288
Views
15
CrossRef citations to date
0
Altmetric
Review

Overcoming barriers to effective management of tardive dyskinesia

Pages 785-794 | Published online: 04 Apr 2019

Abstract

Tardive dyskinesia (TD) is a heterogeneous syndrome of involuntary hyperkinetic movements that is often persistent and occurs belatedly during treatment with antipsychotics. Recent approval of two dopamine-depleting analogs of tetrabenazine based on randomized controlled trials offers an evidence-based therapeutic approach to TD for the first time. These agents are optimally used within the context of a comprehensive approach to patient management that includes a practical screening and monitoring program, sensitive and specific criteria for the diagnosis of TD, awareness of the severity and impact of the disorder, informed discussions with patients and caregivers, and a rational basis for prescribing decisions about continued antipsychotic and adjunctive agents. Areas of limited or inconclusive data, bias and misunderstandings about key aspects, and neglect of training about TD in recent years contribute to barriers in providing effective care and promoting patient safety.

Introduction

The introduction of specific antipsychotic drugs over a half century ago ushered in an era of renewed therapeutic optimism and revitalized interest in understanding the neurobiology of severe mental illness. However, a troubling drawback of this class of agents that are dependent on dopamine-receptor blocking properties was their impact on basal ganglia functions underlying drug-induced movement disorders.Citation1,Citation2 Although most of these adverse effects were acute in onset and reversible after medication cessation, the movements of tardive dyskinesia (TD) that were persistent and associated with prolonged treatment were most concerning.

Until the recent approval of two new tetrabenazine analogs, there was no evidence-based treatments for TD. Even so, there are several areas of misunderstanding, bias, or gaps in knowledge that may limit the effective and safe application of these new drugs. Now that approved treatments are available, it becomes even more important to be aware of the clinical features of TD and when and how these new treatments can be applied to enhance successful outcomes and quality of life.

Frequency and risk of TD

The relative significance of TD as a clinical problem and the need for treatment have been questioned since the initial recognition of the disorder. Following early reports, several authorities questioned whether TD was related to treatment, how common it was and whether it was clinically significant.Citation3Citation8 Early observers assumed that TD was rare and found primarily among elderly women with chronic conditions and brain damage, but this clearly reflected a skewed, selection bias based on studies of long-term residents in institutionalized settings.Citation4,Citation9 Although this bias that TD was uncommon and restricted to chronic mental illness was refuted by many reports among younger people who received dopamine-receptor blocking drugs for pain, gastrointestinal symptoms, or anxiety,Citation3,Citation10Citation12 it may once again become a concern as antipsychotics are widely marketed to an ever-expanding and more functional population of nonpsychotic patients with mood disorders and other indications. It is important to understand that anyone exposed to dopamine-receptor blocking drugs, not just those with chronic mental illness, may be at risk for TD.

In subsequent years, the frequency of TD has been convincingly demonstrated and repeatedly shown.Citation13Citation15 Depending on the risk of the sample population studied and methodology of case detection, studies have shown a cumulative incidence of TD of about 4%–5% annually, with a prevalence rate of 20%–30%.Citation16 Although younger patients are at risk for TD and may be particularly susceptible to more generalized and dystonic movements, older age has consistently been shown to be a major risk factor for TD. The annual incidence of TD in patients over the age of 45 years has been reported as 15%–30% after 1 year of treatment with a prevalence rate of up to 50%–60%.Citation17 Previous studies on TD risk have less consistently suggested an association with female gender, race, higher ratings of negative symptoms and thought disorder, greater cognitive impairments, acute drug-induced movement disorders, substance abuse, and diabetes.Citation18,Citation19 Since both older adults and patients with schizophrenia have been found historically to have a predisposition for spontaneous dyskinesias,Citation3 it is not surprising that they may be at greatest risk for TD.Citation20 However, patients with mood disorders are also at risk and have been considered to be at high risk, although data supporting elevated risk for these patients from early studies may be confounded by the older age of depressed patients, gender bias, and the intermittent use of antipsychotics during sporadic episodes, all known risk factors for TD in their own right.Citation21Citation23 The conclusion for practical purposes must be that regardless of diagnosis, TD is not rare and that anyone exposed to treatment with antipsychotics is at risk.

Differences in liability for TD between older first-generation antipsychotics (FGAs) and newer second-generation antipsychotics (SGAs) have been studied extensively. Comparing the incidence of TD with haloperidol, industry-sponsored trials of SGAs found a 6- to 12-fold reduction in risk for TD with newer agents.Citation24Citation26 Subsequent trials using comparators with less potent dopamine-receptor binding affinity than haloperidol indicated that the advantages of SGAs in reducing risk of TD were diminished.Citation27Citation30 But more recent studies confirmed that the relative risk (RR) of TD with SGAs is significantly less on average than that with FGAs (RR =0.47–0.68 of SGAs vs FGAs).Citation14,Citation31 The risk of TD associated with clozapine is probably least of all.Citation32 The SGAs retain some risk, and since they are more broadly marketed, the absolute number of cases of TD is likely to increase. Thus, while there is some differential risk of TD among antipsychotics depending on dopamine-receptor binding affinity, no currently available antipsychotic is free of liability for TD. Longer duration of antipsychotic treatment and greater cumulative drug doses are considered additional risk factors for the development and persistence of TD.

