psychiatrist

This work may not be copied, distributed, displayed, published, reproduced, transmitted, modified, posted, sold, licensed, or used for commercial purposes. By downloading this file, you are agreeing to the publisher’s Terms & Conditions.

ACADEMIC HIGHLIGHTS

Measurement-Based Diagnosis and Treatment for Tardive Dyskinesia

Christoph U. Correll, MD,a and Leslie Citrome, MD, MPHb

Published: August 31, 2021

Tardive dyskinesia (TD) is an involuntary movement disorder associated with agents that block dopamine receptors, particularly antipsychotics. TD commonly involves the orofacial muscles and extremities, and, because these movements are out of the patient’s control, they can have serious physical and psychological effects. An accurate and early diagnosis of TD is crucial because the risk of permanence increases over time. To minimize the risk of TD development, clinicians should use the lowest necessary doses of dopamine receptor blocking agents, and, if allowed by the treated condition, the dopamine receptor blocking agents should be stopped after the shortest necessary time. Clinicians should try to avoid parkinsonian adverse effects and akathisia and prefer second-generation antipsychotics over first-generation antipsychotics. Moreover, clinicians should differentiate between TD and other drug-induced movement disorders, particularly drug-induced parkinsonism, as anticholinergic treatment can worsen TD. To facilitate measurement-based care, clinicians should use the Abnormal Involuntary Movement Scale examination to screen for and routinely monitor TD, especially when providing treatments intended to decrease the symptoms and impact of TD. Two vesicular monoamine transporter-2 (VMAT2) inhibitors, deutetrabenazine and valbenazine, are approved by the US Food and Drug Administration to treat TD. For patients who have moderate to severe or disabling TD, the American Psychiatric Association recommends treatment with the VMAT2 inhibitors. Clinicians should communicate with patients and care partners about risk factors for and signs of TD, as well as available treatment options for TD and what they can expect in terms of short- and long-term results.


This CME activity is expired. For more CME activities, visit CMEInstitute.com.
Find more articles on this and other psychiatry and CNS topics:
The Journal of Clinical Psychiatry
The Primary Care Companion for CNS Disorders

 

To cite: Correll CU, Citrome LL. Measurement-based diagnosis and treatment for tardive dyskinesia. J Clin Psychiatry. 2021;82(5):NU20016AH2C.
To share: https://doi.org/10.4088/JCP.NU20016AH2C

© Copyright 2021 Physicians Postgraduate Press, Inc.

aZucker School of Medicine at Hofstra/Northwell, Hempstead, New York; Charité Universitätsmedizin Berlin, Germany
bNew York Medical College, Valhalla

Tardive dyskinesia (TD) is a drug-induced movement disorder associated with agents that block dopamine receptors, particularly antipsychotics. TD commonly involves involuntary movements of the orofacial muscles and the extremities. As a consequence, TD can be associated with serious physical and psychological impairments.1 This report, based on presentations given by Christoph U. Correll, MD, and Leslie Citrome, MD, MPH, will address how to use measurement-based care to diagnose TD and manage TD treatment.

MEASUREMENT-BASED DIAGNOSIS

Because TD is brought on by the continued use of dopamine receptor blocking agents, such as antipsychotics, many of those at risk are patients with schizophrenia, bipolar disorder, and major depressive disorder.2 Dr Citrome explained that treating these psychiatric conditions is critical, and many patients may be unable to avoid long-term treatment with these medications despite the risk of TD.

Tardive Dyskinesia Risk Factors and Prevention

Dr Citrome stated that 1 of 4 people who have been exposed to dopamine receptor blocking agents have TD, with a higher percentage among those who have been exposed to first-generation antipsychotics (FGAs).3 A meta-analysis4 of prospective, randomized studies showed that second-generation antipsychotics (SGAs) have a significantly lower risk for TD than FGAs. Among individuals who have been exposed only to SGAs, the incidence rate was about 7%.3 Other risk factors for TD include older age, female sex, presence of a mood disorder, higher cumulative exposure to antipsychotics, emergence of parkinsonism or akathisia, treatment with anticholinergic medications, intermittent antipsychotic treatment, and a history of movement disorders.3,5

Dr Correll noted the importance of managing the risk of TD by educating patients and caregivers about the risks of and alternatives to antipsychotic medication, as well as early signs of TD to watch for if these medications are taken long-term. Clinicians should confirm and document the indication for use of antipsychotics and use conservative maintenance doses; if allowed by the treated condition, the antipsychotic should be stopped after the shortest necessary time.6 Clinicians should also consider the use of SGAs instead of FGAs, especially for patients at high risk for drug-induced movement disorders, including TD.

