Anda di halaman 1dari 6

Journal of the Neurological Sciences 389 (2018) 61–66

Contents lists available at ScienceDirect

Journal of the Neurological Sciences


journal homepage: www.elsevier.com/locate/jns

Review Article

Reprint of: Clinical management of tardive dyskinesia: Five steps to T


success☆,☆☆

Leslie Citrome
New York Medical College, Valhalla, NY, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Tardive dyskinesia (TD) has long been thought to be a generally irreversible consequence of the use of dopamine
Abnormal Involuntary Movement Scale receptor blocking agents. There is now an opportunity to successfully manage this condition with agents ap-
Antipsychotic proved by the US Food and Drug Administration. This is important because TD has not been eliminated with the
Deutetrabenazine use of second-generation antipsychotics, and the expansion of antipsychotics to treat conditions other than
Dopamine
schizophrenia has resulted in millions of additional individuals at risk for developing TD. Recognition of TD
Tardive dyskinesia
requires careful observation; a structured approach using the Abnormal Involuntary Movement Scale is en-
Valbenazine
VMAT2 couraged. Harm reduction can be achieved by using antipsychotics judiciously when possible and by paying
attention to other modifiable risk factors such as drug-induced parkinsonian symptoms and the use of antic-
holinergic medication. Once TD has emerged and is associated with dysfunction or distress, treatment with a
VMAT2 inhibitor such as deutetrabenazine or valbenazine is well supported by several controlled clinical trials.

1. Introduction by involuntary, repetitive, purposeless movements, typically of the


tongue, jaw, lips, face, trunk, upper extremities, and lower extremities,
Tardive dyskinesia (TD) has long been thought to be a generally and these movements can be associated with significant functional
irreversible consequence of the use of dopamine receptor blocking impairment and can be socially stigmatizing [3]. TD movements can
agents. Dopamine receptor blocking agents not only include anti- vary during the day and disappear during sleep.
psychotic medications used for the treatment of psychiatric disorders, Unfortunately, TD is not rare. Fortunately, there are therapeutic
but also antitussive agents such as promethazine and antiemetic med- options that have recently become available for the effective manage-
ications such as metoclopramide used for symptomatic gastro- ment of TD. This review will provide an overview of the management of
esophageal reflux in patients who fail to respond to conventional TD in five steps: 1) Recognition; 2) Assessment; 3) Harm reduction; 4)
therapy and for diabetic gastroparesis. Warnings regarding TD are in- Interventions; and 5) Follow-up.
cluded in US product labeling for all antipsychotics [1]. Within the
product label for metoclopramide, the warning for TD is boxed and 2. Step 1: recognition - admit the possibility that some of your
bolded and reads “…Metoclopramide therapy should be discontinued in patients have tardive dyskinesia
patients who develop signs or symptoms of tardive dyskinesia. Treat-
ment with metoclopramide for longer than 12 weeks should be avoided If you use antipsychotics in your clinical practice, chances are you
in all but rare cases where therapeutic benefit is thought to outweigh have patients with TD. The availability of second-generation anti-
the risk of developing tardive dyskinesia” [2]. In the treatment of psychotics has not eliminated TD. In a meta-analysis of 41 studies
psychotic disorders such as schizophrenia, treatment with anti- published since 2000, the overall mean TD prevalence among 11,493
psychotics is often indefinite, and advice to discontinue this interven- patients exposed to antipsychotics was 25.3%. Although a lower pre-
tion is usually not clinically feasible. valence for those receiving second-generation antipsychotics vs. first-
Although TD is sometimes quite challenging to tease apart from generation antipsychotics was noted (prevalence rates of 20.7% vs.
other movement disorders that may be comorbid, TD is distinguished 30.0%, respectively), and the lowest prevalence (7.2%) was observed

DOI of original article: http://dx.doi.org/10.1016/j.jns.2017.11.019



This article is part of the Special Issue Tardive Syndromes and Their Management edited by Robert A. Hauser and Daniel D. Truong.
☆☆
A publisher's error resulted in this article appearing in the wrong issue. The article is reprinted here for the reader's convenience and for the continuity of the special issue. For
citation purposes, please use; Journal of the Neurological Sciences, 383C, pp. 199-204.

11 Medical Park Drive, Suite 106, Pomona, NY 10970, USA.
E-mail address: citrome@cnsconsultant.com.

https://doi.org/10.1016/j.jns.2018.02.037
Received 13 November 2017; Accepted 15 November 2017
Available online 28 February 2018
0022-510X/ © 2017 Elsevier B.V. All rights reserved.
L. Citrome Journal of the Neurological Sciences 389 (2018) 61–66

