Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Seizure 19 (2010) 375–382

Contents lists available at ScienceDirect

Seizure
journal homepage: www.elsevier.com/locate/yseiz

Review

Antiepileptic treatment in patients with epilepsy and other comorbidities§


J. Ruiz-Giménez a,*, J.C. Sánchez-Álvarez b, F. Cañadillas-Hidalgo c, P.J. Serrano-Castro d
on behalf of the Andalusian Epilepsy Society
a
Unidad de Epilepsia, Servicio de Neurologia, Hospital Universitario Virgen de las Nieves, Granada, España
b
Servicio de Neurologia, Hospital Clinico Universitario San Cecilio, Granada, España
c
Servicio de Neurologı´a, Hospital Universitario Reina Sofia, Cordoba, España
d
Unidad de Neurologia y Neurofisiologı´a, Hospital Torrecardenas, Almeria, España

A R T I C L E I N F O A B S T R A C T

Article history: Background: A high number of patients with epilepsy have comorbidities. The type of comorbidity is an
Received 9 November 2009 important factor in deciding on the most suitable treatment, including that for acute epileptic seizures
Received in revised form 18 April 2010 and chronic antiepileptic treatment. Evidence-based criteria should guide the selection of the
Accepted 20 May 2010
appropriate antiepileptic drugs given specific comorbidities.
Methods: We performed a comprehensive search of the scientific literature on epilepsy treatment in
Keywords: patients with the following comorbidities: heart disease, lung disease, liver disease, kidney disease,
Anticonvulsants
porphyria, organ transplantation, thyroid disease, metabolic disorder, infection, mental disability,
Comorbidity
psychiatric disorder, cognitive impairment, stroke, and brain tumour.
Drug interactions
Epilepsy Results: Most of the studies were case series and retrospective analyses. No randomised controlled trials
Guidelines specifically designed for this type of clinical situation were identified. The level of scientific evidence to
Treatment guide clinical decisions is therefore low.
Conclusions: In this review we make recommendations based on the best scientific evidence available for
treating epilepsy in patients with other comorbidities, including the treatment of epileptic seizures in
acute situations as well as chronic antiepileptic treatment. When no scientific evidence is available, our
recommendations are based on pharmacokinetic criteria and tolerability of antiepileptic drugs, using
accumulated experience and the consensus of the members of the Andalusian Epilepsy Society.
ß 2010 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 376
2. Search strategy and selection criteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 376
3. Cardiovascular disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 376
4. Lung disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 377
5. Liver disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 377
6. Kidney disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378
7. Porphyria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378
8. Organ transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378
9. Thyroid disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378
10. Metabolic disorder . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378
10.1. AEDs and bone metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378
10.2. AEDs and obesity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378

§
This review has been extracted, modified and adapted from the Andalusian Guideline on Epilepsy 2009 (http://www.guiasade.com), written by: O. Alonso-Luengo, E.
Bauzano-Poley, H. Busquier-Hernández, R. Camino-León, F. Cañadillas-Hidalgo, J. Galán-Barranco, F.J. Gascón-Jiménez, A. Galdón-Castillo, D. Garcı́a-Solı́s, J. Gutiérrez-Garcı́a,
M. Ley-Martos, E. López-Laso, J. Martı́nez-Antón, C. Martı́nez-Parra, C. Martı́nez-Quesada, J. Mercadé-Cerdá, M.D. Morales-Martı́nez, V. Moreno-Alegre, M. Nieto-Barrera, E.
Pita-Calandre, M.M. Quesada-Lucas, P.A. Quiroga-Subirana, J. Ramos-Lizana, C. Robles-Vizcaı́no, J.J. Rodrı́guez-Uranga, S. Roldán-Aparicio, M. Rufo-Campos, J. Ruiz-Giménez,
J.C. Sánchez-Álvarez, P.J. Serrano-Castro, F. Villalobos-Chaves.
* Corresponding author at: Hospital de Rehabilitación y Traumatologia, Servicio de Neurologia, Carretera de Jaen, s/n 18013, Granada, Spain. Tel.: +34 659091751;
fax: +34 958021564.
E-mail address: jesusruizgimenez@hotmail.com (J. Ruiz-Giménez).

1059-1311/$ – see front matter ß 2010 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.seizure.2010.05.008
376 J. Ruiz-Giménez et al. / Seizure 19 (2010) 375–382

11. Infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378


12. Mental disability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 379
13. Psychiatric disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 379
14. Cognitive impairment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 380
15. Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 380
16. Brain tumour . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 380
17. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 381
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 381

