Drug-Induced Movement Disorders

25
PHARMACOEPIDEMIOLOGY Drug Safety 1997 Mar: 16 (3): 180-204 © Adis International Limited. All rights reserved. Drug-Induced Movement Disorders Felix Javier Jimenez-Jimenez,l Pedro Jose Garcia-Ruiz 2 and Jose Antonio Molina 3 1 Department of Neurology of Hospital Universitario 'Principe de Asturias', Alcala de Henares, Madrid, Spain 2 Department of Neurology of Fundaci6n Jimenez Diaz, Madrid, Spain 3 Department of Neurology of Hospital Universitario Doce de Octubre, Madrid, Spain Contents Summary 1. Parkinsonism .. 1 .1 Frequency. 1.2 Implicated Drugs 1.3 Clinical Features and Prognosis. 1.4 Prevention 1.5 Therapy ... . 2. Tremor ..... . 2.1 Antipsychotics. 2.2 Calcium Antagonists . 2.3 Other Drugs ..... . 3. Chorea ......... . 3.1 Antiparkinsonian Drugs. 3.2 Anticonvulsant Drugs. 4. Dystonia .......... . 4.1 Antipsychotic Drugs . . 4.2 Orthopramides and Substituted Benzamides . 4.3 Antiparkinsonian Drugs. 5. Tardive Dyskinesia .. 6. Myoclonus. . . . . . . . . . . 6.1 Dopamine Antagonists 6.2 Antidepressant Drugs and Lithium 6.3 Antiparkinsonian Drugs. 6.4 Bismuth Salts . 7. Tics or Tourettism ...... . 8. Akathisia ........ . 8.1 Dopamine Antagonists 8.2 Antidepressant Drugs 9. Conclusion ......... . 180 181 181 181 183 183 184 184 184 184 185 185 185 186 186 186 187 187 187 188 188 188 189 189 189 189 190 190 190 Summary Parkinsonism, tremor, chorea-ballismus, dystonia, tardive dyskinesia, myoc- lonus, tics and akathisia can be induced by many drugs. The drugs that are most frequently implicated in movement disorders are antipsychotics, calcium antag- onists, orthopramides and substituted benzamides (e.g. metoclopramide, sulpir- ide, clebopride, domperidone), eNS stimulants, antidepressants, anticonvulsants, antiparkinsonian drugs and lithium. It is possible for a single drug to induce 2 or

Transcript of Drug-Induced Movement Disorders

PHARMACOEPIDEMIOLOGY Drug Safety 1997 Mar: 16 (3): 180-204 0114~5916/97/0003-0180/$12.50/O

© Adis International Limited. All rights reserved.

Drug-Induced Movement Disorders Felix Javier Jimenez-Jimenez,l Pedro Jose Garcia-Ruiz2 and Jose Antonio Molina3

1 Department of Neurology of Hospital Universitario 'Principe de Asturias', Alcala de Henares, Madrid, Spain

2 Department of Neurology of Fundaci6n Jimenez Diaz, Madrid, Spain 3 Department of Neurology of Hospital Universitario Doce de Octubre, Madrid, Spain

Contents Summary 1. Parkinsonism ..

1 .1 Frequency. 1.2 Implicated Drugs 1.3 Clinical Features and Prognosis. 1.4 Prevention 1.5 Therapy ... .

2. Tremor ..... . 2.1 Antipsychotics. 2.2 Calcium Antagonists . 2.3 Other Drugs ..... .

3. Chorea ......... . 3.1 Antiparkinsonian Drugs. 3.2 Anticonvulsant Drugs.

4. Dystonia .......... . 4.1 Antipsychotic Drugs . . 4.2 Orthopramides and Substituted Benzamides . 4.3 Antiparkinsonian Drugs.

5. Tardive Dyskinesia .. 6. Myoclonus. . . . . . . . . . .

6.1 Dopamine Antagonists 6.2 Antidepressant Drugs and Lithium 6.3 Antiparkinsonian Drugs. 6.4 Bismuth Salts .

7. Tics or Tourettism ...... . 8. Akathisia ........ .

8.1 Dopamine Antagonists 8.2 Antidepressant Drugs

9. Conclusion ......... .

180 181 181 181 183 183 184 184 184 184 185 185 185 186 186 186 187 187 187 188 188 188 189 189 189 189 190 190 190

Summary Parkinsonism, tremor, chorea-ballismus, dystonia, tardive dyskinesia, myoc­lonus, tics and akathisia can be induced by many drugs. The drugs that are most frequently implicated in movement disorders are antipsychotics, calcium antag­onists, orthopramides and substituted benzamides (e.g. metoclopramide, sulpir­ide, clebopride, domperidone), eNS stimulants, antidepressants, anticonvulsants, antiparkinsonian drugs and lithium. It is possible for a single drug to induce 2 or

Drug-Induced Movement Disorders 181

more types of movement disorders in the same patient. Movement disorders are not always reversible after drug withdrawal.

Drug-induced movement disorders include par­kinsonism, tremor, chorea-ballismus, dystonia, tar­dive dyskinesia, myoclonus, tics, or akathisia. Such disorders can be caused by a wide variety of drugs, with parkinsonism, tardive dyskinesia and dysto­nia being the most frequent iatrogenic movement disorders found in clinical practice. Not infre­quently, 2 or more types of drug-induced move­ment disorder coexist in the same patient. This ar­ticle focuses on the distinct types of iatrogenic movement disorders and their associations with various drugs.

1. Parkinsonism

Major signs of parkinsonism include resting tremor, cogwheel rigidity, akinesia and postural instability. Until recently, the term drug-induced parkinsonism was nearly synonymous with anti­psychotic-induced parkinsonism. However, drug­induced parkinsonism is a frequent adverse effect of numerous drugs that interfere with dopamine function in the basal ganglia,[I-4) including calcium antagonists, orthopramides and substituted benza­mides.[2,5-8) Table I summarises the drugs that have been associated with induction or aggravation of parkinsonism.

1.1 Frequency

In Spain, several studies have reported that drug-induced parkinsonism accounts for 24 to 35% of parkinsonian syndromesp,5,7,8) The prevalence of drug-induced parkinsonism in a door-to-door survey conducted in Italy was 32.7 per 100 000 individuals, in other words, approximately 8 times less than that of idiopathic Parkinson's disease.[9) Another door-to-door survey conducted in Ger­many, but restricted to individuals older than 65 years, showed a prevalence of 0.41 % for drug­induced parkinsonism and 0.71 % for Parkinson's disease.! 10)

© Adls Internaflonal Umited. All rights reserved.

Table I. Drugs associated with induction or aggravation of parkinsonism

Drug Antipsychotics[l-l7]

Calcium antagonists: flunarizine, cinnarizine[7,8,18-31]

diltiazem,!32-34] verapamil,[33,35,38] amlodipine,!37] manidipine[38]

Orthopramides and substituted benzamides: metoclopramide[39-48] sulpiride,[1,7,8] clebopride[l,7,8,49-51]

cisapride,[52] domperidone,[53] veralipride!54,55]

Dopamine storage and transport inhibitors: reserpine, [56] tetrabenazine!57]

Antiemeticlantivertiginous agents: thiethylperazine!8,58,59] prochlorperazine[48]

Methyldopa[8,60,61]

Antidepressants:

Evidence

././

././

?

././

./

?

././

././

./

fluoxetine[62-69], paroxetine,!70,71] trazodone[72] ?

phenelzine[73] ?

amfebutomone (bupropion)[74] ? Lithium[75-79] ?

Diazepam[60] ?

Anticonvulsants: valproic acid (sodium valproate),[81-84] phenytoin[85] ?

Cholinergic agents bethanechol, [86] pyridostigmine!87] ?

Other agents: amiodarone,[88-91] procaine,[92] pethidine ? (meperidine),193,94] amphotericin B,[95,96] cefaloridine,[97] naproxen,[98] ethanol,[99] oral contraceptives, [1 DD] indeloxazine, [1 01,102] captopril,[103] cytarabine[104]

Symbols: ././ = well documented or frequent; ./ = relatively well documented or relatively frequent; ? = not well documented or anecdotal reports_

1 ,2 Implicated Drugs

1.2. 1 Antlpsychotlcs Parkinsonism has been a well known adverse

effect of antipsychotic drugs since their introduc­tion for the treatment of patients with psychiatric illnesses.! 11,12) Development of parkinsonism is not infrequent in antipsychotic users, especially in older individuals.[13-17) In our series of patients

Drug Safely 1997 Mar; 16 (3)

182

with drug-induced parkinsonism, based on patients who were referred to a Movement Disorders Unit, 'typical' antipsychotic drugs were implicated in 27.6% of those with parkinsonism.[8] Psychiatrists do not usually refer patients with mild parkinsonism to neurological clinics and, in addition, iatrogenic parkinsonism is not always recognised.[105,106] These 2 reasons may explain the relatively low fre­quency. In a lO-year series at a regional pharmaco­vigilance centre in France, antipsychotics were the offending drug in 39% of 53 patients with drug­induced parkinsonism.l6]

Antipsychotics act as dopamine receptor antag­onists. The development of drug-induced parkin­sonism is usually dose-dependent for each drug, and seems to be related to the blockade of dopa­mine D2 receptors, and with the degree of D2 re­ceptor occupancy in the striatum.l 107] Halogenated and piperazine phenothiazines, and butyrophen­ones, are the antipsychotics with the greatest like­lihood of producing this complication.l12,15, 108, 109] The phenothiazine derivatives thiethylperazine[8,58,59] and prochlorperazine,[46] which are used for the treatment of vertigo, may also induce parkinson­ism.

Although comparisons of absolute prevalence levels for parkinsonism and other movement disor­ders are difficult to make, the risk of developing these movement disorders with the newer 'atypical' antipsychotics (risperidone, remoxipride, clozapine) seems to be lower than that of the 'typical' ones.[lIO-1l71 This is probably linked with a lower affinity for D2 receptors: risperidone binds to sero­tonin (5-hydroxytryptamine; 5-HThA receptors with 20 times greater affinity than it binds to D2 receptors; remoxipride binds selectively, but weakly, to D2 receptors; and clozapine preferentially binds to D4 receptors.[l17]

Early age at onset of schizophrenia,[118] high ventricle: brain ratio,l1l8,119] and severity of negative symptoms of schizophrenia[l19,120] seem to be related to an increased risk and severity of antipsychotic­induced parkinsonism.

In a number of patients, the onset or aggravation of parkinsonian signs has occurred following anti-

© Adis International Lirnited. All rights reserved.

Jimenez-Jimenez et al.

psychotic withdrawaI.lI21,122] Melamed et al,l 1221 suggested that the antipsychotic may precipitate degeneration of vulnerable nigrostriatal neurons by generating cytotoxic free radicals or accelerating neuronal firing rates. Besides the blockade of do­pamine receptors, antipsychotics inhibit complex I of the mitochondrial electron-transport chain in vitro, and patients with antipsychotic-induced par­kinsonism showed decreased complex I activity in platelets.[123] This is a potentially interesting find­ing, since it is known that complex I activity is reduced in the substantia nigra of patients with Parkinson's disease.[124,125]

1.2.2 Calcium Antagonists Since the original description of cinnarizine­

induced parkinsonism by Marti-Masso et al. in 1985,[18] many authors have indicated that calcium antagonists, mainly cinnarizine and flunarizine, are a frequent cause of drug-induced parkinsonism, at least in Europe and South America,[7,8,19-30] where their use is common; cinnarizine was used by 5 to 7% of the elderly Spanish popUlation in 1985.[31] Cinnarizine and flunarizine, taken alone or with other drugs, were the most frequently reported causes of drug-induced parkinsonism in several Spanish series.l5,7,81 Other calcium antagonists have been associated with induction or aggravation of parkinsonism in anecdotal reports (table I).

Calcium antagonists were implicated in 58.1 % of cases of drug-induced parkinsonism in our own series,[8] and were the offending drug in 30% of cases of drug-induced parkinsonism in a lO-year series at a regional pharmacovigilance centre in France.l6] Old age and a family history of essential tremor seem to predispose to calcium antagonist­induced drug-induced parkinsonism.[29,30,126]

Garcia-Ruiz et al,l127] described an experimen­tal model of parkinsonism induced by cinnarizine in aged monkeys. Cinnarizine produced a persist­ent reduction of homo vanillic acid and 5-hydroxy­indoleacetic acid (5-HIAA) levels in the CSF. The authors suggested that this effect is the result of a predominant presynaptic effect of cinnarizine on dopaminergic and serotonergic neurons - resulting

Drug Safety 1997 Mar; 16 (3)

Drug-Induced Movement Disorders

in inhibition of synthesis or release of their respec­tive neurotransmitters - or a possible toxic effect.l127]

Several in vitro studies have shown that calcium antagonists decrease the synthesisl128] and re­leaseI129-13I] of dopamine from the striatum, inhibit [3H]spiperone binding to postsynaptic D2 recep­tors in the striatum,I13I,132] and reduce the levels of catecholamines and [3H] dopamine uptake, with­out neurotoxicity in dopamine-rich neuroblastoma cells.1133] The binding of metabolites offlunarizine and cinnarizine to striatal D2 receptors is higher than that of the parent drug,l134] In addition, like antipsychotics (section 1.2.1), flunarizine and cinnarizine inhibit mitochondrial complex 1,1135]

1.2.3 Orthopramides and Substituted Benzamides Several orthopramides and substituted benza­

mides can induce or aggravate parkinsonism (table I). Like antipsychotics, these drugs block striatal D2 receptors.

1.2.4 Dopamine Storage and Transport Inhibitors Methyldopa has been reported to induce parkin­

sonism in individual patients.18,60,61] Drug-induced parkinsonism is a well known adverse effect of drugs that interfere with the intraneuronal storage of dopamine, such as reserpinel56] and tetrabena­zine.l57] Tetrabenazine also may also block post­synaptic dopamine receptors.

1 .3 Clinical Features and Prognosis

The clinical manifestations of drug-induced parkinsonism are often indistinguishable from those of idiopathic Parkinson's disease, although drug-induced parkinsonism more frequently causes postural tremor and bilateral symptoms,I136] and often coexists with tardive dyskinesia, tardive dys­tonia and/or akathisia,l8,15,19,21,28,137-139]

It has been traditionally assumed that drug­induced parkinsonism was reversible; however, in many patients with parkinsonism-induced by anti­psychoticsl12l ,138,139] or other antidopaminergic drugs, such as flunarizine or cinnarizine,18,25,29,121] the parkinsonian symptoms and signs do not com­pletely disappear, despite withdrawal of the of­fending drug. The persistence of parkinsonian

© Adis International Umited. All rights reserved.

183

symptoms might reflect the fact that the patients have subclinical idiopathic parkinsonism, a condi­tion that was unmasked by the offending drug. On the other hand, this could also represent the onset of Parkinson's disease in the patients while they were taking the drug, and a direct toxic effect on the dopaminergic system cannot be ruled out.

1 .4 Prevention

The best prevention of drug-induced parkinson­ism (and other drug-induced movement disorders) is to avoid the inappropriate use of drugs that are capable of inducing these adverse effects. The pro­longed and, in many cases, unnecessary use of cinna­rizine or flunarizine in elderly patients in Spainl31] resulted in a high frequency of parkinsonism in­duced by these drugs.l5,7,8,18,25,29,30] In our own

drug-induced parkinsonism series, many patients were taking drugs to treat 'vertigo' or 'dyspepsia' for months or even years;18] also, there was fre­quent use of the antipsychotic drug flupenthixol, a thioxanthene that is marketed in combination with a benzodiazepine, with a trade name that could lead general physicians to suppose that he or she is pre­scribing an anxiolytic drug.

The choice and dosage of antipsychotic drugs in the treatment of patients with psychiatric disorders are crucial. It seems reasonable that the dosage of antipsychotic should be kept to the minimum re­quired to achieve and maintain a therapeutic re­sponse, and that those antipsychotics (risperidone, remoxipride and clozapine) with a lower risk of inducing drug-induced parkinsonism (or other movement disorders) should be preferred.

The use of adjunctive anticholinergic drugs to prevent the development of parkinsonism is con­troversial. A consensus statement by the World Health Organization did not recommend the use of these compounds because of their adverse effects, the potentially increased risk of developing tardive dyskinesia, and the fact that many patients taking antipsychotic therapy do not develop drug-induced parkinsonism. I 140,141]

Drug Safety 1997 Mar; 16 (3)

184

1.5 Therapy

The first therapeutic measure for patients with drug-induced parkinsonism should be the discon­tinuation of the offending drug, whenever possible. In those patients with persistent parkinsonian signs, especially elderly individuals with presumed sub­clinical idiopathic parkinsonism that has been un­masked by the offending drug, management should be similar to that of Parkinson's disease.

In psychiatric patients with antipsychotic­induced parkinsonism, the first approach should be to reduce the antipsychotic dosage or switch to an­other antipsychotic with a lower incidence of this adverse effect. If these measures are ineffective, it may be necessary to consider pharmacological treatment, including anticholinergics[l40,141] or amantadine.[140] Levodopa or direct dopamine ag­onists could increase psychotic symptomsY09] and are not useful in many patients.[142]

2. Tremor

Tremor is defined as a rhythmic oscillation of a body part, produced by either alternating or synch­ronous contractions of reciprocally innervated an­tagonistic muscles. Tremor is a common adverse effect of a wide variety of drugs (table II).

Although drugs that induce parkinsonism can produce a typical parkinsonian resting tremor, more characteristically, drug-induced tremors are pos­tural. [I 64] Antipsychotic drugs produce a low­frequency postural tremor, anticonvulsant drugs and lithium produce a high-frequency low-amplitude tremor, while other drugs may enhance physiolog­ical tremor.

2.1 Antipsychotics

Miller and Jankovic[15] reported that 2% of pa­tients with antipsychotic-induced movement disor­ders have postural tremor. Stacy and JankoviC[143] described 5 patients with a predominantly postural tremor ('tardive tremor') that had a frequency range of 3 to 5Hz (a type oftremor that is usually accom­panied by other tardive movement disorders). Moreover, tremor recordings in psychiatric pa-

© Adis internalionai Umited. All rights reserved.

Jimenez-Jimenez et al.

Table II. Drugs associated with induction or aggravation of postural tremor

Drug Antipsychotics[143-147J Lithium[76.146.149]

Anticonvulsant drugs[l50] Tricyclic antidepressants[151.152]

~-Adrenergic agonists[l53] Theophylline[l54] Levodopa[l54]

Amphetamines[l54] Thyroxine[154]

Antihyperglycaemic drugs[154] Caffeine[l54]

Corticosteroids[l54]

Calcium antagonists: flunarizine, cinnarizine[30]

Amiodarone[l54]

Anticonvulsants: phenytoin,1155] zonisamide,[156]lamotrigine[157J

Selective serotonin reuptake inhibitors[l56-160]

Histamine Hl antagonists[161]

Pentavalent antimonials[l62]

Evidence

.1.1 ,/,/

,/,/

,/,/

,/,/

,/,/

.1.1 ,/,/

,/,/

,/,/

,/,/

,/,/

,/

,/

? ?

?

? Cocaine[163] ?

