Anda di halaman 1dari 17

Drugs

https://doi.org/10.1007/s40265-018-0874-x

THERAPY IN PRACTICE

Treatment of Tardive Dyskinesia: A General Overview with Focus


on the Vesicular Monoamine Transporter 2 Inhibitors
Nicki Niemann1 • Joseph Jankovic1

Ó Springer International Publishing AG, part of Springer Nature 2018

Abstract Tardive dyskinesia (TD) encompasses the spec- have compared the various VMAT2 inhibitors. One of the
trum of iatrogenic hyperkinetic movement disorders follow- major advantages of VMAT2 inhibitors over DRBAs, which
ing exposure to dopamine receptor-blocking agents (DRBAs). are still being used by some clinicians in the treatment of
Despite the advent of atypical or second- and third-generation some hyperkinetic disorders, including TD, is that they are
antipsychotics with a presumably lower risk of complications, not associated with the development of TD. We also briefly
TD remains a persistent and challenging problem. Prevention discuss other treatment options for TD, including amantadine,
is the first step in mitigating the risk of TD, but early clonazepam, Gingko biloba, zolpidem, botulinum toxin, and
recognition, gradual withdrawal of offending medications, deep brain stimulation. Treatment of TD and other drug-
and appropriate treatment are also critical. As TD is often a induced movement disorders must be individualized and
persistent and troublesome disorder, specific antidyskinetic based on the severity, phenomenology, potential side effects,
therapies are often needed for symptomatic relief. The and other factors discussed in this review.
vesicular monoamine transporter 2 (VMAT2) inhibitors,
which include tetrabenazine, deutetrabenazine, and valbe-
nazine, are considered the treatment of choice for most
Key Points
patients with TD. Deutetrabenazine—a deuterated version of
tetrabenazine—and valbenazine, the purified parent product
Tardive dyskinesia remains highly prevalent because
of one of the main tetrabenazine metabolites, are novel
of the expanded use of ‘‘second-’’ and ‘‘third’’-
VMAT2 inhibitors and the only drugs to receive approval
generation antipsychotics and other dopamine
from the US FDA for the treatment of TD. VMAT2 inhibitors
receptor-blocking agents.
deplete presynaptic dopamine and reduce involuntary move-
ments in many hyperkinetic movement disorders, particularly Prevention of tardive dyskinesia by judicious use of
TD, Huntington disease, and Tourette syndrome. The active these drugs and early diagnosis is essential in
metabolites of the VMAT2 inhibitors have high affinity for lessening the impact of this iatrogenic disorder on
VMAT2 and minimal off-target binding. Compared with quality of life and in facilitating remission following
tetrabenazine, deutetrabenazine and valbenazine have phar- discontinuation of the offending agent.
macokinetic advantages that translate into less frequent dos- Deutetrabenazine and valbenazine, two vesicular
ing and better tolerability. However, no head-to-head studies monoamine transporter 2 inhibitors approved by the
US FDA, are the mainstay of treatment for tardive
dyskinesia.
& Joseph Jankovic
josephj@bcm.edu; In selected cases, other agents, such as botulinum
http://www.jankovic.org toxin for tardive dystonia, zolpidem for tardive
1
akathisia, amantadine for mild symptoms, or deep
Parkinson’s Disease Center and Movement Disorders Clinic,
brain stimulation for refractory symptoms, may also
Department of Neurology, Baylor College of Medicine, 7200
Cambridge, Suite 9A, Houston, TX 77030, USA be relevant treatment options.
N. Niemann, J. Jankovic

1 Introduction hypo- or hyperkinetic movement caused by DRBAs, is best


avoided in favor of more descriptive terms, based on
specific predominant phenomenology [5, 13, 14]. The Di-
Tardive dyskinesia (TD), a term coined by Faurbye et al.
agnostic and Statistical Manual of Mental Disorders, Fifth
[1] in 1964, represents a group of delayed-onset (i.e.,
Edition (DSM-5) defines TD as ‘‘involuntary athetoid or
‘‘tardive’’), persistent iatrogenic movement disorders sec-
choreiform movements (lasting at least a few weeks)
ondary to exposure to dopamine receptor-blocking agents
generally of the tongue, lower face and jaw, and extremi-
(DRBAs). These drugs have also been referred to as
ties (but sometimes involving the pharyngeal, diaphrag-
‘‘neuroleptics’’, but we prefer the term DRBAs, which we
matic, or trunk muscles) developing in association with the
use in this review. TD is typically manifested by stereo-
use of a neuroleptic medication for at least a few months’’
typies (especially involving the oro-bucco-lingual [O-B-L]
[15]. However, this definition is not sufficiently broad to
muscles), dystonia, chorea, akathisia, myoclonus, tremor,
cover the phenomenological spectrum of tardive movement
or tics [2–5]. All DRBAs, including typical or first-gener-
disorders and does not include TS due to DRBAs that are
ation antipsychotics (FGAs), atypical or second-generation
not antipsychotics [4].
antipsychotics (SGAs), and third-generation antipsychotics
Although some patients have limited insight into their
(TGAs), and drugs used for gastrointestinal indications
condition (and thus may not be bothered by it) [16], TD is
(e.g., metoclopramide, prochlorperazine, and promet-
by no means a benign condition. Other than being
hazine), may potentially cause TD [3]. Although SGAs are
uncomfortable and socially embarrassing, TD can be
associated with a reduced risk of TD, their introduction has
associated with reduced quality of life [3], risk of bodily
had limited impact on the incidence of TD [6]. This is
injury (excoriation of the lips, increased tendency to fall),
partly because use has increased with expanded labeling
and even death (neuroleptic malignant syndrome, respira-
[7]. Many DRBAs are approved for treatment of not only
tory dyskinesias with life-threatening hypo- and hyper-
schizophrenia but also bipolar disorder, depressive epi-
ventilation from uncoordinated movements of the
sodes, or irritability in autistic disorder and are frequently
diaphragm and chest wall) [17].
used off-label (e.g., in the treatment of anxiety, insomnia,
Tardive parkinsonism is a controversial entity where
etc.) [8, 9]. TD remains a challenging disorder for the
parkinsonism persists long after the offending agent has
clinician to manage because of a lack of reliable predictors
been withdrawn [3], unlike drug-induced parkinsonism
to guide risk stratification, variable clinical presentation,
[18]. In a study of 20 patients who developed parkinsonism
and the continued need for DRBAs to control psychiatric or
while being treated with a variety of DRBAs, 123I-ioflu-
gastrointestinal symptoms.
pane single-photon emission computed tomography
The goal of this review is to provide an update on
(SPECT) revealed that 11 (55%) patients had evidence of
recognition and management of TD with a focus on
presynaptic nigrostriatal deficits, suggestive of underlying
vesicular monoamine transporter 2 (VMAT2) inhibitors,
idiopathic Parkinson’s disease [19]. It is therefore possible
especially deutetrabenazine and valbenazine, which
that some patients diagnosed with tardive parkinsonism
recently received US FDA approval for the treatment of
had idiopathic Parkinson’s disease that had been unmasked
TD. Non-FDA-approved drugs, for which the level of
by dopamine receptor blockade. Thus, a more definite
evidence is comparatively lower, are also briefly reviewed.
diagnosis of tardive parkinsonism may require neu-
roimaging demonstrating a lack of striatal dopaminergic
denervation.
2 Definitions and Clinical Phenomenology
Neuroleptic withdrawal-emergent syndrome refers to
dyskinesias (often choreiform), chiefly involving the limbs,
DRBA-related movement disorders include (1) drug-in-
body, and neck, that arise as a withdrawal phenomenon,
duced movement disorders, such as tremors, myoclonus,
occurring most often in children during fast tapering or
parkinsonism, acute dystonic reaction, acute akathisia, and
after abrupt discontinuation of DRBAs. It is always diag-
neuroleptic malignant syndrome, which generally resolve
nosed retrospectively, as it is self-limiting [20].
when the offending drug is discontinued and (2) tardive
The co-occurrence of multiple different phenomenolo-
syndrome (TS), which includes TD and other movement
gies, such as akathisia and stereotypic dyskinesias, in a
disorders, such as parkinsonism, gait disorders, with-
patient with a history of exposure to DRBAs generally
drawal-emergent syndrome, and a variety of sensory
increases the likelihood that the movement disorder rep-
symptoms that persist even when the offending DRBA is
resents TD. When TD is suspected based on presenting
discontinued [2–4, 10, 11]. The terms ‘‘extrapyramidal’’
phenomenology in the absence of history of treatment with
and extrapyramidal syndrome or ‘‘EPS’’, still used even in
DRBAs, it is critical that medical, hospital, and pharmacy
some recent reviews in psychiatric literature [12], for any
records are carefully reviewed for any evidence of prior
Tardive Dyskinesia

