You are on page 1of 16

Handbook of Clinical Neurology, Vol.

119 (3rd series)


Neurologic Aspects of Systemic Disease Part I
Jose Biller and Jose M. Ferro, Editors
© 2014 Elsevier B.V. All rights reserved

Chapter 27

Use of antiepileptic drugs in hepatic and renal disease


JORGE J. ASCONAPÉ*
Department of Neurology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA

INTRODUCTION Dialysis can further modify these pharmacokinetic


parameters or result in significant removal of the drug.
The occurrence of seizures in the presence of hepatic or
The dialyzability of a drug depends on several factors,
liver disease is not uncommon in clinical practice. Patients
including molecular weight, protein binding, plasma
with epilepsy are not immune to hepatic or renal disease,
concentration, blood flow, hematocrit, and the effi-
and patients with hepatic or renal failure often suffer
ciency of the dialyzer (Israni et al., 2006). Dose supple-
from acute reactive seizures secondary to metabolic or
mentation at the time of dialysis may be required for
electrolyte disturbances or other associated comorbidities.
highly extractable drugs.
Since the liver and kidney are the main organs involved in
Most antiepileptic drugs undergo some degree of
the elimination of most drugs (Table 27.1), their dysfunc-
biotransformation, which occurs almost exclusively in
tion can have important effects on the disposition of the
the microsomal mixed-function oxidase system. Phase I
antiepileptic drugs. A clear understanding of these phar-
reactions, including oxidation, reduction, or hydroxyl-
macokinetic effects is essential when selecting a drug in
ation, transform the lipophilic active drug into a more
patients with liver or renal disease. In the past two
polar, water-soluble metabolite. These metabolites may
decades, with the introduction of newer antiepileptic
undergo a subsequent phase II reaction by combining with
drugs, many with a better pharmacokinetic profile than
endogenous substrates, such as glucuronic acid, to form a
the classic drugs, the number of options available has
more polar compound. Liver disease can delay the elimi-
greatly expanded, facilitating the management of these
nation rate of the drug from plasma, leading to a prolon-
patients. The classic antiepileptic drugs, such as phenyt-
gation of the half-life and drug accumulation (Ahmed and
oin, carbamazepine, or valproate, are mostly eliminated
Siddiqui, 2006). Drug metabolism is influenced by hepatic
via hepatic metabolism, therefore, their use is problematic
blood flow, protein binding, degree of drug uptake by the
in patients with liver failure. With most of the newer anti-
hepatocyte, functional capacity of the hepatocyte, and
epileptic drugs, the kidneys play a much more important
patency of the hepatobiliary system. Because of the large
role in their elimination, offering a better alternative to
hepatic reserve capacity, liver disease must be extensive
patients with liver disease.
before drug metabolism is impaired.
Renal disease can prolong the elimination of the par-
ent drug or an active metabolite and lead to accumula-
tion and clinical toxicity (Asconapé and Penry, 1982). INDIVIDUAL ANTIEPILEPTIC DRUGS
Lower doses and longer interdose intervals may be nec-
Phenytoin
essary in these cases. Renal disease can also affect the
protein binding, distribution and metabolism of a drug. Phenytoin (5,5-diphenylhydantoin) is a weak organic acid
The protein binding of anionic acidic drugs, such as phe- with a pKa of 8.31. It is completely but irregularly
nytoin and valproate, can be reduced significantly by absorbed after oral administration, with peak serum
renal failure causing difficulties in the interpretation levels reached 4–8 hours following a single oral dose.
of total serum concentrations commonly used in clinical Phenytoin is 90% protein bound, almost exclusively to
practice. Measuring the free fraction concentrations is the albumin, with a 10% free fraction responsible for
more accurate in these cases. the pharmacologic action. The elimination of phenytoin

*Correspondence to: Jorge J. Asconapé, M.D., Department of Neurology, Maguire Center, Suite 2700, Loyola University Chicago,
2160 South First Avenue, Maywood, Illinois 60153, USA. Tel: þ1-708-216-3407, Fax: þ1-708-216-8028, E-mail: jasconape@lumc.edu
418 J.J. ASCONAPÉ
Table 27.1
Elimination of antiepileptic drugs

Hepatic metabolism Renal elimination


(proportion excreted
Drug CYP 450 Isoenzyme UDPGT Other pathways unchanged by the kidney)

Carbamazepine 85% Major: CYP 3A4 15% Negligible <1%


Minor: CYP 1A2/2C8
Diazepam 80% CYP 2C19, CYP 3A4 Negligible Negligible <5%
Eslicarbazepine – 41% Negligible 52%
Ethosuximide 65% Major: CYP 3A4 Negligible Negligible 20–25%
Minor: CYP 2B/2C/2E1
Ezogabine/ None – Extensive Acetylation 36%
retigabine
Felbamate <20% CYP 3A4/2E1 10% 25% esterase 45–55%
Fosphenytoin – – Negligible Metabolized to phenytoin <5%
by phosphatases (blood,
liver, kidney)
Gabapentin None – None None >95%
Lacosamide 50% CYP 2C19 None None 40%
Lamotrigine 1A4 Negligible 70–80% Negligible 10%
Levetiracetam Negligible – None 24% hydrolysis 66%
(extrahepatic)
Lorazepam Negligible 90% Negligible <1%
Midazolam 85% CYP 3A4 Negligible Negligible <1%
Oxcarbazepine* 4% 49% Cytosol arylketone 27%
reductase (parent drug)
Phenobarbital 20–35% CYP 2C9/19 None Negligible 20–25%
Phenytoin 90% Major: CYP 2C9 Negligible Negligible <5%
Minor: CYP 2C19
Pregabalin Negligible – Negligible Negligible >95%
Primidone 40–60% ? Some Negligible 40–60%
Propofol Negligible CYP 1A9 50% Sulfation <1%
Rufinamide None – 11% 70% carboxylesterase- <2%
mediated hydrolysis
Tiagabine 90% CYP 3A4 Negligible Negligible <2%
Topiramate 20–40% Unknown Unknown 60–70%
Valproate <20% CYP 1A3, 2B7 30–50% 40% b-oxidation <3%
(mitochondrial hepatic
metabolism)
Vigabatrin Negligible – None Negligible 80–95%
Zonisamide 50% CYP 3A4 None 15% N-acetylation 35%
(hepatic)

*Refers to the monohydroxy derivative (MHD).


CYP, cytochrome P450; UDPGT, uridinine diphosphate-glucuronosyltransferase.