among patients receiving second-generation antipsychotics who had During the entire AIMS examination the clinician remains vigilant
never been exposed to first-generation antipsychotics [4], the pre- for any dyskinetic movements, which then get rated using the first 7
valence rates are high enough that TD is a relatively common entity. items of the AIMS, representing 7 muscle groups or body areas, using a
Illustrating the above is a well-executed cohort study conducted at 5-point scale (0 = none, 1 = minimal/extreme normal, 2 = mild,
the Connecticut Mental Health Center that compared the incidence of 3 = moderate, 4 = severe). For each item the rating that is given
TD for second- vs. first-generation antipsychotics using methods like should reflect the most severe movement that was witnessed, regardless
those from a previous prospective cohort study at that site in the 1980s of any activation maneuvers. The total sum of these 7 items constitutes
[5]. Among the 352 initially TD-free outpatients who were examined the AIMS dyskinesia score. However, clinicians should exercise caution
for a new diagnosis of TD every 6 months for up to 4 years, the adjusted in interpreting this total score in the absence of knowing the individual
TD incidence rate-ratio for subjects treated with second-generation item scores. For example, when treating individual patients, an AIMS
antipsychotics alone vs. first-generation antipsychotics alone was 0.68, dyskinesia score of 7 has very different implications when it is based on
suggesting a small advantage for the newer agents. However, the 95% a score of 1 (i.e., minimal/extreme normal) across all 7 items vs. a score
confidence interval of 0.29 to 1.64 includes the possibility that there of 3 (moderate) on 2 items and a 1 on a third item. In the latter patient,
really is no difference. Importantly, the incidence rates did not differ more urgent efforts will need to be made to get those individual item
substantially from previous findings at this site in the 1980s when TD scores of moderate reduced to as low as possible.
was a prominent concern. TD rating scale scores were only slightly To avoid confusing oral movements due to the presence of gum or
lower among incident cases of TD appearing after recent second-gen- candy, these will need to be removed from the mouth. The patient will
eration antipsychotic exposure vs. recent first-generation antipsychotic also need to be asked about any dental problems – these may also lead
exposure. Of note, staff involved in this study were well trained to to oral movements that can be confusing to differentiate from TD. After
identify TD, and systematically looked for it. What would the yield be in completing the first 7 items of the AIMS, the next 3 items document the
your practice if you were to screen your own patients for TD on a overall severity of the abnormal movements (0–4; based on the highest
regular basis? single score in the first 7 items), the degree of incapacitation due to
It is likely that with the expansion of the use of antipsychotics be- abnormal movements (0–4; the patient will need to be asked to what
yond psychotic disorders the number of people at risk to develop TD extent any movements interfere with activities such as eating, drinking,
will continue to increase. Many persons with major depressive disorder speaking, breathing, dressing oneself, writing, working, leisure activ-
receive adjunctive antipsychotic medications in an effort to better ities, being with others, etc.), the patient's awareness (and distress
control their depressive symptoms. This increased use of dopamine level) of the abnormal movements (0–4, with 0 noting no awareness, 1
receptor blocking agents in routine practice makes admitting the pos- noting being aware with no distress, and 2–4 noting awareness and
sibility that your patients may have TD even more important. distress rating from mild to moderate to severe). It is not unusual for
persons with schizophrenia to have little insight into their dyskinetic
3. Step 2: assessment - understanding how to effectively use the movements; however, patients with mood disorders may be better able
Abnormal Involuntary Movement Scale and making a differential to articulate their distress. The last 2 items of the AIMS are yes/no
diagnosis questions regarding dentition status and the use of dentures.
The instructions for the AIMS also include an assessment of upper
Clinically assessing patients for TD requires careful observation. extremity rigidity by flexing and extending the patient's left and right
Although screening for TD can be done without the use of a specific arms, as well as observation of gait, but these are not rated.
scale, quantifying the severity of the TD will necessitate the use of an Nevertheless, findings from these actions may be helpful when de-
agreed-upon measurement. The 12-item Abnormal Involuntary termining if the patient has drug-induced parkinsonian side effects.
Movement Scale (AIMS), developed by the US National Institute of An AIMS score of at least 2 (i.e., mild) in 2 or more body regions or a
Mental Health, is a convenient way to structure a brief evaluation of a score of 3 (moderate) or 4 (severe) in at least one body region in a
patient for TD ([6,7]; see also Supplementary Appendix). Although patient with at least 3 months of cumulative antipsychotic drug ex-
other scales exist, the AIMS is arguably the best known, and has also posure equates to a probable diagnosis of TD according to the Schooler-
served as the primary outcome measure for recent studies of agents Kane criteria [12], provided that other causes of the abnormal move-
recently approved by the US Food and Drug Administration (FDA) for ments have been ruled out. An alternate (but similar) definition of TD is
the treatment of TD [8–11]. The AIMS alone is not sufficient to diagnose the Glazer-Morgenstern criteria [13], where the detection of TD cases is
TD, but can serve as a comprehensive screen and can be used to follow a based on two applications of the AIMS at the beginning and end of each
person's progress over time. visit; the criteria for diagnosing new persistent cases of TD at follow-up
When will a complete AIMS examination be required? If the patient visits are a total AIMS dyskinesia score of 3 or more on both applica-
or caregiver initially complains of abnormal movements or if the clin- tions at two successive visits and at least 1 body area score of 2 or more
ician observes new abnormal movements, a full evaluation is desirable. on both applications at the same two visits.
Since TD can vary over time and can worsen with emotional stress, The definition of TD contained in the American Psychiatric
repeat examinations can be helpful. The AIMS examination begins by Association Diagnostic and Statistical Manual of Mental Disorders, Fifth
observing the patient at rest, which initially can be done unobtrusively Edition, notes that symptoms may develop after a shorter period of
while the patient is in the waiting area. Once in the office or ex- medication use in older persons [14]. Moreover, if the abnormal
amination room it is ideal to have two identical hard, firm chairs with movements appear after discontinuation of an antipsychotic, or after a
no arms placed facing each other – one for the patient and one for the change or reduction in the dosage of the antipsychotic medication, the
examiner. In this manner the clinician can easily model what is being condition is called withdrawal-emergent dyskinesia. However, if the
asked of the patient: sitting with hands on knees, hands hanging un- withdrawal-emergent dyskinesia persists beyond 8 weeks, the patient
supported between the legs or over the knees, opening the mouth, would be considered as having TD.
protruding the tongue, tapping the thumb with each finger, and then In the American Psychiatric Association's practice guideline for the
standing up, extending both arms in front with palms down, and lastly, treatment of patients with schizophrenia [15], patients taking an anti-
walking. The thumb-finger tapping, arm extension and walking are psychotic medication should be regularly monitored for movement
“activation” maneuvers used to elicit abnormal movements in other disorders. The recommended frequency for the assessment of TD is
body areas. An additional activation maneuver that can be used is a every 6 months for persons taking a first-generation antipsychotic or
cognitive task such as asking the patient to count backwards from 100 every 12 months for those receiving second-generation antipsychotics.
or to recite the months of the year in reverse order. However, if patients have additional risk factors for TD, then the