1. Introduction nytoin, primidone, tiagabine, topiramate, valproic acid,


zonisamide, lacosamide) combined with the keywords ‘‘cardio-
Patients with epilepsy often have comorbidities. Several vascular disease’’, ‘‘lung disease’’, ‘‘liver disease’’, ‘‘kidney disease’’,
population surveys have found an increased prevalence of ‘‘porphyria’’, ‘‘organ transplantation’’, ‘‘thyroid disease’’, ‘‘meta-
comorbid conditions in persons with epilepsy as compared with bolic disorder’’, ‘‘infection’’, ‘‘mental disability’’, ‘‘psychiatric
the general population. Two population studies from Canada disorder’’, ‘‘cognitive impairment’’, ‘‘stroke’’ and ‘‘brain tumour’’.
reported a higher prevalence of stroke, diabetes, heart disease, high The Canadian Medical Association (CMA) Infobase (http://
blood pressure, asthma, chronic bronchitis, stomach/intestinal www.cma.ca/cpgs/index.asp), the National Guideline Clearing-
ulcers, arthritis, thyroid conditions, migraine, Alzheimer’s disease house (http://www.guideline.gov/index.asp), the National Library
and cancer in persons with a history of epilepsy.1 Epilepsy was also for Health guidelines finder (http://www.library.nhs.uk/GUIDE-
associated with an increased prevalence of mental health LINESFINDER) and other Web sites were searched for existing
disorders, especially anxiety and depression, compared with the evidence-based practice guidelines.
general population.2 Other studies from England and California Relevant articles and abstracts were selected and reviewed by
also found increased rates of somatic and psychiatric conditions in the authors, and the reference lists from these sources were
persons with epilepsy.3,4 searched for additional trials, as were the reference lists from
Comorbidities can be a direct cause of epilepsy, as with stroke, relevant review articles. Our search covered all relevant data to
be in complex relationship with epilepsy, as with psychiatric October 1, 2009. Most of the studies identified were case series and
disorders, or be associated with no apparent relationship as, for retrospective analyses. No randomised, controlled trials specifical-
example, with heart disease.5 In any case, it has been demonstrated ly designed for this type of clinical situation were identified. The
that people with epilepsy have a poorer health status and are more guidelines on epilepsy treatment identified and reviewed8–10 do
likely to report poor health-related quality of life than the general not provide information about epilepsy treatment in relation to
population. Although seizure freedom should continue to be a comorbidities. The level of scientific evidence to guide clinical
primary clinical goal, optimal care should also include identifica- decisions is therefore low. One exception is the guideline for the
tion and management of comorbidities. Likewise, the antiepileptic management of epilepsy in adults with an intellectual disability
treatment of these patients should be taken under special published by the International Association of the Scientific Study of
consideration given potential side effects and interactions with Intellectual Disability.11
other medications in patients with comorbidities.4,6 In this respect In the absence of human evidence, we used AED pharmacoki-
we believe that bearing in mind the comorbidities of the patients netic criteria and tolerability, as well as the accumulated
with epilepsy at the time of chosing an antiepileptic drug (AED) experience and consensus of the members of the Andalusian
should improve overall health and quality of life in those with Epilepsy Society. The results of the review and recommendations
epilepsy. Until recently, few options were available in the are presented below organized by type of comorbidity.
pharmacological treatment of epilepsy. In the last few years,
however, more than ten AEDs have been introduced. Although 3. Cardiovascular disease
overall these new AEDs have not demonstrated greater efficacy
than older drugs, many of them have an improved pharmacoki- In the acute management of an epileptic seizure, intravenous
netic profile and are better tolerated. These advantages make these phenytoin (PHT) can cause arrhythmias and hypotension. The risk
AEDs potentially useful in patients with epilepsy and other is higher if there is a history of heart disease or if PHT is given at a
comorbidities.7 rapid infusion rate. If intravenous use is necessary in a patient with
Unfortunately, despite this therapeutic diversification and the heart disease, the infusion rate must not exceed 10 mg/min, and
high prevalence of comorbidities in patients with epilepsy, no electrocardiographic and blood pressure monitoring must be
systematic investigations have evaluated the most suitable AEDs performed.12 The use of intravenous PHT or fosphenytoin is
for patients with epilepsy and with other medical problems. In this contraindicated in patients with severe heart disease and second or
review, we searched Medline, Cochrane Library, Guidelines third degree atrioventricular block.13 Valproic acid (VPA) is a good
databases and relevant publications for research studies of alternative to PHT in these patients because no abnormalities have
epilepsy treatment in patients with other comorbidities. Our goal been observed in heart rate or in blood pressure, even when a fast
was to evaluate the evidence and provide recommendations based intravenous infusion rate is used.14 Levetiracetam (LEV) also
on the best available scientific evidence. appears safe, although few studies on its use in emergency
situations are available. Benzodiazepines (BZD) can be used with
2. Search strategy and selection criteria careful monitoring of respiratory function.15–17
In chronic antiepileptic treatment, carbamazepine (CBZ),
MEDLINE and the Cochrane Library databases were searched oxcarbazepine (OXC) and PHT should be used with caution in
using ‘‘anticonvulsant’’ (Medical subject heading (MeSH) ‘‘antiep- patients with heart disease, and they should be avoided in the
ileptic drugs’’ (MeSH) or MeSH terms for AEDs (benzodiazepines, event of atrioventricular conduction dysfunction.18 Pregabalin
carbamazepine, clobazam, ethosuximide, gabapentin, levetirace- (PGB) should be used with care in cases of heart failure due to left
tam, lamotrigine, oxcarbazepine, phenobarbital, pregabalin, phe- ventricular systolic dysfunction.19 In general, enzyme-inducing
J. Ruiz-Giménez et al. / Seizure 19 (2010) 375–382 377

Table 1
Precautions in the management of AEDs together with other commonly used drugs in heart disease.
Antiplatelets Salicylates increase the free fraction of VPA.
Ticlopidine increases the levels of PHT and CBZ.
Antiarrhythmics Enzyme-inducing AEDs increase the metabolism of antiarrhythmics and so it may be necessary to increase the doses of the latter.
Dilthiazem and verapamil increase the levels of CBZ.
Amiodarone increases the levels of PHT.
Antihypertensives Enzyme-inducing AEDs increase the metabolism of beta-blockers and dihydropyridine calcium antagonists
PHT reduces the active metabolite of losartan by up to 63%.
VPA increases levels of nimodipine by 50%.
Oral anticoagulants (OAC) Enzyme-inducing AEDs reduce the anticoagulant effect of OACs
Complex interaction with PHT: PHT increases the initial effect of the OAC and then reduces it. OACs can increase
the levels of PHT. The dose of both drugs must be adjusted if used together.
Diuretics PHT reduces the diuretic response to furosemide.
Use with precaution when associated with CBZ or OXC because of the risk of hyponatraemia.
Digoxin PHT significantly reduces digoxin levels.
Anti-lipidemics In general, enzyme-inducing AEDs stimulate the metabolism of these drugs.

AEDs should be avoided because of their many interactions with Enzyme-inducing AEDs reduce theophyline concentration and
drugs that are commonly used in heart disease20 (Table 1). The theophyline can lower the levels of CBZ and PHT.20
most recommended AEDs are LEV, lamotrigine (LTG), topiramate
(TPM), VPA and zonisamide (ZNS). Gabapentin (GBP) can also be 5. Liver disease
useful as an add-on therapy.
In the case of hepatic dysfunction, the hepatic metabolism of
4. Lung disease some AEDs is impaired, and there may be associated hypoalbu-
minaemia. For this reason it is important to consider the
Parenteral use of barbiturates, BZD and PHT can cause pharmacokinetics of AEDs in the presence of hepatic dysfunction
respiratory depression. When used in patients with respiratory (Table 2). If the hepatic impairment is mild, however, it is generally
impairment, the heart rate, respiratory rate and oximetry should not necessary to adjust the AED dose.23
be monitored. Access to cardiopulmonary resuscitation equipment PB should be avoided in the acute management of epileptic
should also be considered. Parenteral VPA offers a safe and seizures in patients with liver disease because it can trigger or
effective alternative.14,21 LEV can also be effective in these aggravate hepatic encephalopathy. BZDs are metabolised in the
situations.15,17,22 liver and can also cause hepatic encephalopathy. They should only
In the chronic treatment of patients with respiratory im- be used if absolutely needed and at lower doses than normal.24 VPA
pairment, AEDs with a potential for inducing respiratory depres- is contraindicated because of its hepatotoxic capacity in patients
sion, such as barbiturates and BZDs, should be avoided. BZDs, with underlying liver disease.25 PHT should be used with caution
moreover, increase bronchial secretions, particularly in children.21 because, due to its high protein binding, the free fraction is

