Anda di halaman 1dari 11

Epilepsia, 54(3):405414, 2013

doi: 10.1111/epi.12109

CRITICAL REVIEW AND INVITED COMMENTARY

Antiepileptic drug treatment in pregnancy: Changes in drug


disposition and their clinical implications
orn Tomson, Cecilie Johannessen Landmark, and Dina Battino
*Torbj

*Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Pharmacy and Biomedical
Science, Faculty of Health Sciences, Oslo and Akershus University College of Applied Sciences, Oslo, Norway; and
Epilepsy Center, Department of Neurophysiology, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy

Some AEDs, such as carbamazepine seem to be affected


SUMMARY
only marginally by pregnancy. Data on pharmacokinetics
Pregnancy is a state where pharmacokinetic changes during pregnancy are lacking completely for some of the
are more pronounced and more rapid than during any newer generation AEDs: pregabalin, lacosamide, retiga-
other period of life. The consequences of such bine, and eslicarbazepine acetate. Where data are avail-
changes can be far reaching, not least in the management able, the effects of pregnancy on serum concentrations
of epilepsy where the risks with uncontrolled seizures seem to vary considerably individually and are thus diffi-
during pregnancy need to be balanced against potential cult to predict. Although large-scale systematic studies of
teratogenic effects of antiepileptic drugs (AEDs). This the clinical relevance of the pharmacokinetic alterations
article aims to review the literature on gestational are lacking, prospective and retrospective case series
effects on the pharmacokinetics of older and newer have reported an association between declining serum
generation AEDs and discuss the implications for the concentrations and deterioration in seizures control. The
treatment of epilepsy in women during pregnancy. usefulness of routine monitoring of AED serum concen-
Pregnancy can affect the pharmacokinetics of AEDs at trations in pregnancy and of dose adjustments based on
any level from absorption, distribution, metabolism, to falling levels, are discussed in this review. We suggest that
elimination. The effect varies depending on the type monitoring could be important, in particular when
of AED. The most pronounced decline in serum women have been titrated to the lowest effective AED
concentrations is seen for AEDs that are eliminated by dose and serum concentration before pregnancy, and
glucuronidation (UGT), in particular lamotrigine where when that individual optimal concentration can be used as
the effect may be profound. Serum concentrations of reference.
AEDs that are cleared mainly through the kidneys, for KEY WORDS: Epilepsy, Pregnancy, Antiepileptic drugs,
example, levetiracetam, can also decline significantly. Pharmacokinetics.

The pharmacologic treatment of epilepsy is seldom epilepsy. Fetal loss has been reported in conjunction with
more challenging than during pregnancy. Treatment prolonged seizures, such as status epilepticus (Teramo &
involves drug exposure to at least two individuals: the Hiilesmaa, 1982), and frequent tonicclonic seizures dur-
mother with epilepsy and the unborn fetus(es). The mater- ing pregnancy are associated with poorer cognitive devel-
nal and fetal risks imposed by uncontrolled epileptic sei- opment of the child (Meador et al., 2009a). On the other
zures need to be balanced against the possible adverse hand, AEDs can be teratogenic, thereby increasing the
fetal effects of antiepileptic drugs (AEDs). Management risk of congenital malformations as well as of adverse
is further complicated by the fact that effects on the fetus cognitive outcomes (Tomson & Battino, 2012). Recent
can be difficult to monitor and may be irreversible. Major publications from epilepsy and pregnancy registries have
convulsive seizures (generalized tonicclonic seizures) suggested that AEDs differ in their teratogenic potential,
can be harmful to the fetus, in addition to causing medical but also that these adverse effects of AEDs on the fetus
risks and psychosocial consequences for the mother with are dose-dependent (Meador et al., 2009a; Tomson et al.,
2011; Hernandez-Diaz et al., 2012; Mawhinney et al.,
Accepted December 19, 2012; Early View publication January 29, 2013. 2012). Therefore, the usually recommended treatment
Address correspondence to Torbjorn Tomson, Department of Neuro-
logy, Karolinska University Hospital, SE 171 76 Stockholm, Sweden.
strategy is to review and possibly revise treatment well
E-mail: torbjorn.tomson@karolinska.se before conception, selecting the most appropriate AED
Wiley Periodicals, Inc. for the individual woman, taking efficacy as well as tera-
2013 International League Against Epilepsy togenic risks into account, and regardless of which drug is

405
406
T. Tomson et al.

chosen to titrate to the lowest effective dose before


pregnancy (Harden et al., 2009a; National Institute for
Clinical Excellence, 2012; Tomson & Battino, 2012). The
objectives of the further management during pregnancy
are to maintain control of in particular tonicclonic sei-
zures while at the same time keeping exposure to poten-
tially teratogenic drugs to a minimum. This demanding
task is complicated by the fact that pregnancy can signifi-
cantly affect the pharmacokinetics of AEDs with potential
consequences for seizure control as well as drug exposure
to the fetus. So, although the risk of malformations has
been assessed in relation to AED doses at the time of con-
ception (Tomson et al., 2011), or during the first trimester
(Hernandez-Diaz et al., 2012; Mawhinney et al., 2012),
the level of exposure to the fetus can change during the
course of pregnancy. In this article, we review the litera-
ture on the effects of pregnancy on the pharmacokinetics
of different AEDs and discuss the implications for the Figure 1.
treatment of epilepsy in women during pregnancy. Pharmacokinetic changes in drug disposition during pregnancy
and basic pharmacokinetic relationships. CL, total clearance;
AUC, area under the curve of a drug; F, bioavailability; t1/2, half-
Search Strategy life; k, elimination constant and Vd, volume of distribution; Css,
serum concentration at steady state, and , dose frequency).
References for this review were identified from the Epilepsia ILAE
authors files and from a PubMed search (from 1966 to July
2012) using various combinations of the following terms
(by searching as text words) pregnan*, maternal
mother, fetal, fetus with the terms *kinetics, dis- Absorption
position, plasma, utilization and the names of individ- Generally, absorption is extensive and bioavailability
ual AEDs included in the review or the terms high for most AEDs. The rate and extent of absorption
antiepileptic*, anti-epileptic*, anticonvuls*, anti- can, however, vary with the drug formulation. Gastric pH
convuls*, barbit*. and rate of emptying, and small intestine motility may all
In addition, the list of references of the retrieved articles decrease during pregnancy, leading to a decrease in the
was examined for further studies. Moreover, original absorption of AEDs (Pennell, 2003), and the pharmacoki-
research articles and previously published review articles netic parameters peak concentration (Cmax) and time to
were examined. peak concentration (Tmax) (Fig. 1). Malaise and vomiting
Only articles published in English were reviewed. The in early pregnancy are likely to affect the absorption of
review was restricted to studies in humans. AEDs taken orally. An impaired drug absorption would
result in decreased serum concentrations of AEDs and a
possibly decrease inefficacy. However, to the best of our
Mechanisms of Pregnancy- knowledge, there is only one published report of break-
Induced Alterations through seizures due to poor AED absorption during preg-
nancy. This was a single case of phenytoin malabsorption
in Drug Disposition leading to status epilepticus during pregnancy (Ramsay
The physiologic changes that take place during preg- et al., 1978). Hence, it appears that poor drug absorption
nancy can affect any level of the disposition of drugs, from is a rare cause of treatment failure during pregnancy.
absorption, distribution, metabolism, to excretion (ADME,
Fig. 1). It is important to understand the mechanisms by Distribution
which pregnancy alters the pharmacokinetics of AEDs, as During the course of the pregnancy, the blood volume pro-
this determines the possible clinical implications as will be gressively increases, with a decrease in the total serum con-
discussed in more detail below. However, every pregnancy centrations of drugs in parallel. The increase in body water
represents a singular occurrence of genetic variables and and in fat stores leads to an alteration of fat/water ratio that
environmental factors, and AEDs may interact differently may cause an increase in the volume of distribution (Vd). The
with any of these in the individual woman and also differ- overall effects of gestation-induced alterations in Vd appear
ently from pregnancy to pregnancy (Wlodarczyk et al., to be variable between individuals, but the net effect on active
2012). AED serum concentrations is generally limited.

