Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jun 1.
Published in final edited form as: Epilepsy Behav. 2022 Apr 25;131(Pt A):108693. doi: 10.1016/j.yebeh.2022.108693

Movement disorders associated with antiseizure medications: a systematic review

Daniel J Zhou 1, Spriha Pavuluri 1, Isha Snehal 1, Cynthia M Schmidt 2, Miguel Situ-Kcomt 1, Olga Taraschenko 1,*
PMCID: PMC9596228  NIHMSID: NIHMS1796042  PMID: 35483204

Abstract

New onset movement disorders have been frequently reported in association with the use of antiseizure medications (ASMs). The frequency of specific motor manifestations and the spectrum of their semiology for various ASMs have not been well characterized. We carried out a systematic review of literature and conducted a search on CINAHL, Cochrane Library, EMBASE, MEDLINE, PsycINFO, and Scopus from inception to April 2021. We compiled the data for all currently available ASMs using the conventional terminology of movement disorders. Among 5123 manuscripts identified by the search, 437 met the inclusion criteria. The largest number of reports of abnormal movements were in association with phenobarbital, valproic acid, lacosamide, and perampanel, and predominantly included tremor and ataxia. The majority of attempted interventions for all agents were discontinuation of the offending drug or dose reduction which led to the resolution of symptoms in most patients. Familiarity with the movement disorder phenomenology previously encountered in relation with specific ASMs facilitates early recognition of adverse effects and timely institution of targeted interventions.

Keywords: Antiseizure medications, movement disorder, drug-induced, adverse effects

1. Introduction

Antiseizure medications (ASMs) are commonly prescribed for seizure prophylaxis or management, and for other indications, such as bipolar disorder, anxiety, and chronic pain [1]. Both the therapeutic effectiveness and the adverse effects of the ASMs are attributed to their ability to cross the blood brain barrier and act on the targets within the central nervous system (CNS). CNS adverse effects can emerge as cognitive slowing, psychiatric disturbances, or impairment of motor controls [2]. New onset movement disorders are frequently reported in association with the use of ASMs and can manifest with symptoms of vestibular, ocular, and motor dysfunction or global impairment of motor control [3]. Recognition of these symptoms would allow for the implementation of timely interventions aimed at reducing medication toxicities, improving patient compliance, and preventing permanent neurological disability.

The development of adverse effects of ASMs may be defined by drug-specific factors (e.g., mechanism of action and pharmacokinetic profile) or clinical factors (e.g., demographic characteristics and seizure syndrome) [4,5]. Most CNS-related adverse effects, including abnormal movements, are thought to be dose-dependent and triggered by an overall decrease in cerebral neuronal activity rather than by the specific mechanisms of action of these drugs [6]. For example, diplopia and nystagmus frequently encountered during the use of sodium channel blockers for seizure prophylaxis were thought to be due to inhibition of high-frequency action potential firing in vestibular and oculomotor circuits [7]. Concomitant administration of ASMs with other centrally acting drugs may also exacerbate their neurological adverse effects. For example, myoclonus was observed in patients receiving a combination of escitalopram and lamotrigine [8]. The myoclonus was thought to be caused by additive effects of the drugs on serotonin 5-HT1A receptors or by synergetic inhibition of voltage-gated calcium channels [9]. Co-administration of the ASMs with medications that inhibit their metabolism may lead to increased serum ASM levels resulting in more severe manifestations of the CNS toxicity [10].

The objective of this review is to provide an up-to-date, comprehensive summary of the existing literature on the development of new-onset abnormal movements triggered by the administration of ASMs. In the present review, we categorized patients’ symptoms according to the conventional movement disorder terminology and recorded relevant clinical and demographic characteristics of patients presenting with specific movement disorders.

2. Materials and Methods

The systematic review protocol was developed using guidance from the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement [11] and registered in the International Prospective Register of Systematic Reviews (PROSPERO; Center for Reviews and Dissemination number CRD42021242850) [12].

2.1. Search strategy

Literature searches were carried out between April 9 and 11, 2021. EMBASE (embase.com, version including 1974-present), MEDLINE (EBSCOhost), CINAHL (EBSCOhost), PsycINFO (EBSCOHost), Scopus, and The Cochrane Library (including the Cochrane Database of Systematic Reviews and The Cochrane Central Register of Controlled Trials, wiley.com) were searched from inception to the search date.

The list of drugs included in the search and movement disorder keywords were developed by a librarian (C.S.) in consultation with neurologists with subspecialty training in Epilepsy (O.T.) and Movement Disorders (M.S.K.). The ASMs included in the search were first and second generation agents (phenobarbital, phenytoin, primidone, valproate, carbamazepine, and ethosuximide) as well as new generation medications (brivaracetam, cannabidiol, cenobamate, clobazam, clonazepam, diazepam, eslicarbazepine, felbamate, fenfluramine, gabapentin, lacosamide, lamotrigine, levetiracetam, lorazepam, midazolam, oxcarbazepine, perampanel, pregabalin, rufinamide, stiripentol, tiagabine, topiramate, vigabatrin, and zonisamide) [13,14].

Movement disorder phenomenology were described according with the Movement Disorders Society classification [15]. Specifically, tremor (excluding parkinsonism), myoclonus (positive and negative), dystonia (including blepharospasm and spasmodic dysphonia), tics, akathisia, athetosis, ballism, chorea, choreoathetosis, restless leg syndrome (RLS), periodic limb movement disorder (PLMD), eyelid myokymia, and nonspecific dyskinesias were characterized as hyperkinetic movement disorders [15]. Parkinsonism was defined as a hypokinetic movement disorder [15]. Ataxia was a distinct category defined as incoordination of voluntary muscle movement [15]. Nystagmus, oculogyric crisis, opsoclonus, opsoclonus-myoclonus, and ophthalmoplegia were defined as eye movement disorders [16].

Full search strategies are available through UNMC’s digital repository at https://digitalcommons.unmc.edu/search/9. Several types of searches were performed: 1) All databases were searched for articles with a term for the ASM medication class, an ASM family, or one of the included drug’s English generic names in close proximity to one of a list of words for the “induced” concept that was itself in close proximity to a word for the “movement disorder” concept. 2) Databases with heading/subheading systems (EMBASE, MEDLINE, CINAHL, and The Cochrane Library) were searched for articles with a) an ASM heading in combination with an adverse effect-related subheading and b) a movement disorder heading in combination with an etiology-related subheadings. Because of the extensive indexing of EMBASE records, EMBASE searches required that the ASM-related and movement disorder-related headings be major headings. 3) Triple-indexed records in EMBASE were searched for articles indexed with an anti-epileptic drug heading linked to an “adverse drug reactions” subheading, which in turn was linked to a movement disorder-related term. Major heading designation was not required in these searches.

Because no funds were available for translation, search results were limited to English-language articles. Conference abstracts were removed when database filters allowed. Filters in CINAHL and PsycINFO were used to limit results to academic journal articles. Records indexed as concerning animals that were not also indexed as concerning humans were removed from the EMBASE search. Because the lengthy searches taxed the abilities of the other databases’ search engines, no attempt to remove animal studies was made in these databases.

The database searches retrieved 7469 total records (284 from CINAHL, 246 from the Cochrane Library, 4460 from EMBASE, 1190 from MEDLINE, 326 from PsycINFO, and 963 from Scopus). All search results were imported into RefWorks. Of these records, 2346 duplicates were removed using RefWorks’ duplicate detection tool, and 1804 were categorized by RefWorks as review articles, animal studies, and conference abstracts, to be excluded. Consequently, 3319 records for unique publications remained for title/abstract review. Two independent reviewers (D.Z. and S.P.) reviewed the article titles and abstracts and then the selected articles (D.Z. and I.S.). Disagreements between reviewers and inquiries by reviewers were resolved by another reviewer.

2.2. Eligibility criteria

The title, abstract, or full text articles that contained a mention of a new onset movement disorder and ASM were reviewed (Fig. 1). Reports that were not in English, not peer-reviewed (other than letters to the editor), or those that contained no original data were excluded. Reports of exacerbation of a previous movement disorder during the use of the ASM and articles on movements consistent with seizures or those in the settings of severe metabolic derangements known to cause abnormal movements were excluded. Manifestations of abnormal movements reported to be caused by a combination of ASMs were recorded separately. Reports of movement disorders arising in the settings of co-administration of ASM with other agents in which the precipitating factor for abnormal movements was not clear were excluded.

Figure 1.

Figure 1.

Flow diagram of study selection process.

2.3. Data extraction

We extracted the numbers of patients presenting with various movement disorders for all ASMs. If available, we recorded patients’ age, sex, clinical indication of ASM, maximum daily dose and route of administration of ASM, duration of drug use prior to the onset of movement disorder, and serum ASM levels. We also extracted the data on interventions aimed to alleviate the abnormal movements as well as outcomes of these interventions. We categorized duration of ASM prior to the onset of adverse effects as acute (<3 days), subacute (3 days to 3 weeks), and chronic (>3 weeks).

2.4. Quality of evidence

Case reports and case series were evaluated based on completeness and quality of reporting and were excluded if they did not provide the pertinent information [17]. In the large studies, the risk of bias was assessed based on Cochrane Risk of Bias tools for randomized and non-randomized studies [18,19].

2.5. Data synthesis

The information from all reports was consolidated and total counts of movement disorders was recorded for each ASM. Cumulative incidences of movement disorders were calculated, when possible. Mean age for each group was determined through weighted means, when possible. The doses, routes of administration, interventions, and outcomes were consolidated. Comparisons of results between ASMs were descriptive only and not statistically assessed.

3. Results

Our search yielded 5123 manuscripts (duplicates removed), of which 437 met eligibility criteria with publication dates from 1973 to 2021 yielding 4481 patients with new-onset abnormal movements associated with ASMs (Fig. 1). The overall distribution of movement disorders with various phenomenology is summarized in Table 1 while the details of demographic characteristics or treatment regimens and clinical outcomes are reported separately (Table 2).

Table 1.

Antiseizure medications and movement disorder phenomenology.

ASM Number of patients Movement disorders (number of patients)
 Brivaracetam 26 Tremors (9), ataxia (17)
 Carbamazepine 147 Akathisia (3), choreoathetosis (1), dystonia (17), myoclonus (3/+, 6/−), RLS (1), tics (8), tremors (55), other dyskinesia (2), parkinsonism (2), ataxia (48), nystagmus (28), oculogyric crisis (5), ophthalmoplegia (2)
 Cenobamate 13 Ataxia (13)
 Clobazam 18 Dystonia (1), tics (1), tremors (3), parkinsonism (1), ataxia (12)
 Clonazepam 32 Tics (1), ataxia (31)
 Diazepam 12 Tremors (3), other dyskinesia (1), parkinsonism (4), ataxia (3), ophthalmoplegia (1)
 Eslicarbazepine 220 Myoclonus (1/+), tremors (17), ataxia (202)
 Ethosuximide 5 Akathisia (2), chorea (1), other dyskinesia (2), ataxia (2)
 Felbamate 4 Akathisia (1), dystonia (2), tics (1), ataxia (1), nystagmus (1)
 Fosphenytoin 36 Tremors (5), other dyskinesia (2), ataxia (8), nystagmus (30)
 Gabapentin 98 Ballism (1), chorea (4), choreoathetosis (5), dystonia (3), myoclonus (32/+, 20/−), tics (1), tremors (5), other dyskinesia (7), parkinsonism (1), ataxia (32), nystagmus (1), oculogyric crisis (1)
 Lacosamide 452 Myoclonus (5/+), tics (1), tremors (303), other dyskinesia (3), ataxia (140), nystagmus (86)
 Lamotrigine 130 Ballism (1), chorea (2), choreoathetosis (1), dystonia (7), myoclonus (8/+), tics (14), tremors (66), other dyskinesia (1), parkinsonism (3), ataxia (27), nystagmus (5), oculogyric crisis (4), opsoclonus (1)
 Levetiracetam 105 Choreoathetosis (1), tics (2), tremors (71), other dyskinesia (6), parkinsonism (2), ataxia (23)
 Lorazepam 3 Myoclonus (2/+), other dyskinesia (1)
 Midazolam 7 Akathisia (1), athetosis (1), dystonia (1), myoclonus (1/+), RLS (1), tremors (1), other dyskinesia (2), parkinsonism (1)
 Oxcarbazepine 27 Myoclonus (6/+), tremors (8), other dyskinesia (1), parkinsonism (1), ataxia (7), nystagmus (2), oculogyric crisis (1)
 Perampanel 264 Myoclonus (1/−), tremors (10), ataxia (253), oculogyric crisis (1)
 Phenobarbital 1623 Dystonia (3), myoclonus (2/−), tics (6), other dyskinesia (3), ataxia (1614), nystagmus (2), oculogyric crisis (1)
 Phenytoin 209 Athetosis (4), ballism (2), chorea (16), choreoathetosis (25), dystonia (18), myoclonus (2/+, 31/−), tics (1), tremors (16), other dyskinesia (30), parkinsonism (2), ataxia (103), nystagmus (55), ophthalmoplegia (3), opsoclonus-myoclonus (1)
 Pregabalin 183 Akathisia (8), dystonia (3), myoclonus (10/+, 14/−), tremors (18), other dyskinesia (5), parkinsonism (7), ataxia (122), nystagmus (2), ophthalmoplegia (1)
 Primidone 2 Ataxia (2)
 Rufinamide 6 Tremors (1), ataxia (5)
 Stiripentol 44 Tremors (15), ataxia (33)
 Tiagabine 4 Athetosis (1), dystonia (3)
 Topiramate 37 Dystonia (1), eyelid myokymia (9), myoclonus (6/+), RLS (4), PLMD (1), tremors (6), ataxia (11)
 Valproate 623 Akathisia (13), chorea (7), choreoathetosis (1), dystonia (12), myoclonus (2/+, 7/−), tremors (513), other dyskinesia (1), parkinsonism (60), ataxia (52), nystagmus (11)
 Vigabatrin 34 Athetosis (1), chorea (2), choreoathetosis (3), dystonia (2), myoclonus (2/+), tremors (2), other dyskinesia (22), parkinsonism (1), ataxia (4)
Zonisamide 37 RLS (3), tremors (12), parkinsonism (1), ataxia (33)

/+, positive myoclonus. /−, negative myoclonus.

Table 2.

Patient demographics, clinical course, and management outcomes.

Mean daily dose Route of delivery Serum level Monitoring Latency to symptom onset Action taken Outcome Time to improved outcome (days)
Antiseizure medication Cases (n) Mean age (years) % Female mg mg/kg Oral (n) IV (n) Serum levels (n, % normal) <3d 3d-21d >21d D/C ↓ dose No Δ ++ + <3d 3d-21d >21d
Carbamazepine 41 32.0 46% 855 11.2 41 30 (63%) 12 8 21 25 12 4 38 2 1 21 7 6
Clonazepam 1 37.0 0% 8.0 1 1 1 1 1
Diazepam 2 70.0 0% 6.0 2 1 1 2 2 2
Ethosuximide 3 8.7 33% 483 3 3 3 2 1 2 1
Felbamate 3 19.1 0% 1800 41.0 3 2 (50%) 2 2 1 3 1 2
Fosphenytoin 3 17.6 67% 536 3 1 (100%) 2 1 3 3 3
Gabapentin 32 56.8 53% 1120 32 4 (75%) 12 7 9 29 3 32 19 7 4
Lacosamide 3 9.7 67% 9.0 3 1 1 1 1 2 3 2 1
Lamotrigine 41 26.8 56% 254 12.9 41 11 (9%) 9 5 24 23 17 1 35 5 1 11 3 13
Levetiracetam 3 40.7 33% 917 2 1 2 1 2 1 1 2 1 1
Lorazepam 3 20.0 100% 9.0 0.25 1 2 2 1 3 2 1 2 1
Midazolam 7 42.3 43% 5.8 0.15 2* 5 5 1 7 7 6 1
Oxcarbazepine 5 27.2 60% 1050 30.0 5 2 2 1 4 1 5 2 2 1
Phenobarbital 10 10.6 20% 97.4 12.5 9 1 4 (75%) 2 5 2 10 5 4 1 1 4 2
Phenytoin 89 30.2 40% 395 14.5 70 19 80 (36%) 20 19 48 63 17 9 71 16 2 32 31 15
Pregabalin 17 61.3 65% 180 17 9 2 5 17 15 2 5 5 5
Tiagabine 4 21.3 50% 27.5 4 4 1 3 4 2 1
Topiramate 13 35.3 77% 140 13 5 4 4 10 2 1 12 1 7 3 2
Valproate 35 47.4 49% 1223 17.9 34 1 21 (90%) 2 7 25 26 4 5 27 7 1 4 10 15
Vigabatrin 4 4.4 25% 2000 150 2 2 2 1 1 3 1 4 1 2
Zonisamide 2 38.5 100% 300 2 1 (100%) 2 1 1 1 1 1
*

Subcutaneous route.

Intranasal route.

IV, intravenous. <3d, acute (<3 days). 3d-21d, subacute (3 to 21 days). >21d, chronic (>21 days). D/C, discontinued. ↓ dose, decreased dose. No Δ, no change. ++, resolved. +, improved. −, no change.

3.1. Phenobarbital and primidone

Phenobarbital was encountered in the largest number of reports of new onset abnormal movements, with a total of 1623 patients described in 10 papers (Table 1) [2029]. The most frequent manifestation was ataxia (1614 cases, cumulative prevalence of 19.6%) [2729]. Other adverse effects included tics (6 cases), dystonia (3), negative myoclonus (2), oculogyric crisis (1), and other dyskinesia (3).

Among all studies, there were 6 reports of 10 patients for whom further details were available (Table 2) [2025]. The average cumulative daily dose of phenobarbital was 97.4 mg (or 12.5 mg/kg). Serum levels of phenobarbital were obtained in 4 patients and were above therapeutic range in one patient. Phenobarbital was discontinued in all patients which resulted in resolution of symptoms in half of the patients and improvement of movements in 40% of patients. We also identified 2 case reports of ataxia induced by primidone [29].

3.2. Phenytoin and fosphenytoin

Phenytoin was described in association with various acute-onset abnormal movements in 80 reports of 209 patients (Table 1) [26,29107]. Ataxia was the most frequently reported symptom and was described in 103 patients (13.5%). Various hyperkinetic disorders also included myoclonus (33 cases), athetosis (4), chorea (16), choreoathetosis (25), ballism (2), dystonia (18), tremors (16), and other unspecified dyskinesias (30). There were 55 cases of nystagmus, of which four and five presentations were characterized as downbeat [33,36] and horizontal nystagmus [37,45,57,68,75], respectively. Other less commonly encountered complications included tics (1), parkinsonism (2), ophthalmoplegia (3), and opsoclonus-myoclonus (1).

Among all studies, additional information was available in 67 reports of 89 patients (Table 2) [3094,100,107]. These patients with new onset abnormal movements received an average 395 mg (or 14.5 mg/kg) of phenytoin daily prior to the development of adverse effects. Of 80 patients who had serum phenytoin levels recorded, 51 patients (63.8%) were noted to have their levels exceeding the therapeutic range. The majority of patients had a sustained exposure to phenytoin prior to the occurrence of adverse effects. Specifically, among 87 patients for whom the duration of treatment was recorded, 48 patients (55.2%) received phenytoin for over 3 weeks. On the other hand, only 20 patients (23.0%) developed symptoms within 3 days from the initial exposure to phenytoin. The most frequent management approach undertaken to combat these adverse effects was discontinuation of phenytoin (63, 70.8%). This was followed by dose reduction in 17 patients (21.3%). In one case of acute focal dystonia associated with phenytoin, the patient was treated with benztropine [44]. The dystonia resolved, and the patient was continued on the same dose of phenytoin.