Screening for Tardive Dyskinesia

The most recent American Psychiatric Association (APA) guidelines for the treatment of patients with schizophrenia7 recommend routine assessments for abnormal movements at a set frequency with a formal examination. The use of a structured instrument, such as the Abnormal Involuntary Movement Scale (AIMS), is the standard of care.8 The AIMS is an observer-rated, 12-item anchored scale. The AIMS itself is not diagnostic (the clinician needs to consider other explanations for any observable abnormal motor movements), but the AIMS can assist in the quantification of the dyskinetic movements, which can be very helpful in documenting a baseline prior to treatment and in providing patient education.

Dr Citrome stated that follow-up assessments using the AIMS should be conducted on a regular basis. Patients who use FGAs should be examined for TD every 6 months, patients who use SGAs and no concomitant FGAs should be examined annually, and patients at high risk for TD should be examined every 3 months if taking FGAs or every 6 months if taking SGAs.2

To view an in-depth discussion between Drs Citrome and Correll on incorporating the AIMS into clinical practice, see the on-demand presentation titled “Diagnostic and Treatment Fundamentals for Tardive Dyskinesia” in this series at cmeinstitute.com.

Differential Diagnosis

Dr Citrome advised that TD must be differentiated from other movement disorders, particularly drug-induced parkinsonism.9 Other conditions to consider in the differential diagnosis of TD include spontaneous dyskinesias that can be encountered especially in the elderly, drug-induced dyskinesias caused by other agents, chronic motor tic disorder, Tourette syndrome, autism (stereotypies/stimming), restless legs syndrome, and oral movements from ill-fitting dentures and other dental problems, as well as rarer disorders such as Huntington disease/senile chorea, Meige syndrome, Sydenham chorea, Rett syndrome, and Wilson disease.8,10

Patient Perspectives

Here, in an interview with a representative of Mental Health America (MHA) in January 2021, a patient described the impact of TD on her work:

“I am fluent in American Sign Language (ASL). I went to school to be an interpreter. I’m completely deaf in one ear. I can’t be an interpreter anymore because the [TD] movements were so severe I couldn’t hold my hands steady long enough to sign in a way where people could typically understand me. I was also certified to be a body piercer. I can’t do that anymore because it’s a safety hazard. So, I just stopped that. What I was able to do and what I was trained and certified to do, I can no longer do.”

Case Practice Question

You are seeing a 34-year-old man with schizophrenia for an intake visit, and you notice that he has abnormal involuntary movements. His clinical history indicates that he has been treated with second-generation antipsychotic medication since his diagnosis 12 years ago. He has had parkinsonism and akathisia, for which he received anticholinergic treatment. You decide to perform an Abnormal Involuntary Movement Scale (AIMS) evaluation. Which of the following statements about the AIMS is correct?

a. The AIMS is a diagnostic tool for TD.

b. Given the time required, guidelines do not recommend routine AIMS assessments.

c. The AIMS can assist in measurement-based care and patient education.

d. All of the above

Discussion of the Case Practice Question

Preferred response: c. The AIMS can assist in measurement-based care and patient education.

Explanation: The AIMS is not a diagnostic tool, as other medical reasons for the dyskinetic movements need to be ruled out first. This patient is at risk for TD because of long-term antipsychotic treatment, even though he took second-generation medications. Extrapyramidal side effects such as parkinsonism and akathisia are relevant risk factors for TD, and anticholinergic medications for them do not reduce, but potentially further increase, TD risk. The APA guidelines recommend regular use of structured assessments such as the AIMS, in addition to clinical assessments at each visit, to facilitate measurement-based care. The AIMS can be helpful in assisting in the quantification of the dyskinetic movements, which can be useful when documenting a baseline prior to treatment and can aid patient education.

