136
Views
34
CrossRef citations to date
0
Altmetric
Review

Treatment of neurolept-induced tardive dyskinesia

Pages 1371-1380 | Published online: 16 Sep 2013

Abstract

Tardive dyskinesia (TDK) includes orobuccolingual movements and “piano-playing” movements of the limbs. It is a movement disorder of delayed onset that can occur in the setting of neuroleptic treatment as well as in other diseases and following treatment with other drugs. The specific pathophysiology resulting in TDK is still not completely understood but possible mechanisms include postsynaptic dopamine receptor hypersensitivity, abnormalities of striatal gamma-aminobutyric acid (GABA) neurons, and degeneration of striatal cholinergic interneurons. More recently, the theory of synaptic plasticity has been proposed. Considering these proposed mechanisms of disease, therapeutic interventions have attempted to manipulate dopamine, GABA, acetylcholine, norepinephrine and serotonin pathways and receptors. The data for the effectiveness of each class of drugs and the side effects were considered in turn.

Introduction

Tardive dyskinesia (TDK) is a complex involuntary movement disorder that typically occurs in patients treated with antipsychotic drugs, but it usually has a delayed onset. The original description was published by Schonecker in 1957,Citation1 about five years after the commencement of neuroleptic treatment in psychiatry. Neuroleptic drugs are dopamine receptor blocking agents (DRBAs) and include metoclopramide, a dopamine antagonist used as an antiemetic, but which is also an antipsychotic at high doses and has a risk of TDK of 1%–10%.Citation2,Citation3 TDK refers to the orobuccolingual movements, as well as the “piano-playing” movements, of the fingers. Tardive syndrome (TS) includes a wider range of abnormal movements of delayed onset occurring in the setting of treatment with neuroleptic agents. TS includes tardive stereotypy, tardive dystonia, tardive tremor, tardive myoclonus, tardive parkinsonism, and tardive akathisia. TDK occurs in 20%–40% of neurolept-treated patientsCitation4Citation6 with an incidence of 5% per year. Jeste et alCitation7 followed patients at 1–3 month intervals and showed a cumulative incidence of TDK of 26%, 52%, and 60% at 1, 2, and 3 years, respectively. The movements observed need to be distinguished from schizophrenia stereotypies and mannerisms, as well as other causes of orolingual movements, such as loose dentures. Movements similar to those seen in patients with TDK can also occur in other settings (), including Huntington’s disease and Wilson’s disease. These neurodegenerative conditions may manifest with psychiatric symptoms and/or abnormal movements. It is important to note that, in these syndromes, the movements are part of the syndrome and not a drug side effect.

Table 1 Differential diagnosis of tardive dyskinesia

The pathophysiology of TDK and its predilection for orobuccolingual and upper limb muscles are still not fully understood. The basal ganglia and thalamus are the subcortical structures that make up the skeletomotor circuit (SMC). The ventral striatum is primarily involved in the emotional frontal–subcortical functions;Citation8 whereas, the putamen mediates SMC function.Citation9 The SMC is involved in control of voluntary movement, but it does not connect directly with the spinal cord. The SMC facilitates desired motor activity and inhibits activity that is undesirable or in competition with the desired activity.Citation10 Prior to treatment with antipsychotics, activation from the motor cortex leads to increased activity in the select-on circuit in the SMC and to activation of the select-off pathway for competing motor behaviors. Dopaminergic input to the striatum enhances the select-on pathway through D1 receptors. D2 receptors (which function to inhibit) turn the control circuit on and limit competing behaviors. After antipsychotic treatment, D2 blockade results in loss of the control pathway with excess activity of the select-off (competing motor behaviors) circuit, but the D1 select-on function is unchanged. This altered circuitry may explain the acute akinetic effects of antipsychotics. Chronic antipsychotic treatment may result in D2 upregulation and sensitization with resultant excess activity in the control circuit, loss of the select-off circuit activity, and enhanced select-on activity through D1 unblocking, resulting in hyperkinesis and TDK. This focused selection model fits with the D2 upregulation hypothesis for TDK, but it ignores the GABA inhibition from the globus pallidus externa and the possibility of altered cholinergic transmission in TDK. This model, therefore, needs to be further refined to understand all the pathways involved in TDK. Further understanding of these pathways will also explain how deep brain stimulation is effective in treatment of TDK.Citation11

Risk factors for TDK include female sex, older age, higher drug dose, long-term treatment, race, pre-existing mood, movement or cognitive disorder, alcohol use, diabetes, and human immunodeficiency virus (HIV) positivity ().Citation6,Citation12,Citation13 However, unexpected effects of antipsychotics are well-reported with young, robust patients not tolerating small doses of drug, while older, feeble patients demonstrate no toxicity at high doses of antipsychotics.Citation14 It has also been noted that siblings of patients with schizophrenia are more prone to TDK than controls,Citation15 and spontaneous TDK can occur in treatment-naïve schizophrenic patients.Citation16 This increased incidence of TDK in siblings of patients and the occurrence of spontaneous TDK in drug-naïve patients have raised the possibility of a genetic basis for the pathophysiology of TDK, which may reflect variations in genes associated with pharmacokinetic and pharmacodynamic processes of antipsychotic drugs or genetic profiles/polymorphisms that affect responsiveness to treatment. This genetic predisposition/variation may explain some of the heterogeneity of schizophrenia. With regard to genetic polymorphisms of enzymes metabolizing antipsychotic drugs, there have been some studies to show that a poor metabolizer phenotype of CYP2D6 has a greater risk of TDK.Citation17Citation19 Studies of allelic association of CYP2D6 with TDK have, however, provided conflicting results.Citation20Citation22 An association between TDK and a particular CYP1A2 genotype has been suggestedCitation23 but not confirmed.Citation24 A recent meta-analysis of CYP2D6 polymorphisms and risk of TDK suggests a minor effect of alleles with reduced function, but bias could not be excluded.Citation25

Table 2 Risk factors for tardive dyskinesiaCitation6,Citation12Citation25

The neurotransmitters implicated in the pathophysiology of TDK include postsynaptic dopamine receptor hypersensitivity, abnormalities of striatal GABA neurons, and degeneration of striatal cholinergic interneurons.Citation6,Citation26,Citation27 Rosengarten et alCitation28 proposed that TDK may be the result of an imbalance in dopaminergic receptor function. The dopamine receptor hypersensitivity theory proposes that chronic dopamine antagonism results in gradual hypersensitization of dopamine receptors.Citation29 D2 receptor hypersensitivity has been demonstrated in rats,Citation30,Citation31 but direct evidence in humans is less strong; postmortem studies showing similar D2 receptor numbers between TDK and non-TDK patients.Citation32 Biochemical isolation of different dopamine receptor subtypes and demonstration of subtype specific ligandsCitation33 has permitted further investigation of the dopamine pathophysiology of TDK. Malik et alCitation34 studied the effects of D1, D2 and D3 agonists, and D3 antagonists on TDK in Cebus monkeys and demonstrated D3 agonists do indeed have an antidyskinetic effect. However, D3 agonists with D2 agonist effect as well, showed a greater improvement in dyskinesia than pure D3 agonists.