Epilepsia, 54(3):405414, 2013


doi: 10.1111/epi.12109
407
Pharmacokinetics in Pregnancy

Serum albumin and a1-acid glycoprotein levels decline pregnancy (Sturgiss et al., 1994). Increased renal clearance can
as pregnancy progresses, and these are essential transporters affect serum concentrations of AED that are mainly excreted
for AEDs in the bloodstream. A decrease in serum albumin renally, such as gabapentin, levetiracetam, pregabalin, and
and protein binding capacity, and displacement of AEDs by vigabatrin. It has also been suggested as an important contrib-
endogenous compounds, may result in an alteration in the uting cause for the pronounced increase in clearance of lamo-
ratio between the total and the unbound concentration of trigine and its N2-glucuronide metabolite in early pregnancy
highly protein-bound drugs, for example, phenytoin, stirip- (Reimers et al., 2011; Reimers & Brodtkorb, 2012).
entol, tiagabine, and valproate, AEDs with a protein binding Most documented changes can thus be considered the
ranging from 90% to 99% (for review, see (Johannessen consequence of enzyme induction and to some extent
Landmark et al., 2012)). The total serum concentrations of increased renal clearance. Still, several issues are poorly
valproate and phenytoin have been shown to decline in par- investigated, for example, some isoenzymes have been
allel with the falling albumin levels (Yerby et al., 1990, investigated for only one drug, the effect of transport pro-
1992). The net effect of a decrease in protein binding is a fall teins during pregnancy remains to be clarified, as well as
in the total (bound plus unbound) serum concentration of effects of pregnancy on drugs that are eliminated through
the drug, whereas the unbound drug concentration may be non-CYP/UGT or multiple pathways (Anderson, 2005).
essentially unchanged. The unbound concentration is the
pharmacologically active, and also presumably the concen-
tration determining drug exposure to the fetus. A decrease Gestation-Induced Alterations
in protein binding would thus not alter the effect of treat- in Kinetics of Individual AEDs
ment, but total serum concentrations as routinely measured
Published data on some basic pharmacokinetic properties
would be misleading and underestimating the effective
and on alterations in pharmacokinetics of different AEDs dur-
concentration and the fetal exposure. Monitoring of
ing pregnancy are summarized in Table 1. Because lamotri-
free concentrations may be preferable in such situations
gine is the most extensively studied AED in this regard, data
(Johannessen & Tomson, 2006; Patsalos et al., 2008).
on its kinetics during pregnancy are reported separately and
in more detail in Table 2. A brief account of data on individ-
Metabolism
ual AEDs (in alphabetical order) is provided in the following.
The total clearance of AEDs that are metabolized by the
liver depends on the hepatic clearance. The main route is
phase I oxidative metabolism through the cytochrome P450 Carbamazepine
system (CYP). There are different CYP isoenzymes, each of Carbamazepine is approximately 75% protein bound and
which is a specific gene product with characteristic sub- is eliminated by hepatic metabolism through CYP1A2,
strate specificity, where CYP3A4, CYP2C9, CYP2C19 are CYP2C8, and CYP3A4 (Johannessen Landmark et al.,
among the most important for the metabolism of AEDs 2012). Several studies have analyzed the effect of preg-
(Johannessen & Landmark, 2010; Johannessen Landmark nancy on carbamazepine serum concentrations (Lander
& Patsalos, 2010, 2012). et al., 1981; Battino et al., 1985; Yerby et al., 1985;
Phase I CYP enzymes as well as phase II enzymes (uri- Tomson et al., 1994a,b; Bernus et al., 1995), and the results
dine glucuronyl transferases, UGTs) can be induced during are slightly conflicting. Reported declines in total concen-
pregnancy. Such enzyme induction is the clinically most trations from prepregnancy to late pregnancy have ranged
important mechanism for declining serum concentrations of from 0% to 42%. The fall in unbound concentrations has
AEDs during pregnancy, as it will result in a fall in unbound been more limited: 028% (Lander et al., 1981; Battino
as well as total serum concentrations. Enzyme induction et al., 1985; Yerby et al., 1985; Tomson et al., 1994a,b;
appears to be most pronounced for AEDs that are metabo- Bernus et al., 1995). In fact, the largest study of women on
lized through glucuronidation such as lamotrigine, monotherapy reported a very limited decline in total con-
oxcarbazepine (or rather its active MHD-derivative), and centrations during the second and third trimesters (912%),
valproate. The extent to which pregnancy affects AED whereas unbound concentrations remained essentially
metabolism varies considerably between patients and is unchanged (Tomson et al., 1994b).
therefore difficult to predict (Tomson & Battino, 2007;
Patsalos et al., 2008; Johannessen Landmark et al., 2012). Gabapentin
Gabapentin is absorbed form the gastrointestinal tract
Excretion through saturable active transportation (Gidal et al., 2000),
During pregnancy, there is a significant increase in renal but there are no data on the effects of pregnancy on its
blood flow and glomerular filtration rate, up to 5080% of the absorption. Gabapentin is not bound to serum proteins; it is
prepregnancy rate (Sturgiss et al., 1994; Pennell, 2003). This not metabolized but eliminated by renal excretion (Johann-
process starts shortly after conception and continues throughout essen Landmark et al., 2012). No studies have followed the
the second trimester and then declines in the last part of the course of gabapentin serum concentrations throughout

Epilepsia, 54(3):405414, 2013


doi: 10.1111/epi.12109
408
T. Tomson et al.

