Download Epilepsy, sex hormones and antiepileptic drugs in female patients

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Hormone replacement therapy (menopause) wikipedia , lookup

Hormone replacement therapy (male-to-female) wikipedia , lookup

Polycystic ovary syndrome wikipedia , lookup

Hyperandrogenism wikipedia , lookup

Transcript
Review
For reprint orders, please contact [email protected]
Epilepsy, sex hormones and
antiepileptic drugs in
female patients
Expert Rev. Neurother. 9(12), 1803–1814 (2009)
Alberto Verrotti†,
Claudia D’Egidio,
Giangennaro Coppola,
Pasquale Parisi and
Francesco Chiarelli
Author for correspondence
Department of Pediatrics,
University of Chieti, Via dei
Vestini 5, 66100 Chieti, Italy
Tel.: +39 087 135 8015
Fax: +39 087 157 4831
[email protected]
†
Women with epilepsy have a higher incidence of reproductive endocrine disorders than the
general female population. These alterations include polycystic ovary syndrome,
hyperandrogenemia, infertility, hypothalamic amenorrhea and hyperprolactinemia. Reproductive
dysfunction is attributed both to epilepsy itself and to antiepileptic drugs (AEDs). Focal epileptic
discharges from the temporal lobe may have a direct influence on the function of the
hypothalamic–pituitary axis, thus altering the release of sex steroid hormones, including the
production of luteinizing hormone, follicle-stimulating hormone, gonadotropin-releasing
hormone and prolactin. AEDs may modulate hormone release from the hypothalamic–pituitary–
gonadal axis and they may alter the metabolism of sex hormones and their binding proteins.
Hepatic enzyme-inducing AEDs, such as carbamazepine and phenytoin, may be most clearly
linked to altered metabolism of sex steroid hormones, but valproic acid, an enzyme inhibitor,
has also been associated with a frequent occurrence of polycystic ovary syndrome and
hyperandrogenism in women with epilepsy. Therefore, treatment of epilepsy and selection of
AEDs are important for reproductive health in female patients. The aim of the present review
is to critically evaluate the recently published data concerning the interactions between sex
hormones, epilepsy and AEDs.
Keywords : amenorrhea • antiepileptic drug • epilepsy • hyperandrogenemia • hyperprolactinemia
• polycystic ovary syndrome • sex hormone dysfunction
Epilepsy is a common neurologic disorder affecting women during their reproductive years, and
is associated with hormonal disturbances resulting in altered endocrine reproduction and sexual
function [1,2] . A possible role for seizure disorder
or, alternatively, of the use of antiepileptic drugs
(AEDs), have been suggested as the patho­genetic
mechanisms. The brain regulates hormonal
secretion and is sensitive to hormonal feedback;
the neuroendocrine feedback system includes the
hypothalamus, pituitary, gonads and amygdala.
The amygdala is linked to the hypothalamic–
pituitary axis (HPA) [3] , which is involved in the
regulation, production and secretion of ovarian
steroids. Reproductive endocrine disorders associated with epilepsy can be expected in view of
the complex interconnections between the HPA
and the limbic system; the involvement of medial
temporal lobe regions in epilepsy may cause
changes in sex hormone secretion and, consequently, reproductive function [4] . Moreover,
AEDs may also alter hormone levels and interfere
with reproductive function [5,6] .
www.expert-reviews.com
10.1586/ERN.09.112
This review evaluates whether and to what
degree epilepsy and its treatment can affect
reproductive function. For this purpose, this
review is subdivided into two main sections: the
effects of epilepsy on reproductive function and
the effects of AEDs on sex hormones.
Effects of epilepsy on
reproductive function
Reproductive endocrine disorders, such as menstrual cycle disturbances and ovulatory dysfunctions, have frequently been reported in epileptic
women [1–3,5–13] . These abnormalities usually
result from abnormal reproductive endocrine
function and are associated with lower rates
of ovulation and higher rates of infertility [12] .
Menstrual disorders are currently estimated to
occur in a third of women with epilepsy compared with 12–14% of women in the general
population [13] . Anovulation, hypothalamic
amenorrhea (HA), hyperandrogenemia, poly­
cystic ovary syndrome (PCOS), functional
hyperprolactinemia, premature menopause and
© 2009 Expert Reviews Ltd
ISSN 1473-7175
1803
Review
Verrotti, D’Egidio, Coppola, Parisi & Chiarelli
risk of infertility are over-represented in women with epilepsy
(Box 1) [1,9,14–20] . Epilepsy itself may have effects on sex hormone
levels and reproductive endocrine function [1,5,21–22,23] .
Animal studies have demonstrated that reproductive endocrine and sexual dysfunction is more common in partial compared with generalized epilepsy, especially in temporal lobe epilepsy [24–26] ; animal investigations demonstrated that induction
of temporo–limbic seizures can cause changes in reproductive
hormone levels and reproductive function [24,25,27] . In particular,
amygdala seizures in female rodents can cause anovulation, elevated serum testosterone levels and the accumulation of follicular
cystis in the ovaries [24] .
These findings have also been confirmed in human studies
[14,28] . More than a third of cycles in women with localizationrelated epilepsy are anovulatory, compared with 8–10% in controls [11,13,29–33] . Partial epilepsy, mainly when originating in
the temporal lobe, is the form most frequently associated with
reproductive dysfunction [1,13,19,34] . However, other studies
demonstrated that more than 50% of patients with generalized
convulsions may develop reproductive and sexual abnormalities [21,29,35] ; in particular, hyperandrogenemia and PCOS are
more frequent in women with idiopathic generalized epilepsy
than in localization-related epilepsy [14,29,36] . Interestingly,
Herzog et al., examined 50 women with tempolimbic epilepsy
and found that 28 patients had amenorrhea, oligoamenorrhea,
or abnormally long or short menstrual cycle intervals; 19 out of
the 28 women with epilepsy and menstrual disorders had readily
identifiable reproductive endocrine disorders: PCOS in ten, HA
in six, premature menopause in two and hyperprolactinemia in
one [1] . Since no significant relationship between the occurrence
of menstrual disorders and the use of AEDs has been found, it
is suggested that epilepsy may have an independent effect on
reproductive function. This is in agreement with a previous
study describing a significantly higher frequency of ovulatory
dysfunction in female patients, not correlated with the use of
any specific AED [35] .
Moreover, high levels of luteinizing hormone (LH) and an
altered LH-to-follicle-stimulating hormone (FSH) ratio have been
reported in women with epilepsy [17–19] . The presence of elevated
LH pulse frequency in untreated or epileptic women with regular menstrual cycles supports the notion that epilepsy itself may
contribute to reproductive abnormalities [21,35] .
Box 1. Frequent reproductive dysfunctions in
women with epilepsy.
• Hyperandrogenism [29,36,46,94]
• High levels of luteinizing hormone and luteinizing hormone-tofollicle-stimulating hormone ratio [16,17,36,55]
• Polycystic ovary syndrome [1,13,14,30,31,46,47,92–93]
• Menstrual irregularities [1,17,32,35,92]
• Premature menopause [1,15,16]
• Infertility [7,9,110,149]
• Hyperprolactinemia [42,53,150,151]
• Sexual dysfunction [14,50,52,152,153]
1804
Interestingly, a recent cross-sectional observational study of
Murialdo et al. evaluated the relationships between sex steroids
and seizure frequency in women with partial epilepsy on AED
treatments [37] . The authors found that estrodiol, free estrodiol and
progesterone levels were lower in both ovarian phases in patients,
whereas those of sex hormone-binding globulin (SHBG) were
higher than in controls. When changes in hormone levels were compared with those in healthy controls, luteal-free estrodiol and progesterone levels were chiefly impaired in women with more frequent
seizures, mostly undergoing AED polytherapies, but not in those
with absent or rarer seizures. Furthermore, an inverse relationship
was found between free estrodiol levels and seizure frequency scores
during the follicular phase, and with disease duration in the luteal
phase. However, the authors suggested that these findings may be
attributed to the longer duration of AED treatments and the more
frequent use of enzyme-inducing AEDs (EIAEDs) in subjects with
more severe seizures. Accordingly, a previous retrospective study
reported that menstrual disorders were more frequent in patients
with high seizure frequency (less than five seizures per year) [38] .
The mechanisms underlying the association between epilepsy
and reproductive endocrine disorders are still not clear. HPA dysfunction is suggested by observations that pituitary release of
LH in women with epilepsy is altered both spontaneously and
in response to gonadotropin-releasing hormone (GnRH) [1,30,31] .
