ANTICONVULSANTS
BY
DR. UMARUDEEN AJIBOLA M.
DEPT. OF PHARMACOLOGY, CHS, UNIABUJA
ANTICONULSANTS: DEFN.
Chemical compds capable of:
preventing/reducing/Stopping/arr
esting seizures
What is a seizure/convulsion?
Seizure = convulsion
Convulsion: result of abnormal bursts of
electrical discharges of brain neurons
Recurrent/chronic seizures = Epilepsies
Seizure/Epilepsy
Partial – localized seizure foci/one
hemisphere
- consciousness preserved
Generalized - Seizure foci multiple/ both
hemispheres/loss of
consciousness
Seizure mech./epileptogenesis
Loss of balance bt. excitatory & inhibitory
brain neuronal electrical discharges
Excessive glutamate-mediated neuronal
excitation
Loss of GABA-A-mediated neuronal inhibition
Causes of seizures/Epilepsies
Idiopathic
Infections e.g. meningitis
Hereditary factors e.g. dominant gene defects
Head injuries
Cerebral tumours
Metab. Dxs e.g. hypoglycemia, hypocalcemia
Degenerative diseases
Partial seizure classification:
Simple partial seizures – seizure foci localized
to a brain portion; consciousness preserved
Complex partial seizures – seizure foci more
generalized; involves limbic system and
temporal lobe; consciousness impaired
Partial seizures secondarily generalized –
seizure foci localized initially and become
generalized subsequently –even may involve
both hemispheres; consciousness impaired
Generalized seizures
Generalized tonic-clonic (grand mal) seizures
– tonic-clonic jerks
Absence (petit mal) seizures – no jerks but
zoning or passing out
Tonic seizures – sustained muscular
contraction
Atonic seizures – atonic muscular paralysis
Clonic and myoclonic seizures
Infantile spasms
Therapeutic essence of seizure
classification:
Petil mal or absence seizures respond well to
ethosuximide & valproate but exacerbated by
phenytoin and carbamazepine.
In contrast, generalized tonic-clonic seizures
and complex partial seizures respond well to
phenytoin, carbamazepine, and lamotrigine
MECHANISMS OF ACTION OF ANTICONVULSANT
DRUGS
↑ of GABAergic (↓) brain ↓neuronal electrical
transmission
↓ of glutamate-mediated excitatory brain
neuronal electrical transmission
Modulation of ionic conductance of the brain
neurons
Pre-synaptic modulation of transmitter
release
Chemistry of Anti-seizure Drugs
Anticonvulsant drugs: sev. chemical classes
For many of these drugs: strong SAR
Most of anticonvulsants: apolar in nature
Chemical classes of anticonvulsants
Barbiturates e.g. Phenobarbital
Hydantoins e.g. Phenytoin
Bromides e.g. Potassium bromide
Fatty acids e.g. Valproic acid, Sodium
valproate. Tiagabine
Benzodiazepine e.g. diazepam, clobasam,
clonazepam, lorazepam
Chemical classes of anticonvulsants
GABA analogs e.g. Gabapentin
Fructose derivatives e.g. Topiramate
Pryrimidinediones e.g. Primidone
Oxazolidinediones e.g. Ethadione
Carboxamides e.g. carbamazepine,
oxcarbazepine
Succinimides e.g. ethosuximide
Aromatic allylic alcohols e.g. Stiripentol
Chemical classes of anticonvulsants
Carbamates e.g. Felbamate
Propiionates e.g. Beclamide
Aldehydes e.g. Paraldehyde
Sulfonamides e.g. Acetazolamide
Triazines e.g. lamotrigine
Ureas e.g. Pheturide
Pyrrolidines e.g Levetiracetam
Pharmacokinetics of Anticonvulsant
drugs
P/kinetics according to chemistry
Mostly apolar /limited water solubility
Most readily absorbed across cell membranes
High oral bio-availability
Most of anti-seizure drugs not protein-
bound
Except phenytoin/tiagabine/valproic acid
Most liver-metabolized/cleared
INDIVIDUAL DRUGS: PHENYTOIN
Phenytoin: A diphenylhydantoin
Phenytoin: prototype of the grp.
Other hydantoin derivatives: fosphenytoin,
mephenytoin, ethotoin & phenacemide
Marktd. as Eptoin/Phenytek/ Dilantin
Avail. as oral and IV formulations
Pharmacokinetics of Pheytoin
Good GIT absorp. c ≈ 100% bio-availabilty
Peak plasma levels from oral dose at 3-
12hours
IM Absorption unpredictable/IM precipitation
IM route not recommended for phenytoin
In contrast, fosphenytoin good IM absorption
Phenytoin: high plasma protein binding
Plasma levels ↓ c uremia/ hypoalbuminemia
Drug CSF levels = free plasma level
Phenytoin accumul. in brain/liver/muscle/fat
Pharmacokinetics of Pheytoin
Inactive metabolites excreted in the urine
Only a very small proportion of dose excreted unchanged
Elimin. dose-dependent
At very low blood levels, elimin. 1st order kinetics
High plasma levels: zero order kinetics & t1/2 ↑
Toxicity risks
Normal t1/2 from 12 to 36 hours
At low blood levels, it takes 5–7 days to reach steady-
state
At higher levels: 4–6 weeks before blood levels are
stable
Mechanism of Action of Phenytoin:
↓ of high-frequency firing of neurons through
action on voltage-gated (VG) Na+ channels
↓ of synaptic release of excitatory glutamate
neurotransmitter
Clinical Uses of Phenytoin:
Partial seizures / generalized tonic-clonic
seizures (pry or 2ndary to another seizure
type)
Focal seizures - Mainly used to protect against
the development of focal seizures with
complex symptomatology (psychomotor
and temporal lobe seizures)
Also effective in controlling partial seizures
with autonomic symptoms
Used as prevention and Rx of seizures
occurring during/after neurosurgery
Clinical Uses of Phenytoin:
Status epilepticus - Considered after failed
treatment using a benzodiazepine due to slow onset
of action
Abnormal heart rhythms- may be used in the Rx
of ventricular tachycardia and sudden episodes
of atrial tachycardia after other antiarrhythmic
medications or cardioversion has failed. It is a class
1b antiarrhythmic
Digoxin toxicity - IV phenytoin is drug of choice for
arrhythmias caused by cardiac glycoside toxicity
Trigeminal neuralgia - 2nd choice drug to
carbamazepine
Therapeutic Levels & Dosage of Phenytoin:
The therapeutic plasma level of phenytoin for most
patients is between 10 and 20 mcg/mL. A loading
dose can be given either orally or intravenously; the
latter, using fosphenytoin, is the method of choice for
convulsive status epilepticus.