MANAGEMENT APPROACHES

In his presentation, Dr Correll discussed approaches to managing TD. If possible, clinicians may stop the antipsychotic or reduce the dose.11 Increasing the dose could mask TD, but symptoms may re-emerge, leading to symptom perpetuation.12 Switching from an FGA to an SGA, or from one SGA antipsychotic to another, are other interventions to consider.11 Dr Correll also highlighted other antidyskinesia agents that may treat TD, all of them with varying degrees of evidence supporting their use, such as vitamin E, vitamin B6, ginkgo biloba, and eicosapentaenoic acid, as well as melatonin, clonazepam, amantadine, donepezil and branched-chain amino acids.11–13 Reserpine, an irreversible vesicular monoamine transporter-2 (VMAT2) inhibitor, has too many adverse effects to be considered, and tetrabenazine, an older reversible VMAT2 inhibitor, also is a potential treatment option, but rigorous trials for TD are missing for tetrabenazine.13 These treatments are not approved by the US Food and Drug Administration (FDA) to treat TD.11

FDA-Approved Treatments

Dr Correll focused his presentation on 2 FDA-approved agents to treat TD that are recommended by the American Psychiatric Association for the treatment of moderate to severe or disabling TD.7,14 Both drugs, deutetrabenazine and valbenazine, are reversible VMAT2 inhibitors that reduce the uptake of biogenic amines, particularly dopamine, into presynaptic vesicles.15 Studies have demonstrated that both medications are effective in reducing symptoms of TD acutely and long-term without increased risk of depression or suicidality. 14 Dr Correll compared key differentiating features of the 2 treatments (Figure 1).16 Regardless of which treatment is selected, routine monitoring using the AIMS is highly desirable and is essential to providing measurement-based care.7


Figure 1. Key Features  of Deutetrabenazine and Valbenazine

Reprinted with permission from Citrome and Saklad.16


Deutetrabenazine. Deutetrabenazine is closely related chemically to tetrabenazine but is a deuterated drug—selected hydrogen atoms on the tetrabenazine molecule have been replaced with deuterium (a stable, naturally occurring, nonradioactive isotope of hydrogen).17 Deuteration alters the pharmacokinetics of tetrabenazine so that deutetrabenazine has a longer half-life and a lower maximum plasma concentration than tetrabenazine. Increased tolerability and twice-daily dosing (vs 3 doses per day with tetrabenazine) may increase treatment adherence among patients. The initial dose of deutetrabenazine is 6 mg twice per day.14 The dose may be increased by increments of 6 mg/d at weekly intervals with a maximum dose of 24 mg twice a day.

In acute studies, deutetrabenazine was superior to placebo in reducing the severity of dyskinetic movements, measured with the AIMS.14 In an open-label extension study,18 deutetrabenazine showed improvement in both clinician- and patient-rated Global Impression of Change scale scores. At week 6, more than 50% of patients had achieved a “much improved” or “very much improved” outcome. Two-thirds of patients achieved that status by 9 to 12 months, and 3 of 4 patients had done so by the end of 2 years.18

Valbenazine. Valbenazine is a highly selective VMAT2 inhibitor. Valbenazine and its principal active metabolite have selective VMAT2 binding,19 limiting off-target receptor binding. Its half-life is 20 hours, which allows for once-daily dosing. The initial dose  for valbenazine is 40 mg/d, and the maintenance target dose is 80 mg/d, which can be achieved 1 week after the initial dose.14,19

In acute studies, valbenazine was superior to placebo in reducing the severity of dyskinetic movements, measured with the AIMS.14 An extension study20 showed that at the end of one year, 3 of 4 patients receiving the higher dose of 80 mg/d achieved sustained improvement in both clinician and patient ratings. The lower dose of 40 mg/d led to similar improvement in 60% of patients.