62
L. Citrome Journal of the Neurological Sciences 389 (2018) 61–66

recommended monitoring is more frequent: every 3 months for first- Because age is a strong risk factor for TD, if antipsychotics are to be
generation antipsychotics and every 6 months for second-generation used in the elderly, low but effective doses of single drugs should be
antipsychotics. Although the AIMS is mentioned as a possible means of employed together with regular and specific assessments for early TD.
assessing TD, the complete AIMS examination was not explicitly noted
as being the only means of clinically assessing TD. It may be possible to 5. Step 4: interventions
conduct an “abbreviated” AIMS consisting of examination of the face
and arms, and with activation maneuvers; this abbreviated version of Up until recently there were few options to manage TD, none were
the AIMS is intended for screening purposes and has not been system- approved by regulatory authorities, and this lack of effective inter-
atically compared with the utility of doing the complete examination. ventions led to substantial therapeutic nihilism [3,18,21]. A common
The most common obstacle in making a correct diagnosis of TD is management strategy for TD would be to gradually switch a patient
confusion with drug-induced parkinsonism. Parkinsonian tremor is from a first-generation antipsychotic to a second-generation anti-
rhythmic and faster (3–6 Hz) compared to the slow arrhythmic move- psychotic (other than clozapine) and discontinue any anticholinergic
ments typical of TD [7]. Drug-induced parkinsonism typically occurs medications. If the TD did not improve, a switch to another second-
soon after an antipsychotic is initiated or when the dose is raised, and is generation antipsychotic (other than clozapine) was attempted, and if
often lessened when the antipsychotic dose is reduced [16]. TD occurs that failed, a switch to clozapine was recommended. A last choice
later (“tardive” means delayed) and is often made worse (at least would be to consider so-called “suppression therapy” where the TD
temporarily) when the antipsychotic dose is reduced, and the dyskinetic would be masked by adding a potent first-generation antipsychotic
movements appear improved (“masked”) when the antipsychotic dose medication; this was generally only contemplated for severe TD that
is increased; this pattern of apparent worsening or improvement of TD substantially impaired function or was potentially life-threatening (as
with a decrease or increase of antipsychotic dose, respectively, is the when TD can interfere with breathing because of involvement of the
reverse of what is expected with drug-induced parkinsonism. Moreover, diaphragm muscle). A nutritional intervention was also commercia-
anticholinergic medications, such as benztropine, will treat drug-in- lized: a combination of the branched-chain amino acids L-leucine, L-
duced parkinsonian tremor but may make TD worse [17]. valine, and L-isoleucine. This intervention was approved by the FDA as
Other tardive syndromes may be present, for example tardive dys- a medical food for the dietary management of TD in male patients based
tonia and tardive akathisia [18]. Conditions that may resemble TD in- on a double-blind study where one-third of the treatment group had a
clude spontaneous dyskinesias not associated with antipsychotic ex- reduction of 60% or more in dyskinetic movements, compared with
posure, as well as conditions such as Huntington disease and Wilson none of the subjects receiving placebo [22]; the branded product is no
disease that are generally accompanied by a variety of other signs and longer available but compounding pharmacies can make it with the
symptoms. Some drugs or substances can also be associated with ab- same ratio of ingredients tested in the clinical trial.
normal movements; these include stimulants or excessive caffeine use. In 2013, guidelines from the American Academy of Neurology [23]
opined that data were insufficient as to whether stopping the medica-
4. Step 3: harm reduction – reducing risk tion associated with TD would suppress symptoms and the strategy of
switching from a first- to second-generation antipsychotic was also
Ideally it is best to prevent TD. There are many different risk factors determined to have insufficient evidence. Other strategies that have
for TD. Older age and antipsychotic medication exposure (dose/dura- come and gone because of insufficient data to support (or refute) their
tion) are well established risk factors and other risk factors include use include acetazolamide, bromocriptine, thiamine, baclofen, vitamin
female sex, African American ethnicity, pre-existing mood disorder, E, vitamin B6, selegiline, melatonin, nifedipine, levetiracetam, bus-
cognitive disturbance, alcohol or substance abuse, treatment with first- pirone, yi-gan san, biperiden discontinuation, botulinum toxin type A,
generation antipsychotics, use of lithium or antiparkinsonian agents, electroconvulsive therapy, α-methyldopa, reserpine, and pallidal deep
early occurrence of drug-induced parkinsonian symptoms, diabetes, brain stimulation. The guidelines explicitly did not recommend dil-
and HIV positivity [19]. tiazem, galantamine, or eicosapentaenoic acid.
Avoiding the use of dopamine receptor blocking agents is not fea- According to the American Academy of Neurology guidelines, 4
sible when managing patients with schizophrenia; for schizophrenia, interventions could be recommended, based on evidence of varying
antipsychotic medication is currently the only foundational treatment quality: clonazepam, Ginkgo biloba, amantadine, and tetrabenazine.
available. Antipsychotic medication may also be difficult to avoid in Clonazepam is a benzodiazepine with indirect GABA agonist activity
persons with bipolar disorder who cannot be stabilized or maintained and has been explored as a treatment for TD since the 1970s [24].
on mood stabilizers alone; again, attention to antipsychotic dose is re- Unfortunately, although in one study clonazepam reduced dyskinesia
quired. For adjunctive use in major depressive disorder, antipsychotic scores by approximately 37% (using the Maryland Psychiatric Research
medications have been studied principally as an acute treatment and Center Movement Disorder Scale), tolerance developed in patients who
thus there are few data available regarding long-term exposure in the received clonazepam in the long-term extension after 5 to 8 months of
maintenance phase of treatment. Other uses of antipsychotic medica- use [25]. Gingko biloba, an antioxidant, appeared effective in reducing
tions such as for sleep, post-traumatic stress disorder, or anxiety, are TD as measured by the AIMS in a meta-analysis of three 12-week,
currently “off-label,” and thus risk for TD makes this choice of treat- randomized trials from China [26]. Amantadine, a blocker of N-methyl-
ment for these conditions problematic, especially in the face of alter- D-aspartate glutamate receptors, has a modest effect in reducing TD
native interventions. (reduction of AIMS dyskinesia score by 15–22%) [27,28]; an extended-
Minimizing drug-induced parkinsonian symptoms by selecting release formulation of amantadine recently received approval for the
agents with lower risk for this problem may be helpful in reducing the treatment of dyskinesia in patients with Parkinson's disease receiving
risk of eventually developing TD. This is a rationale for using second- levodopa-based therapy, with or without concomitant dopaminergic
generation antipsychotics, and for switching antipsychotic medications medications [29]. Tetrabenazine was originally developed in the 1950s
should drug-induced parkinsonian symptoms occur. The use of antic- and was approved in the US in 2008 as an orphan drug for the treat-
holinergics such as benztropine to address drug-induced parkinsonian ment of choreiform movements associated with Huntington disease
symptoms can place the patient at additional risk for developing TD, [30]. Tetrabenazine is a reversible and specific inhibitor of vesicular
and can worsen existing TD [17]. Moreover, anticholinergic medication monoamine transporter 2 (VMAT2), a protein in the brain that trans-
has a deleterious effect on cognition and can further disadvantage a ports monoamine neurotransmitters such as dopamine, norepinephrine,
person with schizophrenia who already is struggling with cognitive serotonin and histamine into vesicles for release into the synapse when
impairment [20]. the neuron fires [31,32]. With less dopamine in the vesicle, less