is almost exclusively by hepatic biotransformation, an inactive metabolite (Bochner et al., 1973). Approxi-
with < 5% of a given dose eliminated unchanged in mately 96% of plasma pHPPH is conjugated to glucuro-
the urine (Butler, 1957). Phenytoin is metabolized by nides, with 60–80% of a single dose of PHT excreted as
the cytochrome P450 system, with the CYP2C9 being pHPPH glucuronide. Phenytoin undergoes glomerular
the primary isoenzyme, and CYP2C19 playing a more filtration and subsequent tubular reabsorption; pHPPH,
important role when phenytoin serum concentration a more polar metabolite, is handled almost exclusively by
exceeds 18 mg/mL. The main metabolic pathway of phe- glomerular filtration (Borga et al., 1979). One of the
nytoin is parahydroxylation, resulting in the formation major limitations with the clinical use of phenytoin is
of 5-(p-hydroxyphenyl)-5-phenylhydantoin (pHPPH), its capacity of saturating the metabolic pathways at
USE OF ANTIEPILEPTIC DRUGS IN HEPATIC AND RENAL DISEASE 419
serum concentration within the therapeutic range of 10– fraction between 1 and 2 mg/mL, the usual therapeutic
20 mg/mL. The elimination of phenytoin follows a dose- range of 10–20 mg/mL will be reduced to about
dependent, nonlinear kinetics (Michaelis–Menten) 5–10 mg/mL in patients with end-stage renal disease.
model, which can result in a disproportionate increase Equations used to normalize total phenytoin concentra-
in the serum concentration with relatively small incre- tions in the presence of renal failure or hypoalbumine-
ments in the dose. mia have been found to be inaccurate in a significant
Plasma of patients with chronic renal failure has a proportion of patients, resulting in either over- or under-
decreased binding capacity for phenytoin (Odar- prediction of the normalized phenytoin concentrations
Cederl€ of et al., 1970; Reidenberg et al., 1971; Hooper (Mauro et al., 1989). The use of free phenytoin levels
et al., 1974; Reidenberg and Drayer, 1978; Vanholder in this setting is highly recommended. Salivary
et al., 1988). The percentage of unbound fraction can levels have also been proven to adequate for monitoring
be as high as 30%, and the degree of impairment of bind- purposes (McAuliffe et al., 1977).
ing capacity correlates with the level of blood urea nitro- The half-life of phenytoin is decreased to approxi-
gen, serum creatinine, creatinine clearance, and the mately 8 hours in patients with chronic renal failure
degree of physical disability (Reidenberg et al., 1971; (Letter et al., 1971; Odar-Cederl€of and Borga, 1974).
Olsen et al., 1975). In a study of patients with acute renal A single daily dose of phenytoin, therefore, should be
failure from different etiologies, the free fraction of discouraged in uremic patients. The elimination of phe-
phenytoin was found to range from 14% to 45%; how- nytoin is essentially liver-dependent, with < 5% of the
ever, no clear correlation with the above-mentioned drug eliminated unchanged in the urine. Thus, signifi-
markers of renal failure was found (Tiula et al., 1987). cant accumulation of phenytoin even in the presence
In this study, the serum albumin concentration was the of end-stage renal disease does not occur, and dose
best predictor of the free phenytoin concentrations adjustments are not necessary. The main metabolite of
(Tiula et al., 1987). The mechanism of the decreased phenytoin, pHPPH glucuronide, undergoes renal excre-
binding capacity in uremia is not entirely clear, with tion and significantly accumulates in renal failure,
possible explanations including: accumulation of com- whereas the unconjugated pHPPH does not accumulate
petitive or noncompetitive inhibitors in the serum, an (Letter et al., 1971; Borga et al., 1979). However, pHPPH
alteration in the structure or conformation of the albu- is an inactive metabolite, and its accumulation appears to
min molecule, and hypoalbuminemia. Hippuric acid, cer- be of no clinical consequence.
tain organic acids, and peptides include ligands that have Given its high protein binding and poor water solubil-
been shown to displace phenytoin from the albumin mol- ity, phenytoin is poorly dialyzable, and in most cases
ecule in renal failure (Depner and Gulyassy, 1980; supplemental doses after dialysis are not necessary.
Kinniburgh and Boyd, 1981; Gulyassy et al., 1983). An early study indicated that only 2–4% of an intrave-
Hypoalbuminemic patients have been shown to have a nous dose was recovered in the dialysate of seven uremic
greater unbound fraction of phenytoin and an increased patients, with the dialyzer extracting about 4.5% of the
risk of clinical toxicity (Boston Collaborative Drug phenytoin presented to it (Martin et al., 1977). However,
Surveillance Program, 1973); a significant correlation with the use of newer high-efficiency dialyzers, signifi-
between albumin levels and the degree of impairment cant drops in phenytoin serum concentrations, resulting
of the binding capacity has been demonstrated in uremia in breakthrough seizures, have been reported (Frenchie
(Hooper et al., 1974; Reidenberg and Drayer, 1978) and in and Bastani, 1998). Hemodialysis can affect the protein
the nephrotic syndrome with normal renal function binding of phenytoin, but reports have been conflicting.
(Gugler et al., 1975; Gugler and Azarnoff, 1976). How- A decrease in the binding capacity of phenytoin follow-
ever, decreased plasma protein binding of PHT is seen ing hemodialysis has been observed, probably due to the
in uremic patients with normal albumin levels, indicating displacement of phenytoin from the albumin molecule
that hypoalbuminemia is probably only a contributing by elevated levels of nonesterified fatty acids resulting
factor. from the use of heparin (Dromgoole, 1973; Adler
The decreased phenytoin binding capacity results in et al., 1975). However, an increase in the binding capacity
an increase in the apparent volume of distribution (Vd) of phenytoin during dialysis has also been reported
of the drug, with lower total serum concentrations for (Steele et al., 1979).
a given dose (Odar-Cederl€ of and Borga, 1974; Van Plasma from patients with liver failure has a reduced
Peer et al., 1981). The unbound fraction concentration, binding capacity for phenytoin (Reidenberg and
however, remains essentially unchanged, and the ratio Affrime, 1973; Hooper et al., 1974; Olsen et al., 1975).
of free to total drug is increased. Therefore, caution Although the exact mechanism remains unclear, the
should be used when interpreting total serum phenytoin degree of binding decrease has a good correlation with
concentration in uremic patients, since, for a free albumin concentration in plasma (Lunde et al., 1970;
420 J.J. ASCONAPÉ
Rane et al., 1971; Hooper et al., 1974; Blaschke et al., concentrations. Elimination half-life is between 10 and
1975) and also with levels of total plasma bilirubin 16 hours in monotherapy, but is reduced to 6–8 hours with
(Rane et al., 1971). The correlation was even higher when the concomitant use of enzyme-inducing drugs.
both variables were considered together (Lunde et al., Hepatic metabolism is the main route of elimination, with
1970; Rane et al., 1971). The degree of impairment of only 1–3% of a given dose excreted unchanged in the
the phenytoin-binding capacity, however, is not a severe urine. Metabolic pathways include glucuronidation,
as that observed in renal disease, and is usually of no b-oxidation, and !-oxidation. Some of the metabolites
clinical significance. Adjustments in the therapeutic of valproic acid are biologically active. Valproic acid
range of the total phenytoin serum concentration are has no enzyme-inducing properties, but inhibits the
usually not needed as suggested by studies in alcoholic metabolism of lamotrigine, phenobarbital, and the 10-11-
cirrhosis (Affrime and Reidenberg, 1975) and acute viral carbamazepine epoxide. Total plasma concentration
hepatitis (Blaschke et al., 1975). In hyperbilirubinemic between 50 and 100 mg/mL or free plasma concentrations
neonates, however, the reduction in phenytoin binding of 5–10 mg/mL are considered optimal in clinical practice.
may be substantial. In this situation, if the dose of phe- The effects of renal disease on the pharmacokinetics
nytoin is estimated on the basis of bodyweight, a free of valproic acid are very similar to the ones described for
fraction within the toxic range can be achieved (Rane phenytoin, reflecting the fact that both drugs are weak
et al., 1971). organic acids with very high (90%) protein binding under
The effects of liver failure on the elimination of phe- usual clinical conditions. The plasma protein binding is
nytoin have not been adequately studied. Kutt et al. decreased in patients with uremia. The degree of impair-
reported four patients with liver disease receiving phe- ment was found to correlate well with serum creatinine,
nytoin and phenobarbital who developed clinical toxicity blood urea nitrogen, uric acid concentrations, and
as hepatic impairment increased (Kutt et al., 1964). creatinine clearance, and was independent of the albu-
Accumulation of the parent drugs was observed, as well min and total protein concentrations in plasma. The pres-
as reduced excretion of the corresponding metabolites in ence of hypoalbuminemia, acting as a contributing
the urine. Blaschke et al. found no changes in the half- factor, further impairs the plasma binding capacity,
life or plasma clearance of phenytoin in patients with and may be an important factor in patients with the
acute viral hepatitis (Blaschke et al., 1975). nephrotic syndrome. Protein binding of valproic acid
In the presence of renal failure, quantitative adjust- was found to be further reduced during hemodialysis
ments in the dose of phenytoin are usually not necessary. (Bruni et al., 1980). As is the case with phenytoin, the
Total phenytoin serum concentrations need to be inter- decrease in the protein binding of valproic acid results
preted with caution given the increase in the free to total in an increased apparent volume of distribution, with
phenytoin concentration ratio in severe renal failure. lower steady-state total plasma concentrations for a
Free phenytoin levels are highly recommended for mon- given dose, while the free levels remain unchanged.
itoring purposes. Removal of phenytoin during dialysis Accumulation of valproic acid is unlikely in the presence
is negligible and supplemental dosing after dialysis is of renal failure since its elimination is almost exclusively
usually not necessary. by hepatic biotransformation (Kandrotas et al., 1990).
In the presence of severe hepatic disease the clearance However, the possibility of accumulation of active
of phenytoin can be reduced resulting in clinical intoxi- metabolites is a potential concern that has not been prop-
cation. This situation may be aggravated by the nonlinear erly studied. A slight reduction (about 27%) in the
kinetics of phenytoin. Since no simple markers of liver unbound clearance of valproic acid has been observed
function exist, phenytoin should be used with caution in patients with severe renal failure (creatinine clearan-
in liver disease. Frequent serum level determinations ce < 10 mL/min) (Kandrotas et al., 1990). Approximately
are recommended. 20% of a dose of valproic acid is extracted by
conventional, low-efficiency hemodialysis, resulting in
