CLINICAL PHARMACOLOGY
Mechanism Of Action
The precise mechanism(s) by which lamotrigine exerts its
anticonvulsant action are unknown. In animal models designed to detect
anticonvulsant activity, lamotrigine was effective in preventing seizure spread
in the maximum electroshock (MES) and pentylenetetrazol (scMet) tests, and
prevented seizures in the visually and electrically evoked after-discharge
(EEAD) tests for antiepileptic activity. Lamotrigine also displayed inhibitory
properties in the kindling model in rats both during kindling development and
in the fully kindled state. The relevance of these models to human epilepsy,
however, is not known.
One proposed mechanism of action of lamotrigine, the
relevance of which remains to be established in humans, involves an effect on
sodium channels. In vitro pharmacological studies suggest that lamotrigine
inhibits voltage-sensitive sodium channels, thereby stabilizing neuronal
membranes and consequently modulating presynaptic transmitter release of
excitatory amino acids (e.g., glutamate and aspartate).
Effect Of Lamotrigine On N-Methyl d-Aspartate-Receptor–Mediated
Activity
Lamotrigine did not inhibit N-methyl d-aspartate
(NMDA)-induced depolarizations in rat cortical slices or NMDA-induced cyclic
GMP formation in immature rat cerebellum, nor did lamotrigine displace
compounds that are either competitive or noncompetitive ligands at this
glutamate receptor complex (CNQX, CGS, TCHP). The IC50 for lamotrigine effects
on NMDA-induced currents (in the presence of 3 μM of glycine) in cultured
hippocampal neurons exceeded 100 μM.
The mechanisms by which lamotrigine exerts its
therapeutic action in bipolar disorder have not been established.
Pharmacodynamics
Folate Metabolism
In vitro, lamotrigine inhibited dihydrofolate reductase,
the enzyme that catalyzes the reduction of dihydrofolate to tetrahydrofolate.
Inhibition of this enzyme may interfere with the biosynthesis of nucleic acids
and proteins. When oral daily doses of lamotrigine were given to pregnant rats
during organogenesis, fetal, placental, and maternal folate concentrations were
reduced. Significantly reduced concentrations of folate are associated with
teratogenesis [see Use In Specific Populations]. Folate concentrations
werealso reduced in malerats givenrepeated oral doses of lamotrigine. Reduced concentrations
were partially returned to normal when supplemented with folinic acid.
Accumulation In Kidneys
Lamotrigine accumulated in the kidney of the male rat,
causing chronic progressive nephrosis, necrosis, and mineralization. These
findings are attributed to α-2 microglobulin, a species-and sex-specific
protein that has not been detected in humans or other animal species.
Melanin Binding
Lamotrigine binds to melanin-containing tissues, e.g., in
the eye and pigmented skin. It has been found in the uveal tract up to 52 weeks
after a single dose in rodents.
Cardiovascular
In dogs, lamotrigine is extensively metabolized to a
2-N-methyl metabolite. This metabolite causes dose-dependent prolongation of
the PR interval, widening of the QRS complex, and, at higher doses, complete AV
conduction block. Similar cardiovascular effects are not anticipated in humans
because only trace amounts of the 2-N-methyl metabolite (<0.6% of
lamotrigine dose) have been found in human urine.
However, it is conceivable that plasma concentrations of this metabolite could
be increased in patients with a reduced capacityto glucuronidate
lamotrigine(e.g., in patientswithliverdisease, patientstaking concomitant
medications that inhibit glucuronidation).
Pharmacokinetics
The pharmacokinetics of lamotrigine have been studied in
subjects with epilepsy, healthy young and elderly volunteers, and volunteers
with chronic renal failure. Lamotrigine pharmacokinetic parameters for adult
and pediatric subjects and healthy normal volunteers are summarized in Tables
14 and 16.
Table 14: Mean Pharmacokinetic Parametersa in
Healthy Volunteers and Adult Subjects with Epilepsy
Adult Study Population |
Number of Subjects |
Tmax: Time of Maximum Plasma Concentration (h) |
t½: Elimination Half-life (h) |
CL/F: Apparent Plasma Clearance (mL/min/kg) |
Healthy volunteers taking no other medications: |
Single-dose LAMICTAL |
179 |
2.2 (0.25-12.0) |
32.8 (14.0-103.0) |
0.44 (0.12-1.10) |
Multiple-dose LAMICTAL |
36 |
1.7 (0.5-4.0) |
25.4 (11.6-61.6) |
0.58 (0.24-1.15) |
Healthy volunteers taking valproate: |
Single-dose LAMICTAL |
6 |
1.8 (1.0-4.0) |
48.3 (31.5-88.6) |
0.30 (0.14-0.42) |
Multiple-dose LAMICTAL |
18 |
1.9 (0.5-3.5) |
70.3 (41.9-113.5) |
0.18 (0.12-0.33) |
Subjects with epilepsy taking valproate only: |
Single-dose LAMICTAL |
4 |
4.8 (1.8-8.4) |
58.8 (30.5-88.8) |
0.28 (0.16-0.40) |
Subjects with epilepsy taking carbamazepine, phenytoin, phenobarbital, or primidoneb plus valproate: |
Single-dose LAMICTAL |
25 |
3.8 (1.0-10.0) |
27.2 (11.2-51.6) |
0.53 (0.27-1.04) |
Subjects with epilepsy taking carbamazepine, phenytoin, phenobarbital, or primidone:b |
Single-dose LAMICTAL |
24 |
2.3 (0.5-5.0) |
14.4 (6.4-30.4) |
1.10 (0.51-2.22) |
Multiple-dose LAMICTAL |
17 |
2.0 (0.75-5.93) |
12.6 (7.5-23.1) |
1.21 (0.66-1.82) |
a The majority of parameter means determined
in each study had coefficients of variation between 20% and 40% for half-life
and CL/F and between 30% and 70% for Tmax. The overall mean values were
calculated from individual study means that were weighted based on the number
of volunteers/subjects in each study. The numbers in parentheses below each parameter
mean represent the range of individual volunteer/subject values across studies.