63
L. Citrome Journal of the Neurological Sciences 389 (2018) 61–66

Table 1
Deutetrabenazine and valbenazine for tardive dyskinesia, information from product labeling [8,9] and summary data from systematic reviews [10,11].

Deutetrabenazine Valbenazine

Brand name Austedo Ingrezza


Date approved by US FDA for TD August 2017 April 2017
Dose/formulation Tablets: 6 mg, 9 mg, 12 mg Capsules: 40 mg, 80 mg
Other indications Chorea associated with Huntington's disease None
Design rationale Deuteration results in slower drug metabolism Parent drug of [+]-α-HTBZ; no β-HTBZ
Metabolites Active deuterated dihydro metabolites (HTBZ): α-HTBZ and β- Active metabolite: ([+]-α-HTBZ)
HTBZ
Half-life Total (α + β)-HTBZ from deutetrabenazine: 9–10 h Valbenazine and [+]-α-HTBZ: 15–22 h
Boxed bolded warnings relevant to TD None None
Contraindications relevant to TD Hepatic impairment; taking reserpine, MAOIs, tetrabenazine, None
or valbenazine
Warnings and precaution contained in QT prolongation; neuroleptic malignant syndrome; akathisia, Somnolence; QT prolongation
Highlights of Prescribing Information agitation, restlessness, and parkinsonism (latter not applicable
to TD); sedation/somnolence
Dosing recommendations Initial dose 12 mg/d, target dose 12–48 mg/d, administer BID Initial dose 40 mg/d, target dose 80 mg, administer once daily with
with food; titrate at weekly intervals by 6 mg/d based on or without food; titrate to 80 mg/d after one week on 40 mg/d
reduction of TD and tolerability
CYP2D6 poor metabolizers Maximum recommended dosage in poor CYP2D6 metabolizers Consider dose reduction based on tolerability
is 36 mg/d
Drug-drug interactions Strong CYP2D6 inhibitors: maximum recommended dose is MAOIs: avoid; strong CYP3A4 inducers: not recommended; strong
36 mg/d; alcohol or other sedating drugs may have additive CYP3A4 inhibitors: reduce dose to 40 mg; strong CYP2D6
sedation and somnolence inhibitors: consider dose reduction based on tolerability
Hepatic impairment Contraindicated Recommended dose for patients with moderate or severe hepatic
impairment is 40 mg/d
Renal impairment No clinical studies have been conducted to assess the effect of No dosage adjustment is necessary for patients with mild to
renal impairment moderate renal impairment; use is not recommended in patients
with severe renal impairment
QT prolongation recommendations For patients at risk for QT prolongation, assess the QT interval For patients at increased risk of a prolonged QT interval, assess the
before and after increasing the total dosage above 24 mg/d QT interval before increasing the dosage
Most common AEsa and rate vs. placebo Nasopharyngitis (4% vs. 2%), insomnia (4% vs. 1%) Somnolence (10.9% vs. 4.2%)
Responderb rates, pooled 30.0% vs. 14.8% 36.5% vs. 12.4%
NNT (95% CI) vs. placebo 7 (4–18) 5 (3–7)
CGI responderc rates, pooled 46.9% vs. 33.0% 40.4% vs. 18.6%
NNT (95% CI) vs. placebo 8 (4–45) 5 (4–9)
Discontinuation rates due to an AE, 3.6% vs. 3.1% 2.6% vs. 1.6%
pooled
NNH (95% CI) vs. placebo 189 (not significant) 76 (not significant)

