDs) with different mechanisms of action have reached the market, resulting in a greatly increased range of treat- ment options for patients and prescribers.102,103 Some second-­ generation ASDs have shown advantages in drug tolerability, drug-­ drug interactions, and teratogenicity and thus offer valuable individualized options in treating epilepsy. Disappointingly, however, Target and mechanisma   Antiseizure drug Inhibition of voltage-­ gated sodium channels Phenytoin, fosphenytoin, carbamazepine, cenobamate, lamotrigine, oxcarbazepine, eslicarbazepine, lacosamide, and possibly topiramate, zonisamide, rufinamide, and phenobarbital Inhibition of α2δ subunit of voltage-­ gated calcium channels Gabapentin, pregabalin Inhibition of T type voltage-­ gated calcium channels Ethosuximide Activation of GABAA receptor Phenobarbital, benzodiazepines, and possibly topiramate, felbamate, retigabine, and stiripentol. Inhibition of GABA transporter (selective) Tiagabine Inhibition of GABA transaminase enzyme Vigabatrin Modulation of synaptic vesicle protein 2A Levetiracetam, brivaracetam Various actions on multiple targets Valproate, felbamate, topiramate, zonisamide, and cannabidiol Opening KCNQ2-­ 5 (Kv7.2-­ Kv7.5) voltage-­ gated potassium channels Retigabine (ezogabine)b   Inhibition of NMDA-­ type glutamate receptors Felbamate, topiramate and phenobarbital Inhibition of AMPA-­ type glutamate receptors Perampanel a Fenfluramine's mechanism of action for the treatment of seizures associated with Dravet syndrome is unknown. b Production of the drug retigabine has been discontinued by the manufacturer, and it is no longer available. TA B LE 2 Mechanistic categorization of current antiseizure drugs based on foremost targets at therapeutic concentrations
  • 10.     | 345 HAKAMI none of these medications appear to be more efficacious than first-­ generation ASDs, and none have substantially reduced the propor- tion of patients with drug-­ resistant epilepsy, highlighting the need for novel strategies in epilepsy drug development. Given the favorable pharmacokinetic characteristics and adverse-­ effect profiles for the third-­ generation medications, additional evidence to further define their efficacy is crucial to the future treatment of epilepsy. The high incidence of drug-­ resistance and unwanted adverse effects caused by taking long-­ term ASDs raise substantial concern. In particular, there is an urgent need for developing novel drugs that act beyond the membrane ion channels and neural transmissions. Deciphering the genetics that underpins the mechanisms that generate seizures is one of the promising areas in the field.104 Many epilepsy genes have been identified, including genes that increase the risk of dif- ferent types of epilepsy, such as generalized and focal epilepsy and developmental and epileptic encephalopathies. These genes have TA B LE 3 Efficacy of antiseizure drugs against common seizure types and epilepsy syndromes Antiseizure drug/ seizure type Focal seizures GTCS Absence Myoclonic Lennox–­Gastaut syndrome Infantile spasm Dravet's syndrome Brivaracetam + + + Cannabidiol + +a   + Carbamazepine + + –­ –­ –­ Cenobamate + Clobazam + + + +e   Clonazepam + + + Eslicarbazepine + –­ –­ Ethosuximide + Felbamate +b   + Fenfluramine + Gabapentin + ?+ –­ –­ –­ Lacosamide + ?+ Lamotrigine + + + ?+c   + Levetiracetam + + ?+ + Oxcarbazepine + + –­ –­ –­ Perampanel + + Phenobarbital + + –­ ?+ Phenytoin + + –­ –­ –­ Pregabalin + –­ Primidone + + –­ Retigabineg   + Rufinamide + + Stiripentol +f   Tiagabine + –­ –­ Topiramate + + + + + +e   Valproate + + + +d   + + +e   Vigabatrin + ?+ –­ –­ +a   Zonisamide + + ?+ + + Note: GTCS; generalized tonic-­ clonic seizure Note that although there is evidence to support the use of these drugs for these seizure types, the drugs may not be indicated for this use by the US Food and Drug Administration. +Effective;?+ possibly effective; –­worsen seizure. a Especially when associated with tuberous sclerosis complex. b Can cause aplastic anemia and severe hepatitis, used only for patients who respond poorly to other agents. c Possibly effective but may worsen myoclonic seizures in some cases. d Preferred in patients with concomitant GTCS or myoclonic seizures (myoclonic absence seizure). e None of these is very effective in Dravet's syndrome. f In combination with clobazam and valproate. g Has been discontinued by the manufacturer, and it is no longer available.