exposure to DRBAs. Other etiologies should also be con- regarding TD severity were insufficient, the authors could
sidered, such as edentulous oral dyskinesias [21], other not determine whether the observed 9.3% difference in
drug-induced dyskinesias (including oral dyskinesias sec- prevalence rates translated into meaningful clinical sig-
ondary to anticholinergic medications and levodopa-in- nificance. As is evident from these studies, considerable
duced dyskinesias) that resolve following drug controversy still exists as to whether and how much SGAs
discontinuation [22, 23], Huntington disease [24, 25], reduce the risk of TD. Although the preponderance of
Wilson’s disease and other metabolic diseases [26], Tour- evidence is that SGAs lower the risk of TD, there are many
ette syndrome [27], and immune-mediated chorea [28]. reports of TD secondary to SGA use, including clozapine
Some patients treated with DRBAs for their hyperkinetic [45] (in some cases with limited or no prior exposure to
disorder, such as Huntington disease and Tourette syn- DRBAs [46] and in others with prior exposure [47, 48]),
drome, develop TD in addition to the underlying movement and the TGA aripiprazole [49].
disorder [29, 30]. The risk of TD in pediatric populations has been con-
sistently reported to be lower than in adult populations.
Studies of TD in children have yielded prevalence rates
3 Epidemiology and Risk Factors ranging from 7.6 to 20.4% [50–54]. A systematic review
[42] reported the annual incidence rate at just 0.35% in
Early studies of the risk of TD due to DRBA (specifically children. However, in a recent prospective study of 265
antipsychotic drugs) exposure indicated overall prevalence DRBA-naı̈ve or ‘‘quasi-naı̈ve’’ (less than 30 days of prior
rates close to 30% [31–33]. The annual incidence rate of DRBA exposure) children, 5.8% were found to meet cri-
TD is approximately 5% with FGAs, followed by a roughly teria for TD at 1-year follow-up after starting an SGA,
linear increase in incidence rates, at least during the initial suggesting that the risk of TD in children may not be
5 years of exposure [31, 34, 35]. However, in one study of significantly different from the risk in adults, although the
362 psychiatric outpatients followed for up to 25 years, dropout rate was 59% [55].
incidence rates were 32% after 5 years, 57% after 15 years, Risk stratification in the era of SGA is difficult for
and 68% after 25 years of exposure [34]. Elderly patients several reasons. Most studies of SGA are short term,
appear to be at much higher risk of early-onset TD due to employ restrictive enrollment criteria (thus potentially
exposure to FGAs, with 1-year incidence rates as high as excluding the patients at highest risk of TD), include
26% reported in prospective studies of patients with mean patients with prior exposure to or current use of FGAs, and
age[65 years [36, 37]. In a review of the literature from often use haloperidol as the comparator or no comparator at
2004, annual rates of TD associated with SGAs were as all [6, 17]. Further, earlier clinical trials of SGAs used
low as 0.8% in adults and 5.3% in patients aged[54 years higher than recommended doses of haloperidol, thus
[38]. This suggests that the SGAs reduced the incidence of potentially exaggerating the initially perceived benefit of
TD compared with FGAs, particularly in elderly popula- SGAs [38, 56]. Aside from type of DRBA and older age,
tions [39]. Coupled with lower rates of early parkinsonism other risk factors for the development of TD include
[40] and acute dystonic reactions with SGAs [41], these cumulative exposure to DRBAs (duration and dose)
findings helped fuel the expectation that the introduction of [57, 58]. Paradoxically, repeated interruptions of DRBA
SGAs would herald a vast reduction in rates of TD. treatment (e.g., drug holidays) increase the risk of TD [59].
Several studies have examined the relative frequency of Classic TD (O-B-L stereotypy) tends to occur more fre-
TD related to FGAs versus SGAs. The annual incidence of quently in elderly women, whereas tardive dystonia is more
TD was found to be 5.5% with FGAs and 3.9% with SGAs common in young men [4, 32, 60, 61]. Other risk factors
based on a systematic review of studies published between for TD include non-white race, mood disorders, diabetes
2004 and 2008 [42]. The same study also found lower mellitus, intellectual disability, prior history of brain
prevalence rates of TD with SGAs (13.1%) compared to injury, parkinsonism, and acute dystonic reactions
FGAs (32.4%). However, the US-based CATIE (Clinical [3, 17, 62].
Antipsychotic Trials of Intervention Effectiveness) [43] Although metoclopramide was the most common cause
and an intention-to-treat analysis of the UK-based of TD in patients followed in a movement disorders clinic
CUtLASS-1 (Cost Utility of the Latest Antipsychotics in about 10 years ago, the rate of TD secondary to this drug
Schizophrenia Study Band 1) [44] did not demonstrate a has substantially decreased, partly because of black box
significant difference in TD rates between FGAs and warning and increased awareness about this adverse effect
SGAs. Most recently, a meta-analysis of 41 studies yielded among internists and gastroenterologists [63–65]. Meto-
TD prevalence rates of 20.7 and 30.0% with current use of clopramide has also been reported to cause classic TD in
SGAs and FGAs, respectively [6]. However, because data children, even in an infant [53, 66].
N. Niemann, J. Jankovic

4 Pathophysiology without sacrificing the antipsychotic effect of the drug


[67, 68]. These effects may reduce the risk of TD due to
Drugs with the potential to cause TD include all FGAs, less D2 receptor upregulation [2]. Another theory proposes
SGAs, and TGAs, antiemetics (metoclopramide, prochlor- that chronic dopamine receptor blockade leads to increased
perazine, promethazine, clebopride, cisapride), anti-manic metabolism of dopamine, with production of neurotoxic
agents (lithium), calcium channel blockers (cinnarizine, free radicals resulting in nigrostriatal neurodegeneration
flunarizine), and serotonin reuptake and serotonin nore- [73].
pinephrine reuptake inhibitors (duloxetine, citalopram) In addition to the dopaminergic and serotonergic sys-
[2, 3]. tem, the GABAergic system may also play a role in the
Movements are regulated predominantly by two parallel pathophysiology of TD. For example, MSNs in the indirect
striato-pallidal pathways [67, 68]. The globus pallidus pathway receive feedforward inhibition from GABAergic
interna (GPi) provides an inhibitory input to the thalamus. interneurons, which are thought to be damaged or dys-
Increased or decreased activity in the GPi thus inhibits or functional (thus, promoting movement) in patients who
facilitates, respectively, thalamo-cortical activity and develop TD [74]. On the other hand, GABAergic drugs,
therefore movement. The GPi is chiefly regulated by the such as baclofen or benzodiazepines, have not been found
‘‘direct’’ pathway, a single neuron pathway from the motor to produce consistent benefit in patients with TD [3].
striatum to the GPi, which is inhibitory and therefore Genetic factors may also play an important role in the
facilitates movement, and by the ‘‘indirect’’ pathway, a risk of TD. For instance, polymorphisms in several genes,
three-neuron pathway from the motor striatum via the including those associated with drug metabolism;
globus pallidus externa (GPe) and subthalamic nucleus dopaminergic, GABAergic, and serotonergic transmission;
(STN) to the GPi, which is excitatory and therefore inhibits neuronal functioning and connectivity; and oxidative
movement. Dopaminergic stimulation of these pathways stress, have been implicated as susceptibility factors for TD
regulates (and facilitates) movements by exciting striatal [75].
medium spiny neurons (MSNs) in the ‘‘direct’’ pathway Synaptic plasticity is a dynamic process that allows
(expressing dopamine D1 receptors) and by inhibiting neurotransmission to be altered by prior experience [74].
those in the ‘‘indirect’’ pathway (expressing D2 receptors). This process is acutely impaired in the neocortex in
TD has been traditionally attributed to hypersensitivity humans after administration of DRBAs and deranged in
and upregulation of dopamine D2 receptors in the motor several other hyperkinetic movement disorders (such as
striatum due to chronic iatrogenic dopamine receptor Huntington disease and levodopa-induced dyskinesias) and
blockade, although this theory does not explain why TD in patients with schizophrenia after multiple episodes of
persists after withdrawal of the offending agent [68]. In one psychosis compared with patients with schizophrenia with
study, the dose of haloperidol correlated with the frequency first-time psychosis. Chronic dopamine receptor blockade
of vacuous chewing movements and extent of D2 receptor may thus lead to maladaptive synaptic plasticity associated
occupancy in rats [69]. A study utilizing positron emission with an imbalance between the direct and indirect basal
tomography in human subjects with schizophrenia ganglia pathway, resulting in the inability to unlearn
demonstrated increased dopamine D2 receptor binding in improper motor programs that are associated with abnor-
patients with a history of chronic DRBA exposure com- mal movements. This serves as a possible unifying theory
pared with controls [70]. D2 receptor hypersensitivity may to explain why TD most often persists months and years
enhance the inhibitory effect of dopamine in the ‘‘indirect’’ after withdrawal of DRBAs.
pathway and lead to hyperkinetic movement disorders.
Clinical evidence lends support to this theory. For instance,
increasing the dose of a DRBA will suppress involuntary 5 Rating the Severity of Tardive Dyskinesia (TD)
movements associated with TD, at least in the short-term,
whereas sudden withdrawal or dose reduction may pre- Although several TD rating scales exist, the Abnormal
cipitate or worsen TD [3]. Compared with FGAs, SGAs Involuntary Movement Scale (AIMS), developed in the
(particularly clozapine and quetiapine) have lower D2 1970s to evaluate abnormal movements related to TD
receptor affinity [71] and dissociate faster from the D2 [76–78], is the most frequently used scale in studies eval-
receptors (as quickly as 12–24 h vs. several days) [72]. uating treatments of TD [79]. The AIMS is a 12-item
They also antagonize 5-HT2A/C receptors, which is asso- clinician-administered rating scale that assesses the sever-
ciated with reduced D2 antagonist binding in the motor ity of abnormal movements in the face, neck, trunk, and
striatum relative to the nucleus accumbens, which may extremities (items 1–7); the severity, functional impact,
translate into less risk of drug-induced parkinsonism and patient awareness of their abnormal movements (items
Tardive Dyskinesia

8–10); and dental status and use of dentures (item 11–12), which has not yet been found effective outside of PD-
which may be associated with edentulous dyskinesia [21] related psychosis [90]) may be considered, based on a
and misdiagnosed as classic TD [77]. Items 1–10 are likely reduced risk of TD [3], although further studies are
scored along a 5-point scale (0 = none, 5 = severe), needed to support this approach [89]. For patients requiring
whereas items 11–12 require yes/no answers. Experienced treatment of nausea, antiemetics without dopamine recep-
raters generally have higher interrater reliability and record tor-blocking activity should be considered first line.
more consistent scores over time [80]. Although the AIMS Variable TD remission rates have been reported in the
has been used in most studies assessing response to phar- literature. In a study of 49 psychiatric patients with TD, 36
macologic treatment of TD, another validated rating scale were able to completely stop DRBAs. During follow-up
is the St. Hans Rating Scale [81]. ranging from 1 to 59 months (mean * 40 weeks), 2% had
complete resolution of TD, whereas 20% were noted to
have significant improvement of symptoms [91]. In a large
6 Prevention and Remission cohort of 108 patients with TD and mean DRBA expo-
sure of * 4.8 years, the majority had been treated with
Judicious use of DRBAs is the first step in preventing TD. DRBAs for mood disorders or metoclopramide for gas-
Unfortunately, SGAs are increasingly prescribed across all trointestinal illness (94% non-psychotic indication) [92].
age groups [82–84] and off-label uses have expanded to The inciting drug was completely withdrawn in all cases.
include depressive disorder, a variety of behavioral prob- After a mean follow-up of 3.1 years, complete remission
lems, insomnia, anxiety, and head trauma [85]. Conversely, was seen in 13% of patients, but only 2.8% were not
prescriptions for metoclopramide, an antiemetic and gas- receiving symptomatic treatment. In contrast, remission
trointestinal prokinetic agent with D2 receptor-blocking rates up to 62% have been reported in other studies where
properties, one of the most common cause of TD a few patients continued treatment with DRBAs after developing
years ago [63], have decreased since February 2009, when TD [93, 94], likely due to masking of symptoms secondary
the FDA issued a black box warning regarding the risk of to dopamine receptor blockade.
TD [86].
According to American Psychiatric Association guide-
lines, patients should be examined for signs of TD before 7 Treatment of TD
initiating DRBA therapy, at least biannually in patients
taking DRBAs, and upon changes in DRBA dose or agent Figure 1 outlines a general approach to the treatment of
or observation of previously undetected movements [87]. TD. Although, historically, many different drugs have been
Furthermore, patients should be clinically assessed for used in the treatment of TD, as outlined in Fig. 1 and in the
abnormal involuntary movements if they are taking FGAs text, the authors currently treat TD almost exclusively
every 6 months, or every 3 months if at increased risk; if using VMAT2 inhibitors because of their well-documented
they are taking SGAs they should be assessed every efficacy, tolerability, safety, and their FDA approval.
12 months, or every 6 months if at increased risk. Informed Table 1 compares the clinical pharmacology of the
consent when prescribing a DRBA is prudent practice and VMAT2 inhibitors, and Table 2 details a general outline
may prevent legal consequences. for the management of potential side effects related to
To mitigate the risk of TD, DRBAs with a lower risk of treatment with VMAT2 inhibitors. Evidence based guide-
TD should be preferentially used and at the lowest effective lines regarding the treatment of TD have previously been
dose for the shortest period of time with frequent critical published [89, 95].
appraisal of the continued need for treatment. Patients who
are maintained chronically on DRBAs should frequently be 7.1 Drugs with High Level of Evidence for Use
reassessed for early signs of TD. When TD is identified, it in TD
is important to try and withdraw DRBAs in patients who
can tolerate it. DRBAs should be withdrawn slowly, as 7.1.1 Dopamine-Depleting Agents: The Vesicular
sudden withdrawal may worsen or precipitate TD [3] and Monoamine Transporter 2 (VMAT2) Inhibitors
worsen the underlying psychiatric disorder [88]. The SGAs
risperidone and olanzapine may be effective in suppressing VMAT2 transports cytoplasmic monoamines, including
TD, but the American Academy of Neurology (AAN) 2013 dopamine, histamine, norepinephrine, and serotonin, into
guidelines cautioned against the use of SGAs in the treat- presynaptic vesicles by secondary active transportation
ment of TD as they may themselves cause TD [89]. In using the electrochemical proton gradient generated by the
patients who require continued antipsychotic treatment, vesicular membrane H?-ATPase [96]. Monoamines pre-
clozapine and quetiapine (and possibly pimavanserin, sent in the cytosol are degraded by monoamine oxidase.
N. Niemann, J. Jankovic

