You are on page 1of 10

Clinical Neurology and Neurosurgery 112 (2010) 110

Contents lists available at ScienceDirect

Clinical Neurology and Neurosurgery


journal homepage: www.elsevier.com/locate/clineuro

Review

Bone and calcium metabolism and antiepileptic drugs


Alberto Verrotti a, , Giangennaro Coppola b , Pasquale Parisi c , Angelika Mohn a , Francesco Chiarelli a
a
Department of Pediatrics, University of Chieti, Italy
b
Department of Child Neuropsychiatry, University of Naples, Italy
c
II University of Rome La Sapienza, Italy

a r t i c l e i n f o a b s t r a c t

Article history: There is increasing evidence suggesting that epilepsy and its treatment can affect bone mineralization
Received 2 April 2009 and calcium metabolism. Many studies have shown a signicant reduction in bone mineral density
Received in revised form 21 August 2009 in patients treated with classic (phenobarbital, carbamazepine, valproate, etc.) and with new (oxcar-
Accepted 10 October 2009
bazepine, gabapentin) antiepileptic drugs. In spite of data about the possible effects of the antiepileptic
Available online 12 November 2009
drugs on calcium metabolism, the mechanisms of this important side effect remain to be dened. The
abnormalities of calcium metabolism were thought to result from the cytochrome P450 enzyme-inducing
Keywords:
properties of some antiepileptic drugs and the resultant reduction in vitamin D levels, but the effect of
Bone mineral density
Calcium
many medications (e.g., valproate) cannot be readily explained by vitamin D metabolism.
Vitamin D In this article, the literature related to the effects of classic and new antiepileptic drugs on bone health
Parathyroid hormone and calcium metabolism is reviewed.
Valproate 2009 Elsevier B.V. All rights reserved.
Carbamazepine
Phenobarbital

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Classic antiepileptic drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Benzodiazepines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2. Carbamazepine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.3. CBZ and BMD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.4. CBZ and bone metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.5. Phenytoin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.6. Phenobarbital . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.7. Primidone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.8. Valproic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
2.9. VPA and BMD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
2.10. VPA and bone metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3. Newer AEDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.1. Gabapentin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.2. Lamotrigine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.3. Levetiracetam . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.4. Oxcarbazepine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.5. Topiramate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.6. Zonisamide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8

Corresponding author at: Department of Pediatrics, University of Chieti, Poli-


clinico Universitario, Colle dellAra Via dei Vestini 5, 66100 Chieti, Italy. Tel.: +39
0871 358015; fax: +39 0871 574831.
E-mail address: averrott@unich.it (A. Verrotti).

0303-8467/$ see front matter 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.clineuro.2009.10.011
2 A. Verrotti et al. / Clinical Neurology and Neurosurgery 112 (2010) 110

1. Introduction [4]. Similarly, Farhat et al. have determined the effect of AED on
vitamin D levels and bone density in ambulatory patients and to
The adverse effects of antiepileptic drugs (AEDs) on bone health compare the effects of enzyme-inducing and noninducing AED
were rst reported nearly four decades ago, and, since then, a grow- (such clonazepam) of single vs. multiple therapy on bone density.
ing body of literature indicates that patients taking AEDs are at No signicant difference in 25OHD levels was observed between
increased risk for low bone mineral density and metabolic bone dis- patients on enzyme-inducing and noninducing AED. In this study
ease including changes in bone turnover, osteoporosis, alterations a signicant proportion (>50%) of patients in both age groups had
in bone quality, and, most importantly, fracture. low 25OHD levels and low BMD in adults of both sexes, indepen-
First of all, it can be useful to focus on the basic bone physiology. dent of vitamin D levels [5]. In contrast, in another study, BZPs did
Maintenance of optimal bone health depends on an adequate sup- not seem to have an effect on BMD [6] and thus did not induce
ply of calcium and on the effects of some hormones. Parathyroid the modest increase in fracture risk observed in a prior study [7]. In
hormone (PTH) is a peptide hormone that can alter serum calcium conclusion, BZP can adversely affect bone health with a consequent
via actions on three target organs: bone, intestinal mucosa and risk of fractures.
kidney. PTH increases bone turnover and causes loss of calcium
from bone through increases in osteoclast number and activ-
2.2. Carbamazepine
ity. This hormone also increases intestinal calcium absorption
and acts in the kidney on the distal tubule to promote calcium
Carbamazepine (CBZ) is one of the front line AEDs for treat-
reabsorption and on the proximal tubule to decrease phosphate
ment of partial seizures as well as secondary generalized seizures in
absorption.
adults and children. It mainly acts on voltage-gated sodium chan-
Vitamin D is metabolized in the liver to 25-hydroxyvitamin
nels that are stabilized in their inactivated state [11,12].
D (25OHD), which is in turn metabolized in the kidney to 1,25-
dihydroxyvitamin D (1,25-(OH)2 D), the biologically active form.
Drugs that increase liver hydroxylases can reduce the amount of 2.3. CBZ and BMD
25OHD. 1,25-(OH)2 D synthesis is stimulated by hypocalcemia, PTH
and hypophosphataemia. Vitamin D stimulates intestinal calcium Several studies on adults and children have shown that CBZ
absorption and promotes mineralization of the skeleton. treatment induces a state of decreased BMD in the lumbar spine
Bone remodeling is a lifelong process by which skeleton is being [5,1319], femoral neck [5,1921], forearm [19,22] and calcaneus
continuously resorbed and replaced to maintain skeletal integrity; [23], but there have been some conicting results, especially in
serum markers of bone formation are serum bone specic alkaline children.
phosphatase (bALP), osteocalcin (OC), carboxy-terminal propep- Two recent studies by Kumandas et al. [15] and Kim et al. [23]
tide of type I procollagen (PICP), amino-terminal propeptide of type showed that the decrease in BMD was related to reduced levels of
III procollagen (PIIINP), and markers of bone resorption are serum vitamin D secondary to the property of the drug to activate spe-
carboxy-terminal telopeptide of type 1 collagen (ICTP) and urinary cic cytochrome P450 (CYP-450) isoenzymes, which are involved
N-telopeptide of type 1 collagen bone (NTx). in vitamin D catabolism [24].
Several studies which assessed bone metabolism status and The work by Kumandas et al. was a cross-sectional, retrospective
bone mineral density (BMD) in adults and children on AEDs have study that examined the effects of at least 2 years of CBZ therapy
shown rather controversial results. on bone mineral density in 33 preadolescent patients. They showed
This review analyzes the main data of literature in order to sum- that these patients had reduced BMD in the lumbar spine [15].
marize the principal side adverse effects on bone induced by the The report by Kim et al. was a longitudinal study that showed
most frequently used AEDs. a signicant reduction in BMD in the right calcaneus in 10 adult
patients after 6 months of CBZ monotherapy [23]. In both studies,
reduced BMD was associated with lower levels of 25(OH)D.
2. Classic antiepileptic drugs In contrast, 6 other cross-sectional studies [5,14,16,19,20,25]
and 2 longitudinal studies [13,21] have demonstrated a lack of
2.1. Benzodiazepines correlation between serum levels of vitamin D and reduced BMD,
suggesting that the loss of bone mass observed in patients treated
The most common benzodiazepines (BZP) used are clonazepam, with CBZ may not be explained by an effect of drug on vitamin D
diazepam and lorazepam. These are distinct allosteric binding metabolism. Rather, it may be due at least in part to direct effects of
sites on the GABAa receptor chloride ionophore to enhance GABA- CBZ on bone cell proliferation, leading to reduced growth of human
mediated increases in chloride conductances [13]. bone cells [20,26]. Among these studies, the largest and most recent
Some studies have demonstrated a limited increase in the risk was a study by El-Hajj Fuleihan et al. [19], who studied 225 ambu-
of fractures, even at very low doses, for several types of BZPs. There latory patients (137 adults and 88 children) treated with different
was a trend toward increasing fracture risk with increasing dose AEDs, including CBZ monotherapy. The results indicated that the
[810], especially in the spine [8]. Also, a trend toward higher frac- adult patients on enzyme-inducing drugs such as CBZ tended to
ture risk was seen with increasing half-life of the drugs. BZPs with have lower BMD in the lumbar spine and total hip compared to
a shorter half-life may thus be preferred to reduce the risk of frac- those on noninducers; BMD was negatively correlated with the
tures, but it should be noted that lower half-life may not completely duration of treatment in adults and with polytherapy in children.
abolish the increased risk of fractures, although the relative risk is In agreement with these results, 2 previous studies [5,20]
rather limited [9]. demonstrated that the duration of CBZ therapy is an independent
A few studies investigate effects of BZP on bone metabolism. predictor of BMD in adults.
Kulak et al. evaluating 58 young adults with epilepsy on chronic Gender differences in the effects of CBZ on bone mineral density
AEDs therapy, have demonstrated signicant abnormalities of bone have been analyzed. In several studies, the effect was more marked
metabolism, characterized by reduced BMD, reduced 25OHD and in female patients [2729]. In contrast, 2 studies [14,21] showed
increased alkaline phosphatase (ALP). Mean results of routine that the incidence of osteopenia was signicantly greater in males
biochemical testing, such as total calcium (Ca), phosphorus (P), than in females. Other studies did not show a sex-related difference
magnesium and PTH, were normal and did not differ statistically [13,19,23].
A. Verrotti et al. / Clinical Neurology and Neurosurgery 112 (2010) 110 3