Table 2
Pharmacokinetic properties of AEDs of particular interest in the case of hepatic impairment, renal impairment and/or haemodialysis.

Hepatic metabolism Plasma protein Dose adjustment in renal Dose adjustment in haemodialysis (HD)
binding (%) impairment (RI)

BZD ++ = =
CBZ ++ 75 = =
ESM ++ 0 # 25% of dose if Crt < 10% 50% can be eliminated in 6 h of HD.
Best given after HD
GBP – 0 Crt>80: = 200–300 mg after HD in a single
dose or 100–150 mg/day + SD of 125–250 after HD
Crt 50–79: 200–600 mg/8 h
Crt 30–49: 100–300 mg/8 h
Crt 15–29: 300/48 h to 600/24 h
Crt < 15: 300/48 h to 300/24 h
LEV + <10 Crt 50–79: 0.5–1 g/12 h 250–500 mg after HD
Crt 30–49: 250/750 mg/12 h
Crt < 30: 250–500 mg/12 h
LTG ++ 55 # dose in moderate and severe RI 20% is eliminated in 4 h of HD. Best given after HD
OXC ++ 40 # 50% of dose if Crt < 30% ? Avoid in HD due to insufficient data
PB/PRM ++ 45 # Normal dose SD
PHT ++ 90 = =
PGB  0 Crt > 60: = Single SD of 25–100 mg after HD
Crt 30–59: 25–100/8 h
Crt 15–30: 25–50/8 h
Crt < 15: 25–75/day (3 intakes)
TGB ++ 96 = =
TPM + 15 # 50% in moderate and severe RI. 50–100 mg/12 h
50–100 mg after HD
VPA ++ 90 = SD may be needed
ZNS + 40 ? Slower dose adjustment 200–400 mg/day after HD
SD of 100–200 mg the morning before HD

Mainly hepatic metabolism: ++; partly hepatic metabolism: +; extrahepatic metabolism: ; Crt: creatinine clearance in ml/min; (?) no data available, use with caution; = no
dose adjustment required; SD: supplementary dose.
378 J. Ruiz-Giménez et al. / Seizure 19 (2010) 375–382

increased in the presence of hypoalbuminaemia with the corticosteroids and so it may be necessary to increase the dose of
possibility of intoxication.27 LEV is the most recommended these drugs. VPA only causes a minimal reduction of cyclospor-
therapeutic alternative in the acute phase of epileptic seizures ine and tacrolimus levels. Cyclosporine binds largely to plasma
in patients with liver disease.15,22,26 proteins, and this can significantly increase the free fraction of
In chronic treatment, the most suitable AEDs are those with a AEDs that have a high percentage of protein binding. Azathio-
low level of protein binding and limited hepatic metabolism: GBP, prine, mycophenolate and muromonab-CD3 (OKT3) are not
LEV, OXC, PGB and TPM.27 However, in the event of severe liver significantly affected by AEDs.20 In this respect, the good
disease, it may also be necessary to adjust the dose of these AEDs pharmacokinetic properties of second-generation AEDs make
because of the renal dysfunction that is often concomitant. For this them very useful in transplant recipients.37
reason, in the presence of severe liver disease it is advisable to 3. AED side effects may have a negative impact on the transplanted
reduce the normal dose of LEV by 50% and the normal dose of TPM tissue. VPA should be avoided in liver transplantation and in the
by 30%.28 OXC has low protein binding and a lower potential for engraftment phase of bone marrow transplantation (the first 2–
enzyme induction compared to CBZ. BZD, CBZ, ethosuximide 6 weeks).38 CBZ, OXC, PB and PRM should be avoided in bone
(ESM), phenobarbital (PB), PHT, primidone (PRM), tiagabine (TGB) marrow transplant cases.39
and ZNS can be used under close surveillance. The use of LTG and
VPA is not recommended.27
Given these factors, we conclude that GBP, LEV, PGB and TPM
6. Kidney disease are the most appropriate AEDs to treat epilepsy in patients with
liver transplantation. BZD, LTG and VPA are the most appropriate in
In patients with renal impairment, AEDs which are mainly patients with kidney transplantation. GBP, LEV, LTG and TPM are
eliminated by the kidneys will have a longer half life and tend to the most appropriate in bone marrow transplantation.
accumulate in the blood because of the reduced glomerular
filtration rate and tubular secretion.29 For this reason, LEV is not 9. Thyroid disease
recommended in acute management.15,30 Care should be taken
with the chronic use of GBP, LEV, LTG, OXC, PB, PGB, PRM, TPM and Enzyme-inducing AEDs (CBZ, PB, PHT, PRM) influence thyroid
ZNS because these AEDs are eliminated to a greater or lesser extent hormone metabolism, causing a decrease in total and free thyroxin
by the kidneys. They are not strictly contraindicated, but major levels. This modification is usually subclinical and reverses when
dose adjustments will be needed, along with close surveillance AEDs are withdrawn, particularly in healthy patients. However, it
because of the risk of toxicity. Moreover, after haemodialysis, may be clinically significant in patients with hypothyroidism who
plasma levels of these AEDs change and supplementary doses will are on replacement therapy.40,41 Likewise, VPA can cause a
be required. The AEDs most affected by haemodialysis are the most subclinical, reversible increase in TSH.42 Although data on the
hydrosoluble ones, the ones with a low level of protein binding and effect of second-generation AEDs are currently insufficient, it is
those with a lower distribution volume31 (Table 2). Furthermore, likely that the AEDs with a moderate enzyme-inducing effect (OXC,
TPM and ZNS should be avoided in the event of nephrolithiasis or TPM) will also affect thyroid hormones, while the non-enzyme-
when there is a possibility of developing it.32,33 The most inducing AEDs will not affect them.43
recommended AEDs in renal impairment and haemodialysis are
those that are mainly eliminated by the liver, such as BZD, CBZ, 10. Metabolic disorder
ESM, PHT, TGB and VPA.31
10.1. AEDs and bone metabolism