The brain controls reproductive function primarily through
hypothalamic regulation of pituitary secretion [39] . The hypo­
thalamus receives direct connections from the cerebral hemispheres, especially from temporolimbic structures that are
commonly involved in epilepsy and, most notably, from the
amygdala [1,40,41] . In the majority of cases, the epileptic activity
in the frontal lobe exhibits a propagation towards the temporal
lobe and, thereby, influences the hormonal pathway of prolactin
(PRL). [42] . Clinically, there is evidence to suggest that epileptiform discharges may disrupt the temporolimbic modulation of
hypothalamic–pituitary function [14,26] and, ultimately, lead to
changes in ovarian steroidogenesis and morphology [43] . There are
also important clinical findings that indicate that the laterality
and focality of epilepsy may be important determinants of certain
reproductive endocrine disorders [1,14,28,44–46] . In fact, unilateral
temporolimbic discharges are associated with laterally differing
changes in hormonal secretion at all levels of the neuroendocrine
reproductive axis. Consequently, different reproductive disorders
may develop in relation to left- and right-sided temporolimbic epilepsy; in particular, left unilateral temporolimbic epilepsy is associated with a higher occurrence of PCOS [14,47] . Indeed, seizures in
the left side of the limbic system increase the pulse frequencies of
GnRH secretion that, in turn, increases the LH/FSH ratios and
testosterone levels. By contrast, right unilateral temporolimbic
epilepsy is associated with lower GnRH pulse frequency, which
causes a decrease in LH and estrogen levels [1,14] . These hormonal
changes are characteristic of HA [9,28,48] .
Nevertheless, a recent experiment using the amygdala kindling
model in rats demonstrated that, in female rats, seizures that
originate in the left or right amygdala do not result in lateralized
effects on the reproductive system [27] .
Expert Rev. Neurother. 9(12), (2009)
Epilepsy, sex hormones & antiepileptic drugs in female patients
Epileptic discharges in limbic structures may also contribuite
to sexual dysfunction in women, such as lack of sexual interest
and high rates of orgasmic dysfunction, including anorgasmia,
dyspareunia, vaginism or insuffcient vaginal lubrication [49,50] .
Herzog et al. found that right temporal epileptic discharges in
women were associated with hypogonadotropic hypogonadism,
including decreased sexual interest [1] . Sexuality in people with
epilepsy may be adversely affected by alterations in the pituitary
gonado­tropins, PRL and sex steroid hormones [1,12] . Limbic efferents seem to exhibit an impact on the HPA even in the interictal phase of the disorder, and post-ictally elevated PRL serum
concentrations can be detected. In correlation with the type of
seizures, evidence of elevated PRL is found in 88% of seizures
following generalized tonic–clonic seizures, in 78% of seizures
following complex partial seizures and in 22% of seizures following simple partial seizures [51] . The epileptic activity in the frontal
lobe exhibits propagation towards the temporal lobe and thereby
influences the hormonal pathway of PRL, which can also show
tendencies to post-ictal elevation [42] .
In women (and men) with epilepsy, reduced genital blood
flow during erotic stimulation was found via video. The women
with epilepsy who were examined showed a reduced elevation of
genital blood flow during the presentation of erotic video movies
in comparison with healthy controls [52] . Irrespective of gender,
sexual questionnaires revealed reduced libido more commonly in
the right- than in the left-sided temporal lobe seizure origin 44] .
However, compelling explanation remains to be found. It is possible that the right sided temporal lobe disturbance may have a
more pronounced influence on emotions and, therefore, indirectly
on libido [53] .
Baird et al. reported that a third of their 58 lobectomy patients
had an increase in sexual activity after surgery, compared with
a quarter of patients having decreased postsurgical sexual activity [45] . Furthermore, Manna et al. recently���������������������
demonstrated
��������������������
a relationship between a serotonin transporter genetic polymorphism
and temporal lobe epilepsy, suggesting that the serotonin transporter gene may play a role in the development of temporal lobe
epilepsy [54] . These findings indicate that some patients with epilepsy may be genetically susceptible to sexual dysfunction through
a serotonin transport mechanism.
In conclusion, an increased frequency of reproductive endocrine disorders, secondary to pituitary hormone changes, has been
reported in women with epilepsy, especially among those with
partial epilepsy of temporal lobe origin. Therefore, regular monitoring of reproductive function, including menstrual disorders,
infertility, hirsutism and galactorrhea, are recommended.
Effects of antiepileptic drugs on sex hormone levels
A large body of literature addresses the presence of alterations in
sex hormone levels in women taking AEDs [2,5,11,17,18,55–62] .
Antiepileptic drugs may have an influence on the metabolism of the central and/or peripheral endocrine hormones and
their binding proteins. Hepatic EIAEDs, such as phenobarbital
(PB), phenytoin (PHT) and carbamazepine (CBZ), are most
clearly linked to altered metabolism of sex steroid hormones.
www.expert-reviews.com
Review
In fact, in the late 1970s and 1980s, serum sex hormone levels
were reported to be abnormal in female patients treated with
EIAEDs [55,56] .
Victor et al. were the first to report elevated serum SHBG
concentrations in women treated with PHT [57] , and this finding was later confirmed [55,56] . Low dehydroepiandrosterone sulphate (DHEAS) levels have also been reported in women taking PHT [58] , but its clinical significance is unknown. However,
similarly impaired steroid levels have also been shown in women
with epilepsy treated with other EIAEDs [60,63] . In particular,
Galimberti et al. reported decreased DHEAs levels and increased
cortisol levels in women treated with CBZ, PHT, PB in monoor poly-therapy, but not in those treated with non-EIAEDs
(NEIAEDs) [63] . The most interesting finding in this study concerned the relation between epilepsy severity, increased cortisol,
and decreased DHEAs levels. In fact, cortisol and DHEAS levels
were directly and indirectly correlated with seizure frequency,
respectively, suggesting that changes in adrenal steroid levels must
be ascribed mainly to the frequency of seizures, which are known
to activate the hypothalamus–pituitary–adrenal axis [64] .
The most recent cross-sectional study showed that women with
epilepsy taking EIAED polytherapies had lower levels of estradiol
and a lower free estrogen index (FEI) than patients treated with
a single EIAED [59] . EIAEDs induce hepatic cytocrome P450dependent steroid hormone breakdown and production of SHBG,
thereby reducing biologically active sex hormone serum concentrations, such as estrodiol and testosterone [5,18,60–62] . Galimberti
et al. reported that the older patients and those with longer disease duration showed higher levels of SHBG [59] . This can be
attributed to the physiologic increase in binding protein levels
with aging and to the use of EIAEDs. These changes in serum
SHBG levels, and the lowered total estrodiol levels, implied a
global decrease in biologically active free estrodiol. Conversely,
significant differences in estrodiol levels and in FEI were found
between women on different AED regimens. Patients treated with
EIAED polytherapies showed estrodiol and FEI values that were
lower than those recorded both in patients treated with a single
EIAED or NEIAED, or in those on combined (EIAED plus
NEIAED) therapies.
Recently, Jacobsen et al. reported that PB, PHT, valproic acid
(VPA), oxcarbazepine (OXC) and lamotrigine (LTG), but not
CBZ, inhibit the aromatase complex (CYP19) activity that converts testosterone into estrodiol in vitro [65] . Accordingly, a more
recent study demonstrated that VPA and LTG reduced CYP19
aromatase activity in human ovarian follicular cells, but only in
FSH-stimulated cells and at higher concentrations [66] . Additive
enzyme inhibition has been observed in combination experiments with multiple AEDs; therefore, aromatase inhibition might
explain the decreased production of estrodiol observed in vivo,
mainly when combined AED therapies were employed [59] .
Many studies that have evaluated the reproductive endocrine
effects of CBZ in female patients have consistently reported
increased serum SHBG levels and other sex hormone abnormalities [2,62,65,67–71] . Interestingly, Hamed et al. reported elevated
levels of LH and FSH in 28.1 and 34.4% of CBZ-treated patients,
1805
Review
Verrotti, D’Egidio, Coppola, Parisi & Chiarelli
respectively [17] . Dana-Haeri et al. observed that epileptic patients
on CBZ had elevated baseline LH levels and a exaggerated LH
response to the GnRH and thyrotropin-releasing hormones
stimulation [72] . This exaggerated LH response may be related
to a positive-feedback mechanism. Furthermore, they reported
slightly higher PRL levels 2 h after stimulation with GnRH and
thyrotropin-releasing hormone. These changes have not been
confirmed by other studies [60,72–74] . Recently, a double-blind,
randomized, controlled withdrawal study, reported that CBZ
withdrawal was associated with a significant decrease in SHBG
levels and a increase in serum testosterone concentrations, free
androgen index (FAI) and estrogen/SHBG ratio; these results
demonstrated that potential changes in sex steroid levels, induced
by CBZ treatment, could be reversible [71] .
However, most of the studies have not shown an increased frequency of menstrual disorders or hyperandrogenism and polycystic
ovaries (PCO) in women taking CBZ [2,60,67,75,76] , despite alterations in serum sex hormone concentrations. In fact, single abnormal laboratory findings without symptoms may not constitute a
clinically relevant endocrine disorder.