Differential diagnosis: Stop


No Yes
TD? anticholinergic
• Edentulous dyskinesias (unless tardive
• Drug-induced dyskinesias dystonia)
(e.g. levodopa)
• Hunngton disease and
other genec choreas • Reduce dose
Chronic
• Wilson’s disease and other No Yes • Consider
DRBA use
metabolic disorders Taper off necessary? clozapine,
• Touree syndrome slowly queapine, or
• Immune-mediated choreas possibly
pimavanserin
Persistent TD

Tardive Tardive Tardive Dose Comments


stereotypy dystonia akathisia (mg/day)
and/or chorea
Tetrabenazine 25-200 Monitor for akathisia,
X
depression, insomnia,
Deutetrabenazine 12-48 parkinsonism, sedation*
X
Valbenazine X 40-80
Deep brain Target: Experienced centers only
X (refractory) X
stimulation GPi
Botulinum toxin X X Variable 1st line for focal dystonia
Amantadine 100-300 Monitor for cognitive
X X X dysfunction and
hallucinations
Baclofen X 10-80 Monitor for sedation
Clonazepam 80-240 Monitor for sedation. May
X X
develop tolerance
Gingko biloba 0.25-4** Caution with use of
X X
(Egb-761) antiplatelets/anticoagulants
Propranolol 30-160 Monitor heart rate and blood
X X
pressure
Zolpidem X X 10-30 Monitor for sedation
Trihexyphenidyl 1-20 mg Monitor for sedation and
X
anticholinergic side effects
Fig. 1 Management of tardive dyskinesia. DRBA dopamine receptor- may be better tolerated than tetrabenazine. **240 mg/day used in
blocking agent, GPi globus pallidus interna, TD tardive dyskinesia, randomized controlled trial
VMAT2 vesicular monoamine transporter 2. *Similar side effects but

Inhibition of VMAT2 thus reduces presynaptic storage and hyperkinetic movement disorders, is therefore associated
release of monoamines, particularly dopamine (Fig. 2 with multiple peripheral side effects related to peripheral
[97]). Whereas VMAT2 is present only in the central VMAT1 inhibition, such as bronchospasm, nausea, vom-
nervous system, VMAT1 is present both centrally and iting, diarrhea, hypotension, itching, and nasal stuffiness
peripherally [98]. Reserpine, a nondiscriminatory VMAT1/ [99]. The selective VMAT2 inhibitors tetrabenazine,
2 inhibitor previously used to treat hypertension and deutetrabenazine, and valbenazine are less likely to be
Tardive Dyskinesia

Table 1 Vesicular monoamine transporter 2 inhibitors


Characteristic Tetrabenazine (XenazineÒ) Deutetrabenazine (AustedoÒ) Valbenazine (IngrezzaÒ)

FDA approval (year) TD (off-label), HD chorea (2008) TD (2017), HD chorea TD (2017)


Pivotal trials leading to FDA approval Not applicable ARM-TD, AIM-TD KINECT-2, KINECT-3
for TD
Mechanism of action Reversible inhibition of VMAT2 Reversible inhibition of Reversible inhibition of
VMAT2 VMAT2
Active metabolites Yes Yes Yes
Half-life (h) of active metabolites 5–7 9–10 15–22
Long-term efficacy and safety data * 20 *1 *1
(years)
Special considerations regarding None Take with fooda None
administration
Initial dose (mg/day) 12.5 6 40
Titration (mg/week) 12.5 6 40
Usual dose range (mg/day) 50–100 (maximum 200) divided 24–48c divided in two doses 40 or 80 as one dose
in three doses a dayb a day a day
CYP cytochrome P450, FDA US Federal Drug Administration, HD Huntington disease, TD tardive dyskinesia, VMAT2 vesicular monoamine
transporter 2
a
Increases the peak plasma concentration (Cmax) of the active metabolites
b
Consider CYP2D6 genotyping for doses[50 mg/day
c
In some cases, higher doses, up to 72 mg as used in some HD studies, may be required

Table 2 Practical management of side effects related to vesicular monoamine transporter 2 inhibitors
Side effect Management strategy

Medication Review co-administered medications, switch to alternative agents if interactions are found
interactionsa
Any side effect Consider dose reduction
Akathisia Clonidine, gabapentin, propranolol, zolpidem
Anxiety and Selective serotonin reuptake inhibitors, serotonin and norepinephrine reuptake inhibitors, tricyclic antidepressants,
depression mirtazapine
Insomnia Eszopiclone, zaleplon, zolpidem, trazodone, mirtazapine
Parkinsonism Amantadine, ropinirole, pramipexole, rotigotine, levodopa
Sedation Armodafinil, modafinil, methylphenidate, lisdexamfetamine
CYP cytochrome P450, VMAT2 vesicular monoamine transporter 2
a
Caution with concomitant use of CYP2D6 inhibitors (e.g., fluoxetine, paroxetine, cinacalcet, mirabegron [may increase concentration of active
metabolites of all VMAT2 inhibitors]), CYP3A4 inhibitors (e.g., clarithromycin, itraconazole, fluconazole, ketoconazole, verapamil, cimetidine
[may increase concentration of valbenazine and its metabolite, [?]-a-DHTBZ]), and QTc-prolonging agents (e.g., amitriptyline, citalopram,
escitalopram, quetiapine, venlafaxine, or CYP2D6 inhibitors by virtue of increasing VMAT2 inhibitor metabolites [all VMAT2 inhibitors])

associated with peripheral side effects, as they do not 7.1.2 How Do VMAT2 Inhibitors Reduce Unwanted
inhibit peripheral VMAT1 and are therefore better toler- Movements in TD?
ated. VMAT2 inhibitors have never been associated with
the development of TD. A single published report [100] is As discussed in the pathophysiology section, the brain of a
difficult to accept as a case of tetrabenazine-induced TD patient with TD has been postulated to compensate for
[99]. chronic dopamine receptor blockade by upregulating and
raising the sensitivity of D2 receptors to dopamine (‘‘D2
receptor hypersensitivity’’), amongst other changes,
including alteration of synaptic plasticity. In effect,
N. Niemann, J. Jankovic

Fig. 2 Mechanism of action of VMAT2 inhibitors a VMAT2 degraded by monoamine oxidase. In contrast, dopamine receptor-
sequesters dopamine in presynaptic vesicles. Upon fusion of the blocking agents work post-synaptically by blocking membrane-bound
vesicle with the membrane, dopamine is released into the synaptic dopamine receptors. VMAT2 vesicular monoamine transporter 2.
cleft to interact with postsynaptic dopamine receptors. b VMAT2 Reproduced from Jankovic [97] with permission
inhibitors block vesicular storage of dopamine. Instead, dopamine is

excessive stimulation of D2 receptors leads to excessive [99]. In turn, this leads to less stimulation of hyper-
inhibition of the indirect pathway, which serves to reduce sensitive D2 receptors and therefore less inhibition of
or stop movements, leading to relative dominance of the the indirect pathway, which leads to a reduction of
direct pathway [68]. The sum of these changes are exces- movements [102]. Less dopamine will also be avail-
sive movements. At least three ways of addressing the able to stimulate postsynaptic D1 receptors in the
imbalance between the direct and indirect pathway exist: direct pathway, which provides another mechanism of
action of VMAT2 inhibitors in TD. However, the
1. Stopping the offending agent with the goal of allowing
response to a specific dose of any of the VMAT2
the D2 receptor sensitivity to normalize and the brain
inhibitors is likely to be variable, in part depending on
to unlearn the maladaptive motor programs associated
the amount of dopamine present in the presynaptic
with TD, although this is rarely effective as discussed
terminal and the synaptic cleft, which could be altered
in previous sections.
by dopamine-reuptake inhibitors and releasers, such as
2. Raising the dose of the offending DRBA, or, if the
amphetamines, and monoamine oxidase inhibitors or
patient is not currently on a DRBA, starting a DRBA.
concomitant use of a DRBA.
This would serve to further block the hypersensitive
D2 receptors with the hope of ‘‘normalizing’’ the
activity in the indirect pathway. However, as previ- 7.1.3 Tetrabenazine
ously stated, such a course of action, while likely
reducing excessive movements in the short term, is Tetrabenazine was originally developed in the 1950–1960s
likely to cause other side effects (such as parkinson- as a novel antipsychotic and antihypertensive agent. It was
ism, sedation, and metabolic changes) and may approved in the UK in 1971 for the treatment of hyperki-
eventually worsen TD [101]. netic movement disorders, including TD [99]. It was
3. Reducing striatal dopamine using a VMAT2 inhibitor. approved by the US FDA in 2008 for the treatment of
Rather than treating the disorder where the problem is, chorea associated with Huntington disease and is currently
i.e., at the postsynaptic D2 receptor, a VMAT2 being used off-label in the treatment of TD. At least 75% of
inhibitor preferentially reduces the amount of dopa- tetrabenazine is absorbed after oral ingestion, although it is
mine stored in presynaptic vesicles and thus the generally undetectable (doses of 12.5–50 mg) due to rapid
amount of dopamine released into the synaptic cleft metabolism, mainly by carbonyl reductase [99], to positive
Tardive Dyskinesia