Patient Perspectives

In an interview conducted by MHA in January 2021, a patient described the relief from symptoms that she experienced after receiving effective TD medication:

“I’m currently on [an FDA-approved VMAT 2 inhibitor]. It’s taken my movements from a 9 to like a 4 or a 5. [My TD is now] much more tolerable. It’s to where people don’t notice it as much, that they’re not looking at my hands or my legs, focusing in. I don’t have vocal tics anymore unless I’m incredibly stressed out.”

Patient and Care Partner Communication and Education

According to Dr Correll, clinical experience suggests that most patients with moderate to severe or disabling TD would be willing to take medication to achieve a reduction in their symptoms, particularly if the medication is well tolerated.21 Evidence indicates that the potential benefits of VMAT2 inhibitors for the treatment of TD far outweigh the potential harms.14

Dr Correll concluded by emphasizing good communication with patients and caregivers about risk factors for TD, signs of TD, and treatment options. It is important for clinicians to inquire about and address TD symptoms with each patient and jointly develop treatment goals. The AIMS examination can provide a backdrop for these discussions. Dr Correll offered these example questions: “What is it that you would want the most for yourself (or your loved one) from an effective treatment? What are your goals? How can we measure the outcome to see whether we reach your goals?” Dr Correll also emphasized the importance of setting expectations with patients about medications and their side effects.

Clinical Points

  • Differentiate between TD and other drug-induced movement disorders, particularly drug-induced parkinsonism.
  • Use the AIMS examination to screen for and routinely monitor TD, especially when providing treatments intended to decrease the symptoms of TD.
  • For patients who have moderate to severe or disabling TD, the APA recommends treatment with FDA-approved VMAT2inhibitors deutetrabenazine and valbenazine.
  • Discuss treatment goals and expectations with patients and care partners.

References

  1. Jain R, Correll CU. Tardive dyskinesia: recognition, patient assessment, and differential diagnosis. J Clin Psychiatry. 2018;79(2):nu17034ah1c. PubMed
  2. Citrome L. Clinical management of tardive dyskinesia: five steps to success. J Neurol Sci. 2017;383:199–204. PubMed CrossRef
  3. Carbon M, Hsieh C-H, Kane JM, et al. Tardive dyskinesia prevalence in the period of second-generation antipsychotic use: a meta-analysis. J Clin Psychiatry. 2017;78(3):e264–e278. PubMed CrossRef
  4. Carbon M, Kane JM, Leucht S, et al. Tardive dyskinesia risk with first- and second-generation antipsychotics in comparative randomized controlled trials: a meta-analysis. World Psychiatry. 2018;17(3):330–340. PubMed CrossRef
  5. Solmi M, Pigato G, Kane JM, et al. Clinical risk factors for the development of tardive dyskinesia. J Neurol Sci. 2018;389:21–27. PubMed CrossRef
  6. Caroff SN, Miller DD, Dhopesh V, et al. Is there a rational management strategy for tardive dyskinesia? Curr Psychiatr. 2011;10(10):22–32.
  7. Keepers GA, Fochtmann LJ, Anzia JM, et al. The American Psychiatric Association Practice Guideline for the Treatment of Patients With Schizophrenia. 3rd ed. American Psychiatric Association Publishing; 2021.
  8. Kane JM, Correll CU, Nierenberg AA, et al. Revisiting the Abnormal Involuntary Movement Scale: proceedings from the Tardive Dyskinesia Assessment Workshop. J Clin Psychiatry. 2018;79(3):17cs11959. PubMed CrossRef
  9. Ward KM, Citrome L; Tardive Dyskinesia-Key Differences in Pathophysiology and Clinical Management. Antipsychotic-related movement disorders: drug-induced Parkinsonism vs tardive dyskinesia-key differences in pathophysiology and clinical management. Neurol Ther. 2018;7(2):233–248. PubMed CrossRef
  10. Kane JM. Assessing patients for tardive dyskinesia. J Clin Psychiatry. 2017;78(9):e1428. PubMed CrossRef
  11. Kaspar R, Ellingrod VL. Strategies for managing drug-induced tardive dyskinesia. Curr Psychiatr. 2014;13(3):44–46.
  12. Bhidayasiri R, Fahn S, Weiner WJ, et al; American Academy of Neurology. Evidence-based guideline: treatment of tardive syndromes: report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology. 2013;81(5):463–469. PubMed CrossRef
  13. Kim J, Macmaster E, Schwartz TL. Tardive dyskinesia in patients treated with atypical antipsychotics: case series and brief review of etiologic and treatment considerations. Drugs Context. 2014;3:212259. PubMed CrossRef
  14. Solmi M, Pigato G, Kane JM, et al. Treatment of tardive dyskinesia with VMAT-2 inhibitors: a systematic review and meta-analysis of randomized controlled trials. Drug Des Devel Ther. 2018;12:1215–1238. PubMed CrossRef
  15. Bernstein AI, Stout KA, Miller GW. The vesicular monoamine transporter 2: an underexplored pharmacological target. Neurochem Int. 2014;73:89–97. PubMed CrossRef
  16. Citrome L, Saklad SR. Revisiting tardive dyskinesia: focusing on the basics of identification and treatment. J Clin Psychiatry. 2020;81(2):TV18059AH3C. PubMed CrossRef
  17. Citrome L. Breakthrough drugs for the interface between psychiatry and neurology. Int J Clin Pract. 2016;70(4):298–299. PubMed CrossRef
  18. Fernandez HH, Stamler D, Davis MD, et al. Long-term safety and efficacy of deutetrabenazine for the treatment of tardive dyskinesia. J Neurol Neurosurg Psychiatry. 2019;90(12):1317–1323. PubMed CrossRef
  19. O’Brien CF, Jimenez R, Hauser RA, et al. NBI-98854, a selective monoamine transport inhibitor for the treatment of tardive dyskinesia: a randomized, double-blind, placebo-controlled study. Mov Disord. 2015;30(12):1681–1687. PubMed CrossRef
  20. Factor SA, Remington G, Comella CL, et al. The effects of valbenazine in participants with tardive dyskinesia: results of the 1-year KINECT 3 extension study. J Clin Psychiatry. 2017;78(9):1344–1350. PubMed CrossRef
  21. Caroff SN, Citrome L, Meyer J, et al. A modified Delphi consensus study of the screening, diagnosis, and treatment of tardive dyskinesia. J Clin Psychiatry. 2020;81(2):19cs12983. PubMed CrossRef