7. Porphyria Enzyme-inducing AEDs and PHT in particular accelerate


vitamin D catabolism and increase bone turnover.44 VPA interferes
The induction of hepatic haemosynthesis on the part of with osteoblast function. Although LEV and LTG do not appear to
enzyme-inducing AEDs can exacerbate the symptoms of porphyr- have a significant effect in this respect, a case–control study
ia. Treatment with CBZ, PB, PHT, PRM, TPM, VPA and ZNS should be revealed that enzyme-inducing and non-enzyme-inducing AEDs
avoided. A porphyrinogenic capacity has also been shown in in alike are an independent risk factor for osteoporosis.45
vitro studies on LTG and TGB.34
The use of non-enzyme-inducing AEDs such as GBP, LEV and 10.2. AEDs and obesity
PGB is recommended. OXC has been used successfully in an
isolated case of porphyria cutanea tarda and also in another case of Some AEDs are associated with weight gain (CBZ, CLB, GBP, PGB
intermittent acute porphyria.35 If parenteral treatment is neces- and VPA) and others with weight loss (TPM and ZNS). The majority
sary, the use of LEV should be considered. Intravenous magnesium have a neutral effect in this respect.46 Some studies with small
sulphate and BZDs have also been used in isolated cases of status sample size found that patients on VPA treatment who gained
epilepticus, although in theory BZDs can worsen porphyria weight showed insulin resistance that was reversible on with-
symptoms.36 drawal of VPA.47

8. Organ transplantation 11. Infection

To optimise AED treatment in transplant recipients, we must There are important interactions between AEDs and anti-
consider three fundamental factors: infective agents that should be considered in the management of
infectious diseases (particularly those requiring prolonged treat-
1. The possible presence and degree of hepatic or renal dysfunction ment) and of symptomatic seizures from infectious processes.48
in patients who are liver or kidney recipients. In the treatment of neurocysticercosis, enzyme-inducing AEDs
2. Pharmacological interactions between AEDs and immunosup- reduce the concentration of praziquantel and albendazole by over
pressive drugs. Enzyme-inducing AEDs (CBZ, PB, PHT, PRM) can 50%. In the treatment of tuberculosis, isoniazid inhibits the
reduce plasma levels of cyclosporine, tacrolimus, sirolimus and metabolism of CBZ, PHT and VPA, and can cause toxicity.
J. Ruiz-Giménez et al. / Seizure 19 (2010) 375–382 379

Table 3
Principal pharmacological interactions between AEDs and commonly used antibiotics.

CBZ LTG PB PHT VPA

Albendazole # = # = # =
Carbapenems #
Ciprofloxacin #"
Clarithromycin "
Doxycycline # # #
Erythromycin "
Fluconazole " " "
NRTI = = = = = = = = =
NNRTI # " = " # " # " = "
PI # " = " # " # " = "
Isoniazid " " "
Ketoconazole "
Metronidazole # " # #
Praziquantel # = # = # =
Rifampicin # # # # #
Sulphonamides "

The arrows on the left hand side of the boxes indicate the effect of interaction on the plasma levels of the antibiotics and the ones on the right hand side indicate the effect on
plasma levels of AEDs. (") Increased plasma level. (#) Decreased plasma level. (=) No significant effect. NRTI: nucleoside reverse transcriptase inhibitors. NNRTI: non-
nucleoside reverse transcriptase inhibitors. PI: protease inhibitors.

Table 4
Optimisation of treatment in patients with epilepsy with psychiatric comorbidity.

Antiepileptic treatment Psychiatric treatment

Recommended To be avoided Recommended To be avoided

Depression CBZ, GBP, LTG, OXC, PGB, VPA PB, PHT, PRM, TGB, TPM Citalopram Escitalopram Sertraline Amoxapine Maprotiline Bupropion
Trazodone Venlafaxine
Anxiety BZD, GBP, PGB, VPA LEV BZD SSRIs –
Psychosis LTG, OXC, VPA ESM, LEV, TPM Olanzapine Quetiapine Risperidone Chlorpromazine Clozapine