and negative (?/-) isomers of a- and b-dihydrotetra- (p\0.001), after treatment with tetrabenazine for an
benazine (DHTBZ), which reach their peak plasma con- average of 20.3 weeks at a mean daily dose of tetra-
centration (Cmax) within 1–1.5 h after each dose [103]. a- benazine 57.9 mg/day. All patients who completed the
and b-DHTBZ have VMAT2-binding affinities (Ki) of 3.1 study (95%) reported subjective improvement in symp-
and 20 nM, respectively, compared with 100 nM for toms, and 85% rated their improvement as moderate or
tetrabenazine [103, 104]. a- and b-DHTBZ subsequently marked on a global response scale (GRS; where
undergo conjugation with sulfate and glucuronide, medi- 1 = marked improvement, 4 = no change, 5 = worse).
ated by the cytochrome P450 (CYP) enzyme system, The only other prospective and single-blind study included
mainly CYP2D6, prior to renal excretion (75% of each 24 chronic psychiatric patients with TD [110]. The study
dose). Tetrabenazine metabolites have limited or no off- consisted of a 4-week baseline period during which med-
target binding at adrenergic, dopaminergic, and/or sero- ications were not altered (14 patients were taking DRBAs),
tonergic receptors, although the negative isomer of b- followed by 4 weeks on placebo during which all DRBAs
DHTBZ has a Ki of 53 nM at the D2 receptor, which is of were withdrawn, then 6 weeks’ treatment with tetra-
unclear significance [105]. The half-lives of a- and b- benazine (starting at 50 mg/day with titration to
DHTBZ are 7 and 5 h, respectively [103]. Since poor 100–150 mg/day), and finally a 2-week tetrabenazine
metabolizers of the active metabolites of tetrabenazine may washout period, during which patients were given placebo.
be at higher risk of treatment-associated side effects for a At the end of the tetrabenazine period, eight patients had
given dose than intermediate, extended, and ultra-rapid complete resolution of symptoms, six had marked
metabolizers, the FDA has recommended CYP2D6 geno- improvement, six had little or no change, and four patients
typing for doses of tetrabenazine exceeding 50 mg/day dropped out. The frequency of oral dyskinesias was
[106]. In a study of 127 patients with a range of hyperki- reduced by 64% in comparison with placebo.
netic movement disorders, 11 patients were categorized as In two large retrospective series including patients with
poor, 14 as intermediate, 100 as extensive, and two as various hyperkinetic movement disorders treated with
ultra-rapid metabolizers [106]. The titration period was tetrabenazine for up to 21.6 years,[84% of patients with
significantly longer for ultra-rapid metabolizers than for the TD (total of 242 patients) rated their improvement as either
other groups (p\0.01), although there were no significant moderate or marked on a GRS [111, 112]. In these two
differences in the total daily dose, treatment response, or studies, the most common side effects of tetrabenazine (all
frequency of adverse events between the groups [106]. The indications) included sedation (25.0–36.5%), parkinsonism
authors concluded, ‘‘these results do not support the need to (15.4–28.5%), depression (7.6–15.0%), insomnia
systematically genotype all patients prescribed more than (4.9–11.0%), anxiety (5.1–10.3%), and akathisia
50 mg/day of tetrabenazine, and this recommendation (7.6–9.5%), all of which occurred in a dose-dependent
should be reconsidered’’. Of similar concern for all the fashion. While there is a higher risk of depression while
specific VMAT2 inhibitors (tetrabenazine, deutetra- being treated with tetrabenazine in patients with a pre-
benazine, and valbenazine), studies have shown that co- existing history of depression, this symptom can be man-
administration with strong CYP2D6 inhibitors (e.g., aged by dose reduction or addition of an antidepressant
quinidine, paroxetine, fluoxetine) may significantly [113]. Tetrabenazine has received a level C recommenda-
increase the area under the curve (AUC), half-life, and tion and may be considered for treatment of TD according
Cmax of the active metabolites, which may increase side to the AAN 2013 guideline [89] as well as in a recently
effects [103, 107, 108]. Other potential safety concerns for published systematic review [95].
these drugs as a group include the propensity for QTc- Given the need for frequent dosing and the considerable
prolongation, particularly in individuals with poor side effects associated with tetrabenazine, novel VMAT2
CYP2D6 activity, in the presence of strong CYP2D6 inhibitors have garnered significant interest in recent years,
inhibitors, or when co-administered with other agents that and these are discussed in the subsequent sections.
have QTc-prolonging effects (e.g., DRBAs, moxifloxacin,
quinidine). As such, monitoring of the QTc-interval may be 7.1.4 Deutetrabenazine
warranted.
Most studies of tetrabenazine in TD have been relatively Deutetrabenazine, a version of tetrabenazine containing
small. A single-blind prospective study of tetrabenazine in deuterium (also known as ‘‘heavy hydrogen’’: hydrogen
TD included 20 patients (15 female) who were assessed via with one neutron rather than none) as two trideuter-
a randomized videotape protocol before and after treatment omethoxy groups in place of two methoxy groups, is a
[109]. One patient did not tolerate tetrabenazine because of novel and highly specific VMAT2 inhibitor [99]. Deuter-
sedation and did not complete the study. The average ation does not alter the target pharmacology of a drug
improvement on the AIMS was 54.2%, from 17.9 to 8.2 [114]. At least 80% of deutetrabenazine is absorbed after
N. Niemann, J. Jankovic

oral ingestion, although it is generally undetectable due to 24, or 36 mg daily (divided in two doses) for 8 weeks after
rapid metabolism, also by carbonyl reductase, to deuterated a 4-week titration period [117]. LS mean AIMS scores
a- and b-DHTBZ, which reach Cmax within 3–4 h after improved by - 3.2 (p = 0.003) and - 3.3 (p = 0.001) in
each dose [107]. These metabolites are then further the groups receiving 24 and 36 mg/day, respectively,
metabolized by CYP2D6. More than 75% of each dose is compared with - 1.4 for placebo. Improvement in AIMS
excreted renally. The half-life of the active metabolites is scores for the group receiving 12 mg/day were insignificant
9–10 h according to the prescribing information [107]. (p = 0.217). Treatment success was defined as ‘‘much
There is no significant difference in off-target binding or improved’’ or ‘‘very much improved’’ on the CGIC and
VMAT2-binding affinity (Ki) for the deuterated than for was achieved in 49% of patients receiving 24 mg/day
the non-deuterated forms of a- and b-DHTBZ (3.8 vs. (p = 0.014) and 44% of those receiving 36 mg/day
3.1 nM and 22 vs. 20 nM, respectively) [104]. By (p = 0.059) compared with 26% with placebo, whereas
strengthening the carbon–hydrogen bond, deuteration there was no significant difference in outcome on the PGIC
slows metabolism of the active metabolites, resulting in of deutetrabenazine compared with placebo. In a pooled
longer half-lives (from 4.8 to 8.6 h), larger AUC, and lower analysis (not including patients receiving 12 mg/day) of
Cmax than an identical dose of tetrabenazine, according to the results of both ARM-TD and AIM-TD, 30.0% of
Stamler et al. [115]. Food administration increases the Cmax patients receiving deutetrabenazine (vs. 14.8% for placebo)
of the active metabolites deutetrabenazine (but not tetra- had a[50% reduction of the AIMS score, equal to a
benazine) by 50% [103, 107]. Clinically, the pharmacoki- number needed to treat (NNT) of seven (95% confidence
netic advantages of deutetrabenazine translate into less interval [CI] 4–18) [118]. For CGIC and PGIC, the results
frequent dosing (twice vs. three times daily) and lower dose were 46.9% (vs. 33.0%) with an NNT of eight (95% CI
per drug administration (6 mg is roughly equivalent to 4–45) and 42.5% (vs. 30.4%) with an NNT of nine (95% CI
12.5 mg) than for tetrabenazine to achieve the same clin- 5–145), respectively, for a favorable outcome, i.e., out-
ical effect, which may translate into fewer adverse effects comes of ‘‘very much improved’’ (score 1) or ‘‘much
due to lower peak plasma concentrations [99]. This may improved’’ (score 2). Deutetrabenazine was well-tolerated,
further result in less need for CYP2D6 genotyping, if such with low rates of side effects in both ARM-TD and AIM-
testing is pursued at all [106]. Potential safety concerns for TD. In fact, the cumulative side effects of deutetrabenazine
deutetrabenazine with respect to concomitant use of strong versus placebo reported in both ARM-TD and AIM-TD
CYP2D6 inhibitors and QTc-prolonging agents are other- suggest minimal differences between the two groups in
wise similar to those for tetrabenazine (see Sect. 7.1.3). terms of the frequency of reported side effects (sedation,
Deutetrabenazine received FDA approval for the treat- fatigue, diarrhea, dry mouth, anxiety, akathisia, and
ment of TD on 30 August 2017 based on two pivotal depression), all of which occurred at a rate of B 4.3% in the
double-blind, randomized, placebo-controlled trials: the deutetrabenazine group [118]. Insomnia occurred in 6.9
phase II/III trial ARM-TD (Aim to Reduce Movements in versus 1.7% in ARM-TD but was not reported in AIM-TD
TD) and the phase III trial AIM-TD (Addressing Involun- [116, 117]. Deutetrabenazine did not result in prolongation
tary Movements in TD) [107]. In both studies, patients of the QTc interval relative to placebo. A 159-week
could continue use of DRBAs or antidepressants provided extension study of ARM-TD and AIM-TD is currently
there had been no recent change in these medications. underway to assess safety, adverse effects, and efficacy
ARM-TD randomized 117 patients 1:1 to receive [119]. An interim analysis of this study at 54 weeks
deutetrabenazine or placebo starting at 12 mg daily (di- showed that deutetrabenazine was well-tolerated long term,
vided in two doses) followed by titration with 6 mg/day/ with low rates of adverse effects [120]. Deutetrabenazine
week for up to 6 weeks until dyskinesia was controlled, has received a level A (established as effective) recom-
uncontrollable side effects occurred, or a maximum dose of mendation for the treatment of TD [95].
48 mg/day was reached [116]. This was followed by a A recent study demonstrated that overnight conversion
6-week maintenance period. The mean daily dose was of tetrabenazine to deutetrabenazine followed by dose
38.8 mg at the end of the study period. Deutetrabenazine titration to maximize efficacy is well-tolerated and asso-
reduced least-squared (LS) mean AIMS scores by - 3.0 ciated with improved motor outcome in patients with
compared with - 1.6 for placebo (p = 0.019). Further, chorea associated with Huntington disease [121].
there was a nonsignificant difference in the Clinical Global Deutetrabenazine is also FDA approved for the treatment
Impression of Change (CGIC) and Patient Global Impres- of Huntington disease [122] and, based on positive pilot
sion of Change (PGIC), both rated from 1 (very much data [123], is being investigated as a treatment for Tourette
improved) to 7 (very much worse), in favor of deutetra- syndrome.
benazine over placebo. AIM-TD randomized 298 patients
1:1:1:1 to receive either placebo or deutetrabenazine 12,
Tardive Dyskinesia