Seriousness and impact of TD

Dismissal or neglect of TD and the need for treatment also derive from the impression in early studies that patients seemed unaware of or denied concern about TD.Citation3,Citation33,Citation34 The resulting conventional wisdom that TD is unimportant may be an artifact of patient selection bias or the lack of rigorous investigations. Early reports that patients were indifferent to TD were based on older patients with chronic illnesses confined to institutions who were limited by psychosis and negative symptoms and impoverished by cognitive impairments, lack of insight, denial, and few opportunities for social engagement. While up to two thirds of these patients seemed unaware of TD movements, others admitted to being embarrassed by them.Citation35Citation37 By contrast, a recent survey of outpatients with possible TD revealed that 70%–80% were aware of their movements and 50%–60% felt self-conscious or embarrassed by them.Citation20

In assessing the seriousness of TD and need for treatment, clinicians should consider the objective severity of the movements and the functional impact on affected patients.Citation3 Mild cases of TD may be suppressed, acceptable to the patient, or not readily noticeable apart from everyday habits, tics, and mannerisms that are commonly ignored even by normal individuals. However, moderate to severe movements may be intolerable, painful, physically disabling and eventuate in depression and even suicide.Citation38,Citation39 As antipsychotics are prescribed for more functional patients, however, TD is likely to be experienced as intolerable even in mild cases as a result of social embarrassment. Early impressions in chronic schizophrenia, that patients are unconcerned about TD, may be outdated given the evidence that it has physical, emotional, and social consequences that bear directly on clinical outcomes, functional recovery, and survival.Citation40,Citation41

A recent review summarized correlations between TD and impaired cognition, poor response to treatment, risk of relapse, longer hospital stays, lower quality of life, and increased mortality.Citation42 However, these correlations are confounded by the effect of the underlying psychiatric illness, the severity of which contributes to the risk of both TD and poor outcomes. In other words, the association between TD, psychopathology, and poor response could be an artifact of treatment reflecting the fact that patients with severe psychoses and poor prognoses receive higher doses of antipsychotics for longer periods with poor adherence, resulting secondarily in a greater incidence of TD.Citation3,Citation42 There remains a pressing need to assess the psychological, physical, social, and economic burden of TD.Citation43,Citation44 This requires asking concrete, descriptive questions to patients and caregivers regarding specific circumstances in social situations where odd postures and movements invite ridicule and ostracism, and induce shame and withdrawal.Citation3 Complacency is unacceptable regarding even mild cases of TD which can seriously impede social rehabilitation and recovery.

Onset of TD and screening methods

Given that TD may become persistent and irreversible, prevention by conservative use of antipsychotics and early detection is critical. Early diagnosis leading to antipsychotic drug cessation may increase the chances that movements will be transient and reversible.Citation33,Citation45,Citation46 Therefore, the frequency and methods for screening should reflect the biology and time of onset of TD. Although evidence suggests that most cases of TD develop over months to years, with the risk correlating with duration and cumulative dose of antipsychotic treatment,Citation47,Citation48 there are reports of persistent movements arising within days to weeks after starting antipsychotics particularly among susceptible patients such as the elderly.Citation49,Citation50 Delineation of the time of onset of TD is confounded by the fact that TD could be masked or suppressed by antipsychotics and may first become apparent or worsen only after drug withdrawal in 5%–67% of patients.Citation3,Citation51,Citation52 Although not accepted as a completely valid model for TD,Citation53 dopamine-receptor supersensitivity in animal models usually develops quickly after 1 week of treatment and has occurred even after a single injection of an antipsychotic.Citation54,Citation55

By contrast, some criteria for the diagnosis of TD suggest a minimum duration of antipsychotic treatment of at least 3 months.Citation56,Citation57 Even less consistent with biological findings, guidelines for screening of TD suggest clinical assessment every 6 or 12 months for patients receiving FGAs or SGAs, respectively, reduced to 3 or 6 months for patients at heightened risk.Citation58,Citation59 But assessments conducted every 3 months or more are likely to miss a percentage of early cases of TD, preventing timely intervention and reducing chances of reversal. Paradoxically, the same guidelines recommend clinical assessment at every clinical visit to detect acute drug-induced movement disorders, for example, akathisia. These contradictions led the majority of experts in a recent Delphi consensus project on guidelines for managing TD to suggest that the lower limit of exposure to antipsychotics for TD to develop may be unknown but could reasonably be set at 1 month and that screening assessments therefore should be conducted at every clinical encounter.Citation60

Regarding instruments for assessing the severity and extent of TD, the Abnormal Involuntary Movement Scale (AIMS) has achieved widespread acceptance as an easy-to-use and objective measure.Citation61 The AIMS has been effective at enhancing compliance with routine monitoring and screening for TD and at fostering standardization and comparability between research trials.Citation62,Citation63 Of note, a recent expert consensus process on reporting outcomes of clinical trials based on AIMS data in clinically relevant terms concluded that no single analysis can be considered the standard of clinical efficacy and multiple analytic approaches are recommended.Citation62 For clinicians, the complete AIMS examination can be time-consuming to conduct formally in a busy office practice, and efforts have been suggested to develop an abridged, simpler AIMS or other semi-structured examination and screening instrument that could be used more frequently and routinely during all clinical encounters.Citation20,Citation60

Diagnosis of TD

Research criteria for the diagnosis of TD based on AIMS scores developed by Schooler and Kane, which require a rating of moderate movements in at least one body area or ratings of at least mild in two body areas, played an invaluable role in standardizing the diagnosis across virtually all subsequent clinical and epidemiologic studies.Citation56 A modification suggested by Glazer et al, which required only a total score of three with at least one body area rated as mild, offered a lower threshold but never gained currency in research studies.Citation64 However, observation of even mild movements in one body area (ie, an AIMS score of 2) in a clinical practice setting may be significant and sufficient for the diagnosis of TD. For example, patients who become aware of and embarrassed by mild lip-smacking cannot be ignored. Recognition of incipient signs of TD such as tic-like orofacial or lingual movements or increased eye blink frequency may be critical in allowing early treatment intervention and prevention of worsening or generalization of movements.