Fosphenytoin was reported to induce abnormal movements in 36 patients (Table 1) [108111]. In a clinical trial of 60 patients who received intramuscular fosphenytoin, 28 patients developed nystagmus, while 8 and 5 manifested ataxia and tremor, respectively [111]. Three case reports described the presence of dystonia and two reports described nystagmus, one of which was characterized as peduncular; the symptoms resolved following discontinuation of the drug [108110].

3.3. Valproate

We identified 78 studies of 623 patients who received valproate (in various preparations such as valproic acid, sodium valproate, and divalproex sodium) and developed new onset abnormal movements (Table 1) [27,95,9799,101103,112181]. The most frequent movement disorders were tremor (513, 11.6%), parkinsonism (60, 6.1%), and ataxia (52, 9.9%). Other manifestations of hyperkinetic disorders included akathisia, chorea, choreoathetosis, dystonia, myoclonus, and other dyskinesias in 43 patients. In a large prospective cohort study, the odds of developing parkinsonism in patients taking valproate were 5 times higher than in those taking all other ASMs, including carbamazepine, phenytoin, topiramate, clobazam, and lamotrigine [103].

We extracted further details from a subset of these studies (27 case reports of 35 patients) (Table 2) [112138]. The mean cumulative daily dose of valproate in these patients was 1223 mg, with 34 cases of it administered orally and one administered intravenously (IV). Serum levels were available in 21 patients (60%) among whom only 2 patients (9.5%) had levels above the therapeutic range. More than two thirds of patients (73.5%) developed adverse effects following prolonged exposure to valproate for 3 or more weeks while 2 patients (5.9%) manifested the symptoms acutely, within 3 days from initiation of the drug. The interventions included discontinuation and dose reduction of valproate in 74.2% and 11.4% of patients, respectively. Of the patients who had an improvement with these interventions, the latency to improvement was at least 3 weeks in more than half of the patients (51.7%). Additionally, 7 patients with valproate-induced parkinsonism and one with chorea were treated with levodopa [123,126,130] and benzodiazepines and haloperidol [124], respectively.

3.4. Carbamazepine

We identified 43 reports of 147 patients with carbamazepine-induced abnormal movements, which included tremor (55, 12.2%), ataxia (48, 3.7%), nystagmus (28), and dystonia (17, 5.8%) (Table 1) [26,95,103,141,157,178,181217]. In one pediatric study, 19 children (24.7%) developed nystagmus in the settings of accidental or intentional ingestion of carbamazepine [182].

Further details of clinical characteristics were available in 29 reports of 41 of the patients (Table 2) [189217]. Specifically, these patients developed akathisia (3 cases), choreoathetosis (1), blepharospasm (1), other dystonia (7), myoclonus (8), restless leg syndrome (1), orofacial dyskinesia (2), parkinsonism (2), nystagmus (9), oculogyric crisis (5), and ophthalmoplegia (2). The nystagmus was characterized as downbeat, gaze-evoked, and horizontal with direction-changing or torsional components in 3 patients in each category [190,196,197,206,212,213,216]. Patients treated with carbamazepine received a mean cumulative daily dose of 855 mg (or 11.2 mg/kg) prior to onset of the abnormal movements. Of the 30 patients for whom the serum levels were reported, 11 patients (36.7%) had levels above therapeutic range. The symptoms manifested after chronic use (i.e., >21 days) in over half of patients (51.2%). The complications developed acutely in 29.3% of these patients and subacutely in 19.5% of patients. To remediate the adverse effects, carbamazepine was discontinued in 61% of patients and its dose was reduced in 29.3% of patients. This resulted in resolution of symptoms in most patients (92.7%). Carbamazepine-induced dystonia described in 4 cases was additionally treated with promethazine, diazepam, trihexyphenidyl, or gastric lavage [192,195,198,213].

3.5. Benzodiazepines

Clonazepam was reported to cause ataxia (31) and Tourette syndrome (1) (Table 1) [165,218,219]. Diazepam was reported to be associated with abnormal movements in 12 patients, including parkinsonism (4, 14.8%), ataxia (3, 2.3%), tremor (3, 2.3%), divergence paralysis (1), and oral-buccal dyskinesia (1) (Table 1) [220223]. There were 7 case reports of midazolam-induced akathisia, athetosis, dystonia, myoclonus, RLS, tremor, parkinsonism, and orofacial or lingual dyskinesias, all of which resolved following midazolam discontinuation (Table 2) [224230]. Lorazepam was reported to be associated with myoclonus (2) and orofacial dyskinesias (1) (Table 1) [231,232].

3.6. Clobazam and ethosuximide

There were five reports of 18 patients with clobazam-induced ataxia (12), tremors (3), dystonia (1), tics (1), and parkinsonism (1) (Table 1) [95,103,155,233,234]. Ethosuximide was reported in association with ataxia (2), akathisia along with orofacial dyskinesias (2), or generalized chorea (1) (Table 1) [29,235,236].

3.7. Lacosamide

We identified 26 reports of 452 patients with new-onset abnormal movements associated with the use of lacosamide (Table 1) [186,237261]. The most common presentations were tremor (303, 7.1%) and ataxia (140, 6.3%). There were 86 reported cases of nystagmus, 52 (60.5%) of which were also included in cases of ataxia. Other hyperkinetic disorders included tics (1), positive myoclonus (5), and unspecified dyskinesias (3). Further, lacosamide triggered orofacial and limb dyskinesia in 3 patients [237].

3.8. Perampanel

We identified 19 studies of 264 patients with abnormal movements triggered by perampanel (Table 1) [262280]. The most frequently reported manifestation was ataxia (253, 4.8%). Other movement disorders included tremor (10), negative myoclonus (1), and oculogyric crisis (1).

3.9. Gabapentin and pregabalin

We identified 36 studies of 98 patients with gabapentin-associated abnormal movements, including myoclonus (52, 12.5%) and ataxia (32, 6.7%) (Table 1) [98,103,281314]. Among these studies, further details were available for 32 patients (Table 2) [289313]. Specifically, 12 patients developed positive myoclonus, of which 7 had elevated serum creatinine and blood urea nitrogen (BUN) levels. Three other patients had negative myoclonus. Other manifestations included ballism (1 case), chorea (4), choreoathetosis (5), dystonia (3), tics (1), tremors (5), oculogyric crisis (1), parkinsonism, and other dyskinesias (7). The mean cumulative daily dose of gabapentin was 1120 mg. Nearly 40% of patients for whom the onset of symptoms was documented developed adverse effects within 3 days from the initiation of treatment while the remaining patients had subacute (25%) or delayed (32.1%) onset of these complications. The gabapentin dose was reduced in 3 patients while drug was discontinued in all other cases. In addition, some patients with myoclonus underwent dialysis [295,315,316] or received haloperidol, diphenhydramine, lorazepam, or procyclidine [289,290,304,305]. In all instances, the interventions resulted in resolution of symptoms. The recovery was achieved in nearly two-thirds of the patients (63.3%) within 3 days after the intervention and was delayed in the remaining patients.

There were 32 reports of 183 patients with abnormal movements associated with pregabalin, mostly including ataxia (122, 7.6%), myoclonus (24, 2.1%), and tremor (18, 10.6%) (Table 1) [282,283,317346]. These included case reports of 17 patients with mean daily dose of pregabalin of 180 mg (Table 2) [330346]. In these studies, myoclonus developed in 8 patients, of whom 4 had elevated serum creatinine and BUN while downbeat nystagmus emerged in 2 patients [331,334]. Other manifestations included ataxia (5), akathisia (1), blepharospasm (1), and parkinsonism (4). Motor symptoms developed acutely in more than a half of the patients (56.3%). All patients discontinued pregabalin which led to either improvement or complete resolution of symptoms. Abnormal movements improved within 3 days in 33% of patients.

3.10. Oxcarbazepine and eslicarbazepine

We identified 10 studies of 27 patients with abnormal movements triggered by oxcarbazepine, most commonly tremors (8, 0.8%), ataxia (7, 2.1%), and positive myoclonus (6, 0.7%) (Table 1) [95,347355]. They also included two incidences of jerky see-saw and multidirectional nystagmus which resolved after carbamazepine was discontinued [351,355]. Other rare manifestations included tardive dyskinesia (1), parkinsonism (1) and oculogyric crisis (1) [352354]. We also identified 11 reports of 220 patients with ataxia (202, 3.2%), tremor (17, 2.1%), and positive myoclonus (1, 5.6%) following administration of eslicarbazepine (Table 1) [184,356365].

3.11. Lamotrigine

Lamotrigine-induced abnormal movements were encountered in 38 studies reporting 130 patients whose manifestations included tremor (66, 12.1%), ataxia (27, 11.2%), tics (14), and myoclonus (8) (Table 1) [95,101,103,105,155,173,178,181,366395]. Additional data were available in 41 of these patients (Table 2) [366390]. Specifically, 14 patients developed tics, 10 patients had abnormal eye movements, and 7 suffered from various forms dystonia. Five of these patients developed nystagmus with downbeat (3) or horizontal gaze-evoked components (1) [367,374,379,380]. The mean cumulative daily dose of lamotrigine in this subset of patients was 254 mg (or 12.9 mg/kg). Serum levels were obtained in 11 patients and were above therapeutic range in 10 patients (90.9%). The movement disorders developed acutely in 9 patients (23.7%), subacutely in 5 patients (13.2%), and after chronic use in 23 patients (60.5%). To treat these complications, lamotrigine was discontinued in over half of the patients (56.1%), and its dose was reduced in 41.4% of patients. These measures lead to resolution or significant improvement of symptoms in the majority of instances (97.6%). One patient with simultaneous manifestations of blepharospasm, complex tics, and obsessive-compulsive disorder was also treated with risperidone [368]. There was variability in times to resolution of symptoms; 40.7% of patients have recovered within 3 days after the intervention and 48.1% of patients still experienced symptoms after 3 weeks.

3.12. Levetiracetam and brivaracetam

There were 13 reports of 105 patients with new onset movements triggered by administration of levetiracetam (Table 1) [95,155,157,173,178,183,187,392,396400]. These manifestations included tremor (71, 8.2%) and ataxia (23, 20.2%). Other symptom included parkinsonism (2), which improved after adjustment of the dose or discontinuation of the drug [398,400]. Brivaracetam was found to be associated with tremor (9, 2.7%) or ataxia (17, 2.0%) in large observational studies (Table 1) [401405].

3.13. Topiramate, zonisamide, and stiripentol

We identified 16 studies of 37 patients with new-onset abnormal movements induced by topiramate (Table 1) [103,173,406419]. These manifestations included ataxia (11, 8.5%), eyelid myokymia (9, 5.7%), tremor (6, 3.0%), myoclonus (6), and RLS (4). Among these, in 13 patients (Table 2) [406414], topiramate was administered at a mean dose of 140 mg daily for the prophylaxis of seizures or migraine headache as well as treatment of eating disorders. Symptoms developed acutely, subacutely, or were delayed in similar proportions of patients. To treat these adverse effects, topiramate was discontinued in 76.9% of patients, and its dose was reduced in 15.4% of patients. One patient with multifocal myoclonus was additionally treated with cyproheptadine [406], and another patient with new onset restless leg syndrome was administered cabergoline [411]. The abnormal movements resolved in 12 patients (92.3%) and significantly improved in the other patient (7.7%). Over half of the patients achieved recovery within 3 days from interventions.

Zonisamide was associated with 37 cases of abnormal movements, which primarily included ataxia (33, 4.4%) and tremor (12, 3.0%), RLS (3, 2.5%), or parkinsonism (1) (Table 1) [103,420427]. In two case reports, RLS resolved in one of the patients following drug cessation and remained unchanged in the other patient despite dose decrease [426,427]. Among 44 patients treated with stiripentol, 33 (29.8%) developed ataxia and 12 (12.1%) developed tremor (Table 1) [428432].

3.14. Vigabatrin and tiagabine

There were 9 reports of 34 patients with vigabatrin-induced abnormal movements, most of which were hyperkinetic and manifested as athetosis (1), chorea (2), choreoathetosis (3), dystonia (2), myoclonus (1), tremors (2), or other unspecified dyskinesias (22) (Table 1) [103,433440]. Some patients improved after dose reduction or discontinuation of vigabatrin (Table 2).

Tiagabine was found to be associated with athetosis in 1 patient and dystonia in 3 patients (Table 1) [441,442]. In all cases, the movement disorders resolved following discontinuation or dose reduction of the tiagabine (Table 2).

3.14.1. Other antiseizure medications

In a randomized control trial, cenobamate was found to be associated with ataxia in 13 patients (4.0%) (Table 1) [443]. Rufinamide was associated with ataxia in 5 patients (5.6%) [444,445] and tremor in 1 patient (0.6%) (Table 1) [446]. Felbamate was associated with tics in 1 patient (2.6%) [447] while ataxia with downbeat nystagmus was reported in another patient with felbamate intoxication [448]; dystonia was reported in 2 patients (Table 1) [449].

3.15. Movement disorders associated with combinations of ASMs

We identified 16 reports of 80 patients with new onset abnormal movements that developed during the administration of ASM combinations [98,168,185,212,379,401,450459]. The common combination was valproate and lamotrigine, representing 46 patients with manifestations of abnormal movements which included tremor (42, 20.5%), ataxia (5), opsoclonus (2), nystagmus (2), and tics (2) [168,185,379,454,457,459].

Four other case reports described 8 patients with uncommon manifestations of abnormal movements that developed in patients treated with various ASM combinations. In one patient, the combination of IV phenytoin and diazepam administration for seizures triggered an acute reversible opsoclonus, ataxia, and dysarthria [450]. Primidone administered with phenobarbital over 8 years resulted in periodic alternating nystagmus in one patient [451]. Intentional ingestion of toxic doses of carbamazepine and phenytoin triggered choreoathetosis, dystonia, and nystagmus in another patient, and the symptoms were successfully treated with induced vomiting and administration of magnesium sulfate-charcoal [212]. Combined lamotrigine and phenytoin induced chorea in 3 patients [453].

Other larger studies included various combinations of phenytoin, valproate, carbamazepine, gabapentin, lacosamide, levetiracetam, brivaracetam, and tiagabine causing ataxia, tremor, or asterixis [98,401,455,458]. Furthermore, while not matched to specific ASMs and therefore not included in our counts of abnormal movements, in a large single-center study of 201 patients, Zadikoff et al. described 89 patients (44.3%) who developed postural or action tremor [103].

4. Discussion

In the present systematic review of literature, we compiled the available data on association of currently used ASMs with new onset movement disorders of various phenomenology. The spectrum of manifestations of abnormal movements was broad, and the latency to onset of symptoms has varied from a few hours to several days. The majority of adverse effects were reversible and resolved following the discontinuation or dose reduction of ASMs.

A previous review of the abnormal motor manifestations associated with 13 different ASMs was compiled more than 15 years ago [3]. The categorization of the abnormal movements used in the previous systematic review was similar to the system employed in our study. However, we used a more granular approach and applied additional and more specific movement disorder subcategories, such as restless legs syndrome and eyelid myokymia, to highlight the entire spectrum of motor manifestations of the ASM toxicity. We also explored ASM dosing, identified toxic levels, and timing of onset and resolution based on duration of ASM use and timing after ASM dose changes. In the previous review, the authors found that phenytoin and carbamazepine were frequently reported in association with ataxia and dyskinesias while valproate was associated with tremor and parkinsonism [3]. Our present findings are largely in agreement with those reported in the previous review. In the present review, we also included 18 additional ASMs and reviewed 311 new relevant reports that have appeared in the literature since the previously published review. To our knowledge there were no other systematic reviews of the literature on this topic.

Understanding of pathophysiological mechanisms underlying the development of new abnormal movements can help to anticipate their emergence. However, the knowledge of these mechanisms is currently lacking, and the available data are largely restricted to the animal studies. Given the higher propensity of specific movement disorders to appear following administration of certain ASMs, such as parkinsonism with valproate or tics with lamotrigine, it is likely that the development of adverse effects is related to the specific mechanisms of action for each ASM. Likewise, while the same movement disorder may appear during treatment with various ASMs, such as myoclonus with gabapentin or phenytoin, the pathophysiology of these manifestations is likely distinct for different drugs. Since the majority of symptoms have resolved after adjustment of the medication doses, these presentations appeared to be most consistent with a type A category of adverse effects, which are dose-dependent and related to the pharmacologic properties of the drug [460]. However, in some cases where the movement disorders persistent despite the adjustment of the ASM dose, a type B (idiosyncratic) adverse effects could be present. In many patients the relevant data were missing; therefore, an accurate categorization of adverse effects was not feasible.

Phenobarbital and phenytoin were among the oldest ASMs that were notorious for causing ataxia. It is proposed that ataxia in phenobarbital and phenytoin use is triggered by folate depletion [165]. Chronic phenytoin use is also known to be associated with reduced cerebellar volume, that can further contribute to worsening of ataxia [96,461]. Despite the high number of reports of phenytoin-induced dyskinesia, the pathophysiology of this manifestation remains poorly understood. In preclinical studies, a two-week long exposure to phenytoin in rats induced a higher number of stereotypies following a challenge with apomorphine, a dopamine receptor agonist [462].

Valproate was frequently reported in association with tremor and parkinsonism. The putative pathophysiologic mechanisms of valproate-induced parkinsonism included an enhancement of the GABAergic signaling in the basal ganglia and altered gene expression in dopamine signaling by upregulation of the expression of dopamine transporter, thus leading to increased dopamine reuptake and decreased availability in the synaptic cleft [463]. The valproate-induced unmasking or potentiation of the overt pre-existing degeneration of dopaminergic neurons in the substantia nigra was also proposed to contribute to the manifestations of parkinsonism [463]. Tremor and various other hyperkinesias, particularly choreoathetoid movements, were thought to result from the valproate-induced increase in inhibitory tone of the basal ganglia neurons and resultant disinhibition of the efferent pathways to the motor cortex [116,117,122].

Carbamazepine-induced hyperkinetic disorders are thought to be related to its effects on dopaminergic neurotransmission in the basal ganglia pathways. More specifically, carbamazepine induced a biphasic effect on extracellular dopamine release in the striatum of rats such that dopamine levels were increased following the administration of lower doses of carbamazepine and decreased in response to the toxic doses of drug [464]. Other animal studies similarly reported that administration of carbamazepine appears to enhance dopamine release in the striatum [465,466]. Carbamazepine was also noted to increase serotonin levels, measured in the hippocampus of rats [467], which could lead to manifestations of myoclonus. Ataxia and nystagmus which were often reported in patients treated with carbamazepine were thought to be caused by a direct transient effect of the drug on brainstem nuclei and cerebellum [206].