Conversely, rifampicin reduces the plasma concentration of CBZ, these patients, and therefore patients with impaired coordination
LTG, PB, PHT and VPA.20 or gait should avoid the use of CBZ, LTG, OXC, PB, PHT and PRM and
AED treatment in HIV infection depends on whether or not the patients with severe behavioural problems should avoid the use of
patient is on antiretroviral therapy. If the patient is not on LEV, TPM and ZNS.54
antiretrovirals, any AED can be prescribed. Some studies suggest
that VPA might stimulate the viral replication of latent HIV in vitro,
but more recent results are negative and there is thus no evidence 13. Psychiatric disorders
against it.49–51 If the patient is on antiretroviral treatment, then the
reciprocal interactions between AEDs and antiretrovirals must be To optimise antiepileptic treatment in patients with concomi-
taken into consideration (Table 3). In these cases, non-enzyme- tant psychiatric disorders, the following should be considered:
inducing AEDs that are not metabolised in the liver are recom-
mended. The most suitable AEDs for HIV patients are LEV, PGB and 1. Possible influence of AEDs on psychiatric symptoms. Depending
TPM. GBP can be useful too, but usually as a secondary therapy line.48 on the type of psychiatric comorbidity, some AEDs are more
Any AEDs can be used in general and neuromeningeal appropriate than others57–59 (Table 4).
infections, although it is advisable to select ones that have few 2. Pharmacological interactions. Enzyme-inducing AEDs can lower
interactions. Table 3 summarises the most significant interactions the plasma levels of other psychotropic drugs (neuroleptics,
between AEDs and antimicrobial agents.20,52 tricyclic and selective serotonin reuptake inhibitor antidepres-
sants), hindering the control of psychiatric symptoms. The
12. Mental disability interaction between VPA and amitriptyline or nortriptyline can
cause an increase of up to 60% in the plasma levels of these
There is a much higher incidence of epilepsy amongst patients drugs, causing intoxication. There are no significant interactions
with mental disabilities than in the general population. AED between AEDs and lithium.20
efficacy is similar in patients with and without mental disabilities Tricyclic antidepressants (TCAs) can inhibit AED metabolism,
although patients with mental disability are generally more causing toxicity symptoms. The same occurs with some
susceptible to AED side effects and often require continued selective serotonin reuptake inhibitor antidepressants (SSRIs)
treatment over longer periods.53 Therefore, monotherapy should such as fluoxetine, paroxetine and fluvoxamine, although these
be used whenever possible.54 drugs have a better pharmacokinetic profile than TCAs. Other
AEDs with sedative effect or potential cognitive reduction, such antidepressants such as citalopram, escitalopram, sertraline,
as BZD, CBZ, PB, PHT, PRM and TPM should be avoided for long term trazodone and venlafaxine do not have a significant effect on
treatment. However, the use of Diazepam (oral or rectal route) or AED metabolism.20,58,59
Midazolam (buccal or intranasal route) is recommended for acute Most antipsychotic drugs interfere with the hepatic metab-
treatment of prolonged and cluster seizures.11 Whenever possible, olism of AEDs to a variable degree. Clozapine should be avoided
AEDs that have a lower rate of cognitive adverse effects such as in patients with epilepsy, while olanzapine, quetiapine and
GBP, LEV, LTG, OXC and VPA should be used.11,55,56 One should also risperidone do not usually require dose adjustment, even when
consider that other side effects in AEDs are often potentiated in used in combination with enzyme-inducing AEDs.58
380 J. Ruiz-Giménez et al. / Seizure 19 (2010) 375–382

Table 5
Common side effects that can be potentiated must also be
Pharmacological interactions between AEDs and cytotoxic and corticosteroid drugs.
taken into consideration. SSRIs can cause hyponatraemia and so
they must be used with caution when combined with CBZ or CBZ PB PHT VPA
OXC.57 Anthracyclines # # # # # # #
3. Potential epileptogenic effect of antidepressants and antipsy- Cyclophosphamide # # #
chotics. As a general rule the epileptogenic risk is low for TCAs, Cisplatin # # # #"
Corticosteroids # # # #"
SSRIs and antipsychotics alike. The risk is especially minimised
Etoposides # # #
when these drugs are used within the therapeutic range, with a Fluorpyrimidines # "
slow dose titration and avoiding complex combinations Irinotecan topotecan # # #
between them. There are, however, exceptions, and certain Methotrexate # # # #
antidepressants such as amoxapine, maprotiline and bupropion Nitrosureas # # # " "
Taxoides # # #
and certain neuroleptics such as clozapine and chlorpromazine Vincristine # # #
have a higher epileptogenic risk.59
The arrows on the left hand side of the boxes indicate the effect of interaction on the
plasma levels of the cytotoxic and corticosteroid drugs and the ones on the right
hand side indicate the effect on plasma levels of AEDs. (") Increased plasma level. (#)
14. Cognitive impairment Decreased plasma level. (=) No significant effect.

Cognitive impairment as a consequence of reduced brain


functional reserve or degenerative disease can be aggravated by have suffered a stroke.67 In a prospective, randomised study with
AEDs. Before starting treatment with an AED, especially in the 64 stroke patients who had suffered a first seizure, it was
elderly, the patient’s cognitive condition should be assessed to demonstrated that LTG was better tolerated than CBZ, and no
avoid using AEDs that can aggravate incipient mental impairment. significant differences were observed in efficacy.68
Among the AEDs with the most severe impact on cognitive
function are BZD, PB and PRM, and to a lesser degree PHT and TPM. 16. Brain tumour
The AEDs with the least impact on cognitive function are GBP, LEV,
LTG and PGB. In general, high doses and combination AED therapy In patients with brain tumours, pharmacological interactions
aggravate the harmful potential of the drugs from a cognitive between AEDs and cytotoxic agents and corticosteroids should be
standpoint.60,61 considered (Table 5). In a retrospective study of patients with
glioblastoma multiforme treated with conventional chemothera-
15. Stroke py, patients treated with enzyme-inducing AEDs had lower
survival rates than patients treated with non-enzyme-inducing
For different reasons conventional AEDs such as BZD, CBZ, PHT, AEDs.69
PB and VPA are in general less recommended in patients with AED tolerability must be considered in patients with brain
stroke. Firstly, a delay in functional recovery has been reported as a tumours because they appear to suffer more side effects than the
result of their use in patients with stroke.62,63 Furthermore, they general population, including more frequent skin rashes. Severe
can have significant interactions with salicylates and oral antic- cutaneous reactions have been reported in patients treated with
oagulants20 (Table 1). In spite of these disadvantages, the CBZ, PHT or PB who were being given brain radiotherapy.70 As is
availability of PHT and VPA in IV form makes them still a good the case with stroke, the availability of an IV form of PHT makes it
choice for use in acute situations in these patients. still a good choice for use in acute situations in these patients.
Some recent AEDs such as GBP, LEV, LTG, OXC and TPM do not Response to conventional AEDs seems to be variable, with 70%
seem to have interactions with antiplatelets or anticoagulants or to of patients with brain tumours treated with CBZ, 51% of those
affect functional prognosis following a stroke.20 Both GBP and LEV treated with PHT, and 44% of those treated with VPA having
have been evaluated specifically in stroke patients and they have persistent seizures. This suggests that VPA could be the
been shown to be safe and effective in these patients.64–66 conventional AED of choice in patients with brain tumours.71
There is evidence from clinical trials that LTG and GBP are more LEV has shown good efficacy and tolerability in monotherapy in
effective than CBZ in elderly patients with epilepsy, many of whom prospective studies in patients with brain tumours.72 In a recent

Table 6
Recommendations for the use of AEDs in patients with epilepsy and other comorbidities.