Symbols: ,/,/ = well documented or not infrequent; ,/ = relatively well documented or relatively frequent; ? = not well documented or anecdotal reports.

tients treated with antipsychotics have shown that many of these patients had postural tremor of low frequency.[145] There have been reports of single cases of tongue tremor[l44] and rubral like trem­or[146] in patients exposed to antipsychotics. Finally, antipsychotics can induce postural tremor in all 4 limbs of guinea-pigs)l47]

2.2 Calcium Antagonists

Postural tremor seems to be a frequent clinical feature of calcium antagonist-induced parkinson­ism. In our experience, this type of parkinsonism improves after withdrawal of the offending drug, but a coarse mixed tremor persists for many months, despite disappearance of parkinsonism. [30] We suggest that this persistent tremor is the equiv­alent of the antipsychotic-induced 'tardive tremor' reported by Stacy and Jankovic (section 2.1))143] A

Drug Safety 1997 Mar; 16 (3)

Drug-Induced Movement Disorders

case of tongue tremor has been reported in a patient treated with flunarizine.[145]

2.3 Other Drugs

Tremor is a common problem in patients taking lithiump6] Studies show a wide range of incidences of lithium-induced tremor - from 4 to 65% - as a result of differences in defining and reporting this adverse effect.[l48] Tremor often decreases as dura­tion of lithium therapy increases.[l49]

Postural tremor appears in 20 to 25% of patients treated with valproic acid (sodium valproate),[145] and more infrequently, in patients treated with other anticonvulsant drugs (table II).

3. Chorea

Chorea refers to irregular, unpredictable, brief, jerky movements that flit from one part of the body to another in a random sequence. Table III summa­rises the drugs that can induce chorea. The chore­iform movements induced by dopamine antago­nists are part of the spectrum of tardive dyskinesia, and are discussed in section 5.

3.1 Antiparkinsonian Drugs

Choreiform dyskinesias are a well known ad­verse effect of levodopa,[165-171] dopamine ago­nists[l72-176] and, less frequently, of anticholinergic drugS[l85-188] and amantadineP26] Choreiform dys­kinesias associated with levodopa therapy in pa­tients with Parkinson's disease usually follow 2 patterns: 'peak-dose' and 'diphasic' (at the onset and the end of the beneficial effect).

3.1.1 Peak-Dose Dyskineslas Peak-dose dyskinesias can improve after reduc­

tion of the levodopa dosage, but this measure re­sults in increased parkinsonismP45.246] The redis­tribution of the total dose of levodopa to more frequent and lower individual doses, together with the addition of a dopamine agonist, or a change to a liquid formulation of levodopa, should be useful for the treatment of this pattern of dyskinesiasP47]

When the patient is treated simultaneously with anticholinergics, reduction of dosage or withdrawal

© Adis Internafional Umited. All rights reserved.

185

of these drugs should contribute to improvement of the dyskinesias. Recently, improvement of dys­kinesias has been reported with the addition of clozapine,[248] buspirone,[249.250] and fluoxetine}251] In contrast, cholinergic agents[252.253] and drugs that are active at y-aminobutyric acid (GAB A) sites[254.255] do not improve dyskinesias, and the lat­ter can aggravate parkinsonism.l256]

Isoniazid can improve dyskinesias, but also ag­gravates parkinsonian signs.l257] Improvement of levodopa-induced dyskinesias is one of the most im­portant benefits of posteroventral pallidotomy. [258-260]

3.1.2 Diphasic Dysklnesias Diphasic dyskinesias, which can be choreiform

or dystonic, are present at the beginning and at the end of the levodopa response cycle, coinciding

Table III. Drugs associated with induction of chorea

Drugs

Dopamine antagonists (including antipsychotics)

Dopamine agonists: levodopa,[165-171] direct dopamine agonists]172-176]

eNS stimulants:

Evidence ,/,/

,/,/

amphetamines,[177-180] pemoline.]181] ,/,/ methylphenidate, [182] cocaine, [163.183] xanthines]184]

Anticholinergics]185-188] ,/

H1 antihistamines]189] ,/

H2 antihistamines[190] ,/ Oral contraceptives[191-201] ,/

Anticonvulsants: phenytoin[202-209] ,/

phenobarbital (phenobarbitone),[202] ? carbamazepine,[210,211] ethosuximide,[212] valproic acid (sodium valproate),[213] felbamate,[214] gabapentin[215]

Withdrawal of diazepam + pentobarbital[216] ?

Antidepressants: tricyclic antidepressants,[217-219] fluoxetine[220] ?

Lithium[221-225] ?

Other drugs: amantadine,[226] anabolic steroids,[227] ? methadone,[228] baclofen,[229] benzodiazepines,[230] methyldopa, [231,232] digoxin, [233.234] diazoxide,l235] gold salts,1236-238] cyproheptadine,1239] cyclosporin,[240] sulfasalazine,[241] aciclovir,[242] propofol[243.244]

Symbols: ,/,/ = well documented or frequent; ,/ = relatively well documented or relatively frequent; ? = not well documented or anecdotal reports_

Drug Safety 1997 Mar: 16 (3)

186

with the rise and fall of plasma levodopa concen­trationsP61,262] The management ofthis pattern of dyskinesias is very difficult. The best measure is to administer more frequent doses of levodopa, in an attempt to overlap the end-of-dose period with the beginning of the levodopa response cycle, and the addition of a dopamine agonist.[246,247,262] Admin-istration of subcutaneous apomorphine at the be­ginning of the levodopa response cycle decreases the duration, but not the intensity of the dyskine­siasP63,264]

3.2 Anticonvulsant Drugs

Patients who are treated with anticonvulsant drugs, usually at toxic concentrations, can develop chorea. The anticonvulsant drug that has most fre­quently been associated with chorea is pheny­toin,[202-209] although this adverse effect has been also reported with phenobarbital (phenobarbi­tone), [202] carbamazepine, [210,211] ethosuximide, [212] valproic acid,[213] felbamate[214] and gabapen­tin,[215] and after withdrawal of combined therapy with pentobarbital and diazepamPl6]

4. Dystonia

Dystonia is a syndrome dominated by sustained muscle contractions, frequently causing twisting and repetitive movements or abnormal postures. Acute and tardive (or chronic) dystonia can be in­duced by the drugs that are listed in table IV.

4.1 Antipsychotic Drugs

Antipsychotic drugs can induce acute and, less frequently, tardive dystonia. First reported by Burke et al. in 1982,[265] tardive dystonia usually develops within 6 years of starting antipsychotic treat­ment, [266] although it can appear after a short period of exposure. Miller and lankovicf l5] reported that 24% of patients with antipsychotic-induced move­ment disorders have dystonia. The frequency of de­velopment of antipsychotic-induced acute and tar­dive dystonia has been reported to range from 5.3 to 66.7%,[267-271] and 0.4 to 21 %,[272-276] respec-

© Adls International Limited. All rlghts reserved.

Jimenez-Jimenez et al.

tively. These wide ranges result from the various diagnostic criteria that are used.

Butyrophenones, and halogenated and pipera­zine phenothiazines, are the antipsychotics with the greatest likelihood of producing these compli­cations, but the newer 'atypical' antipsychotics, such as risperidone and clozapine, are not devoid of this adverse effect.[117,339-345] The phenothiazine thi­

ethylperazine, which is used for the therapy of ver­tigo,[298,346,347] and the butyrophenone droperidol,

used in epidural anaesthesia,[348] can also induce dystonia.

Table IV. Drugs associated with induction of acute and/or tardive dystonia

Drug Evidence Antipsychotic drugs[lS.265.276] ,/,/

Orthopramides and substituted benzamides: metoclopramide[45-47,2n] ,/,/

su]piride,[278-28l] tiapride,[279] cisapride,[282] ? domperidone, [283,284] vera]ipride[28S]

Dopamine agonists: ]evodopa[170,17l ,286-289] ,/ ,/

Direct dopamine agonists[290] ,/

Antidepressants: selective serotonin reuptake inhibitors[291-301] ,/

tricyclic antidepressants,[302.303] monoamine oxidase inhibitors[304,30S]

Anticonvu]sants:

?

carbamazepine,[306-308] phenytoin[309] ,/

phenobarbital (phenobarbitone)[310] ?

Anticholinergics[311] ?

Anxiolytics: buspirone,[293.300,3l2] diazepam,[3l3] ? bromazepam,[314] midazolam[31S]

CNS stimulants: amphetamines,[3l6.3l7] methylphenidate,[3l8] ? cocaine (and cocaine withdrawal)[163.319-323]

anti migraine drugs ergotamine,[324,32S] sumatriptan[326] ?

Calcium antagonists: cinnarizine,[8.28] flunarizine,[8.28] nifedipine[327] ?

Histamine antagonists (H1 and H2)[328-332] ?

Other drugs: a-Methylparatyrosine,[333] tetrabenazine,[S7] ? bethanechol chloride,[334] flecainide,[33S] chloroquine,[338] etoposide,[337] alfentanil[338]

Symbols: ,/,/ = well documented or frequent; ,/ = relatively well documented or relatively frequent; ? = not well documented or anecdotal reports.

Drug Safely 1997 Mar; 16 (3)

Drug-Induced Movement Disorders

Risk factors for the development of acute and tard­ive dystonia in patients receiving antipsychotic treat­ment include younger age,l240,245,246,265,270,27 1,349-352] male gender, [271,349,353] severity of schizophrenia, [271] and the presence of negative symptoms of schizo­phrenia at baseline.l271 ] Although some authors have reported that patients with mania were more likely to develop antipsychotic-induced dystonia than those with schizophrenia,[267,269] others have found no such differences.[267] Concomitant treat­ment with anticholinergics seems to prevent the development of dystonia. [270]

4.2 Orthopramides and Substituted Benzamides

Several orthopramides and substituted benz a­rnides, such as sulpiride,[278-281] tiapride,[279] meto­cloprarnide,[45,47,277] cisapride,[282] domperidone,[283,284]

and veralipride, a drug used in the treatment of postmenopausal symptoms, can induce tardive dystonia. [285]

4.3 Antiparkinsonian Drugs

Dystonia can be induced by levodopa,[170,171,286-289] anticholinergic agents[311] and dopamine agonists.[290] Dystonias associated with levodopa therapy can follow 3 patterns: 'early morning', 'peak-dose' and 'off-period' dystonias. The therapy of peak-dose dystonia is similar to that previously described for peak-dose choreiform dyskinesias (section 3.1).

Off-period dystonias usually affect the toes or legs, and less frequently, the face and neck.[260,289] They are usually painful, are asscciated with low plasma levodopa concentrations and coincide with the 'off' periods of motor fluctuationsP60]

In certain patients, painful dystonia of the lower extremities is a particular problem on waking in the morning (early morning dystonia). Early morning dystonia can be avoided by administering a dose of sustained-release carbidopa/levodopa or a long­acting dopamine agonist at bedtime, or a dose of standard carbidopa/levodopa in the early morning or on waking.[246] Subcutaneous apomorphine,[354] oral lithium,[355,356] baclofen,[356] anticholiner­gics[356] or benzodiazepines, or (if dystonia is sus-

© Adis International Limited. All rights reserved.

187

tained) botulinum toxin injections,[356] can be use­ful alternatives.

5. Tardive Dyskinesia

The term 'tardive dyskinesia' is used to describe all persistent, occasionally reversible, abnormal in­voluntary movements caused by prolonged expo­sure to anti psychotics or other dopamine antago­nists. The most common form of tardive dyskinesia presents as orofacial stereotypy ('buccolinguo­masticatory syndrome'), at times associated with choreatic movements of the extremities and trunkJ357] In this section, we will review the data regarding this common form of tardive dyskinesia. Other tardive syndromes such as tremor (section 2), dystonia (section 4), myoclonus (section 6) and akathisia (section 8) are described elsewhere in this review.

Tardive dyskinesia was first described by Faur­bye et al. in 1964.[358] It usually develops after a minimum of 1 to 2 years of antipsychotic therapy. Tardive dyskinesia accounted for 14% of patients with drug-induced movement disorders in a French study at a regional pharmacovigilance centre[359] and 63% of such patients referred to a Movement Disorders Unit in the US.[15]

The prevalence of tardi ve dyskinesia is reported to range from 3.7% and 60%, depending on demo­graphic factors, inclusion criteria, and study de­signP76,360-384] Studies in Asiatic countries have shown low prevalence ratesP76,369,370]

The newer atypical anti psychotics risperidone, remoxipride, and clozapine, are associated with a lower incidence of tardive dyskinesia than typical anti psychotics, but are not devoid of this adverse effect. [114, 117,342,385-390] The anti vertiginous pheno-

thiazine thiethylperazine can also induce tardive dyskinesia.l59] Besides antipsychotics, other drugs can induce tardive dyskinesia (table V).

Proposed risk factors for tardive dyskinesia in­clude: • advanced age[36I,362,368,382,384,435-437] • female gender[365,369,380,437-439]

• prolonged duration of exposure to antipsychot­ics[362,363,365,381]

Drug Safety 1997 Mar: 16 (3)

188

• high antipsychotic dosage, and use of 'depot' antipsychotics and high potency antipsychot­iCS[361,384,439]

• presence of subtle movement disorders before initiation of antipsychotic therapy[380,384,440]

• diabetes mellitus[380,440-442] • 'organic' brain dysfunction[369,380,443] and brain

atrophy [444]

• affective disorders,[365,380,436] especially in pa­tients taking concomitant lithium therapy[445]

• negative schizophrenic symptoms[446] and a poor prognosis of schizophrenia[363]

• family history of mental illness[372] and • alcohol abuse/dependence)384,447]

There is debate over whether early or late onset psychosis is a risk factor for tardive dyskinesia.[448] The concomitant use of anticholinergic drugs does not seem to increase the risk of tardive dyskine­sia. [368.449]

Preventive measures against tardive dyskinesia are similar to those for drug-induced parkinsonism (section 1.4). Drugs that have been used or pro-

Table V. Drugs associated with the development of tardive dyskinesia

Drugs Evidence

Antipsychotic drugs

Orthopramides and substituted benzamides: metoclopramideI42,4S-47,391-401] ,(,(

clebopride,[SO,Sl] su[piride,[403,404J veralipride[40SJ ?

Calcium antagonists: flunarizine, cinnarizine[19,21 ,28,402J ,(

Antidepressants:

cyclic antidepressants[406-414J ,(

fluoxetine,[41S] paroxetine,[416] fluvoxamine[417,418] ? Lithium[419-421]

Anxiolytics: buspirone,[312,422-42S] lorazepam[426,427]

Anticholinergics:

carbamazepine, [420J mesuximide (methsuximide )[428J

Anticholinergic agents[429,430]

Other drugs:

?

?

?

?

pethidine (meperidine),1431] pemoline,1432] ? histamine Hl antagonists,[433] clembuterol1434]

Symbols: ,( ,( = well documented or frequent; ,( = relatively well documented or relatively frequent; ? = not well documented or anecdotal reports_

© Adis International Limited. All rights reserved,

Jimenez-Jimenez et al.

posed for use in the treatment of patients with tar­dive dyskinesia, such as antidopaminergics, choli­nomimetics, GABA agonists, calcium antagonists, buspirone, clozapine and tocopherol, have been ex­tensively reviewed by Cavallaro and Smeraldi.[450]

6. Myoclonus

Myoclonus is defined as a sudden, brief, shock­like involuntary movement caused by active mus­cular contractions.l45 ]] Asterixis, which results from brief pauses in muscular activity causing pos­turallapses, may be regarded as a form of 'negative myoclonus' .[451] Drugs that may induce myoclonus are listed in table VI.

6,1 Dopamine Antagonists

Several types of dopamine-blocking agents, such as antipsychotics[452-457] (including the atypical antipsychotic clozapine)[496,541-544] and metoclo­pramide[277,545] have been reported to induce my­oclonus.

Tominaga et aI,[455] described 'tardive myoclo­nus' as persistent postural myoclonus resulting from a late complication of antipsychotic treat­ment. Later on, the same group reported that 38% of patients taking long term antipsychotic therapy developed postural myoclonus, which was more frequent in male patients. In addition, patients with myoclonus had been given significantly higher doses of antipsychotics than those without myoc­lonus.[456] Little and Jankovic reported tardive my­oclonus within 5 months of antipsychotic with­drawal.[457]

6.2 Antidepressant Drugs and Lithium

Cyclic antidepressant drugs, alone or in combi­nation with lithium, can induce reversible action myoclonus, even when used at nontoxic dos­ages.l452,453,458-469] Garvey et aI,[462] reported that

30% of patients treated with cyclic antidepressants developed myoclonus, although in only 9% was this adverse effect clinically significant. Myoclo­nus may be associated with electroencephalo­graphic abnormalities[461] and with enlarged corti-

Drug Safety J 997 Mar: 16 (3)

Drug-Induced Movement Disorders

cal somatosensory evoked potentials,[467] although such abnormalities were not found by other re­searchers in a single-patient study.[465]

Other drugs that can also induce myoclonus are selective serotonin reuptake inhibitors (SSRIs),[470-474] monoamine oxidase inhibitors (MAOIs)[471,475-477] and lithium.[464,496-499]

6.3 Antiparkinsonian Drugs

Myoclonus[478,479] and asterixis[480] can occur as a

adverse effect of long term levodopa therapy; it usu­ally involves the legs or trunk, and occurs at night. In rare instances, bromocriptine can induce myo­clonus.[516]

6.4 Bismuth Salts

Abuse of bismuth can induce a toxic encepha­lopathy with myoclonus.l481-491] Although this en­cephalopathy improves after drug withdrawal, a 10% rate of mortality and/or tardive psychic dis­abilities has been reported.l482] Neurochemical findings include increased CSF 5-HIAAlevels.[485] The metal chelator dimercaprol increases the renal clearance and improves the clinical signs of bismuth induced encephalopathy.l483,484,487]

7. Tics or Tourettism

Tics are recurrent, involuntary, repetitive, rapid, purposeless movements affecting various muscle groups (most often in the face or the head) or pro­ducing vocalisations. Since the original descrip­tion by Klawans et al. in 1978,[546] there have been a number of additional reports of tics or tourettism after exposure to antipsychotic drugs,[547-561] in­cluding the atypical antipsychotic clozapine.l562] In addition, tardive tourettism has been induced in animal models after prolonged antipsychotic expo­sure.l563]

There have been published anecdotal reports of induction of tics by drugs other than antipsychot­ics, including: CNS stimulants, such as cocaine,[163.564-566] or amphetamines, methylphenidate or pemo­line, which are used for the treatment of attention deficit hyperactivity disorder;[557,567-5721 opiate ther-

© Adls Internofionol Umlted. All rights reserved.

189

Table VI. Drugs that can induce myoclonus

Drug Evidence Antipsychotics[452-457]

Antidepressants: cyclic antidepressantsI452,453,45B-469]

selective serotonin reuptake inhibitors,[470-474] monoamine oxidase inhibitors[471,475-477]

Levodopa[478-480]

Bismuth salts[481-491]

Anticanvulsants:[492-495]

.1.1

.1.1

.1.1

valproic acid (sodium valproate), carbamazepine, .!.! phenytoin

Lithium[464,496.499] .!

Morphine or its derivatives, administered .! intravenously or intrathecallyl500-510]

Antineoplastic drugs[511.515] .!

Bromocriptine[516] ?

Anxiolytics: buspirone,[472,517] lorazepam,[518] midazolam[519] ?