Gabapentin and pregabalin were found to be frequently associated with new onset myoclonus. The mechanism of this manifestation remains unclear [282]. One theory suggests that myoclonus emerges due to the action of these drugs on voltage-gated calcium channels in the thalamocortical pathways [283]. Another thought was that gabapentin and pregabalin could cause an increase in serotonin levels in the brain and peripheral nervous system, thereby promoting the development of myoclonus [468470]. In animal studies, the use of serotonin receptor agonists precipitated the myoclonus [471,472]. Myoclonus was frequently encountered in patients with high serum levels of gabapentin or pregabalin and poor renal function and symptoms have subsided following the dose reduction or institution of renal dialysis.

Lamotrigine was associated with tremor, ataxia, tics, dystonia, and abnormal eye movements. The pathophysiologic mechanism underlying the emergence of tics was proposed to be related to blockade of presynaptic glutamate release by lamotrigine leading to enhancement of dopaminergic transmission in the substantia nigra and striatum [383]. Similar indirect effect of lamotrigine on the dopaminergic pathways may also be associated with manifestations of tremors, oculogyric crisis, and dystonia [371,378,385]. At toxic levels, lamotrigine can cause a transient dysfunction of the vestibulo-cerebellar pathways, which may explain some presentations of ataxia and nystagmus [367,380].

Topiramate and zonisamide were found to be associated with restless leg syndrome and periodic limb movements of sleep. It was proposed that topiramate and zonisamide can modulate the neurotransmission in dopaminergic pathways indirectly by enhancing the GABAergic neurotransmission and inhibiting the glutamatergic neurotransmission in the brain [411,426,427,473]. Benzodiazepines and barbiturates that are GABAA receptor agonists are thought to trigger dyskinesia via potentiation of the inhibitory tone in the basal ganglia pathways [25,225,227]. Levetiracetam-induced parkinsonism was attributed to the indirect inhibition of dopamine release in patients with Huntington disease [398,400]. Perhaps in a similar way, dysregulation of dopaminergic signaling may also result in hyperkinetic disorders during treatment with levetiracetam [399].

We identified the reports of various forms of nystagmus in patients treated with ASMs, including downbeat nystagmus and gaze evoked nystagmus. Nystagmus can be generated by disruption of the fixation, vestibulo-ocular reflex or gaze-holding mechanisms. The directionality of nystagmus may provide clues to localization of dysfunction within the CNS. For example, gaze-evoked nystagmus involves disruption of neurotransmission within the horizontal and vertical gaze centers in the caudal pons and midbrain, respectively and was reported during exposure to exogenous GABA [474]. This form of nystagmus was found in patients exposed to phenytoin, carbamazepine, lamotrigine, and oxcarbazepine [31,42,53,57,60,74,81,196,206,212,216,355,374]. On the other hand, downbeat nystagmus is thought to occur in dysfunction of vestibulo-cerebellar pathways and caudal medulla. We found several reports describing downbeat nystagmus in association with phenytoin, carbamazepine, lamotrigine, pregabalin, and felbamate [33,36,196,212,213,331,334,367,380,448,452,475]. Periodic alternating nystagmus was found to be associated with phenobarbital and primidone and was thought to be caused by the medication effects on the cerebellar nodulus [451,475]. One patient taking oxcarbazepine was found to have jerky see-saw nystagmus, which in other disorders is associated with dysfunction of the medial longitudinal fasciculus and lesions to the midbrain, pons, or cerebellum [351]. Pendular nystagmus that developed acutely following the administration of IV fosphenytoin was thought to be resulting from blockade of sodium conductance in the brainstem neurons [109]

Our study has several limitations. Because of the limitations of the literature database search engines and the number of ASMs under consideration only drug class names, English generic names and subject headings were used to identify the drugs in the searches performed. Unindexed records utilizing proprietary names, scientific names, some of the foreign generic names, or investigational names for the ASMs would have been missed. Despite the rigorous approach taken in categorization of movement disorders, it is possible that we did not capture all manifestations of abnormal movements. While we made all attempt to recognize the description of neurological comorbidities that may have led to manifestations of abnormal movements, these conditions were not always explicitly reported. Therefore, it is possible that some patients have had additional factors independently contributing to the development of abnormal movements. Finally, in the larger studies, patients were likely taking multiple ASMs; therefore, it is possible that their adverse effects were complex and more than one ASM contributed to the toxicity.

5. Conclusions

This systematic review provides an up-to-date summary of the movement disorders reported in association of ASMs. The most frequently encountered manifestations of abnormal movements were tremor, ataxia, nystagmus, and various hyperkinetic disorders, as well as parkinsonism which were largely reversed following the removal of the offending drugs. The knowledge of the described associations and their clinical phenotypes may facilitate early diagnosis and treatment of these disorders.

Highlights.

  • Administration of antiseizure medications (ASMs) is often associated with the development of new onset movement disorders

  • Phenobarbital and valproic acid, are most frequently associated with various movement disorders, including tremor and ataxia

  • Nearly all patients with ASM-induced movement disorders improved following the cessation of drugs or their dose adjustment.

  • Familiarity with the phenomenology of motor adverse events in respect to specific ASMs facilitates their early recognition and triggers targeted interventions.

Funding Statement

O.T. received grant support from the American Epilepsy Society-NORSE Institute (Seed grant) and NIH P20GM130447 (Cognitive Neuroscience and Development of Aging (CoNDA) Award). This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Financial Disclosures:

O.T. received grant support from the American Epilepsy Society-NORSE Institute (Seed grant) and NIH P20GM130447 (Cognitive Neuroscience and Development of Aging (CoNDA) Award). All other authors report no disclosures.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Cascade E, Kalali AH, Weisler RH. Varying uses of anticonvulsant medications. Psychiatry (Edgmont) 2008;5:31–3. [PMC free article] [PubMed] [Google Scholar]
  • [2].Zaccara G, Gangemi PF, Cincotta M. Central nervous system adverse effects of new antiepileptic drugs. A meta-analysis of placebo-controlled studies. Seizure 2008;17:405–21. [DOI] [PubMed] [Google Scholar]
  • [3].Zaccara G, Cincotta M, Borgheresi A, Balestrieri F. Adverse motor effects induced by antiepileptic drugs. Epileptic Disord 2004;6:153–68. [PubMed] [Google Scholar]
  • [4].French JA, Kanner AM, Bautista J et al. Efficacy and tolerability of the new antiepileptic drugs II: Treatment of refractory epilepsy: Report of the therapeutics and technology assessment subcommittee and quality standards subcommittee of the american academy of neurology and the american epilepsy society. Neurology 2004;62:1261–73. [DOI] [PubMed] [Google Scholar]
  • [5].Panayiotopoulos CP, Benbadis SR, Covanis A et al. Efficacy and tolerability of the new antiepileptic drugs I: Treatment of new onset epilepsy: Report of the therapeutics and technology assessment subcommittee and quality standards subcommittee of the american academy of neurology and the american epilepsy society. Neurology 2005;64:172, 4; author reply 172. [PubMed] [Google Scholar]
  • [6].Sills G Mechanisms of action of antiepileptic drugs. In: Sander JW, Walker MC, Smalls JE, editors. From science to society. A practical guide to epilepsy. London, UK: ILAE (UK Chapter) and the National Society for Epilepsy; 2011. p. Chapter 25. [Google Scholar]
  • [7].Gittis AH, Moghadam SH, du Lac S. Mechanisms of sustained high firing rates in two classes of vestibular nucleus neurons: Differential contributions of resurgent na, Kv3, and BK currents. J Neurophysiol 2010;104:1625–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Rosenhagen MC, Schmidt U, Weber F, Steiger A. Combination therapy of lamotrigine and escitalopram may cause myoclonus. J Clin Psychopharmacol 2006;26:346–7. [DOI] [PubMed] [Google Scholar]
  • [9].Yitzhak S, Krivoy N. Safety and efficacy of lamotrigine in older adults with epilepsy and comorbid depressive symptoms. 2009;1. [Google Scholar]
  • [10].Perucca E Clinically relevant drug interactions with antiepileptic drugs. Br J Clin Pharmacol 2006;61:246–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Moher D, Shamseer L, Clarke M et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev 2015;4:1–4053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Zhou D, Schmidt C, Taraschenko O. Movement disorders associated with anticonvulsants: A systematic review. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].French JA, Gazzola DM. New generation antiepileptic drugs: What do they offer in terms of improved tolerability and safety? Ther Adv Drug Saf 2011;2:141–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Perucca E, Brodie MJ, Kwan P, Tomson T. 30 years of second-generation antiseizure medications: Impact and future perspectives. Lancet Neurol 2020;19:544–56. [DOI] [PubMed] [Google Scholar]
  • [15].Martino D, Epsay A, Fasano A, Morgante F. Disorders of movement: A guide to diagnosis and treatment. Berlin, Heidelberg: Springer; 2016. [Google Scholar]
  • [16].Wong A Eye movement disorders. Oxford, UK: Oxford University Press; 2008. [Google Scholar]
  • [17].Murad MH, Sultan S, Haffar S, Bazerbachi F. Methodological quality and synthesis of case series and case reports. BMJ Evid Based Med 2018;23:60–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Sterne JA, Hernán MA, Reeves BC et al. ROBINS-I: A tool for assessing risk of bias in non-randomised studies of interventions. BMJ 2016;355 :i4919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Sterne JAC, Savović J, Page MJ et al. RoB 2: A revised tool for assessing risk of bias in randomised trials. BMJ 2019;366:14898. [DOI] [PubMed] [Google Scholar]
  • [20].Burd L, Kerbeshian J, Fisher W, Gascon G. Anticonvulsant medications: An iatrogenic cause of tic disorders. Can J Psychiatry 1986;31:419–23. [DOI] [PubMed] [Google Scholar]
  • [21].Lacayo A, Mitra N. Report of a case of phenobarbital-induced dystonia. Clin Pediatr 1992;31:252. [DOI] [PubMed] [Google Scholar]
  • [22].Lightman SL. Phenobarbital dyskinesia. Postgrad Med J 1978;54:114–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Sandyk R Phenobarbital-induced tourette-like symptoms. Pediatr Neurol 1986;2:54–5. [DOI] [PubMed] [Google Scholar]
  • [24].Sechi GP, Piras MR, Rosati G et al. Phenobarbital-induced buccolingual dyskinesia in oral apraxia. Eur Neurol 1988;28:139–41. [DOI] [PubMed] [Google Scholar]
  • [25].Wiznitzer M, Younkin D. Phenobarbital-induced dyskinesia in a neurologically-impaired child. Neurology 1984;34:1600–1. [DOI] [PubMed] [Google Scholar]
  • [26].Chadwick D, Reynolds EH, Marsden CD. Anticonvulsant induced dyskinesias: A comparison with dyskinesias induced by neuroleptics. J NEUROL NEUROSURG PSYCHIATRY 1976;39:1210–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Li J, Yang D, Zhao D, Li N, Lin W. Efficacy of phenobarbital and sodium valproate in treating convulsive epilepsy in rural northeast china. Seizure 2019;71:207–13. [DOI] [PubMed] [Google Scholar]
  • [28].Si Y, Liu L, Tian L et al. A preliminary observation of the adverse effects of phenobarbital among patients with convulsive epilepsy in rural west china. Epilepsy Behav 2016;54:65–70. [DOI] [PubMed] [Google Scholar]
  • [29].Muñoz-Garcia D, Del Ser T, Bermejo F, Portera A. Truncal ataxia in chronic anticonvulsant treatment. association with drug-induced folate deficiency. J Neurol Sci 1982;55:305–11. [DOI] [PubMed] [Google Scholar]
  • [30].Acar T, Alkan G, Çaksen H et al. Phenytoin induced dystonia. Turk J Pediatr 2018;60:111–2. [DOI] [PubMed] [Google Scholar]
  • [31].Ahmad S, Laidlaw J, Houghton GW, Richens A. Involuntary movements caused by phenytoin intoxication in epileptic patients. J NEUROL NEUROSURG PSYCHIATRY 1975;38:225–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Algahtani H, Shirah B, Alqahtani AJ, Al-Malki A. Irreversible cerebellar atrophy as a complication of short-term phenytoin exposure: Clinical improvement following discontinuation of the culprit. J Epilepsy Res 2020;10:96–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Alpert JN, Downbeat nystagmus due to anticonvulsant toxicity. Ann Neurol 1978;4:471–3. [DOI] [PubMed] [Google Scholar]
  • [34].Asadi-Pooya A, Petramfar P, Taghipour M. Refractory hiccups due to phenytoin therapy. Neurol India 2011;59:68. [DOI] [PubMed] [Google Scholar]
  • [35].Barvaliya M, Sanmukhani J, Patel TK, Tripathi CB. Phenytoin induced chorea in a pediatric patient: An interaction between phenytoin, phenobarbital and clobazam. Indian J Pharmacol 2011;43:731–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Berger JR, Kovacs AG. Downbeat nystagmus with phenytoin. J CLIN NEURO-OPHTHALMOL 1982;2:209–11. [PubMed] [Google Scholar]
  • [37].Betteridge T, Fink J. Phenytoin toxicity and thyroid dysfunction. New Zealand Med J 2009;122:102–4. [PubMed] [Google Scholar]
  • [38].Buchanan N, Rosen E, Rabinowitz L. Athetosis and phenytoin toxicity. Am J Dis Child 1977;131:105. [DOI] [PubMed] [Google Scholar]
  • [39].Caksen H, Odabaş D, Anlar O. Use of biperiden hydrochloride in a child with severe dyskinesia induced by phenytoin. J Child Neurol 2003;18:494–6. [DOI] [PubMed] [Google Scholar]
  • [40].Chalhub EG, DeVivo DC. Letter: Phenytoin-induced choreoathetosis. J Pediatr 1976;89:153–4. [DOI] [PubMed] [Google Scholar]
  • [41].Chalhub EG, Devivo DC, Volpe JJ. Phenytoin induced dystonia and choreoathetosis in two retarded epileptic children. Neurology 1976;26:494–8. [DOI] [PubMed] [Google Scholar]
  • [42].Chaudhary N, Ravat SH, Shah PU. Phenytoin induced dyskinesia. Indian Pediatr 1998;35:274–6. [PubMed] [Google Scholar]
  • [43].Chi WM, Chua KSG, Kong K-. Phenytoin-induced asterixis - uncommon or under-diagnozed? Brain Inj 2000;14:847–50. [DOI] [PubMed] [Google Scholar]
  • [44].Choonara IA, Rosenbloom L. Focal dystonic reaction to phenytoin. Dev Med Child Neurol 1984;26:677–8. [DOI] [PubMed] [Google Scholar]
  • [45].Corey A, Koller W. Phenytoin-induced dystonia. Ann Neurol 1983;14:92–3. [DOI] [PubMed] [Google Scholar]
  • [46].Dasari JR, Vurumadla S, Prasad OP. A case report on phenytoin induced ataxia. Asian J Pharm Clin Res 2016;9:5–6. [Google Scholar]
  • [47].Dorado P, López-Torres E, Peñas-Lledó EM, Martínez-Antón J, Llerena A. Neurological toxicity after phenytoin infusion in a pediatric patient with epilepsy: Influence of CYP2C9, CYP2C19 and ABCB1 genetic polymorphisms. Pharmacogenomics J 2013;13:359–61. [DOI] [PubMed] [Google Scholar]
  • [48].Duarte J, Sempere AP, Cabezas MC, Marcos J, Clavería LE. Postural myoclonus induced by phenytoin. Clin Neuropharmacol 1996;19:536–8. [DOI] [PubMed] [Google Scholar]
  • [49].Ertan S, Ulu MO, Hanimoglu H et al. Phenytoin-induced parkinsonism. Singapore Med J 2006;47:981–3. [PubMed] [Google Scholar]
  • [50].Filloux F, Thompson JA. Transient chorea induced by phenytoin. J Pediatr 1987;110:639–41. [DOI] [PubMed] [Google Scholar]
  • [51].Finsterer J, Keller H, Reining-Festa A, Enzelsberger B, Weidinger F. Phenytoin-induced choreoathetosis after serial seizures due to traumatic brain injury and chronic alcoholism. Clin Case Rep 2018;6:2316–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Gill D, Lyons M, Allam F. Phenytoin induced chorea: A case report. Am J Ther 2018;25:e390. [DOI] [PubMed] [Google Scholar]
  • [53].Girija AS. Paroxysmal dyskinesia in phenytoin toxicity. J Assoc Physicians India 2002;50:1449–50. [PubMed] [Google Scholar]
  • [54].Goñi M, Jimenez M, Feijoo M. Parkinsonism induced by phenytoin. Clin Neuropharmacol 1985;8:383–4. [DOI] [PubMed] [Google Scholar]
  • [55].González Otárula KA, Ugarnes G, Rossi M, Ballesteros D, D’Giano C. Phenytoin-induced chorea: Drug interaction or genetic predisposition? Clin Neuropharmacol 2016;39:120. [DOI] [PubMed] [Google Scholar]
  • [56].Gunduz T, Kocasoy-Orhan E, Hanagasi HA. Orolingual dyskinesia and involuntary neck movements caused by phenytoin intoxication. J Neuropsychiatry Clin Neurosci 2013;25:E51. [DOI] [PubMed] [Google Scholar]
  • [57].Gupta M, Patidar Y, Khwaja GA, Chowdhury D, Batra A, Dasgupta A. Persistent cerebellar ataxia with cerebellar cognitive affective syndrome due to acute phenytoin intoxication: A case report. Neurol Asia 2013;18:107–11. [Google Scholar]
  • [58].Gupta V, Yadav TP, Yadav A. Phenytoin toxicity presenting as acute meningo-encephalitis in children. Neurol India 2011;59:66–7. [DOI] [PubMed] [Google Scholar]
  • [59].Haider Y, Abbott RJ. Phenytoin-induced choreoathetosis. Postgrad Med J 1990;66:1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Herishanu Y, Osimani A, Louzoun Z. Unidirectional gaze paretic nystagmus induced by phenytoin intoxication. Am J Ophthalmol 1982;94:122–3. [DOI] [PubMed] [Google Scholar]
  • [61].Howrie DL, Crumrine PK. Phenytoin-induced movement disorder associated with intravenous administration for status epilepticus. Clin Pediatr 1985;24:467–9. [DOI] [PubMed] [Google Scholar]
  • [62].Jenkins A A case of phenytoin toxicity in a patient with advanced lung cancer. Palliative Med 2006;20:479–80. [DOI] [PubMed] [Google Scholar]
  • [63].Juhi S, Singh TS, Singh JM, Muhammed R, Manik C, Kumar TR. Ataxia, manifestation of phenytoin toxicity: A case report. J Young Pharm 2019;11:112–3. [Google Scholar]
  • [64].Kaur U, Chakrabarti SS, Gambhir IS. Orofacial dyskinesias by phenytoin in an elderly female: The dangers of poor therapeutic monitoring. Epilepsy Behav 2016;59:155–6. [DOI] [PubMed] [Google Scholar]
  • [65].Koukkari MW, Vanefsky MA, Steinberg GK, Hahn JS. Phenytoin-related chorea in children with deep hemispheric vascular malformations. J Child Neurol 1996;11:490–1. [DOI] [PubMed] [Google Scholar]
  • [66].Krishnamoorthy KS, Zalneraitis EL, Young RSK, Bernad PG. Phenytoin-induced choreoathetosis in infancy: Case reports and a review. Pediatrics 1983;72:831–4. [PubMed] [Google Scholar]
  • [67].Kurata K, Kido H, Kobayashi K, Yamaguchi N. Long-lasting movement disorder induced by intravenous phenytoin administration for status epilepticus. A case report. Clin Neuropharmacol 1988;11:467–71. [DOI] [PubMed] [Google Scholar]
  • [68].Lazaro RP. Involuntary movements induced by anticonvulsants drugs. Mt Sinai J Med 1982;49:274–81. [PubMed] [Google Scholar]
  • [69].Lee C-, Li J-. Phenytoin intoxication and upper facial dyskinesia: An unusual presentation. Mov Disord 2008;23:1188–9. [DOI] [PubMed] [Google Scholar]
  • [70].Lucey BP. Teaching video NeuroImages: Phenytoin-induced orofacial dyskinesias. Neurology 2012;79:e177. [DOI] [PubMed] [Google Scholar]
  • [71].Luhdorf K, Lund M. Phenytoin induced hyperkinesia. Epilepsia 1977;18:409–15. [DOI] [PubMed] [Google Scholar]
  • [72].Maiti B, Saha P. Phenytoin intoxication with activated seizure and dyskinesia. J Assoc Physicians India 1987;35:598–9. [PubMed] [Google Scholar]
  • [73].Mauguiere F, Dalery J, De Villard R, Courjon J. Transient hyperkinesia after a single intravenous perfusion of diphenylhydantoin. report of a case associated with nontoxic plasma levels of diphenylhydantoin. Eur Neurol 1979;18:116–23. [DOI] [PubMed] [Google Scholar]
  • [74].McLellan DL, Swash M. Choreo-athetosis and encephalopathy induced by phenytoin. Br Med J 1974;2:204–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Menon VB, Kurian J, Undela K, Ramesh M, Gowdappa HB. Phenytoin toxicity: A case report. J Young Pharm 2015;7:272–5. [Google Scholar]
  • [76].Montenegro MA, Scotoni AE, Cendes F. Dyskinesia induced by phenytoin. Arq Neuro-Psiquiatr 1999;57:356–60. [DOI] [PubMed] [Google Scholar]
  • [77].Moss W, Ojukwu C, Chiriboga CA. Phenytoin-induced movement disorder: Unilateral presentation in a child and response to diphenhydramine. Clin Pediatr 1994;33:634–6. [DOI] [PubMed] [Google Scholar]
  • [78].Namjoshi AA, Aruna AS. Multiple episodes of phenytoin toxicity in an elderly patient. J Pharm Technol 1997;13:122–6. [Google Scholar]
  • [79].Nausieda PA, Koller WC, Weiner WJ, Klawans HL. Clinical and experimental studies of phenytoin-induced hyperkinesias. J NEURAL TRANSM GEN SECT 1979;45:291–305. [DOI] [PubMed] [Google Scholar]
  • [80].Patel DM, Gurumukhani JK, Patel MV, Patel GR. Phenytoin induced chorea: A rare adverse effect of the drug. Curr Drug Saf 2019;14:51–2. [DOI] [PubMed] [Google Scholar]
  • [81].Praveen-Kumar S, Desai M. Ocular motor abnormalities in a patient with phenytoin toxicity - case report and minireview. Clin Neurol Neurosurg 2014;127:116–7. [DOI] [PubMed] [Google Scholar]
  • [82].Puri V, Chaudhry N. Total external ophthalmoplegia induced by phenytoin: A case report and review of literature. Neurol India 2004;52:386–7. [PubMed] [Google Scholar]
  • [83].Rajasekharan C, Tina AM, Renjith SW. Orofaciolingual dyskinesia due to diphenylhydantoin sodium. BMJ Case Rep 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Rajkumar D, Manokaran RK, Shubha S, Shruthi TK. Phenytoin induced status dystonicus: A rare manifestation of phenytoin toxicity in a child with autism spectrum disorder. Indian J Pediatr 2021;88:85–6. [DOI] [PubMed] [Google Scholar]
  • [85].Reilly K, Collins D, Macdonald DR. Permanent ataxia from phenytoin toxicity. Can J Hosp Pharm 1990;43:27–9. [Google Scholar]
  • [86].Robertson K, Von Stempel CB, Arnold I. When less is more: A case of phenytoin toxicity. BMJ Case Rep 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Saito Y, Oguni H, Awaya Y, Hayashi K, Osawa M. Phenytoin-induced choreoathetosis in patients with severe myoclonic epilepsy in infancy. Neuropediatrics 2001;32:231–5. [DOI] [PubMed] [Google Scholar]
  • [88].Shulman LM, Singer C, Weiner WJ. Phenytoin-induced focal chorea. Mov Disord 1996;11:111–4. [DOI] [PubMed] [Google Scholar]
  • [89].Sivathanu S, Sampath S, David HS, Rajavelu KK. Myristicin and phenytoin toxicity in an infant. BMJ Case Rep 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Tomson T Choreoathetosis induced by ordinary phenytoin levels, explained by high free fraction? - A case report. Ther Drug Monit 1988;10:239–41. [DOI] [PubMed] [Google Scholar]
  • [91].Verma R, Kumar S, Biyani S, Singh A. Opsoclonus - myoclonus syndrome induced by phenytoin intoxication. J Neurosci Rural Pract 2014;5:S109–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Yoshida M, Yamada S, Ozaki Y, Nakanishi T. Phenytoin-induced orofacial dyskinesia. A case report. J Neurol 1985;231:340–2. [DOI] [PubMed] [Google Scholar]
  • [93].Zinsmeister S, Marks RE. Acute athetosis as a result of phenytoin toxicity in a child. Am J Dis Child 1976;130:75–6. [DOI] [PubMed] [Google Scholar]
  • [94].Harrison MB, Lyons GR, Landow ER. Phenytoin and dyskinesias: A report of two cases and review of the literature. Mov Disord 1993;8:19–27. [DOI] [PubMed] [Google Scholar]
  • [95].Kumar S, Sarangi SC, Tripathi M, Gupta YK. Evaluation of adverse drug reaction profile of antiepileptic drugs in persons with epilepsy: A cross-sectional study. Epilepsy Behav 2020;105. [DOI] [PubMed] [Google Scholar]
  • [96].Shanmugarajah PD, Hoggard N, Aeschlimann DP et al. Phenytoin-related ataxia in patients with epilepsy: Clinical and radiological characteristics. Seizure 2018;56:26–30. [DOI] [PubMed] [Google Scholar]
  • [97].Anderson M, Egunsola O, Cherrill J, Millward C, Fakis A, Choonara I. A prospective study of adverse drug reactions to antiepileptic drugs in children. 2015;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Pal G, Lin MM, Laureno R. Asterixis: A study of 103 patients. Metab Brain Dis 2014;29:813–24. [DOI] [PubMed] [Google Scholar]
  • [99].Bansal D, Azad C, Kaur M, Rudroju N, Vepa P, Guglani V. Adverse effects of antiepileptic drugs in north indian pediatric outpatients. Clin Neuropharmacol 2013;36:107–13. [DOI] [PubMed] [Google Scholar]
  • [100].Kuruvilla SM, Mukherjee SD. Phenytoin toxicity in a patient receiving 5-fluorouracil-based chemotherapy for metastatic colorectal cancer. Curr Oncol 2011;18:264–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Zeng K, Wang X, Xi Z, Yan Y. Adverse effects of carbamazepine, phenytoin, valproate and lamotrigine monotherapy in epileptic adult chinese patients. Clin Neurol Neurosurg 2010;112:291–5. [DOI] [PubMed] [Google Scholar]
  • [102].Hussein A, Abdulgalil A, Omer F et al. Correlation between serum level of antiepileptic drugs and their side effects. Oman Med J 2010;25:17–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Zadikoff C, Munhoz RP, Asante AN et al. Movement disorders in patients taking anticonvulsants. J Neurol Neurosurg Psychiatry 2007;78:147–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Benvenuti F, Bandinelli S, Mencarelli MA et al. Alterations of ballistic movements in epileptic patients with phenytoin intoxication. Epilepsia 1992;33:376–88. [DOI] [PubMed] [Google Scholar]