Most recommended AEDs Less recommended AEDs AEDs to be avoided

Heart disease LEV, LTG, TPM, VPA, ZNS. GBP* CBZ, OXC, PGB, PHT _
Lung disease LEV, LTG, OXC, PGB, TPM, VPA, ZNS. GBP* CBZ, PHT BZD, PB, PRM
Hepatic impairment LEV, OXC, PGB, TPM. GBP* BZD, CBZ, ESM, PB, PHT, PRM, TGB, ZNS LTG, VPA
Renal impairment BZD, CBZ, ESM, PHT, TGB, VPA GBP, LEV, LTG, OXC, PB, PGB, PRM, TPM, ZNS _
Porphyria LEV, OXC, PGB. GBP* BZD CBZ, LTG, PB, PHT, PRM,
TGB, TPM, VPA, ZNS
Liver transplantation LEV, PGB, TPM. GBP* CBZ, PB, PHT, PRM VPA
Kidney transplantation BZD, LTG, VPA AEDs with renal excretion _
Bone marrow transplantation LEV, LTG, TPM. GBP* _ CBZ, OXC, PB, PRM, VPA
Hypothyroidism BZD, LEV, LTG, PGB, ZNS. GBP* OXC, TPM, VPA CBZ, PB, PHT, PRM
Osteoporosis BZD, LEV, LTG, PGB, ZNS. GBP* VPA CBZ, PB, PHT, PRM
Obesity TPM, ZNS CBZ, CLB GBP, PGB, VPA
HIV LEV, PGB, TPM. GBP* BZD, LTG, OXC, VPA, ZNS CBZ, PB, PHT, PRM
Mental disability LEV, LTG, OXC, VPA. GBP* PGB, ZNS BZD, CBZ, PB, PHT, PRM, TPM
Cognitive impairment LEV, LTG, PGB. GBP* CBZ, OXC, VPA, ZNS BZD, PB, PHT, PRM, TPM
Stroke LEV, LTG. GBP* CBZ, OXC, PHT, TPM, VPA BZD, PB, PRM
Brain tumour LEV, VPA. GBP*, PGB*, ZNS* CBZ, LTG, OXC, PHT, TPM PB, PRM

(*) Useful as add-on therapy. (The medications are ordered alphabetically and not necessarily by order of recommendation.)
J. Ruiz-Giménez et al. / Seizure 19 (2010) 375–382 381