Cardiovascular drugs: propafenone,[520.522] flecainide,[523] diltiazem,[524] ? nifedipine,[525] buflomedil[526]

Antibacterials: cefmetazole,[527] penicillin,[528-531] pefloxacin,[532] ? isoniazid[533]

Other drugs: veratramine,[534] propofol,[535,536] ? physostigmine,[537] norpethidine,[538] pseudoephedrine,[539] tryptophan[540]

Symbols: .!.! = well documented or frequent; .! = relatively well documented or relatively frequent; ? = not well documented or anecdotal reports,

apy,[5731 opiate withdrawal[5741 and withdrawal of opiate antagonist therapy;[5751 anticonvulsants, such as carbamazepine or phenobarbital;[576-5791 clonazepam;[580] and the SSRI fluoxetine.l581,5821 Levodopa was reported to exacerbate tics in a pa­tient with coexistent Gilles de la Tourette syn­drome and Parkinson's disease.[5831 The present au­thors have observed a patient with Parkinson's disease who developed motor and vocal tics during a treatment with trihexyphenidyl; the tics disap­peared after withdrawal of this drug (unpublished observation).

8. Akathisia

Akathisia is a subjective sensation of restless­ness, often associated with an inability to keep still

Drug Safety 1997 Mor; 16 (3)

190

or with unpleasant subjective promptings to moveJ584] Akathisia may be confused with a sim­ilar disorder described as 'restless legs syndrome' , which is characterised by sensory symptoms lo­cated mainly in the calves, primarily at rest, and associated with restlessness in the legs that leads to irresistible urge to move themJ584] Drugs that have been associated with induction of akathisia are summarised in table VII.

8.1 Dopamine Antagonists

Acute and tardive akathisia are well known ad­verse effects of antipsychotic therapy. The preva­lence of akathisia among antipsychotic-treated pa­tients ranges from 8 to 76%, the wide variation reflecting different criteria for definition, and vari­ations in the type and dosage of antipsychotic drugs.[l6,585-590] Akathisia accounted for 7% of drug-induced movement disorders in a Movement Disorders Unit.115] The newer atypical antipsychot­ics, risperidone, remoxipride and clozapine, seem to have a lower risk of inducing akathisia than typ­ical antipsychotics.11l6,1l7,591-596] Epidural droperidol can also induce akathisiaJ597,598]

The potential risk factors for antipsychotic­induced akathisia are not well established, although they are likely to include dosage, rate of dosage in­crements and potency of the antipsychoticJ588,589] The role of old age, female gender, negative symp­toms or cognitive dysfunction associated with schizophrenia, and a diagnosis of affective disorder are not well known.1590] The suggestion by a num­ber of investigatorsI638-64I] that decreased serum iron levels and percentage of saturation were re­lated to the risk of antipsychotic-induced akathisia has not been confirmed by others.1590,642,643]

Antiemetics with dopamine-blocking action (mainly metoclopramide) are also associated with the in­duction of akathisia.£38,4o,403,609-615]

8.2 Antidepressant Drugs

Akathisia can be induced by tricyclic antide­pressants,1151,621-623] MAOIs such as phenelzinel624] and moclobemide,1625] and (more frequently) by serotonergic agents, including fluoxetine,I66,605-6l2]

© Adis Intematlonal Limited. All rights reserved.

Jimenez-Jimenez et al.

Table VII. Drugs associated with induction of akathisia

Drug Evidence Antipsychotics[117.585.5981 .1.1 Metoc[opramide[45,47,400,599-6041 .1.1

Dopamine storage and transport inhibitors:

a-methyltyrosine, reserpine, tetrabenazine[584] .1.1 Levodopa and dopamine agonists[584] .I

Antidepressants:

selective serotonin reuptake inhibitors, [66,298,605-620] tricyclic antidepressants[151,621-623]

phene[zine,[624] moc[obemide[625]

Calcium antagonists: f[unarizine, cinnarizine[19,21,27,28,626,627]

Lithium[628,629]

Anxio[ytics: buspirone,[517.630] midazolam,[631-633] iorazepam[634]

Anticonvu[sants: phenytoin,[635] va[proic acid (sodium va[proate )[6361

Methysergide[637]

Pethidine (meperidine)[625]

?

.I ?

?

?

?

?

Symbols: .1.1 = well documented or frequent; .I = relatively well documented or relatively frequent; ? = not well documented or anecdotal reports.

sertraline,[298,613-617] paroxetine,[618,619] and venla­

faxine.l603,620] However, akathisia associated with serotonergic drugs is a controversial subject, and some authors consider that this may be a variant of the 'serotonin syndrome' J520,525]

9. Conclusion

Many distinct types of drugs can induce move­ment disorders. The most frequent types of drug­induced movement disorders are parkinsonism (24 to 35% of cases of parkinsonism in Spain are induced or unmasked by drugs), tardive dyskinesia and dys­tonia. The drugs that are more frequently involved include antipsychotics, calcium antagonists, ortho­prarnides and substituted benzarnides, eNS stimu­lants, antidepressants, anticonvulsants, antiparkin­sonian drugs and lithium. More than one type of movement disorder can coexist in the same patient. Many of these movement disorders improve after withdrawal of the offending drug.

Drug Safety 1997 Mar: 16 (3)

Drug-Induced Movement Disorders

Acknowledgements

This work was supported in part by grant FIS 95/0269.

References I. Marti-Masso JF. Carrera N, Urtasun M. Newer drugs inducing

parkinsonism [abstract]. 9th International Symposium on Parkinson's disease: 1988 Jun 5-9; Jerusalem, Israel: 137

2. Marti-Masso JF, Carrera N, Urtasun M. Drug-induced parkin­sonism: a growing list [letter]. Mov Disord 1993; 8: 125

3. Gershanik OS. Drug-induced movement disorders. Curr Opin Neurol Neurosurg 1993; 6: 369-76

4. Gershanik OS. Drug-induced parkinsonism in the aged: recog­nition and prevention. Drugs Aging 1994; 5: 127-32

5. Perez-Gilabert Y, Mateo D, Gimenez-Roldan S. Actividad as­istencial en una consulta hospital aria especializada en en­fermedad de Parkinson y transtornos del movimiento: un estudio prospectivo durante anos. Neurologia (Spain) 1994; 9: 317-23

6. Llau ME, Nguyen L, Senard JM, et al. Syndromes parkin­soniens d'origine medicamenteuse: experience d'un centre regional de pharmacovigilance sur dix ans. Rev Neurol (Paris) 1994; 150: 757-76

7. Marti-Masso JF, Poza J1. Parkinsonismo inducido 0 agravado por farrnacos: caracteristicas clinicas y evolucion historica de los farrnacos implicados. Neurologia (Spain) 1996; II: 10-5

8. Jimenez-Jimenez FJ, Orti-Pareja M, Ayuso-Peralta L, et al. Drug-induced parkinsonism in a movement disorders unit. A four-year survey. Parkinsonism Rei Disord 1996; 2: 145-9

9. Morgante L, Rocca WA, Di Rosa AE, et al. Prevalence of par­kinson's disease and other types of parkinsonism: a door-to­door survey in three Sicilian municipalities. The Sicilian neuro-epidemiologic study (SNES) group. Neurology 1992; 42: 1901-7

10. Trenkwalder C, Schwarz J, Gebhard J, et al. Starnberg trial on epidemiology of parkinsonism and hypertension in the elder­ly. Prevalence of parkinson's disease and related disorders assessed by a door-to-door survey of inhabitants older than 65 years. Arch Neurol 1995; 52: 1017-22

II. Hubble JP. Drug-induced parkinsonism. In: Stern MB, Koller WC, editors. Parkinsonian syndromes. New York: Marcel Dekker Inc., 1993: 111-22

12. Ayd Jr FJ. A survey of drug-induced extrapyramidal reactions. JAMA 1961; 175: 1054-60

13. Murphy JE, Stewart RB. Efficacy of antiparkinsonian agents in preventing antipsychotic-induced extrapyramidal symptoms. Am J Hosp Pharm 1979; 36: 641-4

14. Binder RL, Kazamatsuri H, Nishimura T, et al. Tardive dyski­nesia and neuroleptic-induced parkinsonism in Japan. Am J Psychiatry 1987; 144: 1494-6

15. Miller LG, Jankovic J. Neurologic approach to drug-induced movement disorders: a study of 125 patients. South Med J 1990; 8: 525-32

16. Ganzini L, Heintz R, Hoffman WF, et al. Acute extrapyramidal syndromes in neuroleptic-treated elders: a pilot study. J Geriatr Psychiatry Neuro11991; 4: 222-5

17. Avorn J, Bohn RL, Mogun H, et al. Neuroleptic drug exposure and treatment of parkinsonism in the elderly: a case-control study. Am J Med 1995; 99: 48-54

18. Marti-Masso JF, Carrera N, de la Puente E. Posible parkin­sonismo por cinaricina. Med Clin (Barc) 1985; 85: 614-6

© Adis International limited. All rights reserved.

191

19. Chouza C, Scarameli A, Caamano JL, et al. Parkinsonism, tar­dive dyskinesia, akathisia, and depression-induced by flunarizine. Lancet 1986; I: 1300-4

20. Di Rosa AE, Morgante L, Meduri M, et al. Parkinson-like side effects during prolonged treatment with flunarizine. Funct Neurol 1987; 2: 47-50

21. Micheli F, Pardal MF, Gatto M, et al. Flunarizine- and cinnariz­ine-induced extrapyramidal reactions. Neurology 1987; 7: 881-4

22. De Michele G, Filla A, Coppola N, et al. Extrapyramidal side­effects of flunarizine. Acta Neurol [Quad] (Napoli) 1987; 9: 230-3

23. Fernandez-Pardal M, Fernandez-Pardal J, Micheli F. Aggrava­tion of parkinson's disease by cinnarizine [letter]. J Neurol Neurosurg Psychiatry 1988; 51: 158-9

24. Moretti A, Lucantoni C. Flunarizine-induced parkinsonism: clinical report. Ital J Neurol Sci 1988; 9: 295-7

25. Capella D, Laporte JR, Castel JM, et al. Parkinsonism, tremor, and depression-induced by cinnarizine and flunarizine [let­ter]. BMJ 1988; 297: 722-3

26. Benvenutti F, Baroni A, Bandinelli S, et al. Flunarizine-induced parkinsonism in the elderly. J Clin Pharmacol 1988; 28: 600-8

27. Kuzuhara S, Kohara N, Ohkawa Y, et al. Parkinsonism, depres­sion and akathisia-induced by flunarizine, a calcium entry blockade. Report of 31 cases. Rinsho Shinkeigaku 1989; 29: 681-6

28. Micheli FE, Fernandez Pardal M, Giannaula R, et al. Movement disorders and depression due to flunarizine and cinnarizine. Mov Disord 1989; 4: 139-46

29. Gimenez-Roldan S, Mateo D. Cinnarizine-induced parkinson­ism. Susceptibility related to aging and essential tremor. Clin Neuropharmacol 1991; 14: 156-64

30. Garcia-Ruiz P, Garcia de Wbenes J, Jimenez-Jimenez FJ, et al. Parkinsonism associated to calcium channel blockers (CCB). A prospective follow-up study. Clin Neuropharmacol 1992; 15: 19-26

31. Laporte JR, Capella D. Useless drugs are not placebos: lessons from flunarizine and cinnarizine [letter]. Lancet 1986; I: 853-4

32. Dick RS, Barold SS. Diltiazem-induced parkinsonism [letter]. Am J Med 1989; 87: 95-6

33. Malaterre HR, Lauribe P, Paganelli F, et al. Syndrome parkin­sonien, effet indesirable possible des inhibiteurs calciques. Arch Mal Coeur Vaiss 1992; 85: 1335-7

34. Graham DF, Stewart-Wynne EG. Diltiazem-induced acute par­kinsonism. Aust N Z J Med 1994; 24: 70

35. Garcia-Albea E, Jimenez-Jimenez FJ, Ayuso-Peralta L, et al. Parkinsonism unmasked by verapamil. Clin Neuropharmacol 1993; 16: 263-5

36. Padrell MD, Navarro M, Faura CC, et al. Verapamil-induced parkinsonism [letter]. Am J Med 1995; 99: 436

37. Sempere AP, Duarte J, Cabezas C, et al. Parkinsonism-induced by amlodipine. Mov Disord 1995; 10: 115-6

38. Nakashima K, Shimoda M, Kuno N, et al. Temporary symptom worsening caused by manidipine hydrochloride in two pa­tients with parkinson's disease. Mov Disord 1994; 9: 106-7

39. Grimes JD. Parkinsonism and tardive dyskinesia associated with long-term metoclopramide therapy. N Engl J Med 1981; 305: 1417

40. Grimes JD, Hassan MN, Preston DN. Adverse neurologic ef­fects of me to clop rami de. Can Med Assoc J 1982; 126: 23-5

41. Indo T, Ando K. Metoc1opramide-induced parkinsonism. Clin­ical characteristics of ten cases. Arch Neurol 1982; 39: 494-6

42. Bateman DN, Rawlins MD, Simpson JM. Extrapyramidal re­actions with metoclopramide. BMJ 1985; 291: 930-2

Drug Safety 1997 Mar: 16 (3)

192

43. Yamamoto M, Ujike H, Ogawa N. Metoclopramide-induced parkinsonism. Clin Neuropharmacol1987; 10: 287-9

44. Sethi KD, Patel B, Meador KJ. Metoclopramide-induced par­kinsonism [letter). South Med J 1989; 82: 1581-2

45. Miller LG, Jankovic J. Metoclopramide-induced movement disorders. Clinical findings with a review of the literature. Arch Intern Med 1989; 149: 2486-92

46. Bateman DN, Darling WM, Boys R, et al. Extrapyramidal re­actions to metoclopramide and prochlorperazine. Q J Med 1989; 71: 307-11

47. Ganzini L, Casey DE, Hoffman WF, et al. The prevalence of metoclopramide-induced tardive dyskinesia and acute extra­pyramidal movement disorders. Arch Intern Med 1993; 153: 1469-75

48. Avorn J, Gurwitz JH, Bohn RL, et al. Increased incidence of levodopa therapy following metoclopramide use. JAMA 1995; 274: 1780-2

49. Montagna P, Gabellini AS, Monari L, et al. Parkinsonian syn­drome after long-term treatment with clebopride. Mov Disord 1992; 7: 89-90

50. Jimenez-Jimenez FJ, Cabrera-Valdivia F, Ayuso-Peralta L, et al. Persistent parkinsonism and tardive dyskinesia-induced by clebopride. Mov Disord 1993; 8: 246-7

51. Sempere AP, Duarte J, Palomares JM, et al. Parkinsonism and tardive dyskinesia after chronic use of clebopride. Mov Dis­ord 1994; 9: 114-5

52. Naito Y, Kuzuhara S. Parkinsonism-induced or worsened by cisapride. Nippon Ronen Igakkai Zasshi 1994; 31: 899-902

53. Micheli F, Gatto E, Fermindez-Pardal M, et al. Domperidona y enfermedad de parkinson [letter). Medicina (B Aires) 1988; 48:218

54. Milandre L, Ali-Cherif A, Khalil R. Syndrome parkinsonien au cours d'un traitement par Ie veralipride. Rev Med Interne 1991; 12: 157-8

55. Franchignoni FP, Tesio L. Sindrome parkinsoniana indotta da veralipride. Minerva Ginecol 1995; 47: 277-9

56. North RR. Drug-induced movement disorders. Postgrad Med 1971; 97: 882

57. Jankovic J, Orman J. Tetrabenazine therapy of dystonia, chorea, tics, and dyskinesias. Neurology 1988; 38: 391-4

58. Prusinski A, Kozubski W, Szymanska R, et al. Neurological side effects after using thietylperazine. Pol Tyg Lek 1983; 38: 1227-8

59. Bremnum J. Extrapyramidal side effects after long-term treat­ment with thiethylperazine. Ugeskr Laeger 1988; 150: 2827

60. Rosenblum AM, Montgomery EB. Exacerbation of parkinson­ism by methyldopa [letter). JAMA 1980; 244: 2727-8

61. Gillman MA, Sandyk R. Parkinsonism-induced by methyldopa. S Afr Med J 1984; 65: 194

62. Bouchard RH, Pourcher E, Vincent P. Fluoxetine and extra­pyramidal side effects [letter). Am J Psychiatry 1989; 146: 1352-3

63. Tate JL. Extrapyramidal symptoms in a patient taking haloper­idol and fluoxetine. Am J Psychiatry 1989; 146: 399

64. Gernaat HBPE, Van de Woude J, Touw DJ. Fluoxetine and par­kinsonism in patients taking carbamazepine [letter]. Am J Psychiatry 1991; 141: 118-9

65. Jansen Steur ENH. Increase of parkinson disability after fluoxetine medication. Neurology 1993; 43: 211-3

66. Daric C, Dollfus S, Mihout B, et al. Fluoxetine et symptomes extrapyramidaux. A propos de deux observations. Encephale 1993; 19: 61-2

© Adis International Umited. All rights reserved.

Jimenez-Jimenez et al.