Table 1. Pharmacokinetic data and effects of pregnancy on serum concentrations of antiepileptic drugs
Pregnancies
with published
pharmacokinetic Protein Effect of pregnancy
References Antiepileptic drug data (n) Elimination binding on serum concentrations
Bardy et al. (1982); Battino et al. Carbamazepine 157 Almost completely metabolized, About 75% Decrease by 042%
(1985); Bernus et al. (1995); the most important pathway
Lander and Eadie (1991); being the formation of
Tomson et al. (1994a); and carbamazepine epoxide
Yerby et al. (1992, 1985)

Ohman et al. (2005) Gabapentin 3 Unchanged via kidney Not bound No decline
Johannessen et al. (2005); Levetiracetam 45 Two thirds unchanged in Not bound Decrease by 4060%
Pennell et al. (2005); urine, one-third
Tomson et al. (2007); and metabolized by peripheral
Westin et al. (2008) hydrolysis
Christensen et al. (2006); Oxcarbazepine 27 Pre-systemic 10-keto reduction About 40% Decrease by 3038%
Mazzucchelli et al. (2006); to the two enantiomers of
and Petrenaite et al. (2009) MHD than glucuronidation
of the active metabolite
MHD via UGT
Bardy et al. (1987); Battino Phenytoin 307 Mainly via hepatic microsomal- 8590% Decrease by 5661%
et al. (1982); Dansky et al. mixed function oxidase
(1982); Lander and Eadie (1991); reaction, with subsequent
and Tomson et al. (1994a) glucuronidation and
excretion of hydroxylated
derivative
Bardy et al. (1982); Battino et al. Phenobarbital 56 Hepatic oxidation, 5060% Decrease by 5055%
(1984a); Lander and Eadie (1991); glucosidation, hydroxylation
and Yerby et al. (1992) and subsequent conjugation
Battino et al. (1984a); and Rating Primidone-derived 23 Decreased by 70%
et al. (1982) phenobarbital

Ohman et al. (2009, 2002); and Topiramate 34 Mainly unchanged in urine, About 15% Decrease by 1340%
Westin et al. (2009) 2030% hepatic
biotransformation
Battino et al. (1982); Lander and Valproate 68 Hepatic metabolism, 8595% Decrease by 028%
Eadie (1991); Omtzigt et al. mainly by glucuronidation
(1992); Philbert et al. (1985);
and Yerby et al. (1992)
Kawada et al. (2002); and Zonisamide 2 Mainly hepatic 4060% Decreased by 4050%
Oles and Bell (2008); biotransformation,
1530% unchanged in urine

pregnancy. Available data are limited to maternal plasma Lamotrigine is by far the most extensively studied newer-
concentrations at delivery and 3 weeks after delivery in generation AED in conjunction with pregnancy, and the
three women who were taking gabapentin in combination results are summarized in Table 2. In patients taking lamo-

with other AEDs (Ohman et al., 2005). These limited data trigine, monotherapy serum concentrations usually decline
do not suggest lower gabapentin levels in late pregnancy markedly as pregnancy progresses. A decrease in serum
compared to 2 to 3 weeks after. The only potential antici- concentration can be seen already in the first trimester but is
pated effects of pregnancy on its kinetics are those related to most marked in the midthird trimester (Ohman et al.,
changes in glomerular filtration. The reported few cases, 2000; Tran et al., 2002; de Haan et al., 2004; Pennell et al.,
however, do not suggest any major effects on the plasma 2004; Petrenaite et al., 2005; Reimers et al., 2011). On

concentration (Ohman et al., 2005) (Table 1). average, lamotrigine serum concentrations decrease by 50
60%, but an even more pronounced decline has been
Lamotrigine reported in individual patients (Tomson et al., 1997). The
The serum protein binding of lamotrigine is approxi- effects of pregnancy on lamotrigine disposition, however,
mately 55% (Fitton & Goa, 1995). The drug is metabolized vary considerably between individuals and are thus difficult
mainly by glucuronidation catalyzed by UDP-glucuronosyl- to predict (Petrenaite et al., 2005; Franco et al., 2008; Pen-
transferase (UGT), UGT1A4 and 2B7 (Hussein & Posner, nell et al., 2008). The effects are much less pronounced in
1997). women taking lamotrigine in combination with valproate

Epilepsia, 54(3):405414, 2013


doi: 10.1111/epi.12109
Table 2. Effects of pregnancy on the disposition of lamotrigine
No. of
pregnancies on LTG Time of
monotherapy/total sampling after
Authors pregnancies Data presented as Preconception Trimester 1 Trimester 2 Trimester 3 delivery Postpartum
Tran 2/14 Mean apparent clearance 1.2 0.37 1.97 0.53 2.31 0.91 2.26 0.94 236 weeks 1.22 0.54
et al. (2002) (L/[kg/day])SD
% change apparent >65% p < 0.05 >65% p < 0.05 vs.
clearance vs. PC and PP PC and vs. PP
Fotopoulou 9/9 Median dose/serum 39 (3941) 77 (68154) 92 (76167) 97 (74110) On average 3 weeks 35 (3536)
et al. (2009) concentration
(2575% percentile)
197% NS 236% p < 0.05 248% p < 0.05
Pennell 14/14 Apparent 52.9 20.8 89 41 133 71 171 100 212 weeks 66 29
et al. (2004)a clearance mg/(mg/L) (from figure)
% change apparent 191 107 249 142 361 194 150 69
clearance
Reimers 17/21 Mean doses 244 144 342 180 At least 4 weeks
et al. (2011) (mg/day)  SD
% change mean >34% vs. PC
doses
Petrenaite 11/11 Mean ratio LTG 63.5 30.8 46.7 18.3 22.1 5.4 21.7 7.1 6 weeks (from 70 10
et al. (2005) plasma level-to-dose
(lmol/l/mg)  SD figure)
27% NS 65% p < 0.01 66% p < 0.01
b
Ohman 8/8 Mean 2-N-GLUC/LTG 1.02 0.27 Up to 3 months 0.4 0.06
et al. (2008b) ratio
% change 2-N-GLUC/LTG 154% >PP p < 0.01
ratio

Ohman 17/17b Mean dose/plasma 227.1 74 At least 1 month 66.5 17.9
et al. (2008a) concentrations
% change mean 250% >PP
dose/plasma
concentration
LTG 2-N-glucuronide/LTG 0.349 0.141
concentrations lmol/L
147% >PP
de Haan 12/12 Weeks 110 Weeks 1120 Weeks 2130 Weeks 3140 PP
et al. (2004) Mean level-to-dose 82% 14% 51% 14% 40% 8% 97% 15% 48% 10%
ratios (presented as
percentage of
baseline  SD) for
consecutive
10-week periods
LTG, lamotrigine; PC, preconception; PP, postpartum; 2-N-GLUC, lamotrigine-2-Nglucuronide.
a
Post hoc analyses of % change of apparent clearance: first trimester versus second trimester: p < 0.0001; first trimester versus third trimester: p < 0.0001; first trimester versus postpartum: p < 0.03; second tri-
mester versus third trimester: p < 0.04; second trimester versus postpartum: p < 0.0001; third trimester versus postpartum: p < 0.0001.
b
Pregnancies on LTG monotherapy (or without concomitant use of other drugs known to interact with lamotrigine).
Pharmacokinetics in Pregnancy

Epilepsia, 54(3):405414, 2013


409

doi: 10.1111/epi.12109
410
T. Tomson et al.