b Carbamazepine, phenytoin, phenobarbital, and primidone have been
shown to increase the apparent clearance of lamotrigine. Estrogen-containing
oral contraceptives and other drugs, such as rifampin and protease inhibitors
lopinavir/ritonavir and atazanavir/ritonavir, that induce lamotrigine
glucuronidation have also been shown to increase the apparent clearance of
lamotrigine [see DRUG INTERACTIONS]. |
Absorption
Lamotrigine is rapidly and completely absorbed after oral
administration with negligible first-passmetabolism (absolute bioavailabilityis
98%). Thebioavailabilityisnotaffected by food. Peak plasma concentrations occur
anywhere from 1.4 to 4.8 hours following drug administration.
Thelamotriginechewable/dispersibletablets werefound to beequivalent, whether
administered as dispersed in water, chewed and swallowed, or swallowed whole,
to the lamotrigine compressed tablets in terms of rate and extent of
absorption. In terms of rate and extent of absorption, lamotrigine orally
disintegrating tablets, whether disintegrated in the mouth or swallowed whole
with water, were equivalent to the lamotrigine compressed tablets swallowed
with water.
Dose Proportionality
In healthy volunteers not receiving any other medications
and given single doses, the plasma concentrations of lamotrigine increased in
direct proportion to the dose administered over the range of 50 to 400 mg. In 2
small studies (n = 7 and 8) of patients with epilepsy who were maintained on
other AEDs, there also was a linear relationship between dose and lamotrigine
plasma concentrations at steady state following doses of 50 to 350 mg twice
daily.
Distribution
Estimates of the mean apparent volume of distribution
(Vd/F) of lamotrigine following oral administration ranged from 0.9 to 1.3
L/kg. Vd/F is independent of dose and is similar following single and multiple
doses in both patients with epilepsy and in healthy volunteers.
Protein Binding
Data from in vitro studies indicate that lamotrigine is
approximately 55% bound to human plasma proteins at plasma lamotrigine
concentrations from 1 to 10 mcg/mL (10 mcg/mL is 4 to 6 times the trough plasma
concentration observed in the controlled efficacy trials). Because lamotrigine
is not highly bound to plasma proteins, clinically significant interactions
with other drugs through competition for protein binding sites are unlikely.
The binding of lamotrigine to plasma proteins did not change in the presence of
therapeutic concentrations of phenytoin, phenobarbital, or valproate.
Lamotrigine did notdisplaceotherAEDs(carbamazepine, phenytoin, phenobarbital)
from protein-binding sites.
Metabolism
Lamotrigine is metabolized predominantly by glucuronic
acid conjugation; the major metabolite is an inactive 2-N-glucuronide
conjugate. After oral administration of 240 mg of 14C-lamotrigine
(15 μCi) to 6 healthy volunteers, 94% was recovered in the urine and 2%
was recovered in the feces. The radioactivity in the urine consisted of
unchanged lamotrigine (10%), the 2-N-glucuronide (76%), a 5-N-glucuronide
(10%), a 2-N-methyl metabolite (0.14%), and other unidentified minor
metabolites (4%).
Enzyme Induction
The effects of lamotrigine on the induction of specific
families of mixed-function oxidase isozymes have not been systematically
evaluated.
Following multiple administrations (150 mg twice daily)
to normal volunteers taking no other medications, lamotrigineinduced its own
metabolism, resultingin a 25% decreasein t½ and a 37% increase in CL/F at steady
state compared with values obtained in the same volunteers following a single
dose. Evidence gathered from other sources suggests that self-induction by
lamotrigine may not occur when lamotrigine is given as adjunctive therapy in
patients receiving enzyme-inducing drugs such as carbamazepine, phenytoin,
phenobarbital, primidone, or other drugs such as rifampin and the protease
inhibitors lopinavir/ritonavir and atazanavir/ritonavir that induce lamotrigine
glucuronidation [see DRUG INTERACTIONS].
Elimination
The elimination half-life and apparent clearance of
lamotrigine following oral administration of LAMICTAL to adult subjects with
epilepsy and healthy volunteers is summarized in Table 14. Half-life and
apparent oral clearance vary depending on concomitant AEDs.
Drug Interactions
The apparent clearance of lamotrigine is affected by the
coadministration of certain medications [see WARNINGS AND PRECAUTIONS, DRUG
INTERACTIONS].
The net effects of drug interactions with lamotrigine are
summarized in Tables 13 and 15, followed by details of the drug interaction
studies below.