  • 11. 346  |     HAKAMI a role in ion channel and synaptic dysfunction in addition to tran- scriptional regulation. Most mutations occur in the protein-­ coding exons, the sodium and potassium channel, and glutamate receptors.32 Ganaxolone, sirolimus, everolimus, cannabidiol, fenfluramine, stirip- entol, and memantine are new selective disease-­ modifying therapies recently approved for severe forms of genetic epilepsies.32 Data to support gene-­ therapy treatments need to be established in future re- search, and there are often other disorders that these therapies can improve, including acquired epilepsies.104 Furthermore, with a deeper understanding of the cellular and molecular mechanisms of epilep- togenesis, non-­ classical novel antiseizure agents, such as neuroster- oid and allopregnanolone, with their effect on both GABAergic and TA B LE 4 Recommendations for add-­ on and monotherapy in adults and pediatric patients 4 years of age with new-­ onset epilepsy based on an assessment of current literature and published guidelines Seizure type First-­ line (Monotherapy or add-­ on) Second-­ line (Monotherapy or add-­ on) Third-­ line (Add-­ on) Focal-­ onset seizures, including focal to bilateral tonic-­ clonic seizure Lamotriginea   Carbamazepine Levetiracetam Zonisamide Phenytoin Valproate Topiramateb   Oxcarbazepineb   Gabapentinb   Phenobarbitalc   Brivaracetamd   Eslicarbazepined   Lacosamided   Perampaneld   Cenobamatee   Clobazam Retigabine Felbamate Rufinamide Pregabalin Tiagabine Vigabatrin Generalized tonic-­ clonic seizures (GTCS) Valproatef   Carbamazepine Phenytoin Lamotrigine Topiramate Levetiracetam Brivaracetam Perampanel Zonisamide Clobazam Phenobarbital Myoclonic seizure Valproate Lamotrigineg   Topiramate Levetiracetam Brivaracetam Clonazepam Zonisamide Absence seizures Ethosuximide Valproate Lamotrigine Clonazepam Levetiracetam Unclassified seizuresh   Valproate Lamotrigine Levetiracetam Topiramate Zonisamide b Evidence is insufficient to consider gabapentin, oxcarbazepine, or topiramate instead of lamotrigine in patients with new-­ onset focal epilepsy. Gabapentin may be considered first-­ line monotherapy in patients aged ≥60 years. c Often regarded as second-­ line treatment in adults because of sedation and behavioral problems. d Received FDA approval for extrapolation of efficacy as monotherapy across individuals with focal seizures (Kanner AM, et al Neurology 2018; 91:82-­ 90). e Evidence is insufficient to consider the use of clobazam, felbamate, tiagabine, vigabatrin, or third-­ generation antiseizure drugs as monotherapies in treating new-­ onset focal epilepsy. f Valproate should be avoided, if possible, in women of childbearing potential. g May worsen myoclonic seizures in some cases. h Evidence is insufficient to support efficacy of newer antiseizure drugs in unclassified generalized tonic-­ clonic seizures. a Lamotrigine was superior to levetiracetam and zonisamide for time to 12-­ month remission and should remain a first-­ line treatment for new-­ onset focal epilepsy.
  • 12.     | 347 HAKAMI TA B LE 5 Common and serious adverse effects of selected antiseizure drugs Antiseizure drug Systemic adverse effects Neurologic adverse effects Rare idiosyncratic reactions Brivaracetam Nausea, vomiting, constipation, fatigue Headache, somnolence, dizziness, abnormal coordination, nystagmus, mood changes Carbamazepine Nausea, vomiting, diarrhea, a plastic anemia, leukopenia, hyponatremia (common reason for discontinuation), hepatotoxicity, rash, pruritus Ataxia, dizziness, blurred vision, diplopia, headache Erythematous maculopapular rash (Steven-­ Johnson syndrome and toxic epidermal necrolysis), teratogenicity Cenobamate Nausea, vomiting, fatigue, hyperkalemia, QT shortening Somnolence, dizziness, headache, balance disorder, diplopia Drug reaction with eosinophilia and systemic symptoms (DRESS)/ multiorgan hypersensitivity (at high doses) Eslicarbazepine Nausea, vomiting, diarrhea, hyponatremia, rash Dizziness, drowsiness, headache, somnolence, diplopia, ataxia, blurred vision, tremor Ethosuximide Nausea, vomiting Sleep disturbance, drowsiness, hyperactivity Felbamate Nausea, vomiting, anorexia, weight loss Insomnia, dizziness, headache, ataxia Aplastic anemia, severe hepatitis/ hepatic failure Gabapentin Infrequent Somnolence, dizziness, ataxia, headache, tremor, and fatigue Lacosamide Nausea, vomiting, increased cardiac conduction (PR interval) Dizziness, ataxia, diplopia, headache Lamotrigine