(Tomson et al., 2006). Although the dose-to-plasma con- cies, confirm that serum concentrations of MHD decline by
centration ratio of lamotrigine increased from baseline to on average 3040%, hence slightly less than what is seen for
midgestation by 295% in women on lamotrigine monothera- lamotrigine (Christensen et al., 2006; Mazzucchelli et al.,
py; the increase was only 60% among women on lamotri- 2006; Petrenaite et al., 2009). An increase in MHD serum
gine in combination with valproate. concentrations was observed within 78 days after delivery
The decline in lamotrigine levels is probably mainly due (Mazzucchelli et al., 2006).
to enhanced metabolic elimination by induction of the
UGT. This hypothesis is supported by observations of Phenytoin
increased ratios in late pregnancy between the lamotrigine- Phenytoin is approximately 90% bound to serum pro-
2-Nglucuronide metabolite and lamotrigine in plasma and teins. It is eliminated by oxidative metabolism through

in urine (Ohman et al., 2008a,b). However, a more detailed CYP2C9 and CYPC19 (Johannessen Landmark et al.,
analysis of the ratio lamotrigine-2-Nglucuronide/lamotri- 2012). Pregnancy can thus affect the disposition of phenyt-
gine throughout pregnancy suggests that an enhanced renal oin by decrease in protein binding as well as by induced
excretion may contribute in particular to the fall in lamotri- metabolism. The decrease of phenytoin serum concentra-
gine concentrations in early pregnancy (Reimers et al., tions generally starts during the first trimester and becomes
2011). Regardless of whether the mechanism is by increased more evident in the third when total concentrations are
hepatic or renal clearance, the net effect is a decline on total decreased by 5561%, on average. However, the unbound
as well as unbound lamotrigine concentrations. and pharmacologically active concentrations decline to a
Serum lamotrigine concentrations return rapidly to prep- lesser extent, by 1631% (Battino et al., 1982; Dansky
regnancy values after pregnancy. The process starts within et al., 1982; Bardy et al., 1987; Yerby et al., 1992; Tom-
days after delivery and is complete 2 to 3 weeks postpartum son et al., 1994a).

(Ohman et al., 2000; Tran et al., 2002; de Haan et al.,
2004). Phenobarbital
Phenobarbital is approximately 50% bound to serum pro-
Levetiracetam teins, and is eliminated through metabolism by CYP2C19
Levetiracetam is not bound to serum proteins and its and 2E1. Phenobarbital levels have been reported to decline
elimination is primarily renal, although nonoxidative by 5055% during pregnancy (Battino et al., 1984b; Yerby
metabolism by hydrolysis accounts for approximately 30% et al., 1990), whereas the decline has been suggested to be
of its elimination. (Patsalos, 2000; Radtke, 2001). more pronounced, up to 70%, for primidone-derived pheno-
Two reports based on 15 (Tomson et al., 2007) and 21 barbital (Rating et al., 1982; Battino et al., 1984b).
(Westin et al., 2008) pregnancies observed a significant
decline in plasma concentrations during pregnancy by 40% Topiramate
to 60% at the end of pregnancy compared to baseline levels Topiramate is only 15% bound to serum proteins and
before or after pregnancy. A decrease in serum concentra- eliminated mainly unchanged via the kidneys, but a small
tions, or increase in apparent clearance, is observed already fraction undergoes hepatic CYP-dependent metabolism
in the first trimester (Pennell et al., 2005). As with lamotri- (Langtry et al., 1997).
gine, the effects of pregnancy on levetiracetam disposition Two studies have assessed the effects of pregnancy on to-
vary considerably between individuals (Westin et al.,
piramate serum concentrations (Ohman et al., 2009; Westin
2008). Serum concentrations have been found to increase et al., 2009). These reports suggest an average decline in to-
rapidly after delivery to reach prepregnancy levels within piramate levels of 30% to 40% during the third trimester
the first week (Westin et al., 2008). Although the mecha- compared with before or after pregnancy, but with a consid-
nisms have not been studied in detail, it is likely that the erable interindividual variation (Table 1). These effects are
decline in levetiracetam levels is caused by a combination most likely explained by increased renal excretion.
of an increased renal elimination and enhanced enzymatic
hydrolysis. Valproate
Valproate is highly protein bound, approximately 90%,
Oxcarbazepine and is cleared through hepatic metabolism, mainly by
Oxcarbazepine is a prodrug, which is almost completely glucuronidation through UGT1A3 and 2B7 in addition to
metabolized to mono-hydroxycarbazepine (MHD). MHD is several CYPs. Data on the effects of pregnancy on the
responsible for the effects of oxcarbazepine. The protein disposition of valproate are limited. Total concentrations
binding of MHD is about 40%, and it is cleared from the have been reported to fall in late pregnancy by up to 40%
human body mainly by glucuronidation (May et al., 2003). compared with before pregnancy, but this appears to be due
Because its elimination is by the same metabolic route as mainly to decreased protein binding, since unbound valpro-
for lamotrigine, significant effects of pregnancy can be ate concentrations remain essentially unchanged (Philbert
expected. Three studies, comprising altogether 27 pregnan- et al., 1985; Omtzigt et al., 1992; Yerby et al., 1992).
Epilepsia, 54(3):405414, 2013
doi: 10.1111/epi.12109
411
Pharmacokinetics in Pregnancy