Among NEIAEDs, VPA appears to be associated with frequent
reproductive endocrine disorders, such as polycystic changes in
the ovaries, high serum testosterone concentrations and amenorrhea [2,38,77] . These abnormalities are especially common among
women who have gained weight during VPA therapy [61] . In 1993,
Isojarvi et al. through a cross-sectional study showed that menstrual disorders were common among women taking VPA monotherapy for epilepsy and that they were frequently associated with
PCOs and/or hyperandrogenism [2] . PCOs and hyperandrogenism
were expecially common if VPA medication was started before
the age of 20 years [70] . An increased number of ovarian cysts
and PCOS in women on VPA monotherapy was subsequently
confirmed [75,78,79] . In another study of women taking VPA currently or within the preceding 3 years, 38.1% had experienced at
least one anovulatory cycle in contrast to 10.7% of women not
using VPA within the preceding 3 years [11] . Interestingly, women
with idiopathic generalized epilepsy receiving VPA were at highest
risk for anovulatory cycles, polycystic ovaries, elevated BMI and
hyperandrogenism.
A more recent prospective study by Morrell et al. reported a
higher incidence of hyperandrogenism or ovulatory dysfunction in
women taking VPA for epilepsy than in women taking LTG [80] .
Moreover, a significant increase in serum testosterone concentration
was especially prominent in women initiating treatment before the
age of 20 years, suggesting that young women with epilepsy seem
to be especially vulnerable to the effects of VPA on ovarian function [70] . By contrast, in two prospective randomized studies by
Kwan et al. [81] and Stephen et al. [82] , comparing women receiving
VPA and LTG monotherapy, no significant difference in change of
testosterone levels from baseline was found between the VPA and
LTG groups. This discrepancy is probably owing two main reasons.
First, the sample size in these two studies [81,82] were comparatively
smaller than in the study by Morrell et al. [80] . Second, the mean
age of female subjects in the study by Kwan et al. [81] was more than
10 years older than that of subjects in the study by Morrell et al. [80] .
1806
Alterations in serum androgen levels have even been detected
before and during pubertal development in young girls taking
VPA [83] . The mean serum testosterone levels and FAI were high
in all pubertal phases in girls on VPA, even if the frequency
of hyperandrogenism increased with pubertal development.
Therefore, it is likely that VPA may affect steroid metabolism
during the sensitive period, at the onset of puberty. Accordingly,
hyperandrogenism observed in most adult women on VPA therapy can already be induced before the emergence of clinical signs
of puberty. A recent cross-sectional study showed that VPA treatment was only associated with higher testosterone levels in girls
after menarche, suggesting that the sensitivity to VPA-induced
hyperandrogenism is a function of sexual maturation [6] . In postmenarcheal girls, the presence of hyperandrogenemia without
clinical hyperandrogenism may reflect the increase in testosterone that precedes the appearance of clinical signs (i.e., hirsutism, acne, menstrual disorders and PCOs). This emphasizes
the importance of careful endocrine observation of girls treated
with VPA.
However, it is encouraging to note that the reproductive endocrine effects of VPA may be reversible after the medication is
discontinued; a 5 year follow-up study in young women showed
that the 60% of the patients who were administered VPA had
PCOS compared with 5.5% of the girls whose medication had
been discontinued [82] .
The mechanism by which VPA could cause hyperandrogenism
and related reproductive disorders is still unclear. It may be that
VPA can have a direct effect on ovarian androgen production or
that, acting as enzyme inhibitor, VPA may inhibit the metabolism
of sex hormones and thereby lead to increased serum androgen
levels [53,84,85] . Moreover, obesity (induced by VPA treatment) and
associated hyperinsulinemia could be implicated in the development
of PCOS and hyperandrogenism (see later) [2,70] .
Recently, it has been shown that VPA blocks the androgen
and progesterone receptors, but not the estrogen receptor [86] .
The strong progesterone receptor antagonism by VPA suggests
that some biological effects in women may be at least partly due
to impaired progesterone receptor-mediated progesterone action
on reproductive tissue (a previously unrecognized factor contri­
buting to the low fertility of women with epilepsy). In addition,
antiprogestin effects may contribute to the higher frequency of
anovulation among VPA-treated women with epilepsy [57,77] .
The reproductive endocrine effects of the new AEDs have not
been widely studied. However, most of the data suggest that OXC
therapy is not associated with changes in reproductive function
in epileptic patients [83,87] . Only one recent study reported low
serum testosterone concentrations, low FAI, elevated levels of
androstendione and DHEAS and increased prevalence of PCO in
women during OXC treatment [88] . A low prevalence of reproductive disorders has been reported during LTG therapy [5,73] , and
replacement of VPA with LTG resulted in normalization of endocrine function in women with previously identified endocrine
disorders (PCO, hyperandrogenism and increase in bodyweight),
probably related to VPA medication [89] ; serum insulin and testosterone levels returned to normal within 2 months following
Expert Rev. Neurother. 9(12), (2009)
Epilepsy, sex hormones & antiepileptic drugs in female patients
VPA replacement, and the levels remained normal thereafter.
Consistent with these findings, Morrell et al. reported that
women taking LTG had lower serum testosterone levels than
women taking VPA [90] .
In conclusion, careful endocrine observation of girls treated
with EIAED and VPA is important, while it seems that the new
AEDs may offer an alternative if reproductive endocrine problems
emerge during treatment with the older AEDs. Ovarian function
should be monitored closely as part of the comprehensive care of
women with epilepsy to prevent adverse effects of ovarian failure,
mainly in patients with more frequent seizures who need AED
polytherapies and/or in patients treated with old AEDs.
AEDs & PCOS
The most common reproductive endocrine disorder in women
with epilepsy is PCOS [13,21,75] . PCOS occurs in 10–20% of
women with epilepsy compared with 5–6% of women in the
general population [9,13,14,75] .
Polycystic ovary syndrome represents the failure of the ovarian follicle to complete normal maturation during the menstrual
cycle or a series of cycles, a failure that is perhaps related to the
presence of inadequate levels of pituitary FSH, while levels of LH
are normal or elevated [61] . These conditions can produce a failure
of ovulation and the partially developed follicle is retained in the
ovary in the form of a tiny cyst [46,47] . This partially developed
follicle is secretory but it is deficient in aromatase, the enzyme
that converts testosterone to estrogen and, therefore, this follicle
has testosterone as its principal secretory product. Testosterone
may increase the positive feedback of estrogen on pituitary LH
secretion, resulting in increased ovarian steroid secretion, and
can result in hyperandrogenism. Testosterone is aromatized in
peripheral adipose tissue, generally producing high–normal levels of estrogen [61] . Women with this syndrome frequently have
elevated cholesterol with abnormal lipid profiles, elevated insulin
and glucose intolerance [90,91] .
Although women with epilepsy may already have a higher
instance of the PCOS, it has been suggested that treatment with
certain AEDs, particularly VPA, increases this risk further [2,70,89] .
The role of VPA in the pathogenesis of PCOS has been first raised
by Isojärvi et al., who showed that PCOS and hyper­androgenism
may be more common in women with epilepsy who were treated
with VPA than in women treated with other AEDs [2] . Other studies confirmed these findings [69,70,75,92] . Interestingly, Gorkemli
et al. found no statistically significant duration-related rise of risk
for patients who developed PCOS; the lack of a duration-related
reproductive dysfunction may support the hypothesis of early
occurring VPA-associated endocrine changes [93] .
However, recent research did not substantiate the role of
VPA [60,76,94–96] ; the relationship between PCOS and AED
treatment remains controversial [75,95,97] .
The pathogenesis of PCOS appears to be multifactorial, including
genetic predisposition and the intervention of environmental factors.
An important risk factor for PCOS is weight gain [98–101] , which is a
common and undesirable effect of certain AEDs, notably VPA and,
to a lesser extent, CBZ, vigabatrin and gabapentin [102–107] . Weight
www.expert-reviews.com
Review
gain may lead to the expression or exacerbation of PCOS in some
genetically predisposed women, reducing insulin sensitivity. Thus,
AED-related weight increases could trigger the manifestation of a
clinically relevant endocrine disorder; in particular, VPA-induced
weight gain and the related endocrine abnormalities (e.g., hyperinsulinemia and hyperandrogemia) may induce the development
of PCOS [18] .
The pathogenic mechanisms underlying weight gain during
VPA treatment are still unclear [70,108–111] . The observation that
VPA-treated epileptic patients who reported weight gain developed increased appetite, thirst and quenching with calorie-rich
beverages supports the hypothesis of a stimulating effect of VPA
on the hypothalamus [89,109,112,113] .
It has been also suggested that VPA-related obesity may be
associated with elevated serum leptin levels in women with epilepsy [70,102] , but it is unclear whether leptin behavior in VPAinduced obesity is similar to that in any other obesity situation, as
supported by some data [114] . VPA can modify leptin levels through
the increase of bodyweight, and this modification is related to BMI.
Moreover, it is likely that obesity is the cause of insulin resistance
and related hyperinsulinemia [103] , which are often associated
with PCOS [115] . By suppressing the synthesis of SHBG, insulin
increases free androgen concentrations, subsequently increasing
the degree of hirsutism. The positive correlation between insulin
resistance and obesity means that in some women with PCO, the
clinical manifestation of menstrual irregularity and hirsutism
will only become apparent if there is an increase in weight and
associated metabolic changes [98] .
It has also been proposed that weight gain is associated with
low serum IGF-binding protein 1 levels, which may lead to
hyperandrogenism and PCO [70] . In addition, a significant
weight gain was recorded in a retrospective ana­lysis of hospital records of the women taking VPA who also had PCO and
hyperandrogenism; these women had higher insulin and lower
IGF-binding protein 1 levels.