Table 15: Summary of Drug Interactions with
Lamotrigine
Drug |
Drug Plasma Concentration with Adjunctive Lamotriginea |
Lamotrigine Plasma Concentration with Adjunctive Drugsb |
Oral contraceptives (e.g., ethinylestradiol/ levonorgestrel)c |
↔d |
↓ |
Aripiprazole |
Not assessed |
↔e |
Atazanavir/ritonavir |
↔ |
↓ |
Bupropion |
Not assessed |
↔ |
Carbamazepine |
↔ |
↓ |
Carbamazepine epoxideg |
? |
|
Felbamate |
Not assessed |
↔ |
Gabapentin |
Not assessed |
↔ |
Lacosamide |
Not assessed |
↔ |
Levetiracetam |
↔ |
↔ |
Lithium |
↔ |
Not assessed |
Lopinavir/ritonavir |
↔e |
↓ |
Olanzapine |
↔ |
↔e |
Oxcarbazepine |
↔ |
↔ |
10-Monohydroxy oxcarbazepine metaboliteh |
↔ |
|
Perampanel |
Not assessed |
↔e |
Phenobarbital/primidone |
↔ |
↓ |
Phenytoin |
↔ |
↓ |
Pregabalin |
↔ |
↔ |
Rifampin |
Not assessed |
↓ |
Risperidone |
↔ |
Not assessed |
9-Hydroxyri speridonei |
↔ |
|
Topiramate |
↔j |
↔ |
Valproate |
↓ |
↑ |
Valproate + phenytoin and/or carbamazepine |
Not assessed |
↔ |
Zonisamide |
Not assessed |
↔ |
a From adjunctive clinical trials and
volunteer trials.
b Net effects were estimated by comparing the
mean clearance values obtained in adjunctive clinical trials and volunteer
trials.
cThe effect of other hormonal contraceptive
preparations or hormone replacement therapy on  the pharmacokinetics of
lamotrigine has not been systematically evaluated in clinical trials, Â although
the effect may be similar to that seen with the ethinylestradiol/levonorgestrel
 combinations.
d Modest decrease in levonorgestrel.
e Slight decrease, not expected to be clinically meaningful.
f Compared with historical controls.
gNot administered, but an active metabolite of carbamazepine.
h Not administered, but an active metabolite of oxcarbazepine.
i Not administered, but an active metabolite of risperidone.
j Slight increase, not expected to be clinically meaningful. |
Estrogen-Containing Oral Contraceptives
In 16 female volunteers, an oral contraceptive
preparation containing 30 mcg ethinylestradiol and 150 mcg levonorgestrel
increased the apparent clearance of lamotrigine (300 mg/day) by approximately
2-fold with mean decreases in AUC of 52% and in Cmax of 39%. In this study,
trough serum lamotrigine concentrations gradually increased and
wereapproximately 2-fold higher on average at the end of the week of the
inactive hormone preparation compared with trough lamotrigine concentrations at
the end of the active hormone cycle.
Gradual transient increasesinlamotrigine
plasmalevels(approximate 2-fold increase)occurred duringtheweek of
inactivehormone preparation (pill-freeweek) for womennotalso takinga drug that
increased the clearance of lamotrigine (carbamazepine, phenytoin, phenobarbital,
primidone, or other drugs such as rifampin and the protease inhibitors
lopinavir/ritonavir and atazanavir/ritonavir that induce lamotrigine
glucuronidation) [see DRUG INTERACTIONS]. The increase in lamotrigine
plasma levels will be greater if the dose of LAMICTAL is increased in the few
days before or during the pill-free week. Increases in lamotrigine plasma
levels could result in dose-dependent adverse reactions.
In the same study, coadministration of lamotrigine (300
mg/day) in 16 female volunteers did not affect the pharmacokinetics of the
ethinylestradiol component of the oral contraceptive preparation. There were
mean decreases in the AUC and Cmax of the levonorgestrel component of 19% and
12%, respectively. Measurement of serum progesterone indicated that there was
no hormonal evidence of ovulation in any of the 16 volunteers, although
measurement of serum FSH, LH, and estradiol indicated that there was some loss
of suppression of the hypothalamic-pituitary-ovarian axis.
The effects of doses of lamotrigine other than 300 mg/day
have not been systematically evaluated in controlled clinical trials.
The clinical significance of the observed hormonal
changes on ovulatory activity is unknown. However, the possibility of decreased
contraceptive efficacy in some patients cannot be excluded. Therefore, patients
should be instructed to promptly report changes in their menstrual pattern
(e.g., break-through bleeding).
Dosage adjustments may be necessary for women receiving
estrogen-containing oral contraceptive preparations [see DOSAGE AND
ADMINISTRATION].
Other Hormonal Contraceptives Or Hormone Replacement
Therapy
The effect of other hormonal contraceptive preparations
or hormone replacement therapy on the pharmacokinetics of lamotrigine has not
been systematically evaluated. It has been reported that ethinylestradiol, not
progestogens, increased the clearance of lamotrigine up to 2-fold, and the
progestin-only pills had no effect on lamotrigine plasma levels. Therefore,
adjustments to the dosage of LAMICTAL in the presence of progestogens alone
will likely not be needed.