AE – adverse event; CI – confidence interval; MAOI – monoamine oxidase inhibitor; NNH – number needed to harm; NNT – number needed to treat; TD – tardive dyskinesia.
a
As identified in product labeling in Highlights of Prescribing Information; for deutetrabenazine definition is 4% and greater than placebo; for valbenazine definition is ≥ 5% and
twice the rate of placebo.
b
Response defined as a ≥ 50% decrease in the AIMS dyskinesia score (sum of items 1–7) from baseline to endpoint (12 weeks in the deutetrabenazine studies and 6 weeks in the
valbenazine studies); for deutetrabenazine the fixed dose study arm of 12 mg/d was excluded was excluded from the calculations.
c
CGI response defined as a score of 1 (very much improved) or 2 (much improved) at endpoint (12 weeks in the deutetrabenazine studies and 6 weeks in the valbenazine studies); for
deutetrabenazine the fixed-dose study arm of 12 mg/d was excluded from the calculations.

dopamine is released into the synapse, and there is thus a lower like- low affinity) and is metabolized slowly to [+]-α-dihydrotetrabenazine
lihood of triggering an abnormal movement in a system made “hy- ([+]-α-HTBZ), a potent and selective VMAT2 antagonist. [+]-α-HTBZ
persensitive” due to chronic post-synaptic dopamine receptor blockade is one of the metabolites of tetrabenazine and thus valbenazine and
and subsequent upregulation of both number and function of post-sy- tetrabenazine have this metabolite in common. The half-life of valbe-
naptic dopamine receptors. There are a limited number of studies of nazine and [+]-α-HTBZ approaches 1 day and thus daily dosing is re-
tetrabenazine for TD, the first one published in 1972 [33], but overall commended (with or without food). Genotyping is not required but
these studies have demonstrated antidyskinetic effects [23,33–35]. The dosing adjustments may be needed in the presence of CYP 2D6 or CYP
short half-life of tetrabenazine, sensitivity to CYP 2D6 metabolism 3A4 modulators. Efficacy is optimal at the 80 mg/d dose and is reached
(genotyping is required for tetrabenazine doses above 50 mg/d), and its after one week on 40 mg/d. In a placebo-controlled, double-blind,
relatively poor tolerability profile have led to drug development efforts flexible dose (25–75 mg/d) study of 6 weeks in duration, the least-
to improve upon tetrabenazine. Two different approaches have been squares mean change in AIMS dyskinesia scores from baseline to end-
used and have resulted in two new VMAT2 inhibitors that are FDA- point was significantly greater with valbenazine (− 2.6) than placebo
approved for the treatment of TD in adults: deutetrabenazine [9,11] (− 0.2; P = 0.0005) and no patients discontinued because of an ad-
and valbenazine [8,10]. Table 1 provides an overview of both products. verse event [36]. After titration, 76% of those taking valbenazine
Both deutetrabenazine and valbenazine were approved on the basis of reached the 75 mg/d dose. In a 6-week, double-blind, fixed-dose study
placebo-controlled randomized clinical trials whose primary outcome of valbenazine (40 and 80 mg/d) vs. placebo, the least-squares mean
was change on the AIMS dyskinesia score (total score on items 1–7) change from baseline to endpoint was significantly greater for valbe-
from baseline to end point (Week 6 for valbenazine studies and Week nazine 80 mg/d (− 3.2) vs. placebo (− 0.1; P ≤ 0.001), as was also
12 for deutetrabenazine studies), as assessed by blinded video raters observed for the 40 mg/d dose (− 1.9; P = 0.0021) [37]. Dis-
who were movement disorder specialists otherwise not involved in the continuation because of an adverse event occurred in 3.8% of those
study. taking valbenazine (both doses pooled) versus 2.6% of those taking
The first medication to be approved was valbenazine in April 2017 placebo.
[8]. Valbenazine is itself a selective VMAT2 inhibitor (with relatively The second medication to be approved for TD was deutetrabenazine