7.1.5 Valbenazine valbenazine (40 or 80 mg/day) compared with placebo,


although further analyses in the per-protocol population
Valbenazine is the purified parent prodrug of the (?)-iso- showed significant improvement for valbenazine (both
mer of a-DHTBZ [85, 124, 125]. Approximately 49% of p = 0.011 for 40 and 80 mg/day) compared with placebo.
ingested valbenazine is absorbed following oral adminis- In a pooled analysis (all doses) of the results of KINECT-2
tration [108]. Valbenazine is metabolized via two routes: and -3, a total of 36.5% of patients in the valbenazine group
(1) hydrolysis of the valine ester leads to conversion to a- (vs. 12.4% for placebo) had a[50% reduction of AIMS
DHTBZ, the primary VMAT2-inhibitor (Ki = 1–4.2 nM), scores, yielding an NNT of five (95% CI 3–7) [128]. The
which is further metabolized by CYP2D6, and (2) mono- pooled results of patients achieving a favorable response
oxidative metabolism of valbenazine by CYP3A4/5, which (i.e., score 1 or 2) on the CGI-TD was 40.4% for valbe-
generates the mono-oxy metabolite NBI-13611 nazine (vs. 18.6% for placebo), yielding an NNT of five
(Ki = 160–220 nM), both of which have no off-target (95% CI 4–9). The most common side effects reported in
binding [85, 105, 108, 126]. Cmax of valbenazine and a- both studies (valbenazine vs. placebo) were somnolence
DHTBZ is reached 0.5–1 and 4–8 h, respectively, after (5.4 vs. 3.2%), headache (4.5 vs. 3.2%), fatigue (4.0 vs.
ingestion. Concomitant intake of fat significantly decreases 2.4%), dry mouth (4.0 vs. 0.8%), vomiting (3.0 vs. 0%),
the Cmax (47%) and AUC (13%) of valbenazine, but the and urinary tract infection (2.5 vs. 4.8%) [128]. In
Cmax and AUC of a-DHTBZ is unchanged. Valbenazine KINECT-3, up to 4.2% of patients taking valbenazine
itself has low affinity for VMAT2 (Ki = 110–190 nM). reported akathisia and suicidal ideation (vs. 1.3 and 5.3%,
The half-life of valbenazine and a-DHTBZ is 15–22 h. respectively, for placebo) [127]. A recently published
These pharmacodynamic and -kinetic advantages allow for pooled analysis of cardiovascular outcomes showed no
once-daily dosing and potentially better tolerability com- significant differences between the placebo and valbe-
pared with tetrabenazine. Valbenazine metabolites pre- nazine groups [129]. The 1-year KINECT-3 extension
dominantly undergo renal excretion (60%). Safety study demonstrated long-term efficacy, safety, and tolera-
concerns with regards to concomitant use of strong bility of valbenazine for the treatment of TD [130]. Val-
CYP2D6 inhibitors or QTc-prolonging agents are other- benazine has received a level A recommendation for the
wise similar to those with tetrabenazine and deutetra- treatment of TD [95].
benazine, with the additional warning that co- Valbenazine is currently also being investigated in the
administration of strong CYP3A4 inhibitors (e.g., keto- treatment of Tourette syndrome.
conazole) may increase the Cmax and AUC of a-DHTBZ The role of VMAT2 inhibitors in the treatment of
and potentially increase the risk of clinically significant hyperkinetic movement disorders is evolving. Their future
side effects. use may include other indications, including treatment of
KINECT-2, a phase II, double blind, randomized, pla- self-injurious behaviors and possibly even as antipsy-
cebo-controlled dose-titration study, randomized 102 chotics. Indeed, tetrabenazine was initially developed as an
patients to placebo or valbenazine 25 mg/day with antipsychotic. Finally, the hypothesis that one of the
biweekly increments of 25 mg up to 75 mg/day (76% of authors (JJ) proposed decades ago, that co-administration
patients reached the maximum dose) [126]. At week 6, LS of VMAT2 inhibitors with DRBAs may prevent or reduce
mean AIMS scores were reduced by - 2.6 for the valbe- the risk of developing TD, should be tested in a prospective
nazine group compared with - 0.2 for the placebo group longitudinal study.
(p = 0.0005). Results on the CGI-TD and PGIC identified
as ‘‘much improved’’ or ‘‘very much improved’’ were also 7.2 Drugs with Lower Level of Evidence for Use
significantly higher for valbenazine than for placebo (66.7 in TD
vs. 15.9%, p\0.0001, and 57.8 vs. 31.8%, p = 0.001,
respectively). Valbenazine received FDA approval for the 7.2.1 Amantadine
treatment of TD on 11 April 2017 [108] based largely on
the positive results of KINECT-3, a phase III, double-blind, Amantadine is a non-competitive N-methyl-D-aspartate
randomized, placebo-controlled trial [127]. In this study, (NMDA) antagonist with anti-glutamatergic properties
234 patients, of which 86% received concomitant DRBAs, frequently used in the management of levodopa-induced
were randomized 1:1:1 to receive once-daily placebo or dyskinesias [23]. It was evaluated in the treatment of TD in
valbenazine (40 or 80 mg) for 6 weeks. LS mean AIMS two small (16 and 22 patients) randomized placebo-con-
scores improved by - 1.9 for valbenazine 40 mg/day trolled trials. The studies demonstrated 15–22% (both
(p = 0.002) and - 3.2 for valbenazine 80 mg/day p\0.05) reduction of AIMS scores, supporting the short-
(p\0.001) compared with - 0.1 for placebo. No signifi- term use of amantadine in the treatment of TD when used
cant difference on CGI-TD scores were seen at 6 weeks for in conjunction with DRBAs [131, 132]. Amantadine has
N. Niemann, J. Jankovic

Table 3 Active phase II or III trials for treatment of tardive dyskinesiaa


Study Status; Phase n Design Intervention Outcome
duration

NCT03254186 Recruiting; II/III 15 Randomized, Propranolol Primary: change in AIMS score. Secondary: Change in
Sep 2017– crossover, hydrochloride CGI-S and CGI-I score
Feb 2019 blinded, PC 20 mg QID vs.
PL
NCT03176771 Recruiting; II/III 240 Multicenter, (1) 40 or Primary: change in AIMS score
Jun 2017– randomized, 80 mg/day MT-
Jul 2019 blinded, PC 5199b vs. PL
(2) Extension
study: 40 or
80 mg MT-
5199
NCT01804920 Not II/III 16 Randomized, D-serine (up to Primary: change in AIMS score
recruiting; blinded 4 g/day) vs. PL
Mar 2013–
Jan 2018
NCT02198794 Not III 343 Open-label SD-809c Primary: long-term incidence of AE, SAE, drug-related
(RIM-TD) recruiting; extension AE, AE leading to drug withdrawal. Secondary:
Oct 2014– study change in AIMS score, QOL, PGIC, proportion of
Sep 2019 responders
AE adverse effect, AIMS Abnormal Involuntary Movement Scale, CGI-I Clinical Global Impression of Improvement, CGI-S Clinical Global
Impression of Severity, PC placebo-controlled, PGIC Patient Global Impression of Change, PL placebo, QID four times daily, QOL quality of
life, SAE serious adverse effect
a
From http://www.clinicaltrials.gov using search terms ‘‘tardive dyskinesia,’’ ‘‘not yet recruiting,’’ ‘‘recruiting,’’ ‘‘active, not recruiting,’’ ‘‘phase
2, 3’’
b
MT-5199 = valbenazine
c
SD-809 = deutetrabenazine

received a level C (possibly effective) recommendation for experienced a[30% (p\0.001) reduction in AIMS scores
the treatment of TD [95] compared with 5.1% of patients in the placebo group [135].
Interestingly, a polymorphism in the gene for brain-derived
7.2.2 Clonazepam neurotrophic factor (Val/Val genotype) seems to predict a
favorable response to EGb-761 [136]. Gingko biloba has
Clonazepam, a GABAA agonist (benzodiazepine), was received a level B recommendation for the treatment of TD
evaluated in a 12-week double-blind, randomized, cross- [95].
over trial that included 19 patients with TD [133]. An
overall 37% (p\0.001) decrease in the Maryland Psychi- 7.3 Other Drugs with Minimal or No Evidence
atric Research Center Movement Disorders Scale was seen for Use in TD
at 12 weeks. Tolerance developed within 3–9 months, so
clonazepam is therefore recommended only for short-term Several oral agents either have minimal benefit, insufficient
(\3 months) treatment of TD. Clonazepam has received a evidence to support or refute their use, or evidence to
level B (probably effective) recommendation for the support not using them as treatment for TD. These medi-
treatment of TD [95]. cations include acetazolamide, anticholinergics (e.g., ben-
ztropine), cholinergics (e.g., donepezil), antioxidants,
7.2.3 Gingko Biloba buspirone, calcium channel blockers, eicosapentaenoic
acid, levetiracetam, melatonin, pyridoxine, propranolol,
A Gingko biloba extract (EGb-761) with antioxidative thiamine and other vitamins, and other prescription and
properties has been found to be beneficial in the treatment non-prescription treatments [3, 89, 95]. Although anti-
of patients with schizophrenia with TD [134]. In a double- cholinergics may be useful in tardive dystonia, their routine
blind, randomized, placebo-controlled trial of 157 patients use in the treatment of so-called extrapyramidal symptoms
with TD, treatment with EGb-761 reduced AIMS scores by should be discouraged because of cognitive side effects
an average of - 2.13 (p\0.0001) compared with ?0.1 for [137, 138] and worsening of O-B-L stereotypies [139].
placebo, whereas 51.3% of patients in the EGb-761 group Zolpidem was reported to be effective for the treatment of
Tardive Dyskinesia