Aripiprazole
In 18 patients with bipolar disorder on a stable regimen
of 100 to 400 mg/day of lamotrigine, the lamotrigine AUC and Cmax were reduced
by approximately 10% in patients who received aripiprazole 10 to 30 mg/day for
7 days, followed by 30 mg/day for an additional 7 days. This reduction in
lamotrigine exposure is not considered clinically meaningful.
Atazanavir/Ritonavir
In a study in healthy volunteers, daily doses of
atazanavir/ritonavir (300 mg/100 mg) reduced the plasma AUC and Cmax of
lamotrigine (single 100-mg dose) by an average of 32% and 6%, respectively, and
shortened the elimination half-lives by 27%. In the presence of
atazanavir/ritonavir (300 mg/100 mg), the metabolite-to-lamotrigine ratio was
increased from 0.45 to 0.71 consistent with induction of glucuronidation. The
pharmacokinetics of atazanavir/ritonavir were similar in the presence of
concomitant lamotrigine to the historical data of the pharmacokinetics in the
absence of lamotrigine.
Bupropion
The pharmacokinetics of a 100-mg single dose of
lamotrigine in healthy volunteers (n = 12) were not changed by coadministration
of bupropion sustained-release formulation (150 mg twice daily) starting 11
days before lamotrigine.
Carbamazepine
Lamotrigine has no appreciable effect on steady-state
carbamazepine plasma concentration. Limited clinical data suggest there is a
higher incidence of dizziness, diplopia, ataxia, and blurred vision in patients
receiving carbamazepine with lamotrigine than in patients receiving other AEDs
with lamotrigine [see ADVERSE REACTIONS]. The mechanism of this
interaction is unclear. The effect of lamotrigine on plasma concentrations of
carbamazepine-epoxide is unclear. In a small subset of patients (n = 7) studied
in a placebo-controlled trial, lamotrigine had no effect on
carbamazepine-epoxide plasma concentrations, but in a small, uncontrolled study
(n = 9), carbamazepine-epoxide levels increased.
The addition of carbamazepine decreases lamotrigine
steady-state concentrations by approximately 40%.
Felbamate
In a trial in 21 healthy volunteers, coadministration of
felbamate (1,200 mg twice daily) with lamotrigine (100 mg twice daily for 10
days) appeared to have no clinically relevant effects on the pharmacokinetics
of lamotrigine.
Folate Inhibitors
Lamotrigine is a weak inhibitor of dihydrofolate
reductase. Prescribers should be aware of this action when prescribing other
medications that inhibit folate metabolism.
Lacosamide Plasma concentrations of lamotrigine were not
affected by concomitant lacosamide (200, 400, or 600 mg/day) in
placebo-controlled clinical trials in patients with partial-onset seizures.
Gabapentin
Based on a retrospective analysis of plasma levels in 34
subjects who received lamotrigine both with and without gabapentin, gabapentin
does not appear to change the apparent clearance of lamotrigine.
Levetiracetam
Potential drug interactions between levetiracetam and
lamotrigine were assessed by evaluating serum concentrations of both agents
during placebo-controlled clinical trials. These data indicate that lamotrigine
does not influence the pharmacokinetics of levetiracetam and that levetiracetam
does not influence the pharmacokinetics of lamotrigine.
Lithium
The pharmacokinetics of lithium were not altered in
healthy subjects (n = 20) by coadministration of lamotrigine (100 mg/day) for 6
days.
Lopinavir/Ritonavir
The addition of lopinavir (400 mg twice daily)/ritonavir
(100 mg twice daily) decreased the AUC, Cmax, and elimination half-life of
lamotrigine by approximately 50% to 55.4% in 18 healthy subjects. The
pharmacokinetics of lopinavir/ritonavir were similar with concomitant
lamotrigine, compared with that in historical controls.
Olanzapine
The AUC and Cmax of olanzapine were similar following the
addition of olanzapine (15 mg once daily) to lamotrigine (200 mg once daily) in
healthy male volunteers (n = 16) compared with the AUC and Cmax in healthy male
volunteers receiving olanzapine alone (n = 16).
In the same trial, the AUC and Cmax of lamotrigine were
reduced on average by 24% and 20%, respectively, following the addition of
olanzapine to lamotrigine in healthy male volunteers compared with those
receiving lamotrigine alone. This reduction in lamotrigine plasma
concentrations is not expected to be clinically meaningful.
Oxcarbazepine
The AUC and Cmax of oxcarbazepine and its active 10-monohydroxy
oxcarbazepine metabolite were not significantly different following the
addition of oxcarbazepine (600 mg twice daily) to lamotrigine (200 mg once
daily) in healthy male volunteers (n = 13) compared with healthy male
volunteers receiving oxcarbazepine alone (n = 13).
In the same trial, the AUC and Cmax of lamotrigine were
similar following the addition of oxcarbazepine (600 mg twice daily) to
lamotrigine in healthy male volunteers compared with those receiving
lamotrigine alone. Limited clinical data suggest a higher incidence of
headache, dizziness, nausea, and somnolence with coadministration of
lamotrigine and oxcarbazepine compared with lamotrigine alone or oxcarbazepine
alone.
Perampanel
In a pooled analysis of data from 3 placebo-controlled
clinical trials investigating adjunctive perampanel in patients with
partial-onset and primary generalized tonic-clonic seizures, the highest
perampanel dose evaluated (12 mg/day) increased lamotrigine clearance by
<10%. An effect of this magnitude is not considered to be clinically
relevant.