Zonisamide in serum AED concentrations in an individual patient is


The serum protein binding of zonisamide is approxi- accompanied by a change in efficacy, and in general an
mately 60%. Zonisamide is extensively metabolized by increased risk of seizures. The usefulness of serum level
acetylation, glucuronide conjugation, and oxidation, and is monitoring and the need to adjust the dose to maintain sta-
eliminated also partly by renal excretion (Seino et al., 1995; ble serum concentrations is nevertheless often questioned
Perucca & Bialer, 1996). (Scottish Intercollegiate Guidelines Network, 2003). Such
There are no systematic studies of the kinetics of zonisa- a position would assume either that pregnancy as such
mide in pregnancy. In a single reported case, zonisamide makes patients less likely to have seizures or more respon-
levels were followed regularly from the fifth gestational sive to the drug treatment, or that the woman enters preg-
week to the end of pregnancy. The patient received 200 mg/ nancy with a higher AED dose than necessary. There is no
day of zonisamide as monotherapy until week 29, when the evidence whatsoever for the former, and the latter is also
dose was increased to 300 mg/day, since plasma concentra- unlikely if the general management policy to titrate to the
tions decreased from values ranging between 7.5 and 10.1 lowest effective AED dose before pregnancy has been fol-
(from week 5 to week 22) to 4.4 lg/ml in week 27 (Oles & lowed.
Bell, 2008). Clinical observations of lamotrigine and oxcarbazepine
Hence, no firm conclusions can be drawn concerning ges- pregnancies also seem to support that the decline in serum
tation-related alterations in zonisamide kinetics, but induc- concentrations is associated with an increased risk of sei-
tion of its metabolism might occur. zures. Increase in lamotrigine dose was considered to be
needed in 11 of 12 pregnancies in one series (Tran et al.,
Other newer AEDs 2002), and dose adjustment in all 14 lamotrigine monothera-
To the best of our knowledge no published data exist on py pregnancies in another (Pennell et al., 2004). Seizure
the pharmacokinetics of felbamate, pregabalin, tiagabine, aggravation was observed in 9 of 12 cases of pregnancies
vigabatrin, lacosamide, retigabine, perampanel, or from a Dutch series (de Haan et al., 2004) and in 5 of 11
eslicarbazepine acetate during pregnancy. pregnancies from Denmark, all of whom were taking lamo-
trigine monotherapy (Petrenaite et al., 2005). Low concen-
trations of the active oxcarbazepine metabolite were
Clinical Implications and associated with emergence of seizures in two of four in an
Italian series (Mazzucchelli et al., 2006). Another Danish
Conclusions case series found that changes in oxcarbazepine disposition
It is thus undisputable that pregnancy can affect the phar- were often associated with worsening in seizure frequency
macokinetics of AEDs and that this effect varies depending in a study of 13 pregnancies, 10 of which were on monother-
on the type of AED. For some, for example, lamotrigine, the apy (Petrenaite et al., 2009), although others have failed to
effect is profound, whereas other AEDs, for example, carba- find an association between changes in serum concentra-
mazepine, seem to be affected only marginally. For some of tions of the active metabolite of oxcarbazepine and seizure
the newer generation AEDs, data are scarce or even lacking control (Christensen et al., 2006). In the prospective obser-
completely. However, some general assumptions can be vational EURAP study, the dosage was increased more
made based on available data. A pronounced decline in often in pregnancies with monotherapy with lamotrigine or
serum concentrations can be expected for AEDs that are oxcarbazepine compared with other treatments (The
eliminated by glucuronidation (UGT). This might be EURAP study group, 2006). The best evidence for an
relevant for eslicarbazepine acetate and possibly also for association between declining serum concentrations and
retigabine (ezogabine) (Bialer et al., 2009). Plasma concen- deterioration in seizure control comes from a prospective
trations of AEDs that are cleared mainly through the study of 36 pregnancies with lamotrigine monotherapy, of
kidneys, for example, gabapentin, pregabalin, and lacosa- which 39% experienced an increase in seizure frequency
mide, can also be expected to decline significantly. during pregnancy (Pennell et al., 2008). The risk was
Perampanel is protein bound to a high degree, 96%, and significantly increased when the lamotrigine serum concen-
metabolized through CYP3A4 (Bialer et al., 2009). A trations dropped by >35% from the individual optimal
displacement can thus be expected during pregnancy, lead- serum concentration before pregnancy.
ing to declining total concentrations, whereas the active Although there seems to be an association between
unbound concentrations could remain fairly stable. For declining serum concentrations of AEDs and deterioration
AEDs whose pharmacokinetic properties are affected, the in seizure control, many women remain seizure free despite
extent varies between individuals and is therefore difficult lower drug concentrations. Some therefore advise against
to predict. Polytherapy makes it even more difficult to drug level monitoring and also against increasing the dose
predict the course of AED concentrations during pregnancy. unless the patient has had deterioration in seizure control
The key issue is if these alterations in serum concentra- (Scottish Intercollegiate Guidelines Network, 2003). This
tions are of clinical relevance. It appears obvious that a fall position rests on the assumption that risks associated with
Epilepsia, 54(3):405414, 2013
doi: 10.1111/epi.12109
412
T. Tomson et al.

an increase of the AED dose outweigh seizure-related pine, and possibly topiramate. The value of monitoring
maternal and fetal risks. We question this assumption. A during pregnancy largely depends on whether the patients
dose increase should aim primarily at maintaining the optimal serum concentration has been determined before
womans individual optimal serum concentration, not pregnancy. The sampling frequency during pregnancy
increase it. In general, organogenesis is already completed depends on the AED as well as on knowledge of the individ-
at the stage of pregnancy when such dose adjustments are ual womans prior sensitivity to dose changes. Sampling
considered, and a dose adjustment would not increase the monthly could be justified for a woman on lamotrigine, who
risk of birth defects. It can be argued that drug exposure later has been carefully titrated to the lowest effective dose (and
in pregnancy can be of relevance for cognitive development. serum concentration) prior to pregnancy, and where a lower
However, adverse effects of AEDs on cognitive outcomes dose in the past has been associated with major seizures. For
have been shown convincingly only for valproate (Meador such women, we would also propose a proactive approach
et al., 2009a), and this is an AED where serum concentra- with dose adjustments before breakthrough seizures, aiming
tions of the active unbound drug is normally unchanged. at maintaining the optimal prepregnancy levels. Less fre-
Seizures probably do not induce birth defects, and a single quent sampling could on the other hand be considered in a
seizure is unlikely to cause significant harm to the fetus. woman who is on a drug with less pronounced gestational
However, five or more tonicclonic seizures during preg- changes in kinetics, who has been seizure free for a long per-
nancy have been associated with poorer cognitive develop- iod prior to pregnancy, and for whom it is possible that the
ment of the child (Adab et al., 2004; Meador et al., 2009b), used dose could be higher than necessary for seizure control.
and a single seizure in a previously seizure free woman can In such one would also be less inclined to increase the dose
have a major impact on that womans everyday life. In based only on a fall in serum concentration. If the AED dose
addition, although rarely, a single tonicclonic seizure can has been increased during pregnancy, one needs to be aware
even be fatal. The Confidential Enquiries into Maternal of the rapid reversal of the kinetics after delivery: dose
Deaths in the United Kingdom noted that of 261 reductions may be necessary during the first few days after
maternal deaths, 14 women died of epilepsy, 5.4% of all delivery to avoid toxicity.
maternal deaths (Cantwell et al., 2011). The potential In conclusion, pregnancy can have profound effects on
consequences of unnecessary seizures are thus significant. AED pharmacokinetics. Declining serum concentrations is
Admittedly, the evidence for an association between likely to have an impact on the efficacy of the treatment,
pharmacokinetic alterations and deterioration in seizure although for obvious reasons, class I evidence is lacking.
control during pregnancy is circumstantial. Class I evidence Drug level monitoring should nevertheless be considered
would require a randomized controlled trial. However, and decisions on dose adjustments made on an individual
based on the available data and on common sense, it is the basis. If monitoring is considered for highly protein-bound
opinion of the present authors that it would be unethical to AEDs, such as phenytoin and valproate, free concentrations
randomize pregnant women to having their AED dose are preferred. The value of monitoring relies on comparison
guided by clinical judgment only or by clinical judgement with the individual optimal serum concentration established
supported by serum concentration data in settings where before pregnancy rather than with general so-called refer-
drug level monitoring is available. We find it difficult to see ence ranges.
the true equipoise in the two management alternatives.
The evidence-based practice parameter update of the
American Academy of Neurology and American Epilepsy Disclosure
Society concluded that monitoring of lamotrigine, carba-
TT has received speakers honoraria or research funding from Bial,
mazepine, and phenytoin levels during pregnancy should be Eisai, GSK, UCB, Sanofi-Aventis, and Novartis. CJL declares no conflicts
considered; monitoring of levetiracetam and oxcarbazepine of interest. DB has received speakers honoraria from Eisai and UCB
(as MHD) levels may be considered, and although there is Pharma. We confirm that we have read the Journals position on issues
involved in ethical publication and affirm that this report is consistent with
insufficient evidence to support or refute changes in those guidelines.
phenobarbital, valproate, primidone, or ethosuximide lev-
els, this lack of evidence should not discourage monitoring
during pregnancy (Harden et al., 2009b). References
We agree that the usefulness of drug level monitoring in Adab N, Kini U, Vinten J, Ayres J, Baker G, Clayton-Smith J, Coyle H,
pregnancy depends on the AED, but also on the individual Fryer A, Gorry J, Gregg J, Mawer G, Nicolaides P, Pickering L,
characteristics of the patient and her epilepsy. Therefore, Tunnicliffe L, Chadwick DW. (2004) The longer term outcome of
children born to mothers with epilepsy. J Neurol Neurosurg Psychiatry
monitoring appears less important for drugs such as carba- 75:15751583.
mazepine and valproate, where alterations in active drug Anderson GD. (2005) Pregnancy-induced changes in pharmacokinetics: a
levels are usually minor. On the other hand, monitoring is mechanistic-based approach. Clin Pharmacokinet 44:9891008.
Bardy A, Teramo K, Hiilesmaa V. (1982) Apparent plasma clearances of
more important for drugs with marked but unpredictable phenytoin, phenobarbitone, primidone, and carbamazepine during
alterations such as lamotrigine, levetiracetam, oxcarbaze- pregnancy: result of the prospective Helsinki study. In Janz D, Dam M,