When oral therapy is started, it is common to
begin adults at a dosage of 300 mg/d, regardless of
body weight. This may be acceptable in some
patients, but it frequently yields steady-state blood
levels below 10 mcg/mL, which is the minimum
therapeutic level for most patients. If seizures
continue, higher doses are usually necessary to
achieve plasma levels in the upper therapeutic range.
Therapeutic Levels & Dosage of Phenytoin:
Because of its dose-dependent kinetics, some toxicity
may occur with only small increments in dosage. The
phenytoin dosage should be increased each time by only
25–30 mg in adults, and ample time should be allowed
for the new steady state to be achieved before further
increasing the dosage. A common clinical error is to
increase the dosage directly from 300 mg/d to 400
mg/d; toxicity frequently occurs at a variable time
thereafter. In children, a dosage of 5 mg/kg/d should be
followed by readjustment after steady-state plasma levels
are obtained.
Fosphenytoin sodium is available for IV or IM use and
replaces iv phenytoin sodium, a much less soluble form
of the drug.
Drug-drug Interactions & Interference
with Laboratory Tests of Phenytoin
Adverse drug interactions prily due to protein binding or
to metabolism
Since phenytoin is 90% bound to plasma proteins, other
highly bound drugs, such as phenylbutazone and
sulfonamides, can displace phenytoin from its binding site
A decrease in protein binding—eg, from hypoalbuminemia
— results in a decrease in the total plasma concentration
of drug but not the free concentration. Intoxication may
occur if efforts are made to maintain total drug levels in
the therapeutic range by increasing the dose
Phenytoin Protein binding ↓ in renal disease
Drug-drug Interactions & Interference
with Laboratory Tests of Phenytoin
The drug has an affinity for thyroid-binding
globulin, which confuses some tests of
thyroid function; the most reliable screening
test of thyroid function in patients taking
phenytoin appears to be measurement of
thyroid stimulating hormone (TSH)
Phenytoin has been shown to induce
microsomal enzymes responsible for the
metabolism of a number of drugs. Auto
stimulation of its own metabolism, however,
appears to be insignificant
Phenytoin Toxicity:
Nystagmus occurs early, as does loss of smooth
extraocular pursuit movements, but neither is an
indication for decreasing the dose.
Diplopia and ataxia are the most common dose-
related adverse effects requiring dosage
adjustment
Sedation occurs only at considerably higher
levels.
Gingival hyperplasia
Hirsutism
Phenytoin Toxicity:
Mild peripheral neuropathy
Vitamin D metabolism, leading to
osteomalacia.
Low folate levels and megaloblastic anemia
Fever
Lymphadenopathy
Agranulocytosis
CARBAMAZEPINE
Carbamazepine, oxycarbazepine &
eslicarbazine
Tricyclic carboxamide derivatives
Structurally similar to imipramine and
phenytoin
Oxycarbazepine & eslicarbazine are prodrugs
with mech. of action, activity spectrums and
p/kinetic profiles similar to carbamazepine
Phenytoin pharmacokinetics:
Absorp. varies among pts, almost complete
absorp. in all pts
Peak levels 6–8 hours after administration
Slowing absorp. by giving the drug after meals
helps pt tolerate larger total daily doses.
Slow distrib. & vol. of distri. ≈ 1 L/kg
≈ 70% protein bound but no displacement of
other drugs from protein binding sites
Very low systemic clearance of ≈ 1 L/kg/d at
the start of therapy
Carbamazepine pharmacokinetics:
Sig. ability to induce microsomal enzymes
Typically, the half-life of 36 hours observed in
subjects after an initial single dose decreases to
as little as 8–12 hours in subjects receiving
continuous therapy.
Carbamazepine also alters the clearance of
other drugs
Carbamazepine is completely metabolized in
humans to several derivatives
One of these, carbamazepine-10, 11-epoxide,
has been shown to have anticonvulsant activity
Mechanism of Action of
carbamazepine:
↓ Na + channels at therapeutic conc.
↓ of high-frequency repetitive firing in brain
neurons
pre-synaptically decreasing synaptic
transmission across brain neurons
potentiation of a voltage-gated K + current in
brain neurons
Clinical Uses of Carbamzepine:
Partial seizures
Generalized tonic-clonic seizures
Trigeminal neuralgia – carbamazepine is drug
of first choice,
Mania in bipolar disorder – Carbamazepine
also useful
Therapeutic Levels & Dosage of
Carbamazepine:
Carbamazepine is available only in oral form
Effective in children: dosage of 15–25 mg/kg/d is appropriate.
In adults, daily doses of 1 g or even 2 g are tolerated
Higher dosage achieved by giving multiple divided doses daily.
Extended release preparations permit bid dosing for most pts
In patients in whom the blood is drawn just before the morning
dose (trough level), the therapeutic level is usually 4–8 mcg/mL.
Although many patients complain of diplopia at drug levels
above 7 mcg/mL, others can tolerate levels above 10 mcg/mL,
especially with monotherapy. Extended-release formulations
that overcome some of these issues are now available
Adverse drug Interactions of
Carbamazepine:
These are majorly related to the drug’s enzyme-inducing
properties. As noted previously, the increased metabolic capacity
of the hepatic enzymes may cause a reduction in steady-state
carbamazepine concentrations and an increased rate of
metabolism of other drugs, eg, primidone, phenytoin,
ethosuximide, valproic acid, and clonazepam
Other drugs such as valproic acid may inhibit carbamazepine
clearance and increase steady-state carbamazepine blood levels
Other anticonvulsants, however, such as phenytoin and
phenobarbital, may decrease steady-state concentrations of
carbamazepine through enzyme induction.
No clinically significant protein-
binding interactions have been reported
Carbamazepone Toxicity:
Diplopia /ataxia. The diplopia often occurs first and
may last less than an hour during a particular time of
day. Rearrangement of the divided daily dose can
often remedy this complaint.