In Southeast Asian populations, there has been a suggestion of an association between certain D3 receptor polymorphisms and the development of TDK; however, this was not confirmed in a case control meta-analysis.Citation35 Similarly, the association between serotonin receptor gene polymorphisms and TDK remains controversial.Citation36Citation38 More recently, glutamatergic genes have also been implicated in the pathogenesis of TDK. Further, in a genome-wide association screening, Syu et alCitation39 have demonstrated that a single nucleotide polymorphism (SNP) in the heparin sulphate proteoglycan 2 gene (HSPG2) is associated with TDK in Japanese schizophrenic patients. This study was replicated in a Caucasian population, and a small association between an HSPG2 SNP and TDK was shown in an Ashkenazi Jewish cohort of schizophrenics.Citation40 Genome-wide studies, however, are flawed by the number of calculations required and statistical corrections needed, as well as the stringent criteria for TDK used in some studies (see Müller et alCitation41 for review). It has been suggested that the unifying theory for all the genes associated with TDK is that they all result in an abnormality of synaptic plasticity. Synaptic plasticity is modified by many factors, including the dopaminergic and GABAergic systems,Citation42Citation44 and a recent study showed that schizophrenic patients with multiple psychotic episodes had impaired synaptic plasticity.Citation45 This altered synaptic plasticity may explain the lack of improvement in TDK after withdrawal of antipsychotic medication, as well as the spontaneous dyskinesia in treatment-naïve schizophrenic patients and higher rate of dyskinesia in siblings.Citation46

The onset of TDK is typically insidious; beginning several years after the initiation of treatment. The Diagnostic and Statistical Manual of Mental Disorders criteria specifies that the shortest duration of exposure to DRBAs is at least 1 month in patients 60 years or older. TDK reaches its maximum severity fairly rapidly and, then, often stabilises. The most common course is a waxing and waning of mild-to-moderate symptoms over many years, and clinical worsening after a period of stabilization is unusual. Approximately 11% of patients improve, usually within 1–2 years of discontinuation of treatment.Citation6 In classic TDK, the movements are rarely disabling and usually do not bother the patient, but they are of concern to family members. If clinically significant, swallowing and speech may be affected and can result in weight loss.

In view of the proposed pathophysiology of TDK, therapeutic interventions have attempted to manipulate dopamine, GABA, acetylcholine, norepinephrine and serotonin, and the data for the effectiveness of each class of drugs will be considered in turn.

Prevention

As TDK is an iatrogenic disorder, the best means of (but not possible) treatment would be prevention. However, patients do require treatment with neuroleptic agents, and these drugs are often the best treatment for long-term psychiatric disorders. Patients (and families) should be advised of the risk of TDK prior to commencing the drug, and the smallest effective dose of the safest drug should be used. Close monitoring for features of TDK or TS should be continued and, if possible, the dose reduced when features are first noticed and/or consideration given to changing to a drug with a lower risk of TDK.

Atypical neuroleptics

Atypical neuroleptics were initially thought to be at lower risk of inducing TDK; however, three prospective studies do not support this notion.Citation47Citation49 Based on the dopamine hypersensitivity hypothesis (ie, prolonged dopamine antagonist therapy leads to dopamine hypersensitivity), both typical and atypical neuroleptic agents should suppress TDK, but the benefit is usually only in the short term and is greater with more potent agents.Citation50 Sakai et alCitation51 also showed that haloperidol upregulated GABA(a) receptor expression in the substantia nigra; whereas, newer generation drugs did not, providing an alternative reason for lower rates of TDK with newer drugs. There is little data to support the notion that atypical neuroleptics reduce TDK. There are case reports that quetiapineCitation52 and olanzapineCitation53 can reduce TDK, but risperidone is the only atypical neuroleptic drug for which reasonable data is available. A single double-blind trial showed benefit with risperidone over haloperidol and placebo. Risperidone treatment resulted in a greater reduction in orobuccolingual movements compared to limb movements.Citation54 However, at high doses of risperidone, there is a substantial risk of TDK.Citation55 Clozapine, a relatively weak dopamine 2 receptor blocker, has also been shown to reduce abnormal movements in several small studies.Citation56Citation59 Aripiprazole was developed to avoid TDK and is a partial D2 receptor agonist, the rationale being that lack of D2 blocking would lessen the amount of D2 hypersensitivity.Citation60Citation63 However, case series of aripiprazole-treated patients do not support this theory.Citation64,Citation65

Despite the lack of substantial evidence, some experts suggest switching to an atypical neuroleptic to treat TDK and the psychiatric disturbance but do not distinguish between the various drugs.Citation66 Atypical agents, however, have other side effects, including sedation, weight gain, diabetes, agranulocytosis, and psychiatric exacerbation. The risks and benefits should, therefore, be considered for each patient on an individual basis, given the varied results of existing studies.