We must underline that some studies have not shown a sig- the contrary, serum levels of ICTP were signicantly higher in
nicant effect of CBZ monotherapy on bone mineral density female patients [43]. These data are in agreement with a previous
[28,3033]. study by Vlimki et al. [29], who suggested that these differences
In conclusion, most evidence has shown that long-term treat- may be partly explained by lower serum 1,25-dihydroxyvitamin
ment with CBZ has a negative effect on BMD, which may be D (1,25-(OH)2 D) levels found in their female patients treated with
a contributing factor to the increased fracture risk observed in CBZ.
patients with epilepsy, postmenopausal women and older men, and Four other studies carried out on ambulatory epileptic chil-
which may accelerate aging-related osteoporosis [8,34]. Therefore, dren [16,31,39,49] showed an increase of bALP, which is a marker
BMD screening with dual-energy X-ray absorptiometry (DEXA), that is considered to be highly sensitive and specic to increased
with intervals varying depending on the fracture risk of the indi- bone turnover [51]. In particular, a recent longitudinal study [39]
vidual patient, is recommended. showed that epileptic patients using CBZ monotherapy had their
metabolism altered early, as indicated by the elevated activities of
2.4. CBZ and bone metabolism serum bALP isoenzyme after 3 months of treatment, because this
effect was independent of the length of therapy.
Different biochemical alterations of bone metabolism have In conclusion, CBZ cause decreased serum levels of 25OHD,
been reported in association with CBZ treatment, both in adults secondary hyperparathyroidism and increased bone turnover. The
[5,20,23,35,36] and in children [15,31,3739]. mechanisms by which CBZ determines such alterations of bone
These alterations include reduced serum levels of biologically metabolism may be multiple: increased catabolism of vitamin D, a
active vitamin D metabolites, hyperparathyroidism and elevated direct effect on function of bone cells [20] and direct inhibition of
levels of markers of bone turnover, such as markers of bone forma- intestinal calcium transport through a mechanism not mediated by
tion and bone resorption. Serum levels of Ca and P were normal in vitamin D [52]. Therefore, regular screening for hypovitaminosis
most of the studies. and monitoring of biochemical markers of bone turnover during
Low levels of biologically active vitamin D in patients on CBZ the treatment period are advisable. Additionally, prophylaxis with
have been demonstrated in many [23,15,40,35,38,19,41], but not calcium supplements and the use of vitamin D supplementation
in all [13,24,31,37,4244] studies. This effect has been attributed could be considered for all patients with epilepsy upon initiation
to changes in metabolism of vitamin D thought CYP-450 in the of CBZ therapy.
liver [45,46]. Vitamin D deciency associated with the use of CBZ
is likely mediated through the orphan nuclear receptor pregnane 2.5. Phenytoin
X receptor (PXR), which can increase the expression of the CYP-
24. This enzyme catalyzes the conversion of 25OHD to its inactive Phenytoin (PHT) was introduced for the treatment of epilepsy
metabolite (calcitroic acid) [47]. This hypothesis is supported by the in 1938 by Merritt and Putnam [53].
recent cross-sectional, retrospective study by Kumandas et al. [15], This drug plays a major role in altering sodium ion movements
who found signicantly decreased lumbar BMD and serum levels across nerve cell membranes [54,55] and is indicated for treat-
of 25OHD in a group of children who had received CBZ therapy for ing generalized tonicclonic seizures [56,57] and status epilepticus
at least 2 years. Furthermore, these patients had serum ALP and [58,59].
PTH levels signicantly higher than those of the control groups. L1- Cross-sectional studies in adults have shown that epileptic
4 BMD z-scores were negatively correlated with serum PTH levels patients treated with PHT experience reduced BMD at the lumbar
and positively correlated with serum levels of 25OH-vitamin D. In spine [5,27,29,7], femur [5,17,20,27,29,7] and hip [5].
this and other studies [1316,23,42,43], no signicant alterations Among these reports, the most recent is a longitudinal study by
in serum levels of Ca and P were observed. Pack et al. [17]. In this study, the authors reported reduced BMD
Recently, an increase in the markers of bone turnover correlated in their patients that was not related to the duration of therapy.
with use of CBZ has been demonstrated in a number of small studies These results are in agreement with 2 previous studies [27,29]. In
[15,16,29,31,35,39,48,49], and this increase has even been seen in contrast, 2 other studies showed a signicant negative correlation
the presence of normal vitamin D and PTH levels [42,43]. Increased between duration of phenytoin use and BMD [5,20].
bone turnover may be an important contributing mechanism for Multiple biochemical abnormalities of bone metabolism have
bone disease in epileptic patients [20]. It is known that AEDs such been reported in association with PHT treatment.
as CBZ, which induce P-450 enzymes, can accelerate vitamin D Several studies reported hypocalcemia and hypophosphatemia
catabolism. This acceleration could lead to decreased absorption [36,40,6066]; hepatic induction of the CYP-450 enzyme system,
of Ca in the gut. The onset of an intermittent hypocalcemia could leading to increased catabolism of vitamin D is the principal mech-
result in compensatory hyperparathyroidism and consequently anism reported [47,67]. Indeed, many reports revealed reduced
increase bone turnover and loss of cortical bone. In severe cases, levels of serum 25OHD in both adults and children taking PHT
this could lead to long-term loss of bone mass [50]. This hypoth- [5,17,20,29,7,61,68]. Evidence also exists for inhibition of the cel-
esis is consistent with some studies [35,39], but not with others lular response to PTH, caused by the inhibitory effect of PHT on
[13,24,29,42,43]. hormone-sensitive adenylate cyclase activity [69,70]. Fetal rats
Currently, few studies have evaluated effects of AEDs on bone treated with PHT demonstrated an impaired osteoblastic response
metabolism by concurrently measuring the markers of bone for- to PTH [71].
mation and resorption. We previously reported the results of a It has also been shown that PHT interferes with cation trans-
prospective longitudinal study of bone mineral status in epileptic port in many tissues, thereby directly inhibiting intestinal calcium
patients before and after treatment with CBZ [42]. We found higher absorption in rats [72]. This effect might also exist in humans, and
values of serum markers of bone formation such as bALP, OC, PICP, it would lead to lower Ca levels in serum, a frequent nding in
PIIINP, and markers of bone resorption such as ICTP and NTx in patients taking PHT.
patients than in controls. CBZ treatment increases bone turnover, In addition, elevated levels of markers of bone turnover are
and these changes are independent of the serum levels of vitamin found in the serum, such as markers of bone formation and
D metabolites and PTH. bone resorption [17,20,29,61,73,74]. One of the mechanisms that
More recently, we showed that markers of bone formation may determine the amount of bone turnover is secondary hyper-
were signicantly higher in male than in female patients; on parathyroidism, which activates bone resorption and secondarily,
4 A. Verrotti et al. / Clinical Neurology and Neurosurgery 112 (2010) 110