Nausea, rash, cardiac arrhythmias Dizziness, tremor, diplopia Steven-­ Johnson syndrome Levetiracetam Fatigue, infection, anemia, leukopenia Somnolence, dizziness, agitation, anxiety, irritability, depression, psychosis Oxcarbazepine Nausea, rash, hyponatremia (more common) Somnolence, headache, dizziness, vertigo, ataxia, diplopia Perampanel Weight gain, fatigue, nausea Dizziness, somnolence, irritability, gait disturbance, falls (with high dose), aggression, mood alteration Phenobarbital Nausea, rash Somnolence, ataxia, dizziness, confusion, cognitive dysfunction, tolerance, dependence Phenytoin Gingival hyperplasia, hirsutism, megaloblastic anemia, peripheral neuropathy, osteoporosis, rash Nystagmus (early sign of phenytoin administration), diplopia, ataxia, somnolence Pregabalin Weight gain, peripheral edema, dry mouth Somnolence, dizziness, ataxia, headache, and tremor Rufinamide Nausea, vomiting, leukopenia, cardiac conduction (QT interval shortening) Somnolence, fatigue, dizziness, ataxia, headache, diplopia Tiagabine Abdominal pain, nausea, lack of energy Dizziness, difficulty concentrating, somnolence, nervousness, tremor, language problems Topiramate Anorexia, weight loss, paresthesia, fatigue Nervousness, psychomotor slowing, language problems, depression, anxiety, mood problems, tremor Acute glaucoma (may require prompt drug withdrawal). Valproate Gastrointestinal irritation, weight gain, hair loss, easy bruising Ataxia, somnolence, tremor Hepatotoxicity, teratogenicity, and thrombocytopenia Vigabatrin Fatigue Somnolence, headache, dizziness, agitation, confusion, psychosis. Irreversible bilateral concentric visual field defect Zonisamide Weight loss, nausea, anorexia Somnolence, dizziness, confusion, headache, psychosis Potentially serious skin rashes
  • 13. 348  |     HAKAMI glutaminergic systems, may shed light on finding a new approach to treat epilepsy, which is not only controlling seizures but also retarding epileptogenesis.105 Novel pharmacological approaches built on gene therapies and epileptogenesis may overcome many of the adverse ef- fects of currently available treatment options. 2 | CONCLUSIONS Antiseizure drugs (ASDs) have many different pharmacologic profiles that are relevant when selecting and prescribing these medications in patients with epilepsy. These include pharmacokinetic properties, propensity for drug-­ drug interactions, and adverse-­ effect profile and toxicities. This article reviewed the current state of the literature about the clinical pharmacology of ASDs, based primarily on the most recent evidence for the efficacy and tolerability of the currently avail- able drugs and the recent US and UK treatment guidelines. It is antici- pated that Table 4 in this review (recommendations for ASD selection) will require updating as future studies yield more detailed results. It is hoped that this review remains a succinct and practical guide to as- sist non-­ neurologists, particularly the primary healthcare practition- ers, in patient care decisions. The medical students may also wish to read this review to improve their understanding of epilepsy and its pharmacological treatment. Finally, the content of this review is not intended to include all legitimate criteria for choosing to use or ex- clude a specific drug, nor recommended as a substitute for current sci- entific and clinical information. Physicians are encouraged to carefully review the literature in addition to the full guidelines to understand all recommendations associated with patient care and be confident that the information contained in this work is accurate, particularly for new or infrequently used drugs. Physicians are also encouraged to obtain information about drug dosing and frequency from the product infor- mation sheet included in each drug package. Complete information on US Food and Drug Administration (FDA) labeling for each drug can be accessed using the FDA searchable database (FDALabel). CONFLICT OF INTEREST The author declares no conflict of interest. AUTHOR CONTRIBUTIONS The author has made substantial contributions to the conception and design of the work, and acquisition, analysis, and interpretation of data for the work; drafting the work and revising it critically for important intellectual content; final approval of the version to be published; and agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. DATA AVAILABILITY STATEMENT Data sharing not applicable to this article. ORCID Tahir Hakami  https://orcid.org/0000-0002-1933-6044 REFERENCES 1. Fisher RS, Acevedo C, Arzimanoglou A, Bogacz A, Cross JH, Elger CE, et al. ILAE official report: a practical clinical definition of epi- lepsy. Epilepsia. 