Dopamine depleting agents

Tetrabenazine (TBZ; Xenazine®) is a potent, selective, reversible depletor of monoamines from nerve terminals. TBZ inhibits the vesicular monoamine transporter type 2 which, in humans, is expressed nearly exclusively in the brain. TBZ is rapidly metabolized in the liver by carbonyl reductase to stereoisomers of hydrotetrabenazine, some of which are potent inhibitors of vesicular monoamine transporter type 2.Citation67 TBZ has been reported to be effective in treatment of TDK and tics, alleviating symptoms in up to 50% of patients. In more severe cases, there appears to be an additive effect, using a combination of tetrabenazine, clozapine, and clonazepam.Citation68 In a recent systematic review, nine of eleven studies showed a benefit of tetrabenazine in the treatment of TDK.Citation69 The largest prospective single-blinded study of TBZ included 20 patients with TDK present for 2–420 months and not responsive to other treatments. Seventeen out of 20 patients showed marked or moderate improvements as per the Abnormal Involuntary Movement Scale (AIMS).Citation70 Reserpine is also an inhibitor of vesicular monoamine transport, thereby depleting stores of presynaptic dopamine. Tetrabenazine, however, has a quicker onset of action and fewer peripheral catecholamine-depleting effects, compared to reserpine. In favor of these drugs, neither has been shown to cause TDK, and while larger trials would be helpful, the available data support the use of tetrabenazine for suppression of TDK.Citation67Citation69 It should, however, be noted that rapid increase in dose of tetrabenazine can cause other side effects, including marked rigidity/parkinsonism.Citation67

GABA agonists

A systematic review of benzodiazepines in the treatment of TDK concluded that there is no clear evidence that benzodiazepines are effective in suppressing TDK, and use of benzodiazepines remains experimental.Citation71 Nevertheless, there are studies of diazepam and clonazepam (GABA–A agonist) that show these drugs have some effect in reducing abnormal motor movements in patients with TDK, with 40%–50% of patients reporting some improvement.Citation72,Citation73 Thaker et alCitation74 conducted a randomized controlled crossover study of 19 patients and showed a 35% reduction in TDK with clonazepam; Bobruff et alCitation75 also reported a significant reduction in TDK with clonazepam. There are conflicting reports on the efficacy of baclofen (a presynaptic GABA–B agonist) with it showing no effect alone, but having significant benefit when combined with neuroleptic agents.Citation76,Citation77

Piracetam is a cyclic derivative of GABA. Its exact mechanism of action is uncertain. A randomized controlled trial of piracetam showed it to be effective in treatment of TDK.Citation78 Abrupt cessation can, however, precipitate seizures.

Levetiracetam, structurally similar to piracetam, has also been shown to be effective in treatment of TDK in case reports, open-label studies, and in a randomized placebo-controlled trial.Citation79 Gabapentin was shown to have a positive effect in an open-label trial, but there are no randomized controlled trials assessing the efficacy of gabapentin.Citation80 A small, open-label trial suggests that zonisamide, a new antiepileptic for the treatment of partial seizures, is effective in treatment of TDK and well-tolerated. However, there is – once again – no large randomized controlled trial to support this effect.Citation81 Valproate, an epileptic with various mechanisms of action, was however found not to be effective in two studies.Citation82,Citation83

A recent Cochrane review (2011) concluded that “evidence of the effects of baclofen, progabide, sodium valproate, or tetrahydroisoxazolopyridine for people with antipsychotic-induced TDK is inconclusive and unconvincing. Any possible benefits are likely to be outweighed by the adverse cognitive effects associated with their use.” Citation84

Antioxidants

Antioxidant drugs have been trialled in TDK after studies of the pathophysiology of the condition showed involvement of free radicals: chronic neuroleptic exposure increases dopamine turnover in the brain with subsequent production of cytotoxic-free radicals. A recent randomized controlled trial of ginkgo biloba versus placebo in Chinese schizophrenic patients demonstrated significant benefit in TDK as measured by the AIMS score.Citation85 Vitamin E, which neutralizes free radicals, has been investigated in multiple studies. Although a large multicenter study showed minimal acute benefit on reducing abnormal motor movements,Citation86Citation88 vitamin E may have more significant benefit at protecting against the deterioration of TDK symptoms over time.Citation89 Melatonin, another antioxidant, was found to be effective in one randomized controlled trial.Citation90 Further, melatonin levels and the melatonin circadian rhythm are significantly low in schizophrenia, so treatment with melatonin may help remedy sleep disorders in schizophrenia, as well as aiding the antipsychotic treatment effect through its anti-inflammatory and antioxidative effect (see Anderson and MaesCitation91 for review). Vitamin B6 was shown to be effective in two crossover studies, both done by the same group.Citation92

Cholinergic versus anticholinergic agents

It has been observed that parkinsonian features of TDK can be improved by dopamine agonists or cholinergic antagonists, suggesting an imbalance between acetylcholine and dopamine as a means of causing dyskinesias. Donepezil is the only drug that has consistently shown some benefit,Citation93,Citation94 despite the numerous studies of acetylcholinesterase inhibitors and cholinomimetic agents. A systematic review of cholinergic agents failed to show a clear-cut benefit for cholinergic agents in the treatment of TDK. However, in some studies, cholinergic agents were shown to be neither helpful nor detrimental. The authors of the systematic review suggested that researchers should not disregard this group of drugs (ie, cholinomimetics and acetylcholinesterase inhibitors), but larger trials of the best-tolerated drugs should be considered, especially in view of the absence of adverse effects with these drugs.Citation95 Anticholinergics agents, however, are not only ineffective but appear to worsen TDK in some patients.Citation96

Dopaminergic agents

Dopaminergic therapy has been studied to a limited extent. Naloxone,Citation97,Citation98 an opioid receptor agonist with dopamine-modulating effects, may have some benefit. Amantadine, which enhances presynaptic dopamine release, has been studied in two randomized controlled trials and has shown a significant reduction in TDK of up to 40%.Citation99,Citation100 BromocriptineCitation101 and selegilineCitation102 are considered to be ineffective. Buspirone, a serotonin receptor agonist that has some dopamine-modulating effects, is well-tolerated but is of variable benefit.Citation103 From the recent research on the effect of different dopamine subtype agonists on TDK,Citation34 it would be hoped that newer, more selective dopaminergic agents may be more effective in the management of TDK.