Mild GIT upset
Unsteady gait
Drowsiness
Hyponatremia and water intoxication
Idiosyncratic blood dyscrasias: erythrmatous skin
rash, leucopenia and fatal cases of aplastic anemia
and agranulocytosis
Hepatic dysfunction
PHENOBARBITAL
(PHENOBARBITONE )
Phenobarbital, mephobarbital, metharbital, and
primidone are barbituric acid derivatives
Phenobarbital – the prototype of the barbiturates
is one of the oldest clinical anticonvulsant drugs
still in use in modern medicine
However, the sedative side effect of the
barbiturates has relegated its anticonvulsant
usage to the background by newer less sedative
anticonvulsant drugs
Used as drugs of choice for seizures only in
infants
Pharmacokinetics of Phenobarbital:
Oral bioavailability of about 90%
Peak plasma conc. 8 to 12 hours after oral
dose
One of the longest-acting barbiturates
available
T1/2 2-7 DAYS
Very low protein binding(20 to 45%)
Hepatic metab.
Induces Cytochrome P450 2B6
Excreted primarily by the kidneys
Mechanism of Action of
Phenobarbital:
The exact mechanism of action of
phenobarbital is unknown
But enhancement of phasic GABAA receptor
responses
Inhibition of glutamate-mediated excitatory
synaptic responses of the brain neurons
Clinical Uses of Phenobarbital:
Partial seizures
Generalized tonic-clonic seizures
Resistant seizures of any type
Therapeutic Levels, & Dosage:
Therap. levels in most pts range from 10
mcg/mL to 40 mcg/mL effective for febrile
seizures
Levels below 15 mcg/mL appear ineffective
for prevention of febrile seizure recurrence
Upper end of therap. range difficult to define
because many pts tolerate chronic levels
above 40 mcg/mL
Adverse effects of Phenobarbital:
Sedation and hypnosis -principal side effects
CNS effects e.g.
izziness, nystagmusand ataxia
Adverse drug-drug interactions:
Phenobarbital is a Cytochrome P450 hepatic
enzyme inducer
Drugs that are metabolized by the CYP450
enzyme system will decrease effectiveness
because of faster clearance from the syste.
Caution is to be used with children on
anticonvulsant drugs: behavioural
disturbances occur most frequently
with clonazepam and phenobarbital
Contra-indications to Phenobabital use:
Acute intermittent porphyria
Hypersensitivity to any barbiturate
Prior dependence on barbiturates
Severe respiratory insufficiency
Hyperkinesia in children
PRIMIDONE:
Another barbiturate anticonulsant
A pro-drug metab. to phenobarbital &
phenylethylmalonamide (PEMA)
Both of which are anticonulsants
Pharmacokinetics of Primidone:
Primidone is completely absorbed, usually reaching peak
concentrations about 3 hours after oral administration, although
considerable variation has been reported
Primidone is generally distributed in total body water, with a
volume of distribution of 0.6 L/kg. It is not highly bound to
plasma proteins; approximately 70% circulates as unbound drug.
Primidone is metabolized by oxidation to phenobarbital, which
accumulates very slowly, and by scission of the heterocyclic ring
to form PEMA
Both primidone and Phenobarbital also undergo subsequent
conjugation and excretion.
Pharmacokinetics of Primidone:
Primidone has a larger clearance than most other antiseizure
drugs (2 L/kg/d), corresponding to a half-life of 6–8 hours
PEMA clearance is approximately half that of primidone, but
phenobarbital has a very low clearance. The appearance of
phenobarbital corresponds to the disappearance of
primidone. Phenobarbital therefore accumulates very slowly
but eventually reaches therapeutic concentrations in most
patients when therapeutic doses of primidone are
administered.
During chronic therapy, phenobarbital levels derived from
primidone are usually two to three times higher than
primidone levels
Mechanism of Action:
Although primidone is converted to
phenobarbital, it exhibits phenytoin-like
mechanism of anticovuksant effect.
Clinical Uses of Primidone and
metabolites:
Partial seizures
Generalized tonic-clonic seizures and may be
more effective than phenobarbital
Complex partial seizures – primidone may be
used after carbamazepine and phenytoin
Therapeutic Levels & Dosage
Most effic. when plasma levels - 8–12 mcg/mL
Concomitant levels of its metabolite, phenobarbital, at
steady state usually vary from 15 to 30 mcg/mL
Dosages of 10–20 mg/kg/d neces. to obtain these
levels
Very important to start primidone at low doses and
gradually increase over days to a few weeks to avoid
prominent sedation and gastrointestinal complaints
When adjusting doses of the drug, it is important to
remember that the parent drug reaches steady state
rapidly (30–40 hours), but the active metabolites
phenobarbital (20 days) and PEMA (3–4 days) reach
steady state much more slowly
Primidone Toxicity
Dose-related primidone toxicities similar to
those of its metabolite, phenobarbital, except
that drowsiness occurs early in treatment and
may be prominent if the initial dose is too
large
Gradual increments are indicated when
starting the drug in either children or adults
GABAPENTIN AND PREGABALIN
GABA analogs effective
against partial seizures
Pharmacokinetics of Gabapentin and Pregabalin:
Gabapentin not metab./not induce hepatic
enzymes
Absorp. non-linear/dose-dependent at very
high doses
But linear elimin. Kinetics
Not bound to plasma protein
Drug-drug interactions negligible
Elimin. Renal & excreted unchanged
Half-life is 5 to 8 hours; the drug is
administered two or three times per day
Pharmacokinetics of Pregabalin
Pregabalin, like gabapentin, not metabolized &
almost entirely excreted unchanged in the
urine
Not protein bound & virtually no drug-drug
interactions, again resembling the
characteristics of gabapentin
Likewise, other drugs do not affect the
pharmacokinetics of pregabalin
Half-life of pregabalin is 4.5 hours to 7.0
hours, thus requiring more than once-daily
dosing in most patients
Mechanism of Action of Gabapentin
and Pregabalin:
In spite of their close structural resemblance to GABA,
gabapentin and pregabalin do not act directly on GABA
receptors
May, however, modify the synaptic or nonsynaptic release
of GABA. An increase in brain GABA concentration is
observed in patients receiving gabapentin. Gabapentin is
transported into the brain by the L -amino acid transporter
Gabapentin and pregabalin bind avidly to the α2 subunit of
voltage-gated Ca2+ channels. This appears to underlie the
main mechanism of action, which is decreasing Ca2+
entry, with a predominant effect on presynaptic N-type
channels. A decrease in the synaptic release of glutamate
provides the antiepileptic effect.
Clinical Uses of Gabapentin and Pregabalin:
Gabapentin is effective as:
An adjunct against partial seizures and
generalized tonic-clonic seizures
Gabapentin is now indicated for postherpetic
neuralgia in adults at doses of 1800mg
Pregabalin is used as:
Adjunctive treatment of partialseizures,
Painful diabetic peripheral neuropathy and
postherpetic neuralgia
Fibromyalgia
Generalized anxiety disorder.