2014;55:475-­ 82. 2. Hauser WA, Beghi E. First seizure definitions and worldwide inci- dence and mortality. Epilepsia. 2008;49(Suppl 1):8-­ 12. 3. Pitkanen A. Therapeutic approaches to epileptogenesis–­ hope on the horizon. Epilepsia. 2010;51(Suppl 3):2-­ 17. 4. Kwan P, Brodie MJ. Early identification of refractory epilepsy. N Engl J Med. 2000;342:314-­ 9. 5. Marson A, Jacoby A, Johnson A, Kim L, Gamble C, Chadwick D, et al. Immediate versus deferred antiepileptic drug treatment for early epilepsy and single seizures: a randomised controlled trial. Lancet. 2005;365:2007-­ 13. 6. Kwan P, Brodie MJ. Drug treatment of epilepsy: when does it fail and how to optimize its use? CNS Spectr. 2004;9:110-­ 9. 7. Kwan P, Arzimanoglou A, Berg AT, Brodie MJ, Allen Hauser W, Mathern G, et al. Definition of drug resistant epilepsy: consen- sus proposal by the ad hoc Task Force of the ILAE Commission on Therapeutic Strategies. Epilepsia. 2010;51:1069-­ 77. 8. Mattson RH, Cramer JA, Collins JF, Smith DB, Delgado-­ Escueta AV, Browne TR, et al. Comparison of carbamazepine, phenobar- bital, phenytoin, and primidone in partial and secondarily general- ized tonic-­ clonic seizures. N Engl J Med. 1985;313:145-­ 51. 9. French JA, White HS, Klitgaard H, Holmes GL, Privitera MD, Cole AJ, et al. Development of new treatment approaches for epilepsy: unmet needs and opportunities. Epilepsia. 2013;54(Suppl 4):3-­ 12. 10. Wilcox KS, Dixon-­ Salazar T, Sills GJ, Ben-­ Menachem E, Steve White H, Porter RJ, et al. Issues related to development of new antiseizure treatments. Epilepsia. 2013;54(Suppl 4):24-­ 34. 11. Stephen LJ, Brodie MJ. Pharmacotherapy of epilepsy: newly ap- proved and developmental agents. CNS Drugs. 2011;25:89-­ 107. 12. French JA, Kanner AM, Bautista J, Abou-­ Khalil B, Browne T, Harden CL, et al. Efficacy and tolerability of the new antiepileptic drugs I: treatment of new onset epilepsy: report of the Therapeutics and Technology Assessment Subcommittee and Quality Standards Subcommittee of the American Academy of Neurology and the American Epilepsy Society. Neurology. 2004;62:1252-­ 60. 13. French JA, Kanner AM, Bautista J, Abou-­ Khalil B, Browne T, Harden CL, et al. Efficacy and tolerability of the new antiepileptic drugs II: treatment of refractory epilepsy: report of the Therapeutics and Technology Assessment Subcommittee and Quality Standards Subcommittee of the American Academy of Neurology and the American Epilepsy Society. Neurology. 2004;62:1261-­ 73. 14. Fisher RS. An overview of the 2017 ILAE operational classification of seizure types. Epilepsy Behav. 2017;70:271-­ 3. 15. Fisher RS, Cross JH, D'Souza C, et al. Instruction manual for the ILAE 2017 operational classification of seizure types. Epilepsia. 2017;58:531-­ 42. 16. Fisher RS, Cross JH, French JA, Higurashi N, Hirsch E, Jansen FE, et al. Operational classification of seizure types by the International League Against Epilepsy: Position Paper of the ILAE Commission for Classification and Terminology. Epilepsia. 2017;58:522-­ 30. 17. Scheffer IE, Berkovic S, Capovilla G, et al. ILAE classification of the epilepsies: position paper of the ILAE commission for classification and terminology. Epilepsia. 2017;58:512-­ 21. 18. Proposal for revised clinical and electroencephalographic classi- fication of epileptic seizures. From the commission on classifica- tion and terminology of the international league against epilepsy. Epilepsia. 1981;22:489-­ 501. 19. Proposal for revised classification of epilepsies and epileptic syn- dromes. Commission on classification and terminology of the in- ternational league against epilepsy. Epilepsia. 1989;30:389-­ 99. 20. Berg AT, Berkovic SF, Brodie MJ, Buchhalter J, Cross JH, van Emde Boas W, et al. Revised terminology and concepts for organization