Phenobarbital, Primidone
The addition of phenobarbital or primidone decreases
lamotrigine steady-state concentrations by approximately 40%.
Phenytoin
Lamotrigine has no appreciable effect on steady-state
phenytoin plasma concentrations in patients with epilepsy. The addition of
phenytoin decreases lamotrigine steady-state concentrations by approximately
40%.
Pregabalin
Steady-state trough plasma concentrations of lamotrigine
were not affected by concomitant pregabalin (200 mg 3 times daily)
administration. There are no pharmacokinetic interactions between lamotrigine
and pregabalin.
Rifampin
In 10 male volunteers, rifampin (600 mg/day for 5 days)
significantly increased the apparent clearance of a single 25-mg dose of
lamotrigine by approximately 2-fold (AUC decreased by approximately 40%).
Risperidone
In a 14 healthy volunteers study, multiple oral doses of
lamotrigine 400 mg daily had no clinically significant effect on the single-dose
pharmacokinetics of risperidone 2 mg and its active metabolite 9-OH
risperidone. Following the coadministration of risperidone 2 mg with
lamotrigine, 12 of the 14 volunteers reported somnolence compared with 1 out of
20 when risperidone was given alone, and none when lamotrigine was administered
alone.
Topiramate
Topiramate resulted in no change in plasma concentrations
of lamotrigine. Administration of lamotrigine resulted in a 15% increase in
topiramate concentrations.
Valproate
When lamotrigine was administered to healthy volunteers
(n = 18) receiving valproate, the trough steady-state valproate plasma
concentrations decreased by an average of 25% over a 3-week period, and then
stabilized. However, adding lamotrigine to the existing therapy did not cause a
change in valproate plasma concentrations in either adult or pediatric patients
in controlled clinical trials.
The addition of valproate increased lamotrigine
steady-state concentrations in normal volunteers by slightly more than 2-fold.
In 1 trial, maximal inhibition of lamotrigine clearance was reached at
valproate doses between 250 and 500 mg/day and did not increase as the
valproate dose was further increased.
Zonisamide
In a study in 18 patients with epilepsy, coadministration
of zonisamide (200 to 400 mg/day) with lamotrigine (150 to 500 mg/day for 35
days) had no significant effect on the pharmacokinetics of lamotrigine.
Known Inducers Or Inhibitors Of Glucuronidation
Drugs other than those listed above have not been
systematically evaluated in combination with lamotrigine. Since lamotrigine is
metabolized predominately by glucuronic acid conjugation, drugs that are known
to induce or inhibit glucuronidation may affect the apparent clearance of
lamotrigine and doses of lamotrigine may require adjustment based on clinical
response.
Other
In vitro assessment of the inhibitory effect of
lamotrigine at OCT2 demonstrate that lamotrigine, but not the N(2)-glucuronide
metabolite, is an inhibitor of OCT2 at potentially clinically relevant concentrations,
with IC50 value of 53.8 μM [see DRUG INTERACTIONS].
Results of in vitro experiments suggest that clearance of
lamotrigine is unlikely to be reduced by concomitant administration of
amitriptyline, clonazepam, clozapine, fluoxetine, haloperidol, lorazepam,
phenelzine, sertraline, or trazodone.
Results of in vitro experiments suggest that lamotrigine
does not reduce the clearance of drugs eliminated predominantly by CYP2D6.
Specific Populations
Patients With Renal Impairment
Twelve volunteers with chronic renal failure (mean
creatinine clearance: 13 mL/min, range: 6 to 23) and another 6 individuals
undergoing hemodialysis were each given a single 100-mg dose of lamotrigine.
The mean plasma half-lives determined in the study were 42.9 hours (chronic
renal failure), 13.0 hours (during hemodialysis), and 57.4 hours (between
hemodialysis) compared with 26.2 hours in healthy volunteers. On average,
approximately 20% (range: 5.6 to 35.1) of the amount of lamotrigine present in
the body was eliminated by hemodialysis during a 4-hour session [see DOSAGE
AND ADMINISTRATION].
Patients With Hepatic Impairment
The pharmacokinetics of lamotrigine following a single
100mg dose of lamotrigine were evaluated in 24 subjects with mild, moderate, and
severe hepatic impairment (Child-Pugh classification system) and compared with
12 subjects without hepatic impairment. The subjects with severe hepatic
impairment were without ascites (n = 2) or with ascites (n = 5). The mean
apparent clearances of lamotrigine in subjects with mild (n = 12), moderate (n
= 5), severe without ascites (n = 2), and severe with ascites (n = 5) liver
impairment were 0.30 ± 0.09, 0.24 ± 0.1, 0.21 ± 0.04, and 0.15 ± 0.09
mL/min/kg, respectively, as compared with 0.37 ± 0.1 mL/min/kg in the healthy
controls. Mean half-lives of lamotrigine in subjects with mild, moderate,
severe without ascites, and severe with ascites hepatic impairment were 46 ±
20, 72 ± 44, 67 ± 11, and 100 ± 48 hours, respectively, as compared with 33 ± 7
hours in healthy controls [see DOSAGE AND ADMINISTRATION].