observational study on patients with glioma, the higuest responder 16. Hulhoven R, Rosillon D, Bridson WE, et al. Effect of levetiracetam on cardiac
repolarization in healthy subjects: a single-dose, randomized, placebo- and
percentage was obtained with a combination of VPA and LEV.73 active-controlled, four-way crossover study. Clin Ther 2008;30(2 (Febru-
OXC monotherapy has showed similar efficacy and a lower ary)):260–70.
incidence of side effects than traditional AEDs in a comparative 17. Nau KM, Divertie GD, Valentino AK, et al. Safety and efficacy of levetiracetam for
critically ill patients with seizures. Neurocrit Care 2009;11(1):34–7.
study.74 LTG is a well-established treatment in focal epilepsy 18. Kaul S, Meena AK, Murthy JM. Carbamazepine induced bradicardia. Neurol
related to brain tumours, although it takes time to attain a India 2000;48(4):403–4.
therapeutic dose and there is an increased risk of exanthema. GBP 19. Murphy N, Mockler M, Ryder M, et al. Decompensation of chronic heart failure
associated with pregabalin in patients with neuropathic pain. J Card Fail
has shown good efficacy and tolerability in add-on therapy in 2007;13:227–9.
patients with brain tumours.70 Preliminary data from recent and 20. Patsalos PN, Perucca E. Clinically important drug interactions in epilepsy:
small clinical series indicate that PGB and ZNS may represent valid interactions between antiepileptic drugs and other drugs. Lancet Neurol
2003;2:473–81.
alternatives as add-ons in brain tumour-related epilepsy, although
21. Mehta V, Singhi P, Singhi S. Intravenous sodium valproate versus diazepam
larger studies are needed.75,76 infusion for the control of refractory status epilepticus in children: a random-
ized controlled trial. J Child Neurol 2007;22:1191–7.
22. Di Bonaventura C, Mari F, Fattouch J, et al. Use of Levetiracetam in treating
17. Conclusions epilepsy associated with other medical conditions. Acta Neurol Scand
2006;113:82–6.
23. Shet RD. Metabolic concerns associated with antiepileptics medications. Neu-
A high percentage of patients with epilepsy have some
rology 2004;63(10 Suppl. 4):S24–9.
comorbidity, and this is an important factor when selecting the 24. Ochs HR, Greenblatt DJ, Eckardt B, et al. Repeated diazepam dosing in cirrhotic
most suitable AED. Although the level of scientific evidence patients: cumulation and sedation. Clin Pharmacol Ther 1983;33(4):471–6.
concerning epilepsy treatment in patients with other associated 25. Gopaul S, Farrell K, Abbot F. Effects of age and polytherapy, risk factors of
valproic acid (VPA) hepatotoxicity, on the excretion of thiol conjugates of (e)-
diseases is low and rarely supported by clinical trials, there is a 2,4-dieneVPA in people with epilepsy taking VPA. Epilepsia 2003;44(3):322–8.
need for evaluating available data that support the use of certain 26. Blio L, Meo R, de Leva MF, et al. Levetiracetam in patients with epilepsy and
AEDs in the presence of specific diseases. Table 6 summarises chronic liver disease: observations in a case series. Clin Neuropharmacol
2008;31(4):221–5.
which AEDs are most recommended in each situation and which 27. Ahmed SN, Siddiqi ZA. Antiepileptic drugs and liver disease. Seizure 2006;15:
should be avoided or are less recommended, based on the best 156–64.
evidence available and the experience and consensus of the 28. Brockmoller J, Thomsen T, Wittstock M, et al. Pharmacokinetics of levetirace-
tam in patients with moderate to severe liver cirrhosis (Child-Pugh classes A, B
members of the Andalusian Epilepsy Society. and C): characterization by dynamic liver function tests. Clin Pharmacol Ther
2005;77:529–41.
29. Gabardi S, Abramson S. Drug dosing in chronic kidney disease. Med Clin North
Conflicts of interest Am 2005;89:645–87.
30. Vulliemoz S, Iwanowski P, Landis T, et al. Levetiracetam accumulation in renal
The authors have received research grants and speaker fees failure causing myoclonic encephalopathy with triphasic waves. Seizure
2000;18(5 June):376–8.
from Eisai, Esteve, GSK, Iquinosa, Janssen-Cilag, Juste, Novartis, 31. Israni RK, Kasbekar N, Haynes K, et al. Use of antiepileptic drugs in patients with
Parke-Davis, Pfizer, Sanofi and UCB-Pharma. kidney disease. Sem Dialysis 2006;19:408–16.
32. Vega D, Maalouf NM, Sakhaee K. Increased propensity for calcium phosphate
kidney stones with topiramate use. Expert Opin Drug Saf 2007;6(5 (Septem-
References ber)):547–57.
33. Faught E. Review of Unites States and European clinical trials of zonisamide
1. Tellez-Zenteno JF, Matijevic S, Wiebe S. Somatic comorbidity of epilepsy in the in the treatment of refractory partial-onset seizures. Seizure 2004;135:
general population in Canada. Epilepsia 2005;46(12):1955–62. S59–65.
2. Tellez-Zenteno JF, Patten SB, Jetté N, Williams J, Wiebe S. Psychiatric comor- 34. Hahn M, Gildemeister OS, Krauss GL, et al. Effects of new anticonvulsant
bidity in epilepsy: a population-based analysis. Epilepsia 2007;48(12 (Decem- medications on porphyrin synthesis in cultured liver cells: potential implica-
ber)):2336–44. tions for patients with acute porphyria. Neurology 1997;49:97–106.
3. Gaitatzis A, Carroll K, Majeed A, Sander WJ. The epidemiology of the comorbidi- 35. Bernadette GH, Rieck K, Runge U. Oxcarbacepine in focal epilepsy and hepatic
ty of epilepsy in the general population. javascript:AL_get(this, ‘jour’, ‘Epilep- porphyria: a case report. Epilepsia 2001;42:793–5.
sia.’). Epilepsia 2004;45(12):1613–22. 36. Zaatreh MM. Levetiracetam in porphyric status epilepticus: a case report. Clin
4. Elliott JO, Lu B, Shneker B, et al. Comorbidiy, health screening and quality of life Neuropharmacol 2005;28:243–4.
among persons with a story of epilepsy. Epilepsy Behav 2009;(14):125–9. 37. Glass GA, Stankiewicz J, Mithoefer A. Levetiracetam for seizures after liver
5. Ryvlin P. Optimizing therapy of seizures in specific clinical situations. Are the transplantation. Neurology 2005;64:1084–5.
exceptions the rule? Neurology 2006;67(Suppl. 4):S1–2. 38. Bjornsonn E. Hepatotoxicity associated with antiepileptic drugs. Acta Neurol
6. Hinnell C, Williams J, Metcalfe A, et al. Health status and health-related Scand 2008;118(5 (November)):281–90.
behaviors in epilepsy compared to other chronic conditions. A national popu- 39. Focosi D, Kast RE, Benedetti E, et al. Phenobarbital-associated bone marrow
lation-based study. Epilepsia 2010 (Article online in advance of print. Published aplasia: a case report and review of the literature. Acta Haematol 2008;119:
Online: January 7 2010). 18–21.
7. Bialer M. New antiepileptic drugs that are second generation to existing 40. Simko J, Horacek J. Carbamazepine and risk of hypothyroidism: a prospective
antiepileptic drugs. Expert Opin Investig Drugs 2006;15(6 (June)):637–47. study. Acta Neurol Scand 2007;116:317–21.
8. NICE. The epilepsies: the diagnosis and management of the epilepsies in adults 41. Ericsson UB, Bjerre I, Forsgren M, et al. Thyroglobulin and thyroid hormones in
and children in primary and secondary care. October 2004. http://www.ni- patients on long-term treatment with phenytoin, carbamazepine, and valproic
ce.org.uk/page.aspx?o=CG020. acid. Epilepsia 1985;26:594–6.
9. SIGN. Diagnosis and Management of Epilepsy in adults. A national clinical 42. Vainionpää LK, Mikkonen K, Rattyä J, et al. Thyroid function in girls with
guideline. April 2003. http://www.sign.ac.uk/pdf/sign70.pdf. epilepsy with carbamazepine, oxcarbazepine, or valproate monotherapy and
10. Singapore Ministry of Health. Epilepsy in adults. Singapore: Singapore Ministry after withdrawal of medication. Epilepsia 2004;45:197–203.
of Health; 2007 Jan. 43 p. http://www.guideline.gov/summary/pdf.aspx?doc_id 43. Perucca E. Clinically relevant drug interactions with antiepileptic drugs. Br J
=10590&stat=1&string=epilepsy. Clin Pharmacol 2006;61:246–55.
11. Clinical guidelines for the management of epilepsy in adults with an intellec- 44. Pack AM, Morrell MJ, Randall A, et al. Bone health in young women with
tual disability. Working Group of the International Association of the Scientific epilepsy after one year of antiepileptic drug monotherapy. Neurology
Study of Intellectual Disability. Seizure 2001;10:401–9. 2008;70:1586–93.
12. Donovan PJ, Cline D. Phenytoin administration by constant intravenous infu- 45. Ensrud KE, Walczak TS, Blackwell TL, et al., Osteoporotic Fractures in Men
sion: selective rates of administration. Ann Emerg Med 1991;20(2 (Febru- (MrOS) Study Research Group. Antiepileptic drug use and rates of hip bone loss
ary)):139–42. in older men: a prospective study. Neurology 2008;71:723–30.
13. Adams BD, Buckley NH, Kim JY, et al. Fosphenytoin may cause hemodynami- 46. Ben-Menachem E. Weight issues for people with epilepsy. A review. Epilepsia
cally unstable bradydisrhythmias. J Emerg Med 2006;30:75–9. 2007;48(Suppl. 9):S42–5.
14. Agarwal P, Kumar N, Chandra R, et al. Randomized study of intravenous 47. Luef G, Abraham I, Hoppichler F, et al. Increase in postprandial serum insulin
valproate and phenytoin in status epilepticus. Seizure 2007;16:527–32. levels in epileptic patients with valproic acid therapy. Metabolism
15. Ramael S, Daoust A, Otoul C, et al. Levetiracetam intravenous infusion: a 2002;51:1274–8.
randomized, placebo controlled safety and pharmacokinetic study. Epilepsia 48. Bhigjee AI, Rosemberg S. Optimizing therapy of seizures in patients with HIV
2006;47:1128–35. and cysticercosis. Neurology 2006;67(Suppl. 4):S19–22.
382 J. Ruiz-Giménez et al. / Seizure 19 (2010) 375–382