through the coupling phenomenon, bone formation. This hypoth- Abnormalities in Ca and bone metabolism can also be associated
esis is supported by the results of Pack et al. [17], who reported with direct inhibition of intestinal calcium absorption [86].
that epileptic patients treated with PHT had lower serum concen- Several studies evaluating vitamin D levels in ambulatory
trations of 25OHD. This is associated with a biochemical pattern patients produced conicting results [5,74,8993].
consistent with hyperparathyroidism that includes higher PTH, Mosekilde et al. showed that chronic PB therapy was associated
bALP, and urine NTx. In a bone biopsy study [75], osteomalacia was with a mild degree of osteomalacia, which was inversely correlated
found in 53% of patients, along with evidence of increased bone with dietary vitamin D intake. Serum 25-hydroxycholecalciferol
resorption (secondary hyperparathyroidism). (25-HCC) was reduced in epileptic patients compared with a con-
Primary hyperparathyroidism has also been suggested as a pos- trol group, and this reduction was positively correlated with dietary
sible mechanism because male patients with normal vitamin D vitamin D intake but not with the severity of bone changes, indicat-
status had evidence of elevated levels of PTH [29]; hyperparathy- ing that factors other than circulating 25-HCC are responsible for
roidism can primarily activate bone resorption. the development of anticonvulsant osteomalacia [89]. In contrast,
However, direct action of PHT on bone resorption and bone Weinstein et al. demonstrated that hypocalcemia and osteopenia
formation has also been suggested [61]. The latter hypothesis is occurred in spite of normal mean levels of serum 25OHD and 1,25-
supported by animal studies [71,76] and experiments on human (OH)2 D. The indirect relationship between serum concentrations
osteoblast-like bone cells [20], where PHT has been demonstrated of antiepileptic drug and the serum ionized Ca level, and the lack
to inhibit the proliferation of human osteoblast-like cells at con- of correlation with vitamin D metabolite levels, suggested that
centrations equal to therapeutic doses. Signicant bone resorption hypocalcemia was independent of the effect of the drug on vitamin
was found in the calvaria of neonatal mice treated with PHT, as D metabolism. Bone biopsies revealed increased osteoid but nor-
demonstrated by signicantly increased calcium in the medium as mal calcication front formation, accelerated mineralization rate,
compared with controls [76]. These results suggest that both bone and decreased mineralization lag time, which were indicative of
resorption and formation may be affected by PHT. increased skeletal turnover rather than osteomalacia [90]. In the
A nal postulated mechanism of increased bone turnover is cal- same way, Farhat et al. found that over 50% of adults and chil-
citonin deciency. Animal studies suggest a direct inhibitory effect dren/adolescents receiving long-term PB therapy had low 25OHD
of PHT on calcitonin secretion [77,78]. This deciency has been levels, but these levels did not correlate with BMD [5]. In par-
demonstrated both in vitro and in vivo [7981]. ticular, Gissel et al. have observed that BMD reduction is more
Additionally, PHT can cause an increase in vitamin K evident only among those who had used PB for more than 2 years
metabolism, resulting in deciency [26]. Vitamin K is an essential [93].
cofactor for post-translational carboxylation of glutamine residues Aysegl et al. evaluated differences in markers of bone forma-
on -carboxylase (Gla)-containing proteins, including osteocalcin. tion and bone resorption between control patients and epileptic
Gla proteins play an important role in the structure and function of patients receiving chronic anticonvulsant therapy. They found that
the skeleton. Thus, inducing deciency in Gla proteins can poten- only the resorption phase of bone turnover is affected by chronic
tially cause alterations in bone structure, resulting in osteopenia administration of PB [94].
and fractures [78]. bALP isoenzyme levels are found to be higher in children receiv-
In conclusion, PHT can be associated with adverse effects on ing antiepileptic drugs such as PB. Therefore, Voudris et al. suggest
bone mineralization and an increased risk of fractures, especially that the bALP isoenzyme could be used as a marker to monitor bone
in postmenopausal women and older men [8,34]. Therefore, moni- metabolism [49].
toring of BMD by DEXA is recommended for patients at high risk for Moreover, in a recent trial, a laboratorial evaluation of bone and
fracture. PHT can induce several abnormalities in bone metabolism mineral metabolism, including measurements of bALP and ICTP,
including hypocalcemia, hypophosphatemia, reduced serum levels showed an increase of ICTP in patients treated with PB compared
of biologically active vitamin D metabolites, hyperparathyroidism to those not receiving this drug. This increase led to a reduction of
and elevated levels of markers of bone turnover. BMD, as measured by DEXA [95].
These ndings suggest that prophylactic therapy with vitamin Finally, in a recent cross-sectional study of 130 epileptic patients
D given beginning in the earliest phases of anticonvulsant therapy treated with PB, BMD was measured by DEXA and a decrease in
could prevent the possible development of bone disease. bone mineralization was also found [96].
In conclusion, a down-regulation of 25-hydroxylation in
2.6. Phenobarbital patients receiving long-term PB therapy may be responsible
for an increased risk of osteomalacia, bone loss and fractures
Phenobarbital (PB) is an effective anticonvulsant for many kinds [8,88], despite no reported alteration in calcium and phosphorus
of seizures such as tonicclonic and focal seizures, and certain clin- metabolism [13,49,97].
ical epilepsy sub-syndromes [82]. Therefore, vitamin D supplementation should be used routinely.
Like other barbiturates, PB enhances -aminobutyric acid
(GABA)-mediated increases in chloride conductance by prolonging 2.7. Primidone
the duration of channel opening [83].
Studies performed during the early 1970s provided the rst sug- Primidone (PRM), classied as an enzyme-inducing antiepilep-
gestion of the adverse effects of PB on bone with consequent high tic drug, was rst synthesized and used as an anticonvulsant in
risk of fractures [8487]. 1952 [98].
These studies showed that PB was involved in causing rick- It is a desoxyphenobarbital differing from PB in the absence of
ets, which was the result of a decrease in vitamin D levels due to the carbonyl group in position 2 of the pyrimidine ring. Thus, it is
the CYP-450 enzyme-inducing properties of this antiepileptic drug. not a true barbiturate but clinically it is considered to be in that
These changes led to impaired intestinal calcium transport result- group [99].
ing in hypocalcemia, and increased PTH levels mobilized Ca from The likelihood of fractures seems to be minimal, although sev-
bone. Consequently, bone mineralization was reduced. eral studies have shown that PRM, such as PHT and PB, affects bone
More recent studies also show that PB causes induction of and mineral metabolism indirectly since the induction of CYP-450
hepatic microsomal enzymes, resulting in increased catabolism of increases vitamin D metabolism, thereby causing vitamin D insuf-
25OHD and the classic bone changes of osteomalacia [4,88]. ciency or deciency [6,73,100,101].
A. Verrotti et al. / Clinical Neurology and Neurosurgery 112 (2010) 110 5

Another mechanism involves direct action on bone cells, thereby components of bone metabolism. In most studies, there were no
increasing bone resorption and formation. In this way, PRM may statistically signicant differences between groups in the mean
inuence bone turnover [29,43,74]. values of Ca and P. None of the patients had hypocalcemia or
Both mechanisms can be associated with a reduction in BMD hypophosphatemia [15,16,23,28,33,38]; similarly, ratios of uri-
[6,21,30,74]. nary Ca to serum creatinine, and urinary P to serum creatinine
In contrast, Filardi et al. led a study to assess BMD and vitamin D stayed within normal ranges [15]. On the other hand, some
metabolism in patients receiving chronic anticonvulsant therapy, studies found results suggesting that serum Ca concentrations
including PRM therapy, and did not nd any signicant differences can be signicantly lower in subjects receiving VPA [13,14,36].
between patients and controls [97]. Furthermore, Ecevit et al. [37] compared healthy subjects and
Despite the introduction of several newer drugs for the treat- patients receiving VPA therapy, and in the VPA-treated group,
ment of epilepsy, primidone is still a drug that should be considered one-fourth of the patients manifested hypocalcaemia and half of
in certain patients [99]. the patients manifested hypophosphatemia. Tsukahara et al. [14]
found that one patient was hypercalciuric (dened as urinary
2.8. Valproic acid Ca > 0.21 mg/mg Cr). Tubular resorption of P was within the normal
range. Hypercalciuria is at present best explained by an imbal-
Valproic acid (VPA) is a broad-spectrum antiepileptic drug that ance of Ca intake and mineral deposition during skeletal growth
has been in use as a rst line agent for both generalized and partial [14].
seizures [102,103]. VPA blocks voltage-dependent sodium chan- Serum 25OHD concentration is the most commonly used index
nels and modies calcium and potassium conductance [104,105]. of vitamin D status. In patients receiving VPA therapy, biologi-
Administration of VPA increases the activity of the GABA-synthetic cally active vitamin D levels within the normal range have been
enzyme glutamic acid decarboxylase and acts as an inhibitor of demonstrated in a number of studies [14,16,17,21,23,36,109,111].
GABA transaminase [106]. Conversely, in a cross-sectional evaluation conducted on 71
patients (adults and children) who had received anticonvulsant
2.9. VPA and BMD therapy for at least 6 months, the mean 25OHD level was in the
insufcient range. Over one-half of the children in the study had
Some studies of VPA therapy in children and adults found no dif- low vitamin D levels [12]. Furthermore, Tekgul et al. and Nico-
ferences in mean BMD values at both the femur neck and lumbar laidou et al. [13,38] noticed a decreasing trend in all seasons in
spine [13,17,23,31,33,36], while many authors noticed a signi- serum 25OHD concentrations from class 0 (before any anticonvul-
cant reduction of BMD in epileptic patients who used VPA for a sant therapy) to class 3 (third year of therapy) patients receiving
prolonged period [15,16,18,21,28,74,107]. In particular, studies in VPA monotherapy. In many studies, serum levels of intact PTH were
epileptic children found a lower BMD compared with matched con- within the normal ranges [13,14,16,17,31,36,109,111,112]. In one
trols at multiple sites including the femoral neck, radius and lumbar study, compared with reference values, plasma intact PTH was in
spine [5,15,16,108110]. Similar results have been described in the lower limits [14]. In contrast with those results, a few studies
adults [107,111]. One study carried out by Farhat et al. [8] found have shown markedly increased PTH levels [15,23,38]. Increases
that in adults, BMD at all skeletal sites was lower compared with in serum levels of total ALP have been reported in several studies.
young adults; in children, BMD was lower in the spine but higher Babayigit et al. studied healthy children and children with idio-
in the total body. Among these studies, Sheth et al. [18] measured pathic epilepsy who had been treated with VPA for more than
axial (second, third, and fourth lumbar vertebrae) and appendic- 1 year. They found that mean serum ALP concentration was sig-
ular (distal third of radius) BMD in children with uncomplicated nicantly higher in the treated patient group as compared to the
idiopathic epilepsy who had been treated with VPA for more than control subjects [16]. Similar ALP values were seen in other studies
18 months. After correction for gender and age, children treated [28,31,109]. Voudris et al. reported an increase in bALP isoenzyme
with VPA had a 14% and 10% reduction in BMD at the axial and values that correlated with the duration of treatment in children
appendicular sites, respectively, and showed a possible increase receiving VPA, without a concomitant signicant elevation of total
in their risk of osteoporotic fractures. In this study, the reduction ALP values. This result suggests that VPA administration does have
in BMD increased with the duration of VPA therapy. In another an impact on bone mineral metabolism in ambulatory children
study [37], children receiving VPA monotherapy had a mean BMD and that normal total ALP levels could not exclude the possibil-
reduction of 31.9% at the femoral neck region compared with the ity that the respective bone fraction may be high. This isoenzyme,
untreated group. The patients had been receiving VPA monother- but not total ALP values, could therefore be used as a marker for
apy for more than 6 months. Thus, decreased BMD was observed the selection of patients who could benet from an evaluation of
in children with epilepsy who had been treated with VPA, even their bone metabolism prole [49]. In contrast with those results,
though treatment was for a rather short time [28,37]. In one study, Elliott et al. [107] found lower serum ALP. Also, serum ALP was neg-
children with epilepsy who had been receiving VPA and/or LTG atively correlated with serum homocysteine in patients receiving
therapy for more than 2 years had overall reduced stature, low bone VPA.
mass, and reduced bone formation; these outcome measures were OC is a noncollagenous protein synthesized by osteoblasts and
particularly compromised in those children whose physical activ- is specic for bone and dentin. In two studies, the OC levels
ity was low and who received a combined therapy of VPA and LTG of the patient group were signicantly higher than the control
[112]. In conclusion, long-term antiepileptic treatment with VPA group [23,74,108]. Another study demonstrated OC levels at the
may cause osteopenia in both sexes [5,14,16,74,108,111]. A very upper limits of the normal range [31]. In contrast, Tsukahara et
limited increased fracture risk for osteoporotic-related trauma can al. have shown lower values of intact OC [14,109], PICP and ICTP
be present in users of VPA [37]. in pediatric patients receiving long-term VPA monotherapy [14].
Similarly, Song et al. [110] stated that serum OC in children who
2.10. VPA and bone metabolism took VPA was lower relative to normal control patients. Urine val-
ues of deoxypyridinoline showed no signicant difference between
Reports of abnormalities in Ca and P associated with VPA use epileptic and healthy children [23,110]. Sato et al. [74] noticed that
are controversial and difcult to interpret because of widely vary- ICTP, a bone resorption marker, signicantly exceeded control val-
ing study designs [108]. Ca and P concentrations are essential ues. They also reported that high ICTP was positively correlated
6 A. Verrotti et al. / Clinical Neurology and Neurosurgery 112 (2010) 110