  • 14.     | 349 HAKAMI of seizures and epilepsies: report of the ILAE Commission on Classification and Terminology, 2005–­ 2009. Epilepsia. 2010;51:676-­ 85. 21. Kanner AM, Ashman E, Gloss D, Harden C, Bourgeois B, Bautista JF, et al. Practice guideline update summary: efficacy and tolerabil- ity of the new antiepileptic drugs II: treatment-­ resistant epilepsy: report of the guideline development, dissemination, and imple- mentation subcommittee of the American Academy of Neurology and the American Epilepsy Society. Neurology. 2018;91:82-­ 90. 22. Kanner AM, Ashman E, Gloss D, Harden C, Bourgeois B, Bautista JF, et al. Practice guideline update summary: efficacy and toler- ability of the new antiepileptic drugs I: treatment of new-­ onset epilepsy: report of the guideline development, dissemination, and implementation subcommittee of the American Academy of Neurology and the American Epilepsy Society. Neurology. 2018;91:74-­ 81. 23. Fiest KM, Sauro KM, Wiebe S, Patten SB, Kwon C-­ S, Dykeman J, et al. Prevalence and incidence of epilepsy: a systematic re- view and meta-­ analysis of international studies. Neurology. 2017;88:296-­ 303. 24. Epilepsy Foundation. Epilepsy Pipeline Tracker, https://www.epile​ psy.com/pipel​ ine-­ listi​ ng-­ page (accessed April 25, 2021) 25. Baker GA, Jacoby A, Buck D, Stalgis C, Monnet D, et al. Quality of life of people with epilepsy: a European study. Epilepsia. 1997;38:353-­ 62. 26. Loscher W, Schmidt D. Modern antiepileptic drug development has failed to deliver: ways out of the current dilemma. Epilepsia. 2011;52:657-­ 78. 27. Marson AG, Kadir ZA, Hutton JL, Chadwick DW, et al. The new antiepileptic drugs: a systematic review of their efficacy and toler- ability. Epilepsia. 1997;38:859-­ 80. 28. Vajda FJ. Pharmacotherapy of epilepsy: new armamentar- ium, new issues. J Clin Neurosci Off J Neurosurg Soci Australa. 2007;14:813-­ 23. 29. International League Against Epilepsy. Epilepsy Syndromes, https://www.epile​ psydi​ agnos​ is.org/syndr​ ome/epile​ psy-­ syndr​ ome-­ group​ overv​ iew.html (accessed April 25, 2021) 30. Zaccara G, Perucca E. Interactions between antiepileptic drugs, and between antiepileptic drugs and other drugs. Epileptic Disord. 2014;16:409-­ 31. 31. Sills GJ, Rogawski MA. Mechanisms of action of currently used an- tiseizure drugs. Neuropharmacology. 2020;168:107966. 32. Devinsky O, Vezzani A, O'Brien TJ, Jette N, Scheffer IE, de Curtis M, et al. Epilepsy. Nat Rev Dis Primers. 2018;4:18024. 33. Karceski S, Morrell M, Carpenter D. The expert consensus guide- line series treatment of epilepsy. Epilepsy Behav. 2001;2:A1-­ A50. 34. Marson AG, Al-­ Kharusi AM, Alwaidh M, Appleton R, Baker GA, Chadwick DW, et al. The SANAD study of effectiveness of carba- mazepine, gabapentin, lamotrigine, oxcarbazepine, or topiramate for treatment of partial epilepsy: an unblinded randomised con- trolled trial. Lancet. 2007;369:1000-­ 15. 35. Marson AG, Al-­ Kharusi AM, Alwaidh M, Appleton R, Baker GA, Chadwick DW, et al. The SANAD study of effectiveness of val- proate, lamotrigine, or topiramate for generalised and unclassifi- able epilepsy: an unblinded randomised controlled trial. Lancet. 2007;369:1016-­ 26. 36. Marson A, Burnside G, Appleton R, Smith D, Leach JP, Sills G, et al. The SANAD II study of the effectiveness and cost-­ effectiveness of valproate versus levetiracetam for newly diagnosed gener- alised and unclassifiable epilepsy: an open-­ label, non-­ inferiority, multicentre, phase 4, randomised controlled trial. Lancet. 2021;397:1375-­ 86. 37. Marson A, Burnside G, Appleton R, Smith D, Leach JP, Sills G, et al. The SANAD II study of the effectiveness and cost-­ effectiveness of levetiracetam, zonisamide, or lamotrigine for newly diagnosed focal epilepsy: an open-­ label, non-­ inferiority, multicentre, phase 4, randomised controlled trial. Lancet. 2021;397:1363-­ 74. 38. Privitera MD, Brodie MJ, Mattson RH, Chadwick DW, Neto W, Wang S, et al. Topiramate, carbamazepine and valproate mono- therapy: double-­ blind comparison in newly diagnosed epilepsy. Acta Neurol Scand. 2003;107:165-­ 75. 39. Rowan AJ, Ramsay RE, Collins JF, Pryor F, Boardman KD, Uthman BM, et al. New onset geriatric epilepsy: a randomized study of gabapentin, lamotrigine, and carbamazepine. Neurology. 2005;64:1868-­ 73. 40. Brodie MJ, Perucca E, Ryvlin P, Ben-­ Menachem E, Meencke H-­ J, et al. Comparison of levetiracetam and controlled-­ release carba- mazepine in newly diagnosed epilepsy. Neurology. 2007;68:402-­ 8. 41. Brodie MJ, Overstall PW, Multicentre GL. Multicentre, double-­ blind, randomised comparison between lamotrigine and carbamazepine in elderly patients with newly diagnosed epi- lepsy. The UK Lamotrigine Elderly Study Group. Epilepsy Res. 1999;37:81-­ 7. 42. Baulac M, Brodie MJ, Patten A, Segieth J, Giorgi L, et al. Efficacy and tolerability of zonisamide versus controlled-­ release carbamaz- epine for newly diagnosed partial epilepsy: a phase 3, randomised, double-­ blind, non-­ inferiority trial. Lancet Neurol. 