49. Maggi JD, Halman MH. The effect of divalproex sodium on HIV replication in 69. Oberdornfer S, Piribauer M, Marosi C, et al. P450 enzyme inducing and non-
vivo. J Neuropsychiatry Clin Neurosci 2007;19(3 (Summer)):326–30. enzyme inducing antiepileptics in glioblastoma patients treated with standard
50. Lehrman G, Hogue B, Palmer SA, et al. Depletion of latent HIV-1 infection in chemotherapy. J Neurooncol 2005;72:255–60.
vivo: a proof-of-concept study. Lancet 2005;366:549–55. 70. Van Breemen M, Wilms EB, Vecht CJ. Epilepsy in patients with brain tumours:
51. Sagot-Lerolle N, Lamine A, Chaix ML, et al. ANRS EP39 study. Prolonged valproic epidemiology, mechanisms and management. Lancet Neurol 2007;6:421–30.
acid treatment does not reduce the size of latent HIV reservoir. AIDS 71. Wick W, Menn O, Meisner C, et al. Pharmacotherapy of epileptic seizures in
2008;22:1125–9. glioma patients: who, when, why and how long? Onkologie 2005;28:391–6.
52. Desai J. Perspectives on interactions between antiepileptic drugs (AEDs) and 72. Lim DA, Tarapore P, Chang E, et al. Safety and feasibility of switching from
antimicrobial agents. Epilepsia 2008;49(Suppl. 6):S47–9. phenytoin to levetiracetam monotherapy for glioma-related seizure control
53. Beavis J, Kerr M, Marson AG. Pharmacological interventions for epilepsy in following craniotomy: a randomized phase II pilot study. J Neurooncol
people with intellectual disabilities. Cochrane Database Syst Rev 2007;18(3): 2009;93(3 (July)):349–54.
CD005399. 73. Van Breemen MS, Rijsman RM, Taphoorn MJ, et al. Efficacy of anti-epileptic
54. Huber B, Hause U, Horstmann V. Seizure freedom with different therapeutic drugs in patients with gliomas and seizures. J Neurol 2009;256(9 (Septem-
regimens in intellectually disabled epileptic patients. Seizure 2005;14:381–6. ber)):1519–26.
55. Huber B, Bommel W, Hauser I, et al. Efficacy and tolerability of levetiracetam in 74. Maschio M, Dinapoli L, Vidiri A, et al. The role side effects play in the choice of
patients with therapy-resistant epilepsy and learning disabilities. Seizure antiepileptic therapy in brain tumor-related epilepsy: a comparative study on
2004;13:168–75. traditional antiepileptic drugs versus oxcarbazepine. J Exp Clin Cancer Res
56. Coppola G, Verrotti A, Resicato G, et al. Topiramate in children and adolescents 2009;28(6 (May)):60.
with epilepsy and mental retardation: a prospective study on behavior and 75. Novy J, Stupp R, Rossetti AO. Pregabalin in patients with primary brain tumors
cognitive effects. Epilpesy Behav 2008;12(2 (February)):253–6. and seizures: a preliminary observation. Clin Neurol Neurosurg 2009;111(2
57. Ovsiew F. Antiepileptic drugs in psychiatry. J Neurol Neurosurg Psychiatry (February)):171–3.
2004;75:1665–8. 76. Maschio M, Dinapoli L, Saveriano F, et al. Efficacy and tolerability of zonisamide
58. Brodtkorb E, Mula M. Optimizing therapy of seizures in adult patients with as add-on in brain tumor-related epilepsy: preliminary report. Acta Neurol
psychiatric co morbidity. Neurology 2006;67(Suppl. 4):S39–44. Scand 2009;120(3 (September)):210–2.
59. Mula M, Monaco F, Trimble MR. Use of psychotropic drugs in patients with
epilepsy: interactions and seizure risk. Expert Rev Neurother 2004;4:953–64.
60. Hommet C, Mondon K, Camus V, et al. Epilepsy and dementia in the elderly. Glossary
Dement Geriatr Cogn Disord 2008;25:293–300.
61. Mendez M, Lim G. Seizures in elderly patients with dementia: epidemiology AED: antiepileptic drug
and management. Drugs Aging 2003;20(11):791–803. BZD: benzodiazepines
62. Goldstein B. Common drugs may influence motor recovery after stroke. Neu- CBZ: carbamazepine
rology 1995;45:865–71. CLB: clobazam
63. Naidech AM, Kreiter KT, Janjua N, et al. Phenytoin exposure is associated with ESM: ethosuximide
functional and cognitive disability after subarachnoid hemorrhage. Stroke GBP: gabapentin
2005;36:583–7.
LEV: levetiracetam
64. Álvarez-Sabin J, Montaner J, Padro L, et al. Gabapentin in late-onset poststroke
LTG: lamotrigine
seizures. Neurology 2002;59:1991–3.
OXC: oxcarbazepine
65. Kutlu G, Gomceli YB, Unal Y, et al. Levetiracetam monotherapy for late post-
PB: phenobarbital
stroke seizures in the elderly. Epilepsy Behav 2008;13:542–4.
66. Belcastro V, Costa C, Galletti F, et al. Levetiracetam in newly diagnosed late- PGB: pregabalin
onset post-stroke seizures: a prospective observational study. Epilepsy Res PHT: phenytoin
2008;82(2–3 (December)):223–6 (Epub 2008 September 30). PRM: primidone
67. Rowan AJ, Ramsay RE, Collins JF, et al. New onset geriatric epilepsy. A random- SSRI: selective serotonin reuptake inhibitors
ized study of Gabapentin, Lamotrigine, and Carbamazepine. Neurology TCA: tricyclic antidepressants
2005;64:1868–73. TGB: tiagabine
68. Gilad R, Sadeh M, Rapaport A, et al. Monotherapy of lamotrigine versus TPM: topiramate
carbamacepine in patients with poststroke seizure. Clin Neuropharmacol VPA: valproic acid
2007;30:189–95. ZNS: zonisamide

You might also like