with ionized Ca. In this case, they indicated that the rate of resorp- To date, no changes in biochemical parameters of bone
tion may be more than that of formation, thus leading to a reduction metabolism have been associated with GBP treatment.
in bone density.
Recently, Kumandas et al. [15] investigated the effect of VPA 3.2. Lamotrigine
and CBZ monotherapies on insulin-like growth factor (IGF)-1 and
IGF binding protein (IGFBP)-3 levels, which are known to affect The principal mechanism of action of lamotrigine (LTG) appears
bone metabolism and BMD. There were no differences in the serum to involve inhibition of voltage-activated sodium channels, result-
concentrations of IGF-1 and IGFBP-3 between the patients taking ing in increased inhibition of action potential-ring activity [124].
antiepileptic drugs and control children. LTG also inhibits high voltage-activated calcium channels that are
Long-term use of VPA is associated with bone metabolism located presynaptically, and consequently it inhibits the release of
abnormalities, which include reduced BMD and changes in bone neurotransmitters such as glutamate [125].
turnover. VPA displayed a doseresponse relation. However, a LTG is used in monotherapy or as an add-on treatment for refrac-
recent study found that subjects receiving VPA monotherapy had tory partial [126] and generalized epilepsies [127129].
less negative effects on bone markers and improved bone density There are no data about a direct association between LTG and
compared to those receiving PHT or CBZ monotherapy [107]. The fractures but some studies have assessed LTG in relation to its effect
effect of VPA cannot be readily explained by vitamin D metabolism, on BMD [23,24,130,111].
since VPA is not an inducer of the CYP-450 system; it is instead Sheth and Hermann [130] examined the effects of LTG
thought to act by stimulating osteoclast activity and may cause an monotherapy on 13 children by measuring total z-scores of BMD.
imbalance between bone formation and resorption, thereby con- The patients were compared with 40 patients receiving polyther-
tributing to bone loss [74]. apy, as well as 36 control subjects. They showed that the z-scores
Finally, in an experimental study, oral administration of VPA to for BMD for LTG and control subjects were similar and were higher
epileptic rats for 6 months resulted in a signicant increase of bALP, than for those receiving polytherapy. Furthermore, increased dura-
OC and NTx relative to the control group. There were increases in tion of therapy was a predictor of lowered bone mineral density for
receptor activator of NFkB ligand (RANKL) and TNF-, and there polytherapy, but not for lamotrigine monotherapy. These ndings
was a decrease in osteoprotegrin compared with normal controls. suggest that LTG is not a drug that alters bone mineralization during
The authors postulated that the increase in bone formation markers childhood.
was caused by increased osteoblast number or activity in order to A recent longitudinal study of premenopausal women with
compensate for increased osteoclastic activity. The high turnover epilepsy receiving AED monotherapy compared a group of 23
state may be also caused by the increase in PTH caused by VPA [113]. patients receiving LTG treatment with a group of 15 patients taking
Effects of VPA on bone may occur as a result of this agents effect on PHT [24]. All enrolled subjects were between 18 and 40 years old
IGF-I, as suggested by Rattya et al. The mechanism remains to be and were receiving a single AED at least 6 months before enroll-
more robustly dened. However, a recent study found that subjects ment; after 1 year of observation, the BMD of the proximal femur
receiving VPA monotherapy had fewer negative effects on bone and lumbar spine was measured. There was signicant femoral
markers and improved bone density compared to those receiving neck bone loss over 1 year in the group that received PHT. In con-
phenytoin and CBZ monotherapy [107]. trast, patients treated with LTG did not lose bone mass at any
In conclusion, long-term antiepileptic drug treatment with VPA site.
has detectable adverse effects on bone turnover and induces a state Interestingly, Guo et al. [112] reported that LTG and/or VPA
of decreased BMD. Therefore, routine monitoring of BMD and bio- therapy lasting more than 2 years was associated with low total
chemical markers of bone turnover are warranted. body BMD, reduced bone formation (reduced plasma OC) and short
stature in children with epilepsy aged 317. In fact, these anoma-
lies were more evident in those children who had lower physical
3. Newer AEDs activity and were receiving a combined therapy (VPA and LTG).
The authors concluded by afrming that the observed abnormali-
3.1. Gabapentin ties were related to reduced physical activity rather than the direct
effect of drug therapy, because patients who had the need for com-
Gabapentin (GBP) was formed by adding a cyclohexyl group bination therapy were also those who had more severe seizures
to GABA, which allowed it to cross the bloodbrain barrier [114]. and, consequently, reduced physical activity.
Despite its structure, GBP does not bind to GABA receptors in the Currently, there are few studies that have assessed markers of
central nervous system [115]. It appears to have an inhibitory effect bone metabolism in epileptic patients receiving LTG monotherapy.
on voltage-gated calcium channels containing the alpha 2-delta Using a cross-sectional study, Pack et al. [36] studied 19 women
subunit [116,117]. It is not metabolized, and it does not induce with epilepsy who received LTG monotherapy for an average
or inhibit hepatic enzymes [118]. GBP, in clinical use since 1993, period of 21 months. They measured indices of mineral metabolism
is indicated as an adjunctive AED for treatment of partial seizure, (25OHD, PTH, Ca, IGF-I, and IGFBP-III), markers of bone forma-
with or without secondary generalization, in patients over 12 years tion (bALP, OC) and urinary NTx, a bone resorption marker. This
of age [119]. Side effects of GBP are generally mild and transient study demonstrated that premenopausal women receiving LTG
[120]; adverse effects on sexual function [121] and weight gain monotherapy had no signicant reductions in Ca and 25OHD or
[122] are reported at higher doses. increased bone turnover. These ndings are in agreement with
There is no study that has examined the relationship between those reported by a previous study by Stephen et al. [27].
bone metabolism and GBP monotherapy. However, there are three A recent longitudinal study [23] prospectively evaluated the
papers [5,19,21] that have studied a group of adult epileptic alterations in BMD and biochemical markers of bone metabolism
patients treated with different AEDs, including GBP. From their in 8 patients with newly diagnosed epilepsy, aged between 18 and
data, there was evidence that long-term GBP therapy may cause 50, before and after 6 months of LTG monotherapy. In this study,
bone loss at the hip and lumbar spine. More recently, Ensrud et al. there were no abnormalities in BMD, Ca, P, vitamin D or urinary
[123] conrmed, in a prospective study, that this AED can cause a Pyrilinks both before and after 6 months of LTG treatment. Instead,
signicant hip bone loss in older men, suggesting that GBP is not PTH and OC levels increased. The authors suggested that LTG may
free from this important adverse effect. affect bone metabolism, inducing the development of resistance to
A. Verrotti et al. / Clinical Neurology and Neurosurgery 112 (2010) 110 7