67. Gatto EM, Fernandez-Pardal M, Micheli F. Agravacion del parkinsonismo por fluoxetina [letter). Medicina (B Aires) 1994;54: 182

68. Caley CF, Friedman JH. Does fluoxetine exacerbate parkinson's disease? J Clin Psychiatry 1992; 53: 278-82

69. Montastruc JL, Fabre N, Blin 0, etal. Does fluoxetine aggravate parkinson's disease? A pilot prospective study. Mov Disord 1995; 10: 355-7

70. Jimenez-Jimenez FJ, Tejeiro J, Martfnez-Junquera G, et al. Parkin­sonism exacerbated by paroxetine. Neurology 1995; 45: 2406

71. Malek-Ahmadi P, Allen SA. Paroxetine-molindone interaction [letter). J Clin Psychiatry 1995; 56: 82-3

72. Albanese A, Rossi P, Altavista MC. Can trazodone induce par­kinsonism? Clin Neuropharmacol1988; ll: 180-2

73. Teusink JP, Alexopoulos GS, Shamoian CA. Parkinsonian side effects-induced by a monoamine oxidase inhibitor [letter). Am J Psychiatry 1984; 141: 118-9

74. Strouse TB, Salehmoghaddam S, Spar JE. Acute delirium and parkinsonism in a bupropion-treated liver transplant recipient [letter). J Clin Psychiatry 1993; 54: 489-90

75. Lutz EG. Acute lithium-induced parkinsonism precipitated by liquid protein diet [letter). J Med Soc N J 1978; 75: 165-6

76. Kane J, Rifkin A, Quitkin F, et al. Extrapyramidal side effects with lithium therapy. Am J Psychiatry 1978; 135: 851-3

77. Tyrer P, Alexander MS, Regan A, et al. An extrapyramidal syn­drome after lithium therapy. Br J Psychiatry 1980; 136: 191-4

78. Gajkowski K, Werkowicz-Pelczyk D, Masiak I, et al. Neuro­logic symptoms in lithium poisoning. Neurol Neurochir Pol 1987; 21: 412-4

79. Holroyd S, Smith D. Disabling parkinsonism due to lithium: a case report [letter). J Geriatr Psychiatry Neuro11995; 8: 118-9

80. Suranyi-Cadotte BE, Nestoros IN, Nair NPV, et al. Parkinson­ism-induced by high doses of diazepam. Bioi Psychiatry 1985; 20: 451-60

81. van der Zwan Jr A. Transient parkinson syndrome and tremor caused by the use of sodium valproate. Ned Tijdschr Genee­skd 1989; 133: 1230-2

82. Alvarez-GomezMJ, Vaamonde J, NarbonaJ, et al. Parkinsonian syndrome in childhood after sodium valproate administration. Clin Neuropharmacol1993; 16: 451-5

83. Sasso E, Delsoldato S, Negrotti A, et al. Reversible valproate­induced extrapyramidal disorders. Epilepsia 1994; 35: 391-3

84. Froomes PR, Stewart MR. A reversible parkinsonian syndrome and hepatotoxocity following addition of carbamazepine to sodium valproate. Aust N Z J Med 1994; 24: 413-4

85. Goiii M, Jimenez M, Seijoo M. Parkinsonism-induced by phenytoin. Clin Neuropharmacol1985; 8: 383-4

86. Fox JH, Bennett DA, Goetz CG, et al. Induction of parkinson­ism by intraventricular bethanechol in a patient with Alz­heimer's disease. Neurology 1989; 9: 1265

87. Iwasaki Y, Wakata N, Kinoshita M. Parkinsonism-induced by pyridostigmine. Acta Neurol Scand 1988; 78: 236

88. LeMarie JF, Autret A, Biziere K, et al. Amiodarone neuropathy. Further arguments for human drug-induced neurolipidosis. Eur Neuro11987; 147: 881-4

89. Palakurthy PR, Iyer V, Meckler RJ. Unusual neurotoxicity as­sociated with amiodarone therapy. Arch Intern Med 1987; 147: 881-4

90. Werner EG, Olanow CWo Parkinsonism and amiodarone ther­apy. Ann Neurol 1989; 25: 630-2

91. Dotti MT, Federico A. Amiodarone-induced parkinsonism: a case report and pathogenetic discussion. Mov Disord 1995; 10: 233-4

Drug Safety 1997 Mar; 16 (3)

Drug-Induced Movement Disorders

92. Gjerris F. Transitory procaine-induced parkinsonism. J Neurol Neurosurg Psychiatry 1971; 34: 20-2

93. Lieberman AN, Goldstein M. Reversible parkinsonism related to meperidine [letter]. N Engl J Med 1985; 8: 509

94. Olive JM, Masana L, Gonzalez J. Meperidine and reversible parkinsonism. Mov Disord 1994; 9: 115-6

95. Fisher JF, Dewald J. Parkinsonism associated with intra-ven­tricular amphotericine B. J Antimicrob Ther 1983; 12: 97-9

96. Balmaceda CM, Walker RW, Castro-Malaspina H, et al. Rever­sal of amphotericin-B-related encephalopathy. Neurology 1994; 44: 1183-4

97. Mintz V, Liberman VA, Vries A. Parkinsonism syndrome due to cephaloridine [letter]. JAMA 1971; 216: 1200

98. Shaunak S, Brown P, Morgan-Hughes JA. Exacerbation of id­iopathic Parkinson's disease by naproxen [letter]. BMJ 1995; 311: 422

99. Fernandez W, Lees AJ. Temporary deterioration in parkinso­nian signs after modest alcohol intake. Mov Disord 1992; 7: 284-5

100. Yasui M, Kihira T, Ota K, et al. A case of parkinsonism-induced by an oral contraceptive. No To Shinkei 1992; 44: 163-6

10 I. Fujisawa Y, Tsuji T, Sasaki K. Parkinsonism-induced by in­deloxazine hydrochloride. Clin Ther 1989; 11: 807-11

102. Ogawa N. Parkinsonism-induced by indeloxazine hydrochlo­ride in the elderly. Clin Ther 1989; II: 802-6

103. Shimoda K, Hikasa C, Nishikawa S, et al. A case report of captopril-induced parkinsonism. Rinsho Shinkeigaku 1987; 27: 366-8

104. Luque FA, Selhorst JB, Petruska P. Parkinsonism-induced by high-dose cytosine arabinoside. Mov Disord 1987; 2: 219-22

105. Weiden PJ, Mann n, Haas G, et al. Clinical nonrecognition of neuroleptic-induced movement disorders: a cautionary study. Am J Psychiatry 1987; 144: 1148-53

106. Hansen TE, Brown WL, Weigel RM, et al. Vnderrecognition of tardive dyskinesia and drug-induced parkinsonism by psychi­atric residents. Gen Hosp Psychiatry 1992; 14: 340-4

107. Farde L, Nordstrom AL, Wiesel AL, et al. Positron emission tomographic analysis of central D I and D2 dopamine recep­tor occupancy in patients treated with classical neuroleptics and cIozapine. J Clin Psychiatry 1992; 9: 538-44

108. Grohmann R, Koch R, Schmidt LG. Extrapyramidal symptoms in neuroleptic recipients. Agents Actions 1990; 29 Suppl.: 71-82

109. Weiner WJ, Lang AE. Drug-induced movement disorders (not including tardive dyskinesia). In: Weiner WJ, Lang AE, edi­tors. Movement disorders. A comprehensive survey. Mount Kisco, New York: Futura Publishing Co., 1989: 599-644

110. Chouinard G. Early phase II clinical trial of remoxipride in treatment of schizophrenia with measurements of prolactin and neuroleptic activity. J Clin Psychopharmacol 1987; 7: 159-64

Ill. Laux G, Klieser E, Schroder HG, et al. A double-blind multi­centre study comparing remoxipride, two and three times daily, with haloperidol in schizophrenia. Acta Psychiatr Scand 1990; 358 Suppl.: 125-9

112. Lindstrom LH, Wieselgren 1M, Struwe G, et al. A double- blind comparative multicentre study of remoxipride and haloperi­dol in schizophrenia. Acta Psychiatr Scand 1990; 358 Suppl.: 130-5

113. Kane JM, Safferman AZ, Pollack S, et al. Clozapine, negative symptoms, and extrapyramidal side effects. J Clin Psychiatry 1994: 55 Suppl. B: 74-7

114. Gerlach J, Peacock L. Motor and mental side effects of c1ozap­ine. J Clin Psychiatry 1994: 55 Suppl. B: 107-9

© Adis Intemational Umited. All rights reserved.

193

115. Bezchlibnyk-Butler KZ, Remington GJ. Antiparkinsonian drugs in the treatment of neuroleptic-induced extrapyramidal symptoms. Can J Psychiatry 1994; 39: 74-84

116. Kurz M, Hummer M, Oberbauer H, et al. Extrapyramidal side effects of cIozapine and haloperidol. Psychopharmacology (BerJ) 1995; 118: 52-6

117. Owens DGC. Adverse effects of antipsychotic agents. Do newer agents offer advantages? Drugs 1996; 51: 895-930

118. Sandyk R, Kay SR. Drug-induced parkinsonism: relationship to age at onset of schizophrenia. Funct Neuro11991; 6: 151-7

119. Hoffman WF, Labs SM, Casey DE. Neuroleptic-induced par­kinsonism in older schizophrenics. BioI Psychiatry 1987; 22: 427-39

120. Prosser ES, Csernansky JG, Kaplan J, et al. Depression, parkin­sonian symptoms, and negative symptoms in schizophrenics treated with neuroleptics. J Nerv Ment Dis 1987; 175: 100-5

121. Nelli AC, Yarden PE, Guazzelli M, et al. Parkinsonism follow­ing neuroleptic withdrawal. Arch Gen Psychiatry 1989; 46: 383-4

122. Melamed E, Achiron A, Shapira A, et al. Persistent and progressive parkinsonism after discontinuation of chronic neuroleptic therapy: an additional tardive syndrome? Clin Neuro­pharrnacoll991; 14: 273-8

123. Burkhardt C, Kelly JP, Lim YH, et al. Neuroleptic medications inhibit complex I of the electron transport chain. Ann Neurol 1993; 33: 512-7

124. Schapira AHV, Cooper JM, Dexter D, et al. Mitochondrial com­plex I deficiency in Parkinson's disease [letter]. Lancet 1989; I: 1269

125. Mizuno Y, Ohta S, Tanaka S, et al. Deficiencies in complex I subunits of the respiratory chain in Parkinson's disease. Biochem Biophys Res Commun 1989; 163: 1450-5

126. Negrotti A, Ca1zetti S, Sasso E. Calcium-entry blockers--in­duced parkinsonism: possible role of inherited susceptibility. Neurotoxicology 1992; 13: 261-4

127. Garcia-Ruiz PJ, Mena MA, Pefiafiel N, et al. Cinnarizine-in­duced parkinsonism in primates. Clin Neuropharmacol 1992; 15: 152-4

128. Pileblad E, Carlsson A. The Ca2+ antagonist nimodipine de­creases and the Ca2+ agonist BAY K 8644 increases catechol­amine synthesis in mouse brain. Neuropharmacology 1987; 26: 101-5

129. Yarom M, Zurgil N, Zisapel N. Calcium permeability changes and neurotransmitter release in cultured brain neurons. II. Temporal analysis of neurotransmitter release. J BioI Chern 1985;260: 16294-302

130. Barochovsky 0, Bradford HF. Development of transmitter- re­leasing capacity in neuron-enriched tissue cultures. J Neu­rochem 1987; 48: 787-97

131. Pardo B, Casarejos MJ, Tabernero C, etal. Farmacologfade los antagonistas del calcio sobre el sistema dopaminergico [ab­stract]. Neurologfa (Spain) 1989; 4: 160

132. De Vries DJ, Beart PM. Competitive inhibition of [3H]spiperone binding to D-2 dopamine receptors in striatal homogenates by organic calcium-channel antagonists and polivalent cations. Eur J Pharmacol1985; 106: 133-9

133. De Yebenes MJG, Casarejos MJ, Mena MA, et al. Effects of calcium antagonists on dopamine-rich human neuroblastoma cells, NB69 [abstract]. Mov Disord 1992; 7 Suppl. 1: 61

134. Kariya S, Isozaki S, Masubuchi Y, et al. Possible pharmacoki­netic and pharmacodynamic factors affecting parkinsonism inducement by cinnarizine and flunarizine. Biochem Phar­maco11995; 50: 1645-50

Drug Safety 1997 Mar: 16 (3)

194

135. Veitch K, Hue L. Flunarizine and cinnarizine inhibit mitochon­drial complexes I and II: possible implication for parkinson­ism. Mol Pharmacol1994; 45: 158-63

136. Montastruc JL, Llau ME, Rascol 0, et al. Drug-induced parkin­sonism: a review. Fundam Clin Pharmacol1994; 8: 293-306

137. Richardson MA, Craig TJ. The coexistence of parkinsonism­like symptoms and tardive dyskinesia. Am J Psychiatry 1982; 19: 341-3

138. Jankovic J, Casabona J. Coexistent tardive dyskinesia and par­kinsonism. Clin Neuropharmacol1987; 10: 511-21

139. Stephen PJ, Williamson 1. Drug-induced parkinsonism in the elderly. Lancet 1984; II: 1082-3

140. Remington G, Bezchlibnyk-Butler K. Management of acute an­tipsychotic-induced extrapyramidal syndromes. CNS Drugs 1996; 5 Suppl. I: 21-35

141. Barnes TRE, McPhillips MA. Antipsychotic-induced extra­pyramidal symptoms. Role of anticholinergic drugs in treat­ment. CNS Drugs 1996; 6: 315-30

142. Hardie RJ, Lees AJ. Neuroleptic-induced Parkinson's syn­drome: clinical features and results of treatment with levo­dopa. J Neurol Neurosurg Psychiatry 1988; 51: 850-4

143. Stacy M, Jankovic 1. Tardive tremor. Mov Disord 1992; 7: 53-7 144. Arblaster LA, Lakie M, Mutch WJ, et al. A study of the early

signs of drug-induced parkinsonism. J Neurol Neurosurg Psy­chiatry 1993; 56: 301-3

145. Gabellini AS, Martinelli P, Coccagna G. Drug-induced tremor of the tongue. Ital J Neurol Sci 1989; 10: 89-91

146. Friedman JH. 'Rubral' tremor-induced by a neuroleptic drug. Mov Disord 1992; 7: 281-2

147. Tan U, Gepdiremen A, Banoglu N, et al. Intermittent tremor­bursts-induced by chlorpromazine and pentobarbitone in guinea pigs: a new tremor model. Int J Neurosci 1991; 61: 221-8

148. Gelenberg AJ, Jefferson JW. Lithium tremor. J Clin Psychiatry 1995; 56: 283-7

149. Schou M, Baastrup PC, GrofP, et al. Pharmacological and clin­ical problems of lithium prophylaxis. Br J Psychiatry 1970; 116: 615-9

150. Karas BJ, Wilder BJ, Hammond EJ, et al. Valproate tremors. Neurology 1982; 2: 428-32

151. Lejoyeux M, Rouillon F, Ades J, et al. Neural symptoms-in­duced by tricyclic antidepressants: phenomenology and pathophysiology. Acta Psychiatr Scand 1992; 85: 249-56

152. Lejoyeux M, Rouillon F, Ades J. Prospective evaluation of the serotonin syndrome in depressed inpatients treated with clomipramine. Acta Psychiatr Scand 1993; 88: 369-71

153. Larsen K, Schmekel B. Tremor in healthy volunteers after bambuterol and terbutaline CR tablets. Eur J Clin Pharmacol 1993; 45: 303-5

154. Weiner WJ, Lang AE. Tremor. In: Weiner WJ, Lang AE, editors. Movement disorders. A comprehensive survey. Mount Kisco, NY: Futura Publishing Co., 1989: 221-56

155. Logan WJ, Freeman JM. Pseudodegenerative disease due to diphenylhydantoin intoxication. Arch Neuro11969; 21: 631-7

156. Taira T. Zonisamide-induced tremor: report of two cases. No To Shinkei 1992; 44: 61-3

157. Reutens DC, Duncan JS, Patsalos PN. Disabling tremor after lamotrigine with sodium valproate [letter]. Lancet 1993; 42: 185-6

158. Kaminski CA, Robbins MS, Weibley RE. Sertraline intoxica­tion in a child. Ann Emerg Med 1994; 23: 1371-4

159. Apseloff G, Wilner KD, von-Deutsch DA, et al. Sertraline does not alter steady-state concentrations or renal clearance oflith­ium in healthy volunteers. J Clin Pharmacol 1992; 32: 643-6

© Adis International Urnited. All rights reserved.

Jimenez-Jimenez et al.

160. Debattista C, Schatzberg AF. Physical symptoms associated with paroxetine withdrawal [letter]. Am J Psychiatry 1995; 152: 1234-5

161. Soto J, Sacristan JA, Alsar MJ, et al. Terfenadine-induced tremor. Ann Neuro11993; 33: 226

162. Laguna-del-Estal P, Calabrese S, Zabala JA, et al. Toxicidad neur610gica por antimoniales pentavalentes durante el tratamiento de la leishmaniasis visceral. Med Clin (Barc) 1994; 102: 276-7

163. Cardoso FE, Jankovic J. Cocaine-related movement disorders. Mov Disord 1993; 8: 175-8

164. Findley U. Tremors: differential diagnosis and pharmacology. In: Jankovic J, Tolosa E, editors. Parkinson's disease and movement disorders, 2nd ed. Baltimore: Williams & Wilkins, 1993: 293-313

165. Cotzias GC, Van Woert MH, Schiffer LM. Aromatic amino acids and modification of parkinsonism. N Engl J Med 1967; 276: 567-72

166. Papavasiliou PS, Cotzias GC, Duby SE, et al. Levodopa in par­kinsonism: potentiation of central effects with a peripheral inhibitor. N Engl J Med 1972; 285: 8-14

167. Barbeau A. Treatment with levodopa and R04-4602: review and present status. Adv Neurol 1973; 2: 173-98

168. Martin WE, Tolosa ES, Loewenson RL, et al. The effect of combining carbidopa with levodopa for Parkinson's disease. Geriatrics 1975; 30: 39-44

169. Sage 11, Mark MH, McHale DM, et al. Benefits of monitoring plasma levodopa in Parkinson's disease patients with drug­induced chorea. Ann Neuro11991; 29: 623-8

170. Marconi R, Lefebvre-Caparros D, Bonnet AM, et al. Levodopa­induced dyskinesias in Parkinson's disease: phenomenology and pathophysiology. Mov Disord 1994; 9: 2-12

171. Zimmerman Jr TR, Sage 11, Lang AE, et al. Severe evening dyskinesias in advanced Parkinson's disease: clinical descrip­tion, relation to plasma levodopa, and treatment. Mov Disord 1994; 9: 173-7

172. Caine DB, Techeyne PF, Claverfa LE, et al. Bromocriptine in Parkinson's disease. BMJ 1974; 4: 442-4

173. Cotzias GC, Papavasiliou PS, Tolosa E, et al. Treatment of Par­kinson's disease with aporphines. Possible role of growth hor­mones. N Engl J Med 1976; 294: 567-72

174. Lieberman A, Goldstein M, Leibowitz M, et al. Treatment of advanced Parkinson's disease with pergolide. Neurology 1981; 31: 675-82

175. McDonald RJ, Horowski L. Lisuride in the treatment of parkin­sonism. EurNeurol1983; 22: 240-55

176. LeWitt P. Clinical and pharmacological aspects of the anti­parkinsonian ergoline lisuride. In: Fahn S, Marsden CD, Jen­ner P, et aI., editors. Recent developments in Parkinson's disease. New York: Raven Press, 1986: 347-54

177. Klawans HL, Weiner WJ. The effect of d-amphetamine on cho­reiform movement disorders. Neurology 1974; 24: 312-8

178. Lundh H, Tunving K. An extrapyramidal choreiform syndrome caused by amphetamine addiction. J Neurol Neurosurg Psy­chiatry 1981; 44: 728-30

179. Rhee KJ, Albertson TE, Douglas JC. Choreoathetoid disorder associated with amphetamine-like drugs. Am J Emerg Med 1988; 6: 131-3

180. Sperling LS, Horowitz JL. Methamphetamine-induced choreoathe­tosis and rhabdomyolysis. Ann Intern Med 1994; 34: 388-90

181. Nausieda PA, Coller WC, Weiner WJ, et al. Pemoline-induced chorea. Neurology 1981; 31: 356-60

Drug Safety 1997 Mar; 16 (3)

Drug-Induced Movement Disorders

182. Weiner WJ, Nausieda PA, Klawans HL. Methylphenidate-in­duced chorea: case report and pharmacologic implications. Neurology 1979; 29: 1041-4

183. Habal R, Sauter 0, Olowe 0, et al. Cocaine and chorea. Am J Emerg Med 1991; 9: 618-20

184. Stuart AM, Worley LM, Spillane J. Choreiform movements ob­served in an 8-year-old child following use of an oral theo­phylline preparation. Clin Pediatr Phila 1992; 1: 692-4

185. Birket-Smith E. Abnormal involuntary movements-induced by anticholinergic therapy. Acta Neurol Scand 1974; 50: 801-11

186. Warne RW, Gubbay SS. Choreiform movements-induced by anticholinergic therapy [letter]. Med J Aust 1979; 1: 465

187. Matsumoto K, Nogaki H, Morimatsu M. A case of choreo-ath­etoid movements-induced by anticholinergic drugs, tryhexy­phenidyl HCI and dosulepin HCl. Nippon Ronen Igakkai Zasshi 1992; 29: 686-9

188. Nomoto M, Thompson PO, Sheehy MP, et al. Anticholinergic­induced chorea in the treatment of focal dystonia. Mov Disord 1987; 2: 53-6