The specific mechanism by which VPA induces insulin resistance remains to be determined. It is likely that, because VPA is a
branched-chain fatty acid, it can compete with free fatty acids for
albumin binding, increasing their local availability and thus their
physiological modulation of insulin secretion [94] . Moreover, it has
been demonstrated that VPA can directly stimulate pancreatic
b-cells [116] and inhibit GLUT-1 activity [117] .
Finally, in animal experiments VPA has been shown to alter
steroidogenesis and increase testosterone-to-estrodiol ratios in
porcine ovarian follicles [118] . Continuous treatment with VPA
increased the number of follicular cysts and altered sex steroid
hormone levels in rats [119] and increased the number of ovarian
follicular cysts in rats [120] . It remains to be proven that these
direct gonadal VPA effects are clinically relevant in humans.
Moreover, VPA is associated with another phenomenon potentially associated with the development of PCOS; it is an AED
with an inhibitory effect on the hepatic P450 enzyme system and,
therefore, it may inhibit the metabolism of sex steroids, such as
testosterone, and thereby lead to increased androgen levels [121] .
On the contrary, EIAEDs, such as CBZ, PHT or PB, may exert
1807
Review
Verrotti, D’Egidio, Coppola, Parisi & Chiarelli
a protective effect against the development of PCOS by reducing
biologically active testosterone in the serum and by increasing the
binding and metabolism of testosterone.
In conclusion, it remains unclear whether VPA is directly
responsible for the development of PCOS from otherwise normal ovaries. Although it is possible that weight gain may alter
the endocrine and biochemical features to produce PCOS, there
is no conclusive evidence to support this hypothesis. For these
reasons, to date, VPA remains a first-line option for treatment of
many epilepsy types, including in young women. However, since
VPA treatment is associated with greater disruption of reproductive endocrine functions than other AEDs, the length of the
menstrual cycles and bodyweight should be monitored in women
with epilepsy after beginning treatment with VPA.
Pregnancy & epilepsy
Good seizure control is paramount during pregnancy, especially
in the light of the observation that maternal mortality rates are
higher in women with epilepsy compared with the general population [122] . Regarding the effects of seizures of the fetus, seizures
other than generalized tonic–clonic seizures are unlikely to cause
harm. Convulsive seizures, on the other hand, induce lactic acidosis, which is transferred to the fetus and may also cause fetal
bradycardia [123,124] . In addition, status epilepticus can result in
intrauterine death [125] . The risk for the latter may have been overestimated, since one large prospective study reported only one case
of intrauterine death and no maternal mortality among 36 cases
with status epilepticus [126] . Furthermore, recent data suggest
that the number of stillbirths is not increased among women
that are adequately treated for their epilepsy during pregnancy
[127,128] . Finally, there is no clear evidence that partial, absence or
myoclonic seizures adversely affect a pregnancy or the developing
fetus, other than by effects of trauma.
In the past, women with epilepsy were considered at greater
risk for obstetric complications, including Cesarean delivery, preeclampsia, pregnancy-related hypertension, premature contractions
or premature labor and delivery. Most recent studies, however, suggest that with appropriate medical management the incidence of
complications in women with epilepsy is similar to that of the general population [128–133] . AED therapy should be optimized at least
6 months before conception. The reduction or withdrawal of AED
therapy during pregnancy is pointless because any congenital malformations occur in very early stages of develop­ment. The reduction
or withdrawal of AED therapy during pregnancy is also hazardous
because, although maternal seizures do not seem to increase the risk
of congenital malformations [134–138] , they may be harmful to both
the mother and the fetus Mothers should be strongly encouraged to
maintain adequate compliance with drug therapy [139] .
On the other hand, the risk of major congenital malformations is
two-to-three-times higher in children of mothers treated for epilepsy
during pregnancy than is expected in the general population [124] .
The reasons for this increased risk are mainly owing to teratogenic
effects of AEDs. In fact, a meta-ana­lysis suggested that the malformation rate among the offspring of women with untreated epilepsy
was similar to that of nonepileptic controls [134] . The incidence of
1808
congenital malformations has been reported to increase with the
number of AEDs used in polytherapy [140,141] . For some AEDs, notably VPA [127,142,143] and possibly LTG [144] , the risk in monotherapy
appears to be dose dependent. Comparative studies suggest that the
risk of malformation, in particular of neural tube defects, seems to
be greater with VPA compared with other AEDs [144–146] .
In conclusion, available information indicates that the risk of
congenital malformations is increased among offspring of women
with epilepsy, and that this increase may be attributed largely to the
effects of AEDs. However, the incidence of congenital malformations varies 20-fold across published studies [147,148] , mainly because
of methodological differences. In fact, there are major differences
in the populations studied, the diagnostic criteria used to identify
abnormalities, exclusion criteria and the denominators used to
calculate the risk of malformations. Therefore, although the use
of AEDs during pregnancy is associated with an increased risk for
birth defects, this risk must be balanced against those associated
with uncontrolled seizures. Furthermore, AED-related risks to the
fetus could probably be reduced by appropriate treatment adjustments (e.g., a switch from polytherapy to mono­therapy, avoidance
of VPA when possible and titration to the lowest effective dosage).
Expert commentary
Sexual dysfunction in women suffering from epilepsy is an important issue and in recent years there has been growing evidence to
support the role of both epilepsy per se and different AEDs in the
development of this comorbidity.
Endocrinologic abnormalities related to epilepsy can reasonably be expected in view of the complex and close interconnections between the HPA and the cortex. Focal epileptic discharges (in particular from the temporal lobe) can affect HPA
function. Consequently, the production of LH, FSH, GnRH
and PRL and the concentrations and metabolism of their end
products (e.g., estrogen, testosterone and DHEAS) appears to
be modified in epileptic women. Most of the older AEDs (PB,
CBZ and PHT) are known to affect levels of steroid hormones
by enhancing their metabolism through activation of the cytochrome P450 oxidative system, thereby also inducing SHBG
synthesis in hepatocytes.
Moreover, VPA is associated with a frequent occurrence of
reproductive endocrine disorders characterized by polycystic
changes in the ovaries, high serum testosterone concentrations
(hyperandrogenism) and menstrual disorders. Young women with
epilepsy seem to be especially vulnerable to the effects of VPA
on serum androgen levels. It is probable that obesity and related
hyperinsulinemia may exacerbate the VPA-related reproductive
endocrine disorders; in the majority of cases these disorders are
reversible. There is a significant lack of data regarding the effects
of new AEDs.
Physicians should be aware of reproductive endocrine dysfunction that may occur in women with epilepsy during treatment. If a reproductive endocrine disorder is found AEDs
should be reviewed in terms of their indication for the particular seizure type and their tolerability vis-à-vis their potential
for contributing to these endocrine problems.
Expert Rev. Neurother. 9(12), (2009)
Epilepsy, sex hormones & antiepileptic drugs in female patients
Five-year view
It is now clear that both seizures and some old AEDs have detrimental effects on reproductive function in epileptic patients.
In the coming years, understanding the underlying mechanisms
of these effects will provide more information about the direct
influence of the epileptogenic lesion and about the central and
peri­pheral effects on endocrine glands, the metabolism of sex
hormones and binding proteins and the secondary endocrine
complications of AED-related weight changes.
One of the challenges of the next few years is to better define the
endocrine and reproductive effects of the new AEDs, especially in
long-term therapy. These findings will change our policy toward
female patients and enable us to better choose the appropriate
AEDs (particularly in subpopulations at risk, e.g., adolescents,
Review
obese patients and/or patients with previous ovarian dysfunctions). Currently, the concept of ‘personalized’ medicine is receiving much attention; as research in this area advances rapidly, it is
probably that the choice of AEDs will be driven by more accurate
and complete considerations to optimize the treatment of epilepsy
for the individual patients.
Financial & competing interests disclosure
The authors have no relevant affiliations or financial involvement with any
organization or entity with a financial interest in or financial conflict with
the subject matter or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or options, expert
testimony, grants or patents received or pending, or royalties.
No writing assistance was utilized in the production of this manuscript.
Key issues
• Reproductive endocrine disorders, including polycystic ovary syndrome (PCOS), have been associated with epilepsy and the incidence of
menstrual irregularities appears to be more common among women with epilepsy than among women without epilepsy.
• Epilepsy itself may directly influence the endocrine control centers in the brain by altering the release of sex steroid hormones including
the production of luteinizing hormone, follicle-stimulating hormone, gonadotropin-releasing hormone and prolactin and the
concentrations and metabolism of its end products such as estrogen and testosterone.
• In localization-related epilepsy, clinical investigations indicate a close relationship between the occurrence of temporolimbic discharges
and pituitary hormone changes, and an association between epilepsy laterality in temporolimbic epilepsy and the occurrence of certain
reproductive endocrine disorders.
• Left unilateral temporolimbic epilepsy is associated with a higher occurrence of PCOS, while right unilateral temporolimbic epilepsy is
associated with hypothalamic amenorrhea.