 

This CME activity is expired. For more CME activities, visit CMEInstitute.com.
Find more articles on this and other psychiatry and CNS topics:
The Journal of Clinical Psychiatry
The Primary Care Companion for CNS Disorders

 

CME Background Information

Overview

Experts offer a review of the fundamentals of diagnosing and treating tardive dyskinesia.

Target Audience

  • Psychiatrists
  • Neurologists
  • Primary Care Clinicians
  • Psychiatric Nurse Practitioners
  • Psychiatric Physician Assistants

Learning Objectives

Select appropriate treatment for patients diagnosed with tardive dyskinesia

Support Statement

Supported by an educational grant from Neurocrine Biosciences, Inc.

Learning Objective

After completing this educational activity, you should be able to:

  • Select appropriate treatment for patients diagnosed with tardive dyskinesia

Release, Review, and Expiration Dates

This brief report activity was published in August 2021 and is eligible for AMA PRA Category 1 Credit™ through August 31, 2023. The latest review of this material was July 2021.

Statement of Need and Purpose

TD must be detected early to minimize the risk of the movements becoming permanent. But many clinicians are unable to identify risk factors for this condition, are unfamiliar with diagnostic criteria, and do not regularly assess patients for TD. Clinicians, therefore, need education on the risk factors that should alert them to monitor certain patients especially closely for TD and strategies to assess all patients being treated with dopamine-blocking agents and provide an accurate diagnosis. In addition, while treatment for TD is available, with evidence-based recommendations and research on long-term safety and efficacy, clinicians may be slow to implement treatment strategies due to underestimation of the social and occupational impact that TD has on patients’ lives and uncertainty about what to do.