Adverse effects of gabapentin and
Pregabalin:
Somnolence
Dizziness
Ataxia
Headache
Tremor
LAMOTRIGINE
Marketed as Lamictal
A phenyltriazine derivative unrelated
chemically with any other class of
anticonvulsants
Exhibits weak anti-folate property..
Therapeutic dosage &
Pharmacokinetics of lamotrigine:
Almost completely absorbed
Vol. of distr. of 1–1.4 L/kg
≈ 55% Protein binding
Linear kinetics & met.d prily by glucuronidation to the 2- N
-glucuronide, which is excreted in the urine
Half-life of ≈ 24 hours: this decreases to 13–15 hours in
patients taking enzyme-inducing drugs
Effective against partial seizures in adults at 100 and 300
mg/d and with a therapeutic blood level near 3 mcg/mL
Valproate causes a twofold increase in the drug’s half-life;
in patients receiving valproate, the initial dosage of
lamotrigine must be reduced to 25 mg every other day
Mechanism of Action of lamotrigine:
Inhibition of sustained rapid firing of neurons
via a voltage- and use-dependent blockade
of Na+ channels
Inhibition of voltage-gated Ca 2+ channels,
particularly the N- and P/Q-type channels,
Inhibition of the synaptic release of glutamate
Clinical Uses of lamotrigine:
Lamotrigine is effective in:
Partial seizures
Absence and myoclonic seizures in children
Seizure controlin the Lennox-Gastaut
syndrome
Bipolar disorder
Adverse effects of lamotrigine:
Dizziness, headache
Diplopia
Nausea
Somnolence
Skin rash
LEVETIRACETAM
Levetiracetam is a piracetam analog
Ineffective against seizures induced by
maximum electroshock or pentylenetetrazol but
has prominent activity in the kindling model
First major drug with this unusual preclinical
profile that is effective against partial seizures
Oral formulations include extended release
tablets
An IV preparation also available
Pharmacokinetics of Levetiracetam
Oral absorption of levetiracetam is nearly
complete
Absorp. Rapid/ unaffected by food
Peak plasma concentrations in 1.3 hours
Kinetics are linear
Protein binding is less than 10%
Half-life is 6–8 hours, but may be longer in the
elderly
Two thirds of the drug excreted unchanged in
urine
Drug has no known active metabolites
Mechanism of Action of Levetiracetam
Binds selectively to the synaptic vesicular
protein SV2A. The function of this protein is
not understood but it is likely that
levetiracetam modifies the synaptic release of
glutamate and GABA through an action on
vesicular function
Clinical Uses of levetiracetam:
Adjunctive treatment of partial seizures in
adults
Primary generalized tonic-clonic in children
seizures
Myoclonic seizures of juvenile myoclonic
epilepsy
Therapeutic dosage of levetiracetam:
Adult dosing can begin with 500 or 1000
mg/d
The dosage can be increased every 2–4
weeks by 1000 mg to a maximum dosage of
3000 mg/d
The drug is dosed twice daily
Adverse effects of levetiracetam:
Somnolence
Asthenia
Ataxia
Dizziness
Psychotic reactions
TOPIRAMATE
A substituted monosaccharide
Structurally different from all other anti-
seizure drugs
Pharmacokinetics of Topiramate:
Rapidly absorbed within 2 hours and i80%
bioavailable.
No food effect on absorption/15% plasma
protein binding
20–50% metabolism
No active metabolites formed
Drug is pryly excreted unchanged in urine
Half-life is 20–30 hours
Birth control pills may be less effective in the
presence of topiramate, and higher estrogen
doses may be required
Mechanism of Action of Topiramate
blockade of voltage-gated Na+ channels
↓ of high-voltage activated (L-type) Ca2+
channels
Potentiation of the inhibitory effect of GABA, at a
site different from benzodiazepine or barbiturate
sites
Depression of the excitatory action of kainate on
glutamate receptors
Multiple effects of topiramate may arise through a
pry action on kinases altering the phosphorylation
of voltage-gated and ligand-gated ion channels
Clinical Uses of Topiramate:
Partial and generalized tonic-clonic seizures
Lennox-Gastaut syndrome
Infantile spasms and even absence seizures
Migraine headaches.
Therapeutic dosages of Topiramate
Dosages typically range from 200 to 600
mg/d, with a few patients tolerating dosages
higher than 1000 mg/d
Most clinicians begin at a low dose (50 mg/d)
and increase slowly to prevent adverse effects
Several studies have used topiramate in
monotherapy with encouraging results.
Adverse effects of Topiramate:
Somnolence
Fatigue
Dizziness
Cognitive slowing
Paresthesias
Nervousness
Confusion
Acute myopia and glaucoma
Urolithiasis
Teratogenic potential
VIGABATRIN
Vigabatrin is a novel anticonvulsant drug
novel molecular targets for therapeutic
efficacy:
GABA agonists and prodrugs
GABA transaminase inhibitors
GABA uptake inhibitors
Mechanism of Action of Vigabatrin:
Vigabatrin is an irreversible inhibitor of GABA
aminotransferase (GABA-T), the enzyme responsible
for the degradation of GABA
It may also inhibit the vesicular GABA transporter
Vigabatrin produces a sustained increase in the
extracellular concentration of GABA in the brain.
This leads to some desensitization of synaptic
GABAA receptors but prolonged activation of
nonsynaptic GABAA receptors that provide tonic
inhibition. A decrease in brain glutamine synthetase
activity is probably secondary to the increased GABA
concentrations
Clinical Uses of Vigabatrin:
Rx of partial seizures
Infantile Spasms (resistant form only)
Therapeutic dosages and
Pharmacokinetics of Vigabatrin:
The half-life is approximately 6–8 hours, but
considerable evidence suggests that the
pharmacodynamic activity of the drug is more
prolonged and not well correlated with the
plasma half-life.
In infants, the dosage is 50–150 mg/d. In
adults, vigabatrin should be started at an oral
dosage of 500 mg twice daily; a total of 2–3 g
(rarely more) daily may be required for full
effectiveness.