Calcium channel blockers, beta blockers, etc

Calcium channel blockers have been tried in the treatment of TDK. An analysis of five small randomized and eight nonrandomized studies could not refute nor support use of these drugs for TDK.Citation104 A more recent systematic review of calcium channel blockers in the treatment of neuroleptic-induced TDK concluded that there were no appropriate studies from which to draw conclusions. Their data search uncovered 15 possible studies, but eight were not randomized controlled trials, and the others were small or did not include patients with defined mental illnesses. The authors concluded that use of calcium channel blockers in the treatment of TDK is experimental, and large randomized controlled trials are needed to assess their effectiveness.Citation105 Propranolol, a postsynaptic beta receptor blocker, has been reported to be effective in the management of TDK in several case reports and small open trials; but the evidence is weak, and abrupt cessation results in rebound of symptoms.Citation106 Clonidine, a presynaptic alpha receptor agonist, has been shown to be effective in reducing TDK in a single randomized controlled trial.Citation107

Botulinum toxin

As in other types of dystonia, botulinum toxin type A has been shown to be of benefit in treatment of tardive dystoniaCitation108 and is often injected into the tongue and masticatory muscles.Citation109 In painful TDK, botulinum toxin is also reported to be of benefit.Citation110

Essential fatty acids

As research studies reported high levels of phenylalanine in males with TDK, it was thought that branched chain amino acids may be effective in the treatment of TDK. A single double blind placebo-controlled trial showed a significant beneficial effect in patients with TDK. This effect is thought to be due to branched chain amino acids decreasing the concentrations of aromatic amino acids. Other essential fatty acids have also been studied but were shown to have no effect on TDK.Citation111

Surgery

As medical treatments of TDK are often disappointing, recent studies of surgical interventions have been conducted. A few case reports have suggested some efficacy of lesioning surgery (ie, pallidotomy or thalamotomy). A much greater number of series (including one controlled study) have assessed the effects of deep brain stimulation applied to the globus interna.Citation112Citation115 All of these studies have shown a marked improvement of motor symptoms without any major psychiatric side effects. A recent systematic review of deep brain stimulation (DBS) in TDKCitation114 concludes that DBS greatly improves motor scores in patients with TDK (and tardive dystonia) and that psychiatric effects are limited, although this aspect was not always assessed. DBS would be considered appropriate treatment for patients who do not respond to or cannot tolerate pharmacological interventions. It is thought that the lack of psychiatric side effects may be that the globus pallidus interna (GPi) has been stimulated in TDK; whereas, in Parkinson’s, the subthalamic nucleus is the target. Pal-lidal stimulation is also extremely effective for severe disabling tardive dystonia unresponsive to other forms of therapy.Citation112Citation115

Animal studies

Ongoing research into the pathophysiology and treatment of TDK has involved predominantly a mouse model of TDK in which vacuous chewing movements are produced by exposure to neuroleptic drugs. Studies of this mouse model suggest that nicotine significantly reduces vacuous chewing movements induced by haloperidol.Citation116 The exact mechanism of action of the nicotine is uncertain. However, this data needs to be treated with caution as a large review of Chinese schizophrenic patients reported that smoking was not protective against the development of TDK, nor did it increase the risk of developing TDK.Citation117 In other mouse studies, fluphenazine induced vacuous chewing movements in 70% of animals, and the animals showed reduced locomotor and exploratory movements. Combining resveratrol (a polyphenol found in grapes and red wine) with fluphenazine reduced the prevalence, but not intensity, of the vacuous chewing movements down to 30% and also lessened the locomotor and exploratory effects of fluphenazine. These findings suggest that resveratrol may be neuroprotective in the presence of fluphenazine treatment.Citation118

Deep brain stimulation has also recently been trialled for treatment resistant drug-induced TDK. In the rat model of vacuous chewing movements, deep brain stimulation has been performed at the entopeduncular nucleus (EPN) and subthalamic nucleus (STN). Stimulation at both sites is effective, and only unilateral stimulation is required. The stimulation parameters, however, vary with higher rates of stimulation more effective at the STN; whereas, at the EPN, low and high rates of stimulation were equally effective.Citation119

Considering the complex motor pathways now described in the possible pathophysiology of TDKCitation10,Citation11 – as well as the evidence of altered synaptic plasticity in brain stimulation studiesCitation46,Citation114 – perhaps it would be more useful for further animal studies to be done on nonhuman primates whose motor control systems mirror more closely those of the human.

TDK in adolescents and children

The risk of TDK is lower with second-generation antipsychotics than first-generation drugs in children, but most of the use of these drugs has been off-label.Citation120 In randomized controlled trials, Haas et alCitation121,Citation122 showed a higher risk of TDK in risperidone-treated children, and the risk of TDK was dose-related. Correll and KaneCitation123 evaluated the risk for TDK after 1 year of atypical antipsychotic exposure in a meta-analysis of ten long-term studies including youths with minimal previous exposure to typical antipsychotics (783 with risperidone, 27 with quetiapine, and 19 with olanzapine). Only three new cases of TDK occurred during up to 3 years of treatment; in two of them, the symptomatology resolved after drug discontinuation. Although it can occur in the first phases of the treatment, the risk of TDK increases with longer treatments. A pilot studyCitation124 and the Treatment of Early Onset Schizophrenia Spectrum Disorders (TEOSS)Citation125 have shown a relatively equal risk of TDK with olanzapine, risperidone, and aripiprazole. Findling et alCitation126 showed an increased risk of TDK when treating schizophrenic children with aripiprazole compared to placebo. There are only open-label studies of quetiapine,Citation127,Citation128 and these do not report an increased risk of TDK in children. However, all these studies have been fairly small. Larger randomized controlled trials in schizophrenic patients are needed to document the true risk of TDK with these new drugs, to determine the drug with the lowest risk of TDK, and to determine the response of TDK to treatment. For now, the mainstay of treatment suggested is levodopa or amantadine, and switching patients to clozapine may be helpful.Citation129

Conclusion

The treatment of TDK remains unsatisfactory, and the benefits of various agents need to be weighed against their side effects. However, given the advances in the pathophysiology of TDK, further therapeutic studies based on these findings may be helpful in optimizing the management of TDK. Clinicians need also to remember the added risk of TDK when combining the use of neuroleptics with metoclopramide in the same patient.Citation2 Further, children appear to be at greater risk of extrapyramidal side effects,Citation130,Citation131 and the use of second-generation antipsychotics is increasing in children.Citation132 Unfortunately, the most effective drugs for the treatment of TDK may have side effects that preclude their use long-term; whereas, less-effective drugs may have a more tolerable side effect profile (). Management is generally complex and needs to be individualized. As TDK is an iatrogenic disorder, it is best to limit the occurrence by prudent use of neuroleptic agents. Measures to limit the risk of TDK include: (1) critical, objective indications for neuroleptic drug use; (2) long-term use only for compelling or research-supported indications, primarily chronic psychotic illness that worsens when neuroleptic treatment is slowly discontinued; (3) using alternative treatments when neuroleptic treatment is elective, or early dyskinesia is identified; (4) using low but effective doses of single drugs, especially in the elderly; and (5) regular and specific examination for early detection of TDK. Once TDK becomes evident, it is prudent to use the lowest dose of drug necessary and avoiding other dopamine-blocking drugs. For severe dyskinesia, tetrabenazine is the most effective on the trial data available but does have side effects.