PTH, but this drug can compensate for bone loss by increasing bone decreased 25OHD levels and decreased BMD, although a signicant
formation. reduction of 25OHD was found.
In summary, the data now available indicate that LTG monother- Mintzer et al. [35] demonstrated that OXC monotherapy induced
apy does not lead to osteopenic effects or signicant alterations in signicant reductions in 25OHD, with a pattern of changes in other
bone metabolism. bone biomarkers suggestive of secondary hyperparathyroidism
such as higher PTH and bALP. This suggested that this drug had sim-
3.3. Levetiracetam ilar effects on bone and 25OHD metabolism as CBZ. These ndings
were unexpected because OXC is only a limited enzyme inducer.
Levetiracetam (LEV) is an enantiomer of the ethyl analog of An explanation could be that OXC may have some dose-dependent
piracetam that has the ability to facilitate cholinergic transmission CYP-450 induction property [146]; in this latter study, OXC dosage
[131]. LEV seems to have several modes of action, such as sup- was found to have a signicant correlation with bALP. An alterna-
pression of negative allosteric modulators of neuronal GABA- and tive hypothesis is that OXC may have a direct effect on osteoblast
glycine-gated currents [132], inhibition of voltage-gated calcium proliferation, as has been proposed for CBZ [20].
channels [133], reduction of voltage-operated potassium currents In summary, OXC seems to be associated with reduced 25OHD
[134] and binding to synaptic vesicle protein 2A (SV2A) [135]. It levels and elevated biomarkers that are consistent with increased
has been proven effective in treatment-resistant partial seizures in bone turnover. Because this AED has been shown to possess mech-
adults [136]. LEV is safe and well tolerated, but has been scarcely anisms that might affect bone, patients with osteopenia diagnosed
studied. A study carried out by Nissen-Meyer et al. [137] evalu- before the initiation of OXC therapy should be followed carefully,
ated the effect of AEDs on bone mass, biomechanical strength and especially in long-term treatment.
bone turnover in rats. LEV did not affect BMC or BMD at either
low or high dose. In the femoral neck, predominantly trabecular 3.5. Topiramate
bone, low-dose LEV signicantly decreased bone biomechanical
strength parameters. However, high-dose LEV treatment seemed to Topiramate is a sulfamate-substituted derivative of the
affect these parameters of bone strength to any signicant extent. monosaccharide d-fructose. Although the precise mechanisms of
In rats treated with low-dose LEV, serum OC levels were signi- action are unknown, it is considered to produce its antiepilep-
cantly reduced relative to controls, whereas RatLaps (a resorption tic effects primarily through enhancement of GABAergic activity,
marker) levels were unaltered, indicating an effect of this drug inhibition of kainite/alfa-amino-3-hydroxy-5-methyllisoxazole-
primarily on bone formation. In contrast, neither OC nor RatLaps 4-proprionic acid-type glutamate receptors and inhibition of
levels were signicantly affected by high-dose LEV under these voltage-sensitive sodium and calcium channels. It also inhibits
experimental conditions. There were no signicant differences in most of the carbonic anhydrase isozymes (except I and III). Top-
serum Ca levels induced by the different treatments compared iramate as monotherapy or adjunctive therapy was effective in
to controls. Ali et al. [138] studied the effects of LEV monother- reducing the frequency of seizures in patients with primary gener-
apy on BMD, vitamin D and OC levels in 16 adult patients. They alized tonicclonic seizures, partial seizures or seizures associated
found no signicant decrease in vitamin D levels or BMD; eleva- with Lennox-Gastaust syndrome [147].
tion of OC levels was noted in 4 patients but all 4 had previous Carbonic anhydrase is a ubiquitous zinc enzyme. One isozyme
prolonged exposure to other AEDs known to cause increased bone (II) catalyzes the reversible hydration of carbon dioxide to give
turnover, such as PHT, CBZ and VPA. In conclusion, it remains to bicarbonate and proton ions. These proton ions provide an acidic
be determined whether LEV does not affect BMD, but it seems to environment during the bone resorption phase of the bone cycle
reduce bone strength and bone formation without altering bone and, thus, carbonic anhydrase is implicated in osteoclast differen-
mass. Based on these few results, epidemiological studies on bone tiation. Moreover, the majority of topiramate recipients developed
turnover in patients treated with LEV are needed, and these patients mild to moderate metabolic acidosis. As chronic metabolic acidosis
should be evaluated regularly to identify possible bone-related side may result in an increased risk of renal calculi, osteomalacia and/or
effects. osteoporosis.
However, it has been reported, in a double-blind, placebo-
3.4. Oxcarbazepine controlled study in obese adults, that bone turnover was similar
in topiramate placebo groups during long-term exposure [148].
Oxcarbazepine (OXC) is a new AED and is indicated for the treat- In conclusion topiramate seems not to be associated with bone
ment of partial and secondarily generalized tonicclonic seizures metabolism alterations but there is a lack of clinical information.
as both a monotherapy and as part of combination therapy in adults
and children with epilepsy [139,140]. The mechanism of action of 3.6. Zonisamide
OXC is similar to that of CBZ, with comparable efcacy but supe-
rior safety, according to controlled clinical trials [141]. OXC and its Zonisamide is a 1,2-benzisoxazole compound with a sulfon-
active metabolite, monohydroxy derivative (MHD), have effects on amide side chain that is structurally different from other AEDs. Its
sodium channels [142] and possibly potassium [143] and calcium exact mechanism of action is not known; however, current experi-
channels [144]. mental evidence suggests that the major mechanisms of action are
To date, few studies in the literature have assessed the effect the blocking of repetitive ring of voltage-gated sodium channels
of OXC on BMD [16,145]. Babayigit et al. [16] measured BMD and [149] and the reduction of T-type calcium channel currents [150].
determined the changes in biochemical markers of bone miner- This drug has been shown to be an effective AED in patients with
alization in 14 patients with idiopathic epilepsy who had received refractory partial [151,152] and generalized tonicclonic seizures
OXC for more than 1 year. They showed that patients had decreased [153,154].
BMD and increased serum alkaline phosphatase concentrations Currently, there is only one study that showed that administra-
compared with controls. There were no signicant differences in tion of zonisamide can decrease BMD signicantly [155] in epileptic
the serum concentration of Ca, P, PTH and 25OHD. patients.
Recently, Cansu et al. [145] reported a slight decrease in lumbar Interestingly, an animal study [156] found signicantly
and femoral BMD in 8% of epilepsy patients studied after receiving decreased BMD at the tibial metaphysis and diaphysis and
18 months of OXC monotherapy. There was no correlation between increased serum pyridinoline level (PYD), a marker of bone resorp-
8 A. Verrotti et al. / Clinical Neurology and Neurosurgery 112 (2010) 110

Table 1 [13] Tekgul H, Serdaroglu G, Huseyinov A, Gkben S. Bone mineral status in


Main effects of classic and new AEDs on bone and calcium metabolism. pediatric outpatients on antiepileptic drug monotherapy. J Child Neurol
2006;21:4114.
Drug BMD 25-OHD Ca/P PTH Bone turnover [14] Tsukahara H, Kimura K, Todoroki Y, Ohshima Y, Hiraoka M, Shigematsu Y, et
Classic AEDs al. Bone mineral status in ambulatory pediatric patients on long-term anti-
Benzodiazepines N N bALP OC ICTP NTx epileptic drug therapy. Pediatr Int 2002;44:24753.