TD and tardive akathisia in a small series of three patients, mandatory. Whenever possible, DRBAs should be slowly
although further studies are needed [140]. withdrawn or switched to an alternative with less or no risk
of TD. When TD does occur, VMAT2 inhibitors, a class of
drugs that now include two FDA-approved medications
8 Other Treatment Options (deutetrabenazine and valbenazine) for which evidence of
long-term efficacy and safety exists, are the mainstay of
8.1 Botulinum Toxin treatment, including in the setting of concomitant DRBA
exposure. In milder cases, use of amantadine, clonazepam,
Although no controlled trials have evaluated botulinum or Gingko biloba may also be considered. Finally, DBS
toxin in the treatment of TD, several small reports describe should be reserved for severe, refractory cases, whereas the
improvement of involuntary tongue movements, O-B-L main indication for botulinum toxin, anticholinergics, and
stereotypies, pain, and craniocervical dystonia muscle relaxants is dystonia, although the evidence is
[2, 3, 141, 142]. Given the absence of controlled trials, scarce.
botulinum toxin currently has a level U (insufficient evi-
dence) recommentation for the treatment of TS [95].
Author contributions NN contributed to the conception, design,
organization, and execution of the study and the writing of the first
8.2 Deep Brain Stimulation and subsequent drafts. JJ contributed to the conception, design,
organization, and execution of the study, to review and critique, and
Deep brain stimulation (DBS) of the GPi is reserved for to writing of the second and subsequent drafts.
medically refractory and severely disabling TD (level C
recommendation) [95]. More than 70 cases of DBS tar- Funding No sources of funding were used to conduct this study or
prepare this manuscript.
geting the GPi for TD have been reported in the literature
as case reports and case series, in many instances demon- Compliance with Ethical Standards
strating[50% long-term improvement of symptoms on
standardized rating scales [143]. Two case reports have Conflict of interest Nicki Niemann has no conflicts of interest that
are directly relevant to the content of this study. Dr. Jankovic has
also reported improvement of tardive dystonia following received research and/or training grants from Adamas Pharmaceuti-
treatment with STN DBS [144, 145]. Given the lack of cals, Inc.; Allergan, Inc.; Biotie Therapies; CHDI Foundation; Civi-
controlled studies, as well as the invasive nature of the tas/Acorda Therapeutics; Dystonia Coalition; Dystonia Medical
treatment, DBS cannot be recommended for routine use in Research Foundation; F. Hoffmann-La Roche Ltd; Huntington Study
Group; Kyowa Haako Kirin Pharma, Inc.; Medtronic Neuromodula-
the treatment of TD. tion; Merz Pharmaceuticals; Michael J. Fox Foundation for Parkinson
Research; National Institutes of Health; Neurocrine Biosciences;
8.3 Electroconvulsive Therapy NeuroDerm Ltd; Parkinson’s Foundation; Nuvelution; Parkinson
Study Group; Pfizer Inc.; Prothena Biosciences Inc.; Psyadon Phar-
maceuticals, Inc.; Revance Therapeutics, Inc.; Sangamo BioSciences,
Finally, a few case reports and retrospective series report Inc.; St. Jude Medical; and Teva Pharmaceutical Industries Ltd. Dr.
improvement of TD with electroconvulsive therapy, Jankovic has served as a consultant or as an advisory committee
although no firm conclusions can be made based on those member for Adamas Pharmaceuticals, Inc.; Allergan, Inc.; Merz
reports [144–148]. Pharmaceuticals; Pfizer Inc.; Prothena Biosciences; Revance Thera-
peutics, Inc.; and Teva Pharmaceutical Industries Ltd. Dr. Jankovic
Table 3 provides a list of active phase II/III studies. has also received royalties or other payments from Cambridge;
Elsevier; Future Science Group; Hodder Arnold; Medlink: Neurol-
ogy; Lippincott Williams and Wilkins; and Wiley-Blackwell.
9 Conclusion

Contrary to expectations, TD has not vanished despite the References


introduction of SGAs and TGAs, which are marketed as
1. Faurbye A, Rasch PJ, Petersen PB, Brandborg G, Pakkenberg H.
having a lower risk of TD than FGAs, although this Neurological symptoms in pharmacotherapy of psychoses. Acta
remains a contentious topic. Instead, changing prescription Psychiatr Scand. 1964;40:10–27.
practices and increased off-label prescription of these drugs 2. Waln O, Jankovic J. An update on tardive dyskinesia: from
have placed more patients than ever at risk of this poten- phenomenology to treatment. Tremor Other Hyperkinetic Mov.
2013;3:1–11.
tially dangerous and disabling condition. Although signif- 3. Vijayakumar D, Jankovic J. Drug-induced dyskinesia, part 2:
icant insights have been gained into the pathophysiology of Treatment of tardive dyskinesia. Drugs. 2016;76:779–87.
TD, much is yet unknown, and there are significant gaps in 4. Savitt D, Jankovic J. Tardive syndromes. J Neurol Sci. 2018 (in
our understanding of risk stratification and optimal man- press).
agement. Prevention by judicious use of DRBAs is
N. Niemann, J. Jankovic

5. Frei K, Truong DD, Fahn S, Jankovic J, Hauser RA. The Movement disorders in childhood. 2nd ed. London: Academic
nosology of tardive syndromes. J Neurol Sci. 2018. https://doi. Press; 2016. p. 337–407.
org/10.1016/j.jns.2018.02.008. 27. Thenganatt MA, Jankovic J. Recent advances in understanding
6. Carbon M, Hsieh CH, Kane JM, Correll CU. Tardive dyskinesia and managing Tourette syndrome [version 1; referees: 3
prevalence in the period of second-generation antipsychotic use: approved]. F1000Research 2016, 5(F1000 Faculty Rev):152.
a meta-analysis. J Clin Psychiatry. 2017;78:e264–78. https://doi.org/10.12688/f1000research.7424.1.
7. Correll CU, Kane JM, Citrome LL. Epidemiology, prevention, 28. Baizabal-Carvallo JF, Jankovic J. Movement disorders in
and assessment of tardive dyskinesia and advances in treatment. autoimmune diseases. Mov Disord. 2012;27:935–46.
J Clin Psychiatry. 2017;78:1136–47. 29. Benazzi F. Rapid onset of tardive dyskinesia in Huntington
8. Pillay J, Boylan K, Carrey N, Newton A, Vandermeer B, Nuspl disease with olanzapine. J Clin Psychopharmacol.
M, et al. First- and second-generation antipsychotics in children 2002;22:438–9.
and young adults: Systematic review update [Internet]. Agency 30. Singh SK, Jankovic J. Tardive dystonia in patients with Tour-
for Healthcare Research and Quality (US); 2017 [cited 2018 Jan ette’s syndrome. Mov Disord. 1988;3:274–80.
20]. Available from: http://www.ncbi.nlm.nih.gov/pubmed/ 31. Kane JM, Woerner M, Lieberman J. Tardive dyskinesia:
28749632. prevalence, incidence, and risk factors. J Clin Psychopharmacol.
9. McDonagh M, Peterson K, Carson S, Fu R, Thakurta S. Drug 1988;8:52S–6S.
class review: Atypical antipsychotic drugs: final update 3 report 32. Yassa R, Jeste DV. Gender differences in tardive dyskinesia: a
[Internet]. Portland (OR): Oregon Health & Science University; critical review of the literature. Schizophr Bull.
2010. Available from: https://www-ncbi-nlm-nih-gov. 1992;18:701–15.
ezproxyhost.library.tmc.edu/books/NBK50583/. 33. Gardos G, Casey DE, Cole JO, Perenyi A, Kocsis E, Arato M,
10. Kwei K, Frucht S. Acute presentation of nonmotor symptoms in et al. Ten-year outcome of tardive dyskinesia. Am J Psychiatry.
parkinson’s disease. Int Rev Neurobiol. 2017;134:973–86. 1994;151:836–41.
11. Patel N, Jankovic J, Hallett M. Sensory aspects of movement 34. Glazer WM, Morgenstern H, Doucette JT. Predicting the long-
disorders. Lancet Neurol. 2014;13:100–12. term risk of tardive dyskinesia in outpatients maintained on
12. Caroff SN, Campbell EC. Drug-induced extrapyramidal syn- neuroleptic medications. J Clin Psychiatry. 1993;54:133–9.
dromes: implications for contemporary practice. Psychiatr Clin 35. Chakos MH, Alvir JM, Woerner MG, Koreen A, Geisler S,
North Am. 2016;39:391–411. Mayerhoff D, et al. Incidence and correlates of tardive dyski-
13. Obeso JA. The movement disorders journal 2016 and onward. nesia in first episode of schizophrenia. Arch Gen Psychiatry.
Mov Disord. 2016;31:1–2. 1996;53:313–9.
14. Walker RH. Thoughts on selected movement disorder termi- 36. Jeste DV, Caligiuri MP, Paulsen JS, Heaton RK, Lacro JP,
nology and a plea for clarity. Tremor Other Hyperkinet Mov (N Harris MJ, et al. Risk of tardive dyskinesia in older patients. A
Y). 2013;3. https://doi.org/10.7916/D8R49PG6. prospective longitudinal study of 266 outpatients. Arch Gen
15. Medication-induced movement disorders and other adverse Psychiatry. 1995;52:756–65.
effects of medication. Diagnostic and Statistical Manual of 37. Woerner MG, Alvir JMJ, Saltz BL, Lieberman JA, Kane JM.
Mental Disorders. 5th ed. American Psychiatric Association; Prospective study of tardive dyskinesia in the elderly: rates and
2013. p. 709–14. risk factors. Am J Psychiatry. 1998;155:1521–8.
16. Emsley R, Niehaus DJH, Oosthuizen PP, Koen L, Chiliza B, 38. Correll CU, Leucht S, Kane JM. Lower risk for tardive dyski-
Fincham D. Subjective awareness of tardive dyskinesia and nesia associated with second-generation antipsychotics: a sys-
insight in schizophrenia. Eur Psychiatry. 2011;26:293–6. tematic review of 1-year studies. Am J Psychiatry.
17. Cloud LJ, Zutshi D, Factor SA. Tardive dyskinesia: therapeutic 2004;161:414–25.
options for an increasingly common disorder. Neurotherapeu- 39. O’Brien A. Comparing the risk of tardive dyskinesia in older
tics. 2014;11:166–76. adults with first-generation and second-generation antipsy-
18. Mehta SH, Morgan JC, Sethi KD. Drug-induced movement chotics: a systematic review and meta-analysis. Int J Geriatr
disorders. Neurol Clin. 2015;33:153–74. Psychiatry. 2016;31:683–93.
19. Lorberboym M, Treves TA, Melamed E, Lampl Y, Hellmann M, 40. Leucht S, Cipriani A, Spineli L, Mavridis D, Örey D, Richter F,
Djaldetti R. [123I]-FP/CIT SPECT imaging for distinguishing et al. Comparative efficacy and tolerability of 15 antipsychotic
drug-induced parkinsonism from Parkinson’s disease. Mov drugs in schizophrenia: a multiple-treatments meta-analysis.
Disord. 2006;21:510–4. Lancet. 2013;382:951–62.
20. Lo Y-C, Peng Y-C. Amisulpride withdrawal dyskinesia: a case 41. Arnt J, Skarsfeldt T. Do novel antipsychotics have similar
report. Ann Gen Psychiatry. 2017;16:25. pharmacological characteristics? A review of the evidence.
21. Blanchet PJ, Popovici R, Guitard F, Rompré PH, Lamarche C, Neuropsychopharmacology. 1998;18:63–101.
Lavigne GJ. Pain and denture condition in edentulous 42. Correll CU, Schenk EM. Tardive dyskinesia and new antipsy-
orodyskinesia: comparisons with tardive dyskinesia and control chotics. Curr Opin Psychiatry. 2008;21:151–6.
subjects. Mov Disord. 2008;23:1837–42. 43. Miller DD, Caroff SN, Davis SM, Rosenheck RA, McEvoy JP,
22. Shin H-Y, Yoon WT, Lee WY. Anticholinergic agents can Saltz BL, et al. Extrapyramidal side-effects of antipsychotics in
induce oromandibular dyskinesia. J Mov Disord. 2009;2:69–71. a randomised trial. Br J Psychiatry. 2008;193:279–88.
23. Vijayakumar D, Jankovic J. Drug-induced dyskinesia, part 1: 44. Peluso MJ, Lewis SW, Barnes TRE, Jones PB. Extrapyramidal
treatment of levodopa-induced dyskinesia. Drugs. motor side-effects of first- and second-generation antipsychotic
2016;76:759–77. drugs. Br J Psychiatry. 2012;200:387–92.
24. Fekete R, Jankovic J. Upper facial chorea in Huntington disease. 45. Hazari N, Kate N, Grover S. Clozapine and tardive movement
J Clin Mov Disord. 2014;1:7. disorders: a review. Asian J Psychiatr. 2013;6:439–51.
25. Cardoso F. Differential diagnosis of Huntington’s disease: what 46. Li C-R, Chung Y-C, Park T-W, Yang J-C, Kim K-W, Lee K-H,
the clinician should know. Neurodegener Dis Manag. et al. Clozapine-induced tardive dyskinesia in schizophrenic
2014;4:67–72. patients taking clozapine as a first-line antipsychotic drug.
26. Singer HS, Mink JW, Gilbert DL, Jankovic J. Inherited meta- World J Biol Psychiatry. 2009;10:919–24.
bolic disorders with associated movement abnormalities. In:
Tardive Dyskinesia