Before selecting treatment options for TD, it is essential to distinguish TD from other movement disorders which may require very different management. In general, the differential diagnosis of movement disorders is complex with a long list of primary and secondary neurodegenerative disorders, structural abnormalities of the brain, and metabolic and toxic etiologies, many of which are quite rare and require collaboration with movement disorder specialists for in-depth evaluation.Citation1 Potential clues to underlying disorders and the need for consultation include sudden onset or atypical or progressive course, positive family history, and associated systemic medical, neurological, laboratory or neuroimaging findings.

For the psychiatrist, the most likely disorders to differentiate from TD are spontaneous dyskinesias and other drug-induced movement disorders. Older people are at risk for spontaneous dyskinesias, especially involving orofacial muscles which may be associated with edentulousness or ill-fitting dentures.Citation65,Citation66 Psychomotor disorders are a core symptom dimension in schizophrenia and were recognized decades before antipsychotics were introduced.Citation3,Citation67Citation71 While TD may present in a variety of forms simulating classical neurological movement disorders (eg, chorea or dystonia), spontaneous movements found among psychiatric patients include a wide range of hyperkinetic (eg, mannerisms, stereotypies, compulsions) or hypokinetic behaviors (eg, psychomotor retardation, mutism, catalepsy).Citation72 Clinicians and supporting staff may be misled by the fact that neurological movement disorders including TD often fluctuate with anxiety, arousal, and distraction, disappear during sleep, and can be suppressed by voluntary exertion, which often leads to misinterpreting drug-induced movement disorders as intentional “acting out” or attention-seeking behaviors.

It is also important to differentiate TD from acute drug-induced movement disorders. For example, drug-induced parkinsonism characterized by bradykinesia and tremor begins days to weeks after antipsychotic initiation and generally improves after antipsychotic discontinuation or with anticholinergic medications.Citation73 Dopamine-depleting agents would significantly worsen parkinsonism. Similar considerations apply to acute drug-induced dystonic reactions and akathisia. Although movement disorders have been associated with other drug classes, TD-like movements that persist are unusual with drugs that do not block dopamine receptors.Citation74

Treatment of TD

Once TD is detected, a practical management strategy entails a progressive series of steps consistent with standard patient care ().Citation2,Citation73,Citation75 First, after visual inspection, a complete AIMS examination should be performed to document the severity and distribution of movements which could be monitored during subsequent treatment. Although not recorded on the AIMS, it would be prudent to also record the character or subtype of movements observed (eg, dystonia versus stereotyped movements). Second, a differential diagnosis should be considered to rule out spontaneous dyskinesias or other acute drug-induced movements, with neurological consultation if other less common disorders are suspected. Once TD is confirmed, a discussion should take place with patients and caregivers to inform them of the diagnosis, prognosis, and treatment options.Citation58

Table 1 Summary of proposed stepwise treatment algorithm for tardive dyskinesia (TD)

A number of pharmacological decisions then follow that are not necessarily mutually exclusive. Re-evaluating antipsychotic treatment is the first priority. Should the current antipsychotic be maintained, discontinued, reduced, or switched? In a patient with severe psychiatric illness at high risk for damaging relapse who is well-managed on current antipsychotic therapy but observed to have mild, localized TD with minimal subjective impact, maintenance of current treatment may be justified. In most cases, TD is not progressive even with continued antipsychotic treatment, although symptoms may worsen in some cases.Citation42,Citation76 Patients should provide informed consent with this plan and should be carefully monitored for any signs of progression. Alternatively and ideally, antipsychotics could be tapered off; in patients without an underlying psychotic disorder, such as those who develop TD while taking adjunctive dopamine antagonists for depression or for medical conditions, antipsychotics may be difficult to justify and could be safely discontinued. However, patients with schizophrenia or some cases of bipolar disorder may incur a significant risk of psychotic relapse and require ongoing maintenance treatment.Citation77 Although drug withdrawal had been routinely recommended in past guidelines, about 33%–53% of patients were reported to experience worsening of dyskinesias initially, while 36%–55% may have shown improvement over time after FGAs were discontinued.Citation78 But potential reversibility of TD after drug cessation has been controversial. While recent surveys suggest a grim prognosis with as few as 2% of patients showing resolution of TD several years after drug withdrawal in some studies,Citation79,Citation80 older reports found remission from TD a few months after drug cessation occurring in 50%–75% of patients if detected early.Citation45,Citation46 While these early cases may represent reversible withdrawal dyskinesias, they nevertheless support the potential efficacy of drug withdrawal in nonpsychotic patients as a management option.

Another recommended option has been dose reduction of antipsychotics. However, there is no evidence that this maneuver improves TD which in fact may temporarily worsen on decreased dosages. In addition, recent evidence suggests that dose reduction like drug cessation may contribute to symptom relapses and possible re-hospitalization, further eliminating this action as a reasonable treatment option for TD without additional evidence.Citation81 Another alternative is to switch antipsychotics if maintenance treatment is necessary or if the patient requests a change from the agent that caused TD. A change to more potent antipsychotics or FGAs may suppress symptoms of TD in up to 67% of patients, although limiting remission of TD and potentially exacerbating parkinsonism and other acute drug-induced movements.Citation82Citation84 One early study of the effects of FGAs on TD suggested that the suppressive effect was only temporary and TD symptoms may re-emerge during follow-up.Citation85 Similarly, several studies of SGAs have shown reduction in TD severity, with some studies showing greater suppression, lesser suppression, or no difference compared with FGAs.Citation42,Citation86Citation88 In fact, studies of SGAs in suppressing TD have shown about a 3–4 point average decrease in the total AIMS severity scores which is comparable to decreases in recent trials of dopamine-depleting analogs of tetrabenazine,Citation42,Citation87 but at a cost of continued dopamine-receptor blockade which perpetuates the etiologic mechanism of TD and presumably restricts chances of remission.