189. Thach BT, Chase TN, Bosma J. Oral facial dyskinesia associ­ated with prolonged use of antihistaminic decongestants [let­ter]. N Engl J Med 1975; 293: 486-7

190. Lehmann AB. Reversible chorea due to ranitidine and cimeti­dine [letter]. Lancet 1988; II: 158

191. Nausieda PA, Koller WC, Weiner WJ, et al. Chorea-induced by oral contraceptives. Neurology 1979; 29: 1605-9

192. Bedard P, Langelier P, Danbova J, et al. Estrogens, progesterone and the extrapyramidal system. Adv Neuro11979; 24: 411-22

193. Galimberti D. Chorea-induced by the use of oral contraceptives. Report of a case and review of the literature.ltal J Neurol Sci 1987; 8: 383-6

194. Leys 0, Destee A, Petit H, et al. Chorea associated with oral contraception. J Neurol 1987; 235: 46-8

195. Driesen JJ, Wolters EC. Oral contraceptive-induced paraball­ism. Clin Neurol Neurosurg 1987; 89: 49-51

196. Mathur AK, Gatter RA. Chorea as the initial presentation of oral contraceptive-induced systemic lupus erythematosus. J Rheumatol 1988; 15: 1042-3

197. Liedholm LJ, Barajas F. Oral contraceptives causing chorea in a young woman [letter]. Lakartidningen 1988; 85: 448

198. Cocito 0, Gallo G. Hemichorea-induced by oral contraceptive use. Ital J Neurol Sci 1989; 10: 217-8

199. Profumo R, Toce S, Kotagal S. Neonatal choreoathetosis fol­lowing prenatal exposure to oral contraceptives. Pediatrics 1990; 86: 648-9

200. Caviness IN, Muenter MD. An unusual cause of recurrent cho­rea. Mov Disord 1991; 6: 355-7

201. Steiger MJ, Quinn NP. Hormone replacement therapy-induced chorea [letter]. BMJ 1991; 302: 762

202. Chadwick 0, Reynolds EG, Marsden CD. Anticonvulsant-in­duced dyskinesias: a comparison with dyskinesias-induced by neuroleptics. J Neurol Neurosurg Psychiatry 1976; 9: 1210-8

203. Filloux F, Thompson JA. Transient chorea-induced by pheny­toin. J Pediatr 1987; 110: 639-41

204. Szczechowski L, Rosciszewska D. Choreoathetotic movements in a patient with epilepsy after treatment with diphenylhydan­toin. Neurol Neurochir Pol 1987; 21: 255-7

205. Martin6n-Sanchez F, Viso-Lorenzo JA. Una observaci6n pediatrica de coreoatetosis por difenilhidantoina. An Esp Pediatr 1990; 32: 554-5

206. Martinez-Orgado J, Garcia-Aparicio J, Cabanillas-Vilaplana L, et al. Coreoatetosis inducida por difenilhidantoina en un

© Adis International Umited. All rights reserved.

195

lactante con sindrome CHARGE. An Esp Pediatr 1990; 33: 384-6

207. Haider Y, Abbott RJ. Phenytoin-induced choreoathetosis [let­ter]. Postgrad Med J 1990; 66: 1089

208. Harrison MD, Lyons GR, Landow ER. Phenytoin and dyskine­sias: a report of two cases and review of the literature. Mov Disord 1993: 8: 19-27

209. Shulman LM, Singer C, Weiner WJ. Phenytoin-induced focal chorea. Mov Disord 1996; 11: 111-4

210. Weaver OF, Camfield P, Fraser A. Massive carbamazepine overdose: clinical and pharmacologic observations in five ep­isodes. Neurology 1988; 38: 755-9

211. Mosquet B, Starace J, Madelaine S, et al. Syndrome choreo­athetosique sous carbamazepine et viloxazine. Consequence d'une interaction medicamenteuse?Therapie 1994; 49: 513-4

212. Kirschberg GJ. Dyskinesia: an unusual reaction to ethosuxi­mide. Arch Neurol 1975; 32: 137-8

213. Lancman ME, Asconape JJ, Penry JK. Choreiform movements associated with the use of valproate. Arch Neurol 1994; 51: 702-4

214. Kerrick JM, Kelley BJ, Maister BH, et al. Involuntary move­ment disorders associated with felbamate. Neurology 1995; 45: 185-7

215. Buetefisch CM, Gutierrez A, Gutmann L. Choreoathetotic movements: a possible side effect of gabapentin. Neurology 1996; 46: 851-2

216. Patrick SJ, Snelling LK, Ment LR. Infantile chorea following abrupt withdrawal of diazepam and pentobarbital therapy. J Toxicol Clin Toxicol1993; 31: 127-32

217. Fann WE, Sullivan JL, Richman BW. Dyskinesias associated with tricyclic antidepressants. Br J Psychiatry 1976; 128: 490-3

218. Sandyk R. Nomifensine-induced orofacial dyskinesia and Sydenham's chorea. Int J Neurosci 1987; 35: 91-2

219. Patterson JP. Amoxapine-induced chorea [letter]. South Med J 1983; 76: 1077

220. Marchioni E, Perucca E, Soragna 0, et al. Choreiform syn­drome associated with fluoxetine treatment in a patient with deficient CYP2D6 activity. Neurology 1996; 46: 853

221. Zorumski CP, Bakris GL. Choreoathetosis associated with lith­ium: case report and literature review. Am J Psychiatry 1983; 140: 1621-2

222. Walevski A, Radwan M. Choreoathetosis as toxic effect of lith­ium treatment. Eur Neurol 1986; 25: 412-5

223. Matsis PP, Fisher RA, Tasman-Jones C. Acute lithium toxicity: chorea, hypercalcemia and hyperamylasemia. Aust N Z J Med 1989; 19: 718-20

224. Reed SM, Wise MG, Timmerman I. Choreoathetosis: a sign of lithium toxicity. J Neuropsychiatry Clin Neurosci 1989; I: 57-60

225. Helmuth 0, Ljaljevic Z, Ramirez L, et al. Choreoathetosis-in­duced by verapamil and lithium treatment. J Clin Psy­chopharmacoI1989;9:454-5

226. Tsuda N, Negoro K, Morimatsu M. A case of multiple cerebral infarctions associated with hemichorea-induced by amantad­ine hydrochloride. Rinsho Shinkeigaku 1994; 34: 388-90

227. Tilzey A, Heptonstall J, Hamblin T. Toxic confusional state and choreiform movements after treatment with anabolic steroids. BMJ 1981; 283: 343-50

228. Wasserman S, Yahr MD. Choreic movements-induced by the use of methadone. Arch Neuro11980; 37: 727-8

229. Crystal HA. Baclofen therapy may be associated with chorea in Alzheimer's disease. Ann Neuro11990; 28: 839

Drug Safety 1997 Mar; 16 (3)

196

230. Kaplan SR, Murkofsky C. Oral-buccal dyskinesia symptoms associated with low-dose benzodiazepine treatment [letter]. Am J Psychiatry 1978; 135: 1558-9

231. Neil EM, Waters AK. Generalized choreiform movements as a complication of methyldopa therapy in chronic renal failure. Postgrad Med J 1981; 57: 732-3

232. Kaminsky P, Cosmidis S, Debouverie M, et al. Syndrome choreiforme provoque par l'alpha-methyldopa [letter]. The­rapie 1988; 43: 239

233. Wedzicha JA, Gibb WR, Lees AJ, et al. Chorea in digoxin tox­icity [letter]. J Neurol Neurosurg Psychiatry 1984; 47: 419

234. Mulder LJ, Van der Mast RC, Meerwaldt JD. Generalised cho­rea due to digoxin toxicity [letter]. BMJ 1988; 296: 1262

235. Neary 0, Thurston H, Pohl JE. Development of extrapyramidal symptoms in hypertensive patients treated with diazoxide [letter]. BMJ 1973; 3: 474-5

236. Vernay D, Dubost n, Thevenet JP, et al. 'Choree fibrillaire de Morvan' followed by Guillain-Barre syndrome in a patient receiving gold therapy [letter]. Arthritis Rheum 1986; 29: 1413-4

237. Aumaitre 0, Vernay D, Marcheix JC, et al. Choree fibrillaire de Morvan secondaire 11 un traitement par sels d'or. Presse Med 1988; 17: 2140

238. Petiot P, Charles N, Vial C, et al. Complications neurologiques des sels d' or: discussion nosologique a propos d'un cas. Rev Neurol (Paris) 1993; 149: 562-5

239. Samie MR, Ashton AK. Choreoathetosis-induced by cypro­heptadine. Mov Disord 1989; 4: 81-4

240. Combarros 0, Fabrega E, Polo JM, et al. Cyclosporine-induced chorea after liver transplantation for Wilson's disease. Ann Neurol 1993; 33: 108-9

241. Quinn AG, Ellis WR, Bum D, et al. Chorea precipitated by sulphasalazine [letter]. BMJ 1991; 302: 1025

242. Burguet A, Menget A, Fromentin C, et al. Effets neurologiques de I' aciclovir apres greffe de moelle allogenique. Arch Fr Pediatr 1988; 45: 343-5

243. McHugh P. Acute choreoathetoid reaction to propofol [letter]. Anaesthesia 1991; 46: 425

244. Borgeat A, Wilder-Smith 0. Acute choreoathetoid reaction to propofol [letter]. Anaesthesia 1991; 46: 797

245. Tanner CM, Melamed E, Lees AJ. Managing motor fluctua­tions, dyskinesias, and other adverse effects in Parkinson's disease. Neurology 1994; 44 Suppl. I: S12-S16

246. Shale H, Tanner C. Pharmacological options for the manage­ment of dyskinesias. Drugs 1996; 52: 849-60

247. Koller WC, Silver DE, Lieberman A. An algorithm for the man­agement of Parkinson's disease. Neurology 1994; 44 Suppl. 10: SI-S52

248. Bennet JP, Landow ER, Schuh LA. Suppression of dyskinesias in advanced Parkinson's disease. II. Increasing daily clozap­ine doses suppress dyskinesias and improve parkinsonism symptoms. Neurology 199; 43: 1551-5

249. Kleerdorfer B, Lees AJ, Stem GM. Buspirone in the treatment of levodopa-induced dyskinesias [letter]. J Neurol Neurosurg Psychiatry 1991; 54: 376-7

250. Bonifati V, Fabrizio E, Cipriani R, et al. Buspirone in levodopa­induced dyskinesias. Clin Neuropharmacol1994; 17: 99-102

251. DurifF. Vidailhet M, Bonnet AM, et al. Levodopa-induced dys­kinesias are improved by fluoxetine. Neurology 1956; 45: 1855-8

252. Tarsy D, LeopoldN, Sax D. Physostigmine in choreiform move­ment disorders. Neurology 1974; 24: 28-34

253. Klawans HL, Topel JL, Bergen D. Deanol in the treatment of levodopa-induced dyskinesias. Neurology 1975; 25: 290-3

© Adis Interna~onal Umlted. All rights reserved.

Jimenez-Jimenez et al.

254. Price PA, Parkes JD, Marsden CD. Sodium valproate in the treatment of levodopa-induced dyskinesias. J Neurol Neuro­surg Psychiatry 1978; 41: 702-6

255. Ziegler M, Fournier V, Bathien N, et al. Therapeutic response to progabide in neuroleptic and levodopa-induced dyskine­sias. Clin Neuropharmacol1987; 10: 238-46

256. Turjanski N, Lees AJ. Gamma vinyl GABAin the treatment of levodopa-induced dyskinesias [letter]. J Neurol Neurosurg Psychiatry 1992; 55: 413

257. Gershanik OS, Luqnin MR, Scipioni 0, et al. Isoniazid therapy in Parkinson's disease. Mov Disord 1988; 3: 133-9

258. Laitinen LV, Bergenheim A, Hariz MI. Leksell's posteroventral pallidotomy in the treatment of Parkinson's disease. J Neuro­surg 1992; 76: 53-61

259. Laitinen LV. Pallidotomy for Parkinson's disease. Neurosurg Clin North Am 1995; 6: 105-12

260. Iacono RP, Shima F, Lonser RR, et al. The results, indications, and physiology of posteroventral pallidotomy for patients with Parkinson's disease. Neurosurgery 1995; 36: 1118-25

261. Obeso JA, Grandas F, Vaamonde J, et al. Motor complications associated with chronic levodopa therapy in Parkinson's dis­ease. Neurology 1989; 39 Suppl. 2: 11-9

262. Muenter MD, Sharpless NS, Tyce GM, et al. Patterns of dysto­nia ('I-D-I' and 'D-I-D') in response to I-dopa therapy for Parkinson's disease. Mayo Clin Proc 1977; 52: 163-74

263. Victor JF, Pollak P, Gervason CL, et al. Levodopa-induced di­phasic dyskinesias improved by subcutaneous apomorphine. Mov Disord 1992; 7: 283-4

264. DurifF, Deffond D, Dordain G, et al. Apomorphine and diphasic dyskinesia. Clin Neuropharmacol1994; 17: 99-102

265. Burke RE, Falm S, Jankovic J, et al. Tardive dystonia: late onset and persistent dystonia caused by antipsychotic drugs. Neu­rology 1982; 32: 1335-46

266. Wojcik JD, Falk WE, Fink JS, et al. A review of 32 cases of tardive dystonia. Am J Psychiatry 1991; 148: 1055-9

267. Nasrallah HA, Churchill CM, Hamdan-Allan GA. Higher fre­quency of neuroleptic-induced dystonia in mania than in schizophrenia [letter]. Am J Psychiatry 1988; 145: 1455-6

268. Remington GJ, Voineskos G, Pollock B, et al. Prevalence of neuroleptic-induced dystonia in mania and schizophrenia. Am J Psychiatry 1990; 147: 1231-3

269. Khanna R, Das A, Damodaran SS. Prospective study of neuro­leptic-induced dystonia in mania and schizophrenia. Am J Psychiatry 1992; 149: 511-3

270. Spina E, Sturiale V, Valvo S, et al. Prevalence of acute dystonic reactions associated with neuroleptic treatment with and with­out anticholinergic prophylaxis. Int Clin Psychopharmacol 1993; 8: 21-4

271. Aguilar EJ, Keshavan MS, Martinez-Quiles MD, et al. Predic­tors of acute dystonia in first-episode psychotic patients. Am J Psychiatry 1994; 151: 1819-21

272. Yassa R, Nair V, Dimitry R. Prevalence of tardive dystonia. Acta Psychiatr Scand 1986; 73: 629-33

273. Friedman JH, Kuchanski LT, Wagner RL. Tardive dystonia in a psychiatric hospital. J Neurol Neurosurg Psychiatry 1987; 50: 801-3

274. Sethi KD, Hess DC, Harp RJ. Prevalence of dystonia in veterans on chronic antipsychotic therapy. Mov Disord 1990; 5: 319-21

275. Chiu H, Shum P, Lau J, et al. Prevalence of tardive dyskinesia, tardive dystonia, and respiratory dyskinesia among Chinese psychiatric patients in Hong Kong. Am J Psychiatry 1992; 149: 1081-5

276. Sachdev P. Tardive and chronically recurrent OCUlogyric crises. Mov Disord 1993; 8: 93-7

Drug Safety 1997 Mar; 16 (3)

Drug-Induced Movement Disorders

277. Lu CS, Chu NS. Acute'dystonic reaction with asterixis and myoclonus following metoclopramide therapy [letter]. J Neu­rol Neurosurg Psychiatry 1988; 51: 1002-3

278. Perez N, Fernandez S, Legarda I, et al. Distonia aguda por neurolepticos en el tratamiento del vertigo: a prop6sito de dos casos. Acta Otorrinolaringol Esp 1992; 43: 287-9

279. Schott K, Ried S, Stevens I, et al. Neuroleptically-induced dys­tonia in Huntington's disease: a case report. Eur Neuro11989; 29: 39-40

280. Miller LG, Jankovic J. Sulpiride-induced tardive dystonia. Mov Disord 1990; 5: 83-4

281. Linazasoro G, Marti-Masso JF, Olasagasti B. Acute dystonia­induced by sulpiride. Clin Neuropharrnacol 1991; 14: 463-4

282. Bucci KK, Haverstick DE, Abercrombie SA. Dystonic-like re­action following cisapride therapy. J Fam Pract 1995; 40: 86-8

283. Shafrir Y, Levy Y, Steinherz R. Acute dystonic reaction due to domperidone in a infant. Harefuah 1986; 1I0: 237-8

284. Bonuccelli U, Nocchiero A, Napolitano A, et aI. Domperidone­induced acute dystonia and polycystic ovary syndrome. Mov Disord 1991; 6: 79-81

285. Gabellini AS, Pezzoli A, De-Massis P, etal. Veralipride-induced tardive dystonia in a patient with bipolar psychosis. Ital J Neurol Sci 1992; 13: 621-3

286. Kidron D, Melamed E. Forms of dystonia in patients with Par­kinson's disease. Neurology 1987; 37: 1009-11

287. McHale DM, Sage JI, Sonsalla PK, et aI. Complex dystonia of Parkinson's disease: clinical features and relation to plasma levodopa profile. Clin Neuropharmacol 1990; 13: 164-70

288. Mark MH, Sage JI. Levodopa-associated hemifacial dystonia. Mov Disord 1991; 6: 383

289. Poewe WHo Clinical aspects of motor fluctuations in Parkin­son's disease. Neurology 1994; 44 Suppl. 6: S6-9

290. Leiguarda R, Merello M, Sabe L, et al. Bromocriptine-induced dystonia in patients with aphasia and hemiparesis. Neurology 199; 4: 219-22

291. Kramer MS, Marcus DJ, DiFerdinando J, et al. Atypical acute dystonia associated with trazodone treatment. J Clin Psy­chopharmacol1986; 6: 117-8

292. Reccoppa L, Welch WA, Ware MR. Acute dystonia and fluoxet­ine [letter]. J Clin Psychiatry 1990; 51: 487

293. Boylan K. Persistent dystonia associated with buspirone. Neu­rology 1990; 40: 1904

294. Stoukides JA, Stoukides CA. Extrapyramidal symptoms upon discontinuation offluoxetine [letter]. Am J Psychiatry 1991; 148: 126

295. Rio J, Molins A, Viguera ML, et al. Distonia aguda por fluoxetina. Med Clin (Barc) 1992; 99: 46-7

296. George MS, Trimble MR. Dystonic reaction associated with fluvoxamine [letter]. J Clin Psychopharmacol 1993; 1: 220-1

297. Dave M. Fluoxetine-associated dystonia [letter]. Am J Psychi­atry 1994; 151: 149

298. Shihabuddin L, Rapport D. Sertraline and extrapyramidal side effects [letter]. Am J Psychiatry 1994; 151: 288

299. Chong SA. Fluvoxamine and mandibular dystonia [letter]. Can J Psychiatry 1995; 40: 430-1

300. Lauterbach EC. Fluoxetine, buspirone, myoclonus, and dysto­nia [letter]. Am J Psychiatry 1995; 152: 1697

301. Hoaken PC. An alert to extrapyramidal side-effects from SSRIs [letter]. Can J Psychiatry 1995; 40: 51

302. Ornadel D, Barnes EA, Dick DJ. Acute dystonia due to amitryptiline [letter]. J Neurol Neurosurg Psychiatry 1992; 55:414

303. Lee HK. Dystonic reactions to amitriptyline and doxepin [let­ter]. Am J Psychiatry 1988; 145: 649

© Adis Internatlonal Umited. All rights reserved.