47. Davé M. Clozapine-related tardive dyskinesia. Biol Psychiatry. metoclopramide. Nat Clin Pract Gastroenterol Hepatol.
1994;35:886–7. 2006;3:138–48.
48. Molho ES, Factor SA. Possible tardive dystonia resulting from 66. Mejia NI, Jankovic J. Metoclopramide-induced tardive dyski-
clozapine therapy. Mov Disord. 1999;14:873–4. nesia in an infant. Mov Disord. 2005;20:86–9.
49. Peña MS, Yaltho TC, Jankovic J. Tardive dyskinesia and other 67. Loonen AJM, Ivanova SA. New insights into the mechanism of
movement disorders secondary to aripiprazole. Mov Disord. drug-induced dyskinesia. CNS Spectr. 2013;18:15–20.
2011;26:147–52. 68. Stahl SM. Neuronal traffic signals in tardive dyskinesia: not
50. Paulson GW, Rizvi CA, Crane GE. Tardive dyskinesia as a enough ‘‘stop’’ in the motor striatum. CNS Spectr.
possible sequel of long-term therapy with phenothiazines. Clin 2017;22:427–34.
Pediatr (Phila). 1975;14:953–5. 69. Turrone P, Remington G, Kapur S, Nobrega JN. The relation-
51. Campbell M, Armenteros JL, Malone RP, Adams PB, Eisenberg ship between dopamine D2 receptor occupancy and the vacuous
ZW, Overall JE. Neuroleptic-related dyskinesias in autistic chewing movement syndrome in rats. Psychopharmacology.
children: a prospective, longitudinal study. J Am Acad Child 2003;165:166–71.
Adolesc Psychiatry. 1997;36:835–43. 70. Silvestri S, Seeman MV, Negrete JC, Houle S, Shammi CM,
52. McAndrew JB, Case Q, Treffert DA. Effects of prolonged Remington GJ, et al. Increased dopamine D2 receptor binding
phenothiazine intake on psychotic and other hospitalized chil- after long-term treatment with antipsychotics in humans: a
dren. J Autism Child Schizophr. 1972;2:75–91. clinical PET study. Psychopharmacology. 2000;152:174–80.
53. Mejia NI, Jankovic J. Tardive dyskinesia and withdrawal 71. Strange PG. Antipsychotic drugs: importance of dopamine
emergent syndrome in children. Expert Rev Neurother. receptors for mechanisms of therapeutic actions and side effects.
2010;10:893–901. Pharmacol Rev. 2001;53:119–33.
54. Wonodi I, Reeves G, Carmichael D, Verovsky I, Avila MT, 72. Seeman P. Dopamine D2 receptors as treatment targets in
Elliott A, et al. Tardive dyskinesia in children treated with schizophrenia. Clin Schizophr Relat Psychoses. 2010;4:56–73.
atypical antipsychotic medications. Mov Disord. 73. Shi J, Tan YL, Wang ZR, An HM, Li J, Wang YC, et al. Ginkgo
2007;22:1777–82. biloba and vitamin E ameliorate haloperidol-induced vacuous
55. Garcia-Amador M, Merchán-Naranjo J, Tapia C, Moreno C, chewingmovement and brain-derived neurotrophic factor
Castro-Fornieles J, Baeza I, et al. Neurological adverse effects expression in a rat tardive dyskinesia model. Pharmacol Bio-
of antipsychotics in children and adolescents. J Clin Psy- chem Behav. 2016;148:53–8.
chopharmacol. 2015;35:686–93. 74. Teo JT, Edwards MJ, Bhatia K. Tardive dyskinesia is caused by
56. Hugenholtz GWK, Heerdink ER, Stolker JJ, Meijer WEE, maladaptive synaptic plasticity: a hypothesis. Mov Disord.
Egberts ACG, Nolen WA. Haloperidol dose when used as active 2012;27:1205–15.
comparator in randomized controlled trials with atypical 75. Lanning RK, Zai CC, Müller DJ. Pharmacogenetics of tardive
antipsychotics in schizophrenia: comparison with officially dyskinesia: an updated review of the literature. Pharmacoge-
recommended doses. J Clin Psychiatry. 2006;67:897–903. nomics. 2016;17:1339–51.
57. Rakesh G, Muzyk A, Szabo ST, Gupta S, Pae C-U, Masand P. 76. Guy W. AIMS: ECDEU assessment manual for psychophar-
Tardive dyskinesia: 21st century may bring new treatments to a macology. Washington, DC: Government Printing Office; 1976.
forgotten disorder. Ann Clin Psychiatry. 2017;29:108–19. p. 534–7.
58. Achalia RM, Chaturvedi SK, Desai G, Rao GN, Prakash O. 77. Munetz MR, Benjamin S. How to examine patients using the
Prevalence and risk factors associated with tardive dyskinesia Abnormal Involuntary Movement Scale. Hosp Community
among Indian patients with schizophrenia. Asian J Psychiatr. Psychiatry. 1988;39:1172–7.
2014;9:31–5. 78. Bark N, Florida D, Gera N, Varardi R, Harghel L, Adlington K.
59. van Harten PN, Hoek HW, Matroos GE, Koeter M, Kahn RS. Evaluation of the routine clinical use of the Brief Psychiatric
Intermittent neuroleptic treatment and risk for tardive dyskine- Rating Scale (BPRS) and the Abnormal Involuntary Movement
sia: Curaçao Extrapyramidal Syndromes Study III. Am J Psy- Scale (AIMS). J Psychiatr Pract. 2011;17:300–3.
chiatry. 1998;155:565–7. 79. Suzuki T. Which rating scales are regarded as ‘‘the standard’’ in
60. Xiang Y-T, Wang C-Y, Si T-M, Lee EHM, He Y-L, Ungvari clinical trials for schizophrenia? A critical review. Psy-
GS, et al. Sex differences in use of psychotropic drugs and drug- chopharmacol Bull. 2011;44:18–31.
induced side effects in schizophrenia patients: findings of the 80. Lane RD, Glazer WM, Hansen TE, Berman WH, Kramer SI.
research on Asia psychotropic prescription (REAP) studies. Aust Assessment of tardive dyskinesia using the Abnormal Involun-
N Zeal J Psychiatry. 2011;45:193–8. tary Movement Scale. J Nerv Ment Dis. 1985;173:353–7.
61. Zhang XY, Chen DC, Qi LY, Wang F, Xiu MH, Chen S, et al. 81. Gerlach J, Korsgaard S, Clemmesen P, Lauersen AM, Magelund
Gender differences in the prevalence, risk and clinical correlates G, Noring U, et al. The St. Hans Rating Scale for extrapyramidal
of tardive dyskinesia in Chinese schizophrenia. Psychopharma- syndromes: reliability and validity. Acta Psychiatr Scand.
cology. 2009;205:647–54. 1993;87:244–52.
62. Tenback DE, van Harten PN, van Os J. Non-therapeutic risk 82. Domino ME, Swartz MS. Who are the new users of antipsy-
factors for onset of tardive dyskinesia in schizophrenia: a meta- chotic medications? Psychiatr Serv. 2008;59:507–14.
analysis. Mov Disord. 2009;24:2309–15. 83. Huskamp HA, Horvitz-Lennon M, Berndt ER, Normand S-LT,
63. Wijemanne S, Jankovic J, Evans RW. Movement disorders from Donohue JM. Patterns of antipsychotic prescribing by physi-
the use of metoclopramide and other antiemetics in the treatment cians to young children. Psychiatr Serv. 2016;67:1307–14.
of migraine. Headache J Head Face Pain. 2016;56:153–61. 84. Hálfdánarson Ó, Zoëga H, Aagaard L, Bernardo M, Brandt L,
64. Kenney C, Hunter C, Davidson A, Jankovic J. Metoclopramide, Fusté AC, et al. International trends in antipsychotic use: a study
an increasingly recognized cause of tardive dyskinesia. J Clin in 16 countries, 2005–2014. Eur Neuropsychopharmacol.
Pharmacol. 2008;48:379–84. 2017;27:1064–76.
65. Pasricha PJ, Pehlivanov N, Sugumar A, Jankovic J. Drug 85. Seeberger LC, Hauser RA. Valbenazine for the treatment of
Insight: from disturbed motility to disordered movement—a tardive dyskinesia. Expert Opin Pharmacother.
review of the clinical benefits and medicolegal risks of 2017;18:1279–87.
N. Niemann, J. Jankovic