If anticholinergic drugs have been prescribed, a decision should also be made about whether continuation is necessary or gradual tapering could be considered. Anticholinergic drugs probably worsen TD generally, such that improvement in TD severity ratings have been noted in up to 60% of patients withdrawn from these agents.Citation75,Citation82,Citation84,Citation89,Citation90 But caution should be taken in decisions to reduce or taper anticholinergics if acute drug-induced movements (eg, parkinsonism, dystonia) or tardive dystonia are present as these disorders could re-emerge or worsen after withdrawal. Amantadine may be a reasonable alternative to anticholinergics in patients with TD and parkinsonism; it is now marketed in a long-acting formulation and evidence suggests it may have beneficial effects on both the movement disorder types.Citation90Citation94

If TD remains a significant problem once antipsychotic treatment is optimized, specific anti-dyskinetic drugs should then be considered. A broad range of agents have been proposed as treatments for TD derived from theories of the underlying pathophysiology that identify potential targets. For example, the prevailing heuristic theory is the dopamine-receptor supersensitivity hypothesis which posits that prolonged dopamine-receptor blockade results in an increase in numbers or sensitivity of post-synaptic receptors.Citation1,Citation53Citation55 While helpful in understanding the causative and suppressive effects of drugs that affect dopaminergic neurotransmission at the clinical level, animal models of dopamine-receptor supersensitivity in rodents and primates are inexact; dyskinesias observed after drug administration in animals may develop sooner than TD, may not be age-related, and invariably resolve after drug cessation. Cholinergic compounds have been tried based on a theorized opposing balance between dopamine and acetylcholine in the basal ganglia and the finding of cholinergic cell loss in the striatum after administration of antipsychotics in rats.Citation95 Similarly, animal studies revealing loss of striatal neurons containing gamma-aminobutyric acid (GABA) after antipsychotic treatment suggested that GABA-ergic compounds may alleviate TD.Citation1 Findings in animal models which showed that dopaminergic blockade results in excess dopamine turnover and oxidative free radical formation formed the basis for trials of antioxidants.Citation1 To synthesize these disparate effects of supersensitivity and interneuronal dysfunction underlying TD, animal models have been invoked to show that these abnormalities may in concert affect synaptic plasticity of glutaminergic synapses on striatal interneurons, resulting in an imbalance of the direct and indirect pathways, and abnormal output to the sensorimotor cortex.Citation1,Citation96,Citation97

The record of clinical trials of agents to treat TD is extensive. Virtually all reviews and meta-analyses of these trials have concluded that the evidence on effectiveness for most of these agents remains preliminary and inconclusive.Citation75,Citation86,Citation98,Citation99 However, these reviews have been of limited usefulness to clinicians because they focus on trial design and statistical validity but less so on tolerability, reliability, and availability of products, while antipsychotics and anticholinergics are analyzed equally with specific antidyskinetics, apart from their psychiatric necessity. They conclude that treatments studied have limited evidence, based on small trials that are often underpowered, uncontrolled, unblinded, from single sites, or unreplicated. Though most agents did not meet the threshold of evidence to justify approval, some may have efficacy for subgroups of patients with TD if more definitive trials could be conducted, that is, absence of evidence is not evidence of absence!

In contrast, recent state-of-the-art randomized controlled trials have provided robust evidence on the efficacy and tolerability of two tetrabenazine analogs, valbenazine, and deutetrabenazine. These drugs act to deplete dopamine and thereby reduce the severity of TD, by inhibiting vesicular monoamine transporter-2 (VMAT-2) in nerve terminals, which ordinarily protects dopamine from metabolic breakdown by sequestering monoamines in protective vesicles. Tetrabenazine was initially synthesized as an antipsychotic and subsequently shown to suppress TD.Citation100Citation102 Despite a lack of controlled trials of tetrabenazine, it is generally regarded as efficacious for the treatment of TD symptoms based on historical, open-label, observational studies, but is limited by a short half-life and side effects of sedation, parkinsonism, akathisia, and depression.Citation103 Valbenazine and deutetrabenazine were designed with improved pharmacokinetics to allow for a longer half-life and reduced fluctuations associated with differences in genetic metabolic rate and drug interactions. Rigorous trials have confirmed that both drugs are more effective than placebo in reducing severity of TD and are better tolerated than tetrabenazine.Citation75,Citation101,Citation102,Citation104 The management of TD is, therefore, likely to be transformed with the introduction of these VMAT-2 inhibitors as approved treatments for TD. These agents have also renewed interest in many questions about the biology and management of TD. For example, although recent short-term and year-long extension trials of VMAT-2 inhibitors offer clear evidence of efficacy and toler-ability in suppressing the severity of TD, even in patients continuing antipsychotic treatment, their impact on real-world, long-term outcomes remains to be investigated. There are as yet no head-to-head comparative trials of tetrabenazine and its analogs, so their relative effectiveness cannot be addressed. Similarly, comparisons between the VMAT-2 inhibitors and SGAs or other antidyskinetic agents have not been conducted, although tetrabenazine at least was shown to be more effective than haloperidol in suppressing TD.Citation85 In addition, the relative cost-effectiveness of approved treatments remains controversial;Citation105 ongoing studies of the social, psychological, and economic impact of TD are likely to provide crucial evidence to better inform cost–benefit treatment decisions in the near future.Citation20

The interaction between the VMAT-2 inhibitors and antipsychotics has also raised interesting questions about synergistic antipsychotic effects. For example, if VMAT-2 inhibitors were added early in treatment in first-episode patients, would that allow lower doses of antipsychotics to be used and therefore possibly reduce the risk of TD? However, there are no animal or human data on the efficacy and safety of VMAT-2 inhibitors used for prophylactic purposes in preventing TD, such that this use is not advisable at this time. On the contrary, dopamine depletion, like denervation, may theoretically add to the risk of supersensitivity, synaptic dysplasticity, and TD from antipsychotic-induced dopamine-receptor blockade.Citation97,Citation106Citation109 But the risk of VMAT-2 inhibitors independently causing TD seems slight given the scarcity of such reports of TD associated with VMAT-2 inhibitors, despite recent and ongoing long-term studies of tetrabenazine and novel VMAT-2 inhibitors. While tetrabenazine may have dopamine-receptor blocking properties associated with a potential risk of TD and other drug-induced movement disorders, the risk may be lower with valbenazine, which was developed specifically without significant off-target receptor-binding properties, and with deutetrabenazine as a result of the lower doses that can be used effectively. Finally, the determination of risk for causing or contributing to TD attributed to VMAT-2 inhibitors is confounded by the fact that patients receiving these agents already have TD and are usually receiving concomitant antipsychotic medications.