Pediatric Patients
The pharmacokinetics of lamotrigine following a single
2-mg/kg dose were evaluated in 2 studies in pediatric subjects (n = 29 for
subjects aged 10 months to 5.9 years and n = 26 for subjects aged 5 to 11
years). Forty-three subjects received concomitant therapy with other AEDs and
12 subjects received lamotrigine as monotherapy. Lamotrigine pharmacokinetic
parameters for pediatric patients are summarized in Table 16.
Population pharmacokinetic analyses involving subjects
aged 2 to 18 years demonstrated that lamotrigine clearance was influenced
predominantly by total body weight and concurrent AED therapy. The oral
clearance of lamotrigine was higher, on a body weight basis, in pediatric
patients than in adults. Weight-normalized lamotrigine clearance was higher in
those subjects weighing <30 kg compared with those weighing >30 kg.
Accordingly, patients weighing <30 kg may need an increase of as much as 50%
in maintenance doses, based on clinical response, as compared with subjects
weighing more than 30 kg being administered the same AEDs [see DOSAGE AND
ADMINISTRATION]. These analyses also revealed that, after accounting for
body weight, lamotrigine clearance was not significantly influenced by age.
Thus, the same weight-adjusted doses should be administered to children
irrespective of differences in age. Concomitant AEDs which influence
lamotrigine clearance in adults were found to have similar effects in children.
Table 16: Mean Pharmacokinetic Parameters in Pediatric
Subjects with Epilepsy
Pediatric Study Population |
Number of Subjects |
Tmax (h) |
t½ (h) |
CL/F (mL/min/kg) |
Ages 10 months-5.3 years |
Subjects taking carbamazepine, phenytoin, phenobarbital, or primidonea |
10 |
3.0 (1.0-5.9) |
7.7 (5.7-11.4) |
3.62 (2.44-5.28) |
Subjects taking antiepileptic drugs with no known effect on the apparent clearance of lamotrigine |
7 |
5.2 (2.9-6.1) |
19.0 (12.9-27.1) |
1.2 (0.75-2.42) |
Subjects taking valproate only |
8 |
2.9 (1.0-6.0) |
44.9 (29.5-52.5) |
0.47 (0.23-0.77) |
Ages 5-11 years |
Subjects taking carbamazepine, phenytoin, phenobarbital, or primidonea |
7 |
1.6 (1.0-3.0) |
7.0 (3.8-9.8) |
2.54 (1.35-5.58) |
Subjects taking carbamazepine, phenytoin, phenobarbital, or primidonea plus valproate b |
8 |
3.3 (1.0-6.4) |
19.1 (7.0-31.2) |
0.89 (0.39-1.93) |
Subjects taking valproate only |
3 |
4.5 (3.0-6.0) |
65.8 (50.7-73.7) |
0.24 (0.21-0.26) |
Ages 13-18 years |
Subjects taking carbamazepine, phenytoin, phenobarbital, or primidonea |
11 |
-c |
-c |
1.3 |
Subjects taking carbamazepine, phenytoin, phenobarbital, or primidonea plus valproate |
8 |
-c |
-c |
0.5 |
Subjects taking valproate only |
4 |
-c |
-c |
0.3 |
a Carbamazepine, phenytoin, phenobarbital, and
primidone have been shown to increase the apparent clearance of lamotrigine.
Estrogen-containing oral contraceptives, rifampin, and the protease inhibitors
lopinavir/ritonavir and atazanavir/ritonavir have also been shown to increase
the apparent clearance of lamotrigine [see DRUG INTERACTIONS].
b Two subjects were included in the calculation for mean Tmax.
c Parameter not estimated. |
Geriatric Patients
The pharmacokinetics of lamotrigine following a single
150-mg dose of lamotrigine were evaluated in 12 elderly volunteers between the
ages of 65 and 76 years (mean creatinine clearance = 61 mL/min, range: 33 to
108 mL/min). The mean half-life of lamotrigine in these subjects was 31.2 hours
(range: 24.5 to 43.4 hours), and the mean clearance was 0.40 mL/min/kg (range:
0.26 to 0.48 mL/min/kg).
Male And Female Patients
The clearance of lamotrigine is not affected by gender.
However, during dose escalation of lamotrigine in 1 clinical trial in patients
with epilepsy on a stable dose of valproate (n = 77), mean trough lamotrigine
concentrations unadjusted for weight were 24% to 45% higher (0.3 to 1.7 mcg/mL)
in females than in males.
Racial Or Ethnic Groups
The apparent oral clearance of lamotrigine was 25% lower
in non-Caucasians than Caucasians.
Clinical Studies
Epilepsy
Monotherapy With LAMICTAL In Adults With Partial-Onset Seizures
Already Receiving Treatment With Carbamazepine, Phenytoin, Phenobarbital, Or Primidone
As The Single Antiepileptic Drug
The effectiveness of monotherapy with LAMICTAL was
established in a multicenter double-blind clinical trial enrolling 156 adult
outpatients with partial-onset seizures. The patients experienced at least 4
simple partial-onset, complex partial-onset, and/or secondarily generalized
seizures during each of 2 consecutive 4-week periods while receiving
carbamazepine or phenytoin monotherapy during baseline. LAMICTAL (target dose
of 500 mg/day) or valproate (1,000 mg/day) was added to either carbamazepine or
phenytoin monotherapy over a 4-week period. Patients were then converted to
monotherapy with LAMICTAL or valproate during the next 4 weeks, then continued
on monotherapy for an additional 12-week period.