Epilepsia, 54(3):405414, 2013


doi: 10.1111/epi.12109
413
Pharmacokinetics in Pregnancy

Bossi L, Helge H, Richens A, Schmidt D (Eds) Epilepsy, pregnancy, Finnell R, Shafer PO, Le Guen C. (2009b) Practice Parameter update:
and the child. Raven Press, New York, pp. 141145. management issues for women with epilepsyfocus on pregnancy (an
Bardy AH, Hiilesmaa VK, Teramo KA. (1987) Serum phenytoin during evidence-based review): vitamin K, folic acid, blood levels, and
pregnancy, labor and puerperium. Acta Neurol Scand 75:374375. breastfeeding. Report of the Quality Standards Subcommittee and
Battino D, Avanzini G, Bossi L, Canger R, Como ML, Croci D, Spina S. Therapeutics and Technology Assessment Subcommittee of the
(1982) Monitoring of antiepileptic drugs plasma levels during American Academy of Neurology and American Epilepsy Society.
pregnancy and puerperium. In Janz D, Dam M, Bossi L, Helge H, Neurology 3:142149.
Richens A, Schmidt D (Eds) Epilepsy, pregnancy and the child. Raven Hernandez-Diaz S, Smith CR, Shen A, Mittendorf R, Hauser WA, Yerby
Press, New York, pp. 147154. M, Holmes LB. (2012) Comparative safety of antiepileptic drugs
Battino D, Binelli S, Bossi L, Canger R, Como ML, Croci D, Cusi C, during pregnancy. Neurology 78:16921699.
De Giambattista M, Pardi GGA. (1984a) Monitoring of antiepileptic Hussein Z, Posner J. (1997) Population pharmacokinetics of lamotrigine
drugs and hormones in pregnant epileptic women. In Porter RJ, Mattson monotherapy in patients with epilepsy: retrospective analysis of routine
RH, Ward AA, Dam M (Eds) Advances in epileptology: the XVth monitoring data. Br J Clin Pharmacol 43:457465.
Epilepsy International Symposium. Raven Press, New York, pp. 227 Johannessen SI, Landmark CJ. (2010) Antiepileptic drug interactions -
232. principles and clinical implications. Curr Neuropharmacol 8:254267.
Battino D, Binelli S, Bossi L, Como ML, Croci D, Cusi C, Avanzini G. Johannessen SI, Tomson T. (2006) Pharmacokinetic variability of newer
(1984b) Changes in primidone/phenobarbitone ratio during pregnancy antiepileptic drugs: when is monitoring needed? Clin Pharmacokinet
and the puerperium. Clin Pharmacokinet 9:252260. 45:10611075.
Battino D, Binelli S, Bossi L, Canger R, Croci D, Cusi C, De Giambattista Johannessen SI, Helde G, Brodtkorb E. (2005) Levetiracetam
M, Avanzini G. (1985) Plasma concentrations of carbamazepine and concentrations in serum and in breast milk at birth and during lactation.
carbamazepine 10,11-epoxide during pregnancy and after delivery. Epilepsia 46:775777.
Clin Pharmacokinet 10:279284. Johannessen Landmark C, Patsalos PN. (2010) Drug interactions involving
Bernus I, Hooper WD, Dickinson RG, Eadie MJ. (1995) Metabolism of the new second and third generation antiepileptic drugs. Expert Rev
carbamazepine and co-administered anticonvulsants during pregnancy. Neurother 10:119140.
Epilepsy Res 21:6575. Johannessen Landmark C, Patsalos PN. (2012) Methodologies used to
Bialer M, Johannessen SI, Levy RH, Perucca E, Tomson T, White HS. identify and characterize interactions among antiepileptic drugs. Expert
(2009) Progress report on new antiepileptic drugs: a summary of the Rev Clin Pharmacol 5:281292.
Ninth Eilat Conference (EILAT IX). Epilepsy Res 83:143. Johannessen Landmark C, Johannessen SI, Tomson T. (2012) Host factors
Cantwell R, Clutton-Brock T, Cooper G, Dawson A, Drife J, Garrod D, affecting antiepileptic drug delivery-pharmacokinetic variability. Adv
Harper A, Hulbert D, Lucas S, McClure J, Millward-Sadler H, Neilson Drug Deliv Rev 64:896910.
J, Nelson-Piercy C, Norman J, OHerlihy C, Oates M, Shakespeare J, Kawada K, Itoh S, Kusaka T, Isobe K, Ishii M. (2002) Pharmacokinetics of
de Swiet M, Williamson C, Beale V, Knight M, Lennox C, Miller A, zonisamide in perinatal period. Brain Dev 24:9597.
Parmar D, Rogers J, Springett A. (2011) Saving Mothers Lives: Lander CM, Eadie MJ. (1991) Plasma antiepileptic drug concentrations
reviewing maternal deaths to make motherhood safer: 20062008. The during pregnancy. Epilepsia 32:257266.
Eighth Report of the Confidential Enquiries into Maternal Deaths in the Lander CM, Livingstone I, Tyrer JH, Eadie MJ. (1981) The clearance of
United Kingdom. BJOG 118(Suppl. 1):1203. anticonvulsant drugs in pregnancy. Clin Exp Neurol 17:7178.
Christensen J, Sabers A, Sidenius P. (2006) Oxcarbazepine concentrations Langtry HD, Gillis JC, Davis R. (1997) Topiramate. A review of its
during pregnancy: a retrospective study in patients with epilepsy. pharmacodynamic and pharmacokinetic properties and clinical efficacy