To what extent might these agents facilitate remission or reversibility of TD long-term? By suppressing TD symptoms without blocking dopamine receptors, these agents might allow patients who do not require maintenance antipsychotics to tolerate drug withdrawal and therefore facilitate natural reversal of the mechanisms underlying TD, that is, as a “bridge to recovery”? As suggestive evidence, approximately 25%–30% of patients enrolled in long-term extension studies of valbenazine continued to show strong response in the reduction of TD even 4 weeks after withdrawal from valbenazine.Citation110 Long-term withdrawal studies may provide answers in the future.

Conclusion

The availability of the VMAT-2 inhibitors may transform the treatment of TD, both in patients who may not require continued antipsychotic treatment and could be withdrawn from antipsychotics and in those with psychotic disorders who require maintenance antipsychotic treatment. Even so, these findings need to be tested in real-world settings over the long-term and taken into consideration as part of a broader, practical treatment algorithm () that provides a stepwise approach to the management of patients affected by TD. Availability of these treatments may also rekindle interest in TD and the need for re-education and development of standardized practice guidelines. Additional research into TD should include investigations of underlying pathophysiological mechanisms, genetic predisposition, and the efficacy of other potential antidyskinetic drugs, especially those targeting non-dopaminergic mechanisms, with the ultimate goal of achieving true reversibility and remission of TD.

Disclosure

Dr Caroff served as a consultant for Neurocrine Biosciences, Teva Pharmaceuticals, Osmotica Pharmaceuticals, and Disper-Sol Technologies and received a research grant from Neurocrine Biosciences. The author reports no other conflicts of interest in this work.