slight drops in the valproic acid serum levels. Plasma
Valproic acid
levels, however, recover a few hours after dialysis, with
Valproic acid (2-propyl-pentanoic acid; dipropyl-acetic no significant differences in the half-life of valproic acid
acid; 2-propyl-valeric acid) is a simple, eight-carbon, between dialysis and nondialysis days (Kandrotas et al.,
branched-chain carboxylic acid with a pKa of 4.9. Well 1990; Dasgupta et al., 1996). Dose supplementation
absorbed after oral administration, peak levels are after hemodialysis is generally not necessary. With the
observed within 0.5–4 hours. Of the total plasma concen- use of newer, high-flux dialyzers, however, the extrac-
tration, 90% is protein bound, with a resulting small vol- tion is significantly increased, and dose supplementation
ume of distribution. The protein binding of valproic acid may be required in some cases (Kane et al., 2000;
is nonlinear, progressively decreasing at higher plasma Kielstein et al., 2003).
USE OF ANTIEPILEPTIC DRUGS IN HEPATIC AND RENAL DISEASE 421
The disposition of valproic acid was studied in are no clear recommendations on the supplemental dos-
patients with alcoholic cirrhosis and acute viral hepatitis ing of phenobarbital following hemo- or peritoneal dial-
(Klotz et al., 1978). Plasma protein binding of valproic ysis. Pre- and postdialysis phenobarbital serum level
acid was reduced in patients with cirrhosis (70  11.3%) determinations can help in individualizing the dosing
and acute hepatitis (78  14.1%) compared to controls requirements.
(88  5.2%). The apparent volume of distribution was The disposition of phenobarbital has not been ade-
increased and lower peak plasma concentrations were quately studied in patients with hepatic disease. Since
observed in both groups compared to controls. Plasma a significant proportion of phenobarbital is eliminated
elimination half-life was prolonged in patients with cir- by direct renal excretion, the drug has a relatively low
rhosis (18.9  5.1 h) and acute hepatitis (17.0  3.7 h) ver- risk of accumulation in liver disease. In a single-dose
sus controls (12.2  3.7 h). The total plasma clearance of study, the half-life of phenobarbital was significantly
valproic acid, however, was not impaired, likely the prolonged in patients with liver cirrhosis (130  15 h)
result of increased penetration of valproic acid into when compared to a control group (86  3 h) (Alvin
the cells as a result of the decreased protein-binding et al., 1975). In the same study, in a group of patients with
capacity. When plasma clearance for free valproic acid acute viral hepatitis, the prolongation of the half-life
was calculated, it was found to be significantly reduced observed did not reach statistical significance but a large
only in patients with cirrhosis, indicating the possibility intersubject variability was found. Drug accumulation
of impaired biotransformation. Finally, these authors was reported in two patients on chronic phenobarbital
found no abnormalities in the pattern of excretion of therapy with chronic cirrhosis (Kutt et al., 1964). Since
valproate metabolites in the urine. Patients in this study biliary excretion of phenobarbital is insignificant in
were suspected to have mild liver failure based on labo- humans, dose adjustment is not necessary in patients
ratory parameters. Other authors have found a signifi- with cholestasis (Alvin et al., 1975).
cant increase in the plasma half-life and a reduction in
the clearance of total drug in a group of patients in
the acute stage of viral hepatitis (Gugler and von
Primidone
Unruh, 1980).
Primidone (2-deoxyphenobarbital) is well absorbed after
oral administration, with about 25% of the drug bound
Phenobarbital
to plasma proteins. It is metabolized in the liver to pheny-
Phenobarbital (5-ethyl-5-phenylbarbituric acid) is a weak lethylmalonamide (PEMA) and phenobarbital, both bio-
acid with a molecular weight of 232.23 and low water sol- logically active. Half-life of primidone is 3–12 hours,
ubility. It is well absorbed after oral administration, and that of PEMA 29–36 hours. Approximately 15–40% of
45–60% of the drug is bound to plasma proteins. Metab- primidone is excreted unchanged in the urine. Renal insuf-
olism of phenobarbital is mostly by hydroxylation ficiency can lead to accumulation of primidone, and
and subsequent glucuronidation (Tang et al., 1977). dose adjustments may be necessary, but no clear guide-
Main metabolites include parahydroxyphenobarbital lines are available. The use of primidone in renal failure
and N-hydroxyphenobarbital, both biologically inactive. is further complicated by the fact that the active metabo-
Between 9% and 33% of phenobarbital is excreted lites, PEMA and phenobarbital, may accumulate dispro-
unchanged in the urine, with the renal clearance affected portionally to the primidone. In the case reported by
by urine pH and flow. Elimination half-life ranges from Heipertz et al., serum levels of PEMA and primidone were
53 to 140 hours. proportionally higher than those of phenobarbital, and
The half-life of phenobarbital has been reported to be were thought to be responsible for the signs of clinical
unchanged in the presence of renal insufficiency intoxication (Heipertz et al., 1979). In the case reported
(Reidenberg and Affrime, 1973). Some drug accumula- by Stern, very high levels of PEMA correlated with clinical
tion, however, is to be expected in the presence of severe signs of toxicity whereas phenobarbital levels were only
renal failure. Hemodialysis removes 20–50% of the drug slightly elevated and primidone levels were within the
during a 4 hour period, depending on the type of dialyzer therapeutic range (Stern, 1977). Serum levels of all three
and blood flow rate. Low-efficiency, low-blood flow compounds may be necessary to properly assess the pos-
dialyzers show a clearance of about 20% (Zawada sibility of primidone intoxication in uremic patients.
et al., 1983), whereas high-efficiency, high-blood flow Hemodialysis removes 20–50% of a primidone dose.
dialysis increases the clearance to about 50% (Palmer, A 30% dose supplementation has been recommended
2000). Peritoneal dialysis has been shown to clear about prior to hemodialysis (Lee et al., 1982; Streete et al., 1990).
35–40% of the daily dose of phenobarbital over a 24 hour There are no studies of the effects of liver disease on
period (Marquardt et al., 1992; Porto et al., 1997). There the pharmacokinetics of primidone.
422 J.J. ASCONAPÉ
Carbamazepine, oxcarbazepine, hepatic failure. Close monitoring of carbamazepine
and eslicarbazepine serum concentration is recommended in this situation.
Carbamazepine, oxcarbazepine, and eslicarbazepine
OXCARBAZEPINE
acetate belong to the dibenzazepine family and are
closely related structurally. In terms of their elimination, Oxcarbazepine (10,11-dihydro-10-oxo-5H-dibenz(b,f )
however, these drugs have important differences. Car- azepine-5-carboxamide) is a keto-analogue of carbamaz-
bamazepine is eliminated almost exclusively by hepatic epine. Oxcarbazepine, a prodrug, undergoes rapid pre-
metabolism whereas oxcarbazepine and eslicarbazepine systemic 10-keto reduction by the cytosol arylketone
are prodrugs whose main active metabolite is eliminated reductase to the 10-monohydroxy derivative (MHD),
mostly by renal excretion. the pharmacologically active compound. MHD is pri-
marily converted to a glucuronide conjugate, with a
small amount further hydroxylated to a dihydroxy deriv-
CARBAMAZEPINE ative (DHD), an inactive metabolite. The two major reac-
tions controlling the disposition of oxcarbazepine and
Carbamazepine (5-H-dibenz[b,f ]azepine-5-carboxamide)
MHD (reduction and glucuronidation) are nonoxidative.
has a molecular weight of 236.3 and very poor water sol-
As a result, oxcarbazepine has a much lower propensity
ubility. Its bioavailability is approximately 80% after oral
to induce P450 oxidative enzymes than carbamazepine,
administration, with peak plasma levels reached in 4–12
and a more favorable drug–drug interaction potential.
hours. About 70–80% of the drug in plasma is protein
Furthermore, the reduction and glucuronidation of
bound, mostly to globulins (Hooper et al., 1975). Elimina-
drugs are not affected as much by hepatic impairment
tion of carbamazepine is almost exclusively by hepatic
as oxidative metabolism. Approximately 38% of MHD
biotransformation via the CYP3A4, with < 1% of the
is bound to plasma proteins. The half-life of MHD is
drug eliminated by direct renal excretion. Epoxidation
8–10 hours (Lloyd et al., 1994).
and hydroxylation are the main metabolic pathways.
In the study by Rouan et al. (1994), there was a linear
The 10-11-carbamazepine epoxide, an active metabolite,
correlation between the creatinine clearance and the
undergoes further hydroxylation with < 2% eliminated
clearance of the MHD and its conjugated metabolites.
as such in the urine. Carbamazepine is a potent inducer
In subjects with severe renal insufficiency (creatinine
of the CYP450 system and is capable of inducing its
clearance < 10 mL/min), the elimination half-life of the
own metabolism as well as the metabolism of other com-
MHD was prolonged to approximately 19 hours, with a
monly used drugs such as oral contraceptives, warfarin,
corresponding 2–2.5 times increase in the mean area
digoxin, ciclosporin, and tacrolimus.
under the curve in the plasma concentration-time curves
Renal disease does not affect the protein binding of
of oxcarbazepine and the MHD compared to healthy con-
carbamazepine (Hooper et al., 1975). Since the drug and
trols (Rouan et al., 1994). The authors concluded that, in
its active metabolite, the 10-11-carbamazepine epoxide,
patients with mild renal failure (creatinine clearan-
are eliminated almost exclusively by biotransformation,
ce > 30 mL/min), no dose adjustments are necessary. In
no significant accumulation is observed even in the pres-
patients with moderate renal impairment (creatinine clear-
ence of severe renal failure, and dose adjustments are
ance 10–30 mL/min), a 50% reduction in the target dose
typically not necessary. Carbamazepine has relatively
of oxcarbazepine is recommended. In patients with end-
low extractability during dialysis and routine dose sup-
stage renal disease (creatinine clearance < 10 mL/min),
plementation is usually not recommended (Lee et al.,
no specific dosage recommendations were given, but an
1980). Hemodialysis, however, has been shown to be
even more conservative dosing is warranted. There are
effective in the management of carbamazepine intoxica-
no data on the disposition of MHD during dialysis.
tion, with 22–50% reductions in the serum concentra-
The pharmacokinetics of oxcarbazepine and MHD
tions of the drug reported, so caution is recommended
are not affected by mild or moderate hepatic dysfunc-
(Schuerer et al., 2000; Kielstein et al., 2002; Tapolyai
tion (Child–Pugh Hepatic Impairment Classification
et al., 2002; Chetty et al., 2003).
types A and B, respectively), and dose adjustments are
In the presence of mild liver insufficiency, the protein
usually not necessary (Asconapé and D’Souza, 2004).
binding of carbamazepine was found to be significantly
Patients with severe hepatic failure (Child–Pugh C) have
decreased when compared to normal controls (Hooper
not been studied adequately.
et al., 1975). The decrease, however, was slight, and
did not correlate with any laboratory parameters. The