2012;11:579-­ 88. 43. Chadwick D. Safety and efficacy of vigabatrin and carbamazepine in newly diagnosed epilepsy: a multicentre randomised double-­ blind study. Vigabatrin European Monotherapy Study Group. Lancet. 1999;354:13-­ 9. 44. Kwan P, Brodie MJ, Kälviäinen R, Yurkewicz L, Weaver J, Knapp LE, et al. Efficacy and safety of pregabalin versus lamotrigine in patients with newly diagnosed partial seizures: a phase 3, double-­ blind, randomised, parallel-­ group trial. Lancet Neurol. 2011;10:881-­ 90. 45. Glauser TA, Cnaan A, Shinnar S, et al. Ethosuximide, valproic acid, and lamotrigine in childhood absence epilepsy: initial monother- apy outcomes at 12 months. Epilepsia. 2013;54:141-­ 55. 46. Saetre E, Perucca E, Isojärvi J, Gjerstad L, et al. An international multicenter randomized double-­ blind controlled trial of lamotrig- ine and sustained-­ release carbamazepine in the treatment of newly diagnosed epilepsy in the elderly. Epilepsia. 2007;48:1292-­ 302. 47. French JA, Krauss GL, Wechsler RT, Wang X-­ F, DiVentura B, Brandt C, et al. Perampanel for tonic-­ clonic seizures in idiopathic general- ized epilepsy A randomized trial. Neurology. 2015;85:950-­ 7. 48. French J, Brandt C, Friedman D, Biton V, Knapp L, Pitman V, et al. Adjunctive use of controlled-­ release pregabalin in adults with treatment-­ resistant partial seizures: a double-­ blind, randomized, placebo-­ controlled trial. Epilepsia. 2014;55:1220-­ 8. 49. Brodie MJ, Rosenfeld WE, Vazquez B, Sachdeo R, Perdomo C, Mann A, et al. Rufinamide for the adjunctive treatment of par- tial seizures in adults and adolescents: a randomized placebo-­ controlled trial. Epilepsia. 2009;50:1899-­ 909. 50. Brodie MJ, Lerche H, Gil-­ Nagel A, Elger C, Hall S, Shin P, et al. Efficacy and safety of adjunctive ezogabine (retigabine) in refrac- tory partial epilepsy. Neurology. 2010;75:1817-­ 24. 51. French JA, Mosier M, Walker S, Sommerville K, Sussman N, et al. A double-­ blind, placebo-­ controlled study of vigabatrin three g/day in patients with uncontrolled complex partial seizures. Vigabatrin Protocol 024 Investigative Cohort. Neurology. 1996;46:54-­ 61. 52. Elger C, Halász P, Maia J, Almeida L, Soares-­ da-­ Silva P, et al. Efficacy and safety of eslicarbazepine acetate as adjunctive treat- ment in adults with refractory partial-­ onset seizures: a random- ized, double-­ blind, placebo-­ controlled, parallel-­ group phase III study. Epilepsia. 2009;50:454-­ 63. 53. French JA, Baroldi P, Brittain ST, Johnson JK, et al. Efficacy and safety of extended-­ release oxcarbazepine (Oxtellar XR) as adjunc- tive therapy in patients with refractory partial-­ onset seizures: a randomized controlled trial. Acta Neurol Scand. 2014;129:143-­ 53.
  • 15. 350  |     HAKAMI 54. French JA, Costantini C, Brodsky A, von Rosenstiel P, et al. Adjunctive brivaracetam for refractory partial-­ onset seizures: a randomized, controlled trial. Neurology. 2010;75:519-­ 25. 55. Baulac M, Rosenow F, Toledo M, Terada K, Li T, De Backer M, et al. Efficacy, safety, and tolerability of lacosamide monotherapy versus controlled-­ release carbamazepine in patients with newly diagnosed epilepsy: a phase 3, randomised, double-­ blind, non-­ inferiority trial. Lancet Neurol. 2017;16:43-­ 54. 56. French JA, Abou-­ Khalil BW, Leroy RF, Yacubian EMT, Shin P, Hall S, et al. Randomized, double-­ blind, placebo-­ controlled trial of ezoga- bine (retigabine) in partial epilepsy. Neurology. 2011;76:1555-­ 63. 57. Dam M, Ekberg R, Loyning Y, et al. A double-­ blind study compar- ing oxcarbazepine and carbamazepine in patients with newly diag- nosed, previously untreated epilepsy. Epilepsy Res. 1989;3:70-­ 6. 58. Kalviainen R, Aikia M, Saukkonen AM, et al. Vigabatrin vs carba- mazepine monotherapy in patients with newly diagnosed epilepsy. A randomized, controlled study. Arch Neurol. 1995;52:989-­ 96. 59. Ramsay E, Faught E, Krumholz A, Naritoku D, Privitera M, Schwarzman L, et al. Efficacy, tolerability, and safety of rapid initiation of topiramate versus phenytoin in patients with new-­ onset epilepsy: a randomized double-­ blind clinical trial. Epilepsia. 2010;51:1970-­ 7. 60. Trinka E, Marson AG, Van Paesschen W, Kälviäinen R, Marovac J, Duncan B, et al. KOMET: an unblinded, randomised, two parallel-­ group, stratified trial comparing the effectiveness of levetiracetam with controlled-­ release carbamazepine and extended-­ release so- dium valproate as monotherapy in patients with newly diagnosed epilepsy. J Neurol Neurosurg Psychiatry. 