Trial endpoints were completion of all weeks of trial
treatment or meeting an escape criterion. Criteria for escape relative to
baseline were: (1) doubling of average monthly seizure count, (2) doubling of
highest consecutive 2-day seizure frequency, (3) emergence of a new seizure type
(defined as a seizure that did not occur during the 8-week baseline) that is
more severe than seizure types that occur during study treatment, or (4)
clinically significant prolongation of generalized tonic-clonic seizures. The
primary efficacy variable was the proportion of patients in each treatment
group who met escape criteria.
The percentages of patients who met escape criteria were
42% (32/76) in the group receiving LAMICTAL and 69% (55/80) in the valproate
group. The difference in the percentage of patients meeting escape criteria was
statistically significant (P = 0.0012) in favor of LAMICTAL. No differences in
efficacy based on age, sex, or race were detected.
Patients in the control group were intentionally treated
with a relatively low dose of valproate; as such, the sole objective of this
trial was to demonstrate the effectiveness and safety of monotherapy with LAMICTAL,
and cannot be interpreted to imply the superiority of LAMICTAL to an adequate
dose of valproate.
Adjunctive Therapy With LAMICTAL In Adults With Partial-Onset
Seizures
The effectiveness of LAMICTAL as adjunctive therapy
(added to other AEDs) was initially established in 3 pivotal, multicenter,
placebo-controlled, double-blind clinical trials in 355 adults with refractory
partial-onset seizures. The patients had a history of at least 4 partial-onset
seizures per month in spite of receiving 1 or more AEDs at therapeutic
concentrations and in 2 of the trials were observed on their established AED
regimen during baselines that varied between 8 to 12 weeks. In the third trial,
patients were not observed in a prospective baseline. In patients continuing to
have at least 4 seizures per month during the baseline, LAMICTAL or placebo was
then added to the existing therapy. In all 3 trials, change from baseline in
seizure frequency was the primary measure of effectiveness. The results given
belowarefor allpartial-onsetseizures in the intent-to-treat population (all
patients who received at least 1 dose of treatment) in each trial, unless
otherwise indicated. The median seizure frequency at baseline was 3 per week
while the mean at baseline was 6.6 per week for all patients enrolled in
efficacy trials.
One trial (n = 216) was a double-blind,
placebo-controlled, parallel trial consisting of a 24-week treatment period.
Patients could not be on more than 2 other anticonvulsants and valproate was
not allowed. Patients were randomized to receive placebo, a target dose of 300
mg/day of LAMICTAL, or a target dose of 500 mg/day of LAMICTAL. The median
reductions in the frequency of all partial-onset seizures relative to baseline
were 8% in patients receiving placebo, 20% in patients receiving 300 mg/day of
LAMICTAL, and 36% in patients receiving 500 mg/day of LAMICTAL. The seizure
frequency reduction was statistically significant in the 500-mg/day group
compared with the placebo group, but not in the 300-mg/day group.
A second trial (n = 98) was a double-blind,
placebo-controlled, randomized, crossover trial consisting of two 14-week
treatment periods (the last 2 weeks of which consisted of dose tapering)
separated by a 4-week washout period. Patients could not be on more than 2
other anticonvulsants and valproate was not allowed. The target dose of
LAMICTAL was 400 mg/day.
When the first 12 weeks of the treatment periods were
analyzed, the median change in seizure frequency was a 25% reduction on
LAMICTAL compared with placebo (P<0.001).
The third trial (n = 41) was a double-blind,
placebo-controlled, crossover trial consisting of two 12-week treatment periods
separated by a 4-week washout period. Patients could not be on more than 2
other anticonvulsants. Thirteen patients were on concomitant valproate; these
patients received 150 mg/day of LAMICTAL. The 28 other patients had a target
dose of 300 mg/day of LAMICTAL. The median change in seizure frequency was a
26% reduction on LAMICTAL compared with placebo (P<0.01).
No differences in efficacy based on age, sex, or race, as
measured by change in seizure frequency, were detected.
Adjunctive Therapy With LAMICTAL In Pediatric Patients With
Partial-Onset Seizures
The effectiveness of LAMICTAL as adjunctive therapy in
pediatric patients with partial-onset seizures was established in a
multicenter, double-blind, placebo-controlled trial in 199 patients aged 2 to
16 years (n = 98 on LAMICTAL, n = 101 on placebo). Following an 8-week baseline
phase, patients were randomized to 18 weeks of treatment with LAMICTAL or
placebo added to their current AED regimen of up to 2 drugs. Patients were
dosed based on body weight and valproate use. Target doses were designed to
approximate 5 mg/kg/day for patients taking valproate (maximum dose: 250
mg/day) and 15 mg/kg/day for the patients not taking valproate (maximum dose:
750 mg/day). The primary efficacy endpoint was percentage change from baseline
in all partial-onset seizures. For the intent-to-treat population, the median
reduction of all partial-onset seizures was 36% in patients treated with
LAMICTAL and 7% on placebo, a difference that was statistically significant (P<0.01).