ESLICARBAZEPINE ACETATE
disposition of carbamazepine has not been adequately
studied in subjects with liver disease, but significant Eslicarbazepine acetate, (S)-(-)-10-acetoxy-10,11-dihy-
accumulation is to be expected in the presence of severe dro-5H-dibenzazepine-5-carboxamide, differs from
USE OF ANTIEPILEPTIC DRUGS IN HEPATIC AND RENAL DISEASE 423
oxcarbazepine in that it is metabolized almost administration, has an apparent volume of distribution
exclusively to the (S)-enantiomer (designated as S-licar- after oral administration of 1.14, and is about 55% pro-
bazepine) with less than a 5% chiral conversion to the tein bound. The elimination half-life is 24–35 hours.
(R)-enantiomer, whereas oxcarbazepine is converted to However, when used concomitantly with enzyme-
both (S)- and (R)- enantiomers in about a 4–5:1 proportion inducing drugs, the half-life is reduced to approximately
(Hainzl et al., 2001). Eslicarbazepine acetate is an oral 14 hours, and, in the presence of valproate, increased to
prodrug that is rapidly and extensively metabolized about 70 hours. Lamotrigine is metabolized into several
by the liver via a hydrolytic first-pass metabolism into inactive compounds, of which the aromatic N-2 glucuro-
S-licarbazepine, the biologically active drug. Eslicarbaze- nide is the most important. In healthy volunteers, 71% of
pine acetate is well absorbed after oral administration the orally administered lamotrigine was recovered as N-2
with a bioavailability about 16% higher than that observed glucuronide, and 10% as unchanged lamotrigine
after an equivalent dose of oxcarbazepine (Almeida and (Dickins and Chen, 2002).
Soares-da-Silva, 2003; Almeida et al., 2005). Peak plasma Lamotrigine has not been adequately studied in
concentrations (tmax) are observed in 1–4 hours. Plasma patients with renal disease. In a small study, the renal
protein binding is low (<40%). Eslicarbazepine acetate clearance of lamotrigine was found to be decreased in
has a half-life of 13–20 hours, and displays linear kinetics patients with chronic renal failure compared to healthy
at doses of 400–1200 mg/day (Almeida and Soares- controls. This difference was not clinically relevant since
da-Silva, 2007). It is eliminated predominantly by renal the renal clearance of lamotrigine has a minor role in the
excretion, with 91% of the drug recovered in urine overall elimination of the drug (Wootton et al., 1997). In
following an oral dose (Almeida and Soares-da-Silva, this study, the mean half-life of lamotrigine was 36 hours,
2007). Eslicarbazepine acetate is rapidly converted by prolonged compared to 28 hours in the healthy volunteers,
hydrolysis to eslicarbazepine. In healthy volunteers, of probably reflecting a larger volume of distribution in the
the dose recovered in the urine, 52% corresponded to esli- patients (Dickins and Chen, 2002). In another study, fol-
carbazepine and 41% to eslicarbazepine-glucuronide lowing the administration of a single 100 mg dose of
(Maia et al., 2008). Eslicarbazepine has a clearance rate lamotrigine in patients with renal failure, the mean lamo-
of 20–30 mL/min, with 20% of the dose recovered in trigine clearance was reduced at 27.9 mL/min and the
the urine after 12 hours and 40% within 24 hours half-life prolonged at 50.7 hours in patients with renal fail-
(Almeida and Soares-da-Silva, 2007). ure not undergoing dialysis compared to the correspond-
In subjects with mild renal impairment eslicarbaze- ing values of healthy controls at 38.5 mL/min and 25.7
pine demonstrated slightly slower elimination rates com- hours, respectively (Fillastre et al., 1993). In dialyzed
pared to normal volunteers (10.2 mL/min versus 17. patients, mean half-life was 59.6 hours off dialysis and
3 mL/min, respectively) (Maia et al., 2008). As expected, 15.5 hours during dialysis (Fillastre et al., 1993). On aver-
patients with moderate or severe renal impairment age, 17% of the lamotrigine body pool was removed for
showed much lower clearance rates (3.7 mL/min and every 4 hour dialysis session (Fillastre et al., 1993). In view
1.5 mL/min, respectively) (Maia et al., 2008). Dose of these findings, daily dosing of lamotrigine may need to
adjustments are generally required in the presence of be reduced in patients with moderate to severe renal fail-
moderate or severe renal impairment (creatinine clearan- ure. Posthemodialysis supplementation is probably not
ce < 50 mL/min). There is no information on the effects necessary in most patients. Clearance with peritoneal dial-
of hemodialysis on the serum concentrations of eslicar- ysis has not been studied.
bazepine, but significant extraction is to be expected and The pharmacokinetics of lamotrigine has not been
dose supplementation may be necessary. well studied in patients with liver failure. In healthy sub-
The clearance of eslicarbazepine was not signifi- jects with unconjugated hyperbilirubinemia (Gilbert syn-
cantly affected in the presence of mild or moderate drome), characterized by a deficiency of the enzyme
hepatic failure, and dose adjustments are usually not bilirubin uridine diphosphate glucuronyl transferase
necessary in this setting (Almeida et al., 2008). The phar- (UGT), which is involved in the metabolism of lamotri-
macokinetics of eslicarbazepine has not been studied in gine, the clearance of lamotrigine was 32% lower and
patients with severe hepatic impairment. its half-life 37% longer than in healthy controls
(Posner et al., 1989). These differences, however, were
considered unlikely to be of clinical significance.
Lamotrigine
Lamotrigine (3,5-diamino-6-(2,3-dicholorophenyl)-1,2,4-
Levetiracetam
triazine) is a phenyyltriazine with a molecular weight
of 256.09 and poor solubility in water (Dickins and Levetiracetam, (S-a-ethyl-2-oxo-1-pyrrolidine acet-
Chen, 2002). Lamotrigine is well absorbed after oral amide), a racemically pure ethyl analog of piracetam,
424 J.J. ASCONAPÉ
is structurally unrelated to other antiepileptic drugs hours. Topiramate is not extensively metabolized. In
(Patsalos, 2004). It is rapidly and almost completely healthy volunteers, about 20% of the administered
absorbed (>95%) after oral administration with a Tmax dose is metabolized; however, about 50% of the drug
of 1.3 hours. It is not protein bound (<10%), has a vol- is metabolized with the concomitant use of enzyme-
ume of distribution of 0.5–0.7 L/kg, and a half-life of inducing antiepileptic drugs (Britzi et al., 2005).
6–8 hours (Patsalos, 2004). Levetiracetam is mostly elim- Serum concentrations of the drug were 40–50% lower
inated through the kidneys. Approximately 95% of the in the presence of enzyme inducers. No active
administered dose was recovered in the urine in healthy metabolites have been identified in humans. The drug
volunteers. The total body clearance is approximately is eliminated predominantly by the kidney, with
0.96 mL/min/kg, and the renal clearance 0.6 mL/min/ 50–80% of the drug recovered unchanged in the urine.
kg in adults, 0.8 mL/min/kg in children, and 0.5 mL/ Oral plasma clearance (CL/F) is approximately
min/kg in the elderly (Patsalos, 2004). Levetiracetam 20–30 mL/min.
undergoes glomerular filtration and approximately Clearance of topiramate was reduced by 42% in mod-
66% of the dose is excreted unchanged in the urine. erate renal failure (creatinine clearance 30–69 mL/min/
The remainder is metabolized, through enzymatic hydro- 1.73 m2) and by 54% in severe renal failure (creatinine
lysis of the acetamide group (<25% of the administered clearance < 30 mL/min/1.73 m2) compared to normal
dose), to an inactive carboxylic acid metabolite. Other renal function subjects (creatinine clearance > 70 mL/
minor inactive metabolites, accounting for < 3% of min/1.73 m2). In subjects with renal failure (creatinine
the administered dose, have been identified as well clearance < 70 mL/min/1.73 m2), a 50% reduction of
(Radtke, 2001). The metabolism of levetiracetam is inde- the usual adult dose is recommended by the manufac-
pendent of the cytochrome P450 system. turer. Topiramate is cleared by hemodialysis at a rate
The elimination of levetiracetam is significantly that is 4–6 times greater than in individuals with normal
delayed in the presence of renal failure, with the clear- renal function.
ance of the drug being directly proportional to the creat- The effects of hepatic disease on the disposition of
inine clearance (French, 2001). The half-life of the drug topiramate have not been adequately studied, but signif-
ranges from approximately 10 hours in the presence of icant accumulation is not to be expected. A 30% increase
mild renal failure to 24 hours in severe renal failure in the drug concentration has been reported in moderate
(French, 2001). Therefore, dose adjustments are usually to severe hepatic failure, but the mechanism is unclear
necessary depending on the severity of the renal failure (Ahmed and Siddiqui, 2006).
(Table 27.2). Levetiracetam is efficiently removed by
hemodialysis, with approximately 50% of the drug pool
Lacosamide
removed by 4 hours of dialysis. A supplemental dose of
250–500 mg per 4 hours of dialysis is recommended by Lacosamide, (R)-2-acetamido-N-benzyl-3-methoxypro-
the manufacturer. prionamide, is a functionalized amino acid with a molec-
Liver failure does not affect the elimination of levetir- ular weight of 250.30. It is a white to light yellow
acetam significantly. A single-dose (1000 mg) study in crystalline powder that is soluble in water and acetoni-
male subjects with varying degrees of hepatic failure has trile, and slightly soluble in ethanol.
been conducted (French, 2001). In patients with mild or Lacosamide is well absorbed after oral administration
moderate hepatic failure (Child–Pugh class A and B with a bioavailabilty of approximately 100% (Hovinga,
respectively), no significant changes in the pharmacokinet- 2003). Food does not affect the rate or extent of the
ics of the drug were found. In patients with severe hepatic absorption (Cawello et al., 2004). The volume of distri-
failure (Child–Pugh class C), clearance was reduced by bution of lacosamide is 0.6 L/kg and the binding to
46% and the plasma concentrations were increased com- plasma proteins is < 15%. Lacosamide is primarily elim-
pared to healthy controls. However, these patients also inated from the systemic circulation by renal excretion
had renal failure, which could account for the pharmaco- and biotransformation. After oral or intravenous admin-
kinetic changes observed. In general, dose adjustments are istration, 95% of the lacosamide is recovered in the urine
not necessary in patients with hepatic disease. and < 0.5% in the feces. The major compounds found in
the urine include: unchanged lacosamide (approximately
40%), O-desmethyl metabolite (approximately 30%),
Topiramate
and a structurally unknown polar fraction (approxi-
Topiramate (2,3:4,5-bis-O-(1-methylethylidene)-b-D- mately 20%). The metabolites have no pharmacologic
fructopyranose sulfamate), is a sulfamate-substituted activity. Metabolism of lacosamide is through the
derivative of D-fructose. It is minimally bound to serum CYP2C19. The half-life of lacosamide is approximately
proteins (15%) and has a half-life of approximately 21 13 hours.
USE OF ANTIEPILEPTIC DRUGS IN HEPATIC AND RENAL DISEASE 425
Table 27.2
Dose adjustments for antiepileptic drugs in adult patients with renal failure