  • [105].Cohen AF, Ashby L, Crowley D. Lamotrigine (BW430C), a potential anticonvulsant. effects on the central nervous system in comparison with phenytoin and diazepam. Br J Clin Pharmacol 1985;20:619–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Luef G, Chemelli A, Birbamer G, Aichner F, Bauer G. Phenytoin overdosage and cerebellar atrophy in epileptic patients: Clinical and MRI findings. Eur Neurol 1994;34:79–81. [DOI] [PubMed] [Google Scholar]
  • [107].Pulliainen V, Jokelainen M, Hedman C, Pammo O. A case of cerebellar atrophy after phenytoin intoxication: Neurologic, neuroradiologic, and neuropsychological findings. J Epilepsy 1998;11:241–7. [Google Scholar]
  • [108].Panachiyil GM, Babu T, Sebastian J, Ravi MD. A case report of fosphenytoin induced orofacial dyskinesia in an 11-month-old baby with post-encephalitic sequelae. J Clin Diagn Res 2019;13:SD07–8. [Google Scholar]
  • [109].Shaikh AG. Fosphenytoin induced transient pendular nystagmus. J Neurol Sci 2013;330:121–2. [DOI] [PubMed] [Google Scholar]
  • [110].Thodeson DM, Reiber DC, Dolce AM, Sirsi D. Fosphenytoin-induced dyskinesias in an infant with sturge-weber syndrome. Neurology 2016;86:1561–2. [DOI] [PubMed] [Google Scholar]
  • [111].Ramsay RE, Wilder BJ, Uthman BM et al. Intramuscular fosphenytoin (cerebyx®) in patients requiring a loading dose of phenytoin. Epilepsy Res 1997;28:181–7. [DOI] [PubMed] [Google Scholar]
  • [112].Alvarez-Gomez M, Vaamonde J, Narbona J et al. Parkinsonian syndrome in childhood after sodium valproate administration. Clin Neuropharmacol 1993;16:451–5. [DOI] [PubMed] [Google Scholar]
  • [113].Botturi A, Silvani A, Pravettoni G, Paoli RA, Lucchiari C. Reversible valproate induced pisa syndrome and parkinsonism in a neuro-oncology patient with depression and epilepsy. Case Rep Neurol 2016;8:115–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Caruana Galizia E, Isaacs JD, Cock HR. Non-hyperammonaemic valproate encephalopathy after 20 years of treatment. Epilepsy Behav Case Report 2017;8:9–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Gardner TM, Aziz R, Muralee S, Tampi RR. Valproic acid-induced myoclonus in a demented patient: A case report. Case Rep Med 2009;2009:392091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Giordano A, Amboni M, Tessitore A. Valproate-induced generalized choreoathetosis. Mov Disord Clin Pract 2014;1:271–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Gunal DI, Guleryuz M, Bingol CA. Reversible valproate-induced choreiform movements. Seizure 2002;11:205–6. [DOI] [PubMed] [Google Scholar]
  • [118].Hassamal S, Waller S, Reese K, Testa C. Reversible valproic acid-induced parkinsonism and cognitive impairment in an elderly patient with bipolar disorder I. 2016;27:213–7. [PubMed] [Google Scholar]
  • [119].Haut F, Clos S. Acute akathisia and sodium valproate. Int J Psychiatry Clin Pract 2001;5:219–22. [DOI] [PubMed] [Google Scholar]
  • [120].Iijima M Valproate-induced parkinsonism in a demented elderly patient [1]. J Clin Psychiatry 2002;63:75. [DOI] [PubMed] [Google Scholar]
  • [121].Khwaja GA, Ranjan R, Gupta M, Chowdhry D, Hirve M. Valproate-induced reversible ‘parkinsonism plus’ syndrome. J Indian Acad Clin Med 2010;11:235–8. [Google Scholar]
  • [122].Lancman ME, Asconape JJ, Penry JK. Choreiform movements associated with the use of valproate. Arch Neurol 1994;51:702–4. [DOI] [PubMed] [Google Scholar]
  • [123].Maximov KG, Maximov GK. Valproate-induced parkinsonism in epilepsy patient - A case report. Acta Med Bulg 2008;35:38–42. [Google Scholar]
  • [124].Morrison PF, Sankar R, Shields WD. Valproate-induced chorea and encephalopathy in atypical nonketotic hyperglycinemia. Pediatr Neurol 2006;35:356–8. [DOI] [PubMed] [Google Scholar]
  • [125].Oh J, Park KD, Cho HJ, Choi KG, Jung SM. Spasmodic dysphonia induced by valproic acid. Epilepsia 2004;45:880–1. [DOI] [PubMed] [Google Scholar]
  • [126].Onofrj M, Thomas A, Paci C. Reversible parkinsonism induced by prolonged treatment with valproate. J Neurol 1998;245:794–6. [DOI] [PubMed] [Google Scholar]
  • [127].Salazar Z, Tschopp L, Calandra C, Micheli F. Pisa syndrome and parkinson secondary to valproic acid in huntington’s disease. Mov Disord 2008;23:2430–1. [DOI] [PubMed] [Google Scholar]
  • [128].Sasso E, Delsoldato S, Negrotti A, Mancia D. Reversible valproate-induced extrapyramidal disorders. Epilepsia 1994;35:391–3. [DOI] [PubMed] [Google Scholar]
  • [129].Sechi GP, Conti M, Sau GF, Cocco GA. Valproate-induced parkinsonism, glial cells and alexander’s disease. Prog Neuro-Psychopharmacol Biol Psychiatry 2008;32:1351–2. [DOI] [PubMed] [Google Scholar]
  • [130].Silver M, Factor SA. Valproic acid-induced parkinsonism: Levodopa responsiveness with dyskinesia. Parkinsonism Relat Disord 2013;19:758–60. [DOI] [PubMed] [Google Scholar]
  • [131].Srinivasan S, Lok AW. Valproate-induced reversible hemichorea. Mov Disord 2010;25:1511–2. [DOI] [PubMed] [Google Scholar]
  • [132].van de Velde K, Cras P, Helsen G. Acute chorea caused by valproate in an elderly. Acta Neurol Belg 2011;111:220–1. [PubMed] [Google Scholar]
  • [133].Yohanan M, Aulakh JS, Weith J, Hawkins JW. Pisa syndrome in a patient in a wheelchair taking valproic acid [2]. Am J Psychiatry 2006;163:325–6. [DOI] [PubMed] [Google Scholar]
  • [134].Gupta A, Kushwaha S. Disabling resting tremors induced by the short-term infusion of valproate: A reversible phenomenon. 2018;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Hommet C, Mondon K, De Toffol B, Constans T. Reversible cognitive and neurological symptoms during valproic acid therapy [6]. J Am Geriatr Soc 2007;55:628. [DOI] [PubMed] [Google Scholar]
  • [136].Kakisaka Y, Ito S, Ohara T, Hino-Fukuyo N, Uematsu M, Kure S. Asymmetric drug-induced tremor: Rare feature of a common event. Pediatr Neurol 2013;48:479–80. [DOI] [PubMed] [Google Scholar]
  • [137].Miyauchi T Valproate intoxication in a patient with bipolar I disorder due to SGLT2 inhibitor-induced weight reduction. DARU 2020;28:419–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Siniscalchi A, Gallelli L, Loizzo S, Tiziana A, Russo E, De Sarro G. Sodium valproate induced tremor in a patient with epilepsy and down’s syndrome. 2013;8:69–71. [DOI] [PubMed] [Google Scholar]
  • [139].Aguglia U, Gambardella A, Zappia M, Valentino P, Quattrone A. Negative myoclonus during valproate-related stupor. neurophysiological evidence of a cortical non-epileptic origin. Electroencephalogr Clin Neurophysiol 1995;94:103–8. [DOI] [PubMed] [Google Scholar]
  • [140].Bondon-Guitton E, Perez-Lloret S, Bagheri H, Brefel C, Rascol O, Montastruc J-. Drug-induced parkinsonism: A review of 17 years’ experience in a regional pharmacovigilance center in france. Mov Disord 2011;26:2226–31. [DOI] [PubMed] [Google Scholar]
  • [141].Easterford K, Clough P, Kellett M, Fallon K, Duncan S. Reversible parkinsonism with normal beta-CIT-SPECT in patients exposed to sodium valproate. Neurology 2004;62:1435–7. [DOI] [PubMed] [Google Scholar]
  • [142].Jamora D, Lim S-, Pan A, Tan L, Tan E-. Valproate-induced parkinsonism in epilepsy patients. Mov Disord 2007;22:130–3. [DOI] [PubMed] [Google Scholar]
  • [143].Masmoudi K, Gras-Champel V, Masson H, Andréjak M. Parkinsonism and/or cognitive impairment with valproic acid therapy: A report of ten cases. Pharmacopsychiatry 2006;39:9–12. [DOI] [PubMed] [Google Scholar]
  • [144].Nouzeilles M, García M, Rabinowicz A, Merello M. Prospective evaluation of parkinsonism and tremor in patients treated with valproate. Parkinsonism Relat Disord 1999;5:67–8. [DOI] [PubMed] [Google Scholar]
  • [145].Novick D, Gonzalez-Pinto A, Haro JM et al. Translation of randomised controlled trial findings into clinical practice: Comparison of olanzapine and valproate in the EMBLEM study. Pharmacopsychiatry 2009;42:145–52. [DOI] [PubMed] [Google Scholar]
  • [146].Schwartz TH, Karpitskiy VV, Sohn RS. Intravenous valproate sodium in the treatment of daily headache. Headache 2002;42:519–22. [DOI] [PubMed] [Google Scholar]
  • [147].Simeon D, Baker B, Chaplin W, Braun A, Hollander E. An open-label trial of divalproex extended-release in the treatment of borderline personality disorder. CNS Spectr 2007;12:439–43. [DOI] [PubMed] [Google Scholar]
  • [148].Alkhalil HJ, Sridhar SB, Rabbani SA, Al Omar A. Intensive monitoring of adverse drug reactions to antiepileptic drugs in neurology department of a secondary care hospital in U.A.E. J Young Pharm 2019;11:192–6. [Google Scholar]
  • [149].Alonso-Juarez M, Baizabal-Carvallo J. Distinguishing features between valproate-induced tremor and essential tremor. Acta Neurol Scand 2018;138:177–81. [DOI] [PubMed] [Google Scholar]
  • [150].Alonso-Juarez M, Torres-Russotto D, Crespo-Morfin P, Baizabal-Carvallo J. The clinical features and functional impact of valproate-induced tremor. Parkinsonism Relat Disord 2017;44:147–50. [DOI] [PubMed] [Google Scholar]
  • [151].Baizabal-Carvallo J, Alonso-Juarez M. Valproate-induced rest tremor and parkinsonism. Acta Neurol Belg 2021;121:515–9. [DOI] [PubMed] [Google Scholar]
  • [152].Bavrasad R, Nejad SEM, Yarahmadi AR, Sajedi SI, Rahim F. Assessment of the middle dose of topiramate in comparison with sodium valproate for migraine prophylaxis: A randomized-double-blind study; rouzdarou pharmaceutical(iran); sobhan(iran). Int J Pharmacol 2010;6:670–5. [Google Scholar]
  • [153].Calabrese JR, Goethe JW, Kayser A et al. Adverse events in 583 valproate-treated patients. Depression 1995;3:257–62. [Google Scholar]
  • [154].Deleu D, Al-Hail H, Mesraoua B, Mahmoud HA. Short-term efficacy and safety of valproate sustained-release formulation in newly diagnosed partial epilepsy (VIPe-study): A multicenter observational open-label study. Saudi Med J 2007;28:1402–7. [PubMed] [Google Scholar]
  • [155].Egunsola O, Choonara I, Sammons HM, Whitehouse WP. Safety of antiepileptic drugs in children and young people: A prospective cohort study. Seizure 2018;56:20–5. [DOI] [PubMed] [Google Scholar]
  • [156].Giri VP, Giri OP, Khan FA, Kumar N, Kumar A, Haque A. Valproic acid versus lamotrigine as first-line monotherapy in newly diagnosed idiopathic generalized tonic —Clonic seizures in adults — A randomized controlled trial. J Clin Diagn Res 2016;10:FC01–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Hakami T, Todaro M, Petrovski S et al. Substitution monotherapy with levetiracetam vs older antiepileptic drugs: A randomized comparative trial. Arch Neurol 2012;69:1563–71. [DOI] [PubMed] [Google Scholar]
  • [158].Hamed SA, Abdellah MM. The relationship between valproate induced tremors and circulating neurotransmitters: A preliminary study. Int J Neurosci 2017;127:236–42. [DOI] [PubMed] [Google Scholar]
  • [159].Houston JP, Tohen M, Degenhardt EK, Jamal HH, Liu LLL, Ketter TA. Olanzapine-divalproex combination versus divalproex monotherapy in the treatment of bipolar mixed episodes: A double-blind, placebo-controlled study. J Clin Psychiatry 2009;70:1540–7. [DOI] [PubMed] [Google Scholar]
  • [160].Hwang H, Kim H, Kim SH et al. Long-term effectiveness of ethosuximide, valproic acid, and lamotrigine in childhood absence epilepsy. Brain Dev 2012;34:344–8. [DOI] [PubMed] [Google Scholar]
  • [161].Kaushik S, Chopra D, Sharma S, Aneja S. Adverse drug reactions of anti-epileptic drugs in children with epilepsy: A cross-sectional study. Curr Drug Saf 2019;14:217–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].McElroy SL, Martens BE, Creech RS et al. Randomized, double-blind, placebo-controlled study of divalproex extended release loading monotherapy in ambulatory bipolar spectrum disorder patients with moderate-to-severe hypomania or mild mania. J Clin Psychiatry 2010;71:557–65. [DOI] [PubMed] [Google Scholar]
  • [163].Mehndiratta MM, Satyawani M, Gupta S, Khwaja GA. Clinical and surface EMG characteristics of valproate induced tremors. Electromyogr Clin Neurophysiol 2005;45:177–82. [PubMed] [Google Scholar]
  • [164].Miró J, Aiguabella M, Veciana M et al. Low-dose sodium valproate in the treatment of idiopathic generalized epilepsies. Acta Neurol Scand 2014;129:e20–3. [DOI] [PubMed] [Google Scholar]
  • [165].Munoz-Garcia D, Del Ser T, Bermejo F, Portera A. Truncal ataxia in chronic anticonvulsant treatment. association with drug-induced folate deficiency. J Neurol Sci 1982;55:305–11. [DOI] [PubMed] [Google Scholar]
  • [166].Paparella G, Angelini L, De Biase A et al. Clinical and kinematic features of valproate-induced tremor and differences with essential tremor. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Park KM, Kim SH, Nho SK et al. A randomized open-label observational study to compare the efficacy and tolerability between topiramate and valproate in juvenile myoclonic epilepsy. J Clin Neurosci 2013;20:1079–82. [DOI] [PubMed] [Google Scholar]
  • [168].Pisani F, Oteri G, Russo MF, Di Perri R, Perucca E, Richens A. The efficacy of valproate-lam otrigine comedication in refractory complex partial seizures: Evidence for a pharmacodynamic interaction. Epilepsia 1999;40:1141–1146. [DOI] [PubMed] [Google Scholar]
  • [169].Rajesh B, Ashalatha R, Sarma S, Radhakrishnan K. An assessment of the clinical equivalence of valproate chrono and extended release divalproex formulations; ranbaxy(india). Ann Indian Acad Neurol 2007;10:169–74. [Google Scholar]
  • [170].Sarchielli P, Messina P, Cupini LM et al. Sodium valproate in migraine without aura and medication overuse headache: A randomized controlled trial. Eur Neuropsychopharmacol 2014;24:1289–97. [DOI] [PubMed] [Google Scholar]
  • [171].Sekhar S, Kalra B, Mendhekar DN, Tekur U. Efficacy of sodium valproate and haloperidol in the management of acute mania: A randomized open-label comparative study. J Clin Pharmacol 2010;50:688–92. [DOI] [PubMed] [Google Scholar]
  • [172].Shah N, Reddy MS, Vohra S, Chaudhuri U, Mohanasundaram S. Safety and effectiveness of divalproex sodium extended release containing regimen in indian patients with bipolar I disorder in continuation phase: Results of EASED registry. Asian J Psychiatry 2016;20:32–8. [DOI] [PubMed] [Google Scholar]
  • [173].Silvennoinen K, de Lange N, Zagaglia S et al. Comparative effectiveness of antiepileptic drugs in juvenile myoclonic epilepsy. 2019;4:420–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Spitz MC, Deasy DN. Conversion to valproate monotherapy in nonretarded adults with primary generalized tonic-clonic seizures. J EPILEPSY 1991;4:33–8. [Google Scholar]
  • [175].Steinhoff BJ, Ueberall MA, Siemes H, Kurlemann G, Schmitz B, Bergmann L. The LAM-SAFE study: Lamotrigine versus carbamazepine or valproic acid in newly diagnosed focal and generalised epilepsies in adolescents and adults. Seizure 2005;14:597–605. [DOI] [PubMed] [Google Scholar]
  • [176].Stephen LJ, Sills GJ, Leach JP et al. Sodium valproate versus lamotrigine: A randomised comparison of efficacy, tolerability and effects on circulating androgenic hormones in newly diagnosed epilepsy. Epilepsy Res 2007;75:122–9. [DOI] [PubMed] [Google Scholar]
  • [177].Verrotti A, Nanni G, Agostinelli S et al. Effects of the abrupt switch from solution to modified-release granule formulation of valproate. Acta Neurol Scand 2012;125:e14–8. [DOI] [PubMed] [Google Scholar]
  • [178].Wieshmann UC, Tan GM, Baker G. Self-reported symptoms in patients on anti epileptic drugs in monotherapy. Acta Neurol Scand 2011;124:355–8. [DOI] [PubMed] [Google Scholar]
  • [179].Xiao Y, Xiong W, Lu L et al. The clinical characteristics and related factors of tremor in patients with epilepsy. Seizure 2019;66:70–5. [DOI] [PubMed] [Google Scholar]
  • [180].Yilmaz U, Yilmaz TS, Dizdarer G, Akinci G, Güzel O, Tekgül H. Efficacy and tolerability of the first antiepileptic drug in children with newly diagnosed idiopathic epilepsy. Seizure 2014;23:252–9. [DOI] [PubMed] [Google Scholar]
  • [181].Zeng Q-, Fan T-, Zhu P et al. Comparative long-term effectiveness of a monotherapy with five antiepileptic drugs for focal epilepsy in adult patients: A prospective cohort study. 2015;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Stremski ES, Brady WB, Prasad K, Hennes HA. Pediatric carbamazepine intoxication. Ann Emerg Med 1995;25:624–30. [DOI] [PubMed] [Google Scholar]
  • [183].Consoli D, Bosco D, Postorino P et al. Levetiracetam versus carbamazepine in patients with late poststroke seizures: A multicenter prospective randomized open-label study (epic project). Cerebrovasc Dis 2012;34:282–9. [DOI] [PubMed] [Google Scholar]
  • [184].Jayasutha J, Bhargavdilip S, Kishore K, Ramasamy C. Comparision of efficacy and safety of carbamazepine and eslicarbazepine in adult partial and generalized seizures. Asian J Pharm Clin Res 2014;7:144–7. [Google Scholar]
  • [185].Lee BI, No SK, Yi S- et al. Unblinded, randomized multicenter trial comparing lamotrigine and valproate combination with controlled-release carbamazepine monotherapy as initial drug regimen in untreated epilepsy. Seizure 2018;55:17–24. [DOI] [PubMed] [Google Scholar]
  • [186].Schmitz B, Dimova S, Zhang Y, Chellun D, De Backer M, Gasalla T. Tolerability and efficacy of lacosamide and controlled-release carbamazepine monotherapy in patients with newly diagnosed epilepsy and concomitant psychiatric conditions: Post hoc analysis of a prospective, randomized, double-blind trial. Epilepsy Res 2020;159. [DOI] [PubMed] [Google Scholar]
  • [187].Sebastian J, Adepu R, Keshava BS, Harsha S. Assessment of antiepileptic drugs usage in a south indian tertiary care teaching hospital. Neurol Asia 2013;18:159–65. [Google Scholar]
  • [188].Sobaniec W, Kulak W, Smigielska-Kuzia J, Bockowski L, Majkowski J, Jedrzejczak J. A multicenter, placebo-controlled, double-blind study of efficacy of a new form of carbamazepine (CarbatrolR) in refractory epileptic patients. Polish journal of pharmacology 2004;56:195–201. [PubMed] [Google Scholar]
  • [189].Aguglia U, Zappia M, Quattrone A. Carbamazepine-induced nonepileptic myoclonus in a child with benign epilepsy. Epilepsia 1987;28:515–8. [DOI] [PubMed] [Google Scholar]
  • [190].Al-Sibahee E, Alkaissi H. Carbamazepine-induced nystagmus in a 29-year-old patient. Neurol Clin Neurosci 2020;8:42–3. [Google Scholar]
  • [191].Arnstein E Oculogyric crisis: A distinct toxic effect of carbamazepine. J Child Neurol 1986;1:289–90. [DOI] [PubMed] [Google Scholar]
  • [192].Bansal S, Gill M, Bhasin C. Carbamazepine-induced dystonia in an adolescent. Indian J Pharmacol 2016;48:329–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Berchou RC. Carbamazepine-induced oculogyric crisis. Arch Neurol 1979;36:522–3. [DOI] [PubMed] [Google Scholar]