Faculty Affiliation

Christoph U. Correll, MD
Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York; and Charité Universitätsmedizin Berlin, Germany

Leslie Citrome, MD, MPH
New York Medical College, Valhalla

Financial Disclosure

The faculty for this CME activity and the CME Institute staff were asked to complete a statement regarding all relevant personal and financial relationships between themselves or their spouse/partner and any commercial interest. The Accreditation Council for Continuing Medical Education (ACCME) defines a commercial interest as any entity producing, marketing, re-selling, or distributing health care goods or services consumed by, or used on, patients. The ACCME defines relevant financial relationships as financial relationships in any amount occurring within the past 12 months that create a conflict of interest. The CME Institute has resolved any conflicts of interest that were identified. No member of the CME Institute staff reported any relevant personal financial relationships. Faculty financial disclosure is as follows:

Dr Correll has received grant/research support and honoraria from AbbVie, Acadia, Alkermes, Allergan, Angelini, Axsome, Gedeon Richter, Gerson Lehrman Group, Indivior, IntraCellular  Therapies, Janssen/J&J, Karuna, LB Pharma, Lundbeck, MedAvante-ProPhase, MedInCell, Medscape, Merck, Mitsubishi Tanabe Pharma, Mylan (Viatris), Neurocrine, Noven, Otsuka, Pfizer, Recordati, Rovi, Servier, Sumitomo Dainippon, Sunovion, Supernus, Takeda, and Teva; has received grant/research support from Janssen and Takeda; is a member of the speakers/advisory boards for Acadia, Alkermes, Allergan, Angelini, Axsome, Gedeon Richter, IntraCellular Therapies, Janssen/J&J, LB Pharma, Lundbeck, MedInCell, Merck, Mylan (Viatris), Neurocrine, Noven, Otsuka, Pfizer, Recordati, Rovi, Servier, Sumitomo Dainippon, Sunovion, Supernus, Takeda, and Teva; and is a stock shareholder of LB Pharma. Dr Citrome is a consultant for AbbVie, Acadia, Alkermes, Allergan, Avanir, Axsome, BioXcel, Cadent Therapeutics, Eisai, Impel, Intra-Cellular Therapies, Janssen, Karuna, Lundbeck, Luye, Merck, Neurocrine, Noven, Osmotica, Otsuka, Sage, Shire, Sunovion, Takeda, and Teva; is a member of the speakers/advisory boards for AbbVie, Acadia, Alkermes, Allergan, Eisai, Intra-Cellular Therapies, Janssen, Lundbeck, Merck, Neurocrine, Noven, Otsuka, Sage, Shire, Sunovion, Takeda, Teva; is a stock shareholder of Bristol-Myers, Squibb, Eli Lilly, J&J, Merck, and Pfizer; and has received royalties from Wiley, UpToDate, Springer Healthcare, and Elsevier.

Disclosure of Off-Label Usage

The chair has determined that, to the best of his knowledge, vitamin E, vitamin B6, ginkgo biloba, eicosapentaenoic acid, melatonin, clonazepam, amantadine, donepezil, branched-chain amino acids, reserpine, and tetrabenazine are not approved by the US Food and Drug Administration for the treatment of tardive dyskinesia.

Review Process

The faculty members agreed to provide a balanced and evidence-based presentation and discussed the topics and CME objectives during the planning sessions. The faculty’s submitted content was validated by CME Institute staff, and the activity was evaluated for accuracy, use of evidence, and fair balance by the Chair and a peer reviewer who is without conflict of interest.

Acknowledgment

This activity is derived from the teleconference series “Revisiting the Fundamentals of Diagnosing and Treating Tardive Dyskinesia,” which was held in March 2021 and supported by an educational grant from Neurocrine Biosciences, Inc. The opinions expressed herein are those of the faculty and do not necessarily reflect the opinions of the CME provider and publisher or the commercial supporter.

Accreditation Statement

The CME Institute of Physicians Postgraduate Press, Inc., is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians.

Credit Designation

The CME Institute of Physicians Postgraduate Press, Inc., designates this enduring material for a maximum of 1.00 AMA PRA Category 1 Credit™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

Note: The American Nurses Credentialing Center (ANCC) and the American Academy of Physician Assistants (AAPA) accept certificates of participation for educational activities certified for AMA PRA Category 1 Credit™ from organizations accredited by the ACCME.

 

Volume: 82

Quick Links:

References