Total daily dose according to GFR

Drug 60–89 mL/min 30–59 mL/min 15–29 mL/min <15 mL/min

Carbamazepine 400–2400 mg No adjustment necessary No adjustment necessary No adjustment necessary


Eslicarbazepine 800–1200 mg Reduce dose by 50% Insufficient data, Insufficient data, use with
use with caution caution
Ethosuximide 500–1500 mg No adjustment Adjustment may be Adjustment may be
necessary necessary necessary
Ezogabine/ 600–1200 mg 300–600 mg (50% 300–600 mg 300–600 mg
retigabine reduction)
Felbamate 1200–3600 mg Reduce dose by 50% Insufficient data, Insufficient data, use
use with caution with caution
Gabapentin 900–3600 mg/ 400–1400 mg/day (BID) 200–700 mg/day (QD) 100–300 mg/day (QD)
day (BID or
TID)
Lacosamide 200–400 mg No adjustment necessary Maximum dose of Maximum dose of
300 mg/day for GFR 300 mg/day for GFR
<30 mL/min <30 mL/min
Lamotrigine 200–400 mg Insufficient data, use Insufficient data, use Insufficient data, use
with caution with caution with caution
Levetiracetam 500–1000 mg 250–750 mg BID 250–500 mg BID 250–500 mg BID
BID
Oxcarbazepine 300–600 mg 300–600 mg BID Reduce dose by 50% Insufficient data, use
BID with caution
Phenobarbital 60–240 mg Insufficient data, use Insufficient data, use Insufficient data, use with
with caution with caution caution. BID dosing
Phenytoin 200–600 mg No adjustment necessary No adjustment necessary No adjustment necessary
Pregabalin 150–600 mg/day 75–300 mg/day 25–150 mg/day (QD 25–75 mg/day (QD)
(BID or TID) (BID or TID) or BID)
Primidone 750–2000 mg Insufficient data, use Insufficient data, use Insufficient data, use
with caution with caution with caution
Rufinamide 800–3200 mg No adjustment necessary No adjustment necessary No adjustment necessary
Tiagabine 32–56 mg No adjustment necessary No adjustment necessary No adjustment necessary
Topiramate 100–200 mg 50–100 mg BID for 50–100 mg BID 50–100 mg BID
BID GFR <70 mL/min
Valproate 1000–4000 mg No adjustment necessary No adjustment necessary No adjustment necessary
Vigabatrin 1000–3000 mg 25% dose reduction for 50% dose reduction for 75% dose reduction for
GFR >50 to 80 mL/min GFR >30 to 50 mL/min GFR >10 to <30 mL/min
Zonisamide 200–400 mg 200–400 mg Insufficient data, use Insufficient data, use
with caution with caution

GFR, glomerular filtration rate; BID, twice daily; QD, once daily; QOD, once every other day.