197

304. Pande AC, Max P. A dystonic reaction occurring during treat­ment with tranylcypromine [letter]. J Clin Psychopharmacol 1989; 9: 229-30

305. Jarecke CR, Reid P1. Acute dystonic reaction-induced by a monoamine oxidase inhibitor. J Clin Psychopharmacol1990; 10: 144-5

306. Critchley EM, Phillips M. Unusual idiosyncratic reactions to carbamazepine [letter]. J Neurol Neurosurg Psychiatry 1988; 51: 1238

307. Lee Jw. Persistent dystonia associated with carbamazepine therapy: a case report. N Z Med J 1994; 107: 360-1

308. Soman P, Jain S, Rajsekbar V, et al. Dystonia: a rare manifesta­tion of carbamazepine toxicity. Postgrad Med J 1994; 70: 54-5

309. Moss W, Ojukwu C, Chiriboga CA. Phenytoin-induced move­ment disorder. Unilateral presentation in a child and response to diphenhydramine. Clin Pediatr (Phila) 1994; 33: 364-8

310. Lacayo A, Mitra N. Report of a case of phenobarbital--induced dystonia [letter]. Clin Pediatr (Phila) 1992; 31: 252

311. Howrie DL, Rowley AH, Krenzelok EP. Benztropine-induced acute dystonic reaction. Ann Emerg Med 1986; 15: 594-6

312. LeWitt PA, Walters A, Hening W, et al. Persistent movement disorders-induced by buspirone. Mov Disord 1993; 8: 331-4

313. Hooker EA, Danzl DE Acute dystonic reaction due to diaze­pam. J Emerg Med 1988; 6: 491-3

314. Perez-Trollen JM, Modrego-Pardo PJ, Vazquez-Andre M, et al. Bromazepam-induced dystonia. Biomed Pharmacother 1992; 46: 375-6

315. Stolarek IH, Ford MJ. Acute dystonia-induced by midazolam and abolished by flumazenil [letter]. BMJ 1990; 300: 614

316. Capstick C, Checkley S, Gray J, et al. Dystonia-induced by amphetamine and haloperidol [letter]. Br J Psychiatry 1994; 165: 276

317. Thiel A, Dressler D. Dyskinesias possibly-induced by norpseudo­ephedrine. J Neuro11994; 241: 167-169

318. Gay CT, Ryan SG. Paroxysmal kinesigenic dystonia after meth­ylphenidate administration. J Child Neurol 1994; 9: 45-6

319. Merab J. Acute dystonic reaction to cocaine [letter]. AmJ Med 1988;84:564

320. Choy-Kwong M, Lipton RB. Dystonia related to cocaine with­drawal: a case report and pathogenic hypothesis. Neurology 1989; 39: 996-7

321. Kumor K. Cocaine withdrawal dystonia. Neurology 1990; 40: 863-4

322. Hegarty AM, Lipton RB, Merriam AE, et al. Cocaine as a risk factor for acute dystonic reactions. Neurology 1991; 41: 1670-2

323. Farrel PE, Diehl AK. Acute dystonic reaction to crack cocaine [letter]. Ann Emerg Med 1991; 20: 322

324. Merello MJ, Nogues MA, Leiguarda RC, et aI. Dystonia and reflex sympathetic dystrophy-induced by ergotamine. Mov Disord 1991; 6: 263-4

325. Olson WL. Dystonia and reflex sympathetic dystrophy-induced by ergotamine. Mov Disord 1992; 7: 188-9

326. Garda G, Kaufman MB, Colucci RD. Dystonic reaction asso­ciated with sumatriptan [letter]. Ann Pharmacother 1994; 28: 1199

327. de Medina A, Biasini 0, Rivera A, et al. Nifedipine and myoclo­nic dystonia [letter]. Ann Intern Med 1986; 104: 125

328. Casteels-Van-Daele M, Eggermont E, Casaer P, et al. Acute dystonic reactions and long-lasting impaired consciousness associated with oxatomide in children [letter]. Lancet 1986; I: 1204-5

329. Santora J, Rozek S, Samie MR. Diphenhydramine-induced dys­tonia [letter]. Clin Pharm 1989; 8: 471

Drug Safety 1997 Mar; 16 (3)

198

330. Roila F, Donati D, Basurto C, et al. Diphenhydramine and acute dystonia [letter]. Ann Intern Med 1989; Ill: 92-3

331. Etzel JV. Diphenhydramine-induced acute dystonia. Pharmaco­therapy 1994; 14: 492-6

332. Romisher S, Felter R, Dougherty J. Tagamet-induced acute dys­tonia. Ann Emerg Med 1987; 16: 1162-4

333. McCann UD, Penetar DM, Belenky G. Acute dystonic reaction in normal humans caused by catecholamine depletion. Clin Neuropharmacol1990; 13: 565-568

334. Shafrir Y, Levy Y, Beharab A, et al. Acute dystonic reaction to bethanechol: a direct acetylcholine receptor agonist. Dev Med Child Neuro11986; 28: 646-8

335. Miller LG, Jankovic J. Persistent dystonia possibly-induced by flecainide. Mov Disord 1992; 7: 62-3

336. Achumba n, Ette EI, Thomas WO, et al. Chloroquine-induced acute dystonic reactions in the presence of metronidazole. Drug Intell Clin Pharm 1988; 22: 308-10

337. Ascher DP, Delaney RA. Acute dystonia from etoposide. Drug Intell Clin Pharm 1988; 22: 41-2

338. Mets B. Acute dystonia after alfentanil in untreated Parkinson's disease [letter]. Anesth Analg 1991; 72: 557-8

339. Lamberti JS, Bellnier T. Clozapine and tardive dystonia. J Nerv Ment Dis 1993; 181: 137-8

340. Kastrup 0, Gastpar M, Schwarz M. Acute dystonia due to c1ozapine. J Neurol Neurosurg Psychiatry 1994; 57: 119

341. Thomas P, Lalaux N, Vaiva G, et al. Dose-dependent stuttering and dystonia in a patient taking c10zapine [letter]. Am J Psy­chiatry 1994; 151: 1096

342. Owens DG. Extrapyramidal side effects and tolerability of ris­peridone: a review. J Clin Psychiatry 1994; 55 Suppl.: 29-35

343. Dickson R, Williams R, Dalby JT. Dystonic reaction and relapse with c10zapine discontinuation and risperidone intiation [let­ter]. Can J Psychiatry 1994; 39: 184

344. Radford JM, Brown TM, Borison RL. Unexpected dystonia while changing from c10zapine to risperidone [letter]. J Clin Psychopharmacol 1995; 15: 225-6

345. Worrall R, Wilson A, Cullen M. Dystonia and drug-induced hepatitis in a patient treated with c10zapine [letter]. Am J Psy­chiatry 1995; 152: 647-8

346. Lupu V, Gheragosian G. Reactie distonica severa in intoxicatia acuta cu Torecan. Rev Pediatr Obstet Ginecol Pediatr 1989; 38: 277-81

347. Jimenez-Jimenez FJ, Vazquez A, Garcfa-Ruiz P, et al. Chronic hemidystonia following acute dystonic reaction to thiethyl­perazine [letter]. J Neurol Neurosurg Psychiatry 1991; 54: 562

348. Shimada J, Akama Y, Tase C, et al. Problems of epidural droperidol administration. Masui 1994; 43: 1248-50

349. Gimenez-Roldan S, Mateo D, Bartolome P. Tardive dystonia and severe tardive dyskinesia. Acta Psychiatr Scand 1985; 71 : 488-94

350. Kang UJ, Burke RE, Fahn S. Natural history and treatment of tardive dystonia. Mov Disord 1986; I: 193-208

351. Gardos GG, Cole JO, Salomon M, et al. Clinical forms of severe tardive dyskinesia. Am J Psychiatry 1987; 144: 895-902

352. Addonizio G, Alexopoulos GS. Drug-induced dystonia in young and elderly patients. Am J Psychiatry 1988; 145: 869-71

353. Yassa R, Nair V, Iskandar H. A comparison of severe tardive dystonia and severe tardive dyskinesia. Acta Psychiatr Scand 1989; 80: 155-9

354. Bosch MF, Ludin HP. Apomorphin als adjuvante behandlung beim idiopathischen parkinsonsyndrom. Schweiz Arch Neu­rol Psychiatr 1994; 145: 8-13

355. Poewe WH, Lees AJ. The pharmacology of foot dystonia in parkinsonism. Clin Neuropharmacol1987; 10: 47-56

© Adis International Umited. All rights reserved.

Jimenez-Jimenez et al.

356. Pacchetti C, Albani G, Martignoni E, et al. 'Off' painful dysto­nia in Parkinson's disease treated with botulinum toxin. Mov Disord 1995; 10: 333-6

357. Tolosa E, Alom J, Marti MJ. Drug-induced dyskinesias. In: Jankovic J, Tolosa E, editors. Parkinson's disease and move­ment disorders. 2nd ed. Baltimore: William & Wilkins, 1993: 375-97

358. Faurbye A, Rasch PJ, Peterson PB, et al. Neurological symp­toms in pharmacotherapy of psychoses. Acta Psychiatr Scand 1964; 40: 10-27

359. Llau ME, Senard JM, Rascol 0, et al. Mouvements anormaux d'origine medicamenteuse: l'experience d'un centre de phar­macovigilance sur cinq ans. Therapie 1995; 50: 425-7

360. Kolakowska T, Williams AO, Ardern M, et al. Tardive dyskine­sia in schizophrenics under 60 years of age. Bioi Psychiatry 1986; 21: 161-9

361. Morgenstern H, Glazer WM, Gibowski LD, et al. Predictors of tardive dyskinesia: results of a cross-sectional study in an outpatient population. J Chronic Dis 1987; 40: 319-27

362. Gureje 0. Tardive dyskinesia in schizophrenics. Prevalence, distribution and relationship to neurological 'soft' signs in Nigerian patients. Acta Psychiatr Scand 1987; 76: 523-8

363. Chouinard G, Annable L, Ross-Chouinard A, et al. A 5-year prospective longitudinal study of tardive dyskinesia: factors predicting appearance of new cases. J Clin Psychopharmacol 1988; 8 (4 Suppl.): 21S-6S

364. Bourgeois M. Les dyskinesies tardives des neuroleptiques en France. Encephale 1988; 14 Spec No: 195-201

365. Moussaoui D, Douki S, Bentounsi B, et al. Epidemiologie des dyskinesies tardives au Maghreb. Encephale 1988; 14 Spec No: 203-8

366. Ko GN, Zhang LD, Yan WW, et al. The Shanghai 800: preva­lence of tardive dyskinesia in a Chinese psychiatric hospital. Am J Psychiatry 1989; 146: 387-9

367. Yassa R, Nair NP, Iskander H, et al. Factors in the development of severe forms of tardive dyskinesia. Am J Psychiatry 1990; 147: 1156-63

368. Altamura AC, Cavallaro R, Regazzetti MG. Prevalence and risk factors for tardive dyskinesia: a study in an Italian population of chronic schizophrenics. Eur Arch Psychiatry Neurol Sci 1990; 240: 9-12

369. Tan CH, Tay LK. Tardive dyskinesia in elderly psychiatric pa­tients in Singapore. Aust N Z J Psychiatry 1991; 25: 119-22

370. Inada T, Ohnishi K, Kamisada M, et al. A prospective study of tardive dyskinesia in Japan. Eur Arch Psychiatry Clin Neu­rosci 1991; 240: 250-4

371. Sramek J, Roy S, Ahrens T, et al. Prevalence of tardive dyski­nesia among three ethnic groups of chronic psychiatric pa­tients. Hosp Community Psychiatry 1991; 42: 590-2

372. Richardson MA, Haugland G, Craig TJ. Neuroleptic use, par­kinsonian symptoms, tardive dyskinesia, and associated fac­tors in child and adolescent psychiatric patients. Am J Psychiatry 1991; 148: 1322-8

373. Saltz BL, Woerner MG, Kane JM, et al. Prospective study of tardive dyskinesia incidence in the elderly. JAMA 1991; 266: 2402-6

374. Yassa R, Nastase C, Dupont D, et al. Tardive dyskinesia in el­derly psychiatric patients: a 5-year study. Am J Psychiatry 1992; 149: 1206-11

375. Sweet RA, Mulsant BH, Rifai AH, et al. Dyskinesia and neuro­leptic exposure in elderly psychiatric inpatients. J Geriatr Psy­chiatry NeuroI1992; 5: 156-61

Drug Safely 1997 Mar; 16 (3)

Drug-Induced Movement Disorders

376. Koshino Y, Madokoro S, Ito T, et al. A survey of tardive dyski­nesia in psychiatric inpatients in Japan. Clin Neuropharmacol 1992; 15: 34-43

377. McCreadie RG. The Nithsdale schizophrenia surveys. An over­view. Soc Psychiatry Psychiatr EpidemioI1992; 27: 40-5

378. Harris MJ, Panton D, Caligiuri MP, et al. High incidence of tardive dyskinesia in older outpatients on low doses of neu­roleptics. Psychopharmacol Bull 1992; 28: 87-92

379. Chiu HF, Wing YK, Kwong PK, et al. Prevalence of tardive dyskinesia in samples of elderly people in Hong Kong. Acta Psychiatr Scand 1993; 87: 266-8

380. Jeste DV, Caligiuri MP. Tardive dyskinesia. Schizophr Bull 1993; 19: 303-15

381. Sweet RA, Mulsant BH, Gupta B, et al. Duration of neuroleptic treatment and prevalence of tardive dyskinesia in late life. Arch Gen Psychiatry 1995; 52: 478-86

382. Miller CH, Simioni I, Oberbauer H, et al. Tardive dyskinesia prevalence rates during a ten-year follow-up. J Nerv Ment Dis 1995; 183: 404-7

383. Dorevitch A, Meretyk I, Umansky Y, et al. Antipsychotic drugs and tardive dyskinesia: preliminary results in an adolescent psychiatric ward. J Clin Pharm Ther 1995; 20: 63-5

384. Jeste DV, Caligiuri MP, Paulsen JS, et al. Risk of tardive dys­kinesia in older patients. A prospective longitudinal study of 266 outpatients. Arch Gen Psychiatry 1995; 52: 756-65

385. Ahlfors UG, Rimon R, AppeIberg B, et al. Remoxipride and haloperidol in schizophrenia: a double-blind multi centre study. Acta Psychiatr Scand 1990; 358 Suppl.: 99-103

386. Walinder J, Holm AC. Experiences oflong-term treatment with remoxipride: efficacy and tolerability. Acta Psychiatr Scand Suppl 1990; 358: 158-63

387. Kane JM, Woerner MG, Pollack S, et al. Does c\ozapine cause tardive dyskinesia? J Clin Psychiatry 1993; 54: 327-30

388. Tamminga CA, Thaker GK, Moran M, et al. Clozapine in tard­ive dyskinesia: observations from human and animal model studies. J Clin Psychiatry 1994; 55 Suppl. B: 102-6

389. Dave N. Clozapine-related tardive dyskinesia. Bioi Psychiatry 1994; 35: 886-7

390. Addington DE, Toews JA, Addington JM. Risperidone and tar­dive dyskinesia: a case report [letter]. J Clin Psychiatry 1995; 56: 484-5

391. Beuc\air L, Fontaine R. Tardive dyskinesia associated with metoc\opramide. Can Med Assoc J 1986; 134: 613-4

392. Lazzara RR, Stoudemire A, Manning D, et al. Metoc\opramide­induced tardive dyskinesia: a case report. Gen Hosp Psychi­atry 1986; 8: 107-9

393. Patel M, Louis S. Long-term neurologic complications of me to­c\opramide [letter]. N Y State J Med 1986; 86: 210

394. Breitbart W. Tardive dyskinesia associated with high-dose in­travenous metoc\opramide [letter]. N Engl J Med 1986; 315: 518-9

395. Mahler Jr JC, Brown RP. More on metoclopramide and tardive dyskinesia [letter]. N Engl J Med 1987; 316: 412-3

396. Sewell DD, Jeste DY. Metoc\opramide-associated tardive dys­kinesia. An analysis of 67 cases. Arch Fam Med 1992; 1: 271-8

397. Sewell DD, Yoshinobu BH, Caligiuri MP, et al. Metoclopram­ide-associated tardive dyskinesia in hemodialysis patients with diabetes mellitus. Two case reports. Gen Hosp Psychia­try 1992; 14: 416-9

398. Putnam PE, Orenstein SR, Wessel HB, et al. Tardive dyskinesia associated with use of metoc\opramide in a child. J Pediatr 1992; 121: 983-5

© Adis Internalional limited. All rights reserved.