Table 3 Hierarchy of strategies in the management of tardive dyskinesia

Benzodiazepines may be useful in less-prominent movements, and botulinum toxin is specifically useful in the presence of pain or dystonia. Deep brain stimulation remains an option in those patients with severe disabling TDK in whom several pharmacological agents, given an adequate duration of treatment, have not been effective.

Disclosure

The author reports no conflict of interest in this work.

References

  • SchoneckerMParoxysmal dyskinesia as the effect of megaphenNervenarzt19572812550553 German13517450
  • KenneyCHunterCDavidsonAJankovicJMetoclopramide, an increasingly recognized cause of tardive dyskinesiaJ Clin Pharmacol200848337938418223146
  • RaoASCamilleriMReview article: metoclopramide and tardive dyskinesiaAliment Pharmacol Ther2010311111919886950
  • WoernerMGKaneJMLiebermanJThe prevalence of tardive dyskinesiaJ Clin Psychopharmacol199111134421674949
  • KaneJMSmithJMTardive dyskinesia: prevalence and risk factors, 1959 to 1979Arch Gen Psychiatry19823944734816121548
  • EberhardJLindströmELevanderSTardive dyskinesia and antipsychotics: a 5-year longitudinal study of frequency, correlates and courseInt Clin Psychopharmacol2006211354216317315
  • JesteDVCaligiuriMPPaulsenJSRisk of tardive dyskinesia in older patients. A prospective longitudinal study of 266 outpatientsArch Gen Psychiatry19955297567657654127
  • HeimerLA new anatomical framework for neuropsychiatric disorders and drug abuseAm J Psychiatry2003160101726173914514480
  • CummingsJLFrontal-subcortical circuits and human behaviorArch Neurol19935088738808352676
  • MinkJWThe basal ganglia: focused selection and inhibition of competing motor programsProg Neurobiol19965043814259004351
  • MarchandWRDildaVNew models of frontal-subcortical skeletomotor circuit pathology in tardive dyskinesiaNeuroscientist200612318619816684965
  • MargoleseHCChouinardGKolivakisTTBeauclairLMillerRAnnableLTardive dyskinesia in the era of typical and atypical antipsychotics. Part 2: Incidence and management strategies in patients with schizophreniaCan J Psychiatry2005501170371416363464
  • ShedlackKJSoldat-CoutureCSwansonCLJrRapidly progressive tardive dyskinesia in AIDSBiol Psychiatry19943521471488167213
  • OhmoriOShinkaiTHoriHMatsumotoCNakamuraJA perspective on molecular genetic studies of tardive dyskinesia: one clue for individualized antipsychotic drug therapyProg Neuropsychopharmacol Biol Psychiatry200327458158612787842
  • Van HartenPNTenbackDETardive dyskinesia: clinical presentation and treatmentInt Rev Neurobiol20119818721021907088
  • KhotVWyattRJNot all that moves is tardive dyskinesiaAm J Psychiatry199114856616661673323
  • ArthurHDahlMLSiwersBSjöqvistFPolymorphic drug metabolism in schizophrenic patients with tardive dyskinesiaJ Clin Psychopharmacol19951532112167635999
  • AndreassenOAMacEwanTGulbrandsenAKMcCreadieRGSteenVMNon-functional CYP2D6 alleles and risk for neuroleptic-induced movement disorders in schizophrenic patientsPsychopharmacology (Berl)199713121741799201806
  • ArmstrongMDalyAKBlennerhassettRFerrierNIdleJRAntipsychotic drug-induced movement disorders in schizophrenics in relation to CYP2D6 genotypeBr J Psychiatry199717023269068770
  • SachseCBrockmöllerJBauerSRootsICytochrome P450 2D6 variants in a Caucasian population: allele frequencies and phenotypic consequencesAm J Hum Genet19976022842959012401
  • OhmoriOKojimoHShinkaiTTeraoTSuzukiTAbeKGenetic association analysis between CYP2D6*2 allele and tardive dyskinesia in schizophrenic patientsPsychiatry Res1999872–323924410579557
  • LamLCGarcia-BarceloMMUngvariGSCytochrome P450 2D6 genotyping and association with tardive dyskinesia in Chinese schizophrenic patientsPharmopsychiatry2001346238241
  • BasileVSOzdemirVMasellisMA functional polymorphism of the cytochrome P450 1A2 (CYP1A2) gene: association with tardive dyskinesia in schizophreniaMol Psychiatry20005441041710889552
  • SchulzeTGSchumacherJMüllerDLack of association between a functional polymorphism of the cytochrome P450 1A2 (CYP1A2) gene and tardive dyskinesia in schizophreniaAm J Med Genet2001105649850111496364
  • PatsopoulosNANtzaniEEZintzarasEIoannidisJPCYP2D6 polymorphisms and the risk of tardive dyskinesia in schizophrenia: a meta-analysisPharmacogenet Genomics200515315115815861039
  • MillerRChouinardGLoss of striatal cholinergic neurons as a basis for tardive and L-dopa-induced dyskinesias, neuroleptic-induced supersensitivity psychosis and refractory schizophreniaBiol Psychiatry199334107137387904833
  • SachdevPSThe current status of tardive dyskinesiaAust N Z J Psychiatry200034335536910881961
  • RosengartenHSchweitzerJWFriedhoffAJInduction of oral dyskinesias in naive rats by D1stimulationLife Sci19833325247924826227782
  • MarsdenCDJennerPThe pathophysiology of extrapyramidal side-effects of neuroleptic drugsPsychol Med198010155726104342
  • CalabresiPDe MurtasMMercuriNBBernardiGChronic neuroleptic treatment: D2 dopamine receptor supersensitivity and striatal glutamatergic transmissionAnn Neurol19923143663731350190
  • TurronePRemingtonGKapurSNobregaJNThe relationship between dopamine D2 receptor occupancy and the vacuous chewing movement syndrome in ratsPsychopharmacology (Berl)2003165216617112417967
  • CrowTJCrossAJJohnstoneECOwenFOwensDGWaddingtonJLAbnormal involuntary movements in schizophrenia: are they related to the disease process or its treatment? Are they associated with changes in dopamine receptors?J Clin Psychopharmacol1982253363407130435