• A possible role for the use of antiepileptic drugs as an alternative pathogenic mechanism has been suggested.
• The use of enzyme-inducing antieplileptic drugs, such as phenobarbital, phenytoin and carbamazepine, increases serum sex hormonebinding globulin concentrations leading to diminished bioactivity of estrodiol, which may result in menstrual disorders in some women,
and thus to reduced fertility.
• The inhibition of the aromatase complex (CYP19) activity, induced by phenobarbital, phenytoin, valproic acid (VPA), oxcarbazepine and
lamotrigine, may explain the decreased production of estrodiol observed in vivo.
• In women, the use of valproic acid (VPA), a non-enzyme-inducing antiepileptic drug, appears to be associated with a frequent
occurrence of menstrual disorders, hyperandrogenisn and polycystic changes. These disorders are especially common among women
who have gained weight during VPA treatment.
• The mechanism of VPA-associated hyperandrogenism may result from inhibition of the conversion of testosterone to estrogen induced
by VPA.
• The mechanisms of VPA-induced PCOS are still controversial. VPA-induced weight gain or obesity and the resultant insulin resistance
have been suggested to predispose patients to PCOS.
References
Papers of special note have been highlighted as:
• of interest
•• of considerable interest
1
•
2
Herzog AG, Seibel MM, Schomer DL,
Vaitukaitis JL, Geschwind N.
Reproductive endocrine disorders in
women with partial seizures of temporal
lobe origin. Arch. Neurol. 43, 341–346
(1986).
Shows the lack of a significant
relationship between the occurrence of
menstrual disorders and the use of
antiepileptic drugs (AEDs) in female
epileptic patients.
Isojärvi JI, Laatikainen TJ, Pakarinen AJ,
Juntunen KT, Myllylä VV. Polycystic
www.expert-reviews.com
ovaries and hyperandrogenism in women
taking valproate for epilepsy. N. Engl. J.
Med. 329, 1383–1388 (1993).
•• Excellent study reporting for the first time
a high incidence of menstrual disorders
linked to obesity, hyperandrogenism and
polycystic ovaries in women taking
valproate (VPA).
3
Klüver H, Bucy PC, Klüver H, Bucy PC.
Preliminary analysis of functions of the
temporal lobes in monkeys.
J. Neuropsychiatry Clin. Neurosci. 9,
606–620 (1939).
4
Herzog AG, Fowler KM. Sexual hormones
and epilepsy: threat and opportunities.
Curr. Opin. Neurol. 18, 167–172 (2005).
5
Isojärvi JI, Taubøll E, Herzog AG. Effect of
antiepileptic drugs on reproductive
endocrine function in individuals with
epilepsy. CNS Drugs 19, 207–223 (2005).
6
de Vries L, Karasik A, Landau Z, Phillip M,
Kiviti S, Goldberg-Stern H. Endocrine
effects of valproate in adolescent girls with
epilepsy. Epilepsia 48, 470–477 (2007).
•
Long-term treatment with VPA in
adolescent girls is associated with
increased testosterone levels after
menarche, without clinical
hyperandrogenism or polycystic ovary
syndrome (PCOS).
7
Wallace H, Shorvon S, Tallis R. Agespecific incidence and prevalence rates of
treated epilepsy in an unselected
1809
Review
Verrotti, D’Egidio, Coppola, Parisi & Chiarelli
population of 2,052,922 and age-specific
fertility rates of women with epilepsy.
Lancet 352, 1970–1973 (1998).
8
9
Dansky LV, Andermann E, Andermann F.
Marriage and fertility in epileptic patients.
Epilepsia 21, 261–271 (1980).
Webber MP, Hauser WA, Ottman R,
Annegers JF. Fertility in persons with
epilepsy: 1935–1974. Epilepsia 27, 746–752
(1986).
21
Bilo L, Meo R, Nappi C et al. Reproductive
endocrine disorders in women with
primary generalized epilepsy. Epilepsia
29, 612–619 (1988).
22
Nappi C, Meo R, Di Carlo C, Estraneo A,
Bilo L. Reuced fertility and
neuroendocrine dysfunction in women
with epilepsy. Gynecol. Endocrinol. 8,
133–145 (1994).
23
Scharfman HE, Kim M, Hintz TM,
MacLusky NJ. Seizures and reproductive
function: insights from female rats with
epilepsy. Ann. Neurol. 64, 687–697
(2008).
10
Hamed SA. Neuroendocrine hormonal
conditions in epilepsy: relationship to
reproductive and sexual functions.
Neurologist 14, 157–169 (2008).
11
Morrell MJ, Giudice L, Flynn KL et al.
Predictors of ovulatory failure in women
with epilepsy. Ann. Neurol. 52, 704–711
(2002).
24
12
Morrell MJ. Reproductive and metabolic
disorders in women with epilepsy. Epilepsia
44, 11–20 (2002).
25
13
Herzog AG, Friedman MN. Menstrual
cycle interval and ovulation in women with
localization-related epilepsy. Neurology 57,
2133–2135 (2002).
Feeney DM, Gullotta FP, Gilmore W.
Hyposexuality produced by temporal lobe
epilepsy in the cat. Epilepsia 39, 140–149
(1998).
26
14
Herzog AG, Coleman AE, Jacobs AR et al.
Interictal EEG discharges, reproductive
hormones, and menstrual disorders in
epilepsy. Ann. Neurol. 54, 625–637 (2003).
Sperling MR, Pritchard PB, Engel J, Daniel
C, Sagel J. Prolactin in partial epilepsy: an
indicator of limbic seizures. Ann. Neurol.
20, 716–722 (1986).
27
•
Shows that the laterality and focality of
epilepsy may be important determinants
of reproductive endocrine function.
Hum KM, Megna S, Burnham WM.
The effects of right and left amygdala
kindling on the female reproductive
system in rats. Epilepsia 50, 880–886
(2009).
28
Herzog AG. A relationship between
particular reproductive endocrine disorders
and the laterality of epileptiform discharges
in women with epilepsy. Neurology 43,
1907–1910 (1993).
15
Klein P, Serje A, Pezzullo JC. Premature
ovarian failure in women with epilepsy.
Epilepsia 42, 1584–1589 (2001).
16
Harden CL, Koppel BS, Herzog AG,
Nikolov BG, Hauser WA. Seizure
frequency is associated with age at
menopause in women with epilepsy.
Neurology 61, 451–455 (2003).
17
Hamed SA, Hamed EA, Shokry M,
Omar H, Abdellah MM. The reproductive
conditions and lipid profile in females with
epilepsy. Acta Neurol. Scand. 115, 112–122
(2007).
18
19
20
Bauer J, Isojärvi JI, Herzog AG et al.
Reproductive dysfunction in women with
epilepsy: recommendations for evaluation
and management. J. Neurol. Neurosurg.
Psychiatr. 73, 121–125 (2002).
Stoffel-Wagner B, Bauer J, Flügel D,
Brennemann W, Klingmüller D, Elger CE.
Serum sex hormones are altered in patients
with chronic temporal lobe epilepsy
receiving anticonvulsant medication.
Epilepsia 39, 1164–1173 (1998).
Morrell MJ. Epilepsy in women. Am. Fam.
Physician 66, 1489–1494 (2002).
1810
29
Edwards HE, Burnham WM, Ng MM,
Asa S, MacLusky NJ. Limbic seizures alter
reproductive function in the female rat.
Epilepsia 40, 1370–1377 (1999).
31
Bazán AC, Montenegro MA, Cendes F,
Min LL, Guerreiro CA. Menstrual cycle
worsening of epileptic seizures in women
with symptomatic focal epilepsy.
Arq. Neuropsiquiatr. 63, 751–756 (2005).
35
Bilo L, Meo R, Valentino R, Di Carlo C,
Striano S, Nappi C. Characterization of
reproductive endocrine disorders in women
with epilepsy. J. Clin. Endocrinol. Metab.
86, 2950–2956 (2001).
•
The presence of increased luteinizing
hormone pulse frequency in untreated or
drug-free epileptic women with regular
menstrual cycles supports the notion that
epilepsy itself may contribute to
reproductive abnormalities.
36
Bilo L, Meo, R, Valentino R, Buscaino GA,
Striano S, Nappi C. Abnormal pattern of
luteinizing hormone pulsatility in women
with epilepsy. Fertil. Steril. 55, 705–711
(1991).
37
Murialdo G, Magri F, Tamagno G et al.
Seizure frequency and sex steroids in
women with partila epilepsy on
antiepileptic therapy. Epilepsia 50,
1920–1926 (2009).
•• Excellent cross-sectional study reporting
more relevant changes in sex hormone
levels in patients with more
frequent seizures.
38
Svalheim S, Taubøll E, Bjørnenak T et al.
Do women with epilepsy have increased
frequency of menstrual disturbances?
Seizure 12, 529–533 (2003).
39
Spratt DI, Finkelstein JS, Butler JP,
Badger TM, Crowley WF. Effects of
increasing the frequency of low doses of
gonadotropin-releasing hormone (GnRH)
on gonadotropin secretion in GnRHdeficient men. J. Clin. Endocrinol. Metab.