References

  • OwensDGCA Guide to the Extrapyramidal Side-Effects of Antipsychotic Drugs2nd edCambridge (UK)Cambridge University Press2014
  • CaroffSNCampbellECDrug-induced extrapyramidal syndromes: implications for contemporary practicePsychiatr Clin North Am20163939141110.1016/j.psc.2016.04.00327514296
  • CaroffSNUngvariGSCunningham OwensDGHistorical perspectives on tardive dyskinesiaJ Neurol Sci20183894910.1016/j.jns.2018.02.01529454494
  • KlineNSOn the rarity of “irreversible” oral dyskinesias following phenothiazinesAm J Psychiatry1968124485410.1176/ajp.124.8S.48
  • AnonymousThe London letter. A side effect of phenothiazinesCan Med Assoc J1964911081
  • AnonymousSide-effects of phenothiazine drugsBr J Med19642141210.1136/bmj.2.5422.1412
  • AnonymousIrreversible side effects of phenothiazinesJAMA19651913333345899755
  • TurekISDrug induced dyskinesia: reality or myth?Dis Nerv Syst197536397399807464
  • BrandonSMcClellandHAProtheroeCA study of facial dyskinesia in a mental hospital populationBr J Psychiatry19711181711845576277
  • EvansJHPersistent oral dyskinesia in treatment with phenothiazine derivativesLancet1965145846014241885
  • CraneGETardive dyskinesia in patients treated with major neuroleptics: a review of the literatureAm J Psychiatry19681248S404810.1176/ajp.124.8S.40
  • KlawansHLBergenDBruynGWPaulsonGWNeuroleptic-induced tardiave dyskinesias in nonpsychotic patientsArch Neurol1974303383394816841
  • KaneJMSmithJMTardive dyskinesia: prevalence and risk factors, 1959 to 1979Arch Gen Psychiatry1982394734816121548
  • CarbonMKaneJMLeuchtSCorrellCUTardive dyskinesia risk with first- and second-generation antipsychotics in comparative randomized controlled trials: a meta-analysisWorld Psychiatry20181733034010.1002/wps.2057930192088
  • CarbonMHsiehC-HKaneJMCorrellCUTardive dyskinesia prevalence in the period of second-generation antipsychotic use: a meta-analysisJ Clin Psychiatry201778e264e27810.4088/JCP.16r1083228146614
  • KaneJMTardive dyskinesia: epidemiological and clinical presentationBloomFEKupferDJPsychopharmacology: The Fourth Generation of ProgressNew YorkRaven Press199514851495
  • JesteDVTardive dyskinesia in older patientsJ Clin Psychiatry200061Suppl 42732
  • MillerDDMcEvoyJPDavisSMClinical correlates of tardive dyskinesia in schizophrenia: baseline data from the CATIE schizophrenia trialSchizophr Res200580334310.1016/j.schres.2005.07.03416171976
  • SolmiMPigatoGKaneJMCorrellCUClinical risk factors for the development of tardive dyskinesiaJ Neurol Sci2018389212710.1016/j.jns.2018.02.01229439776
  • CaroffSNCutlerAJTannerCMAwareness and Impact of Possible Tardive Dyskinesia by Primary Psychiatric Diagnosis in Patients Prescribed Antipsychotics: Results from the RE-KINECT StudyOrlando (FL)Academy of Managed Care Pharmacy (AMCP) Nexus 20182018
  • KeckPEJrMcElroySLStrakowskiSMSoutulloCAAntipsychotics in the treatment of mood disorders and risk of tardive dyskinesiaJ Clin Psychiatry200061Suppl 43338
  • CoplanJGuggerJJTasleemHTardive dyskinesia from atypical antipsychotic agents in patients with mood disorders in a clinical settingJ Affect Disord201315086887110.1016/j.jad.2013.04.05323726783
  • RajanTMBharadwajBRajkumarRPAdolePSFrequency and correlates of tardive dyskinesia in Indian patients with type I bipolar disorderAsian J Psychiatr201832929810.1016/j.ajp.2017.12.00129222987
  • CorrellCULeuchtSKaneJMLower risk for tardive dyskinesia associated with second-generation antipsychotics: a systematic review of 1-year studiesAm J Psychiatry200416141442510.1176/appi.ajp.161.3.41414992963
  • BeasleyCMDellvaMATamuraRNRandomised double-blind comparison of the incidence of tardive dyskinesia in patients with schizophrenia during long-term treatment with olanzapine or haloperidolBr J Psychiatry1999174233010211147
  • JesteDVLacroJPBaileyARockwellEHarrisMJCaligiuriMPLower incidence of tardive dyskinesia with risperidone compared with haloperidol in older patientsJ Am Geriatr Soc19994771671910366172
  • CorrellCUSchenkEMTardive dyskinesia and new antipsychoticsCurr Opin Psychiatry20082115115610.1097/YCO.0b013e3282f5313218332662
  • LiebermanJAStroupTSMcEvoyJPEffectiveness of antipsychotic drugs in patients with chronic schizophreniaN Engl J Med20053531209122310.1056/NEJMoa05168816172203
  • MillerDDCaroffSNDavisSMExtrapyramidal side-effects of antipsychotics in a randomised trialBr J Psychiatry200819327928810.1192/bjp.bp.108.05008818827289
  • LeuchtSWahlbeckKHamannJKisslingWNew generation antipsychotics versus low-potency conventional antipsychotics: a systematic review and meta-analysisLancet20033611581158910.1016/S0140-6736(03)13306-512747876
  • WoodsSWMorgensternHSaksaJRIncidence of tardive dyskinesia with atypical versus conventional antipsychotic medications: a prospective cohort studyJ Clin Psychiatry20107146347410.4088/JCP.07m03890yel20156410
  • KaneJMWoernerMGPollackSSaffermanAZLiebermanJADoes clozapine cause tardive dyskinesia?J Clin Psychiatry1993543273308104929
  • CraneGEPrevention and management of tardive dyskinesiaAm J Psychiatry197212946646710.1176/ajp.129.4.4665071149
  • AlexopoulosGSLack of complaints in schizophrenics with tardive dyskinesiaJ Nerv Ment Dis197916712512733230
  • MacphersonRCollisRTardive dyskinesia. Patients’ lack of awareness of movement disorderBr J Psychiatry19921601101121347479
  • MarsdenCDTarsyDBaldessariniRJSpontaneous and drug-induced movement disorders in psychotic patientsBensonDFBlumerDPsychiatric Aspects of Neurologic DiseaseNew YorkGrune & Stratton1975219253
  • SandykRKaySRAwerbuchGISubjective awareness of abnormal involuntary movements in schizophreniaInt J Neurosci1993691207916006
  • YassaRFunctional impairment in tardive dyskinesia: medical and psychosocial dimensionsActa Psychiatr Scand19898064672763861
  • TarsyDHistory and definition of tardive dyskinesiaClin Neuropharmacol1983691996133621