  • [194].Bimpong-Buta K, Froescher W. Carbamazepine-induced choreoathetoid dyskinesias. J NEUROL NEUROSURG PSYCHIATRY 1982;45:560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Bradbury AJ, Bentick B, Todd PJ. Dystonia associated with carbamazepine toxicity. Postgrad Med J 1982;58:525–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Chrousos GA, Cowdry R, Schuelein M. Two cases of downbeat nystagmus and oscillopsia associated with carbamazepine. Am J Ophthalmol 1987;103:221–4. [DOI] [PubMed] [Google Scholar]
  • [197].Dover K, Stephens JR. A 16-year-old girl with altered mental status, abducens nerve palsy, and ataxia. CLIN PEDIATR 2016;55:1256–9. [DOI] [PubMed] [Google Scholar]
  • [198].Fadare JO, Owolabi LF. Carbamazepine-induced dystonia, a case report. Neurol Asia 2009;14:165–6. [Google Scholar]
  • [199].Gorman M, Barkley GL. Oculogyric crisis induced by carbamazepine. Epilepsia 1995;36:1158–60. [DOI] [PubMed] [Google Scholar]
  • [200].Holtmann M, Korn-Merker E, Boenigk HE. Carbamazepine-induced combined phonic and motor tic in a boy with down’s syndrome. Epileptic Disord 2000;2:39–40. [PubMed] [Google Scholar]
  • [201].Joyce RP, Gunderson CH. Carbamazepine-induced orofacial dyskinesia. Neurology 1980;30:1333–4. [DOI] [PubMed] [Google Scholar]
  • [202].Klrlk S, Yiś U. Carbamazepine-induced nonepileptic myoclonus in a child with autism and epilepsy. J Pediatr Epilepsy 2021. [Google Scholar]
  • [203].Kurlan R, Kersun J, Behr J et al. Carbamazepine-induced tics. Clin Neuropharmacol 1989;12:298–302. [DOI] [PubMed] [Google Scholar]
  • [204].Lee JWY. Persistent dystonia associated with carbamazepine therapy; a case report. NEW ZEALAND MED J 1994;107:360–1. [PubMed] [Google Scholar]
  • [205].Magaudda A, Di Rosa G. Carbamazepine-induced non-epileptic myoclonus and tic-like movements. Epileptic Disord 2012;14:172–3. [DOI] [PubMed] [Google Scholar]
  • [206].Matarazzo M, Sánchez-Seco VG, Méndez-Guerrero AJ et al. Drug-related eyelid nystagmus: Two cases of a rare clinical phenomenon related to carbamazepine and derivatives. Clin Neuropharmacol 2016;39:49–50. [DOI] [PubMed] [Google Scholar]
  • [207].Milne IK. Akathisia associated with carbamazepine therapy. N Z Med J 1992;105:182. [PubMed] [Google Scholar]
  • [208].Prakash S, Bhanvadia RJ, Shah ND. Restless legs syndrome with carbamazepine-induced osteomalacia: Causal or casual association. Gen Hosp Psychiatry 2010;32:228.e1, 228.e3. [DOI] [PubMed] [Google Scholar]
  • [209].Rittmannsberger H, Leblhuber F. Asterixis induced by carbamazepine therapy. Biol Psychiatry 1992;32:364–8. [DOI] [PubMed] [Google Scholar]
  • [210].Robertson PL, Garofalo EA, Silverstein FS, Komarynski MA. Carbamazepine-induced tics. Epilepsia 1993;34:965–8. [DOI] [PubMed] [Google Scholar]
  • [211].Schwarcz G, Gosenfeld L, Gilderman A, Jiwesh J, Ripple RE. Akathisia associated with carbamazepine therapy. Am J Psychiatry 1986;143:1190–1. [PubMed] [Google Scholar]
  • [212].Schwartzman MJ, Leppik IE. Carbamazepine-induced dyskinesia and ophthalmoplegia. CLEVEL CLIN J MED 1990;57:367–72. [DOI] [PubMed] [Google Scholar]
  • [213].Soman P, Jain S, Rajsekhar V, Vineeta S, Sharma BK. Dystonia - a rare manifestation of carbamazepine toxicity [2]. Postgrad Med J 1994;70:54–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Stryjer R, Strous RD, Bar F, Ulman A-, Rabey JM. Segmental dystonia as the sole manifestation of carbamazepine toxicity. Gen Hosp Psychiatry 2002;24:114–5. [DOI] [PubMed] [Google Scholar]
  • [215].Wijdicks EFM, Arendt C, Bazzell MC. Postoperative ophthalmoplegia and ataxia due to carbamazepine toxicity facilitated by diltiazem [2]. J Neuro-Ophthalmol 2004;24:95. [DOI] [PubMed] [Google Scholar]
  • [216].Wirfs L, Whitworth K, Kellar J. Nystagmus associated with carbamazepine toxicity. Clin Pract Cases Emerg Med 2017;1:441–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Rittmannsberger H, Leblhuber F, Sommer R. Asterixis as a side effect of carbamazepine therapy. Klin Wochenschr 1991;69:279–81. [DOI] [PubMed] [Google Scholar]
  • [218].Martin D, Hirt HR. Clinical experience with clonazepam (rivotril) in the treatment of epilepsies in infancy and childhood. Neuropadiatrie 1973;4:245–66. [DOI] [PubMed] [Google Scholar]
  • [219].Gillman MA, Sandyk R. Clonazepam-induced tourette syndrome in a subject with hyperexplexia. Postgrad Med J 1987;63:311–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Suranyi-Cadotte B, Nestoros JN, Nair NPV. Parkinsonism induced by high doses of diazepam. Biol Psychiatry 1985;20:455–7. [DOI] [PubMed] [Google Scholar]
  • [221].Rogin J, Wheless J, Abou-Khalil B et al. Safety and effectiveness of long-term treatment with diazepam auto-injector administered by caregivers in an outpatient setting for the treatment of acute repetitive seizures. Epilepsia 2014;55:1444–51. [DOI] [PubMed] [Google Scholar]
  • [222].Arai M, Fujii S. Divergence paralysis associated with the ingestion of diazepam. J Neurol 1990;237:45–6. [DOI] [PubMed] [Google Scholar]
  • [223].Kaplan SR, Murkofsky C. Oral-buccal dyskinesia symptoms associated with low-dose benzodiazepine treatment. Am J Psychiatry 1978;135:1558–9. [DOI] [PubMed] [Google Scholar]
  • [224].Brown DJF, McArthur D, Moulsdale H. Subcutaneous midazolam as a cause of extrapyramidal side effects in a patient with prostate cancer. J Pain Symptom Manage 2007;34:111–3. [DOI] [PubMed] [Google Scholar]
  • [225].Komur M, Arslankoylu A, Okuyaz C. Midazolam-induced acute dystonia reversed by diazepam. J Anaesthesiol Clin Pharmacol 2012;28:368–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Lauerma H Nocturnal wandering caused by restless legs and short-acting benzodiazepines. Acta Psychiatr Scand 1991;83:492–3. [DOI] [PubMed] [Google Scholar]
  • [227].McConn MM, Gundy JT, Karan SB, Lindenmuth DM. Adverse drug reaction: Midazolam-induced extrapyramidal symptoms: A case report. A A Pract 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Ozcan B, Kavurt S, Yucel H, Bas AY, Demirel N. Rhythmic myoclonic jerking induced by midazolam in a preterm infant. Pediatr Neurol 2015;52:e9. [DOI] [PubMed] [Google Scholar]
  • [229].Stolarek IH, Ford MJ. Acute dystonia induced by midazolam and abolished by flumazenil. Br Med J 1990;300:614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Vorsanger GJ, Roberts JT. Midazolam-induced athetoid movements of the lower extremities during epidural anesthesia reversed by physostigmine. J Clin Anesth 1993;5:494–6. [DOI] [PubMed] [Google Scholar]
  • [231].Sandyk R Orofacial dyskinesias associated with lorazepam therapy. Clin Pharm 1986;5:419–21. [PubMed] [Google Scholar]
  • [232].Lee DSC, Wong HA, Knoppert DC. Myoclonus associated with lorazepam therapy in very-low-birth-weight infants; wyeth. Biol Neonate 1994;66:311–5. [DOI] [PubMed] [Google Scholar]
  • [233].Conry JA, Ng Y-, Paolicchi JM et al. Clobazam in the treatment of lennox-gastaut syndrome. Epilepsia 2009;50:1158–66. [DOI] [PubMed] [Google Scholar]
  • [234].Perry MS, Bailey L, Malik S, Gilson C, Kotecha A, Hernandez A. Clobazam for the treatment of intractable epilepsy in children. J Child Neurol 2013;28:34–9. [DOI] [PubMed] [Google Scholar]
  • [235].Ehyai A, Kilroy AW, Fenichel GM. Dyskinesia and akathisia induced by ethosuximide. Am J Dis Child 1978;132:527–8. [DOI] [PubMed] [Google Scholar]
  • [236].Kirschberg GJ. Dyskinesia: An unusual reaction to ethosuximide. Arch Neurol 1975;32:137–8. [DOI] [PubMed] [Google Scholar]
  • [237].Madani N, O’Malley JA, Porter BE, Baumer FM. Lacosamide-induced dyskinesia in children with intractable epilepsy. J Child Neurol 2020;35:662–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [238].Flores L, Kemp S, Colbeck K et al. Clinical experience with oral lacosamide as adjunctive therapy in adult patients with uncontrolled epilepsy: A multicentre study in epilepsy clinics in the united kingdom (UK). Seizure 2012;21:512–7. [DOI] [PubMed] [Google Scholar]
  • [239].Guilhoto LMFF, Loddenkemper T, Gooty VD et al. Experience with lacosamide in a series of children with drug-resistant focal epilepsy. Pediatr Neurol 2011;44:414–9. [DOI] [PubMed] [Google Scholar]
  • [240].Chung S, Sperling MR, Biton V et al. Lacosamide as adjunctive therapy for partial-onset seizures: A randomized controlled trial; pharma schwarz(germany). Epilepsia 2010;51:958–67. [DOI] [PubMed] [Google Scholar]
  • [241].Ferreira JA, Le Pichon J-, Abdelmoity AT et al. Safety and tolerability of adjunctive lacosamide in a pediatric population with focal seizures – an open-label trial. Seizure 2019;71:166–73. [DOI] [PubMed] [Google Scholar]
  • [242].Fountain NB, Krauss G, Isojarvi J, Dilley D, Doty P, Rudd GD. Safety and tolerability of adjunctive lacosamide intravenous loading dose in lacosamide-naive patients with partial-onset seizures. Epilepsia 2013;54:58–65. [DOI] [PubMed] [Google Scholar]
  • [243].Giráldez BG, Toledano R, García-Morales I et al. Long-term efficacy and safety of lacosamide monotherapy in the treatment of partial-onset seizures: A multicenter evaluation. Seizure 2015;29:119–22. [DOI] [PubMed] [Google Scholar]
  • [244].Halász P, Kälviäinen R, Mazurkiewicz-Beldzińska M et al. Adjunctive lacosamide for partial-onset seizures: Efficacy and safety results from a randomized controlled trial. Epilepsia 2009;50:443–53. [DOI] [PubMed] [Google Scholar]
  • [245].Hillenbrand B, Wisniewski I, Jürges U, Steinhoff BJ. Add-on lacosamide: A retrospective study on the relationship between serum concentration, dosage, and adverse events. Epilepsy Behav 2011;22:548–51. [DOI] [PubMed] [Google Scholar]
  • [246].Husain A, Chung S, Faught E, Isojarvi J, McShea C, Doty P. Long-term safety and efficacy in patients with uncontrolled partial-onset seizures treated with adjunctive lacosamide: Results from a phase III open-label extension trial. Epilepsia 2012;53:521–8. [DOI] [PubMed] [Google Scholar]
  • [247].Pasha I, Kamate M, Suresh DK. Safety of lacosamide in children with refractory partial epilepsy. Saudi Pharm J 2015;23:556–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Rosenow F, Kelemen A, Ben-Menachem E et al. Long-term adjunctive lacosamide treatment in patients with partial-onset seizures. Acta Neurol Scand 2016;133:136–44. [DOI] [PubMed] [Google Scholar]
  • [249].Shaibani A, Biton V, Rauck R, Koch B, Simpson J. Long-term oral lacosamide in painful diabetic neuropathy: A two-year open-label extension trial. Eur J Pain 2009;13:458–63. [DOI] [PubMed] [Google Scholar]
  • [250].Shaibani A, Fares S, Selam J- et al. Lacosamide in painful diabetic neuropathy: An 18-week double-blind placebo-controlled trial. J Pain 2009;10:818–28. [DOI] [PubMed] [Google Scholar]
  • [251].Stephen LJ, Kelly K, Parker P, Brodie MJ. Adjunctive lacosamide in clinical practice: Sodium blockade with a difference? Epilepsy Behav 2011;22:499–504. [DOI] [PubMed] [Google Scholar]
  • [252].Stillman MJ. Efficacy and safety of lacosamide in diabetic neuropathic pain: An 18-week double-blind placebo-controlled trial of fixed-dose regimens. clin J pain. 2009;25:376-385: Comments. Headache 2010;50:335. [DOI] [PubMed] [Google Scholar]
  • [253].Villanueva V, Garcés M, López-Gomáriz E et al. Early add-on lacosamide in a real-life setting: Results of the REALLY study. Clin Drug Invest 2014;35:121–31. [DOI] [PubMed] [Google Scholar]
  • [254].Villanueva V, López FJ, Serratosa JM et al. Control of seizures in different stages of partial epilepsy: LACO-EXP, a spanish retrospective study of lacosamide. Epilepsy Behav 2013;29:349–56. [DOI] [PubMed] [Google Scholar]
  • [255].Villanueva V, López-Gomáriz E, López-Trigo J et al. Rational polytherapy with lacosamide in clinical practice: Results of a spanish cohort analysis RELACOVA. Epilepsy Behav 2012;23:298–304. [DOI] [PubMed] [Google Scholar]
  • [256].Vossler DG, Wechsler RT, Williams P, Byrnes W, Therriault S. Long-term exposure and safety of lacosamide monotherapy for the treatment of partial-onset (focal) seizures: Results from a multicenter, open-label trial. Epilepsia 2016;57:1625–33. [DOI] [PubMed] [Google Scholar]
  • [257].Wechsler RT, Li G, French J et al. Conversion to lacosamide monotherapy in the treatment of focal epilepsy: Results from a historical-controlled, multicenter, double-blind study. Epilepsia 2014;55:1088–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Wechsler RT, Yates SL, Messenheimer J, Leroy R, Beller C, Doty P. Lacosamide for uncontrolled primary generalized tonic-clonic seizures: An open-label pilot study with 59-week extension. Epilepsy Res 2017;130:13–20. [DOI] [PubMed] [Google Scholar]
  • [259].Wymer JP, Simpson J, Sen D, Bongardt S. Efficacy and safety of lacosamide in diabetic neuropathic pain: An 18-week double-blind placebo-controlled trial of fixed-dose regimens. Clin J Pain 2009;25:376–85. [DOI] [PubMed] [Google Scholar]
  • [260].Yorns WR, Khurana DS, Carvalho KS, Hardison HH, Legido A, Valencia I. Efficacy of lacosamide as adjunctive therapy in children with refractory epilepsy. J Child Neurol 2014;29:23–7. [DOI] [PubMed] [Google Scholar]
  • [261].Zadeh WW, Escartin A, Byrnes W et al. Efficacy and safety of lacosamide as first add-on or later adjunctive treatment for uncontrolled partial-onset seizures: A multicentre open-label trial. Seizure 2015;31:72–9. [DOI] [PubMed] [Google Scholar]
  • [262].Brodie MJ, Stephen LJ. Prospective audit with adjunctive perampanel: Preliminary observations in focal epilepsy. Epilepsy Behav 2016;54:100–3. [DOI] [PubMed] [Google Scholar]
  • [263].Datta AN, Xu Q, Sachedina S, Boelman C, Huh L, Connolly MB. Clinical experience with perampanel for refractory pediatric epilepsy in one canadian center. J Child Neurol 2017;32:834–9. [DOI] [PubMed] [Google Scholar]
  • [264].French JA, Krauss GL, Biton V et al. Adjunctive perampanel for refractory partial-onset seizures: Randomized phase III study 304. Neurology 2012;79:589–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [265].Morano A, Fattouch J, Albini M et al. Perampanel as adjunctive therapy in highly refractory epilepsies: Real-world data from an italian tertiary care epilepsy centre. J Neurol Sci 2018;390:67–74. [DOI] [PubMed] [Google Scholar]
  • [266].Gidal BE, Laurenza A, Hussein Z et al. Perampanel efficacy and tolerability with enzyme-inducing AEDs in patients with epilepsy. Neurology 2015;84:1972–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Hwang S-, Lee Y-, Nam SO et al. Real-life effectiveness and tolerability of perampanel in pediatric patients aged 4 years or older with epilepsy: A korean national multicenter study. J Clin Neurol 2020;16:53–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].Ishikawa N, Tateishi Y, Tani H, Kobayashi Y, Kobayashi M. Clinical profiles associated with serum perampanel concentrations in children with refractory epilepsy. Epilepsy Behav 2019;94:82–6. [DOI] [PubMed] [Google Scholar]
  • [269].Kwan P, Brodie MJ, Laurenza A, FitzGibbon H, Gidal BE. Analysis of pooled phase III trials of adjunctive perampanel for epilepsy: Impact of mechanism of action and pharmacokinetics on clinical outcomes. Epilepsy Res 2015;117:117–24. [DOI] [PubMed] [Google Scholar]
  • [270].Lin K-, Lin J-, Chou M- et al. Efficacy and tolerability of perampanel in children and adolescents with pharmacoresistant epilepsy: The first real-world evaluation in asian pediatric neurology clinics. Epilepsy Behav 2018;85:188–94. [DOI] [PubMed] [Google Scholar]
  • [271].Macrohon B, Janette Resurreccion-De La Calzada J, Sanchez-Gan B. Clinical experience on the use of perampanel in epilepsy among child neurologists in the philippines. Brain Dev 2021;43:411–8. [DOI] [PubMed] [Google Scholar]
  • [272].Maurousset A, Limousin N, Praline J, Biberon J, Corcia P, De Toffol B. Adjunctive perampanel in refractory epilepsy: Experience at tertiary epilepsy care center in tours. Epilepsy Behav 2016;61:237–41. [DOI] [PubMed] [Google Scholar]
  • [273].Montouris G, Yang H, Williams B, Zhou S, Laurenza A, Fain R. Efficacy and safety of perampanel in patients with drug-resistant partial seizures after conversion from double-blind placebo to open-label perampanel. Epilepsy Res 2015;114:131–40. [DOI] [PubMed] [Google Scholar]
  • [274].Rektor I, Krauss GL, Bar M et al. Perampanel study 207: Long-term open-label evaluation in patients with epilepsy. Acta Neurol Scand 2012;126:263–9. [DOI] [PubMed] [Google Scholar]
  • [275].Rinaldi F, De Maria G. Safety and efficacy of perampanel as adjunctive therapy in patients with refractory focal epilepsy over 12 months: Clinical experience in a real-world setting. Int J Epilepsy 2018;5:75–9. [Google Scholar]
  • [276].Rodríguez-Osorio X, Lema-Facal T, Rubio-Nazábal E et al. Perampanel effectiveness and safety as early add-on treatment for focal-onset seizures: PEREAGAL study. Epilepsy Res 2021;172. [DOI] [PubMed] [Google Scholar]
  • [277].Rohracher A, Zimmermann G, Villanueva V et al. Perampanel in routine clinical use across europe: Pooled, multicenter, observational data. Epilepsia 2018;59:1727–39. [DOI] [PubMed] [Google Scholar]
  • [278].Santamarina E, Bertol V, Garayoa V et al. Efficacy and tolerability of perampanel as a first add-on therapy with different anti-seizure drugs. Seizure 2020;83:48–56. [DOI] [PubMed] [Google Scholar]
  • [279].Steinhoff BJ, Hamer H, Trinka E et al. A multicenter survey of clinical experiences with perampanel in real life in germany and austria. Epilepsy Res 2014;108:986–8. [DOI] [PubMed] [Google Scholar]
  • [280].Villanueva V, Garcés M, López-González FJ et al. Safety, efficacy and outcome-related factors of perampanel over 12 months in a real-world setting: The FYDATA study. Epilepsy Res 2016;126:201–10. [DOI] [PubMed] [Google Scholar]
  • [281].Asconapé J, Diedrich A, DellaBadia J. Myoclonus associated with the use of gabapentin. Epilepsia 2000;41:479–81. [DOI] [PubMed] [Google Scholar]
  • [282].Desai A, Kherallah Y, Szabo C, Marawar R. Gabapentin or pregabalin induced myoclonus: A case series and literature review. J Clin Neurosci 2019;61:225–34. [DOI] [PubMed] [Google Scholar]
  • [283].Kim JB, Jung J-, Park M-, Lee EJ, Kwon D-. Negative myoclonus induced by gabapentin and pregabalin: A case series and systematic literature review. J Neurol Sci 2017;382:36–9. [DOI] [PubMed] [Google Scholar]
  • [284].Morris GL III. Efficacy and tolerability of gabapentin in clinical practice. Clin Ther 1995;17:891–900. [DOI] [PubMed] [Google Scholar]
  • [285].Rowbotham M, Harden N, Stacey B, Bernstein P, Magnus-Miller L. Gabapentin for the treatment of postherpetic neuralgia: A randomized controlled trial. JAMA 1998;280:1837–42. [DOI] [PubMed] [Google Scholar]
  • [286].Vieta E, Manuel Goikolea J, Martínez-Arán A et al. A double-blind, randomized, placebo-controlled, prophylaxis study of adjunctive gabapentin for bipolar disorder. Journal of clinical psychiatry 2006;67:473–477. [DOI] [PubMed] [Google Scholar]
  • [287].Vuković V, Lovrenčić-Huzjan A, Bosnar-Puretić M, Demarin V. The efficacy of gabapentin in migraine prophylaxis: An observational open label study. Acta Clin Croat 2009;48:145–51. [PubMed] [Google Scholar]
  • [288].Wong JO-, Tan TD-, Tseng K-, Cheu N-, Wu J-. Gabapentin for the treatment of chronic intractable neuropathic pain: A long-term follow-up study. 2005;17:357–65. [Google Scholar]