[15] Kumandas S, Koklu E, Gms H, Koklu S, Kurtoglu S, Karakukcu M, et al. Effect
Carbamazepine N bALP OC ICTP NTx
of carbamezapine and valproic acid on bone mineral density, IGF-I and IGFBP-
Phenytoin bALP NTx
3. J Pediatr Endocrinol Metab 2006;19:52934.
Phenobarbital N bALP ICTP [16] Babayigit A, Dirik E, Bober E, Cakmakci H. Adverse effects of
Primidone N antiepileptic drugs on bone mineral density. Pediatr Neurol 2006;35:
Valproic acid N N N ALP OC 17781.
[17] Pack AM, Morrell MJ, Randall A, McMahon DJ, Shane E. Bone health in young
New AEDs
women with epilepsy after one year of antiepileptic drug monotherapy. Neu-
Gabapentin
rology 2008;29:158693.
Lamotrigine N N N ? N [18] Sheth RD, Wesolowski CA, Jacob JC, Penney S, Hobbs GR, Riggs JE, et al.
Levetiracetam N N N ? Effect of carbamazepine and valproate on bone mineral density. J Pediatr
Oxcarbazepine N bALP 1995;127:25662.
Zonisamidea PYD [19] El-Hajj Fuleihan G, Dib L, Yamout B, Sawaya R, Mikati MA. Predictors of
a bone density in ambulatory patients on antiepileptic drugs. Bone 2008;43:
Results from animal studies.
14955.
[20] Feldkamp J, Becker A, Witte OW, Scharff D, Scherbaum WA. Long-term anti-
tion, in rats after chronic administration of zonisamide. On the convulsant therapy leads to low bone mineral densityevidence for direct
drug effects of phenytoin and carbamazepine on human osteoblast-like cells.
contrary, there was no signicant alteration in OC level. These Exp Clin Endocrinol Diabetes 2000;108:3743.
results suggest that zonisamide may cause bone loss by accelerat- [21] Andress DL, Ozuna J, Tirschwell D, Grande L, Johnson M, Jacobson AF, et
ing bone resorption. The same study demonstrated that combined al. Antiepileptic drug-induced bone loss in young male patients who have
seizures. Arch Neurol 2002;59:7816.
administration of alfacalcidol with zonisamide could prevent this [22] Petty SJ, Paton LM, OBrien TJ, Makovey J, Erbas B, Sambrook P, et al.
bone loss. Effect of antiepileptic medication on bone mineral measures. Neurology
Zonisamide has been found to inhibit carbonic anhydrase in 2005;65:135865.
[23] Kim SH, Lee JW, Choi KG, Chung HW, Lee HW. A 6-month longitudinal study
human erythrocytes [157]; as suggested for topiramate, we spec-
of bone mineral density with antiepileptic drug monotherapy. Epilepsy Behav
ulate that this inhibition may be a potential cause of reduction of 2007;10:2915.
BMD. [24] Pack A. Bone health in people with epilepsy: is it impaired and what are the
risk factors? Seizure 2008;17:1816.
[25] Chou IJ, Lin KL, Wang HS, Wang CJ. Evaluation of bone mineral density in
4. Conclusion children receiving carbamazepine or valproate monotherapy. Acta Paediatr
Taiwan 2007;48:31722.
[26] Fitzpatrick LA. Pathophysiology of bone loss in patients receiving anticonvul-
The treatment of epilepsy can adversely affect bone health and
sant therapy. Epilepsy Behav 2004;5 Suppl 2:S315.
calcium metabolism. The main abnormalities induced by classic [27] Stephen LJ, McLellan AR, Harrison JH, Shapiro D, Dominiczak MH, Sills GJ,
and new AEDs are reported in Table 1. These adverse effects of the et al. Bone density and antiepileptic drugs: a case-controlled study. Seizure
older AEDs (PHT, PB, CBZ and VPA) have been studied in depth and 1999;8:33942.
[28] Kafali G, Erselcan T, Tanzer F. Effect of antiepileptic drugs on bone min-
appear to have been demonstrated. The effects of the new AEDs on eral density in children between ages 6 and 12 years. Clin Pediatr (Phila)
bone and calcium metabolism need to be better dened because 1999;38:938.
the number of studies is limited and more evidence is needed on [29] Vlimki MJ, Tiihonen M, Laitinen K, Thtel R, Krkkinen M, Lamberg-
Allardt C, et al. Bone mineral density measured by dual-energy X-ray
the safety of these new AEDs. absorptiometry and novel markers of bone formation and resorp-
tion in patients on antiepileptic drugs. J Bone Miner Res 1994;9:
References 6317.
[30] Pack AM, Olarte LS, Morrell MJ, Flaster E, Resor SR, Shane E. Bone mineral
[1] Gavish M, Snyder SH. Benzodiazepine recognition sites on GABA receptors. density in an outpatient population receiving enzyme-inducing antiepileptic
Nature 1980;87:6512. drugs. Epilepsy Behav 2003;4:16974.
[2] MacDonald RL, Barker JL. Enhancement of GABA-mediated post- [31] Erbayat Altay E, SerdaroYlu A. Evaluation of bone mineral metabolism in
synaptic inhibition in cultured mammalian spinal cord neurons: children receiving carbamazepine and valproic acid. J Pediatr Endocrinol
a common mode of anticonvulsant action. Brain Res 1979;167: Metab 2000;13:9339.
32336. [32] Hoikka V, Alhava EM, Karjalainen P, Kernen T, Savolainen KE, Riekkinen
[3] Olsen RW. GABAbenzodiazepinebarbiturate receptor interactions. J Neu- P, et al. Carbamazepine and bone mineral metabolism. Acta Neurol Scand
rochem 1981;37:113. 1984;70:7780.
[4] Vestergaard P, Rejnmark L, Mosekilde L. Fracture risk associated with use of [33] Akin R, Okutan V, Sarici U, Altunbas A, Gkcay E. Evaluation of bone
antiepileptic drugs. Epilepsia 2004;45:13307. mineral density in children receiving antiepileptic drugs. Pediatr Neurol
[5] Vestergaard P, Rejnmark L, Mosekilde L. Anxiolytics and sedatives 1998;19:12931.
and risk of fractures: effects of half-life. Calcif Tissue Int 2008;82: [34] Souverein PC, Webb DJ, Weil JG, Van Staa TP, Egberts AC. Use of antiepileptic
3443. drugs and risk of fractures: casecontrol study among patients with epilepsy.
[6] Ensrud KE, Walczak TS, Blackwell T, Ensrud ER, Bowman PJ, Stone KL. Neurology 2006;66:131824.
Antiepileptic drug use increases rates of bone loss in older women: a prospec- [35] Mintzer S, Boppana P, Toguri J, DeSantis A. Vitamin D levels and bone
tive study. Neurology 2004;62:20517. turnover in epilepsy patients taking carbamazepine or oxcarbazepine. Epilep-
[7] Kulak CA, Borba VZ, Bilezikian JP, Silvado CE, Paola L, Boguszewski CL. Bone sia 2006;47:5105.
mineral density and serum levels of 25 OH vitamin D in chronic users of [36] Pack AM, Morrell MJ, Marcus R, Holloway L, Flaster E, Done S, et al. Bone mass
antiepileptic drugs. Arq Neuropsiquiatr 2004;62:9408. and turnover in women with epilepsy on antiepileptic drug monotherapy.
[8] Farhat G, Yamout B, Mikati MA, Demirjian S, Sawaya R, El-Hajj Fuleihan G. Ann Neurol 2005;57:2527.
Effects of antiepileptic drugs on bone density in ambulatory patients. Neurol- [37] Ecevit C, AydoYan A, Kavakli T, Altin z S. Effect of carbamazepine
ogy 2002;58:134853. and valproate on bone mineral density. Pediatr Neurol 2004;31:
[9] Kinjo M, Setoguchi S, Schneeweiss S, Solomon DH. Bone mineral density 27982.
in subjects using central nervous system-active medications. Am J Med [38] Nicolaidou P, Georgouli H, Kotsalis H, Matsinos Y, Papadopoulou A,
2005;118:1414. Fretzayas A, et al. Effects of anticonvulsant therapy on vitamin D sta-
[10] Cummings SR, Nevitt MC, Browner WS, Stone K, Fox KM, Ensrud KE, et al. tus in children: prospective monitoring study. J Child Neurol 2006;21:
Risk factors for hip fracture in white women. Study of Osteoporotic Fractures 2059.
Research Group. N Engl J Med 1995;332:76773. [39] Voudris KA, Attilakos A, Katsarou E, Garou A, Dimou S, Skardoutsou A, et
[11] Talwar D. Mechanisms of antiepileptic drug action. Pediatr Neurol al. Early alteration in bone metabolism in epileptic children receiving carba-
1990;6:28995. mazepine monotherapy owing to the induction of hepatic drug-metabolizing
[12] Kuo CC. A common anticonvulsant binding site for phenytoin, carbamazepine, enzymes. J Child Neurol 2005;20:5136.
and lamotrigine in neuronal Na+ channels. Mol Pharmacol 1998;54: [40] Gough H, Goggin T, Bissessar A, Baker M, Crowley M, Callaghan N. A com-
71221. parative study of the relative inuence of different anticonvulsant drugs, UV
A. Verrotti et al. / Clinical Neurology and Neurosurgery 112 (2010) 110 9