Adverse effects of Vigabatrin:
Drowsiness
Dizziness
Weight gain
Agitation
Confusion
Psychosis; preexisting mental illness is a relative
contraindication. The drug was delayed in its worldwide
introduction by the appearance in rats and dogs of a
reversible intramyelinic edema. This phenomenon has
now been detected in infants taking the drug; the clinical
significance is unknown
Peripheral visual field defects in 30–50% of long term
patients
ETHOSUXIMIDE
Ethosuximide, phensuximide, and
methsuximide: succinimides derivatives
Basic cyclic ureide structure
Ethosuximide is 2-ethyl-2-
methylsuccinimide.
Pharmacokinetics of Ethosuximide:
Absorp.complete ff. oral dosage forms
Peak levels 3–7 hours after oral capsules
Ethosuximide not protein-bound
Drug hydroxylated to inactive metabolites
Ethosuximide has a very low total body
clearance (0.25 L/kg/d)
Half-life of approximately 40 hour
Mechanism of Action of
Ethosuximide:
The anti-absence attack effect of
ethosuximide is achieved by:
Inhibition of the T-type Ca2+ currents are
thought to provide a pacemaker current in
thalamic neurons responsible for generating
the rhythmic cortical discharge of an absence
attack
Inhibition of inwardly rectifying K+ channels
Clinical Uses of Ethosuximide:
Particularly
effective
against absence
seizures
Therapeutic Levels & Dosage of
Ethosuximide:
Therapeutic levels of 60–100 mcg/mL can be
achieved in adults with dosages of 750–1500
mg/d, although lower or higher dosages and
blood levels (up to 125 mcg/mL) may be
necessary and tolerated in some patients
Ethosuximide has a linear relationship
between dose and steady-state plasma levels.
The drug might be administered as a single
daily dose were it not for its adverse
gastrointestinal effects; twice-a-day dosage
is common
Drug Interactions & Toxicity of
Ethosuximide:
Administration of ethosuximide with valproic
acid results in a decrease in ethosuximide
clearance and higher steady-state
concentrations owing to inhibition of
metabolism
No other important drug interactions have
been reported for the succinimides.
Adverse effects of Ethosuximide:
Gastric distress - pain, nausea, and vomiting
Transient lethargy or fatigue
Headache
Dizziness
Hiccup
Euphoria
VALPROIC ACID & SODIUM
VALPROATE:
The anti-seizure property of sodium valproate and valproic acid
was discovered serendipitously when they were used as solvents
for other drugs in the search for therapeutic agents effective
against seizures.
Valproic acid is fully ionized at body pH, and for that reason the
active form of the drug may be assumed to be the valproate ion
regardless of whether valproic acid or a salt of the acid is
administered.
Valproic acid is one of a series of fatty carboxylic acids that have
antiseizure activity; this activity appears to be greatest for
carbon chain lengths of five to eight atoms. The amides and
esters of valproic acid are also active antiseizure agents.
It is available as oral syrups and capsules and IV formulation
Pharmacokinetics of Valproic acid:
Valproate is well absorbed after an oral dose, with bioavailability
greater than 80%. Peak blood levels are observed within 2 hours.
Food may delay absorption, and decreased toxicity may result if the
drug is given after meals.
Valproic acid is 90% bound to plasma proteins, although the fraction
bound is somewhat reduced at blood levels greater than 150
mcg/mL. Since valproate is both
highly ionized and highly proteinbound, its distribution is essentially
confined to extracellular water with a volume of distribution of
approximately 0.15 L/kg. At higher doses, there is an increased free
fraction of valproate, resulting in
lower total drug levels than expected. It may be clinically useful,
therefore, to measure both total and free drug levels.
Clearance for valproate is low and dose
dependent; its half-life varies from 9 to 18 hours. Approximately 20%
of the drug is excreted as a direct conjugate of valproate.
Mechanism of Action of Valproic
acid:
Inhibition of Na + currents
Blockade of NMDA receptor-mediated
excitation
Inhibition of the GABA transporter GAT-1
Inhibition of GABA transaminase and thus
blockade of degradation of GABAin the brain
Clinical Uses of Valproic acid:
Very effective against absence seizures
Absence seizures with concomitant generalized
tonic-clonic attacks: preferred to ethosuximide
Certain types of myoclonic seizures
Effective in tonic-clonic seizures, especially those
that are primarily generalized
Atonic attacks may also respond
Partial seizures
Status epilepticus: IV formulation
Bipolar disorder
Migraine prophylaxis
Therapeutic dosages of valproic
acid:
Dosages of 25–30 mg/kg/d may be adequate
in some patients, but others may require 60
mg/kg/d or even more
Therapeutic levels of valproate range from 50
to 100 mcg/mL.
Drug Interactions of Vaproic acid:
Valproate displaces phenytoin from plasma
proteins In
addition to binding interactions, valproate inhibits
the metabolism of several drugs, including
phenobarbital, phenytoin, and carbamazepine,
leading to higher steady-state concentrations of
these agents
Inhibition of phenobarbital metabolism, for
example, may cause levels of the barbiturate to
rise steeply, causing stupor or coma
Valproate can dramatically decrease the clearance
of lamotrigine.
Toxicity of Vaproic acid:
GIT effects: nausea, vomiting, abdominal pain
and heartburn
Sedation is uncommon with valproate alone but
may be striking when valproate is added to
Phenobarbital
A fine tremor is frequently seen at higher levels
Weight gain, increased appetite, and hair loss
Hepatoxicity
Thrombocytopenia
Teratogenic risk to fetuses of mothers on the
dru
BENZODIAZEPINES
Benzodiazepines are primarily anxiolytic
sedatives whose anticonvulsant usage has
gained currency in the therapy of epilepsy
They are chemically 1,4-benzodiazepines,
and most contain a carboxamide group in the
7-membered heterocyclic ring structure. Six
benzodiazepines are known to be used in the
management of seizures
Pharmacokietics of Benzodiazepines:
The rates of oral absorption of sedative-hypnotics differ depending
on a number of factors, including lipophilicity. For example, the
absorption of triazolam is extremely rapid, and
that of diazepam and the active metabolite of clorazepate is more
rapid than other commonly used benzodiazepines.
Clorazepate, a prodrug, is converted to its active form,
desmethyldiazepam (nordiazepam), by acid hydrolysis in the stomach.
High lipid solubility is responsible for the rapid onset of central
nervous system effects of triazolam, All benzodiazepines cross the
placental barrier during pregnancy. If enzodiazepines are given during
the predelivery period, they may contribute to the depression of
neonatal vital functions. Benzodiazepines are also detectable in breast
milk and may exert depressant effects in the nursing infant.