Neurology 67:14971499. in the management of epilepsy. Drugs 54:752773.
Dansky L, Andermann E, Shervin A, Andermann F. (1982) Plasma levels Mawhinney E, Campbell J, Craig J, Russell A, Smithson W, Parsons L,
of pheytoin during pregnancy ant the puerperium. In Janz D, Dam M, Robertson I, Irwin B, Morrison P, Liggan B, Delanty N, Hunt S,
Bossi L, Helge H, Richens A, Schmidt D (Eds) Epilepsy, pregnancy, Morrow J. (2012) Valproate and the risk for congenital malformations:
and the child. Raven Press, New York, pp. 155162. is formulation and dosage regime important? Seizure 21:215218.
de Haan GJ, Edelbroek P, Segers J, Engelsman M, Lindhout D, Devile- May TW, Korn-Merker E, Rambeck B. (2003) Clinical pharmacokinetics
Notschaele M, Augustijn P. (2004) Gestation-induced changes in of oxcarbazepine. Clin Pharmacokinet 42:10231042.
lamotrigine pharmacokinetics: a monotherapy study. Neurology 63: Mazzucchelli I, Onat FY, Ozkara C, Atakli D, Specchio LM, Neve AL,
571573. Gatti G, Perucca E. (2006) Changes in the disposition of oxcarbazepine
Fitton A, Goa KL. (1995) Lamotrigine. An update of its pharmacology and and its metabolites during pregnancy and the puerperium. Epilepsia
therapeutic use in epilepsy. Drugs 50:691713. 47:504509.
Fotopoulou C, Kretz R, Bauer S, Schefold JC, Schmitz B, Dudenhausen Meador KJ, Baker GA, Browning N, Clayton-Smith J, Combs-Cantrell DT,
JW, Henrich W. (2009) Prospectively assessed changes in lamotrigine- Cohen M, Kalayjian LA, Kanner A, Liporace JD, Pennell PB, Privitera
concentration in women with epilepsy during pregnancy, lactation and M, Loring DW. (2009a) Cognitive function at 3 years of age after fetal
the neonatal period. Epilepsy Res 85:6064. exposure to antiepileptic drugs. N Engl J Med 360:15971605.
Franco V, Mazzucchelli I, Gatti G, Specchio LM, La Neve A, Papantonio Meador KJ, Penovich P, Baker GA, Pennell PB, Bromfield E, Pack A,
A, Ozkaynakci AE, Perucca E. (2008) Changes in lamotrigine Liporace JD, Sam M, Kalayjian LA, Thurman DJ, Moore E, Loring
pharmacokinetics during pregnancy and the puerperium. Ther Drug DW. (2009b) Antiepileptic drug use in women of childbearing age.
Monit 30:544547. Epilepsy Behav 16:609616.
Gidal BE, Radulovic LL, Kruger S, Rutecki P, Pitterle M, Bockbrader HN. National Institute for Clinical Excellence. (2012) The epilepsies: the
(2000) Inter- and intra-subject variability in gabapentin absorption and diagnosis and management of the epilepsies in adults and children in
absolute bioavailability. Epilepsy Res 40:123127. primary and secondary care. NICE Clinical guideline 137. Available at
Harden CL, Meador KJ, Pennell PB, Hauser WA, Gronseth GS, French JA, http://www.nice.org.uk/nicemedia/live/13635/57779/57779.pdf.
Wiebe S, Thurman D, Koppel BS, Kaplan PW, Robinson JN, Hopp J,
Ohman I, Vitols S, Tomson T. (2000) Lamotrigine in pregnancy:
Ting TY, Gidal B, Hovinga CA, Wilner AN, Vazquez B, Holmes L, pharmacokinetics during delivery, in the neonate, and during lactation.
Krumholz A, Finnell R, Hirtz D, Le Guen C. (2009a) Practice Epilepsia 41:709713.
Parameter update: management issues for women with epilepsyfocus
Ohman I, Vitols S, Luef G, Soderfeldt B, Tomson T. (2002) Topiramate
on pregnancy (an evidence-based review): Teratogenesis and perinatal kinetics during delivery, lactation, and in the neonate: preliminary
outcomes. Report of the Quality Standards Subcommittee and observations. Epilepsia 43:11571160.
Therapeutics and Technology Subcommittee of the American
Ohman I, Vitols S, Tomson T. (2005) Pharmacokinetics of gabapentin
Academy of Neurology and American Epilepsy Society. Neurology during delivery, in the neonatal period, and lactation: does a fetal
50:12371246. accumulation occur during pregnancy? Epilepsia 46:16211624.
Harden CL, Pennell PB, Koppel BS, Hovinga CA, Gidal B, Meador KJ,
Ohman I, Beck O, Vitols S, Tomson T. (2008a) Plasma concentrations of
Hopp J, Ting TY, Hauser WA, Thurman D, Kaplan PW, Robinson JN, lamotrigine and its 2-N-glucuronide metabolite during pregnancy in
French JA, Wiebe S, Wilner AN, Vazquez B, Holmes L, Krumholz A, women with epilepsy. Epilepsia 49:10751080.

Epilepsia, 54(3):405414, 2013


doi: 10.1111/epi.12109
414
T. Tomson et al.

Ohman I, Luef G, Tomson T. (2008b) Effects of pregnancy and guideline recommended for use in Scotland. Available at http://www.
contraception on lamotrigine disposition: new insights through analysis sign.ac.uk/guidelines/fulltext/70/index.html.
of lamotrigine metabolites. Seizure 17:199202. Seino M, Naruto S, Ito T, Miyazaki H. (1995) Zonisamide. In Levy RH,