The presence of renal disease may affect the elimina- usually recommended in the presence of severe renal
tion of lacosamide. The area under the curve was failure. Four hours of hemodialysis reduces the area
increased by 25% in mild (creatinine clearance 30– under the curve of lacosamide by about 50%. Therefore,
50 mL/min) and 60% in severe (creatinine clearan- a 25–50% dose supplementation is recommended
ce  30 mL/min) renal insufficiency when compared to following dialysis.
subjects with normal renal function (creatinine clearan- In the presence of mild liver insufficiency (Child–
ce > 80 mL/min). There were no changes observed in the Pugh A) no dose adjustments are usually necessary. In
Cmax. Dose adjustments are usually not necessary in moderate hepatic failure (Child–Pugh B) a 50–60%
the presence of mild and moderate renal insufficiency. increase in the area under the curve was observed
A 75% reduction in the total daily maintenance dose is compared to normal controls. A 75% reduction in the
426 J.J. ASCONAPÉ
usual maintenance dose has been recommended in The elimination half-life of gabapentin is 6–7 hours
these patients. The elimination of lacosamide has not and the clearance 100–300 mL/min.
been studied in patients with severe hepatic failure Renal failure significantly alters the elimination of
(Child–Pugh C). gabapentin and adjustments in the maintenance dosage
are usually necessary (Table 27.2) (Blum et al., 1994).
After a single 400 mg dose of gabapentin in anuric sub-
Tiagabine
jects, the mean gabapentin half-life was 132 hours in
Tiagabine ((-)-(R)-1-[4,4-bis(3methyl-2-thienyl)-3- between hemodialysis periods, increasing during hemo-
butenyl]-3-piperidinecarboxylic acid hydrochloride) is a dialysis to a mean half-life of about 4 hours (Wong et al.,
selective GABA reuptake inhibitor with modest efficacy 1995). Due to its high water solubility, low molecular
for partial seizures. Tiagabine has an absolute oral bioa- weight and negligible protein binding, gabapentin is
vailabilty of 90% and a Tmax of about 1 hour (Adkins and effectively extracted by hemodialysis (Wong et al.,
Noble, 1998). Its volume of distribution is 1 L/kg and 1995). Plasma levels of gabapentin were reduced by
more than 95% of the drug is bound to plasma proteins 40% during the first 2 hours of hemodialysis compared
(Adkins and Noble, 1998). Tiagabine is extensively to predialysis levels (Wong et al., 1995). Approximately
metabolized by the liver (CYP3A4), with 63% of an oral 50% of the body pool of gabapentin is expected to be
dose recovered in the feces and 25% in the urine. Only extracted during a 4 hour session of hemodialysis
1% of the drug is eliminated unchanged in the urine. (Wong et al., 1995). Therefore, a postdialysis dose sup-
The plasma clearance is 12.8 L/h, but increases to plementation is usually necessary to maintain steady-
21.4 L/h when used in conjunction with enzyme-inducing state plasma concentrations (Table 27.3).
drugs. The elimination half-life is 5–8 hours, (2–3 hours Given its pharmacokinetic properties, the disposition
with enzymatic induction). of gabapentin is unlikely to be affected by liver disease.
Impaired renal function and hemodialysis do not affect The pharmacokinetics of gabapentin, however, has not
the pharmacokinetics of tiagabine significantly and dose been systematically studied in patients with liver failure.
adjustments are not necessary (Cato et al., 1998). In the Pregabalin (S-3-(aminomethyl)-5-methylhexanoic
presence of hepatic disease, the clearance of tiagabine is acid) has a molecular weight of 159.23 and is readily sol-
reduced proportionally to the severity of the liver failure uble in water. It is well absorbed (>90%) after oral
and significant accumulation of the drug may occur administration (Ben-Menachem, 2004). Pregabalin
(Lau et al., 1997). In subjects with normal hepatic function, undergoes negligible metabolism in humans and it does
mild and moderate impairment, the elimination half-life not bind to plasma proteins. The elimination half-life of
of tiagabine was 7, 12 and 16 hours, respectively (Lau pregabalin is about 6 hours in healthy subjects. Pregaba-
et al., 1997). The free tiagabine concentrations and the free lin is eliminated from the systemic circulation by direct
fraction were increased in the subjects with hepatic impair- renal excretion as unchanged drug, with a renal clear-
ment. No studies have been conducted in subjects with ance of 67.0–80.9 mL/min in healthy volunteers.
severe hepatic failure. Tiagabine should be used cautiously Pregabalin clearance is nearly proportional to the cre-
in subjects with hepatic failure. atinine clearance, with both the area under the curve and
the elimination half-life increasing with decreasing renal
function (Randinitis et al., 2003). Similarly to gabapen-
Gabapentin and pregabalin
tin, pregabalin is effectively extracted by hemodialysis.
Gabapentin and pregabalin belong to a category of func- The plasma concentrations of pregabalin fall approxi-
tionalized amino acids designed to mimic the chemical mately 50% following 4 hours of hemodialysis. Dose
structure of g-aminobutyric acid (GABA), but lacking adjustments in the maintenance dose and postdialysis
any direct GABAergic effect. dose supplementation are, therefore, necessary in the
Gabapentin (1-(aminomethyl)cyclohexane acetic acid) presence of renal failure (Table 27.3).
is highly soluble in water and has a molecular weight of Liver disease is not expected to affect the disposition
171.34. It has saturable absorption after oral administra- of pregabalin, and dose adjustments are not necessary.
tion, with a progressive decrease in bioavailability as the
dose increases (McLean, 1994). It has almost no protein
Vigabatrin
binding and its volume of distribution standardized for
weight is 0.65–1.4 L/kg. Gabapentin undergoes no Vigabatrin (4-amino-5-hexenoic acid; g-vinyl GABA) is
hepatic metabolism. The absorbed drug is eliminated an irreversible, specific inhibitor of the GABA transam-
entirely by the kidney unchanged. Its elimination rate inase. It is readily soluble in water and the molecular
constant, plasma clearance and renal clearance are line- weight is 129.16. Vigabatrin is completely absorbed fol-
arly related to the creatinine clearance (McLean, 1994). lowing oral administration. It does not bind to plasma
USE OF ANTIEPILEPTIC DRUGS IN HEPATIC AND RENAL DISEASE 427
Table 27.3
Antiepileptic drugs and hemodialysis

Molecular Water Plasma protein Volume of Dose supplementation


Drug weight solubility binding distribution (L/kg) (per 4 hours of hemodialysis)

Carbamazepine 236.3 Very low 70–80% 0.8–1.2 Not necessary


Eslicarbazepine Low 38% (MHD) 2.7 Not studied (probably necessary)
Ethosuximide 141.17 High None 0.65 Necessary: 50% of total daily dose
Ezogabine/retigabine 303.3 Low 80% 2–3 Not studied
Felbamate 238.24 Very low 20–25% 0.75 Not studied (probably not necessary)
Gabapentin 171.34 High None 0.65–1.4 Necessary: 100–200% of total daily dose
Lacosamide 250.3 High <15% 0.6 Necessary: 50% of total daily dose
Lamotrigine 256.09 Low 55% 0.9–1.3 Usually not necessary
Levetiracetam 170.21 High <10% 0.5–0.7 Necessary: 50% of total daily dose
Oxcarbazepine 252.3 Low 38% (MHD) 0.3–0.8 Not studied (probably necessary)
Phenobarbital 232.23 Low 45–60% 0.4–0.7 Probably necessary, but not well
established. Pre- and postdialysis levels
recommended for dosing
Phenytoin 252.26 Low 90% 0.5–0.8 Usually not necessary. However,
70–80% in significant extraction has been reported
ESRD with use of high-efficiency dialyzers
Pregabalin 159.23 High None 0.5 Necessary: 100–200% of total daily dose
Primidone 218.25 Low 25% 0.6–1.0 Necessary: 30% supplemental dose prior
to dialysis
Rufinamide 238.2 Very low 34% 0.8–1.2 Not necessary
Tiagabine 412.0 Low 96% 1.0 Not necessary
Topiramate 339.4 Low 15% 0.6–1.0 Necessary: 50% of total daily dose
Valproate 144.2 Very low 90% 0.1–0.4 Usually not necessary. However,
70–80% in significant extraction has been reported
ESRD with use of high-efficiency dialyzers
Vigabatrin 129.16 High None 0.8 Not studied
Zonisamide 212.23 Low 40–60% 1.09–1.77 May be necessary: 25–50% of total
daily dose

MHD, monohydroxy derivative; ESRD, end-stage renal disease.