199

399. Sewell DD, Kodsi AB, Caligiuri MP, et al. Metoc\opramide and tardive dyskinesia. Bioi Psychiatry 1994; 36: 630-2

400. Robinson D, Omar SJ, Dangel C, et al. Metoc\opramide-in­duced extrapyramidal symptoms in a diabetic patient [letter]. JAm Geriatr Soc 1994; 42: 1307-8

401. Macucci M, Dotti MT, Federico A. Familial acute dystonic­dyskinetic syndrome with dominant inheritance after use of antidopaminergic antiemetic drugs. Mov Disord 1995; 10: 528-9

402. Assmann VC, Perquin WV, Touw DJ. Extrapyramidal move­ment disorders following the use of flunarizine. Ned Tijdschr Geneeskd 1988; 12: 1940-3

403. Sandyk R. Tardive dyskinesia-induced by sulpiride in a patient with hypothyroidism. Clin Neuropharmacol1986; 9: 100-1

404. Achiron A, Zoldan Y, Melamed E. Tardive dyskinesia-induced by sulpiride. Clin Neuropharmacol1990; 13: 248-52

405. Masmoudi K, Decocq G, Chetaille E, et al. Troubles ex­trapyramidaux induits par Ie veralipride (Agreal). A propos de cinq observations. Therapie 1995; 50: 451-4

406. Huang Cc. Persistent tardive dyskinesia associated with amox­apine therapy [letter]. Am J Psychiatry 1986; 143: 1069-70

407. Huang CC. Persistent tardive dyskinesia associated with amoxapine therapy: two case reports. Hillside J Clin Psychi­atry 1986; 8: 209-13

408. Gangat AE, Luiz HA, Kajee AH, et al. TricyC\ic-induced acute tardive dyskinesia: a case report [letter]. S Afr Med J 1987; 71: 729

409. Yassa R, Camille Y, Belzile L. Tardive dyskinesia in the course of antidepressant therapy: a prevalence study and review of the literature. J Clin Psychopharmacol 1987; 7: 243-6

410. Otani K, Kaneko S, Fukushima Y, et al. Involuntary movements associated with mianserin treatment: a case report [letter]. Br J Psychiatry 1989; 154: 113-4

411. Golden RN, Miller H, Evans DL. Trimipramine and dopamine receptor blockade [letter]. J Clin Psychiatry 1989; 50: 392-3

412. Katz SE. Tardive dyskinesia associated with molindone treat­ment [letter]. Am J Psychiatry 1990; 147: 124-5

413. Sparring-Bjorksten K, Walinder J. Does mianserin induce in­voluntary movements in brain damaged patients? Int Clin Psychopharmacol 1993;8: 203-4

414. Gersten SP. Tardive dyskinesia-like syndromes with clomipramine [letter]. Am J Psychiatry 1993; 150: 165-6

415. Budman CL, Bruun RD. Persistent dyskinesia in a patient re­ceiving fluoxetine [letter]. Am J Psychiatry 1991; 148: 1403

416. al-Adwani A. Brain damage and tardive dyskinesia [letter]. Br J Psychiatry 1995; 167: 410-1

417. Wils V. Extrapyramidal symptoms in a patient treated with fluvoxamine [letter]. J Neurol Neurosurg Psychiatry 1992; 55: 330-1

418. Arya DK, Szabadi E. DyskineSia associated with fluvoxamine. J Clin Psychopharmacol 1993; 13: 365-6

419. Sternbach H, Jordan S. Lithium-associated tardive dyskinesia. J Clin Psychopharmacol1990; 10: 143-4

420. Lazarus A. Tardive dyskinesia-like syndrome associated with lithium and carbamazepine. J Clin Psychopharmacol 1994; 14: 146-7

421. Ghadirian AM, Annable L, Belanger MC, et aI. A cross-sec­tional study of parkinsonism and tardive dyskinesia in lith­ium-treated affective disordered patients. J Clin Psychiatry 1996; 57: 22-8

422. Strauss A. Oral dyskinesia associated with buspirone use in an elderly woman [letter]. J Clin Psychiatry 1988; 49: 322-3

423. Simpson GM, Singh H. Buspirone and dyskinesia [letter]. J Clin Psychiatry 1988; 49: 50

Drug Safety 1997 Mar: 16 (3)

200

424. Lydiard RB. Buspirone revisited [letter]. 1 Clin Psychiatry 1989;50:308

425. Rock NL. Possible adverse effects of buspirone when used with other psychotropic drugs. 1 Clin Psychopharmacol 1990; 10: 80-1

426. Sandyk R. Orofacial dyskinesias associated with lorazepam therapy. Clin Pharm 1986; 5: 419-21

427. Sexson WR, Thigpen 1, Stajich GY. Stereotypic movements after lorazepam administration in premature neonates: a series and review of the literature. 1 Perinatol1995; 15: 146-149

428. Dooley 1, Camfield P, Buckley D, et al. Methsuximide-induced movement disorder. Pediatrics 1991; 88: 1291-2

429. Hauser RA, Olanow CWo Orobuccal dyskinesia associated with trihexyphenidyl therapy in a patient with Parkinson's disease. Mov Disord 1993; 8: 512-4

430. Linazasoro G. Anticholinergics and dyskinesia [letter]. Mov Disord 1994; 9: 689

431. Morisy L, Platt D. Hazards of high-dose meperidine [letter]. lAMA 1986; 255: 467-8

432. Merriam AE. Pemoline-induced abnormal involuntary move­ments. 1 Clin Psychopharmacol 1990; 10: 302-3

433. Schuller DE, Turkewitz D. Adverse effects of antihistamines. Postgrad Med 1986; 79: 75-86

434. Micheli F, Gatto E, Gene R, et al. Clenbuterol-induced tardive dyskinesia. Clin Neuropharmacol1991; 14: 427-31

435. Ramsay FM, Millard PH. Tardive dyskinesia in the elderly. Age Ageing 1986; 15: 145-50

436. Morgenstern H, Glazer WM, Niedzwiecki D, et al. The impact of neuroleptic medication on tardive dyskinesia: a meta-anal­ysis of published studies. Am 1 Public Health 1987; 77: 717-24

437. Wolf ME, DeWolfe AS, Mosnaim AD. Risk factors for tardive dyskinesia according to primary psychiatric diagnosis. Hill­side 1 Clin Psychiatry 1987; 9: 3-11

438. Yassa R, Jeste DY. Gender differences in tardive dyskinesia: a critical review of the literature. Schizophr Bull 1992; 18: 701-15

439. Muscettola G, Pampallona S, Barbato G, et al. Persistent tardive dyskinesia: demographic and pharmacological risk factors. Acta Psychiatr Scand 1993; 87: 29-36

440. Ganzini L, Heintz RT, Hoffman WF, et al. The prevalence of tardive dyskinesia in neuroleptic-treated diabetics. A control­led study. Arch Gen Psychiatry 1991; 48: 259-63

441. Ganzini L, Casey DE, Hoffman WF, et al. Tardive dyskinesia and diabetes mellitus. Psychopharmacol Bull 1992; 28: 281-6

442. Woerner MG, Saltz BL, Kane 1M, et al. Diabetes and develop­ment of tardive dyskinesia. Am 1 Psychiatry 1993; 150: 966-88

443. Pourcher E, Baruch P, Bouchard RH, et al. Neuroleptic associ­ated tardive dyskinesias in young people with psychoses. Br 1 Psychiatry 1995; 166: 768-72

444. Ueyama K, Fukuzako H, Takeuchi K, et al. Brain atrophy and intellectual impairment in tardive dyskinesia. Ipn 1 Psychiatry Neuro11993; 47: 99-104

445. Dinan TG, Kohen D. Tardive dyskinesia in bipolar affective disorder: relationship to lithium therapy. Br J Psychiatry 1989; 155: 55-7

446. Waddington lL, Youssef HA. An unusual cluster of tardive dys­kinesia in schizophrenia: association with cognitive dysfunc­tion and negative symptoms. Am 1 Psychiatry 1986; 143: 1162-5

447. Dixon L, Weiden PI, Haas G, et al. Increased tardive dyskinesia in alcohol-abusing schizophrenic patients. Compr Psychiatry 1992; 33: 121-2

448. Yassa R, Nair V, Schwartz G. Early versus late onset psychosis and tardive dyskinesia. Bioi Psychiatry 1986; 21: 1291-7

© Adis International Limited. All rights reserved.

Jimenez-Jimenez et al.

449. Yassa R. Tardive dyskinesia and anticholinergic drugs. A critical review of the literature. Encephale 1988; 14 Spec No: 233-9

450. Cavallaro R, Smeraldi E. Antipsychotic-induced tardive dyski­nesia: recognition, prevention and management. CNS Drugs 1995; 4: 278-293

451. Obeso lA, Artieda 1, Marsden CD. Different clinical presenta­tions of myoclonus. In: lankovic 1, Tolosa E, editors. Parkin­son's disease and movement disorders, 2nd ed. Baltimore: Williams & Wilkins: 315-28

452. Forstl H, Pohlmann-Eden B, Rothenberger A. Veranderungen somatosensibel evozierter Potentiale bei pharmakogener Myoklonie. Nervenarzt 1992; 63: 359-62

453. Brogmus KE, Lesch A. Psychopharmakainduzierte Myo­klonien. Psychiatr Prax 1995; 22: 77-9

454. Drouet A, Valance J. Myoclonies de repos et d'action induites par la piperazine. Rev Med Interne 1994; 15: 364-5

455. Tominaga H, Fukuzako H, Izumi K, et al. Tardive myoclonus [letter]. Lancet 1987; I: 322

456. Fukuzako H, Tominaga H, Izumi K, et al. Postural myoclonus associated with long-term administration of neuroleptics in schizophrenic patients. Bioi Psychiatry 1990; 27: 1116-26

457. Little IT, lankovic 1. Tardive myoclonus. Mov Disord 1987; 2: 307-11

458. Noble 1, Matthew H. Acute poisoning by tricyclic antidepres­sants: clinical features and management of 100 patients. Clin Toxicol 1969; 2: 403-21

459. Burks IS, Walker JE, Rumack BH, et al. Tricyclic antidepres­sant poisoning; reversal of coma, choreoathetosis and myoc­lonus by physostigmine. lAMA 1974; 230: 1405-7

460. Darcourt G, Fadeuilhe A, Lavagna 1, et al. Trois cas de myoclon­ies d'action au cors de traitements par l'imipramine et l'amitriptyline. Rev Neurol (Paris) 1970; 122: 141-2

461. Lippmann S, Moskovitz R, O'Tuama L. Tricyclic-induced my­oclonus [letter]. Am 1 Psychiatry 1977; 134: 90-1

462. Garvey MJ, Tollefson GD. Occurrence of myoclonus in patients treated with cyclic antidepressants. Arch Gen Psychiatry 1987; 44: 269-72

463. Casas M, Garcia-Ribera C, Alvarez E, et al. Myoclonic move­ments as a side-effect of treatment with therapeutic doses of clomipramine. Int Clin Psychopharmacol 1987; 2: 333-6

464. Devanand DP, Sackeim HA, Brown RP. Myoclonus during combined tricyclic antidepressant and lithium treatment. 1 Clin Psychopharmacol 1988; 8: 446-7

465. Fukuzako H, Hokazono Y, Tominaga H, et al. Jerk-locked av­eraging and somatosensory evoked potential in tricyclic­induced myoclonus: a case report. Ipn 1 Psychiatry Neurol 1989; 43: 645-9

466. Forstl H, Pohlmann-Eden B. Amplitudes of somatosensory evoked potentials reflect cortical hyperexcitability in antide­pressant-induced myoclonus. Neurology 1990; 40: 924-6

467. Patterson IE Myoclonus caused by a tricyclic antidepressant. South Med 1 1990; 83: 463-5

468. Lejoyeux M, Ades 1, Rouillon E Myoclonies chez les patients traites par antidepresseurs tricycliques. Evaluation prospec­tive [letter]. Presse Med 1994; 23: 447

469. Black Kl, Kilzieh N. Severe imipramine-induced myoclonus in a patient with psychotic bipolar depression, catatonia, and schizencephaly. Ann Clin Psychiatry 1994; 6: 45-9

470. Patel HC, Bruza D, Yeragani Y. Myoclonus with trazodone [let­ter].l Clin Psychopharmacol 1988; 8: 152

471. Feighner IP, Boyer WF, Tyler DL, et al. Adverse consequences of fluoxetine-MAOI combination therapy. 1 Clin Psychiatry 1990; 51: 222-5

Drug Safety 1997 Mar; 16 (3)

Drug-Induced Movement Disorders

472. Goldberg RJ, Huk M. Serotonin syndrome from trazodone and buspirone [letter]. Psychosomatics 1992; 33: 235-6

473. Jahr JS, Pisto JD, Gitlin MC, et al. The serotonin syndrome in a patient receiving sertraline after an ankle block. Anesth An­alg 1994; 79: 189-91

474. Lauterbach EC. Reversible intermittent rhythmic myoclonus with fluoxetine in presumed Pick's disease. Mov Disord 1994; 9: 343-6

475. White PD. Myoclonus and episodic delirium associated with phenelzine: a case report [letter]. J Clin Psychiatry 1987; 48: 340-1

476. Naylor MW, Grunhaus L, Cameron O. Myoclonic seizures after abrupt withdrawal from phenelzine and alprazolam. J Nerv Ment Dis 1987; 175: 111-4

477. Askenasy JJ, Yahr MD. Is monoamine oxidase inhibitor­induced myoclonus serotoninergically mediated? J Neural Transm 1988; 72: 67-76

478. Klawans HL, Goetz C, Bergen D. Levodopa-induced myoclo­nus. Arch Neurol 1975; 32: 331-4

479. Hirschorn KA, Greenberg HS. Successful treatment of levo­dopa-induced myoclonus and levodopa withdrawal-induced neuroleptic malignant syndrome. A case report. Clin Neu­ropharmacol 1988; II: 278-81

480. Glantz R, Weiner WJ, Goetz CG, et al. Drug-induced asterixis in Parkinson's disease. Neurology 1982; 2: 553-5

481. Goule JP, Husson A, Fondimare A, et al. Encephalopatie aux sels insolubles de bismuth [letter]. Nouv Presse Med 1975; 4: 1366

482. Buge A, Rancurel G, Dechy H. Encephalopathies myocloniques bismuthiques. Formes evolutives, complica­tions tardives, durables ou definitives. A propos de 41 cases. Rev Neurol (Paris) 1977; 133: 401-15

483. Nogue S, Mas A, Pares A, et al. Encefalopatia mioclonica y convulsiva por bismuto. Utilidad del tratamiento con dimer­caprol. Med Clin (Bare) 1985; 84: 530-2

484. Vidailhet M, Le TH, Wechsler B, et al. L'encephalopathie au bismuth n'a pas disparu ... [letter]. Presse Med 1987; 16: 1054

485. Molina JA, Calandre L, Bermejo F. Myoclonic encephalopathy due to bismuth salts: treatment with dimercaprol and analysis ofCSF transmitters. Acta Neurol Scand 1989; 79: 200-3

486. Gomez-Jimenez J, Alvarez J, Boada-Rovira M, et al. Encelafopatia mioclonica por bismuto [letter]. Rev Clin Esp 1989; 184: 212

487. Molina JA, Calandre L, Bermejo F, et al. Encefalopatia mioclonica por sales de bismuto. Eficacia del tratamiento con dimercaprol. Med Clin (Barc) 1989; 93: 20-2

488. Mendelowitz PC, Hoffman RS, Weber S. Bismuth absorption and myoclonic encephalopathy during bismuth subsalicylate therapy [letter]. Ann Intern Med 1990; 112: 140-1

489. Nogue-Xarau S, Sanz-Gallen P. Encefalopatia mioclonica por bismuto [letter]. Rev Clin Esp 1990; 186: 145

490. Jungreis AC, Schaumburg HH. Encephalopathy from abuse of bismuth subsalicylate (Pepto-Bismol). Neurology 1993; 43: 1265

491. Gordon MF, Abrams RI, Rubin DB, et al. Bismuth subsalicylate toxicity as a cause of prolonged encephalopathy with myoc­lonus. Mov Disord 1995; 10: 220-2

492. Klawans HL, Carvey PM, Tanner CM, et al. Drug-induced my­oclonus. Adv Neuro11986; 43: 251-64

493. Aguglia U, Zappia M, Quattrone A. Carbamazepine-induced nonepileptic myoclonus in a child with benign epilepsy. Epilepsia 1987; 28: 515-8

© Adis International Umited. All rights reserved.

201

494. Dhuna A, Pascual-Leone A, Talwar D. Exacerbation of partial seizures and onset of nonepileptic myoclonus with carbam­azepine. Epilepsia 1991; 2: 275-8

495. AgugJia U, Gambardella A, Zappia M, et al. Negative myoclo­nus during valproate-related stupor. Neurophysiological evi­dence of a cortical non-epileptic origin. Electroencepha10gr Clin Neurophysiol1995; 94: 103-8

496. Lemus CZ, Lieberman JA, Johns CA. Myoclonus during treat­ment with clozapine and lithium: the role of serotonin. Hill­side J Clin Psychiatry 1989; II: 127-30

497. Rosen PD, Stevens R. Action myoclonus in lithium toxicity. Ann Neuro11983; 13: 221-2

498. Julius SC, Brenner RP. Myoclonic seizures with lithium. Bioi Psychiatry 1987; 22: 1184-90

499. Prettyman R. Lithium neurotoxicity at subtherapeutic serum levels [letter]. Br J Psychiatry 1994; 164: 123

500. Glavina MJ, Robertshaw R. Myoclonic spasms following in­trathecal morphine. Anaesthesia 1988; 43: 389-90

501. Potter JM, Reid DB, Shaw RJ, et al. Myoclonus associated with treatment with high doses of morphine: the role of supple­mental drugs. BMJ 1989; 299: 150-3

502. Jayawardena B, Hill DJ. Myoclonic spasms after epidural diamorphine infusion. Anaesthesia 1991; 46: 473-4

503. De Conno F, Caraceni A, Martini C, et al. Hyperalgesia and myoclonus with intrathecal infusion of high-dose morphine. Pain 1991; 47: 337-9

504. Franco ML, Berro MJ, Saez JM, et al. Mioclonias de miembros inferiores como complicacion de la morfina espinal. Rev Esp Anestesiol Reanim 1992; 39: 192-3

505. Sjogren P, Jonsson T, Jensen NH, et al. Hyperalgesia and my­oclonus in terminal cancer patients treated with continuous intravenous morphine. Pain 1993; 55: 93-7

506. Cartwright PD, Hesse C, Jackson AO. Myoclonic spasms fol­lowing intrathecal diamorphine. J Pain Symptom Manage 1993; 8: 492-5

507. de-Armendi AJ, Fahey M, Ryan JF. Morphine-induced myoclo­nic movements in a pediatric pain patient. Anesth Analg 1993; 77: 191-2

508. Bowdle TA, Rooke GA. Postoperative myoclonus and rigidity after anesthesia with opioids. Anesth Analg 1994; 78: 783-6

509. Kloke M, Bingel U, Seeber S. Complications of spinal opioid therapy: myoclonus, spastic muscle tone and spinal jerking. Support Care Cancer 1994; 2: 249-52

510. Jacobsen LS, Olsen AK, Sjogren P, et al. Morphine-induced hyperalgesia, allodynia and myoclonus: new side-effects of morphine?UgeskrLaeger 1995; 157: 3307-10

511. Meloni G, Raucci U, Pinto RM, et al. Pretransplant condition­ing with busulfan and cyclophosphamide in acute leukemia patients: neurological and electroencephalographic prospec­tive study. Ann Oncol 1992; 3: 145-8

512. Martin M, Diaz-Rubio E, Casado A, et al. Prednimustine-in­duced myoclonus: a report of three cases. Acta Oncol 1994; 33: 81-2

513. Ammenti A, Reither B, Muller-Wiefel DE. Chlorambucil neu­rotoxicity: report of two cases. Helv Paediatr Acta 1980; 35: 281-7

514. Byrne TN Jr, Moseley TAE lll, Finer MA. Myoclonic seizures following chlorambucil overdose. Ann Neuro11981; 9: 191-4

515. LaDelfa I, Bayer N, Myers R, et al. Chlorambucil-induced myoclonic seizures in an adult. J Clin Onco11985; 3: 1691-2

516. Buchman AS, Bennett DA, Goetz CG. Bromocriptine-induced myoclonus. Neurology 1987; 37: 885

Drug Safety 1997 Mar: 16 (3)

202

517. Ritchie EC, Bridenbaugh RH, Jabbari B. Acute generalized my­oclonus following buspirone administration. J Clin Psychiatry 1988; 49: 242-3

518. Lee DS, Wong HA, Knoppert DC. Myoclonus associated with lorazepam therapy in very-Iow-birth-weight infants. Bioi Ne­onate 1994; 66: 311-5

519. Magny JF, d'Allest AM, Nedelcoux H, et al. Midazolam and myoclonus in neonate. Eur J Pediatr 1994; 15: 389-90

520. Madigand M, Dien J, Pavin G, et al. Encephalopathie myoclonique probablement imputable 11 la propafenone [let­ter]. Presse Med 1988; 17: 538

521. Jeandel C, Bannwarth B, Trechot P, et al. Encephalopathie myoclonique 11 la propafenone. Therapie 1990; 45: 161-2

522. Chua TP, Farrell T, Lipkin DP. Myoclonus associated with pro­pafenone [letter]. BMJ 1994; 308: 113

523. Ghika J, Goy 11, Naegeli C, et al. Acute reversible ataxo­myoclonic encephalopathy with flecainide therapy. Schweiz Arch Neurol Psychiatr 1994; 145: 4-6

524. Jeret JS, Somasundaram M, Asaikar S. Diltiazem-induced my­oclonus. N Y State J Med 1992; 92: 447-8

525. Pedro-Botet ML, Bonal J, Caralps A. Nifedipine and myoclonic disorders [letter]. Nephron 1989; 51: 281

526. Lucas C, Soetaert G, Leys D, et al. Myoclonies au cours d'un traitement par buflomedil. Acta Clin Belg 1989; 44: 360-1

527. Uchihara T, Tsukagoshi H. Myoclonic activity associated with cefmetazole, with a review of neurotoxicity of cephalospo­rins. Clin Neurol Neurosurg 1988; 90: 369-71

528. New PS, Wells CEo Cerebral toxicity associated with massive intravenous penicillin therapy. Neurology 1965; 15: 1052-8

529. Cohill DF, Pezzi PJ, Greenberg SR, Frobese AS. Central nerv­ous system toxicity secondary to massive doses of penicillin 'G' in the treatment of overwhelming infections. Am J Med Sci 1967; 254: 692-4

530. Lerner PI, Smith N, Weinstein L. Penicillin neurotoxicity. Ann NY Acad Sci 1967; 145: 310-8

531. Conway N, Beck E, Somerville J. Penicillin encephalopathy. Postgrad Med J 1968; 44: 891-7

532. Durand JM, Telle H, Quiles N, et al. Syndrome confusionnel, myoclonies et traitement par la pefloxacine. Ann Med Interne (Paris) 1993; 144: 495-6

533. Yagi S, Moriya 0, Nakajima M, et al. A case of tuberculous pleurisy associated with myoclonus and Quincke's edema due to isoniazid and isoniazid sodium methanesulfonate. Kekkaku 1989;64:407-12

534. Fukuda T. Veratramine-induced myoclonus. Seishin Shin­keigaku Zasshi 1990; 92: 865-9

535. Bouly A, Nathan N, Feiss P. Propofol in myotonic dystrophy [letter]. Anaesthesia 1991; 46: 705

536. Hughes NJ, Lyons JB. Prolonged myoclonus and meningism following propofol. Can J Anaesth 1995; 42: 744-6

537. Mayeux R, Albert M, Jenike M. Physostigmine-induced myoc­lonus in Alzheimer's disease. Neurology. 1987; 7: 345-6

538. Reutens DC, Stewart-Wynne EG. Norpethidine-induced myoc­lonus in a patient with renal failure [letter]. J Neurol Neuro­surg Psychiatry 1989; 52: 1450-1

539. Sica DA, Comstock TJ. Pseudoephedrine accumulation in renal failure. Am J Med Sci 1989; 298: 261-3

540. Korthals JK, Gieron-Korthals MA. Eosinophilia-myalgia syn­drome with myoclonic jerks. J Fla Med Assoc 1990; 77: 1031-3

541. Berman I, Zalma A, Durand CJ, et al. Clozapine-induced myoclonic jerks and drop attacks [letter]. J Clin Psychiatry 1992; 53: 329-30

© Adis Internalional Limited. All rights reserved.