64, 1179–1186 (1987).
40
Herzog AG. A hypothesis to integrate
partial seizures of temporal lobe origin and
reproductive endocrine disorders. Epilepsy
Res. 3, 151–159 (1989).
41
Klüver H, Bucy PC, Klüver H, Bucy PC.
Preliminary analysis of functions of the
temporal lobes in monkeys. 1939.
J. Neuropsychiatry Clin. Neurosci. 9,
606–620 (1997).
42
Rao ML, Stefen H, Bauer J. Epileptic but
not psychogenic seizures are accompained
by simultaneous elevation of serum
pituitary hormones and cortisol levels.
Neuroendocrinology 49, 33–39 (1989).
43
Herzog AG. Polycystic ovarian syndrome
in women with epilepsy: epileptic or
iatrogenic? Ann. Neurol. 39, 559–560
(1996).
Löfgren E, Mikkonen K, Tolonen U et al.
Reproductive endocrine function in women
with epilepsy: the role of epilepsy type and
medication. Epilepsy Behav. 10, 77–83
(2007).
•• Excellent study that demonstrated the
relationship between epilepsy treatment,
medication and reproductive disorders.
30
34
Meo R, Bilo L, Nappi C et al.
Derangement of the hypothalamic GnRH
pulse generator in women with epilepsy.
Seizure 2, 241–252 (1993).
Drislane FW, Coleman AE, Schomer DL
et al. Altered pulsatile secretion of
luteinizing hormone in women with
epilepsy. Neurology 44, 306–310 (1994).
32
Cummings LN, Giudice L, Morrell MJ.
Ovulatory function in epilepsy. Epilepsia
36, 355–359 (1995).
33
Herzog AG. Menstrual disorders in women
with epilepsy. Neurology 66, S23–S28
(2006).
Expert Rev. Neurother. 9(12), (2009)
Epilepsy, sex hormones & antiepileptic drugs in female patients
44
Daniele A, Azzoni A, Bizzi A, Rossi A,
Gainotti G, Mazza S. Sexual behavior and
hemispheric laterality of the focus in
patients with temporal lobe epilepsy. Biol.
Psychiatry 42, 617–624 (1997).
45
Baird AD, Wilson SJ, Bladin PF,
Saling MM, Reutens DC. Sexual outcome
after epilepsy surgery. Epilepsy Behav.
4268–4278 (2003).
•
Suggests that right temporal resection
could improve sexual functioning.
46
Herzog AG, Coleman AE, Jacobs AR et al.
Relationship of sexual dysfunction to
epilepsy laterality and reproductive
hormone levels in women. Epilepsy Behav.
4, 407–413 (2003).
47
Herzog AG, Schachter SC. On the
association between valproate and
polycystic ovary syndrome Epilepsia 42,
311–315 (2001).
48
Kalinin VV, Zheleznova EV. Chronology
and evolution of temporal lobe epilepsy and
endocrine reproductive dysfunction in
women: relationships to side of focus and
catameniality. Epilepsy Behav. 11, 185–191
(2007).
49
Harden CL. Sexual dysfunction in women
with epilepsy. Seizure 17, 131–135 (2008).
50
Crawford P, Appleton R, Betts T,
Duncan J, Guthrie E, Morrow J. Best
practice guidelines for the management of
women with epilepsy. The Women with
Epilepsy Guidelines Development group.
Seizure 8, 201–217 (1999).
51
Bauer J, Burr W, Elger CE. Seizure
occurrence during ovulatory and
anovulatory cycles in patients with
temporal lobe epilepsy: a prospective study.
Eur. J. Neurol. 5, 83–88 (1998).
52
Morrell MJ, Sperling MR, Stecker M,
Dichter MA. Sexual dysfunction in partial
epilepsy: a deficit in physiologic sexual
arousal. Neurology 44, 243–247 (1994).
53
54
55
56
Bauer J, Cooper-Mahkorn D. Reproductive
dysfunction in women with epilepsy:
menstrual cycle abnormalities, fertility, and
polycystic ovary syndrome. Int. Rev.
Neurobiol. 83, 135–155 (2008).
Manna I, Labate A, Gambardella A et al.
Serotonin transporter gene (5-Htt):
association analysis with temporal lobe
epilepsy. Neurosci. Lett. 421, 52–56 (2007).
Dana-Haeri J, Oxley J, Richen A. Reduction
of free testosterone by antiepileptic drugs.
Br. Med. J. 284, 85–86 (1982).
Beastall GH, Cowan RA, Gray JM,
Fogelman I. Hormone binding globulins
and anticonvulsant therapy. Scott. Med. J.
30, 101–105 (1985).
www.expert-reviews.com
57
Victor A, Lundberg PO, Johansson ED.
Induction of sex hormone binding globulin
by phenytoin. Br. Med. J. 2, 934–935
(1977).
58
Levesque LA, Herzog AG, Seibe MM.
The effect of phenytoin and carbamazepine
on serum dehydroepiandrosterone sulfate
in men and women who have partial
seizures with temporal lobe involvement.
J. Clin. Endocrinol. Metab. 63, 243–245
(1986).
59
Galimberti CA, Magri F, Copello F et al.
Changes in sex steroid levels in women
with epilepsy on treatment: relationship
with antiepileptic therapies and seizure
frequency. Epilepsia 50, 28–32 (2009).
•• Excellent observational study that showed
decreased sex steroid levels correlated
with AED treatments, mainly when
enzyme-inducing AED polytherapies
were given.
60
Murialdo G, Galimberti CA, Gianelli MV
et al. Effects of valproate, phenobarbital,
and carbamazepine on sex steroid setup in
women with epilepsy. Clin.
Neuropharmacol. 21, 52–58 (1998).
61
Herzog AG. Disorders of reproduction in
patients with epilepsy: primary
neurological mechanisms. Seizure 17,
101–110 (2008).
62
Isojärvi JI. Serum steroid hormones and
pituitary function in female epileptic
patients during carbamazepine therapy.
Epilepsia 31(4), 438–445 (1990).
63
Galimberti CA, Magri F, Copello F et al.
Seizure frequency and cortisol and
dehydroepiandrosterone sulfate (DHEAS)
levels in women with epilepsy receiving
antiepileptic drug treatment. Epilepsia 46,
517–523 (2005).
64
Mejias-Aponte CA, Jimenz-Rivera CA,
Segarra AC. Sex differences in models of
temporal lobe epilepsy: role of testosterone.
Brain Res. 944, 210–218 (2002).
65
Jacobsen NW, Halling-Sorensen B,
Birkved K. Inhibition of human aromatase
complex (CYP19) by antiepileptic drugs.
Toxicol. In Vitro 22, 146–143 (2008).
66
67
Taubøll E, Gregoraszczuk LE, Wojtowicz
AK, Milewicz T. Effects of levetiracetam
and valproate on reproductive endocrine
function studied in human ovarian
follicular cells. Epilepsia 50, 1868–1874
(2009).
Murialdo G, Galimberti CA, Magri F et al.
Menstrual cycle and ovary alterations in
women with epilepsy on antiepileptic
therapy. J. Endocrinol. Invest. 20, 519–526
(1997).
Review
•
Good study that did not show an
increased frequency of menstrual
disorders or hyperandrogenism and
polycystic ovaries in women taking
carbamazepine (CBZ).
68
Rättyä J, Pakarinen AJ, Knip M et al. Early
hormonal changes during valproate or
carbamazepine treatment: a 3-month study.
Neurology 57, 440–444 (2001).
69
Isojärvi JI, Laatikainen TJ, Pakarinen AJ,
Juntunen KT, Myllylä VV. Menstrual
disorders in women with epilepsy receiving
carbamazepine. Epilepsia 36, 676–681
(1995).
•
Longitudinal study reporting that
menstrual disorders in female patients on
CBZ treatment with normal ovaries are
associated with increased serum sex
hormone-binding globulin (SHBG) levels
and low estradiol/SHBG ratio.
70
Isojärvi JI, Laatikainen TJ, Knip M,
Pakarinen AJ, Juntunen KT, Myllylä VV.
Obesity and endocrine disorders in women
taking valproate for epilepsy. Ann. Neurol.
39, 579–584 (1996).
71
Lossius MI, Taubøll E, Mowinckel P,
Mørkrid L, Gjerstad L. Reversible effects of
antiepileptic drugs on reproductive
endocrine function in men and women
with epilepsy – a prospective randomized
double-blind withdrawal study. Epilepsia
48, 1875–1882 (2007).
72
Dana-Haeri J, Oxley J, Richens A.
Pituitary responsiveness to gonadotrophinreleasing and thyrotrophin-releasing
hormones in epileptic patients receiving
carbamazepine or phenytoin. Clin.
Endocrinol. (Oxf.) 20, 163–168 (1984).
73
Isojärvi JI, Myllylä VV, Pakarinen AJ.
Effects of carbamazepine on pituitary
responsiveness to luteinizing hormonereleasing hormone, thyrotropin-releasing
hormone, and metoclopramide in
epileptic patients. Epilepsia 30, 50–56
(1989).