Metabolic transformation to more water-soluble metabolites is
necessary for clearance of benzodiazepines from the body. The
microsomal drug-metabolizing enzyme systems of the liver are
Pharmacokietics of Benzodiazepines:
Hepatic metabolism accounts for the clearance of all
benzodiazepines. The patterns and rates of metabolism depend on
the individual drugs. Most benzodiazepines undergo microsomal
oxidation (phase I reactions), including N- dealkylation and aliphatic
hydroxylation catalyzed by cytochrome P450 isozymes, especially
CYP3A4. The metabolites are subsequently conjugated (phase II
reactions) to form glucuronides that are excreted in the urine.
However, many phase I metabolites of benzodiazepines are
pharmacologically active, some with long half-lives. For example,
desmethyldiazepam, which has an elimination half-life of more than
40 hours, is an active metabolite of chlordiazepoxide, diazepam,
prazepam, and clorazepate. Alprazolam and triazolam undergo α-
hydroxylation, and the resulting metabolites appear to exert short-
lived pharmacologic effects because they are rapidly conjugated to
form inactiveglucuronides. The short elimination half-life of
triazolam (2–3 hours) favors its use as a hypnotic rather than as a
sedative drug.
Pharmacokietics of Benzodiazepines:
The formation of active metabolites has complicated studies on
the pharmacokinetics of the benzodiazepines in humans because
the elimination half-life of the parent drug may have little relation
to the time course of pharmacologic effects. Benzodiazepines for
which the parent drug or active metabolites have long half-lives
are predictably more likely to cause cumulative effects with
multiple doses. Cumulative and residual effects such as excessive
drowsiness appear to be less of a problem with such drugs as
estazolam, oxazepam, and lorazepam, which have relatively short
half-lives and are metabolized directly to inactive glucuronides.
The metabolism of several commonly used benzodiazepines
including diazepam, midazolam, and triazolam isaffected by
inhibitors and inducers of hepatic P450 isozymes
Mechanisms of action of the
benzodiazepines:
Although the benzodiazepines are similar
chemically, subtle structural alterations result
in differences in activity
The main mechanism of anti-seizure action,
which is shown to different degrees by the six
compounds is as follows:
Mechanisms of action of the
benzodiazepines:
Bind to specific GABAA receptor subunits at
central nervous system (CNS) neuronal
synapses facilitating frequency of GABA-
mediated chloride ion channel opening—
enhance membrane hyperpolarization
Diazepam:
Highly effective for stopping continuous
seizure activity, especially generalized tonic-
clonic status epilepticus. It is given IV or PR
Very effective refractory patients who need
acute control of bouts of seizure activity.
Given PR.
Lorazepam:
More effective and longer
acting than diazepam in the
treatment of status
epilepticus and is preferred
by some experts
Clonazepam:
Long-acting drug with efficacy against absence seizures
One of the most potent antiseizure agents known
Effective in some cases of myoclonic seizures
Helpful ininfantile spasms
Sedation is prominent, especially on initiation of therapy
Starting doses should be small. Maximal tolerated doses
are usually in the range of 0.1–0.2 mg/kg, but many weeks
of gradually increasing daily doses may be needed to
achieve these dosages in some patients within a few
months. The remarkable antiseizure potency
Nitrazepam:
Less potent than clonazepam for infantile
spasms and myoclonic seizures
Clorazepate dipotassium:
Adjunct in treatment of complex partial
seizures in adults
Drowsiness and lethargy are common adverse
effects
Clobazam:
It is a 1,5- benzodiazepine (other marketed
benzodiazepines are 1,4-benzodiazepines)
Less sedative potential than others
Half-life of 18 hours
Effective at dosages of 0.5–1 mg/kg/d
Does interact with some other anti-seizure
drugs
Limitations of benzodiazepines in
Seizure treatment
Pronounced sedative effect, which is
unfortunate both in the treatment of status
epilepticus and in chronic therapy. Children
may manifest a paradoxical hyperactivity, as
with barbiturates
Tolerance, in which seizures may respond
initially but recur
ACETAZOLAMIDE
Acetazolamide is a sulfonamide derivative
whose diuretic whose main action is the
inhibition of carbonic anhydrase
Acetazolamide anti-seizure
mechanisms:
Via carbonic anhydrase inhibition/Mild
acidosis in the brain
GABA receptor ion channel-mediated hyper-
polarizing action
Therapeutic indications of Acetazolamide:
Special role in epileptic women who experience
seizure exacerbations at the time of menses
Seizure control may be improved and tolerance
may not develop because the drug is not
administered continuously
The usual dosage is approximately 10 mg/kg/d
to a maximum of 1000 mg/d
Its uses for long-term anti-seizure control
limited largely by tolerance development to the
drugs
FELBAMATE
Felbamate use as anti-seizure agent stopped on the
ground of severe adverse effects such as aplastic
anemia and severe hepatitis at unexpectedly high
rates and has been relegated to the status of a third-
line drug for refractory cases
In spite of the seriousness of the adverse effects,
thousands of patients worldwide utilize this
medication. Usual dosages are 2000–4000 mg/d in
adults, and effective plasma levels range from 30
mcg/mL to 100 mcg/mL. In addition to its usefulness
in partial seizures, felbamate has proved effective
against the seizures that occur in Lennox-Gastaut
syndrome
THERAPEUTIC STRATEGY
It is often clinically cost-effective when
initiating anti-seizure therapy to start with
the use of a single drug, especially in patients
who are not severely affected and who can
benefit from the advantage of fewer adverse
effects using monotherapy
For patients with hard-to-control seizures,
multiple drugs are usually utilized
simultaneously.
THERAPEUTIC STRATEGY
For most of the older anti-seizure drugs, relationships
between blood levels and therapeutic effects have
been characterized to a high degree
The same is true for the
pharmacokinetics of these drugs. These relationships
provide significant advantages in the development of
therapeutic strategies for the treatment of epilepsy
The therapeutic index for most anti-seizure drugs is
low, and toxicity is not uncommon. Thus, effective
treatment of seizures often requires an awareness of
the therapeutic levels and pharmacokinetic profiles of
the drugs in question
MANAGEMENT OF EPILEPSY: PARTIAL SEIZURES
& GENERALIZED TONIC-CLONIC SEIZURES
It is clinically trending to use newer drugs such as
gabapentin, Primidone, sodium valproate, and
topiramate in place of phenytoin, carbamazepine
and barbiturates for most partial and general
seizures, This is because these newer anti-seizure
agents not only have broader activity spectrum but
also safer toxicity profiles than the older group
Whichever anti-seizure drug used, it must be
gradually withdrawn whenever a drug’s use is to be
terminated. When there is no seizure attack for 3-4
years, therapy termination can be iniated albeit
gradually
GENERALIZED SEIZURES
The drugs used for generalized tonic-clonic
seizures are the same as for partial seizures;
in addition, valproate is clearly useful.