  • SibleyDRMonsmaFJJrShenYMolecular biology of dopaminergic receptorsInt Rev Neurobiol1993353914158463063
  • MalikPAndersenMBPeacockLThe effects of dopamine D3 agonists and antagonists in a nonhuman primate model of tardive dyskinesiaPharmacol Biochem Behav200478480581015301939
  • UtsunomiyaKShinkaiTSakataSGenetic association between the dopamine D3 receptor gene polymorphism (Ser9Gly) and tardive dyskinesia in patients with schizophrenia: a reevaluation in East Asian populationsNeurosci Lett20125071525622172931
  • BasileVSOzdemirVMasellisMLack of association between serotonin-2A receptor gene (HTR2A) polymorphisms and tardive dyskinesia in schizophreniaMol Psychiatry20016223023411317228
  • SegmanRHHeresco-LevyUFinkelBAssociation between the serotonin 2A receptor gene and tardive dyskinesia in chronic schizophreniaMol Psychiatry20016222522911317227
  • OhmoriOShinkaiTHoriHNakamuraJGenetic association analysis of 5-HT(6) receptor gene polymorphism (267C/T) with tardive dyskinesiaPsychiatry Res200211029710212057822
  • SyuAIshiguroHInadaTAssociation of the HSPG2 gene with neuroleptic-induced tardive dyskinesiaNeuropsychopharmacology20103551155116420072119
  • GreenbaumLAlkelaiAZozulinskyPKohnYLererBSupport for association of HSPG2 with tardive dyskinesia in Caucasian populationsPharmacogenomics J201212651352021808285
  • MüllerDJChowdhuryNIZaiCCThe pharmacogenetics of antipsychotic-induced adverse eventsCurr Opin Psychiatry201326214415023370274
  • NitscheMAMüller-DahlhausFPaulusWZiemannUThe pharmacology of neuroplasticity induced by non-invasive brain stimulation: building models for the clinical use of CNS active drugsJ Physiol2012590Pt 194641466222869014
  • TeoJTTerranovaCSwayneOGreenwoodRJRothwellJCDiffering effects of intracortical circuits on plasticityExp Brain Res2009193455556319048237
  • KorchounovAZiemannUNeuromodulatory neurotransmitters influence LTP-like plasticity in human cortex: a pharmaco-TMS studyNeuropsychopharmacology20113691894190221544070
  • HasanAMisewitschKNitscheMAImpaired motor cortex responses in non-psychotic first-degree relatives of schizophrenia patients: a cathodal tDCS pilot studyBrain Stimul Epub3222013
  • TeoJTEdwardsMJBhatiaKTardive dyskinesia is caused by maladaptive synaptic plasticity: a hypothesisMov Disord201227101205121522865512
  • WoodsSWMorgensternHSaksaJRIncidence of tardive dyskinesia with atypical versus conventional antipsychotic medications: a prospective cohort studyJ Clin Psychiatry201071446347420156410
  • KahnRSFleischhackerWWBoterHEUFEST study groupEffectiveness of antipsychotic drugs in first-episode schizophrenia and schizophreniform disorder: an open randomised clinical trialLancet200837196181085109718374841
  • LeuchtSKisslingWDavisJMSecond-generation antipsychotics for schizophrenia: can we resolve the conflict?Psychol Med200939101591160219335931
  • EganMFApudJWyattRJTreatment of tardive dyskinesiaSchizophr Bull19972345836099365997
  • SakaiKGaoXMHashimotoTTammingaCATraditional and new antipsychotic drugs differentially alter neurotransmission markers in basal ganglia-thalamocortical neural pathwaysSynapse200139215216011180502
  • VeselyCKüfferleBBrückeTKasperSRemission of severe tardive dyskinesia in a schizophrenic patient treated with the atypical antipsychotic substance quetiapineInt Clin Psychopharmacol20001515760
  • SoutulloCAKeckPEJrMcElroySLOlanzapine in the treatment of tardive dyskinesia: a report of two casesJ Clin Psychopharmacol19991911001019934952
  • ChouinardGEffects of risperidone in tardive dyskinesia: an analysis of the Canadian multicenter risperidone studyJ Clin Psychopharmacol1995151 Suppl 136S44S7537286
  • TarsyDLunguCBaldessariniRJEpidemiology of tardive dyskinesia before and during the era of modern antipsychotic drugsHandb Clin Neurol201110060161621496610
  • FactorSAFriedmanJHThe emerging role of clozapine in the treatment of movement disordersMov Disord19971244834969251065
  • SimpsonGMLeeJHShrivastavaRKClozapine in tardive dyskinesiaPsychopharmacol (Berl)19785617580
  • LiebermanJASaltzBLJohnsCAPollackSBorensteinMKaneJThe effects of clozapine on tardive dyskinesiaBr J Psychiatry19911585035101675900
  • SmallJGMilsteinVMarhenkeJDHallDDKellamsJJTreatment outcome with clozapine in tardive dyskinesia, neuroleptic sensitivity, and treatment-resistant psychosisJ Clin Psychiatry19874872632672885310
  • NatesanSRecklessGENobregaJNFletcherPJKapurSDissociation between in vivo occupancy and functional antagonism of dopamine D2 receptors: comparing aripiprazole to other antipsychotics in animal modelsNeuropsychopharmacology20063191854186316319908
  • ShapiroDARenockSArringtonEAripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacologyNeuropsychopharmacology20032881400141112784105
  • TammingaCACarlssonAPartial dopamine agonists and dopaminergic stabilisers, in the treatment of psychosisCurr Drug Targets CNS Neurol Disord20021214114712769623
  • EvcimenYAEvcimenHHollandJAripiprazole-induced tardive dyskinesia: the role of tamoxifenAm J Psychiatry200716491436143717728431
  • PeñaMSYalthoTCJankovicJTardive dyskinesia and other movement disorders secondary to aripiprazoleMov Disord201126114715220818603
  • SchwartzTRazaSAripiprazole (abilify) and tardive dyskinesiaP T2008331323419749988
  • SimpsonGMThe treatment of tardive dyskinesia and tardive dystoniaJ Clin Psychiatry200061Suppl 4394410739330
  • JankovicJClarence-SmithKTetrabenazine for the treatment of chorea and other hyperkinetic movement disordersExpert Rev Neurother201111111509152322014129