exposure and diet on vitamin D and calcium metabolism in out-patients with [72] Koch HU, Kraft D, von Herrath D, Schaefer K. Inuence of diphenylhydan-
epilepsy. Q J Med 1986;59:56977. toin and phenobarbital on intestinal calcium transport in the rat. Epilepsia
[41] Zerwekh JE, Homan R, Tindall R, Pak CY. Decreased serum 24,25- 1972;13:82934.
dihydroxyvitamin D concentration during long-term anticonvulsant therapy [73] Hahn TJ, Hendin BA, Scharp CR, Boisseau VC, Haddad JG. Serum 25-
in adult epileptics. Ann Neurol 1982;12:1846. hydroxycalciferol levels and bone mass in children on chronic anticonvulsant
[42] Verrotti A, Greco R, Morgese G, Chiarelli F. Increased bone turnover in epileptic therapy. N Engl J Med 1975;292:5504.
patients treated with carbamazepine. Ann Neurol 2000;47(3):3858. [74] Sato Y, Kondo I, Ishida S, Motooka H, Takayama K, Tomita Y, et al. Decreased
[43] Verrotti A, Greco R, Latini G, Morgese G, Chiarelli F. Increased bone turnover bone mass and increased bone turnover with valproate therapy in adults with
in prepubertal, pubertal, and postpubertal patients receiving carbamazepine. epilepsy. Neurology 2001;57:4459.
Epilepsia 2002;43:148892. [75] Bell RD, Pak CY, Zerwekh J, Barilla DE, Vasko M. Effect of phenytoin on bone
[44] Brmswig S, Zittermann A, Berthold HK. Carbamazepine does not alter bio- and vitamin D metabolism. Ann Neurol 1979;5:3748.
chemical parameters of bone turnover in healthy male adults. Calcif Tissue [76] Takahashi A, Onodera K, Shinoda H, Mayanagi H. Phenytoin and its metabolite,
Int 2000;73:35660. 5-(4-hydroxyphenyl)-5-phenylhydantoin, show bone resorption in cultured
[45] Samaniego EA, Sheth RD. Bone consequences of epilepsy and antiepileptic neonatal mouse calvaria. Jpn J Pharmacol 2000;82:824.
medications. Semin Pediatr Neurol 2007;14:196200. [77] Pento JT, Glick SM, Kagan A. Diphenylhydantoin inhibition of calcitonin secre-
[46] Myllynen P, Pienimki P, Raunio H, Vhkangas K. Microsomal metabolism tion in the pig. Endocrinology 1973;92:3303.
of carbamazepine and oxcarbazepine in liver and placenta. Hum Exp Toxicol [78] Scott AK, Haynes BP, Schinkel KD, Ohnhaus EE, Park BK. Hepatic enzyme
1998;17:66876. induction and vitamin K1 elimination in man. Eur J Clin Pharmacol
[47] Valsamis HA, Arora SK, Labban B, McFarlane SI. Antiepileptic drugs and bone 1987;33:935.
metabolism. Nutr Metab 2006;3:36. [79] Bogliun G, Beghi E, Crespi V, Delodovici L, dAmico P. Anticonvulsant drugs
[48] Pack AM. The association between antiepileptic drugs and bone disease. and bone metabolism. Acta Neurol Scand 1986;74:2848.
Epilepsy Curr 2003;3:915. [80] Ohta T, Wergedal JE, Gruber HE, Baylink DJ, Lau KH. Low dose phenytoin is an
[49] Voudris K, Moustak M, Zeis PM, Dimou S, Vagiakou E, Tsagris B, et al. Alkaline osteogenic agent in the rat. Calcif Tissue Int 1995;56:428.
phosphatase and its isoenzyme activity for the evaluation of bone metabolism [81] Vernillo AT, Rifkin BR, Hauschka PV. Phenytoin affects osteocalcin secre-
in children receiving anticonvulsant monotherapy. Seizure 2002;11:37780. tion from osteoblastic rat osteosarcoma 17/2.8 cells in culture. Bone
[50] Sheth RD. Metabolic concerns associated with antiepileptic medications. Neu- 1990;11:30912.
rology 2004;63:S249. [82] Olsen RW. Phenobarbital and other barbiturates. In: Levy RH, Mattson RH,
[51] Looker AC, Bauer DC, Chesnut CH, Gundberg CM, Hochberg MC, Klee G, et al. Meldrum BS, Perucca E, editors. Antiepileptic drugs, vol. 50. Philadelphia:
Clinical use of biochemical markers of bone remodeling: current status and Lippincott Williams & Wilkins Publishers; 2002. p. 48995.
future directions. Osteoporos Int 2000;11:46780. [83] Beghi E. Phenobarbital and other barbiturates. In: Levy RH, Mattson RH,
[52] von Borstel Smith M, Crofoot K, Rodriguez-Proteau R, Filtz TM. Effects of Meldrum BS, Perucca E, editors. Antiepileptic drugs, vol. 54. Philadelphia:
phenytoin and carbamazepine on calcium transport in Caco-2 cells. Toxicol Lippincott Williams & Wilkins Publishers; 2002. p. 5227.
In Vitro 2007;21:85562. [84] Hahn TJ, Birge SJ, Schrp CR, Avioli LV. Phenobarbital-induced alterations in
[53] Delorenzo RJ, Sun DA. Phenytoin and other hydantioins mechanisms of action. vitamin D metabolism. Clin Invest 1972;51:7418.
In: Levy RH, Mattson RH, Meldrum BS, Perucca E, editors. Antiepileptic drugs, [85] Lifshitz F, Maclaren N. Vitamin D-dependent rickets in institutionalized, men-
vol. 57. Philadelphia: Lippincott Williams & Wilkins Publishers; 2002. p. tally retarded children receiving long-term anticonvulsant therapy. I. A survey
55164. of 228 patients. J Pediatr 1973;83:61220.
[54] Delgado-Escueta AV, Horan MP. Phenytoin: biochemical membrane studies. [86] Foss MC, Meneghelli UG, Tolosa Verssimo JM. The effect of the anticon-
Adv Neurol 1980;27:37798. vulsants phenobarbital and diphenylhydantoin on intestinal absorption of
[55] Segal MM, Douglas AF. Late sodium channel openings underlying epilepti- calcium. Acta Physiol Lat Am 1979;29:2238.
form activity are preferentially diminished by the anticonvulsant phenytoin. [87] Weisman Y, Fattal A, Eisenberg Z, Harel S, Spirer Z, Harell A. Decreased serum
J Neurophysiol 1997;77:302134. 24,25-dihydroxy vitamin D concentrations in children receiving chronic anti-
[56] DeLorenzo RJ. The epilepsies. In: Bradley WG, Daroff RB, Fenichel GM, Penry convulsant therapy. Br Med J 1979;2:5213.
JK, Glaser JH, editors. Neurology in clinical practice: principles of diagnosis [88] Hosseinpour F, Ellfolk M, Norlin M, Wikvall K. Phenobarbital suppresses
and management. 4th ed. Philadelphia: Butterworth-Heinermann; 2004. vitamin D3 25-hydroxylase expression: A potential new mechanism for drug-
[57] DeLorenzo RJ, Bowling AC, Taft WC. A molecular approach to the development induced osteomalacia. Biochem Biophys Res Commun 2007;357:6037.
of anticonvulsants. Ann N Acad Sci 1986;477:23846. [89] Mosekilde L, Christensen SC, Lund B, Sorensen OH, Melsen F. The inter-
[58] DeLorenzo RJ. Status epilepticus: concepts in diagnosis and treatment. Semin relationships between serum 25-hydroxychocalciferol, serum parathyroid
Neurol 1990;10:396405. hormone and bone changes in anticonvulsant osteomalacia. Acta Endocrinol
[59] DeLorenzo RJ. Regulation of neuronal excitability: molecular foundations for 1977;84:55965.
the study of alcohol withdrawal. In: Porter RJ, Mattson RH, Cramer JA, Taft [90] Weinstein RS, Bryce FG, Sappington LJ, King DW, Gallagher BB. Decreased
WC, Glaser GH, editors. Alcohol and seizures basic mechanism and clinical serum ionized calcium and normal vitamin D metabolite levels with anticon-
concepts. Philadelphia: FA Davis; 1990. p. 4754. vulsant drug treatment. J Clin Endocrinol Metab 1984;58:10039.
[60] Telci A, Cakatay U, Kurt BB, Kayali R, Sivas A, Akcay T, et al. Changes in bone [91] Ala-Houhala M, Korpela R, Koivikko M, Koskinen T, Koskinen M, Koivula T.
turnover and deoxypyridinoline levels in epileptic patients. Clin Chem Lab Long-term anticonvulsant therapy and vitamin D metabolism in ambulatory
Med 2000;38:4750. pubertal children. Neuropediatrics 1986;17:2126.
[61] Lau KH, Nakade O, Barr B, Taylor AK, Houchin K, Baylink DJ. Phenytoin [92] Holloway L, Paulson A, Seale C, Morell MJ, Marcus R. Skeletal status of women
increases markers of osteogenesis for the human species in vitro and in vivo. with epilepsy. J Bone Miner Res 2000;15(Suppl. 1):SA302 (Abstract).
J Clin Endocrinol Metab 1995;80:234753. [93] Gissel T, Poulsen CS, Vestergaard P. Adverse effects of antiepileptic drugs on
[62] Richens A, Rowe DJ. Disturbance of calcium metabolism by anticonvulsant bone mineral density in children. Expert Opin Drug Saf 2007;6:26778.
drugs. Br Med J 1970;4:736. [94] Aysegl T, Cakatay U, Kurt BB, Kayali R, Sivas A, Akcay T, et al. Changes in
[63] Schmitt BP, Nordlund DJ, Rodgers LA. Prevalence of hypocalcemia and bone turnover and deoxypyridinoline levels in epileptic patients. Clin Chem
elevated serum alkaline phosphatase in patients receiving chronic anticon- Lab Med 2000;38:4750.
vulsant therapy. J Fam Pract 1984;18, 873-. [95] Kulak CAM, Borba VZC, Silvado CE, de Paola L, Seibel MJ, Bilezikian JP, et
[64] Winnacker JL, Yeager H, Saunders JA, Russell B, Anast CS. Rickets in children al. Bone density and bone turnover markers in patients with epilepsy on
receiving anticonvulsant drugs. Biochemical and hormonal markers. Am J Dis chronic antiepileptic drug therapy. Arq Bras Endocrinol Metabol 2007;51:
Child 1977;131:28690. 46671.
[65] Reunanen MI, Sotaniemi EA, Hakkarainen HK. Serum calcium balance during [96] Lado F, Spiegel R, Masur JH, Boro A, Haut NY. Value of routine screening
early phase of diphenylhydantoin therapy. Int J Clin Pharmacol Biopharm for bone demineralization in an urban population of patients with epilepsy.
1976;14:159. Epilepsy Res 2008;78:15560.
[66] Silver J, Davies TJ, Kupersmitt E, Orme M, Petrie A, Vajda F. Prevalence and [97] Filardi S, Guerreiro CAM, Magna LA, Neto JFM. Bone mineral density, vitamin
treatment of vitamin D deciency in children on anticonvulsant drugs. Arch D and anticonvulsant therapy. Arq Neuropsiquiatr 2000;58:61620.
Dis Child 1974;49:34450. [98] Goodman LS, Swinyard EA, Brown WC, Schiffman DO, Grawal MS, Bliss EL.
[67] Pack AM, Morrell MJ. Adverse effects of antiepileptic drugs on bone struc- Anticonvulsant properties of 5-phenyl-ethylhexahydropyridine-4,6-dione, a
ture: epidemiology, mechanisms and therapeutic implications. CNS Drugs new antiepileptic. J Parmacol Exp Therapeut 1953;108:42836.
2001;15:63342. [99] Fincham RW, Schottelius DD. Primidone. In: Levy RH, Mattson RH, Meldrum
[68] Tjellesen L, Christiansen C. Serum vitamin D metabolites in epileptic patients BS, Perucca E, editors. Antiepileptic drugs, vol. 65. Philadelphia: Lippincott
treated with 2 different anti-convulsants. Acta Neurol Scant 1982;66:33541. Williams & Wilkins Publishers; 2002. p. 62135.
[69] Harris M, Jenkins MV, Wills MR. Phenytoin inhibition of parathyroid hormone [100] Christiansen C, Rodbro P, Lund M. Incidence of anticonvulsant osteomalacea
induced bone resorption in vitro. Br J Pharmacol 1974;50:4058. and effect of vitamin D; controlled therapeutic trial. Br Med J 1973;4:695701.
[70] Jenkins MV, Harris M, Wills MR. The effect of phenytoin on parathyroid extract [101] Bouillon R, Reynaert J, Claes JH, Lissens W, De Moor P. The effect of anti-
and 25-hydroxycholecalciferol-induced bone resorption: adenosine 3,5 cyclic convulsant therapy on serum levels of 25-hydroxy-vitamin D, calcium, and
monophosphate production. Calcif Tissue Res 1974;16:1637. parathyroid hormone. J Clin Endocrinol Metab 1975;41:11305.
[71] Hahn TJ, Scharp CR, Richardson CA, Halstead LR, Kahn AJ, Teitelbaum SL. Inter- [102] Bourgeois B, Beaumanoir A, Blajev B, de la Cruz N, Despland PA, Egli M, et
action of diphenylhydantoin (phenytoin) and phenobarbital with hormonal al. Monotherapy with valproate in primary generalized epilepsies. Epilepsia
mediation of fetal rat bone resorption in vitro. J Clin Invest 1978;62:40614. 1987;28:S811.
10 A. Verrotti et al. / Clinical Neurology and Neurosurgery 112 (2010) 110