  • [289].Attupurath R, Aziz R, Wollman D, Muralee S, Tampi RR. Chorea associated with gabapentin use in an elderly man. Am J Geriatr Pharmacother 2009;7:220–4. [DOI] [PubMed] [Google Scholar]
  • [290].Buetefisch CM, Gutierrez A, Gutmann L. Choreoathetotic movements: A possible side effect of gabapentin. Neurology 1996;46:851–2. [PubMed] [Google Scholar]
  • [291].Cho K-, Hong S-. Myoclonus induced by the use of gabapentin. J Korean Neurosurg Soc 2008;43:237–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [292].Chudnow RS, Dewey RB Jr., Lawson CR. Choreoathetosis as a side effect of gabapentin therapy in severely neurologically impaired patients. Arch Neurol 1997;54:910–2. [PubMed] [Google Scholar]
  • [293].Ege F, Koçak Y, Titiz AP, Öztürk SM, Öztürk Ş, Özbakir Ş. Gabapentin-induced myoclonus: Case report. Mov Disord 2008;23:1947–8. [DOI] [PubMed] [Google Scholar]
  • [294].Erol C, Ozben S, Ozer F, Cetin S, Tiras R. Bilateral ballism induced by gabapentin in idiopatic parkinson’s disease. Clin Neurol Neurosurg 2009;111:397. [DOI] [PubMed] [Google Scholar]
  • [295].Guddati AK, Zafar Z, Cheng JT, Mohan S. Treatment of gabapentin-induced myoclonus with continuous renal replacement therapy. Indian J Nephrol 2012;22:59–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [296].Hampton Z, Shahrestani N, Little A. Gabapentin-induced facial myoclonus in the setting of acute on chronic kidney disease. Cureus 2019;11:e4758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [297].Holtkamp M, Halle A, Meierkord H, Masuhr F. Gabapentin-induced severe myoclonus in a patient with impaired renal function [3]. J Neurol 2006;253:382–3. [DOI] [PubMed] [Google Scholar]
  • [298].Hui C-, Leung JK-, Chang RS-, Shea Y-. Reversible dysphagia due to gabapentin-induced jaw myoclonus. Chin Med J 2019;132:1485–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [299].Jacob PC, Chand RP, Omeima E-. Asterixis induced by gabapentin. Clin Neuropharmacol 2000;23:53. [DOI] [PubMed] [Google Scholar]
  • [300].Kaufman KR, Parikh A, Chan L, Bridgeman M, Shah M. Myoclonus in renal failure: Two cases of gabapentin toxicity. Epilepsy Behav Case Report 2014;2:8–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [301].Norton JW, Quarles E. Gabapentin-related [5]. J Clin Psychopharmacol 2001;21:623–4. [DOI] [PubMed] [Google Scholar]
  • [302].Pina MA, Modrego PJ. Dystonia induced by gabapentin. Ann Pharmacother 2005;39:380–2. [DOI] [PubMed] [Google Scholar]
  • [303].Raju PM, Walker RW, Lee MA. Dyskinesia induced by gabapentin in idiopathic parkinson’s disease [6]. Mov Disord 2007;22:288–9. [DOI] [PubMed] [Google Scholar]
  • [304].Reeves AL, So EL, Sharbrough FW, Krahn LE. Movement disorders associated with the use of gabapentin. Epilepsia 1996;37:988–90. [DOI] [PubMed] [Google Scholar]
  • [305].Rohman L, Hebron A. Acute dystonic reaction caused by gabapentin. J Emerg Med 2014;46:e89. [DOI] [PubMed] [Google Scholar]
  • [306].Scullin P, Sheehan P, Kelly S. Myoclonic jerks associated with gabapentin. Palliative Med 2003;17:717–8. [DOI] [PubMed] [Google Scholar]
  • [307].Sechi G, Murgia B, Sau G et al. Asterixis and toxic encephalopathy induced by gabapentin. Prog Neuro-Psychopharmacol Biol Psychiatry 2004;28:195–9. [DOI] [PubMed] [Google Scholar]
  • [308].Shea Y-, Mok MMY, Chang RSK. Gabapentin-induced myoclonus in an elderly with end-stage renal failure. J Formos Med Assoc 2014;113:660–1. [DOI] [PubMed] [Google Scholar]
  • [309].Souzdalnitski D, Chang AK, Guirguis M. Chorea in a chronic pain patient using gabapentin. Ochsner J 2014;14:276–8. [PMC free article] [PubMed] [Google Scholar]
  • [310].Steinhoff BJ, Herrendorf G, Bittermann H-, Kurth C. Isolated ataxia as an idiosyncratic side-effect under gabapentin. Seizure 1997;6:503–4. [DOI] [PubMed] [Google Scholar]
  • [311].Twardowschy CA, Teive HAG, Fernandes AF, Búrigo IP, Lange M, Werneck LC. Chorea due to gabapentin monotherapyin a not encephalopatic patient. Arq Neuro-Psiquiatr 2008;66:107. [DOI] [PubMed] [Google Scholar]
  • [312].Yeddi A, Adam O, Khalid M et al. Myoclonus and altered mental status induced by single dose of gabapentin in a patient with end-stage renal disease: A case report and literature review. Am J Ther 2018;26:e768–70. [DOI] [PubMed] [Google Scholar]
  • [313].Zesiewicz TA, Shimberg WR, Hauser RA, Robinson W, Wilson M-, Sullivan KL. Chorea as a side effect of gabapentin (neurontin®) in a patient with complex regional pain syndrome type 1. Clin Rheumatol 2008;27:389–90. [DOI] [PubMed] [Google Scholar]
  • [314].Robertson KL, Marshman LAG. Gabapentin superadded to a pre-existent regime containing amytriptyline for chronic sciatica. Pain Med 2016;17:2095–9. [DOI] [PubMed] [Google Scholar]
  • [315].Kaufman KR, Parikh A, Chan L, Bridgeman M, Shah M. Myoclonus in renal failure: Two cases of gabapentin toxicity. Epilepsy Behav Case Rep 2013;2:8–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [316].Yeddi A, Adam O, Khalid M et al. Myoclonus and altered mental status induced by single dose of gabapentin in a patient with end-stage renal disease: A case report and literature review. Am J Ther 2019;26:e768–70. [DOI] [PubMed] [Google Scholar]
  • [317].Modur PN, Milteer WE. Adjunctive pregabalin therapy in mentally retarded, developmentally delayed patients with epilepsy. Epilepsy Behav 2008;13:554–6. [DOI] [PubMed] [Google Scholar]
  • [318].Schjerning O, Damkier P, Lykkegaard SE, Jakobsen KD, Nielsen J. Pregabalin for anxiety in patients with schizophrenia — A randomized, double-blind placebo-controlled study. Schizophr Res 2018;195:260–6. [DOI] [PubMed] [Google Scholar]
  • [319].Stephen LJ, Parker P, Kelly K, Wilson EA, Leach V, Brodie MJ. Adjunctive pregabalin for uncontrolled partial-onset seizures: Findings from a prospective audit. Acta Neurol Scand 2011;124:142–5. [DOI] [PubMed] [Google Scholar]
  • [320].Carreño M, Maestro I, Molins A et al. Pregabalin as add-on therapy for refractory partial seizures in every day clinical practice. Seizure 2007;16:709–12. [DOI] [PubMed] [Google Scholar]
  • [321].Elger CE, Brodie MJ, Anhut H, Lee CM, Barrett JA. Pregabalin add-on treatment in patients with partial seizures: A novel evaluation of flexible-dose and fixed-dose treatment in a double-blind, placebo-controlled study. Epilepsia 2005;46:1926–36. [DOI] [PubMed] [Google Scholar]
  • [322].Huber B, Bocchicchio M, Feuerbaum E et al. Efficacy and tolerability of pregabalin in patients with difficult-to-treat epilepsy and intellectual disability. Epilepsy Behav 2008;13:397–401. [DOI] [PubMed] [Google Scholar]
  • [323].Lee BI, Yi S, Hong SB et al. Pregabalin add-on therapy using a flexible, optimized dose schedule in refractory partial epilepsies: A double-blind, randomized, placebo-controlled, multicenter trial. Epilepsia 2009;50:464–74. [DOI] [PubMed] [Google Scholar]
  • [324].Loprinzi CL, Qin R, Baclueva EP et al. Phase III, randomized, double-blind, placebo-controlled evaluation of pregabalin for alleviating hot flashes, N07C1. J Clin Oncol 2010;28:641–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [325].Orita S, Yamashita M, Eguchi Y et al. Pregabalin for refractory radicular leg pain due to lumbar spinal stenosis: A preliminary prospective study. Pain Res Manage 2016;2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [326].Shaheen A, Alam SM, Ahmad A, Khan M. Clinical efficacy and tolerability of gabapentinoid with current prescription patterns in patients with neuropathic pain. Pak J Med Sci 2019;35:1505–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [327].Tsounis S, Kimiskidis VK, Kazis D et al. An open-label, add-on study of pregabalin in patients with partial seizures: A multicenter trial in greece. Seizure 2011;20:701–5. [DOI] [PubMed] [Google Scholar]
  • [328].Van Seventer R, Feister HA, Young JP Jr., Stoker M, Versavel M, Rigaudy L. Efficacy and tolerability of twice-daily pregabalin for treating pain and related sleep interference in postherpetic neuralgia: A 13-week, randomized trial. Curr Med Res Opin 2006;22:375–84. [DOI] [PubMed] [Google Scholar]
  • [329].Zamani G, Tavasoli A, Zare-Shahabadi A, Rezaei N, Ahmadvand A. Efficacy of pregabalin in childhood refractory partial seizure. Iran J Pediatr 2014;24:100–4. [PMC free article] [PubMed] [Google Scholar]
  • [330].Ari B, Domac F, Kenangil G. A case of pregabalin-induced parkinsonism. Neurol India 2020;68:1469–71. [DOI] [PubMed] [Google Scholar]
  • [331].Choi J-, Park Y-, Woo YS, Kim SU, Jung J-, Kwon D-. Perverted head-shaking and positional downbeat nystagmus in pregabalin intoxication. J Neurol Sci 2014;337:243–4. [DOI] [PubMed] [Google Scholar]
  • [332].Courtois F, Borrey D, Haufroid V, Hantson P. Pregabalin-associated myoclonic encephalopathy without evidence of drug accumulation in a patient with acute renal failure. Indian J Nephrol 2014;24:48–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [333].Dag E, Gokce B, Buturak SV, Tiryaki D, Erdemoglu AK. Pregabalin-induced akathisia. Ann Pharmacother 2013;47:592–3. [DOI] [PubMed] [Google Scholar]
  • [334].Ekinci AS, Ciftci S, Cavus B, Aydogdu I. Could pregabalin cause oculomotor symptoms in lower dose? A case with down beat nystagmus as a side effect. Acta Neurol Belg 2017;117:777–8. [DOI] [PubMed] [Google Scholar]
  • [335].Ge L, Li A, Wang N, Li P, Xin H, Li W. Pregabalin-associated stuttering and frequent blepharospasm: Case report and review; shandong zibo xinhua pharmaceutical company(china); pfizer(united states). DARU J Pharm Sci 2020;28:815–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [336].Healy DG, Ingle GT, Brown P. Pregabalin- and gabapentin-associated myoclonus in a patient with chronic renal failure. Mov Disord 2009;24:2028–9. [DOI] [PubMed] [Google Scholar]
  • [337].Heckmann JG, Ulrich K, Dütsch M, Neundörfer B. Pregabalin associated asterixis. Am J Phys Med Rehabil 2005;84:724. [DOI] [PubMed] [Google Scholar]
  • [338].Hellwig S, Amtage F. Pregabalin-induced cortical negative myoclonus in a patient with neuropathic pain. Epilepsy Behav 2008;13:418–20. [DOI] [PubMed] [Google Scholar]
  • [339].Hounnou P, Nicoucar K. Delayed onset of rotatory self-motion perception, dysdiadochokinesia and disturbed eye pursuit caused by low-dose pregabalin. BMJ Case Rep 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [340].Masmoudi I, Gras-Champel V, Barbieux-Vaquez D, Masmoudi K. Pregabalin-induced parkinsonism: A case report. Therapie 2017;72:395–6. [DOI] [PubMed] [Google Scholar]
  • [341].Olszewska DA, Chalissery AJ, Williams J, Lynch T, Smyth S. Speech myoclonus due to probable pregabalin adverse drug-reaction. Parkinsonism Relat Disord 2015;21:823–4. [DOI] [PubMed] [Google Scholar]
  • [342].Ozturk HM, Morkavuk G. Nasal pregabalin overdose and myclonus: A new way of misuse. Psychiatry Clin Psychopharmacology 2019;29:216–9. [Google Scholar]
  • [343].Park K, Kim M, Lee S. Negative myoclonus associated with pregabalin. Yeungnam Univ J Med 2018;35:240–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [344].Perez Lloret S, Amaya M, Merello M. Pregabalin-induced parkinsonism: A case report. Clin Neuropharmacol 2009;32:353–4. [DOI] [PubMed] [Google Scholar]
  • [345].Prado-Mel E, Gil-López M, Navarro-Corrales M. Pregabalin-induced parkinsonism 72 hours after iodinated contrast administration. J Pharm Pract Res 2018;48:368–71. [Google Scholar]
  • [346].Shimizu T, Yoshida T, Kitamura K, Hamada O. Disturbance of consciousness and involuntary movements caused by pregabalin. BMJ Case Rep 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [347].Wang Y, Chen Y-, Zhang Y- et al. Oxcarbazepine oral suspension in pediatric patients with partial seizures and/or generalized tonic-clonic seizures: A multi-center, single arm, observational study in china. World J Pediatr 2017;13:551–9. [DOI] [PubMed] [Google Scholar]
  • [348].Chung SS, Johnson JK, Brittain ST, Baroldi P. Long-term efficacy and safety of adjunctive extended-release oxcarbazepine (oxtellar XR®) in adults with partial-onset seizures; supemus(united states). Acta Neurol Scand 2016;133:124–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [349].Eun S-, Kim HD, Chung HJ et al. A multicenter trial of oxcarbazepine oral suspension monotherapy in children newly diagnosed with partial seizures: A clinical and cognitive evaluation. Seizure 2012;21:679–84. [DOI] [PubMed] [Google Scholar]
  • [350].Vieta E, Cruz N, García-Campayo J et al. A double-blind, randomized, placebo-controlled prophylaxis trial of oxcarbazepine as adjunctive treatment to lithium in the long-term treatment of bipolar I and II disorder. Int J Neuropsychopharmacol 2008;11:445–52. [DOI] [PubMed] [Google Scholar]
  • [351].Adamec I, Nanković S, Zadro I, Hajnšek S, Habek M. Oxcarbazepine-induced jerky see-saw nystagmus. Neurol Sci 2013;34:1839–40. [DOI] [PubMed] [Google Scholar]
  • [352].Gatzonis SD, Georgaculias N, Singounas E, Jenkins A, Stamboulis E, Siafakas A. Elimination of oxcarbazepine-induced oculogyric crisis following vagus nerve stimulation. Neurology 1999;52:1918–9. [DOI] [PubMed] [Google Scholar]
  • [353].Hergüner MÖ, Incecik F, Altunbaşak Ş. Oxcarbazepine-induced tardive dyskinesia: A rare adverse reaction. J Pediatr Neurosci 2010;5:85–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [354].Okuyucu EE, Duman T, Akcin E. Reversible parkinsonism with oxcarbazepine use. Parkinsonism Relat Disord 2009;15:787–8. [DOI] [PubMed] [Google Scholar]
  • [355].Rao S, Harper-Shankie M, Agarwal R. Vertical gaze palsy due to medication error. Epilepsy Behav Case Report 2017;8:33–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [356].Sanchez-Larsen A, Sopelana D, Diaz-Maroto I et al. Assessment of efficacy and safety of eslicarbazepine acetate for the treatment of trigeminal neuralgia. Eur J Pain 2018;22:1080–7. [DOI] [PubMed] [Google Scholar]
  • [357].Elger C, Koepp M, Trinka E et al. Pooled efficacy and safety of eslicarbazepine acetate as add-on treatment in patients with focal-onset seizures: Data from four double-blind placebo-controlled pivotal phase III clinical studies. CNS Neurosci Ther 2017;23:961–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [358].Gama H, Vieira M, Costa R, Graça J, Magalhães LM, Soares-da-Silva P. Safety profile of eslicarbazepine acetate as add-on therapy in adults with refractory focal-onset seizures: From clinical studies to 6 years of post-marketing experience; bial portela and c(portugal); sunovion(united states). Drug Saf 2017;40:1231–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [359].Gómez-Ibáñez A, Serratosa JM, Guillamón E et al. Efficacy and safety of eslicarbazepine-acetate in elderly patients with focal epilepsy: Case series. Seizure 2017;48:53–6. [DOI] [PubMed] [Google Scholar]
  • [360].Hufnagel A, Ben-Menachem E, Gabbai AA, Falcão A, Almeida L, Soares-da-Silva P. Long-term safety and efficacy of eslicarbazepine acetate as adjunctive therapy in the treatment of partial-onset seizures in adults with epilepsy: Results of a 1-year open-label extension study. Epilepsy Res 2013;103:262–9. [DOI] [PubMed] [Google Scholar]
  • [361].Krauss G, Biton V, Harvey JH et al. Influence of titration schedule and maintenance dose on the tolerability of adjunctive eslicarbazepine acetate: An integrated analysis of three randomized placebo-controlled trials. Epilepsy Res 2018;139:1–8. [DOI] [PubMed] [Google Scholar]
  • [362].Rocamora R, Peltola J, Assenza G, McMurray R, Villanueva V. Safety, tolerability and effectiveness of transition to eslicarbazepine acetate from carbamazepine or oxcarbazepine in clinical practice. Seizure 2020;75:121–8. [DOI] [PubMed] [Google Scholar]
  • [363].Sales F, Chaves J, McMurray R, Loureiro R, Fernandes H, Villanueva V. Eslicarbazepine acetate in post-stroke epilepsy: Clinical practice evidence from euro-esli. Acta Neurol Scand 2020;142:563–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [364].Sperling MR, Abou-Khalil B, Harvey J et al. Eslicarbazepine acetate as adjunctive therapy in patients with uncontrolled partial-onset seizures: Results of a phase III, double-blind, randomized, placebo-controlled trial. Epilepsia 2015;56:244–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [365].Villanueva V, Serratosa JM, Guillamón E et al. Long-term safety and efficacy of eslicarbazepine acetate in patients with focal seizures: Results of the 1-year ESLIBASE retrospective study. Epilepsy Res 2014;108:1243–52. [DOI] [PubMed] [Google Scholar]
  • [366].Algahtani HA, Aldarmahi AA, Al-Rabia M, Almalki WH, Bryan Young G. Generalized myoclonus and spasticity induced by lamotrigine toxicity: A case report and literature review. Clin Neuropharmacol 2014;37:52–4. [DOI] [PubMed] [Google Scholar]
  • [367].Alkawi A, Kattah JC, Wyman K. Downbeat nystagmus as a result of lamotrigine toxicity. Epilepsy Res 2005;63:85–8. [DOI] [PubMed] [Google Scholar]
  • [368].Alkin T, Onur E, Özerdem A. Co-occurence of blepharospasm, tourettism and obsessive-compulsive symptoms during lamotrigine treatment. Prog Neuro-Psychopharmacol Biol Psychiatry 2007;31:1339–40. [DOI] [PubMed] [Google Scholar]
  • [369].Centorino MB, Catalano G, Catalano MC. Lamotrigine induced whole body tics: A case report and literature review. Curr Drug Saf 2016;11:189–91. [DOI] [PubMed] [Google Scholar]
  • [370].Crespel A, Genton P, Berramdane M et al. Lamotrigine associated with exacerbation or de novo myoclonus in idiopathic generalized epilepsies. Neurology 2005;65:762–4. [DOI] [PubMed] [Google Scholar]
  • [371].Ebrahimi HA, Ebrahimi S. Dopa-responsive dystonia subsequent to lamotrigine administration: Case reports. Iran J Pharmacol Ther 2013;12:39–41. [Google Scholar]
  • [372].Fernández Corcuera P, Pomarol E, Amann B, McKenna P. Myoclonus provoked by lamotrigine in a bipolar patient. J Clin Psychopharmacol 2008;28:248–9. [DOI] [PubMed] [Google Scholar]
  • [373].Janszky J, Rásonyi G, Halász P et al. Disabling erratic myoclonus during lamotrigine therapy with high serum level - report of two cases. Clin Neuropharmacol 2000;23:86–9. [DOI] [PubMed] [Google Scholar]
  • [374].Jung I, Shin JH, Kim J. Gaze-evoked nystagmus associated with valproic acid-induced lamotrigine toxicity. Clin Neurol Neurosurg 2020;196. [DOI] [PubMed] [Google Scholar]
  • [375].Kim D, Oh S, Kim OJ. A case of lamotrigine-induced excessive involuntary eye blinking. J Clin Neurol 2007;3:93–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [376].Lombroso CT, Lombroso CT. Lamotrigine-induced tourettism. Neurology 1999;52:1191–4. [DOI] [PubMed] [Google Scholar]
  • [377].Lu D, Lin X, Liu X, Su L. Lamotrigine-induced facial tic in a pediatric bipolar disorder patient. J Child Adolesc Psychopharmacol 2013;23:583–4. [DOI] [PubMed] [Google Scholar]
  • [378].Marrero-Gonzalez P, Ruano OL, Catalano G, Catalano MC. Dystonia associated with lamotrigine therapy: A case report and review of the literature. Curr Drug Saf 2014;9:60–2. [DOI] [PubMed] [Google Scholar]
  • [379].Moreira B, Thomé-Souza S, Valente KDR. Late side-effects of valproate and lamotrigine. J Epilepsy Clin Neurophysiol 2007;13:187–9. [Google Scholar]