Adjunctive Therapy With LAMICTAL In Pediatric And Adult
Patients With Lennox-Gastaut Syndrome
The effectiveness of LAMICTAL as adjunctive therapy in
patients with Lennox-Gastaut syndrome was established in a multicenter,
double-blind, placebo-controlled trial in 169 patients aged 3 to 25 years (n =
79 on LAMICTAL, n = 90 on placebo). Following a 4-week, single-blind, placebo
phase, patients were randomized to 16 weeks of treatment with LAMICTAL or
placebo added to their current AED regimen of up to 3 drugs. Patients were
dosed on a fixed-dose regimen based on body weight and valproate use. Target
doses were designed to approximate 5 mg/kg/day for patients taking valproate
(maximum dose: 200 mg/day) and 15 mg/kg/day for patients not taking valproate
(maximum dose: 400 mg/day). The primary efficacy endpoint was percentage change
from baseline in major motor seizures (atonic, tonic, major myoclonic, and
tonic-clonic seizures). For the intent-to-treat population, the median
reduction of major motor seizures was 32% in patients treated with LAMICTAL and
9% on placebo, a difference that was statistically significant (P<0.05).
Drop attacks were significantly reduced by LAMICTAL (34%) compared with placebo
(9%), as were tonic-clonic seizures (36% reduction versus 10% increase for
LAMICTAL and placebo, respectively).
Adjunctive Therapy With LAMICTAL In Pediatric And Adult
Patients With Primary Generalized Tonic-Clonic Seizures
The effectiveness of LAMICTAL as adjunctive therapy in
patients with PGTC seizures was established in a multicenter, double-blind,
placebo-controlled trial in 117 pediatric and adult patients aged 2 years and
older (n = 58 on LAMICTAL, n = 59 on placebo). Patients with at least 3 PGTC
seizures during an 8-week baseline phase were randomized to 19 to 24 weeks of
treatment with LAMICTAL or placebo added to their current AED regimen of up to
2 drugs. Patients were dosed on a fixed-dose regimen, with target doses ranging
from 3 to 12 mg/kg/day for pediatric patients and from 200 to 400 mg/day for
adult patients based on concomitant AEDs.
The primary efficacy endpoint was percentage change from
baseline in PGTC seizures. For the intent-to-treat population, the median
percent reduction in PGTC seizures was 66% in patients treated with LAMICTAL
and 34% on placebo, a difference that was statistically significant (P =
0.006).
Bipolar Disorder
Adults
The effectiveness of LAMICTAL in the maintenance
treatment of bipolar I disorder was established in 2 multicenter, double-blind,
placebo-controlled trials in adult patients (aged 18 to 82 years) who met DSM-IV
criteria for bipolar I disorder. Trial 1 enrolled patients with a current or
recent (within 60 days) depressive episode as defined by DSM-IV and Trial 2
included patients with a current or recent (within 60 days) episode of mania or
hypomania as defined by DSM-IV. Both trials included a cohort of patients (30%
of 404 subjects in Trial 1 and 28% of 171 patients in Trial 2) with rapid
cycling bipolar disorder (4 to 6 episodes per year).
In both trials, patients were titrated to a target dose
of 200 mg of LAMICTAL as add-on therapy or as monotherapy with gradual
withdrawal of any psychotropic medications during an 8-to 16-week open-label
period. Overall 81% of 1,305 patients participating in the open-label period
were receiving 1 or more other psychotropic medications, including
benzodiazepines, selective serotonin reuptake inhibitors (SSRIs), atypical
antipsychotics (including olanzapine), valproate, or lithium, during titration
of LAMICTAL. Patients with a CGI-severity score of 3 or less maintained for at
least 4 continuous weeks, including at least the final week on monotherapy with
LAMICTAL, were randomized to a placebo-controlled double-blind treatment period
for up to 18 months. The primary endpoint was TIME (time to intervention for a
mood episode or one that was emerging, time to discontinuation for either an
adverse event that was judged to be related to bipolar disorder, or for lack of
efficacy). The mood episode could be depression, mania, hypomania, or a mixed
episode.
In Trial 1, patients received double-blind monotherapy
with LAMICTAL 50 mg/day (n = 50), LAMICTAL 200 mg/day (n = 124), LAMICTAL 400
mg/day (n = 47), or placebo (n = 121). LAMICTAL (200-and 400-mg/day treatment
groups combined) was superior to placebo in delaying the time to occurrence of
a mood episode (Figure 1). Separate analyses of the 200-and 400-mg/day dose
groups revealed no added benefit from the higher dose.
In Trial 2, patients received double-blind monotherapy
with LAMICTAL (100 to 400 mg/day, n = 59), or placebo (n = 70). LAMICTAL was
superior to placebo in delaying time to occurrence of a mood episode (Figure
2). The mean dose of LAMICTAL was about 211 mg/day.
Although these trials were not designed to separately
evaluate time to the occurrence of depression or mania, a combined analysis for
the 2 trials revealed a statistically significant benefitfor LAMICTAL over
placebo in delayingthetimeto occurrence ofbothdepressionand mania, although the
finding was more robust for depression.
Figure 1: Kaplan-Meier Estimation of Cumulative
Proportion of Patients with Mood Episode (Trial 1)
Figure 2: Kaplan-Meier Estimation of Cumulative
Proportion of Patients with Mood Episode (Trial 2)