  • Ascher-SvanumHZhuBFariesDPengXKinonBJTohenMTardive dyskinesia and the 3-year course of schizophrenia: results from a large, prospective, naturalistic studyJ Clin Psychiatry2008691580158819192441
  • BrowneSRoeMLaneAQuality of life in schizophrenia: relationship to sociodemographic factors, symptomatology and tardive dyskinesiaActa Psychiatr Scand1996941181248883573
  • CaroffSNDavisVGMillerDDTreatment outcomes of patients with tardive dyskinesia and chronic schizophreniaJ Clin Psychiatry20117229530310.4088/JCP.09m05793yel20816031
  • StrassnigMRosenfeldAHarveyPDTardive dyskinesia: motor system impairments, cognition and everyday functioningCNS Spectr20182337037728877766
  • CloudLJZutshiDFactorSATardive dyskinesia: therapeutic options for an increasingly common disorderNeurotherapeutics20141116617610.1007/s13311-013-0222-524310603
  • QuitkinFRifkinAGochfeldLKleinDFTardive dyskinesia: are first signs reversible?Am J Psychiatry1977134848710.1176/ajp.134.1.8412666
  • JesteDVPotkinSGSinhaSFederSWyattRJTardive dyskinesia–reversible and persistentArch Gen Psychiatry19793658559035116
  • CraneGEPersistent dyskinesiaBr J Psychiatry19731223954054577946
  • FaurbyeARaschPJPetersenPBBrandborgGPakkenbergHNeurological symptoms in pharmacotherapy of psychosesActa Psychiatr Scand196440102714217630
  • SchoneckerMParoxysmal dyskinesia as the effect of megaphenNervenarzt19572855055313517450
  • UhrbrandLFaurbyeAReversible and irreversible dyskinesia after treatment with perphenazine, chlorpromazine, reserpine, and electroconvulsive therapyPsychopharmacologia1960140841810.1007/BF00441188
  • KaneJMWoernerMLiebermanJTardive dyskinesia: prevalence, incidence, and risk factorsJ Clin Psychopharmacol1988852S56S3065365
  • TurekIKurlandAAHanlonTEBohmMTardive dyskinesia: its relation to neuroleptic and antiparkinson drugsBr J Psychiatry19721216056124648433
  • JesteDVLohrJBKauffmannCAWyattRJPathophysiology of tardive dyskinesia; Evaluation of supersensitivity theory and alertnative hypothesesCaseyDEGardosGTardive Dyskinesia and Neuroleptics: From Dogma to ReasonWashington (DC)American Psychiatric Press, Inc19861532
  • MooreKEDyskinesia: animal experimental correlatesActa Psychiatr Scand1981638810210.1111/j.1600-0447.1981.tb02461.x
  • ChristensenAVFjallandBNielsenIMOn the supersensitivity of dopamine receptors, induced by neurolepticsPsychopharmacology197648169655
  • SchoolerNRKaneJMResearch diagnoses for tardive dyskinesiaArch Gen Psychiatry198239486487
  • American Psychiatric AssociationDiagnostic and Statistical Manual of Mental Disorders4th edWashington (DC)American Psychiatric Press, Inc.1994
  • American Psychiatric AssociationTardive Dyskinesia: A Task Force Report of the American Psychiatric AssociationAmerican Psychiatric Press. Inc.Washington (DC)1992
  • LehmanAFLiebermanJADixonLBPractice guideline for the treatment of patients with schizophrenia, second editionAm J Psychiatry2004161156
  • CaroffSNCitromeLMeyerJA Modified Delhi Consensus Approach to Clinical Guidelines for Tardive DyskinesiaNew YorkAnnual Meeting of the American Psychiatric Association2018
  • GuyWAbnormal Involuntary Movement Scale (AIMS) ECDEU Assessment Manual for Psychopharmacology Revised: Alcohol, Drug Abuse and Mental Health AdministrationNational Institute of Mental HealthRockville, MD1976534537
  • KaneJMCorrellCUNierenbergAACaroffSNSajatovicMTardive Dyskinesia Assessment Working Group. Revisiting the abnormal involuntary movement scale: proceedings from the tardive dyskinesia assessment workshopJ Clin Psychiatry201879317cs11959
  • MunetzMRSchulzSCScreening for tardive dyskinesiaJ Clin Psychiatry19864775772867994
  • GlazerWMMorgensternHDoucetteJTPredicting the long-term risk of tardive dyskinesia in outpatients maintained on neuroleptic medicationsJ Clin Psychiatry1993541331398098030
  • LiebermanJKaneJMWoernerMWeinholdPPrevalence of tar-dive dyskinesia in elderly samplesPsychopharmacol Bull19842022266718647
  • KaneJMWeinholdPKinonBWegnerJLeaderMPrevalence of abnormal involuntary movements (“spontaneous dyskinesias”) in the normal elderlyPsychopharmacology1982771051086812127
  • ManschreckTCKopalaLCHonerWGNeurological comorbidity and features in schizophreniaMeyerJMNasrallahHAMedical Illness and SchizophreniaArlington (VA)American Psychiatric Publishing, Inc2003185214
  • PeraltaVCuestaMJMotor abnormalities: from neurodevelopmental to neurodegenerative through “functional” (neuro)psychiatric disordersSchizophr Bull20174395697110.1093/schbul/sbx08928911050
  • PappaSDazzanPSpontaneous movement disorders in antipsychotic-naive patients with first-episode psychoses: a systematic reviewPsychol Med2009391065107610.1017/S003329170800471619000340
  • KhotVWyattRJNot all that moves is tardive dyskinesiaAm J Psychiatry199114866166610.1176/ajp.148.5.6611673323
  • FentonWSPrevalence of spontaneous dyskinesia in schizophreniaJ Clin Psychiatry200061Suppl 41014
  • RichardIO’BrienCKurlanRSpontaneous movement disorders in psychiatric patientsFactorSALangAEWeinerWJDrug Induced Movement Disorders2nd edMalden (MA)Blackwell Publishing Inc.20053052
  • CitromeLReprint of: clinical management of tardive dyskinesia: five steps to successJ Neurol Sci2018389616610.1016/j.jns.2018.02.03729519687
  • D’AbreuAFriedmanJHTardive dyskinesia-like syndrome due to drugs that do not block dopamine receptors: rare or non-existent: literature reviewTremor Other Hyperkinet Mov (New York, NY)20188570
  • CaroffSNCampbellECCarrollBPharmacological treatment of tardive dyskinesia: recent developmentsExpert Rev Neurother20171787188110.1080/14737175.2017.135861628727483
  • MargoliusAFernandezHHCurrent treatment of tardive dyskinesiaParkinsonism Relat Disord2018 Epub121910.1016/j.parkreldis.2018.12.022
  • GilbertPLHarrisMJMcAdamsLAJesteDVNeuroleptic withdrawal in schizophrenic patients. A review of the literatureArch Gen Psychiatry19955217318810.1001/archpsyc.1995.039501500050017872841