proteins and is widely distributed in the body (volume of respectively. The effects of dialysis on the clearance of
distribution: 0.8–1.1 L/kg). Vigabatrin is not significantly vigabatrin have not been studied adequately, but, given
metabolized and its elimination is primarily through its chemical and pharmacokinetic characteristics, it is
renal excretion. The half-life is approximately 5–8 hours, likely that the drug will be significantly extracted. How-
and the total clearance 1.7–1.9 mL/min/kg (Grant and ever, considering that the biologic half-life of vigabatrin,
Heel, 1991). About 80% of the oral dose is eliminated determined by irreversible enzymatic inhibition, is in the
in the urine as unchanged drug. order of several days, fluctuation of serum concentra-
In the presence of renal disease, vigabatrin accumu- tions during hemodialysis are probably not as clinically
lates proportionally to the decrease in the creatinine clear- relevant as with other antiepileptic drugs.
ance (Haegele et al., 1988). In subjects with mild renal The elimination of vigabatrin in the presence of liver
failure (CLcr > 50–80 mL/min), the mean area under failure has not been studied, but is not likely to be
the curve was increased by 30% and the half-life by affected.
55%. In moderate renal failure (CLcr > 30–50 mL/min),
the mean area under the curve and the half-life increased
Zonisamide
twofold. In severe renal failure (CLcr > 10– 30 mL/min),
the area under the curve increased 4.5-fold and the Zonisamide (1,2-benzisoxazole-3-methanesulfonamide)
half-life 3.5-fold. As a result, the maintenance dose of has a molecular weight of 212.23 and a pH-dependent
vigabatrin should be reduced by 25%, 50%, or 75% in water solubility, with low solubility at acidic or neutral
the presence of mild, moderate, or severe renal failure, pH, and increased solubility as the pH increases beyond
428 J.J. ASCONAPÉ
8. Zonisamide is absorbed rapidly and completely after The pharmacokinetics of felbamate has not been
oral administration (Leppik, 2004). The elimination studied in subjects with liver failure. Since the use of
half-life is long, about 50–60 hours. The drug distributes the drug has been associated with fulminant liver failure,
evenly in the body, with a volume of distribution between it is better avoided in patients with hepatic disease.
1.09 and 1.77 L/kg. Zonisamide is 40–60% bound to
plasma proteins, and has an even higher affinity to bind Ethosuximide
to erythrocytes. Zonisamide is extensively metabolized
Ethosuximide (2-ethyl-2-methylsuccinimide) is a water-
in humans, with the main pathways including glucuro-
soluble weak acid with a molecular weight of 141.17. It
nide conjugation, acetylation, and hydroxylation fol-
is well absorbed after oral intake (Buchanan et al.,
lowed by oxidation. After a single dose in humans,
1973). Ethosuximide is not bound to plasma proteins,
approximately 30% of the drug is recovered unchanged
and it is distributed through body water with an apparent
in the urine, 20% as N-acetyl-zonisamide, and 50% as the
volume of distribution of 0.65 L/kg. Ethosuximide is
glucuronide of the open-ring metabolite (Buchanan
eliminated primarily by metabolism, with only about
et al., 1996).
20% of an administered dose recovered unchanged in
The effects of renal failure on the disposition of zoni-
the urine (Buchanan et al., 1973). The elimination half-
samide have not been adequately studied. Significant
life is 40–60 hours, and is generally higher in children
accumulation of the drug is not to be expected in the
than in adults.
presence of mild to moderate renal failure since only
Decreased renal function is not likely to affect the
30% of the drug is eliminated directly by the kidney.
elimination of ethosuximide significantly and dose
Data from the manufacturer indicates that, following
adjustments are probably not needed. Ethosuximide is
a single 300 mg dose of zonisamide, a 35% increase in
removed efficiently by hemodialysis and peritoneal dial-
the area under the curve was observed in subjects with
ysis (Marbury et al., 1981; Marquardt et al., 1992). The
severe renal failure (CLcr < 20 mL/min). In a small
elimination half-life of ethosuximide was reduced to
study, a 50% reduction in the plasma concentration of
3–4 hours during hemodialysis, and approximately
zonisamide was observed following 4–5 hours of hemo-
40–50% of the total daily dose of ethosuximide was
dialysis (Ijiri et al., 2004). Levels of zonisamide remained
removed during a 4 hour session of hemodialysis using
low 5–6 hours after dialysis, suggesting that there was no
a low-efficiency dialyzer (Marbury et al., 1981). A single
redistribution of the drug. A postdialysis supplemental
case report in a child demonstrated that 50% of the total
dose is not routinely recommended, but may be neces-
daily dose of ethosuximide was removed by peritoneal
sary in some cases.
dialysis (Marquardt et al., 1992). Patients on ethosuxi-
The pharmacokinetics of zonisamide has not been
mide will probably need dose supplementation at the
studied in subjects with liver disease. Caution is recom-
time of dialysis, but this had not been systematically
mended when prescribing zonisamide in the presence of
investigated.
moderate to severe hepatic failure.
Because ethosuximide is eliminated predominantly
by metabolism, hepatic disease is expected to impair
Felbamate its elimination. The pharmacokinetics of ethosuximide,
however, has not been studied in subjects with liver
Felbamate (2-phenyl-1,3-propanediol dicarbamate) has a
failure.
molecular weight of 238.24 and very low water
solubility. It is well absorbed (90%) after oral administra-
Rufinamide
tion (Palmer and McTavisk, 1993). Approximately 20–
25% of the drug is bound to serum proteins. Of the Rufinamide (1-((2,6-difluorophenyl)methyl)-1H-1,2,3-
absorbed drug, 40–50% is eliminated by direct renal triazole-4 carboxamide) is a triazole derivative with a
excretion and the rest undergoes biotransformation, molecular weight of 238.2 and very poor water solubility.
resulting in mostly inactive or weakly active Rufinamide is well absorbed after oral administration up
metabolites (Shumaker et al., 1990). Mean elimination to doses of 1600 mg, with a nonlinear decrease in bio-
half-life was 16–22 hours in healthy volunteers availability observed at higher doses (Perucca et al.,
(Shumaker et al., 1990). 2008). A small proportion (34%) of the drug is bound
Clearance of felbamate is decreased in the presence to plasma proteins, mostly albumin. Elimination half-life
of renal failure and the drug is likely to accumulate in is 6–10 hours, with an apparent volume of distribution of
patients with moderate to severe renal failure (Glue 50 L at a dose of 3200 mg/day. Rufinamide is exten-
et al., 1997). The dosing of felbamate in the presence sively metabolized via hydrolysis by carboxylesterases
of renal failure has not been adequately studied to pro- to a pharmacologically inactive carboxylic acid deriva-
vide specific recommendations. tive. The metabolism of rufinamide does not involve
USE OF ANTIEPILEPTIC DRUGS IN HEPATIC AND RENAL DISEASE 429
the cytochrome P450. Less than 2% of a given dose is limited information on the effects of hemodialysis on
eliminated unchanged in the urine. The pharmacokinet- the disposition of ezogabine/retigabine.
ics of rufinamide is not affected by impaired renal func- The effects of varying degrees of liver impairment on
tion and the drug is not significantly extracted by the pharmacokinetics of ezogabine/retigabine were stud-
hemodialysis (Perucca et al., 2008). A regimen of three ied following a single 100 mg dose. Mild liver failure
hemodialyses a week carried out 3 hours after dosing (Child–Pugh score 5–6) had no significant effect on
of rufinamide was predicted to reduce the average area the elimination of ezogabine/retigabine. In subjects with
under the curve over 1 week by 12% compared to subjects moderate (Child–Pugh score 7–9) and severe (Child–
not undergoing dialysis (Perucca et al., 2008). Therefore, Pugh score > 9) hepatic impairment there was a 50%
dose supplementation in patients on hemodialysis is gen- and 100% increase in the area under the curve, respec-
erally not required. tively. There was an approximately 30% increase in the
The effects of hepatic failure on the pharmacokinet- exposure of the N-acetyl metabolite in subjects with
ics of rufinamide have not been studied. The drug should moderate to severe hepatic impairment. Dose reductions
be used with caution in patients with mild or moderate are recommended in moderate to severe hepatic impair-
liver disease and probably avoided in the presence of ment. Initial dose should not exceed 150 mg/day in these
severe hepatic impairment. groups. Maximal doses recommended are 750 mg/day in
moderate and 600 mg/day in severe liver failure.