Jimenez-Jimenez et al.

542. Malow BA, Reese KB, Sato S, et al. Spectrum of EEG abnor­malities during clozapine treatment. Electroencephalogr Clin Neurophysiol1994; 91: 205-11

543. Meltzer HY, Ranjan R. Valproic acid treatment of clozapine-in­duced myoclonus [letter]. Am J Psychiatry 1994; 151: 1246-7

544. Bak TH, Bauer M, Schaub RT, et al. Myoclonus in patients treated with clozapine: a case series. J Clin Psychiatry 1995; 56: 418-22

545. Eisele G. Neurologic complications of metoclopramide therapy [letter]. N Y State J Med 1988; 88: 332

546. Klawans HL, Falk DK, Nausieda PA, et al. Gilles de la Tourette syndrome after long-term chlorpromazine therapy. Neurology 1978; 28: 1064-8

547. De Veaugh-Geiss J. Tardive Tourette syndrome. Neurology 1980; 30: 562-3

548. Stahl SM. Tardive Tourette syndrome in an autistic patient after long-term neuroleptic administration [letter]. Am J Psychiatry 1980; 30: 562-3

549. Fog R, Pakkenberg H. Theoretical and clinical aspects of the Tourette syndrome (chronic multiple tic). J Neura! Transm 1980; 16 Suppl.: 211-5

550. Seeman MV, Patel J, Pyke J. Tardive dyskinesia with Tourette­like syndrome [letter]. J Clin Psychiatry 1981; 42: 357-8

551. Singer DW. Transient Gilles de la Tourette syndrome after chronic neuroleptic withdrawal. Dev Med Child Neuro11981; 23: 518-30

552. Mueller J, AminoffMA. Tourette-like syndrome after long-term neuroleptic drug treatment. Br J Psychiatry 1982; 141: 191-3

553. Klawans HL, Nausieda PA, Goetz CC, et al. Tourette-like symp­toms following chronic neuroleptic therapy. Adv Neurol 1982; 35: 415-8

554. Jeste DV, Cutler NR, Kaufmann CA. Low-dose apomorphine and bromocriptine in neuroleptic-induced movement disor­ders. Bioi Psychiatry 1983; 18: 1085-91

555. Munetz MR, Slawsky RC, Neil JF. Tardive Tourette's syndrome treated with clonidine and mesoridazine. Psychosomatics 1985; 26: 254-7

556. La! S, Al Ansari E. Tourette-like syndrome following low dose short-term neuroleptic treatment. Can J Neurol Sci 1986; 13: 125-8

557. Casat CD, Wilson DC III. Tics with combined thioridazine­methylphenidate therapy: case report. J Clin Psychiatry 1986; 47: 44-5

558. Gualtieri CT, Patterson DR. Neuroleptic-induced tics in two hyperactive children. Am J Psychiatry 1986; 143: 1176-7

559. Karagianis JL, Nagpurkar R. A case of Tourette syndrome de­veloping during haloperidol treatment. Can J Psychiatry 1990; 35: 228-32

560. Kuniyoshi M, Inanaga K, Arikawa K, et al. A case of tardive Tourette-like syndrome. Jpn J Psychiatry Neurol 1992; 46: 67-70

561. Bharucha KJ, Sethi KD. Tardive tourettism after exposure to neuroleptic therapy. Mov Disord 1995; 10: 791-3

562. Lindenmayer JP, Da-Silva D, Buendia A, et al. Tic-like syn­drome after treatment with clozapine [letter]. Am J Psychiatry 1995; 152:649

563. Weinstein D, See RE, Ellison G. Delayed appearance of facial tics following chronic fluphenazine administration to guinea pigs. Pharmacol Biochem Behav 1989; 32: 1057-60

564. Factor SA, Sanchez-Ramos JR, Weiner WJ. Cocaine and Tourette's syndrome [letter]. Ann Neurol 1988; 23: 423-4

565. Pascual-Leone A, Dhuna A. Cocaine-associated multifocal tics. Neurology 1990; 40: 999-1000

Drug Safety 1997 Mar; 16 (3)

Drug-Induced Movement Disorders

566. Attig E, Amyot R, Botez T. Cocaine-induced chronic tics [let­ter]. J Neurol Neurosurg Psychiatry 1994; 57: 1143-4

567. Golden GS. The relationship between stimulant medication and tics. Pediatr Ann 1988; 17: 405-6

568. Sverd J, Gadow KD, Paolicelli LM. Methylphenidate treatment of attention-deficit hyperactivity disorder in boys with Tourette's syndrome. J Am Acad Child Adolesc Psychiatry 1989; 28: 574-9

569. Chandler ML, Barnhill JL, Gualtieri CT, et al. Tryptophan an­tagonism of stimulant-induced tics. J Clin Psychopharmacol 1989; 9: 69-70

570. Stevenson RD, Wolraich ML. Stimulant medication therapy in the treatment of children with attention deficit hyperactivity disorder. Pediatr Clin North Am 1989; 6: 1183-97

571. Borcherding BG, Keysor CS, Rapoport JL, et al. Motor/vocal tics and compulsive behaviors on stimulant drugs: is there a common vulnerability? Psychiatry Res 1990; 33: 83-94

572. Lipkin PH, Goldstein 11, Adesman AR. Tics and dyskinesias associated with stimulant treatment in attention-deficit hyper­activity disorder. Arch Pediatr Adolesc Med 1994; 148: 859-61

573. Bruun R, Kurian R. Opiate therapy and self-harming behavior in Tourette's syndrome. Mov Disord 1991; 6: 184-5

574. Lichter D, Majumdar L, Kurian R. Opiate withdrawal unmasks Tourette's syndrome. Clin Neuropharmacol1988; 11: 559-64

575. Sandyk R. Naloxone withdrawal exacerbates Tourette syn­drome. J Clin Psychopharmacol 1986; 6: 58-9

576. Sandyk R: Phenobarbital-induced Tourette-Iike symptoms. Pediatr Neurol 1986; 2: 54-5

577. Burd L, Kerbeshian J, Fisher W, et al. Anticonvulsant medica­tions: an iatrogenic cause of tic disorders. Can J Psychiatry 1986; 31: 419-23

578. Kurian R, Kersun J, Behr J, et al. Carbamazepine-induced tics. Clin Neuropharmacol1989; 12: 298-302

579. Robertson PL, Garofalo EA, Silverstein FS, et al. Carbamaze­pine-induced tics. Epilepsia 1993; 34: 965-8

580. Gillman MA, Sandyk R. Clonazepam-induced Tourette syn­drome in a subject with hyperexplexia. Postgrad Med J 1987; 63: 311-2

581. Eisenhauer G, Jermain DM. Fluoxetine and tics in an adoles­cent. Ann Pharmacother 1993; 27: 725-6

582. Cunningham M, Cunningham K, Lydiard RB. Eye tics and sub­jective hearing impairment during fluoxetine therapy [letter]. Am J Psychiatry 1990; 147: 947-8

583. Shale H, Fabn S, Mayeux R. Tics in a patient with Parkinson's disease. Mov Disord 1986; 1: 79-83

584. Lang AE. Akathisia and the restless legs syndrome. In: Jankovic J, Tolosa E, editors. Parkinson's disease and movement dis­orders, 2nd ed. Baltimore: Williams & Wilkins: 399-418

585. Sandyk R, Kay SR. Relationship of neuroleptic-induced akathi­sia to drug-induced parkinsonism. Ital J Neurol Sci 1990; 11: 439-42

586. Barnes TR, Halstead SM, Little PW. Relationship between iron status and chronic akathisia in an in-patient population with chronic schizophrenia. Br J Psychiatry 1992; 161: 791-6

587. Berardi D, Zanfini R, Alessi S, et al. Effetti disforizzanti dei neurolettici in pazienti non schizofrenici. Minerva Psichiatr 1993; 34: 125-33

588. Sachdev P, Kruk J. Clinical characteristics and predisposing factors in acute drug-induced akathisia. Arch Gen Psychiatry 1994; 51: 963-74

589. Sachdev P. The epidemiology of drug-induced akathisia: Part I. Acute akathisia. Schizophr Bull 1995; 21: 431-49

© Adis International Umited. All rights reserved.

203

590. Sachdev P. The epidemiology of drug-induced akathisia: Part II. Chronic, tardive, and withdrawal akathisias. Schizophr Bull 1995; 21: 451-61

591. Cohen BM, Keck PE, Satlin A, et al. Prevalence and severity of akathisia in patients on c1ozapine. BioI Psychiatry 1991; 29: 1215-9

592. Fagan D, Scott DB, Mitchell M, et al. Effects of remoxipride on measures of psychological performance in healthy volun­teers. Psychopharmacology (Berl) 1991; 105: 225-9

593. Safferman AZ, Lieberman JA, Pollack S, et al. Akathisia and c10zapine treatment. J Clin Psychopharmacol1993; 13: 286-7

594. Friedman JH. Akathisia with c10zapine [letter]. BioI Psychiatry 1993; 33: 852-3

595. Chengappa KN, Shelton MD, Baker RW, et al. The prevalence of akathisia in patients receiving stable doses of c1ozapine. J Clin Psychiatry 1994; 55: 142-5

596. Byerly MJ, Greer RA, Evans DL. Behavioral stimulation asso­ciated with risperidone initiation [letter]. Am J Psychiatry 1995; 152: 1096-7

597. Ward NG. Akathisia associated with droperidol during epidural anesthesia. Anesthesiology 1989; 71: 786-7

598. Athanassiadis C, Karamanis A. Akathisia after long-term epi­dural use of droperidol: a case report. Pain 1992; 50: 203-4

599. Hamilton PA. Metoclopramide-induced akathisia. Mil Med 1987; 152: 585-6

600. Sorbe B, Hallen C. Antiemetic treatment of chemotherapy­induced nausea in ovarian carcinoma patients. Gynecol Oncol 1989; 34: 141-4

601. Beightol RW, Coupal JJ, Shih WJ. Prevention ofmetoclopram­ide-induced akathisia during gastric emptying imaging. J Nucl Med 1991; 32: 1644-5

602. Cory DA. Adverse reaction to metocIopramide during en­terocIysis [letter]. Am J Roentgenol1994; 163: 480

603. Fleishman SB, Lavin MR, Sattler M, et al. Antiemetic-induced akathisia in cancer patients receiving chemotherapy. Am J Psychiatry 1994; 151: 763-5

604. Weimer S. Akathisia in a cancer patient treated with antiemetics [letter]. Am J Psychiatry 1995; 152: 960-1

605. Lipinski Jr JF, Mallya G, Zimmerman P, et al. Fluoxetine-in­duced akathisia: clinical and theoretical implications. J Clin Psychiatry 1989; 50: 339-42

606. Maany I, Dhopesh V. Akathisia and fluoxetine. J Clin Psychia­try 1990; 51: 210-2

607. Friedman EH. Fluoxetine-induced akathisia in male OCD pa­tients [letter]. J Clin Psychiatry 1990; 51: 212

608. Rothschild AJ, Locke CA. Reexposure to fluoxetine after seri­ous suicide attempts by three patients: the role of akathisia. J Clin Psychiatry 1991; 52: 491-3

609. Fleischhacker WW. Propranolol for fluoxetine-induced akathi­sia [letter]. BioI Psychiatry 1991; 30: 531-2

610. Bertschy G, Vandel S. Fluoxetine-related indifference and akathisia. A case report. Therapie 1993; 48: 158-9

611. Marshall RD, Printz D, Cardenas D, et al. Adverse events in PTSD patients taking fluoxetine [letter]. Am J Psychiatry 1995; 152: 1238-9

612. Ketai R. Interaction between fluoxetine and neuroleptics [let­ter]. Am J Psychiatry 1993; ISO: 836-7

613. KIee B, Kronig MH. Case report of probable sertraline-induced akathisia [letter]. Am J Psychiatry 1993; 150: 986-7

614. LaPorta LD. Sertraline-induced akathisia. J Clin Psy­chopharmacol 1993; 13:219-20

615. Settle EC Jr. Akathisia and sertraline [letter]. J Clin Psychiatry 1993;54: 321

Drug Safety 1997 Mar: 16 (3)

204

616. Opler LA. Sertraline and akathisia [letter]. Am J Psychiatry 1994; 151: 620-1

617. Altshuler LL, Pierre JM, Wirshing WC, et al. Sertraline and akathisia. J Clin Psychopharmacol 1994; 14: 278-9

618. Phillips SD. A possible paroxetine withdrawal syndrome [let­ter]. Am J Psychiatry 1995; 152: 645-6

619. Adler LA, Angrist BM. Paroxetine and akathisia [letter]. Bioi Psychiatry 1995; 37: 336-7

620. Klysner R, Larsen JK, Sorensen P, et al. Toxic interaction of venlafaxine and isocarboxazide [letter]. Lancet 1995; 346: 1298-9

621. Zubenko GS, Cohen BM, Lipinski JF Jr. Antidepressant-related akathisia. J Clin Psychopharmacol 1987; 7: 254-7

622. Yeragani VK, Pohl R, Balon R. TricyC\ics, akathisia, and jitteri­ness [letter]. J Clin Psychopharmacol 1988; 8: 295

623. Sabaawi M, Richmond DR, Fragala MR. Akathisia in associa­tion with nortriptyline therapy. Am Fam Physician 1993; 48: 1024-6

624. Phillips SD, Ringo P. Phenelzine and venlafaxine interaction [letter]. Am J Psychiatry 1995; 152: 1400-1

625. Gillman PK. Possible serotonin syndrome with moclobemide and pethidine [letter]. Med J Aust 1995; 162: 554

626. Bakchine S, Lacomblez L, Soubrie C. Syndrome extrapyra­midal au cours d'un traitement par la flunarizine. Rev Neurol (Paris) 1988; 144: 833-4

627. Rodriguez-Garda JL, Arechaga-Uriarte S. Akatisia inducida por flunarizina [letter]. Rev Clin Esp 1991; 188: 384

628. Price WA, Zimmer B. Lithium-induced akathisia [letter]. J Clin Psychiatry 1987; 48: 81

629. Patterson JE Lithium-induced akathisia [letter]. J Clin Psy­chopharmacoI1988;8:445

630. Patterson JF. Akathisia associated with buspirone. J Clin Psy­chopharmacol 1988; 8: 296-7

631. Lawrence K. Reversal of midazolam sedation. Crit Care Med 1993; 21: 1407-8

© Adis International Umited. All rights reserved.

Jimenez-Jimenez et al.

632. Honan VJ. Paradoxical reaction to midazolam and control with flumazenil. Gastrointest Endosc 1994; 40: 86-8

633. Hollman GA. Oral midazolam and emergence deli.rium. Ann Emerg Med 1995; 25: 853-4

634. Yassa R, Groulx B. Lorazepam in the treatment of lithium­induced akathisia. J Clin Psychopharmacol 1989; 9: 70-1

635. Phelps S1, Baldree LA, Boucher BA, et al. Neuropsychiatric toxicity of phenytoin. Importance of monitoring phenytoin levels. Clin Pediatr (Phila) 1993; 32: 107-10

636. Riche H, Salord F, Jourdan C, et al. Neurological side effects related to valproate after neurosurgery [letter]. Acta Neu­rochir (Wien) 1994; 127: 240

637. Bernick C. Methysergide-induced akathisia. Clin Neu­ropharmacol 1988; 11:87-9

638. Brown KW, Glen SE, White T. Low serum iron status and akathisia. Lancet 1987; I: 1234-6

639. Barton A, Bowie J, Ebmeier K. Low plasma iron status and akathisia. J Neurol Neurosurg Psychiatry 1990; 53: 671-4

640. O'Loughlin V, Dickie AC, Ebmeier KP. Serum iron and trans­ferrin in acute neuroleptic-induced akathisia [letter]. J Neurol Neurosurg Psychiatry 1991; 54: 363-4

641. Horiguchi J. Low serum iron in patients with neuroleptic-in­duced akathisia and dystonia under antipsychotic drug treat­ment. Acta Psychiatr Scand 1991; 84: 301-3

642. Sachdev P, Loneragan C. Acute drug-induced akathisia is not associated with low serum iron status. Psychopharmacology (Berl) 1991; 103: 138-9

643. Soni SD, Tench D, Routledge RC. Serum iron abnormalities in neuroleptic-induced akathisia in schizophrenic patients. Br J Psychiatry 1993; 163: 669-72

Correspondence and reprints: Dr Felix Javier Jimenez­Jimenez, C/ Corregidor Jose de Pasamonte 24, 3° D, E-28030 Madrid, Spain.

Drug Safely 1997 Mar: 16 (3)