2013;84:1138-­ 47. 61. Trinka E, Ben-­ Menachem E, Kowacs PA, Elger C, Keller B, Löffler K, et al. Efficacy and safety of eslicarbazepine acetate versus controlled-­ release carbamazepine monotherapy in newly diag- nosed epilepsy: a phase III double-­ blind, randomized, parallel-­ group, multicenter study. Epilepsia. 2018;59:479-­ 91. 62. Hakami T, Todaro M, Petrovski S, MacGregor L, Velakoulis D, Tan M, et al. Substitution monotherapy with levetiracetam vs older antiepileptic drugs: a randomized comparative trial. Arch Neurol. 2012;69:1563-­ 71. 63. Lattanzi S, Zaccara G, Giovannelli F, Grillo E, Nardone R, Silvestrini M, et al. Antiepileptic monotherapy in newly diagnosed focal epilepsy. A network meta-­ analysis. Acta Neurol Scand. 2019;139:33-­ 41. 64. Lattanzi S, Trinka E, Del Giovane C, Nardone R, Silvestrini M, Brigo F, et al. Antiepileptic drug monotherapy for epilepsy in the elderly: A systematic review and network meta-­ analysis. Epilepsia. 2019;60:2245-­ 54. 65. Sharawat IK, Panda PK, Kasinathan A, Panda P, Dawman L, Joshi K, et al. Efficacy and tolerability of fenfluramine in patients with Dravet syndrome: A systematic review and meta-­ analysis. Seizure. 2021;85:119-­ 26. 66. Lattanzi S, Brigo F, Trinka E, Zaccara G, Striano P, Del Giovane C, et al. Adjunctive cannabidiol in patients with dravet syndrome: a systematic review and meta-­ analysis of efficacy and safety. CNS Drugs. 2020;34:229-­ 41. 67. Veroniki AA, Cogo E, Rios P, Straus SE, Finkelstein Y, Kealey R, et al. Comparative safety of anti-­ epileptic drugs during pregnancy: a systematic review and network meta-­ analysis of congenital mal- formations and prenatal outcomes. BMC Med. 2017;15:95. 68. Lattanzi S, Brigo F, Cagnetti C, Trinka E, Silvestrini M, et al. Efficacy and safety of adjunctive cannabidiol in patients with Lennox-­ Gastaut syndrome: a systematic review and meta-­ analysis. CNS Drugs. 2018;32:905-­ 16. 69. Brigo F, Del Giovane C, Nardone R, Trinka E, Lattanzi S, et al. Intravenous antiepileptic drugs in adults with benzodiazepine-­ resistant convulsive status epilepticus: a systematic review and network meta-­ analysis. Epilepsy Behav. 2019;101:106466. 70. Krumholz A, Shinnar S, French J, et al. Evidence-­ based guideline: management of an unprovoked first seizure in adults: report of the guideline development subcommittee of the American Academy of Neurology and the American Epilepsy Society. Neurology. 2015;85:1526-­ 7. 71. Glauser T, Ben-­ Menachem E, Bourgeois B, Cnaan A, Chadwick D, Guerreiro C, et al. ILAE treatment guidelines: evidence-­ based analysis of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes. Epilepsia. 2006;47:1094-­ 120. 72. Glauser T, Ben-­ Menachem E, Bourgeois B, Cnaan A, Guerreiro C, Kälviäinen R, et al. Updated ILAE evidence review of antiepileptic drug efficacy and effectiveness as initial monotherapy for epilep- tic seizures and syndromes. Epilepsia. 2013;54:551-­ 63. 73. Appleton RE, Freeman A, Cross JH. Diagnosis and management of the epilepsies in children: a summary of the partial update of the 2012 NICE epilepsy guideline. Arch Dis Child. 2012;97:1073-­ 6. 74. Nunes VD, Sawyer L, Neilson J, Sarri G, Cross JH, et al. Diagnosis and management of the epilepsies in adults and children: summary of updated NICE guidance. BMJ. 2012;344:e281. 75. Network SIG. SIGN 143-­ Diagnosis and management of epilepsy in adults. 2018. 76. Network SIG. Diagnosis and Management of Epilepsy in adults. SIGN national clinical guideline 143. Edinburgh: SIGN; 2015. 77. Stephen LJ, Sills GJ, Leach JP, Butler E, Parker P, Hitiris N, et al. Sodium valproate versus lamotrigine: a randomised comparison of efficacy, tolerability and effects on circulating androgenic hor- mones in newly diagnosed epilepsy. Epilepsy Res. 2007;75:122-­ 9. 78. Brodie MJ, Richens A, Yuen AW. Double-­ blind comparison of la- motrigine and carbamazepine in newly diagnosed epilepsy. UK Lamotrigine/Carbamazepine Monotherapy Trial Group. Lancet. 1995;345:476-­ 9. 79. Reunanen M, Dam M, Yuen AW. A randomised open multicentre comparative trial of lamotrigine and carbamazepine as mono- therapy in patients with newly diagnosed or recurrent epilepsy. Epilepsy Res. 1996;23:149-­ 55. 80. Nieto-­ Barrera M, Brozmanova M, Capovilla G, Christe W, Pedersen B, Kane K, et al. A comparison of monotherapy with lamotrigine or carbamazepine in patients with newly diagnosed partial epilepsy. Epilepsy Res. 2001;46:145-­ 55. 