Ohman I, Sabers A, de Flon P, Luef G, Tomson T. (2009) Pharmacokinetics Mattson RH, Meldrum BS (Eds) Antiepileptic drugs. 4th ed. Raven
of topiramate during pregnancy. Epilepsy Res 87:124129. Press, New York, pp. 10011024.
Oles KS, Bell WL. (2008) Zonisamide concentrations during pregnancy. Sturgiss SN, Dunlop W, Davison JM. (1994) Renal haemodynamics and
Ann Pharmacother 42:11391141. tubular function in human pregnancy. Baillieres Clin Obstet Gynaecol
Omtzigt JG, Nau H, Los FJ, Pijpers L, Lindhout D. (1992) The disposition 8:209234.
of valproate and its metabolites in the late first trimester and early Teramo K, Hiilesmaa V. (1982) Pregnancy and fetal complications in
second trimester of pregnancy in maternal serum, urine, and amniotic epileptic pregnancies. In Janz D, Dam M, Bossi L, Helge H, Richens A,
fluid: effect of dose, co-medication, and the presence of spina bifida. Schmidt D (Eds) Epilepsy, pregnancy, and the child. Raven Press, New
Eur J Clin Pharmacol 43:381388. York, pp. 5359.
Patsalos PN. (2000) Pharmacokinetic profile of levetiracetam: toward ideal The EURAP study. (2006) Seizure control and treatment in pregnancy:
characteristics. Pharmacol Ther 85:7785. observations from the EURAP epilepsy pregnancy registry. Neurology
Patsalos PN, Berry DJ, Bourgeois BF, Cloyd JC, Glauser TA, Johannessen 66:354360.
SI, Leppik IE, Tomson T, Perucca E. (2008) Antiepileptic drugsbest Tomson T, Battino D. (2007) Pharmacokinetics and therapeutic drug
practice guidelines for therapeutic drug monitoring: a position paper by monitoring of newer antiepileptic drugs during pregnancy and the
the subcommission on therapeutic drug monitoring, ILAE Commission puerperium. Clin Pharmacokinet 46:209219.
on Therapeutic Strategies. Epilepsia 49:12391276. Tomson T, Battino D. (2012) Teratogenic effects of antiepileptic drugs.
Pennell PB. (2003) Antiepileptic drug pharmacokinetics during pregnancy Lancet Neurol 11:803813.
and lactation. Neurology 61:S35S42. Tomson T, Lindbom U, Ekqvist B, Sundqvist A. (1994a) Disposition
Pennell PB, Newport DJ, Stowe ZN, Helmers SL, Montgomery JQ, Henry of carbamazepine and phenytoin in pregnancy. Epilepsia 35:131
TR. (2004) The impact of pregnancy and childbirth on the metabolism 135.
of lamotrigine. Neurology 62:292295. Tomson T, Lindbom U, Ekqvist B, Sundqvist A. (1994b) Epilepsy and
Pennell PB, Koganti A, Helmers S, Beach A, Newman M, Newport DJ, pregnancy: a prospective study of seizure control in relation to free and
Stowe ZN. (2005) The impact of pregnancy and childbirth on the total plasma concentrations of carbamazepine and phenytoin. Epilepsia
elimination of levetiracetam [abstract]. Epilepsia 46:89. 35:122130.
Pennell PB, Peng L, Newport DJ, Ritchie JC, Koganti A, Holley DK, Newman
Tomson T, Ohman I, Vitols S. (1997) Lamotrigine in pregnancy and
M, Stowe ZN. (2008) Lamotrigine in pregnancy: clearance, therapeutic lactation: a case report. Epilepsia 38:10391041.
drug monitoring, and seizure frequency. Neurology 70:21302136. Tomson T, Luef G, Sabers A, Pittschieler S, Ohman I. (2006) Valproate
Perucca E, Bialer M. (1996) The clinical pharmacokinetics of the newer effects on kinetics of lamotrigine in pregnancy and treatment with oral
antiepileptic drugs. Focus on topiramate, zonisamide and tiagabine. contraceptives. Neurology 67:12971299.
Clin Pharmacokinet 31:2946. Tomson T, Palm R, Kallen K, Ben-Menachem E, Soderfeldt B, Danielsson
Petrenaite V, Sabers A, Hansen-Schwartz J. (2005) Individual changes in B, Johansson R, Luef G, Ohman I. (2007) Pharmacokinetics of
lamotrigine plasma concentrations during pregnancy. Epilepsy Res levetiracetam during pregnancy, delivery, in the neonatal period, and
65:185188. lactation. Epilepsia 48:11111116.
Petrenaite V, Sabers A, Hansen-Schwartz J. (2009) Seizure deterioration in Tomson T, Battino D, Bonizzoni E, Craig J, Lindhout D, Sabers A, Perucca
women treated with oxcarbazepine during pregnancy. Epilepsy Res E, Vajda F. (2011) Dose-dependent risk of malformations with
84:245249. antiepileptic drugs: an analysis of data from the EURAP epilepsy and
Philbert A, Pedersen B, Dam M. (1985) Concentration of valproate during pregnancy registry. Lancet Neurol 10:609617.
pregnancy, in the newborn and in breast milk. Acta Neurol Scand Tran TA, Leppik IE, Blesi K, Sathanandan ST, Remmel R. (2002)
72:460463. Lamotrigine clearance during pregnancy. Neurology 59:251255.
Radtke RA. (2001) Pharmacokinetics of levetiracetam. Epilepsia 42(Suppl. Westin AA, Reimers A, Helde G, Nakken KO, Brodtkorb E. (2008) Serum
4):2427. concentration/dose ratio of levetiracetam before, during and after
Ramsay RE, Strauss RG, Wilder BJ, Willmore LJ. (1978) Status epilepticus pregnancy. Seizure 17:192198.
in pregnancy: effect of phenytoin malabsorption on seizure control. Westin AA, Nakken KO, Johannessen SI, Reimers A, Lillestolen KM,
Neurology 28:8589. Brodtkorb E. (2009) Serum concentration/dose ratio of topiramate
Rating D, Nau H, Jager-Roman E, Gopfert-Geyer I, Koch S, Beck- during pregnancy. Epilepsia 50:480485.
Mannagetta G, Schmidt D, Helge H. (1982) Teratogenic and Wlodarczyk BJ, Palacios AM, George TM, Finnell RH. (2012)
pharmacokinetic studies of primidone during pregnancy and in the Antiepileptic drugs and pregnancy outcomes. Am J Med Genet A
offspring of epileptic women. Acta Paediatr Scand 71:301311. 158A:20712090.
Reimers A, Brodtkorb E. (2012) Second-generation antiepileptic drugs and Yerby MS, Friel PN, Miller DQ. (1985) Carbamazepine protein binding
pregnancy: a guide for clinicians. Expert Rev Neurother 12:707717. and disposition in pregnancy. Ther Drug Monit 7:269273.
Reimers A, Helde G, Brathen G, Brodtkorb E. (2011) Lamotrigine and its Yerby MS, Friel PN, McCormick K, Koerner M, Van Allen M, Leavitt AM,
N2-glucuronide during pregnancy: the significance of renal clearance Sells CJ, Yerby JA. (1990) Pharmacokinetics of anticonvulsants in
and estradiol. Epilepsy Res 94:198205. pregnancy: alterations in plasma protein binding. Epilepsy Res 5:223228.
Scottish Intercollegiate Guidelines Network. (2003) Guideline No. 70. Yerby MS, Friel PN, McCormick K. (1992) Antiepileptic drug disposition
Diagnosis and management of epilepsy in adults: a national clinical during pregnancy. Neurology 42:1216.

Epilepsia, 54(3):405414, 2013


doi: 10.1111/epi.12109
This document is a scanned copy of a printed document. No warranty is given about the accuracy of the copy.
Users should refer to the original published version of the material.

Anda mungkin juga menyukai