64
L. Citrome Journal of the Neurological Sciences 389 (2018) 61–66

in August 2017 [9]. Deutetrabenazine is related to tetrabenazine in that was similar to that found for the NNT vs. placebo for Clinical Global
deuterium is substituted for hydrogen at the key sites of primary me- Impression (CGI) response (very much improved or much improved): 8
tabolism, altering the pharmacokinetics of drug metabolism by slowing [11]. The analogous results from the pooled valbenazine trials were
it down. Thus, the active metabolites of deutetrabenazine, deuterated similar: NNT for AIMS response was 5 and NNT for CGI response was
α-HTBZ and β-HTBZ, have longer half-lives compared to the analogous also 5 [10].
non-deuterated active metabolites of tetrabenazine. Deuterium is a Although each agent can exhibit adverse effects, the rates for spe-
stable, nonradioactive, nontoxic, naturally occurring isotope of hy- cific adverse effects in the clinical trials were generally low, with
drogen and has the same size and shape as a hydrogen atom, differing number needed to harm (NNH) values > 10, denoting an intervention
only in forming stronger chemical bonds. Genotyping is not required that would be generally expected to be tolerable on that domain [41].
but dosing adjustments may be needed in the presence of CYP 2D6 The adverse event with the largest drug-placebo difference for deute-
modulators. Twice daily dosing with food is recommended. In contrast trabenazine was insomnia (4% vs. 1%), with a resultant NNH of 34
to the fixed target dose for valbenazine (80 mg/d), deutetrabenazine [11]. That for valbenazine was somnolence (11% vs. 4%), with a re-
dose is determined individually for each patient based on reduction of sultant NNH of 15 [10]. Overall tolerability can be quantified by
TD and tolerability. The recommended starting dose of deute- comparing rates of discontinuation because of an adverse event. A total
trabenazine for TD is 6 mg twice daily, and can be increased at weekly of 3.6% of patients receiving deutetrabenazine (all doses pooled) dis-
intervals in increments of 6 mg/d to a maximum recommended daily continued because of an adverse event, compared with 3.1% for pla-
dosage of 48 mg. In a placebo-controlled, double-blind, flexible dose cebo, resulting in a NNH of 189 [11]. A total of 2.9% of patients re-
(12–48 mg/d) study of 12 weeks in duration, the least-squares mean ceiving valbenazine (all doses pooled) discontinued because of an
change in AIMS dyskinesia scores from baseline to endpoint was sig- adverse event, compared with 1.6% for placebo, resulting in a NNH of
nificantly greater with deutetrabenazine (−3.0) than placebo (− 1.6; 76 [10]. Thus both deutetrabenazine and valbenazine are far more
P = 0.019) and more patients discontinued because of an adverse event likely to result in a response (≥50% reduction in the AIMS dyskinesia
with placebo (3.4%) than with deutetrabenazine (1.7%) [38]. After score) than a discontinuation because of an adverse event. For deute-
titration, the mean total daily dose was 38.8 mg/d. In a 12-week, trabenazine or valbenazine for TD there does not appear to be an ex-
double-blind, fixed-dose study of deutetrabenazine (12, 24 and 36 mg/ acerbation of underlying psychopathology, nor are there any clinically
d) vs. placebo, the least-squares mean change from baseline to endpoint relevant signals regarding depression or suicidality. Warnings regarding
was significantly greater for deutetrabenazine 36 mg/d (− 3.3) vs. QT prolongation do not preclude the use of concomitant agents that
placebo (−1.4; P = 0.001), as was also observed for the 24 mg/d dose may also prolong QT (a common occurrence as many antipsychotics or
(− 3.2; P = 0.003), but not for the 12 mg/d dose (− 2.1; P = 0.217) antidepressants are associated with prolongation of the QT interval);
[39]. The percentages of patients discontinuing because of an adverse however, ECG monitoring may be required in at-risk situations when
event were 4% for deutetrabenazine (all doses pooled) versus 3% for undergoing dose titration (see Table 1).
placebo. How long should VMAT2 inhibitors be used? At this point in time,
Clinically relevant reductions in dyskinetic movements can be ex- indefinite duration of therapy is recommended. As evidenced in the
pected in persons receiving either deutetrabenazine or valbenazine. long-term data available from valbenazine, dyskinetic movements re-
Note that participants who entered the studies on antipsychotic medi- turned within 4 weeks after discontinuation [8].
cations continued to receive them at the same dose. Using the outcome Both deutetrabenazine and valbenazine require prescribing through
of a 50% or greater reduction in baseline AIMS dyskinesia score as a specialty pharmacies to assist in navigating the approval process for
desired response, number needed to treat (NNT) can be calculated to these relatively expensive medications and to also minimize potential
compare the active drug vs. placebo. NNT describes the number of patient co-pays.
patients that needed to be randomized to active drug rather than pla-
cebo before expecting to encounter one additional responder [40]. NNT 6. Step 5: follow-up - check your work
values < 10 generally denote interventions that are found to be
clinically useful [41]. Fig. 1 illustrates the NNT for AIMS response from When treatment is initiated, a baseline assessment should be ob-
the two pivotal fixed-dosed studies of deutetrabenazine [39] and val- tained and the AIMS examination is recommended for this purpose.
benazine [37]. The NNT values for deutetrabenazine 24 and 36 mg/d Advantages to using the AIMS include its ubiquity both in the clinic and
(pooled) and valbenazine 40 and 80 mg/d (pooled) are the same: 5. The in drug development. Follow-up assessments should be done on a reg-
NNT for AIMS response vs. placebo when pooling both of the available ular basis, and the AIMS in this case can facilitate communication
deutetrabenazine studies (excluding the dose of 12 mg/d) was 7, and among providers. Lack of documented improvement can trigger addi-
tional enquiries regarding adherence or unidentified drug-drug inter-
actions, as well as inform decisions on dose titration or changing the
intervention. Measurement-based care has the potential to improve
outcomes, as has been demonstrated in the treatment of major de-
pressive disorder [42].
Measuring the AIMS dyskinesia items alone is inadequate. Follow-
up questions regarding functional impairments attributable to TD need
to be asked (e.g., interference with activities such as eating, drinking,
speaking, breathing, dressing oneself, writing, working, leisure activ-
ities, being with others). Fortunately, the AIMS contains items that
remind the clinician to evaluate impact on function and to determine
the degree of associated distress.

7. Summary

In the past, suggested treatments for TD have been largely in-


Fig. 1. ≥ 50% reduction in AIMS dyskinesia score from baseline to endpoint, NNT vs. effective or limited. There are now 2 FDA-approved treatments for TD:
placebo and 95% CIs, for the Phase III fixed-dose studies of deutetrabenazine (12 weeks) deutetrabenazine and valbenazine. Both are efficacious and tolerable.
and valbenazine (6 weeks); reproduced with permission from [11].
They differ in terms of labeled instructions for frequency of