81. Tanganelli P, Regesta G. Vigabatrin vs. carbamazepine monother- apy in newly diagnosed focal epilepsy: a randomized response conditional cross-­ over study. Epilepsy Res. 1996;25:257-­ 62. 82. Aikia M, Jutila L, Salmenpera T, Mervaala E, Kalviainen R, et al. Comparison of the cognitive effects of tiagabine and carbamaz- epine as monotherapy in newly diagnosed adult patients with partial epilepsy: pooled analysis of two long-­ term, randomized, follow-­ up studies. Epilepsia. 2006;47:1121-­ 7. 83. Christe W, Krämer G, Vigonius U, Pohlmann H, Steinhoff BJ, Brodie MJ, et al. A double-­ blind controlled clinical trial: oxcarba- zepine versus sodium valproate in adults with newly diagnosed epilepsy. Epilepsy Res. 1997;26:451-­ 60. 84. Tomson T, Battino D, Perucca E. Teratogenicity of antiepileptic drugs. Curr Opin Neurol. 2019;32:246-­ 52. 85. Roger JP, Michael AR. Antiseizure drugs. In: Katzung BG, editor. Basic clinical pharmacology. McGraw-­ Hill; 2018, p. 409-­ 39. 86. French JA. Seizure exacerbation by antiepileptic drugs. Epilepsy Curr. 2005;5:192-­ 3. 87. Silvennoinen K, Lange N, Zagaglia S, Balestrini S, Androsova G, Wassenaar M, et al. Comparative effectiveness of antiepileptic drugs in juvenile myoclonic epilepsy. Epilepsia Open. 2019;4:420-­ 30. 88. Wallace A, Wirrell E, Kenney-­ Jung DL. Pharmacotherapy for dravet syndrome. Paediatr Drugs. 2016;18:197-­ 208. 89. Lagae L, Sullivan J, Knupp K, Laux L, Polster T, Nikanorova M, et al. Fenfluramine hydrochloride for the treatment of seizures in
  • 16.     | 351 HAKAMI Dravet syndrome: a randomised, double-­ blind, placebo-­ controlled trial. Lancet. 2019;394:2243-­ 54. 90. Trinka E, Cock H, Hesdorffer D, Rossetti AO, Scheffer IE, Shinnar S, et al. A definition and classification of status epilepticus–­ report of the ILAE task force on classification of status epilepticus. Epilepsia. 2015;56:1515-­ 23. 91. Glauser T, Shinnar S, Gloss D, Alldredge B, Arya R, Bainbridge J, et al. Evidence-­ based guideline: treatment of convulsive status ep- ilepticus in children and adults: report of the guideline committee of the American Epilepsy Society. Epilepsy Curr. 2016;16:48-­ 61. 92. Haut SR. Seizure clusters: characteristics and treatment. Curr Opin Neurol. 2015;28:143-­ 50. 93. Werhahn KJ, Trinka E, Dobesberger J, Unterberger I, Baum P, Deckert-­ Schmitz M, et al. A randomized, double-­ blind comparison of antiepileptic drug treatment in the elderly with new-­ onset focal epilepsy. Epilepsia. 2015;56:450-­ 9. 94. Ettinger AB. Psychotropic effects of antiepileptic drugs. Neurology. 2006;67:1916-­ 25. 95. Tomson T, Battino D, Bonizzoni E, Craig J, Lindhout D, Sabers A, et al. Dose-­ dependent risk of malformations with antiepileptic drugs: an analysis of data from the EURAP epilepsy and pregnancy registry. Lancet Neurol. 2011;10:609-­ 17. 96. Patorno E, Bohn RL, Wahl PM, et al. Anticonvulsant medications and the risk of suicide, attempted suicide, or violent death. JAMA. 2010;303(14):1401-­ 9. 97. Gibbons RD, Hur K, Brown CH, Mann JJ, et al. Relationship be- tween antiepileptic drugs and suicide attempts in patients with bipolar disorder. Arch Gen Psychiatry. 2009;66:1354-­ 60. 98. Shiek Ahmad B, Hill KD, O'Brien TJ, Gorelik A, Habib N, Wark JD, et al. Falls and fractures in patients chronically treated with antie- pileptic drugs. Neurology. 2012;79:145-­ 51. 99. Chukwu J, Delanty N, Webb D, Cavalleri GL, et al. Weight change, genetics and antiepileptic drugs. Expert Rev Clin Pharmacol. 2014;7:43-­ 51. 100. French JA, Perucca E, Sander JW, Bergfeldt L, Baulac M, Auerbach DS, et al. FDA safety warning on the cardiac effects of lamotrigine: an advisory from the Ad Hoc ILAE/AES task force. Epilepsy Curr. 2021: 1535759721996344. 101. Dixon R, Alexander S, Brickel N. Effect of lamotrigine on the PR interval in healthy subjects. Br J Clin Pharmacol. 2011;71:961-­ 2. 102. Perucca E, Brodie MJ, Kwan P, Tomson T, et al. 30 years of second-­ generation antiseizure medications: impact and future perspec- tives. Lancet Neurol. 2020;19:544-­ 56. 103. Chen Z, Brodie MJ, Kwan P. What has been the impact of new drug treatments on epilepsy? Curr Opin Neurol. 2020;33:185-­ 90. 104. Drew L. Gene therapy targets epilepsy. Nature. 2018;564:S10-­ 1. 105. Rogawski MA, Zolkowska D. Anticonvulsant activity of steroids. Google Patents. 2021. How to cite this article: Hakami T. Neuropharmacology of Antiseizure Drugs. Neuropsychopharmacol Rep. 2021;41:​ 336–­351. https://doi.org/10.1002/npr2.12196