At least three drugs are effective against
absence seizures. Two are non-sedating and
therefore preferred: ethosuximide and
valproate.
Clonazepam is also highly effective but has
disadvantages of dose-related adverse effects
and development of tolerance. Lamotrigine
and topiramate may also be useful.
GENERALIZED SEIZURES
Specific myoclonic syndromes are usually treated with
valproate; an intravenous formulation can be used
acutely if needed
It is non-sedating and can be dramatically effective.
Other patients respond to clonazepam, nitrazepam, or
other benzodiazepines, although high doses may be
necessary, with accompanying
drowsiness. levetiracetam may be useful.
Another specific myoclonic
syndrome, juvenile myoclonic epilepsy, can be
aggravated by phenytoin or carbamazepine; valproate
is the drug of choice followed by lamotrigine and
topiramate
GENERALIZED SEIZURES
Atonic seizures are often refractory to all available medications,
although some reports suggest that valproate may be beneficial,
as may lamotrigine
Benzodiazepines have been reported to improve seizure control
in some of these patients but may worsen the attacks in others
Felbamate has been demonstrated to be effective in some
patients, although the drug’s idiosyncratic toxicity limits its use.
If the loss of tone
appears to be part of another seizure type (e.g. absence or
complex partial seizures), every effort should be made to treat
the other seizure type vigorously
This is done hoping for simultaneous alleviation of the atonic
component of the seizure
The ketogenic diet may also be useful
GENERALIZED SEIZURES
DRUGS USED IN INFANTILE SPASMS
Repository corticotrophin
Oral or parenteral prednisone
Benzodiazepines e.g. clonazepam or
nitrazepam; their efficacy in this
heterogeneous syndrome may be nearly as
good as that of corticosteroids
Vigabatrin is effective and is considered the
drug of choice by many pediatric neurologists
STATUS EPILEPTICUS
Some physicians prefer lorazepam, which is
equivalent to diazepam in effect and perhaps
somewhat longer acting
For patients who are not actually in the throes
of a seizure, diazepam therapy can be
omitted and the patient treated at once with a
long-acting drug such as phenytoin
STATUS EPILEPTICUS
There are many forms of status epilepticus
The most common, generalized tonic-clonic
status epilepticus, is a life-threatening
emergency, requiring immediate
cardiovascular, respiratory, and metabolic
management as well as pharmacologic therapy
IV Diazepam is the most effective drug in most
patients for stopping the attacks and is given
directly by IV push to a maximum total dose of
20–30 mg in adults
STATUS EPILEPTICUS
Until the introduction of fosphenytoin, the mainstay of
continuing therapy for status epilepticus was
intravenous phenytoin, which is effective and
nonsedating. It can be given as a loading dose of 13–
18 mg/kg in adults; the usual error is to give too little.
Administration should be at a maximum rate of 50
mg/min. It is safest to give the drug directly by
intravenous push, but it can also be diluted in saline; it
precipitates rapidly in the presence of glucose. Careful
monitoring of cardiac rhythm and blood pressure is
necessary, especially in elderly people. At least part of
the cardiotoxicity is from the propylene glycol in which
the phenytoin is dissolved
STATUS EPILEPTICUS
Fosphenytoin, which is freely soluble in intravenous
solutions without the need for propylene glycol or
other solubilizing agents, is a safer parenteral
agent. Because of its greater molecular weight, this
prodrug is two thirds to three quarters as potent as
phenytoin on a milligram basis
In previously treated epileptic patients, the
administration of a large loading dose of phenytoin
may cause some dose-related toxicity such as
ataxia. This is usually a relatively minor problem
during the acute status episode and is easily
alleviated by later adjustment of plasma levels
STATUS EPILEPTICUS
Although other drugs such as lidocaine have
been recommended for the treatment of
generalized tonic-clonic status epilepticus,
general anesthesia is usually necessary in
highly resistant cases
For patients in absence status,
benzodiazepines are still drugs of first choice
Rarely, intravenous valproate may be required
STATUS EPILEPTICUS
IV diazepam may depress respiration (less
frequently, cardiovascular function), and
facilities for resuscitation must be
immediately at hand during its
administration. The effect of diazepam is not
lasting, but the 30- to 40-minute seizure-
free interval allows more definitive therapy to
be initiated
STATUS EPILEPTICUS
For patients who do not respond to
phenytoin, Phenobarbital can be given in
large doses: 100–200 mg intravenously to a
total of 400–800 mg. Respiratory depression
is a common complication, especially if
benzodiazepines have already been given,
and there should be no hesitation in
instituting intubation and ventilation
Non-seizure therapeutic uses of
Anticonvulsant drugs:
Although seizure control or epilepsy has
become the major clinical indication for
anticonvulsant therapy, there are other non-
seizure/non-epileptic clinical indications for
which anticonvulsant drugs are deployed
These include: trigeminal neuralgia, migraine,
chronic neuropathic pain syndromes,
essential tremors, bipolar disorders and
post-herpetic neuralgia
Non-seizure therapeutic uses of
Anticonvulsant drugs:
Trigeminal neuralgia – Carbamazepine, Primidone
Migraine prophylaxis - Valproic acid (sodium valproate)
Essential tremor - Valproic acid (sodium valproate) is
commonly prescribed as first-line therapy for patients
with trigeminal neuralgia. has been recently marketed
for the management of
Diabetic neuropathic pain syndromes - Gabapentin
particularly diabetic neuropathy and
Post-herpetic neuralgia – Gabapentin
Bipolar disorders - Carbamazepine, Lamotrigine,
oxcarbazepine, gabapentin and topiramate valproic
acid
Non-drug anticonvulsant therapies:
Vagal nerve stimulation
Ketogenic diets e.g. high dose omega-3 fatty
acids
Epilepsy and Special situations
Pregnancy
Contraception
Anesthesia
Surgery
Epilepsy and Contraception
Potential adverse drug-drug interactions between epilepsy
drugs that induce liver enzymes and can lead to increased
hepatic metabolism of oral contraceptives and hence their
lower plasma levels and risks of failure of contraception.