65
L. Citrome Journal of the Neurological Sciences 389 (2018) 61–66

administration (twice daily for deutetrabenazine vs. once daily for of patients with schizophrenia, second edition, Am. J. Psychiatry 161 (2 Suppl)
valbenazine), titration requirements (dose to efficacy/tolerability for (2004) 1–56.
[16] D.C. Mamo, R.A. Sweet, M.S. Keshavan, Managing antipsychotic-induced parkin-
deutetrabenazine vs. titrate to target dose of 80 mg/d for valbenazine), sonism, Drug Saf. 20 (3) (1999) 269–275.
need for food (administer deutetrabenazine with food), drug-drug in- [17] Merck, COGENTIN (Benztropine Mesylate Injection). Prescribing Information,
teractions (consider CYP 2D6 modulators for deutetrabenazine vs. both Available at https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/
012015s026lbl.pdf, (April 2013) (Accessed 18 October 2017).
CYP 2D6 and CYP 3A4 for valbenazine), contraindications (hepatic [18] L. Citrome, R. Dufresne, J.M. Dyrud, Tardive dyskinesia: minimizing risk and im-
impairment for deutetrabenazine), and minor differences in adverse proving outcomes in schizophrenia and other disorders, Am. J. Manag. Care 13
event profile. However, the clinical usefulness of these treatments is (Suppl) (2007) 1–12.
[19] C.U. Correll, J.M. Kane, L.L. Citrome, Epidemiology, prevention, and assessment of
rendered moot if TD goes unrecognized. Clinicians are encouraged to tardive dyskinesia and advances in treatment, J. Clin. Psychiatry. 78 (8) (2017)
assume that TD exists in their practice, that there is a need for harm 1136–1147.
reduction, and to use assessments such as the AIMS for the evaluation of [20] S. Vinogradov, M. Fisher, H. Warm, C. Holland, M.A. Kirshner, B.G. Pollock, The
cognitive cost of anticholinergic burden: decreased response to cognitive training in
TD and ongoing monitoring of TD treatment.
schizophrenia, Am. J. Psychiatry 166 (9) (2009) 1055–1062.
[21] H.C. Margolese, G. Chouinard, T.T. Kolivakis, L. Beauclair, R. Miller, L. Annable,
Disclosures Tardive dyskinesia in the era of typical and atypical antipsychotics. Part 2: in-
cidence and management strategies in patients with schizophrenia, Can. J.
Psychiatr. 50 (11) (2005) 703–714.
This research did not receive any specific grant from funding [22] M.A. Richardson, M.L. Bevans, L.L. Read, et al., Efficacy of the branched-chain
agencies in the public, commercial, or not-for-profit sectors. In the past amino acids in the treatment of tardive dyskinesia in men, Am. J. Psychiatry 160 (6)
48 months Leslie Citrome has engaged in collaborative research with, (2003) 1117–1124.
[23] R. Bhidayasiri, S. Fahn, W.J. Weiner, et al., Evidence-based guideline: treatment of
or received consulting or speaking fees, from: Acadia, Alexza, Alkermes, tardive syndromes: report of the Guideline Development Subcommittee of the
Allergan, AstraZeneca, Avanir, Boehringer Ingelheim, Bristol-Myers American Academy of Neurology, Neurology 81 (5) (2013) 463–469.
Squibb, Eli Lilly, Forum, Genentech, Intra-Cellular Therapeutics, [24] P.P. Lerner, C. Miodownik, V. Lerner, Tardive dyskinesia (syndrome): current
concept and modern approaches to its management, Psychiatry Clin. Neurosci. 69
Janssen, Jazz, Lundbeck, Merck, Medivation, Mylan, Neurocrine, (6) (2015) 321–334.
Novartis, Noven, Otsuka, Pfizer, Reckitt Benckiser, Reviva, Shire, [25] G.K. Thaker, J.A. Nguyen, M.E. Strauss, R. Jacobson, B.A. Kaup, C.A. Tamminga,
Sunovion, Takeda, Teva, Valeant, Vanda. Clonazepam treatment of tardive dyskinesia: a practical GABAmimetic strategy,
Am. J. Psychiatry 147 (4) (1990) 445–451.
[26] W. Zheng, Y.-Q. Xiang, C.H. Ng, G.S. Ungvari, H.F. Chiu, Y.T. Xiang, Extract of
Appendix A. Supplementary data ginkgo biloba for tardive dyskinesia: meta-analysis of randomized controlled trials,
Pharmacopsychiatry 49 (3) (2016) 107–111.
[27] S. Pappa, S. Tsouli, G. Apostolou, V. Mavreas, S. Konitsiotis, Effects of amantadine
Supplementary data to this article can be found online at https://
on tardive dyskinesia: a randomized, double-blind, placebo-controlled study, Clin.
doi.org/10.1016/j.jns.2017.11.019. Neuropharmacol. 33 (6) (2010) 271–275.
[28] S. Angus, J. Sugars, R. Boltezar, S. Koskewich, N.M. Schneider, A controlled trial of
References amantadine hydrochloride and neuroleptics in the treatment of tardive dyskinesia,
J. Clin. Psychopharmacol. 17 (2) (1997) 88–91.
[29] Adamas Pharma, GOCOVRI (Amantadine) Extended Release Capsules, for Oral Use.
[1] L. Citrome, H.A. Nasrallah, On-label on the table: what the package insert informs Prescribing Information, Available at https://www.accessdata.fda.gov/drugsatfda_
us about the tolerability profile of oral atypical antipsychotics, and what it does not, docs/label/2017/208944lbl.pdf, (August 2017) (Accessed 16 October 2017).
Expert. Opin. Pharmacother. 13 (11) (2012) 1599–1613. [30] Lundbeck, XENAZINE (Tetrabenazine) Tablets, for Oral Use. Prescribing
[2] A.N.I. Pharmaceuticals, REGLAN (Metoclopramide) Tablets, for Oral Use. Information, Available at https://www.accessdata.fda.gov/drugsatfda_docs/label/