74
Bonuccelli U, Murialdo G, Martino E et al.
Effects of carbamazepine on prolactin
secretion in normal subjects and in epileptic
subjects. Clin. Neuropharmacol. 8, 165–174
(1985).
75
Isojärvi JI, Taubøll E, Pakarinen AJ et al.
Altered ovarian function and
cardiovascular risk factors in valproatetreated women. Am. J. Med. 111, 290–296
(2001).
76
Bauer J, Jarre A, Klingmüller D, Elger CE.
Polycystic ovary syndrome in patients with
focal epilepsy: a study in 93 women.
Epilepsy Res. 41, 163–167 (2000).
1811
Review
77
78
79
80
81
82
83
Verrotti, D’Egidio, Coppola, Parisi & Chiarelli
Stephen LJ, Kwan P, Shapiro D,
Dominiczak M, Brodie MJ. Hormone
profiles in young adults with epilepsy
treated with sodium valproate or
lamotrigine monotherapy. Epilepsia 42,
1002–1006 (2001).
87
Betts T, Yarrow H, Dutton N, Greenhill L,
Rolfe T. A study of anticonvulsant
medication on ovarian function in a group
of women with epilepsy who have only ever
taken one anticonvulsant compared with a
group of women without epilepsy. Seizure
12, 323–329 (2003).
88
Prabhakar S, Sahota P, Kharbanda PS et al.
Sodium valproate, hyperandrogenism and
altered ovarian function in Indian women
with epilepsy: a prospective study. Epilepsia
48, 1371–1377 (2007).
Morrel M, Bhatt M, Ozkara C et al.
Higher incidence of symptoms of polycystic
ovary syndrome in women with epilepsy
treated with valproate versus lamotrigine.
Neurology 64, 428 (2005).
Kwan P, Yip FP, Hui AC et al. Effects of
valproate or lamotrigine monotherapy on
the reproductive endocrine and insulinrelated metabolic profile in Chinese adults
with epilepsy: a prospective randomized
study. Epilepsy Behav. 14, 610–616
(2009).
Stephen LJ, Sills GJ, Leach JP et al. Sodium
valproate versus lamotrigine: a randomised
comparison of efficacy, tolerability and
effects on circulating androgenic hormones
in newly diagnosed epilepsy. Epilepsy Res.
75, 122–129 (2007).
84
Verrotti A, Ciampani M, Pomilio M,
Latini G, Chiarelli F. Endocrine
abormalities induced by antiepileptic
drugs. Curr. Pediatr. Rev. 2, 85–92 (2006).
85
Death AK, McGrath KC, Handelsman DJ.
Valproate is an anti-androgen and
anti-progestin. Steroids 70, 946–953
(2005).
86
Lydon JP, DeMayo FJ, Conneely OM,
O’Malley BW. Reproductive phenotpes of
the progesterone receptor null mutant
mouse. J. Steroid Biochem. Mol. Biol. 56,
67–77 (1996).
1812
Löfgren E, Tapanainen JS, Koivunen R,
Pakarinen A, Isojärvi JI. Effects of
carbamazepine and oxcarbazepine on the
reproductive endocrine function in women
with epilepsy. Epilepsia 47, 1441–1446
(2006).
89
Isojärvi JI, Rättyä J, Myllylä VV et al.
Valproate, lamotrigine, and insulinmediated risks in women with epilepsy.
Ann. Neurol. 43, 446–451 (1998).
90
Morrell MJ, Isojärvi J, Taylor AE et al.
Higher androgens and weight gain with
valproate compared with lamotrigine for
epilepsy. Epilepsy Res. 54, 189–199 (2003).
91
Opaleke A, Helmers SL. Hormonal
consequences of epilepsy. Semin. Pediatr.
Neurol. 14, 189–195 (2007).
92
Sahota P, Prabhakar S, Kharbanda PS et al.
Seizure type, antiepileptic drugs, and
reproductive endocrine dysfunction in
Indian women with epilepsy: a crosssectional study. Epilepsia 49, 2069–2077
(2008).
93
Gorkemli H, Genc BO, Dogan E, Genc E,
Ozdemir S. Long-term effects of valproic
acid on reproductive endocrine functions in
Turkish women with epilepsy. Gynecol.
Obstet. Invest. 67, 223–227 (2009).
94
Vainionpää LK, Rättyä J, Knip M et al.
Valproate-induced hyperandrogenism
during pubertal maturation in girls with
epilepsy. Ann. Neurol. 45, 444–450 (1999).
•• Demonstrates an increasing frequency of
hyperandrogenism with pubertal
development, suggesting that the
sensitivity to VPA-induced
hyperandrogenism is a function of
sexual maturation.
Rättya J, Vainionpää L, Knip M,
Lanning P, Isojärvi JI. The effects of
valproate, carbamazepine, and
oxcarbazepine on growth and sexual
maturation in girls with epilepsy. Pediatrics
103, 588–593 (1999).
Luef G, Abraham I, Trinka E et al.
Hyperandrogenism, postprandial
hyperinsulinism and the risk of PCOS in a
cross sectional study of women with
epilepsy treated with valproate. Epilepsy Res.
48, 91–102 (2002).
95
Genton P, Bauer J, Duncan S et al. On the
association between valproate and
polycystic ovary syndrome. Epilepsia 42,
295–304 (2001).
96
Luef G, Abraham I, Haslinger M et al.
Polycystic ovaries, obesity and insulin
resistance in women with epilepsy. A
comparative study of carbamazepine and
valproic acid in 105 women. J. Neurol. 249,
835–841 (2002).
97
Azziz R, Woods KS, Reyna R, Key TJ,
Knochenhauer ES, Yildiz BO. The
prevalence and features of the polycystic
ovary syndrome in an unselected
population. J. Clin. Endocrinol. Metab. 89,
2745–2749 (2004).
98
Polson DW. Polycystic ovary syndrome and
epilepsy – a gynaecological perspective.
Seizure 12, 397–402 (2003).
99
Rasgon N. The relationship between
polycystic ovary syndrome and antiepileptic
drugs: a review of the evidence. J. Clin.
Psychopharmacol. 24, 322–334 (2004).
100
Duncan S. Polycystic ovarian syndrome in
women with epilepsy: a review. Epilepsia
42, 60–65 (2001).
101
Betts T, Dutton N, Yarrow H. Epilepsy and
the ovary (cutting out the hysteria). Seizure
10, 220–328 (2001).
102
Verrotti A, Basciani F, Morresi S,
de Martino M, Morgese G, Chiarelli F.
Serum leptin changes in epileptic patients
who gain weight after therapy with
valproic acid. Neurology 53, 230–232
(1999).
103
Verrotti A, la Torre R, Trotta D, Mohn A,
Chiarelli F. Valproate-induced insulin
resistance and obesity in children. Horm.
Res. 71, 125–131 (2009).
104
Jallon P, Picard F. Bodyweight gain and
anticonvulsants: a comparative review.
Drug Saf. 24, 969–978 (2001).
105
Biton V, Mirza W, Montouris G, Vuong A,
Hammer AE, Barrett PS. Weight change
associated with valproate and lamotrigine
monotherapy in patients with epilepsy.
Neurology 56, 172–177 (2001).
106
Wirrell EC. Valproic acid-associated weight
gain in older children and teens with
epilepsy. Pediatr. Neurol. 28, 126–129
(2003).
107
Mikkonen K, Vainionpää LK,
Pakarinen AJ et al. Long-term reproductive
endocrine health in young women with
epilepsy during puberty. Neurology 62,
445–450 (2004).
•• Demonstrates the reversibility of the
reproductive endocrine effects of VPA
after discontinuation of the treatment.
108
Egger J, Brett EM. Effects of sodium
valproate in 100 children with special
reference to weight. Br. Med. J. 283,
577–581 (1981).
109
Dinesen H, Gram L, Andersen T, Dam M.
Weight gain during treatment with
valproate. Acta Neurol. Scand. 70, 65–69
(1984).
110
Breum L, Astrup A, Gram L et al.
Metabolic changes during treatment with
valproate in humans: implication for
untoward weight gain. Metab. Clin. Exp.
41, 666–670 (1992).
111
Gidal BE, Anderson GD, Spencer NW
et al. Valproate-associated weight gain:
potential relation to energy expenditure
and metabolism in patients with epilepsy.
J. Epilepsy 9, 234–241 (1996).
Expert Rev. Neurother. 9(12), (2009)
Epilepsy, sex hormones & antiepileptic drugs in female patients
112
Demir E, Aysun S. Weight gain associated
with valproate in childhood. Pediatr.
Neurol. 22, 361–364 (2000).
113
Covanis A, Gupta AK, Jeavons PM.
Sodium valproate: monotherapy and
polytherapy. Epilepsia 23, 693–720 (1982).
114
115
116
125
Pylvänen V, Knip M, Pakarinen A, Kotila
M, Turkka J, Isojärvi JI. Serum insulin and
leptin levels in valproate-associated obesity.
Epilepsia, 43, 514–517 (2002).
126
Dunaif A. Insulin resistance and the
polycystic ovary syndrome: mechanism and
implications for pathogenesis. Endocr. Rev.