  • KimiagarIDobronevskyEProkhorovTMiniovitzARabeyJMRapid improvement of tardive dyskinesia with tetrabenazine, clonazepam and clozapine combined: a naturalistic long-term follow-up studyJ Neurol2012259466066422068977
  • ChenJJOndoWGDashtipourKSwopeDMTetrabenazine for the treatment of hyperkinetic movement disorders: a review of the literatureClin Ther20123471487150422749259
  • OndoWGHannaPAJankovicJTetrabenazine treatment for tardive dyskinesia: assessment by randomized videotape protocolAm J Psychiatry199915681279128110450276
  • SoaresKVMcGrathJJDeeksJJGamma-aminobutyric acid agonists for neuroleptic-induced tardive dyskinesia [review]Cochrane Database Syst Rev20012CD00020311405955
  • ThakerGKNguyenJAStraussMEJacobsonRKaupBATammingaCAClonazepam treatment of tardive dyskinesia: a practical GABAmimetic strategyAm J Psychiatry199014744454511969244
  • SinghMMBeckerREPitmanRKNasrallahHALalHSustained improvement in tardive dyskinesia with diazepam: indirect evidence for corticolimbic involvementBrain Res Bull19831121791856313141
  • ThakerGKTammingaCAAlphsLDLaffermanJFerraroTNHareTABrain gamma-aminobutyric acid abnormality in tardive dyskinesia. Reduction in cerebrospinal fluid GABA levels and therapeutic response to GABA agonist treatmentArch Gen Psychiatry19874465225293034188
  • BobruffAGardosGTarsyDRapkinRMColeJOMoorePClonazepam and phenobarbital in tardive dyskinesiaAm J Psychiatry19811382189193
  • StewartRMRollinsJBeckhamBRoffmanMBaclofen in tardive dyskinesia patients maintained on neurolepticsClin Neuropharmacol1982543653736130838
  • GerlachJRyeTKristjansenPEffect of baclofen on tardive dyskinesiaPsychopharmacol (Berl)1978562145151
  • LibovIMiodownikCBersudskyYDwolatzkyTLernerVEfficacy of piracetam in the treatment of tardive dyskinesia in schizophrenic patients: a randomized, double-blind, placebo-controlled crossover studyJ Clin Psychiatry20076871031103717685739
  • WoodsSWSaksaJRBakerCBCohenSJTekCEffects of levetiracetam on tardive dyskinesia: a randomized, double-blind, placebo-controlled studyJ Clin Psychiatry200869454655418312060
  • HardoyMCCartaMGCarpinielloBGabapentin in antipsychotic-induced tardive dyskinesia: results of 1-year follow-upJ Affect Disord200375212513012798252
  • IwataYIrieSUchidaHEffects of zonisamide on tardive dyskinesia: a preliminary open-label trialJ Neurol Sci20123151–213714022285275
  • FiskGGYorkSMThe effect of sodium valproate on tardive dyskinesia – revisitedBr J Psychiatry19871505425463117156
  • NasrallahHADunnerFJMcCalley-WhittersMA placebo-controlled trial of valproate in tardive dyskinesiaBiol Psychiatry19852022052083918588
  • AlabedSLatifehYMohammadHARifaiAGamma-aminobutyric acid agonists for neuroleptic-induced tardive dyskinesia [review]Cochrane Database Syst Rev20114CD00020321491376
  • ZhangWFTanYLZhangXYChanRCWuHRZhouDFExtract of Ginkgo biloba treatment for tardive dyskinesia in schizophrenia: a randomized, double-blind, placebo-controlled trialJ Clin Psychiatry201172561562120868638
  • EganMFHydeTMAlbersGWTreatment of tardive dyskinesia with vitamin EAm J Psychiatry199214967737771350428
  • AdlerLAPeselowERotrosenJVitamin E treatment of tardive dyskinesiaAm J Psychiatry19931509140514078102511
  • AdlerLARotrosenJEdsonRVitamin E treatment for tardive dysklinesia. Veterans Affairs Cooperative Study #394 Study GroupArch Gen Psychiatry199956983684112892048
  • Soares-WeiserKMaayanNMcGrathJVitamin E for neuroleptic-induced tardive dyskinesia [review]Cochrane Database Syst Rev20112CD000209
  • ShamirEBarakYShalmanIMelatonin treatment for tardive dyskinesia: a double-blind, placebo-controlled, crossover studyArch Gen Psychiatr200158111049105211695951
  • AndersonGMaesMMelatonin: an overlooked factor in schizophrenia and in the inhibition of anti-psychotic side effectsMetab Brain Dis201227211311922527998
  • LernerVMiodownikCKaptsanAVitamin B6 treatment for tardive dyskinesia: a randomized, double-blind, placebo-controlled, crossover studyJ Clin Psychiatry200768111648165418052557
  • BergmanJDwolatzkyTBrettholzILernerVBeneficial effect of donepezil in the treatment of elderly people with tardive movement disordersJ Clin Psychiatry200566110711015669896
  • CaroffSNCampbellECHaveyJCSullivanKAKatzIRMannSCTreatment of tardive dyskinesia with donepezilJ Clin Psychiatry200162212812911247101
  • TammenmaaIASailasEMcGrathJJSoares-WeiserKWahlbeckKSystematic review of cholinergic drugs for neuroleptic-induced tardive dyskinesia: a meta-analysis of randomized controlled trialsProg Neuropsychopharmacol Biol Psychiatry20042871099110715610922
  • DesmaraisJEBeauclairLMargoleseHCAnticholinergics in the era of atypical antipsychotics: short-term or long-term treatment?J Psychopharmacol20122691167117422651987
  • BlumINisipeanuPFRobertsENaloxone in tardive dyskinesiaPsychopharmacol (Berl)1987934538
  • LindenmayerJPGardnerEGoldbergEHigh-dose naloxone in tardive dyskinesiaPsychiatry Res198826119283070611
  • AngusSSugarsJBoltezarRKoskewichSSchneiderNMA controlled trial of amantadine hydrochloride and neuroleptics in the treatment of tardive dyskinesiaJ Clin Psychopharmacol1997172889110950469
  • PappaSTsouliSApostolouGMavreasVKonitsiotisSEffects of amantadine on tardive dyskinesia: a randomized, double-blind, placebo-controlled studyClin Neuropharmacol2010336271275
  • LiebermanJAAlvirJMukherjeeSKaneJMTreatment of tardive dyskinesia with bromocriptine. A test of the receptor modification strategyArch Gen Psychiatry198946109089132572205