  • CaseyDEGerlachJTardive dyskinesia: what is the long-term outcome?CaseyDEGardosGTardive Dyskinesia and Neuroleptics: From Dogma to ReasonWashington (DC)American Psychiatric Press, Inc19867697
  • ZutshiDCloudLJFactorSATardive syndromes are rarely reversible after discontinuing dopamine receptor blocking agents: experience from a university-based movement disorder clinicTremor Other Hyperkinet Mov (N Y)2014426625374768
  • GlazerWMMooreDCSchoolerNRBrennerLMMorgensternHTardive dyskinesia. A discontinuation studyArch Gen Psychiatry1984416236276145399
  • CaroffSNMuFAyyagariRSchillingTAblerVCarrollBHospital utilization rates following antipsychotic dose reductions: implications for tardive dyskinesiaBMC Psychiatry20181830610.1186/s12888-018-1889-230249218
  • EganMFApudJWyattRJTreatment of tardive dyskinesiaSchizophr Bull1997235836099365997
  • MentzelCLBakkerPRvan OsJEffect of antipsychotic type and dose changes on tardive dyskinesia and parkinsonism severity in patients with a serious mental illness: the curacao extrapyramidal syndromes study XIIJ Clin Psychiatry201778e279e28510.4088/JCP.16m1104928199071
  • Soares-WeiserKFernandezHHTardive dyskinesiaSemin Neurol20072715916910.1055/s-2007-97116917390261
  • KazamatsuriHChienCPColeJOLong-term treatment of tardive dyskinesia with haloperidol and tetrabenazineAm J Psychiatry197313047948310.1176/ajp.130.4.4794570750
  • BhidayasiriRFahnSWeinerWJGronsethGSSullivanKLZesiewiczTAEvidence-based guideline: treatment of tardive syndromes: report of the Guideline Development Subcommittee of the American Academy of NeurologyNeurology20138146346910.1212/WNL.0b013e31829d86b623897874
  • MentzelTQvan der SnoekRLieverseRClozapine mono-therapy as a treatment for antipsychotic-induced tardive dyskinesia: a meta-analysisJ Clin Psychiatry201879617r1185210.4088/JCP.su17023ah3c
  • TarsyDBaldessariniRJTaraziFIEffects of newer antipsychotics on extrapyramidal functionCNS Drugs200216234510.2165/00023210-200216010-0000311772117
  • GreilWHaagHRossnaglGRutherEEffect of anticholinergics on tardive dyskinesia. A controlled discontinuation studyBr J Psychiatry19841453043106148119
  • WardKMCitromeLAntipsychotic-related movement disorders: drug-induced parkinsonism vs. tardive dyskinesia-key differences in pathophysiology and clinical managementNeurol Ther2018723324810.1007/s40120-018-0105-030027457
  • PappaSTsouliSApostolouGMavreasVKonitsiotisSEffects of amantadine on tardive dyskinesia: a randomized, double-blind, placebo-controlled studyClin Neuropharmacol20103327127521121175
  • ElkurdMTBahrooLBPahwaRThe role of extended-release amantadine for the treatment of dyskinesia in Parkinson’s disease patientsNeurodegener Dis Manag20188738010.2217/nmt-2018-000129564954
  • FannWELakeCRAmantadine versus trihexyphenidyl in the treatment of neuroleptic-induced parkinsonismAm J Psychiatry197613394094310.1176/ajp.133.8.940782262
  • DiMascioABernardoDLGreenblattDJMarderJEA controlled trial of amantadine in drug-induced extrapyramidal disordersArch Gen Psychiatry1976335996025066
  • MargoleseHCChouinardGKolivakisTTBeauclairLMillerRTardive dyskinesia in the era of typical and atypical antipsychotics. Part 1: pathophysiology and mechanisms of inductionCan J Psychiatry20055054154710.1177/07067437050500090716262110
  • TeoJTEdwardsMJBhatiaKTardive dyskinesia is caused by maladaptive synaptic plasticity: a hypothesisMov Disord2012271205121510.1002/mds.2510722865512
  • BorgkvistALiebermanOJSulzerDSynaptic plasticity may underlie l-DOPA induced dyskinesiaCurr Opin Neurobiol201848717810.1016/j.conb.2017.10.02129125979
  • BhidayasiriRJitkritsadakulOFriedmanJHFahnSUpdating the recommendations for treatment of tardive syndromes: a systematic review of new evidence and practical treatment algorithmJ Neurol Sci2018389677510.1016/j.jns.2018.02.01029454493
  • Soares-WeiserKRathboneJOgawaYShinoharaKBergmanHMiscellaneous treatments for antipsychotic-induced tardive dyskinesiaCochrane Database Syst Rev20183Cd00020829552749
  • BrandrupETetrabenacine treatment in persisting dyskinesia caused by psychopharmacaAm J Psychiatry196111855155210.1176/ajp.118.6.55113872499
  • CaroffSNAggarwalSYonanCTreatment of tardive dyskinesia with tetrabenazine or valbenazine: a systematic reviewJ Comp Eff Res2018713514810.2217/cer-2017-006528965423
  • ScorrLMFactorSAVMAT2 inhibitors for the treatment of tardive dyskinesiaJ Neurol Sci2018389434710.1016/j.jns.2018.02.00629433808
  • JankovicJClarence-SmithKTetrabenazine for the treatment of chorea and other hyperkinetic movement disordersExpert Rev Neurother2011111509152310.1586/ern.11.14922014129
  • CitromeLTardive dyskinesia: placing vesicular monoamine transporter Type 2 (VMAT2) inhibitors into clinical perspectiveExpert Rev Neurother20181832333210.1080/14737175.2018.145550429557243
  • AtlasSJAgboolaFCurfmanGEffectiveness and value of 2 novel treatments for tardive dyskinesiaJAMA Intern Med20181781110111210.1001/jamainternmed.2018.246329913006
  • LeWittPATardive dyskinesia caused by tetrabenazineClin Neuropharmacol201336929310.1097/WNF.0b013e318290cd4123673911
  • MandelRJHartgravesSLSeversonJAWoodwardJJWilcoxRERandallPKA quantitative estimate of the role of striatal D-2 receptor proliferation in dopaminergic behavioral supersensitivity: the contribution of mesolimbic dopamine to the magnitude of 6-OHDA lesion-induced agonist sensitivity in the ratBehav Brain Res19935953648155293
  • MoySSCriswellHEBreeseGRDifferential effects of bilateral dopamine depletion in neonatal and adult ratsNeurosci Biobehav Rev1997214254359195600
  • KostrzewaRMBrusRLifelong rodent model of tardive dyskinesia-persistence after antipsychotic drug withdrawalCurr Top Behav Neurosci20162935336210.1007/7854_2015_39526472552
  • CaroffSNLindenmayerJPFarahmandKBurkeJSiegertSCharacteristics of patients with tardive dyskinesia who maintain treatment response after discontinuing long-term valbenazine: pooled analysis of two trialsThe American College of Neuropsychopharmacology Annual Meeting2018Hollywood, FL