18, 774–800 (1997).
127
Luef GJ, Lechleitner M, Bauer G,
Trinka E, Hengster P. Valproic acid
modulates islet cell insulin secretion: a
possible mechanism of weight gain in
epilepsy patients. Epilepsy Res. 55, 53–58
(2003).
128
Teramo K, Hiilesmaa V. Pregnancy and
fetal complications in epileptic pregnancies.
In: Epilepsy, Pregnancy, and the Child.
Janz D, Dam M, Bossi L, Helge H,
Richens A, Schmidt D (Eds). Raven, NY,
USA 53–59 (1982).
The EURAP study group. Seizure control
and treatment in pregnancy: observations
from the EURAP Epilepsy Pregnancy
Registry. Neurology 66, 354–360 (2006).
Katz O, Levy A, Wizniter A et al.
Pregnancy and perinatal outcome in
epileptic women: a population-based study.
J. Matern. Fetal Neonatal Med. 19, 21–25
(2006).
Richmond JR, Krishnamoorthy P,
Andermann E, Benjamin A. (2004)
Epilepsy and pregnancy: an obstetric
perspective. Am. J. Obstet. Gynecol. 190,
371–379 (2004).
Olafsson E, Hallgrimsson JT, Hauser WA.
Pregnancies of women with epilepsy: a
population-based study in Iceland.
Epilepsia 39, 887–892 (1998).
Review
138
Sabers A, Dam M, Rogvi-Hansen B,
Boas J, Sidenius P, Laue FM. Epilepsy and
pregnancy: lamotrigine as main drug used.
Acta Neurol. Scand. 10, 9–13 (2004).
139
Aguglia U, Barboni G, Battino D et al.
Italian Consensus Conference on Epilepsy
and Pregnancy, Labor and Puerperium.
Epilepsia 50, 7–23 (2009).
140
Tomson T. Gender aspects of
pharmacokinetics of new and old AEDs:
pregnancy and breast-feeding. Ther. Drug
Monit. 27, 718–721 (2005).
141
Veiby G, Daltveit KA, Engelsen AB,
Gilhus EN. Pregnancy, delivery, and
outcome for the child in maternal epilepsy.
Epilepsia 50, 2130–2139 (2009).
142
Artama M, Auvinen A, Raudaskoski T,
Isojarvi I, Isojarvi J. Antiepileptic drug use
of women with epilepsy and congenital
malformations in offspring. Neurology 64,
1874–1878 (2005).
143
Vajda FJ, Eadie MJ. Maternal valproate
dosage and foetal malformations. Acta
Neurol. Scand. 112, 137–143 (2005).
144
Morrow J, Russell A, Guthrie E et al.
Malformation risks of antiepileptic drugs
in pregnancy: a prospective study from the
UK Epilepsy and Pregnancy Register.
J. Neurol. Neurosurg. Psychiatry 77,
193–198 (2006).
Wong HY, Chu TS, Lai JC et al. Sodium
valproate inhibits glucose transport and
exacerbates Glut-1 deficiency in vitro.
J. Cell. Biochem. 96, 775–785 (2005).
129
Gregoraszczuk E, Wójtowicz AK, Taubøll
E, Ropstad E. Valproate-induced
alterations in testosterone, estradiol and
progesterone secretion from porcine
follicular cells isolated from small- and
medium-sized ovarian follicles. Seizure 9,
480–485 (2000).
130
Fairgrieve SD, Jackson M, jonas P et al.
Population based, prospective study of the
care of women with epilepsy in pregnancy.
BMJ 321, 674–675 (2000).
131
Crawford P. Best practice guidelines for the
management of women with epilepsy.
Epilepsia 46, 117–124 (2005).
145
119
Taubøll E, Isojärvi JI, Flinstad Harbo H,
et al. Long-term valproate treatment
induces changes in ovarian morphology
and serum sex steroid hormone levels in
female Wistar rats. Seizure 8, 490–493
(1999).
132
Viinikainen K, Heinonen S, Eriksson K,
Kalviainen R. Community-based,
prospective, controlled study of obstetric
and neonatal outcome of 179 pregnancies
in women with epilepsy. Epilepsia 47,
186–192 (2006).
Wide K, Winbladh B, Kallen B. Major
malformations in infants exposed to
antiepileptic drugs in utero, with emphasis
on carbamazepine and valproic acid: a
nation-wide, population-based register
study. Acta Paediatr. 9, 174–176 (2004).
146
120
Røste LS, Taubøll E, Berner A, Isojärvi JI,
Gjerstad L. Valproate, but not lamotrigine,
induces ovarian morphological changes in
Wistar rats. Exp. Toxicol. Pathol. 52,
545–552 (2001).
133
Wyszynski DF, Nambisan M, Surve T,
Alsdorf RM, Smith CR, Holmes LB.
Increased rate of major malformations in
offspring exposed to valproate during
pregnancy. Neurology 64, 961–965 (2005).
147
121
Isojärvi J. Disorders of reproduction in
patients with epilepsy: antiepileptic drug
related mechanisms. Seizure 17, 111–119
(2008).
Harden LH, Hopp J, Ting YT et al.
Management issues for women with
epilepsy – focus on pregnancy (an
evidence-based review): I. Obstetrical
complications and change in seizure
frequency. Epilepsia 50, 1229–1236 (2009).
Barrett C, Richens A. Epilepsy and
pregnancy: report of an Epilepsy Research
Foundation Workshop. Epilepsy Res. 52,
147–187 (2003).
148
Olafsson E, Hauser WA, Gudmundsson G.
Fertility in patients with epilepsy: a
population-based study. Neurology 51,
71–73 (1998).
149
Harden CL. Sexuality in women with
epilepsy. Epilepsy Behav 7, S2–S6 (2005).
150
Bauer J, Stefan H, Schrell U, Sappke U,
Uhlig B. Neurophysiologic principles and
clinical value of post-convulsive serum
prolactin determination in epileptic seizure.
Fortschr. Neurol. Psychiatr. 57, 457–468
(1989).
151
Bauer J. Epilepsy and prolactin in adults: a
clinical review. Epilepsy Res. 24, 1–7 (1996).
117
118
122
123
124
Adab N, Kini U, Vinten J et al. The long
term outcome of children born to mothers
with epilepsy. J. Neurol. Neurosurg.
Psychiatry 75, 1575–1583 (2004).
Teramo K, Hiilesmaa V, Bardy A,
Saarikoski S. Fetal heart rate during a
maternal grand mal epileptic seizure.
J. Perinat. Med. 7, 3–6 (1979).
Tomson T, Battino D. Pharmacokinetics
and therapeutic drug monitoring of newer
antiepileptic drugs during pregnancy and
puerperium. Clin. Pharmacokinet. 46,
209–219 (2007).
www.expert-reviews.com
134
135
Fried S, Kozer E, Nulman I, Einarson TR,
Koren G. Malformation rates in children of
women with untreated epilepsy: a metaanalysis. Drug Saf. 27, 197–202 (2004).
Fonager K, Larsen H, Pedersen L,
Sørensen HT. Birth outcomes in women
exposed to anticonvulsant drugs. Acta
Neurol. Scand. 101, 289–294 (2000).
136
Holmes LB, Harvey EA, Coull BA et al.
The teratogenicity of anticonvulsant
drugs. N. Engl. J. Med. 344, 1132–1138
(2001).
137
Kaaja E, Kaaja R, Matila R, Hiilesmaa V.
Enzyme-inducing antiepileptic drugs in
pregnancy and the risk of bleeding in the
neonate. Neurology 58, 549–553 (2002).
1813
Review
Verrotti, D’Egidio, Coppola, Parisi & Chiarelli
152
Duncan S, Blacklaw J, Beastall GH,
Brodie MJ. Sexual function in women with
epilepsy. Epilepsia 38, 1074–1081 (1997).
153
Morrell MJ, Guldner GT. Self-reported
sexual function and sexual arousability in
women with epilepsy. Epilepsia 37,
1204–1210 (1996).
•
•
Affiliations
•
Alberto Verrotti
Department of Pediatrics, University of
Chieti, Via dei Vestini 5, 66100 Chieti,
Italy
Tel.: +39 087 135 8015
Fax: +39 087 157 4831
[email protected]
1814
Claudia D’Egidio
Department of Pediatrics, University of
Chieti, Via dei Vestini 5, 66100 Chieti,
Italy
Tel.: +39 087 135 8015
Fax: +39 087 157 4831
[email protected]
Giangennaro Coppola
Clinic of Child Neuropsychiatry, Second
University of Naples, Italy
Tel.: +39 081 566 6695
Fax: +39 081 566 6694
[email protected]
•
Pasquale Parisi
Department of Child Neurology and Sleep
Centre, University of Rome, Italy
Tel.: +39 068 213 529
Fax: +39 068 245 556l
•
Francesco Chiarelli
Department of Pediatrics, University of
Chieti, Via dei Vestini 5, 66100 Chieti,
Italy
Tel.: +39 087 135 8015
Fax: +39 087 157 4831
[email protected]
Expert Rev. Neurother. 9(12), (2009)