  • GoffDCRenshawPFSarid-SegalODreyfussDAAmicoETCirauloDAA placebo-controlled trial of selegiline (L-deprenyl) in the treatment of tardive dyskinesiaBiol Psychiatry199333107007068102552
  • MossLENeppeVMDrevetsWCBuspirone in the treatment of tardive dyskinesiaJ Clin Psychopharmacol19931332042098102622
  • Soares-WeiserKKRathboneJCalcium channel blockers for neuroleptic-induced tardive dyskinesia [review]Cochrane Database Syst Rev20041CD00020614973950
  • EssaliADeirawanHSoares-WeiserKAdamsCECalcium channel blockers for neuroleptic-induced tardive dyskinesia [review]Cochrane Database Syst Rev201111CD00020622071797
  • AiaPGRevueltaGJCloudLJFactorSATardive dyskinesiaCurr Treat Options Neurol201113323124121365202
  • BrowneJSilverHMartinRHartRMergenerMWilliamsPThe use of clonidine in the treatment of neuroleptic-induced tardive dyskinesiaJ Clin Psychopharmacol19866288922871058
  • JankovicJBrinMFTherapeutic uses of botulinum toxinN Engl J Med199132417118611942011163
  • RapaportASadehMSteinDBotulinum toxin for the treatment of oro-facial-lingual-masticatory tardive dyskinesiaMov Disord200015235235510752596
  • TschoppLSalazarZMicheliFBotulinum toxin in painful tardive dyskinesiaClin Neuropharmacol200932316516619483486
  • EmsleyRNiehausDJKoenLThe effects of eicosapentaenoic acid in tardive dyskinesia: a randomized, placebo-controlled trialSchizophr Res200684111212016632329
  • EltahawyHAFeinsteinAKhanFSaint-CyrJLangAELozanoAMBiltaeral globus pallidus internus deep brain stimulation in tardive dyskinesia: a case reportMov Disord200419896997215300668
  • DamierPThoboisSWitjasTFrench Stimulation for Tardive Dyskinesia (STARDYS) Study GroupBilateral deep brain stimulation of the globus pallidus to treat tardive dyskinesiaArch Gen Psychiatry200764217017617283284
  • MentzelCLTenbackDETijssenMAVisser-VandewalleVEvan HartenPNEfficacy and safety of deep brain stimulation in patients with medication-induced tardive dyskinesia and/or dystonia: a systemic reviewJ Clin Psychiatry201273111434143823218160
  • KefalopoulouZPaschaliAMarkakiEVassilakosPEllulJConstantoyannisCA double-blind study on a patient with tardive dyskinesia treated with pallidal deep brain stimulationActa Neurol Scand2009119426927318976318
  • BordiaTMcIntoshJMQuikMNicotine reduces antipsychotic-induced orofacial dyskinesia in ratsJ Pharmacol Exp Ther2012340361261922144565
  • ZhangXYYuYQSunSSmoking and tardive dyskinesia in male patients with chronic schizophreniaProg Neuropsychopharmacol Biol Psychiatry20113571765176921723906
  • BusanelloAPerozaLRWagnerCResveratrol reduces vacuous chewing movements induced by acute treatment with fluphenazinePharmacol Biochem Behav2012101230731022266770
  • CreedMHamaniCNobregaJNDeep brain stimulation of the subthalamic or entopeduncular nucleus attenuates vacuous chewing movements in a rodent model of tardive dyskinesiaEur Neuropsychopharmacol201121539340020624675
  • PringsheimTDojaABelangerSPattenSCanadian Alliance for Monitoring Effectiveness and Safety of Antipsychotics in Children (CAMESA) guideline groupTreatment recommendations for extrapyramidal side effects associated with second-generation antipsychotic use in children and youthPaediatr Child Health201116959059823115503
  • HaasMEerdekensMKushnerLEfficacy, safety and tolerability of two dosing regimens in adolescent schizophrenia: double-blind studyBr J Psychiatry2009194215816419182179
  • HaasMUnisASArmenterosJCopenhaverMDQuirozJAKushnerSFA 6-week, randomized, double-blind placebo-controlled study of the efficacy and safety of risperidone in adolescent with schizophreniaJ Child Adolesc Psychopharmacol200919661162120035579
  • CorrellCUKaneJMOne-year incidence rates of tardive dyskinesia in children and adolescents treated with second-generation antipsychotics: a systematic reviewJ Child Adolesc Psychopharmacol200717564765617979584
  • SikichLHamerRMBashfordRASheitmanBBLiebermanJAA pilot study of risperidone, olanzapine, and haloperidol in psychotic youth: a double-blind, randomized, 8-week trialNeuropsychopharmacology200429113314514583740
  • SikichLFrazierJAMcClellanJDouble-blind comparison of first- and second-generation antipsychotics in early-onset schizophrenia and schizo-affective disorder: findings from the treatment of early-onset schizophrenia spectrum disorder (TEOSS) studyAm J Psychiatry2008165111420143118794207
  • FindlingRLRobbANyilasMA multiple-center, randomized, double-blind, placebo-controlled study of oral aripiprazole for treatment of adolescents with schizophreniaAm J Psychiatry2008165111432144118765484
  • McConvilleBJArvanitisLAThyrumPTPharmacokinetics, tolerability, and clinical effectiveness of quetiapine fumarate: an open-label trial in adolescents with psychotic disordersJ Clin Psychiatry200061425226010830145
  • SchimmelmannBGMehler-WexCLambertMA prospective 12-week study of quetiapine in adolescents with schizophrenia spectrum disordersJ Child Adolesc Psychopharmacol200717676877818315449
  • MasiGLiboniFManagement of schizophrenia in children and adolescents: focus on pharmacotherapyDrugs201171217920821275445
  • CorrellCUSafety and tolerability of antipsychotic treatment in young patients with schizophreniaJ Clin Psychiatry2011728e2621899814
  • RasimasJJLiebeltELAdverse effects and toxicity of the atypical antipsychotics: what is important for the pediatric emergency medicine practitionerClin Pediatr Emerg Med201213430031023471213
  • PattenSBWaheedWBreseeLA review of pharmacoepidemiologic studies of antipsychotic use in children and adolescentsCan J Psychiatry2012571271772123228229