  • [380].Oh S, Kim JS, Lee YH, Lee AY, Kim J, Kim JM. Downbeat, positional, and perverted head-shaking nystagmus associated with lamotrigine toxicity. J Clin Neurol 2006;2:283–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [381].Sabolek M, Runge U. Lamotrigine induced dyskinesia in parkinson’s disease; bristol myers squibb(germany); glaxo SmithKline(germany); lundbeck(germany); orion(germany). Eur J Neurol 2010;17:e37. [DOI] [PubMed] [Google Scholar]
  • [382].Santens P, Claeys I, Vonck K, Boon P. Parkinsonism due to lamotrigine [9]. Mov Disord 2006;21:2269–70. [DOI] [PubMed] [Google Scholar]
  • [383].Sotero De Menezes MA, Rho JM, Murphy P, Cheyette S. Lamotrigine-induced tic disorder: Report of five pediatric cases. Epilepsia 2000;41:862–7. [DOI] [PubMed] [Google Scholar]
  • [384].Tombini M, Pellegrino G, Assenza G, Di Lazzaro V. De novo multifocal myoclonus induced by lamotrigine in a temporal lobe epilepsy case. J Neurol Sci 2017;373:31–2. [DOI] [PubMed] [Google Scholar]
  • [385].Veerapandiyan A, Gallentine WB, Winchester SA, Baker J, Kansagra SM, Mikati MA. Oculogyric crises secondary to lamotrigine overdosage. Epilepsia 2011;52:e4–6. [DOI] [PubMed] [Google Scholar]
  • [386].Verma A, Miller P, Carwile ST, Husain AM, Radtke RA. Lamotrigine-induced blepharospasm. Pharmacotherapy 1999;19:877–80. [DOI] [PubMed] [Google Scholar]
  • [387].Yetimalar Y, Seçkin M, Seçil Y, Başoǧlu M. Lamotrigine-induced bilateral ballism [4]. Mov Disord 2007;22:1832–3. [DOI] [PubMed] [Google Scholar]
  • [388].Zesiewicz TA, Sullivan KL, Hauser RA. Chorea induced by lamotrigine [1]. J Child Neurol 2006;21:357. [DOI] [PubMed] [Google Scholar]
  • [389].Musiek ES, Anderson CT, Dahodwala NA, Pollard JR. Facial tic associated with lamotrigine in adults. Mov Disord 2010;25:1512–3. [DOI] [PubMed] [Google Scholar]
  • [390].Yang J, Chung S, Kim J. Action tremor associated with lamotrigine monotherapy. J Mov Disord 2010;3:18–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [391].Kovács A, Farkas Z, Kelemen A, Juhos V, Szűcs A, Kamondi A. Lamotrigine induces tremor among epilepsy patients probably via cerebellar pathways. Tohoku J Exp Med 2019;248:273–84. [DOI] [PubMed] [Google Scholar]
  • [392].Rosenow F, Schade-Brittinger C, Burchardi N et al. The LaLiMo trial: Lamotrigine compared with levetiracetam in the initial 26 weeks of monotherapy for focal and generalised epilepsy - an open-label, prospective, randomised controlled multicenter study. J Neurol Neurosurg Psychiatry 2012;83:1093–8. [DOI] [PubMed] [Google Scholar]
  • [393].Fallah R, Akhavan KS, Golestan M. Efficacy and safety of lamotrigine in lennox-gastaut syndrome. Iran J Child Neurol 2009;3:33–8. [Google Scholar]
  • [394].Schindler F, Anghelescu IG. Lithium versus lamotrigine augmentation in treatment resistant unipolar depression: A randomized, open-label study. Int Clin Psychopharmacol 2007;22:179–82. [DOI] [PubMed] [Google Scholar]
  • [395].Sander JWAS, Trevisol-Bittencourt P, Hart YM, Patsalos PN, Shorvon SD. The efficacy and long-term tolerability of lamotrigine in the treatment of severe epilepsy. Epilepsy Res 1990;7:226–9. [DOI] [PubMed] [Google Scholar]
  • [396].Cardona AF, Rojas L, Wills B et al. Efficacy and safety of levetiracetam vs. other antiepileptic drugs in hispanic patients with glioblastoma. J Neuro-Oncol 2018;136:363–71. [DOI] [PubMed] [Google Scholar]
  • [397].Montazerlotfelahi H, Amanat M, Tavasoli AR et al. Levetiracetam for prophylactic treatment of pediatric migraine: A randomized double-blind placebo-controlled trial. Cephalalgia 2019;39:1509–17. [DOI] [PubMed] [Google Scholar]
  • [398].Gatto EM, Roca CU, Etcheverry JL, Fadel D. Levetiracetam-induced parkinsonism in a huntington disease patient. Clin Neuropharmacol 2006;29:303–4. [DOI] [PubMed] [Google Scholar]
  • [399].Yim SH, Choi YH, Heo K, Cho KH. A case of dyskinesia after levetiracetam administration. BMC Neurol 2019;19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [400].Zesiewicz TA, Sanchez-Ramos J, Sullivan KL, Hauser RA. Levetiracetam-induced parkinsonism in a huntington disease patient; UCB(united states). Clin Neuropharmacol 2005;28:188–90. [DOI] [PubMed] [Google Scholar]
  • [401].Brodie MJ, Fakhoury T, McDonough B et al. Brivaracetam-induced elevation of carbamazepine epoxide levels: A post-hoc analysis from the clinical development program. Epilepsy Res 2018;145:55–62. [DOI] [PubMed] [Google Scholar]
  • [402].Fonseca E, Guzmán L, Quintana M et al. Efficacy, retention, and safety of brivaracetam in adult patients with genetic generalized epilepsy. Epilepsy Behav 2020;102. [DOI] [PubMed] [Google Scholar]
  • [403].Steinhoff BJ, Bacher M, Bucurenciu I et al. Real-life experience with brivaracetam in 101 patients with difficult-to-treat epilepsy—A monocenter survey. Seizure 2017;48:11–4. [DOI] [PubMed] [Google Scholar]
  • [404].Steinig I, von Podewils F, Möddel G et al. Postmarketing experience with brivaracetam in the treatment of epilepsies: A multicenter cohort study from germany. Epilepsia 2017;58:1208–16. [DOI] [PubMed] [Google Scholar]
  • [405].Yates SL, Fakhoury T, Liang W, Eckhardt K, Borghs S, D’Souza J. An open-label, prospective, exploratory study of patients with epilepsy switching from levetiracetam to brivaracetam. Epilepsy Behav 2015;52:165–8. [DOI] [PubMed] [Google Scholar]
  • [406].Alonso-Navarro H, Jiménez-Jiménez FJ. Reversible tremor, myoclonus, and fasciculations associated with topiramate use for migraine. Clin Neuropharmacol 2006;29:157–9. [DOI] [PubMed] [Google Scholar]
  • [407].Ginsberg DL. Topiramate-induced facial myoclonus. Prim psychiatry 2007;14:22–3. [Google Scholar]
  • [408].Khalkhali M Topiramate-induced persistent eyelid myokymia. Case Rep Psychiatry 2016;2016:7901085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [409].Kutluay E, Pakoz B, Beydoun A. Reversible facial myoclonus with topiramate therapy for epilepsy [2]. Epilepsia 2007;48:2001–2. [DOI] [PubMed] [Google Scholar]
  • [410].Miller AD, Prost VM, Bookstaver PB, Gaines KJ. Topiramate-induced myoclonus and psychosis during migraine prophylaxis. Am J Health-Syst Pharm 2010;67:1178–80. [DOI] [PubMed] [Google Scholar]
  • [411].Romigi A, Izzi F, Placidi F, Sperli F, Cervellino A, Marciani MG. Topiramate-induced restless legs syndrome: A report of two cases [3]. J Neurol 2007;254:1120–1. [DOI] [PubMed] [Google Scholar]
  • [412].Romigi A, Vitrani G, D’Aniello A, Di Gennaro G. Topiramate-induced periodic limb movement disorder in a patient affected by focal epilepsy. Epilepsy Behav Case Report 2014;2:121–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [413].Bermejo PE. Restless legs syndrome induced by topiramate: Two more cases. J Neurol 2009;256:662–3. [DOI] [PubMed] [Google Scholar]
  • [414].Lin G, Lawrence R. Pediatric case report of topiramate toxicity. Clin Toxicol 2006;44:67–9. [DOI] [PubMed] [Google Scholar]
  • [415].Medrano-Martínez V, Pérez-Sempere A, Moltó-Jordá JM et al. Eyelid myokymia in patients with migraine taking topiramate. Acta Neurol Scand 2015;132:143–6. [DOI] [PubMed] [Google Scholar]
  • [416].Clark AM, Kriel RL, Leppik IE et al. Intravenous topiramate: Comparison of pharmacokinetics and safety with the oral formulation in healthy volunteers; janssen(united states). Epilepsia 2013;54:1099–105. [DOI] [PubMed] [Google Scholar]
  • [417].Hufnagel A, Kowalik A, Rettig K, Schreiner A, Schäuble B. Long-term assessment of topiramate for epilepsy: An open-label, single-arm, multicentre, prospective study in a naturalistic setting; janssen cilag(germany). Clin Drug Invest 2011;31:779–90. [DOI] [PubMed] [Google Scholar]
  • [418].Ramsay RE, Uthman B, Pryor FM et al. Topiramate in older patients with partial-onset seizures: A pilot double-blind, dose-comparison study. Epilepsia 2008;49:1180–5. [DOI] [PubMed] [Google Scholar]
  • [419].Lindley SE, Carlson EB, Hill K. A randomized, double-blind, placebo-controlled trial of augmentation topiramate for chronic combat-related posttraumatic stress disorder. J Clin Psychopharmacol 2007;27:677–81. [DOI] [PubMed] [Google Scholar]
  • [420].Mohammadianinejad SE, Abbasi V, Sajedi SA et al. Zonisamide versus topiramate in migraine prophylaxis: A double-blind randomized clinical trial. Clin Neuropharmacol 2011;34:174–7. [DOI] [PubMed] [Google Scholar]
  • [421].Cross JH, Auvin S, Patten A, Giorgi L. Safety and tolerability of zonisamide in paediatric patients with epilepsy. Eur J Paediatr Neurol 2014;18:747–58. [DOI] [PubMed] [Google Scholar]
  • [422].Fallah R, Divesalar S, Babaei A. The efficacy and safety of zonisamide as an add-on drug in the treatment of lennox-gastaut syndrome. Iran J Child Neurol 2010;4:45–50. [Google Scholar]
  • [423].Hubert K, Knake S, Bauer S, Voss M, Rosenow F, Strzelczyk A. Treatment of status epilepticus with zonisamide: A multicenter cohort study of 34 patients and review of literature. Epilepsy Behav 2020;109. [DOI] [PubMed] [Google Scholar]
  • [424].Schmidt D, Jacob R, Loiseau P et al. Zonisamide for add-on treatment of refractory partial epilepsy: A european double-blind trial; parke davis(germany). Epilepsy Res 1993;15:67–73. [DOI] [PubMed] [Google Scholar]
  • [425].Stephen LJ, Kelly K, Wilson EA, Parker P, Brodie MJ. A prospective audit of adjunctive zonisamide in an everyday clinical setting. Epilepsy Behav 2010;17:455–60. [DOI] [PubMed] [Google Scholar]
  • [426].Chen JT, Garcia PA, Alldredge BK. Zonisamide-induced restless legs syndrome. Neurology 2003;60:147. [DOI] [PubMed] [Google Scholar]
  • [427].Velasco PEB, Goiburu JAZ, Pinel RS. Restless legs syndrome induced by zonisamide [4]. Mov Disord 2007;22:1517–8. [DOI] [PubMed] [Google Scholar]
  • [428].Balestrini S, Sisodiya SM. Audit of use of stiripentol in adults with dravet syndrome. Acta Neurol Scand 2017;135:73–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [429].Inoue Y, Ohtsuka Y, Ikeda H et al. Long-term safety and efficacy of stiripentol for the treatment of dravet syndrome: A multicenter, open-label study in japan. Epilepsy Res 2015;113:90–7. [DOI] [PubMed] [Google Scholar]
  • [430].Inoue Y, Ohtsuka Y, Oguni H et al. Stiripentol open study in japanese patients with dravet syndrome. Epilepsia 2009;50:2362–8. [DOI] [PubMed] [Google Scholar]
  • [431].Myers KA, Lightfoot P, Patil SG, Cross JH, Scheffer IE. Stiripentol efficacy and safety in dravet syndrome: A 12-year observational study. Dev Med Child Neurol 2018;60:574–8. [DOI] [PubMed] [Google Scholar]
  • [432].Yıldız EP, Ozkan MU, Uzunhan TA et al. Efficacy of stiripentol and the clinical outcome in dravet syndrome. J Child Neurol 2019;34:33–7. [DOI] [PubMed] [Google Scholar]
  • [433].Dill P, Datta AN, Weber P, Schneider J. Are vigabatrin induced T2 hyperintensities in cranial MRI associated with acute encephalopathy and extrapyramidal symptoms? Eur J Paediatr Neurol 2013;17:311–5. [DOI] [PubMed] [Google Scholar]
  • [434].Jongsma MJ, Laan LAEM, Van Emde Boas W, Meinardi H. Reversible motor disturbances induced by vigabatrin [28]. Lancet 1991;338:893. [DOI] [PubMed] [Google Scholar]
  • [435].Dulac O, Chiron C, Luna D et al. Vigabatrin in childhood epilepsy. J Child Neurol 1991;6:2S30–7. [PubMed] [Google Scholar]
  • [436].Fong CY, Osborne JP, Edwards SW et al. An investigation into the relationship between vigabatrin, movement disorders, and brain magnetic resonance imaging abnormalities in children with infantile spasms. Dev Med Child Neurol 2013;55:862–7. [DOI] [PubMed] [Google Scholar]
  • [437].Matilainen R, Pitkanen A, Ruutiainen T, Mervaala E, Riekkinen P. Vigabatrin in epilepsy in mentally retarded patients. Br J Clin Pharmacol 1989;27:113S–8S. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [438].Matilainen R, Pitkanen A, Ruutiainen T, Mervaala E, Sarlund H, Riekkinen P. Effect of vigabatrin on epilepsy in mentally retarded patients: A 7-month follow-up study. Neurology 1988;38:743–7. [DOI] [PubMed] [Google Scholar]
  • [439].Tartara A, Manni R, Galimberti CA et al. Six-year follow-up study on the efficacy and safety of vigabatrin in patients with epilepsy. Acta Neurol Scand 1992;86:247–51. [DOI] [PubMed] [Google Scholar]
  • [440].Uldall P, Alving J, Gram L, Beck S. Vigabatrin in pediatric epilepsy - an open study. J Child Neurol 1991;6:2S38–44. [PubMed] [Google Scholar]
  • [441].Tombini M, Pacifici L, Passarelli F, Rossini PM. Transient athetosis induced by tiagabine. Epilepsia 2006;47:799–800. [DOI] [PubMed] [Google Scholar]
  • [442].Wolańczyk T, Grabowska-Grzyb A. Transient dystonias in three patients treated with tiagabine. Epilepsia 2001;42:944–6. [DOI] [PubMed] [Google Scholar]
  • [443].Krauss GL, Klein P, Brandt C et al. Safety and efficacy of adjunctive cenobamate (YKP3089) in patients with uncontrolled focal seizures: A multicentre, double-blind, randomised, placebo-controlled, dose-response trial. Lancet Neurol 2020;19:38–48. [DOI] [PubMed] [Google Scholar]
  • [444].Coppola G, Zamponi N, Kluger G et al. Rufinamide for refractory focal seizures: An open-label, multicenter european study. Seizure 2013;22:33–6. [DOI] [PubMed] [Google Scholar]
  • [445].McMurray R, Striano P. Treatment of adults with Lennox–Gastaut syndrome: Further analysis of efficacy and safety/tolerability of rufinamide; eisai. Neurol Ther 2016;5:35–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [446].Brodie MJ, Rosenfeld WE, Vazquez B et al. Rufinamide for the adjunctive treatment of partial seizures in adults and adolescents: A randomized placebo-controlled trial. Epilepsia 2009;50:1899–909. [DOI] [PubMed] [Google Scholar]
  • [447].Zupanc ML, Roell Werner R, Schwabe MS et al. Efficacy of felbamate in the treatment of intractable pediatric epilepsy. Pediatr Neurol 2010;42:396–403. [DOI] [PubMed] [Google Scholar]
  • [448].Hwang T-, Still CN, Jones JE. Reversible downbeat nystagmus and ataxia in felbamate intoxication. Neurology 1995;45:846. [DOI] [PubMed] [Google Scholar]
  • [449].Kerrick JM, Kelley BJ, Maister BH, Graves NM, Leppik IE. Involuntary movement disorders associated with felbamate. Neurology 1995;45:185–7. [DOI] [PubMed] [Google Scholar]
  • [450].Dehaene I, Van Vleymen B. Opsoclonus induced by phenytoin and diazepam. Ann Neurol 1987;21:216. [DOI] [PubMed] [Google Scholar]
  • [451].Schwankhaus JD, Kattah JC, Lux WE, Masucci EF, Kurtzke JF. Primidone/phenobarbital-induced periodic alternating nystagmus. Ann Ophthalmol 1989;21:230–2. [PubMed] [Google Scholar]
  • [452].Wu D, Thijs RD. Anticonvulsant-induced downbeat nystagmus in epilepsy. Epilepsy Behav Case Report 2015;4:74–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [453].Zaatreh M, Tennison M, D’Cruz O, Beach RL. Anticonvulsants-induced chorea: A role for pharmacodynamic drug interaction? Seizure 2001;10:596–9. [DOI] [PubMed] [Google Scholar]
  • [454].Thome-Souza S, Moreira B, Valente KD. Late adverse effects of the coadministration of valproate and lamotrigine. Pediatr Neurol 2012;47:47–50. [DOI] [PubMed] [Google Scholar]
  • [455].Chang RS-, Lui HKK, Lui HTC, Leung CYW, Leung YHI, Wang YO. Efficacy upon 12-weeks after achievement of maximal dose and tolerability of lacosamide as an adjunctive therapy in epilepsy: Real world clinical experience. J Neurol Sci 2020;409. [DOI] [PubMed] [Google Scholar]
  • [456].Egunsola O, Choonara I, Sammons HM. Safety of lamotrigine in paediatrics: A systematic review. 2015;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [457].Grisotto KP, Bruck I, Antoniuk SA, Santos LHC. Association of lamotrigine and valproate in refractory epilepsies of children and adolescents. Arq Neuro-Psiquiatr 2008;66:477–81. [DOI] [PubMed] [Google Scholar]
  • [458].Gustavson LE, Cato A III, Boellner SW et al. Lack of pharmacokinetic drag interactions between tiagabine and carbamazepine or phenytoin. Am J Ther 1998;5:9–16. [DOI] [PubMed] [Google Scholar]
  • [459].Moeller JJ, Rahey SR, Sadler RM. Lamotrigine-valproic acid combination therapy for medically refractory epilepsy. Epilepsia 2009;50:475–9. [DOI] [PubMed] [Google Scholar]
  • [460].Perucca P, Gilliam FG. Adverse effects of antiepileptic drugs. Lancet Neurol 2012;11:792–802. [DOI] [PubMed] [Google Scholar]
  • [461].Ney GC, Lantos G, Barr WB, Schaul N. Cerebellar atrophy in patients with long-term phenytoin exposure and epilepsy. Arch Neurol 1994;51:767–71. [DOI] [PubMed] [Google Scholar]
  • [462].Lalonde R Dopaminergic supersensitivity after long-term administration of phenytoin in rats. Epilepsia 1985;26:81–4. [DOI] [PubMed] [Google Scholar]
  • [463].Brugger F, Bhatia KP, Besag FM. Valproate-associated parkinsonism: A critical review of the literature. CNS Drugs 2016;30:527–40. [DOI] [PubMed] [Google Scholar]
  • [464].Okada M, Hirano T, Mizuno K et al. Biphasic effects of carbamazepine on the dopaminergic system in rat striatum and hippocampus. Epilepsy Res 1997;28:143–53. [DOI] [PubMed] [Google Scholar]
  • [465].Elphick M Effects of carbamazepine on dopamine function in rodents. Psychopharmacology (Berl) 1989;99:532–6. [DOI] [PubMed] [Google Scholar]
  • [466].Barros HM, Braz S, Leite JR. Effect of carbamazepine on dopamine release and reuptake in rat striatal slices. Epilepsia 1986;27:534–7. [DOI] [PubMed] [Google Scholar]
  • [467].Dailey JW, Reith ME, Yan QS, Li MY, Jobe PC. Carbamazepine increases extracellular serotonin concentration: Lack of antagonism by tetrodotoxin or zero Ca2+. Eur J Pharmacol 1997;328:153–62. [DOI] [PubMed] [Google Scholar]
  • [468].Pranzatelli MR. Serotonin and human myoclonus. rationale for the use of serotonin receptor agonists and antagonists. Arch Neurol 1994;51:605–17. [DOI] [PubMed] [Google Scholar]
  • [469].Rao ML, Clarenbach P, Vahlensieck M, Krätzschmar S. Gabapentin augments whole blood serotonin in healthy young men. J Neural Transm 1988;73:129–34. [DOI] [PubMed] [Google Scholar]
  • [470].Brawek B, Löffler M, Dooley DJ, Weyerbrock A, Feuerstein TJ. Differential modulation of K(+)-evoked (3)H-neurotransmitter release from human neocortex by gabapentin and pregabalin. Naunyn Schmiedebergs Arch Pharmacol 2008;376:301–7. [DOI] [PubMed] [Google Scholar]
  • [471].Luscombe G, Jenner P, Marsden CD. 5-HT-mediated myoclonus in the guinea pig as a model of brainstem 5-HT and tryptamine receptor action. Adv Neurol 1986;43:529–43. [PubMed] [Google Scholar]
  • [472].Pratt JA, Rothwell J, Jenner P, Marsden CD. P,p′-DDT-induced myoclonus in the rat and its application as an animal model of 5-HT-sensitive action myoclonus. Adv Neurol 1986;43:577–88. [PubMed] [Google Scholar]
  • [473].Sills GJ, Leach JP, Kilpatrick WS, Fraser CM, Thompson GG, Brodie MJ. Concentration-effect studies with topiramate on selected enzymes and intermediates of the GABA shunt. Epilepsia 2000;41:30–4. [DOI] [PubMed] [Google Scholar]
  • [474].Stahl JS, Averbuch-Heller L, Leigh RJ. Acquired nystagmus. Arch Ophthalmol 2000;118:544–9. [DOI] [PubMed] [Google Scholar]
  • [475].Furman JM, Wall C, Pang DL. Vestibular function in periodic alternating nystagmus. Brain 1990;113 (Pt 5):1425–39. [DOI] [PubMed] [Google Scholar]

RESOURCES