Prescribing Information, Available at https://www.accessdata.fda.gov/drugsatfda_ 2015/021894s010lbl.pdf, (June 2015) (Accessed 19 October 2017).
docs/label/2017/017854s062lbl.pdf, (August 2017) (Accessed 18 October 2017). [31] D.R. Guay, Tetrabenazine, a monoamine-depleting drug used in the treatment of
[3] H.C. Margolese, G. Chouinard, T.T. Kolivakis, L. Beauclair, R. Miller, Tardive dys- hyperkinetic movement disorders, Am. J. Geriatr. Pharmacother. 8 (4) (2010)
kinesia in the era of typical and atypical antipsychotics. Part 1: pathophysiology 331–373.
and mechanisms of induction, Can. J. Psychiatr. 50 (9) (2005) 541–547. [32] J.J. Chen, W.G. Ondo, K. Dashtipour, D.M. Swope, Tetrabenazine for the treatment
[4] M. Carbon, C.H. Hsieh, J.M. Kane, C.U. Correll, Tardive dyskinesia prevalence in of hyperkinetic movement disorders: a review of the literature, Clin. Ther. 34 (7)
the period of second-generation antipsychotic use: a meta-analysis, J. Clin. (2012) 1487–1504.
Psychiatry 78 (2017) e264–e278. [33] H. Kazamatsuri, C. Chien, J.O. Cole, Treatment of tardive dyskinesia, I: clinical
[5] S.W. Woods, H. Morgenstern, J.R. Saksa, et al., Incidence of tardive dyskinesia with efficacy of a dopamine-depleting agent, tetrabenazine, Arch. Gen. Psychiatry 27 (1)
atypical versus conventional antipsychotic medications: a prospective cohort study, (1972) 95–99.
J. Clin. Psychiatry 71 (4) (2010) 463–474. [34] H. Kazamatsuri, C.P. Chien, J.O. Cole, Long-term treatment of tardive dyskinesia
[6] W. Guy, ECDEU Assessment Manual for Psychopharmacology, US Department of with haloperidol and tetrabenazine, Am. J. Psychiatry 130 (4) (1973) 479–483.
Health, Education, and Welfare, Washington, DC, 1976. [35] W.G. Ondo, P.A. Hanna, J. Jankovic, Tetrabenazine treatment for tardive dyski-
[7] M.R. Munetz, S. Benjamin, How to examine patients using the Abnormal nesia: assessment by randomized videotape protocol, Am. J. Psychiatry 156 (8)
Involuntary Movement Scale, Hosp. Commun. Psychiatry 39 (11) (1988) (1999) 1279–1281.
1172–1177. [36] C.F. O'Brien, R. Jimenez, R.A. Hauser, et al., NBI-98854, a selective monoamine
[8] Neurocrine Biosciences, INGREZZA (valbenazine) Capsules, for Oral Use. transport inhibitor for the treatment of tardive dyskinesia: a randomized, double-
Prescribing Information, Available at https://ingrezza.com/HCP/PI, (October blind, placebo-controlled study, Mov. Disord. 30 (12) (2015) 1681–1687.
2017) (Accessed 18 October 2017). [37] R.A. Hauser, S.A. Factor, S.R. Marder, et al., KINECT 3: a phase 3 randomized,
[9] Teva Pharmaceuticals, AUSTEDO (Deutetrabenazine) Tablets, for Oral Use. double-blind, placebo-controlled trial of valbenazine for tardive dyskinesia, Am. J.
Prescribing information, Available at https://www.accessdata.fda.gov/drugsatfda_ Psychiatry 174 (5) (2017) 476–484.
docs/label/2017/209885lbl.pdf, (August 2017) (Accessed 18 October 2017). [38] H.H. Fernandez, S.A. Factor, R.A. Hauser, et al., Randomized controlled trial of
[10] L. Citrome, Valbenazine for tardive dyskinesia: a systematic review of the efficacy deutetrabenazine for tardive dyskinesia: the ARM-TD study, Neurology 88 (21)
and safety profile for this newly approved novel medication - what is the number (2017) 2003–2010.
needed to treat, number needed to harm and likelihood to be helped or harmed? Int. [39] K.E. Anderson, D. Stamler, M.D. Davis, et al., Deutetrabenazine for treatment of
J. Clin. Pract. 71 (7) (2017) e12964. involuntary movements in patients with tardive dyskinesia (AIM-TD): a double-
[11] L. Citrome, Deutetrabenazine for tardive dyskinesia: a systematic review of the blind, randomised, placebo-controlled, phase 3 trial, Lancet Psychiatry 4 (8) (2017)
efficacy and safety profile for this newly approved novel medication - what is the 595–604.
number needed to treat, number needed to harm and likelihood to be helped or [40] L. Citrome, Compelling or irrelevant? Using number needed to treat can help de-
harmed? Int. J. Clin. Pract. 71 (11) (2017) e13030. cide, Acta Psychiatr. Scand. 117 (6) (2008) 412–419.
[12] N.R. Schooler, J.M. Kane, Research diagnoses for tardive dyskinesia, Arch. Gen. [41] L. Citrome, T.A. Ketter, When does a difference make a difference? Interpretation of
Psychiatry 39 (4) (1982) 486–487. number needed to treat, number needed to harm, and likelihood to be helped or
[13] H. Morgenstern, W.M. Glazer, Identifying risk factors for tardive dyskinesia among harmed, Int. J. Clin. Pract. 67 (5) (2013) 407–411.
long-term outpatients maintained with neuroleptic medications. Results of the Yale [42] A.S. Yeung, Y. Jing, S.K. Brenneman, et al., Clinical Outcomes in Measurement-
Tardive Dyskinesia Study, Arch. Gen. Psychiatry 50 (9) (1993) 723–733. based Treatment (Comet): a trial of depression monitoring and feedback to primary
[14] American Psychiatric Association, Diagnostic and Statistical Manual of Mental care physicians, Depress Anxiety 29 (10) (2012) 865–873.
Disorders, Fifth edition, American Psychiatric Publishing, May 2013.
[15] A.F. Lehman, J.A. Lieberman, L.B. Dixon, et al., Practice guideline for the treatment

66

Anda mungkin juga menyukai