These drugs include:
Carbamazepine Clobazam Eslicarbazepine
Felbamate Lamotrigine at doses of 300 mg daily or more
Oxcarbazepine Phenobarbital Phenytoin
Primidone Rufinamide
Topiramate at doses of 200 mg daily or more.
It is important to note that a woman already on any of these
drugs is placed on oral contraceptives she should be started
on a higher dose in order to avoid unwanted pregnancy.
Adverse effects of oral
contraceptives on lamotrigine
Conversely, Lamotrigine plasma kevels can be
lowered by oral contraceptives since they too
are liver enzymes inducers. This scenario can
lead to breakthrough seizures when
lamotrigine dosage is not adjusted upwards
in an epilepsy patient whose seizure is well
controlled and subsequently placed on an
oral contraceptive
Anticonvulsant drugs with no adverse drug-
drug interactions with contraceptives
Acetazolamide/ Clonazepam/ Diazepam
Clorazepate/ valproic acid/sodium valproate
Ethosuximide/ Gabapentin / Zonisamide
Lamotrigine at doses less than 300 mg daily
Lacosamide / Levetiracetam / Lorazepam
Pregabalin / Tiagabine / Vigabatrin
Effect of antiepileptic Drugs (AEDs) on anaesthesia
There are important pharmacokinetic and
pharmacodynamic interactions between AEDs
and drugs commonly used in anaesthesia
These affect both drug efficacy and the risk
of seizure activity intraoperatively
Induction and inhibition of the cytochrome
P450 isoenzymes in hepatic metabolism
constitutes the most significant mech. of
drug interactions involving AEDs
AEDs with potent enzyme-inducing properties
Carbamazepine, phenytoin, phenobarbital,
Primidone, Topiramate , Oxcarbazepine and
eslicarbazepine
They cause decreased plasma warfarrin,-p
antibacterials, immunosuppressants, and cvs
drugs, e.g. amiodarone, propranolol,
metoprolol, nifedipine, felodipine,
nimodipine, and verapamil concentrations
Important to monitor plasma levels of
warfarrin and other drugs if co-administered
with any of these AEDs in peri-operative
period
AEDs with hepatic microsomal
enzyme systems inhibition
Valproate
May reduce the clearance of many
concurrently administered medications and
even AEDs
AEDs with no effects on hepatic
microsomal enzyme systems
Gabapentin
Lamotrigine
Levetiracetam
Tiagabine
Vigabatrin
Antibiotics with potent inhibition or induction
of hepatic enzymatic metabolism of AEDs
Macrolide antibiotics, particularly
erythromycin, are potent inhibitors of
carbamazepine metabolism and can lead to
carbamazepine toxicity
Concomitant use of carbapenem antibiotics
can lead to a significant decrease in serum
valproate
Effect of anaesthetic agents on epilepsy
Many of the agents used possess
both pro-convulsant and
anticonvulsant properties, which
could impact on the choice of
anaesthetic
Inhalational anaesthetics
Nitrous oxide (N2O) inhalation, Isoflurane,
isoflurane and desflurane suppress seizures
But N2O withdrawal, sevoflurane, enflurane
promote seizures
Hyperbaric N2O in combination with
isoflurane or halothane causes myoclonus
Effects of opioids on anaesthesia
Meperidine, remifentanil, fentanyl,
alfentanil, sufentanil, and morphine
have been associated with increased
seizure activity during anaesthesia
I.V. anaesthetic agents
The barbiturates (thiopental, methohexital, and
pentobarbital) and propofol are well established as agents
for Rx of refractory status epilepticus
All agents have been reported to produce excitatory activity,
such as myoclonus, opisthotonus, and rarely generalized
seizures on induction of anaesthesia
The highest incidence of seizure induction appears to be
with etomidate, followed by thiopental, methohexital, and
propofol
However, at higher doses, all agents act as anticonvulsants
Ketamine is a non-competitive glutamate antagonist acting
at NMDA receptors, a property which could be beneficial in
Rx of status epilepticus refractory to other agents
As with the other i.v. agents, low doses may facilitate
seizures, but at doses that produce sedative or anaesthetic
effects, ketamine shows anticonvulsant properties
Effects of the Benzodiazepines on epilepsy
All benzodiazepines in clinical practice
possess potent anticonvulsant properties
Diazepam, midazolam, and lorazepam are
widely used to terminate episodes of status
epilepticus
Local anaesthetics
LA agents readily cross the blood–brain
barrier, causing sedation and analgesia ff by
generalized convulsions at higher doses
High blood levels result from an accidental
i.v. administration or rapid systemic
absorption from a highly vascular area
I.V. lidocaine has been used to Rx status
epilepticus mainly in children
Neuromuscular blocking agents on anesthesia
None of the neuromuscular blocking agents
e.g. atracurium, Laudanosine or
Succinylcholine appear to have any pro-
convulsant or anticonvulsant effects
Effects of Anticholinergics and anticholinesterases on
anaesthesia
The increase in acetylcholine via
administration of atropine or scopolamine
can produce central cholinergic blockade (or
central anticholinergic syndrome)
This manifests as agitation with seizures,
hallucinations, and restlessness or stupor,
coma, and apnoea
Most effective Rx for this is physostigmine
Glycopyrrolate does not cross the blood–brain
barrier, so does not produce these effects
Perioperative management of AEDs
In pts with well-controlled epilepsy, it is vital that efforts
are made to avoid disruption of antiepileptic medication
perioperatively
Patients should be advised to take their regular
medications on the morning of surgery and regular dosing
should be re-established as early as practicable after
surgery. If a single dose is missed (such as that might
occur with day-case surgery), it should be taken as soon
as possible after surgery
Where multiple doses are likely to be missed, AEDs should
be administered parenterally where possible. I.V. forms of
phenytoin, sodium valproate, and levetiracetam are
available (where i.v. doses are equivalent to oral doses)
and carbamazepine is available as a suppository
If the patient's regular AED is not available in parenteral
formulation, advice should be sought from a neurologist
regarding alternatives that may be used to cover the
perioperative period
Perioperative management of AEDs
In general, routine drug level monitoring is not
required perioperatively as anaesthetic agents
do not significantly alter the pharmacokinetics
of AEDs
However, prolonged intensive care unit (ICU)
stay, with attendant changes in serum pH and
albumin levels and also the use of drugs that
interact with AEDs, can affect their serum
concentrations
Of the commonly used AEDs, phenytoin
presents the greatest challenge because of its
unique pharmacokinetic properties
In patients admitted to ICU, it is necessary to
check serum concentrations of phenytoin daily