[103] de Silva M, MacArdle B, McGowan M, Hughes E, Stewart J, Neville BG, et al. [130] Sheth RD, Hermann BP. Bone mineral density with lamotrigine monotherapy
Randomised comparative monotherapy trial of phenobarbitone, phenytoin, for epilepsy. Pediatr Neurol 2007;37:2504.
carbamazepine, or sodium valproate for newly diagnosed childhood epilepsy. [131] Wlfert E, Hanin I, Verloes R. Facilitation of calcium-dependent cholinergic
Lancet 1996;347:70913. function by ucb L059, a new second generation nootropic agent. Psy-
[104] Mattson RH, Cramer JA, Williamson PD, Novelly RA. Valproic acid in epilepsy: chopharmacol Bull 1989;25:498502.
clinical and pharmacological effects. Ann Neurol 1978;3:205. [132] Rigo JM, Hans G, Nguyen L, Rocher V, Belachew S, Malgrange B, et al. The anti-
[105] McLean MJ, Macdonald RL. Sodium valproate, but not ethosuximide, produces epileptic drug levetiracetam reverses the inhibition by negative allosteric
use- and voltage-dependent limitation of high frequency repetitive ring of modulators of neuronal GABA- and glycine-gated currents. Br J Pharmacol
action potentials of mouse central neurons in cell culture. J Pharmacol Exp 2002;136:65972.
Ther 1986;237:100111. [133] Lukyanetz EA, Shkryl VM, Kostyuk PG. Selective blockade of N-type calcium
[106] Lscher W. Valproate induced changes in GABA metabolism at the subcellular channels by levetiracetam. Epilepsia 2002;43:918.
level. Biochem Pharmacol 1981;1:13646. [134] Madeja M, Margineanu DG, Gorji A, Siep E, Boerrigter P, Klitgaard H, et al.
[107] Elliott JO, Jacobson MP, Haneef Z. Homocysteine and bone loss in epilepsy. Reduction of voltage-operated potassium currents by levetiracetam: a novel
Seizure 2007;16:2234. antiepileptic mechanism of action? Neuropharmacology 2003;45:66171.
[108] Oner N, Kaya M, Karasalihoglu S, Karaca H, Celtik C, Ttncler F. Bone mineral [135] Gillard M, Chatelain P, Fuks B. Binding characteristics of levetiracetam to
metabolism changes in epileptic children receiving valproic acid. J Paediatr synaptic vesicle protein 2A (SV2A) in human brain and in CHO cells expressing
Child Health 2004;40:4703. the human recombinant protein. Eur J Pharmacol 2006;536:1028.
[109] Rieger-Wettengl G, Tutlewski B, Stabrey A, Rauch F, Herkenrath P, Schauseil- [136] Cereghino JJ, Biton V, Abou-Khalil B, Dreifuss F, Gauer LJ, Leppik I. Levetirac-
Zipf U, et al. Analysis of the musculoskeletal system in children and etam for partial seizures: results of a double-blind, randomized clinical trial.
adolescents receiving anticonvulsant monotherapy with valproic acid or car- Neurology 2000;55:23642.
bamazepine. Pediatrics 2001;108:E107. [137] Nissen-Meyer LS, Svalheim S, Taubll E, Reppe S, Lekva T, Solberg LB, et al.
[110] Song XQ, Wang ZP, Bao KR, Zhang JM, Wu J, Yan CH, et al. Effect of car- Levetiracetam, phenytoin, and valproate act differently on rat bone mass,
bamazepine and valproate on bone metabolism in children with epilepsy. structure, and metabolism. Epilepsia 2007;10:185060.
Zhonghua Er Ke Za Zhi 2005;43:72832. [138] Ali II, Herial NA, Horrigan T, Kellough L, Tietjen GE. Measurement of bone
[111] Boluk A, Guzelipek M, Savli H, Temel I, OziYik HI, Kaygusuz A. The effect of mineral dendity in patients on levetiracetam monotherapy. Am Epilepsy Soc
valproate on bone mineral density in adult epileptic patients. Pharmacol Res Abstr 2006;2:150.
2004;50:937. [139] Wellington K, Goa KL. Oxcarbazepine: an update of its efcacy in the man-
[112] Guo CY, Ronen GM, Atkinson SA. Long-term valproate and lamotrigine treat- agement of epilepsy. CNS Drugs 2001;15:13763.
ment may be a marker for reduced growth and bone mass in children with [140] Bang L, Goa K. Oxcarbazepine: a review of its use in children with epilepsy.
epilepsy. Epilepsia 2001;42:11417. Paediatr Drugs 2003;5:55773.
[113] Elwakkad AS, El Elshamy KA, Sibaii H. Fish liver oil and propolis as [141] Freidel M, Krause E, Kuhn K, Peper R, Vogel H. Oxcarbazepine in the treatment
protective natural products against the effect of the anti-epileptic drug val- of epilepsy. Fortschr Neurol Psychiatr 2007;75:1006.
proate on immunological markers of bone formation in rats. Epilepsy Res [142] Kalis MM, Huff NA. Oxcarbazepine, an antiepileptic agent. Clin Ther
2008;80:4756. 2001;23:680700.
[114] Petroff OA, Hyder F, Rothman DL, Mattson RH. Effects of gabapentin on [143] Herranz JL, Argumosa A. Characteristics and indications of oxcarbazepine. Rev
brain GABA, homocarnosine, and pyrrolidinone in epilepsy patients. Epilepsia Neurol 2002;35:S1019.
2000;41:67580. [144] Stefani A, Pisani A, De Murtas M, Mercuri NB, Marciani MG, Calabresi P. Action
[115] Goa KL, Sorkin EM, Gabapentin. A review of its pharmacological properties of GP 47779, the active metabolite of oxcarbazepine, on the corticostriatal
and clinical potential in epilepsy. Drugs 1993;46:40927. system. II. Modulation of high-voltage-activated calcium currents. Epilepsia
[116] Striano P, Striano S. Gabapentin: a Ca2+ channel alpha 2-delta ligand far 1995;36:9971002.
beyond epilepsy therapy. Drugs Today 2008:4435368. [145] Cansu A, Yesilkaya E, SerdaroYlu A, HirfanoYlu TL, Camurdan O, G lbahar
[117] Bryans JS, Davies N, Gee NS, Dissanayake VU, Ratcliffe GS, Horwell DC, et O, et al. Evaluation of bone turnover in epileptic children using oxcarbazepine.
al. Identication of novel ligands for the gabapentin binding site on the Pediatr Neurol 2008;39:26671.
alpha2delta subunit of a calcium channel and their evaluation as anticon- [146] Patsalos PN, Zakrzewska JM, Elyas AA. Dose dependent enzyme induction by
vulsant agents. J Med Chem 1998;41:183845. oxcarbazepine? Eur J Clin Pharmacol 1990;39:1878.
[118] LaRoche SM, Helmers SL. The new antiepileptic drugs: scientic review. JAMA [147] Lyseng-Williamson KA, Yang LPH. Topiramate. A review of its use in the treat-
2004;291:60514. ment of epilepsy. Drugs 2007;67:223156.
[119] Morris GL. Gabapentin. Epilepsia 1999;40:S6370. [148] Ramsay ER, Leung AT. Effect of topiramate on bone resorption in adults.
[120] McLean MJ, Morrell MJ, Willmore LJ, Privitera MD, Faught RE, Holmes GL, Epilepsia 2006;47(S4):171.
et al. Safety and tolerability of gabapentin as adjunctive therapy in a large, [149] Schauf CL. Zonisamide enhances slow sodium inactivation in Myxicola. Brain
multicenter study. Epilepsia 1999;40:96572. Res 1987;413:1858.
[121] Dalal A, Zhou L. Gabapentin and sexual dysfunction: report of two cases. [150] Suzuki S, Kawakami K, Nishimura S, Watanabe Y, Yagi K, Seino M, et al. Zon-
Neurologist 2008;14:501. isamide blocks T-type calcium channel in cultured neurons of rat cerebral
[122] DeToledo JC, Toledo C, DeCerce J, Ramsay RE. Changes in body weight with cortex. Epilepsy Res 1992;12:217.
chronic, high-dose gabapentin therapy. Therpeut Drug Monit 1997;19:3946. [151] Baulac M, Leppik IE. Efcacy and safety of adjunctive zonisamide therapy for
[123] Ensrud KE, Walczak TS, Blackwell TL, Ensrud ER, Barrett-Connor E, Orwoll ES. refractory partial seizures. Epilepsy Res 2007;75:7583.
Antiepileptic drug use and rates of hip bone loss in older men: a prospective [152] Brodie MJ, Duncan R, Vespignani H, Solyom A, Bitenskyy V, Lucas C. Dose-
study. Neurology 2008;71:72330. dependent safety and efcacy of zonisamide: a randomized, double-blind,
[124] Meldrum BS. Update on the mechanism of action of antiepileptic drugs. placebo-controlled study in patients with refractory partial seizures. Epilepsia
Epilepsia 1996;37:S411. 2005;46:3141.
[125] Landmark CJ. Targets for antiepileptic drugs in the synapse. Med Sci Monit [153] Leppik IE, Willmore LJ, Homan RW, Fromm G, Oommen KJ, Penry JK, et al.
2007;13:17. Efcacy and safety of zonisamide: results of a multicenter study. Epilepsy Res
[126] Duchowny M, Pellock JM, Graf WD, Billard C, Gilman J, Casale E, et al. A 1993;14:16573.
placebo-controlled trial of lamotrigine add-on therapy for partial seizures [154] You SJ, Kang HC, Kim HD, Lee HS, Ko TS. Clinical efcacy of zonisamide
in children. Lamictal Pediatric Partial Seizure Study Group. Neurology in Lennox-Gastaut syndrome: Korean multicentric experience. Brain Dev
1999;53:172431. 2008;30:28790.
[127] Gericke CA, Picard F, de Saint-Martin A, Strumia S, Marescaux C, Hirsch E. [155] Takahashi A. Effects of antiepileptics phenytoin, zonisamideand valproate on
Efcacy of lamotrigine in idiopathic generalized epilepsy syndromes: a video- bone metabolism in growing rats. Inter Congress Ser 2005;1284:878.
EEG-controlled, open study. Epileptic Disord 1999;1:15965. [156] Takahashi A, Onodera K, Kamei J, Sakurada S, Shinoda H, Miyazaki S, et al.
[128] Motte J, Trevathan E, Arvidsson JF, Barrera MN, Mullens EL, Manasco P. Lamot- Effects of chronic administration of zonisamide, an antiepileptic drug, on
rigine for generalized seizures associated with the Lennox-Gastaut syndrome. bone mineral density and their prevention with alfacalcidol in growing rats.
Lamictal Lennox-Gastaut Study Group. N Engl J Med 1998;339:8512. J Pharmacol Sci 2003;91:3138.
[129] Frank LM, Enlow T, Holmes GL, Manasco P, Concannon S, Chen C, et al. Lamictal [157] Masuda Y, Karasawa T. Inhibitory effect of zonisamide on human carbonic
(lamotrigine) monotherapy for typical absence seizures in children. Epilepsia anhydrase in vitro. Arzneimittelforschung 1993;43:4168.
1999;40:9739.

You might also like