Ezogabine/retigabine
CONCLUSIONS
Ezogabine/retigabine (N-(2-amino-4-(4-fluorobenzyla-
mino)-phenyl) carbamic acid ethyl ester) was recently Based on their route of elimination, antiepileptic drugs can
approved for the adjunctive treatment of partial epilepsy. be divided into three main groups. Drugs that are elimi-
Known worldwide as retigabine (INN), it is called ezoga- nated unchanged by the kidneys or undergo minimal
bine (USAN) in the US. Its mechanism of action is not metabolism, drugs that are removed almost exclusively
fully understood, but it appears to exert its main anticon- by hepatic metabolism, and drugs that are eliminated by
vulsant effect by enhancing potassium currents (Kv7.2 to a combination of renal and nonrenal routes. Drugs in
7.5) through activation of the voltage-gated potassium the first group include gabapentin, pregabalin, vigabatrin,
channels (KCNQ2/3 and KCNQ3/5) (Plosker and Scott, and topiramate when used as monotherapy or associated
2006). It has a molecular weight of 303.3 and low water with nonenzyme-inducing drugs. These drugs will require
solubility. It is rapidly absorbed after oral administration. major dose adjustments in patients with severe renal insuf-
Ezogabine/retigabine is approximately 80% bound to ficiency and may be better avoided in these cases. Drugs
plasma proteins and has a volume of distribution of eliminated predominantly by biotransformation include
2–3 L/kg. Ezogabine/retigabine undergoes extensive phenytoin, valproate, carbamazepine, tiagabine, and
phase II metabolism via glucuronidation and acetylation. rufinamide. These drugs are at significant risk of
Its monoacetylated metabolite, N-acetyl ezogabine/retiga- accumulation with advanced hepatic disease. Drugs in
bine, is biologically active, but less potent than the parent the intermediate group include levetiracetam, lacosamide,
compound (Blackburn-Munro et al., 2005). The elimina- zonisamide, primidone, phenobarbital, ezogabine/retiga-
tion of ezogabine/retigabine is predominantly renal, with bine, oxcarbazepine, eslicarbazepine, ethosuximide, and
84% of an administered dose recovered in the urine felbamate. These drugs can be used cautiously in patients
(Hermann et al., 2003). The renal clearance following with either renal or liver failure. Lamotrigine, oxcarbaze-
intravenous administration is 0.4–0.6 L/h/kg. About pine, and eslicarbazepine mostly undergo extensive phase
36% of the drug is recovered unchanged in the urine, II metabolism. Since the reactions involving phase II
and 18% as the N-acetyl metabolite. The elimination metabolism are very ubiquitous in extrahepatic tissues,
half-life is 7–11 hours. the elimination of these drugs is not significantly affected
Data provided by the manufacturer indicate that, fol- by liver disease.
lowing a single dose of 100 mg of ezogabine/retigabine, Antiepileptic drugs that are at high risk of being
the area under the curve was increased by about 30% in extracted by hemodialysis include ethosuximide,
patients with mild renal insufficiency (creatinine gabapentin, lacosamide, levetiracetam, pregabalin, and
clearance  50 to  80 mL/min) and increased by about topiramate. Dose supplementation is usually necessary
100% with moderate renal insufficiency (creatinine in these cases. Pre- and postdialysis serum concentrations
clearance  30 to  50 mL/min). A reduction of 50% in can be helpful in estimating the need for supplementation.
the total daily dose has been recommended for patients The use of antiepileptic drugs in the presence of
with a creatinine clearance  50 mL/min. There is very hepatic or renal disease is complex and requires great
430 J.J. ASCONAPÉ
familiarity with the pharmacokinetics of these agents in patients with renal failure. Clin Pharmacol Ther 26:
(Lacerda et al., 2006). Closer follow-up of the patients 306–314.
and more frequent monitoring of serum concentrations Boston Collaborative Drug Surveillance Program (1973).
are required to optimize clinical outcomes. Diphenylhydantoin side effects and serum albumin levels.
Clin Pharmacol Ther 14: 529–532.
Britzi M, Perucca E, Soback S et al. (2005). Pharmacokinetic
REFERENCES and metabolic investigation of topiramate disposition in
healthy subjects in the absence and in the presence of
Adkins JC, Noble S (1998). Tiagabine: a review of its pharma- enzyme induction by carbamazepine. Epilepsia 46:
codynamic and pharmacokinetic properties and therapeutic 378–384.
potential in the management of epilepsy. Drugs 55: Bruni J, Wang LH, Marbury TC et al. (1980). Protein binding
437–460. of valproic acid in uremic patients. Neurology 30:
Adler DS, Martin E, Gambertoglio JG et al. (1975). 557–559.
Hemodialysis of phenytoin in a uremic patient. Clin Buchanan RA, Kinkel AW, Smith TC (1973). The absorption
Pharmacol Ther 18: 65–69. and excretion of ethosuximide. Int J Clin Pharmacol 7:
Affrime M, Reidenberg MM (1975). The protein binding of 213–218.
some drugs in plasma from patients with alcoholic liver dis- Buchanan R, Bockbrader H, Chang T et al. (1996). Single- and
ease. Eur J Clin Pharmacol 8: 267–269. multiple-dose pharmacokinetics of zonisamide. Epilepsia
Ahmed SN, Siddiqui ZA (2006). Antiepileptic drugs and liver 37 (Suppl. 5): 172.
disease. Seizure 15: 156–164. Butler TC (1957). The metabolic conversion of 5,5-
Almeida L, Soares-da-Silva P (2003). Safety, tolerability and diphenylhydantoin to 5(p-hydroxyphenyl)-5 phenyl-
pharmacokinetic profile of BIA 2-093, a novel putative hydantoin. J Pharmacol Exp Ther 119: 1–11.
antiepileptic agent, during first administration to humans. Cato A III, Gustavson LE, Qian JX et al. (1998). Effect of renal
Drugs R D 4: 269–284. impairment on the pharmacokinetics and tolerability of tia-
Almeida L, Soares-da-Silva P (2007). Eslicarbazepine acetate gabine. Epilepsia 39: 43–47.
(BIA 2-093). Neurotherapeutics 4: 88–96. Cawello W, Kropeit D, Schiltmeyer B et al. (2004). Food does
Almeida L, Falcao A, Maia J et al. (2005). Single-dose and not affect the pharmacokinetics of SPM-927 (Abstract).
steady-state pharmacokinetics of eslicarbazepine acetate Epilepsia 45 (Suppl. 7): 307.
(BIA 2-093) in healthy elderly and young subjects. J Clin Chetty M, Sarkar P, Aggarwal A et al. (2003). Carbamazepine
Pharmacol 45: 1062–1066. poisoning treatment with haemodialysis. Nephrol Dial
Almeida L, Potgieter JH, Maia J et al. (2008). Transplant 18: 220–221.
Pharmacokinetics of eslicarbazepine acetate in patients Dasgupta A, Jacques M, Malhotra D (1996). Diminished pro-
with moderate hepatic impairment. Eur J Clin Pharmacol tein binding capacity of uremic sera for valproate following
64: 267–273. hemodialysis: the role of free fatty acids and uremic com-
Alvin J, McHorse T, Hoyumpa A et al. (1975). The effect of pounds. Am J Nephrol 16: 327–333.
liver disease in man on the disposition of phenobarbital. Depner TA, Gulyassy PF (1980). Plasma protein binding in
J Pharmacol Exp Ther 192: 224–235. uremia: extraction and characterization of an inhibitor.
Asconapé J, D’Souza J (2004). No dose adjustments of oxcar- Kidney Int 18: 86–94.
bazepine required in mild or moderate hepatically impaired Dickins M, Chen C (2002). Lamotrigine: chemistry, biotrans-
patients. Epilepsia 45 (Suppl. 7): 305. formation and pharmacokinetics. In: RH Levy,
Asconapé JJ, Penry JK (1982). Use of antiepileptic drugs in the RH Mattson, BS Meldrum et al. (Eds.), Antiepileptic
presence of liver and kidney diseases: a review. Epilepsia Drugs. 5th edn. Lippincott, Williams and Wilkins,
23 (Suppl. 1): S65–S79. Philadelphia, pp. 370–379.
Ben-Menachem E (2004). Pregabalin pharmacology and its Dromgoole SH (1973). The effect of haemodialysis on the
relevance to clinical practice. Epilepsia 45 (Suppl. 6): 13–18. binding capacity of albumin. Clin Chim Acta 46: 469–472.
Blackburn-Munro G, Dalby-Brown W, Mirza NR et al. (2005). Fillastre JP, Taburet AM, Fialaire A et al. (1993).
Retigabine: chemical synthesis to clinical application. CNS Pharmacokinetics of lamotrigine in patient with renal
Drug Rev 11: 1–20. impairment: influence of haemodialysis. Drugs Exp Clin
Blaschke TF, Meffin PJ, Melmon KL et al. (1975). Influence of Res 19: 25–32.
acute viral hepatitis on phenytoin kinetics and protein bind- French J (2001). Use of levetiracetam in special populations.
ing. Clin Pharmacol Ther 17: 685–691. Epilepsia 42 (Suppl. 4): 40–43.
Blum RA, Sica DA, Comstock TJ et al. (1994). Frenchie D, Bastani B (1998). Significant removal of phenyt-
Pharmacokinetics of gabapentin in subjects with various oin during high flux dialysis with cellulose triacetate dia-
degrees of renal function. Clin Pharmacol Ther 58: 154–159. lyzer. Nephrol Dial Transplant 13: 817–818.
Bochner F, Hooper WD, Sutherland JM et al. (1973). The renal Glue P, Sulowicz W, Colucci R et al. (1997). Single-dose phar-
handling of diphenylhydantoin and 5(p-hydroxyphenyl)-5- macokinetics of felbamate in patient with renal dysfunc-
phenyl-hydantoin. Clin Pharmacol Ther 14: 791–796. tion. Br J Clin Pharmacol 44: 91–93.
Borga O, Hoppel C, Odar-Cederl€of I et al. (1979). Plasma Grant SM, Heel RC (1991). Vigabatrin: a review of its pharma-
levels and renal excretion of phenytoin and its metabolites codynamic and pharmacokinetic properties and therapeutic
USE OF ANTIEPILEPTIC DRUGS IN HEPATIC AND RENAL DISEASE 431
potential in epilepsy and disorders of motor control. Drugs Kutt H, Winters W, Scherman R et al. (1964).
4: 889–926. Diphenylhydantoin and phenobarbital toxicity. The role
Gugler R, Azarnoff DL (1976). Drug protein binding and the of liver disease. Arch Neurol 11: 649–656.
nephrotic syndrome. Clin Pharmacokinet 1: 25–35. Lacerda G, Krummel T, Sabourdy C et al. (2006). Optimizing
Gugler R, von Unruh GE (1980). Clinical pharmacokinetics of therapy of seizures in patients with renal or hepatic dys-
valproic acid. Clin Pharmacokinet 5: 67–83. function. Neurology 67 (Suppl. 4): S28–S33.
Gugler R, Shoeman DW, Huffman DH et al. (1975). Lau AH, Gustavson LE, Sperelakis R et al. (1997).
Pharmacokinetics of drugs in patients with the nephrotic Pharmacokinetics and safety of tiagabine in subjects
syndrome. J Clin Invest 55: 1182–1183. with various degrees of hepatic function. Epilepsia 38:
Gulyassy PF, Bottini AT, Jarrard EA et al. (1983). Isolation of 445–451.
inhibitors of ligand: albumin-binding from uremic body Lee CS, Wang LH, Marbury TC et al. (1980). Hemodialysis
fluids and normal urine. Kidney Int 24 (Suppl. 16): S238–S242. clearance and total body elimination of carbamazepine dur-
Haegele KD, Huebert ND, Ebel M et al. (1988). ing chronic hemodialysis. Clin Toxicol 17: 429–438.
Pharmacokinetics of vigabatrin: implications of creatinine Lee CS, Marbury TC, Perchalski RT et al. (1982).
clearance. Clin Pharmacol Ther 44: 558–565. Pharmacokinetics of primidone elimination in uremic
Hainzl D, Parada A, Soares-da-Silva P (2001). Metabolism of patients. J Clin Pharmacol 22: 301–308.
two new antiepileptic drugs and their principal metabolites Leppik IE (2004). Zonisamide: chemistry, mechanism of
S(þ)- and R(-)-10,11-dihydro-10-hydroxy carbamazepine. action, and pharmacokinetics. Seizure 13 (Suppl. 1): S5–S9.
Epilepsy Res 44: 197–206. Letter JM, Mellk H, Louis S et al. (1971). Diphenylhydantoin
Heipertz R, Guthoff A, Bernhardt W (1979). Primidone metab- metabolism in uremia. N Engl J Med 285: 648–652.
olism in renal insufficiency and acute intoxication. J Neurol Lloyd P, Flesch G, Dieterle W (1994). Clinical pharmacology
221: 101–104. and pharmacokinetics of oxcarbazepine. Epilepsia 35
Hermann R, Ferron GM, Erb K et al. (2003). Effects of age and (Suppl. 3): S10–S13.
sex on the disposition of retigabine. Clin Pharmacol Ther Lunde PKM, Rane A, Yaffee SJ et al. (1970). Plasma protein
73: 61–70. binding of diphenylhydantoin in man: interaction with
Hooper WD, Bochner F, Eadie MJ et al. (1974). Plasma protein other drugs and the effect of temperature and plasma dilu-
binding of diphenylhydantoin. Effects of sex hormones, tion. Clin Pharmacol Ther 11: 846–855.
renal and hepatic disease. Clin Pharmacol Ther 15: 276–282. McAuliffe JJ, Sherwin AL, Leppik IE et al. (1977). Salivary
Hooper DW, Dubetz DK, Bochner F et al. (1975). Plasma pro- levels of anticonvulsants: a practical approach to drug mon-
tein binding of carbamazepine. Clin Pharmacol Ther 17: itoring. Neurology 27: 409–413.
433–440. McLean MJ (1994). Clinical pharmacokinetics of gabapentin.
Hovinga CA (2003). SPM-927 (Schwarz Pharma). IDrugs 6: Neurology 44 (Suppl. 5): S17–S22.
479–485. Maia J, Almeida L, Falcao A et al. (2008). Effect of renal
Ijiri Y, Fukuda F, Suzuki K et al. (2004). Dialyzability of the impairment on the pharmacokinetics of eslicarbazepine
antiepileptic drug zonisamide in patients undergoing acetate. Int J Clin Pharmacol Ther 46: 119–130.
hemodialysis. Epilepsia 45: 924–927. Marbury TC, Lee CC, Perchalski RJ (1981). Hemodialysis
Israni RK, Kasbekar N, Haynes K et al. (2006). Use of antie- clearance of ethosuximide in patients with chronic renal
pileptic drugs in patients with kidney disease. Semin Dial failure. Am J Hosp Pharm 38: 1757–1760.
19: 408–416. Marquardt ED, Ishisaka DY, Barra KK et al. (1992). Removal
Kandrotas RJ, Love JM, Gal P et al. (1990). The effect of of ethosuximide and phenobarbital by peritoneal dialysis in
hemodialysis and hemoperfusion on serum valproic acid a child. Clin Pharm 11: 1030–1031.
concentration. Neurology 40: 1456–1458. Martin E, Gambertoglio JG, Adler DS et al. (1977). Removal
Kane SL, Constantiner M, Staubus AE et al. (2000). High-flux of phenytoin by hemodialysis in uremic patients. JAMA
hemodialysis without hemoperfusion is effective in 238: 1750–1753.
acute valproic acid overdose. Ann Pharmacother 34: 1146–1151. Mauro LS, Mauro VF, Bachmann KS et al. (1989). Accuracy
Kielstein JT, Schwarz A, Arnavaz A et al. (2002). High-flux of two equations in determining normalized phenytoin con-
hemodialysis: an effective alternative to hemoperfusion centrations. DICP 23: 64–68.
in the treatment of carbamazepine intoxication. Clin Odar-Cederl€ of I, Lunde P, Sjoqvist F (1970). Abnormal phar-
Nephrol 57: 484–486. macokinetics of phenytoin in a patient with uraemia.
Kielstein RJ, Woywodt A, Schumann G et al. (2003). Lancet 2: 831.
Efficiency of high-flux hemodialysis in the treatment of Odar-Cederl€ of I, Borga O (1974). Kinetics of diphenylhydan-
valproic acid intoxication. J Toxicol Clin Toxicol 41: toin in uraemic patients: consequences of decreased plasma
873–876. protein binding. Eur J Clin Pharmacol 7: 31–37.
Kinniburgh DW, Boyd ND (1981). Isolation of peptides from Olsen GD, Bennett WM, Porter GA (1975). Morphine and phe-
uremic plasma that inhibit phenytoin binding to plasma nytoin binding to plasma proteins in renal and hepatic fail-
proteins. Clin Pharmacol Ther 30: 276–280. ure. Clin Pharmacol Ther 17: 677–684.
Klotz U, Rapp T, Muller WA (1978). Disposition of valproic Palmer BF (2000). Effectiveness of hemodialysis in the extra-
acid in patients with liver disease. Eur J Clin Pharmacol 13: corporeal therapy of phenobarbital overdose. Am J Kidney
55–60. Dis 36: 640–643.
432 J.J. ASCONAPÉ
Palmer KJ, McTavisk D (1993). Felbamate: a review of its Schuerer DJ, Brophy PD, Maxvold NJ et al. (2000). High-
pharmacodynamic and pharmacokinetic properties, and efficiency dialysis for carbamazepine overdose. J Toxicol
therapeutic efficacy in epilepsy. Drugs 45: 1041–1065. Clin Toxicol 38: 321–323.
Patsalos PN (2004). Clinical pharmacokinetics of levetirace- Shumaker RC, Fantel C, Kelton E et al. (1990). Evaluation of
tam. Clin Pharmacokinet 43: 707–724. the elimination of [C14] felbamate in healthy men.
Perucca E, Cloyd J, Critchley D et al. (2008). Rufinamide: Epilepsia 31: 642.
clinical pharmacokinetics and concentration-response Steele WWH, Lawrence JR, Elliot HL et al. (1979).
relationships in patients with epilepsy. Epilepsia 49: Alterations of phenytoin protein binding with in vivo hae-
1123–1141. modialysis in dialysis encephalopathy. Eur J Clin
Plosker GL, Scott LJ (2006). Retigabine in partial seizures. Pharmacol 15: 69–71.
CNS Drugs 20: 601–608. Stern EL (1977). Possible phenylethylmalondiamide (PEMA)
Porto I, John EG, Heilliezer J (1997). Removal of phenobarbi- intoxication. Ann Neurol 2: 356–357.
tal during continuous cycling peritoneal dialysis in a child. Streete JM, Berry DJ, Jones JA et al. (1990). Clearance of phe-
Pharmacotherapy 17: 832–835. nylethylmalonamide during haeamodialysis of a patient
Posner J, Cohen AF, Land G et al. (1989). The pharmacokinet- with renal failure. Ther Drug Monit 12: 281–283.
ics of lamotrigine (BW430C) in healthy subjects with Tang BK, Inaba T, Kalow W (1977). N-hydroxyphenobarbital.
unconjugated hyperbilirubinemia. Br J Clin Pharmacol The major metabolite of phenobarbital in man. Fed Proc 36:
28: 117–120. 966.
Radtke RA (2001). Pharmacokinetics of levetiracetam. Tapolyai M, Campbell M, Dailey K et al. (2002).
Epilepsia 42 (Suppl. 4): 24–27. Hemodialysis is as effective as hemoperfusion for drug
Randinitis EJ, Posvar EL, Alvey CW et al. (2003). removal in carbamazepine poisoning. Nephron 90: 213–215.
Pharmacokinetics of pregabalin in subjects with Tiula E, Haapanen EJ, Neuvonen PJ (1987). Factors affecting
various degrees of renal function. J Clin Pharmacol 43: serum protein binding of phenytoin, diazepam and propran-
277–283. olol in acute renal diseases. Int J Clin Pharmacol Ther
Rane A, Lunde PKM, Jalling B et al. (1971). Plasma protein Toxicol 25: 469–475.
binding of diphenylhydantoin in normal and hyperbilirubi- Van Peer AP, Belpaire FM, Rosseel MT et al. (1981).
nemic infants. J Pediatr 78: 877–882. Distribution of antipyrine, phenylbutazone, and phenytoin
Reidenberg MM, Affrime M (1973). Influence of disease on in experimental renal failure. Pharmacology 22: 139–145.
binding of drugs to plasma proteins. Ann N Y Acad Sci Vanholder R, Van Landschoot N, De Smet R et al. (1988).
226: 115–126. Drug protein binding in chronic renal failure: evaluation
Reidenberg MM, Drayer DE (1978). Effects of of nine drugs. Kidney Int 33: 996–1004.
renal disease upon drug disposition. Drug Metab Rev 8: Wong MO, Eldon MA, Keane WF et al. (1995). Disposition of
293–302. gabapentin in anuric subjects on hemodialysis. J Clin
Reidenberg MM, Odar-Cederl€of I, Von Bahr C et al. (1971). Pharmacol 35: 622–626.
Protein binding of diphenylhydantoin and desmethylimi- Wootton R, Soul-Lawton J, Rolan PE et al. (1997).
pramine in plasma from patients with poor renal function. Comparison of the pharmacokinetics of lamotrigine in
N Engl J Med 285: 264–267. patients with chronic renal failure and healthy volunteers.
Rouan MC, Lecaillon JB, Godbillon J et al. (1994). The Br J Clin Pharmacol 43: 23–27.
effect of renal impairment on the pharmacokinetics of Zawada ET, Nappi J, Done G et al. (1983). Advances in the
oxcarbazepine and its metabolites. Eur J Clin Pharmacol hemodialysis management of phenobarbital overdose.
47: 161–167. South Med J 76: 6–8.

You might also like