86. Ehrenpreis ED, Deepak P, Sifuentes H, Devi R, Du H, Leikin 104. Austedo (deutetrabenazine) Tablets, Medical Review(s). [cited
JB. The metoclopramide black box warning for tardive dyski- 2018 Jan 4]; Available from: https://www.accessdata.fda.gov/
nesia: effect on clinical practice, adverse event reporting, and drugsatfda_docs/nda/2017/208082Orig1s000TOC.cfm.
prescription drug lawsuits. Am J Gastroenterol. 105. Grigoriadis DE, Smith E, Hoare SRJ, Madan A, Bozigian H.
2013;108:866–72. Pharmacologic characterization of valbenazine (NBI-98854) and
87. Lehman AF, Lieberman JA, Dixon LB, McGlashan TH, Miller its metabolites. J Pharmacol Exp Ther. 2017;361:454–61.
AL, Perkins DO, et al. Practice guideline for the treatment of 106. Mehanna R, Hunter C, Davidson A, Jimenez-Shahed J, Jankovic
patients with schizophrenia, second edition. Am J Psychiatry. J. Analysis of CYP2D6 genotype and response to tetrabenazine.
2004;161:1–56. Mov Disord. 2013;28:210–5.
88. Viguera AC, Baldessarini RJ, Hegarty JD, van Kammen DP, 107. Teva Pharmaceuticals. Austedo (deutetrabenazine), Highlights
Tohen M. Clinical risk following abrupt and gradual withdrawal of Prescribing Information. [cited 2018 Jan 4]; Available from:
of maintenance neuroleptic treatment. Arch Gen Psychiatry. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/
1997;54:49–55. 208082s000lbl.pdf.
89. Bhidayasiri R, Fahn S, Weiner WJ, Gronseth GS, Sullivan KL, 108. Neurocrine Biosciences. Ingrezza (valbenazine), Highlights of
Zesiewicz TA, et al. Evidence-based guideline: treatment of Prescribing Information. [cited 2018 Jan 4]; Available from:
tardive syndromes: report of the Guideline Development Sub- https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/
committee of the American Academy of Neurology. Neurology. 209241lbl.pdf.
2013;81:463–9. 109. Ondo WG, Hanna PA, Jankovic J. Tetrabenazine treatment for
90. Sahli ZT, Tarazi FI. Pimavanserin: novel pharmacotherapy for tardive dyskinesia: assessment by randomized videotape proto-
Parkinson’s disease psychosis. Expert Opin Drug Discov. col. Am J Psychiatry. 1999;156:1279–81.
2018;13:103–10. 110. Kazamatsuri H, Chien C, Cole JO. Treatment of tardive dyski-
91. Glazer WM, Morgenstern H, Schooler N, Berkman CS, Moore nesia. I. Clinical efficacy of a dopamine-depleting agent, tetra-
DC. Predictors of improvement in tardive dyskinesia following benazine. Arch Gen Psychiatry. 1972;27:95–9.
discontinuation of neuroleptic medication. Br J Psychiatry. 111. Kenney C, Hunter C, Jankovic J. Long-term tolerability of
1990;157:585–92. tetrabenazine in the treatment of hyperkinetic movement disor-
92. Zutshi D, Cloud LJ, Factor SA. Tardive syndromes are rarely ders. Mov Disord. 2007;22:193–7.
reversible after discontinuing dopamine receptor blocking 112. Jankovic J, Beach J. Long-term effects of tetrabenazine in
agents: experience from a university-based movement disorder hyperkinetic movement disorders. Neurology. 1997;48:358–62.
clinic. Tremor Other Hyperkinetic Mov (N Y). 2014;4:266. 113. Kenney C, Hunter C, Mejia N, Jankovic J. Is history of
93. Modestin J, Wehrli MV, Stephan PL, Agarwalla P. Evolution of depression a contraindication to treatment with tetrabenazine?
neuroleptic-induced extrapyramidal syndromes under long-term Clin Neuropharmacol. 2006;29:259–64.
neuroleptic treatment. Schizophr Res. 2008;100:97–107. 114. Gant TG. Using deuterium in drug discovery: leaving the label
94. Fernandez HH, Krupp B, Friedman JH. The course of tardive in the drug. J Med Chem. 2014;57:3595–611.
dyskinesia and parkinsonism in psychiatric inpatients: 14-year 115. Stamler D, Bradbury M, Brown F. The pharmacokinetics and
follow-up. Neurology. 2001;56:805–7. safety of deuterated-tetrabenazine. Neurology.
95. Bhidayasiri R, Jitkritsadakul O, Friedman JH, Fahn S. Updating 2013;80:P07.210.
the recommendations for treatment of tardive syndromes: a 116. Fernandez HH, Factor SA, Hauser RA, Jimenez-Shahed J, Ondo
systematic review of new evidence and practical treatment WG, Jarskog LF, et al. Randomized controlled trial of
algorithm. J Neurol Sci. 2018. https://doi.org/10.1016/j.jns.2018. deutetrabenazine for tardive dyskinesia. Neurology.
02.010. 2017;88:2003–10.
96. Lawal HO, Krantz DE. SLC18: vesicular neurotransmitter 117. Anderson KE, Stamler D, Davis MD, Factor SA, Hauser RA,
transporters for monoamines and acetylcholine. Mol Aspects Isojärvi J, et al. Deutetrabenazine for treatment of involuntary
Med. 2013;34:360–72. movements in patients with tardive dyskinesia (AIM-TD): a
97. Jankovic J. Progress in Parkinson disease and other movement double-blind, randomised, placebo-controlled, phase 3 trial.
disorders. Nat Rev Neurol. 2017;13:76–8. Lancet Psychiatry. 2017;4:595–604.
98. Benarroch EE. Monoamine transporters: structure, regulation, 118. Citrome L. Deutetrabenazine for tardive dyskinesia: a system-
and clinical implications. Neurology. 2013;81:761–8. atic review of the efficacy and safety profile for this newly
99. Jankovic J. Dopamine depleters in the treatment of hyperkinetic approved novel medication-What is the number needed to treat,
movement disorders. Expert Opin Pharmacother. number needed to harm and likelihood to be helped or harmed?
2016;17:2461–70. Int J Clin Pract. 2017;71:e13030.
100. LeWitt PA. Tardive dyskinesia caused by tetrabenazine. Clin 119. ClinicalTrials.gov. Reducing involuntary movements in tardive
Neuropharmacol. 2013;36:92–3. dyskinesia. NCT02198794. [Internet]. Auspex Pharm. Inc. 2017
101. Caroff SN, Davis VG, Miller DD, Davis SM, Rosenheck RA, [cited 2017 Nov 6]. Available from: https://clinicaltrials.gov/
McEvoy JP, et al. Treatment outcomes of patients with tardive ct2/show/NCT02198794.
dyskinesia and chronic schizophrenia. J Clin Psychiatry. 120. Anderson K, Stamler D, Davis M, Factor S, Hauser R, Isojärvi J,
2011;72:295–303. et al. Long-term safety of deutetrabenazine for the treatment of
102. Stahl SM. Mechanism of action of vesicular monoamine trans- tardive dyskinesia: results from an open-label, long-term study
porter 2 (VMAT2) inhibitors in tardive dyskinesia: reducing [abstract]. Mov Disord. 2017;32 (suppl 2). [Internet]. [cited
dopamine leads to less ‘‘go’’ and more ‘‘stop’’ from the motor 2018 Jan 4]. Available from: http://www.mdsabstracts.org/
striatum for robust therapeutic effects. CNS Spectr. abstract/long-term-safety-of-deutetrabenazine-for-the-treatment-
2017;23:1–6. of-tardive-dyskinesia-results-from-an-open-label-long-term-
103. Lundbeck. Xenazine (tetrabenazine), Highlights of Prescribing study/.
Information. [cited 2018 Jan 4]; Available from: https://www. 121. Frank S, Stamler D, Kayson E, Claassen DO, Colcher A, Davis
accessdata.fda.gov/drugsatfda_docs/label/2015/021894s010lbl. C, et al. Safety of converting from tetrabenazine to deutetra-
pdf. benazine for the treatment of chorea. JAMA Neurol.
2017;74:977–82.
Tardive Dyskinesia

122. Bashir H, Jankovic J. Treatment options for chorea. Expert Rev 135. Zhang W-F, Tan Y-L, Zhang X-Y, Chan RCK, Wu H-R, Zhou
Neurother. 2018;18:51–63. D-F. Extract of Ginkgo biloba treatment for tardive dyskinesia
123. Jankovic J, Jimenez-Shahed J, Budman C, Coffey B, Murphy T, in schizophrenia: a randomized, double-blind, placebo-con-
Shprecher D, et al. Deutetrabenazine in tics associated with trolled trial. J Clin Psychiatry. 2011;72:615–21.
tourette syndrome. Tremor Other Hyperkinetic Mov (N Y). 136. Zhang XY, Zhang W-F, Zhou D-F, Chen DC, Xiu MH, Wu
2016;6:422. H-R, et al. Brain-derived neurotrophic factor levels and its
124. Müller T. Valbenazine for the treatment of tardive dyskinesia. Val66Met gene polymorphism predict tardive dyskinesia treat-
Expert Rev Neurother. 2017;17:1135–44. ment response to Ginkgo biloba. Biol Psychiatry.
125. Caroff SN, Aggarwal S, Yonan C. Treatment of tardive dyski- 2012;72:700–6.
nesia with tetrabenazine or valbenazine: a systematic review. 137. Desmarais JE, Beauclair L, Annable L, Bélanger M-C, Koli-
J Comp Eff Res. 2017;6:422. vakis TT, Margolese HC. Effects of discontinuing anticholin-
126. O’Brien CF, Jimenez R, Hauser RA, Factor SA, Burke J, Mandri ergic treatment on movement disorders, cognition and
D, et al. NBI-98854, a selective monoamine transport inhibitor psychopathology in patients with schizophrenia. Ther Adv
for the treatment of tardive dyskinesia: A randomized, double- Psychopharmacol. 2014;4:257–67.
blind, placebo-controlled study. Mov Disord. 2015;30:1681–7. 138. Vinogradov S, Fisher M, Warm H, Holland C, Kirshner MA,
127. Hauser RA, Factor SA, Marder SR, Knesevich MA, Ramirez Pollock BG. The cognitive cost of anticholinergic burden:
PM, Jimenez R, et al. KINECT 3: a phase 3 randomized, double- Decreased response to cognitive training in schizophrenia. Am J
blind, placebo-controlled trial of valbenazine for tardive dyski- Psychiatry. 2009;166:1055–62.
nesia. Am J Psychiatry. 2017;174:476–84. 139. Aquino CCH, Lang AE. Tardive dyskinesia syndromes: current
128. Citrome L. Valbenazine for tardive dyskinesia: A systematic concepts. Parkinsonism Relat Disord. 2014;20:S113–7.
review of the efficacy and safety profile for this newly approved 140. Waln O, Jankovic J. Zolpidem improves tardive dyskinesia and
novel medication-What is the number needed to treat, number akathisia. Mov Disord. 2013;28:1748–9.
needed to harm and likelihood to be helped or harmed? Int J Clin 141. Jankovic J. An update on new and unique uses of botulinum
Pract. 2017;71:e12964. toxin in movement disorders. Toxicon. 2017. https://doi.org/10.
129. Thai-Cuarto D, O’Brien CF, Jimenez R, Liang GS, Burke J. 1016/j.toxicon.2017.09.003.
Cardiovascular profile of valbenazine: analysis of pooled data 142. Tan EK, Jankovic J. Tardive and idiopathic oromandibular
from three randomized, double-blind, placebo-controlled trials. dystonia: a clinical comparison. J Neurol Neurosurg Psychiatry.
Drug Saf. 2017. https://doi.org/10.1007/s40264-017-0623-1. 2000;68:186–90.
130. Factor SA, Remington G, Comella CL, Correll CU, Burke J, 143. Morigaki R, Mure H, Kaji R, Nagahiro S, Goto S. Therapeutic
Jimenez R, et al. The effects of valbenazine in participants with perspective on tardive syndrome with special reference to deep
tardive dyskinesia: results of the 1-year KINECT 3 extension brain stimulation. Front Psychiatry. 2016;7:207.
study. J Clin Psychiatry. 2017;78:1344–50. 144. Sun B, Chen S, Zhan S, Le W, Krahl SE. Subthalamic nucleus
131. Angus S, Sugars J, Boltezar R, Koskewich S, Schneider NM. A stimulation for primary dystonia and tardive dystonia. Acta
controlled trial of amantadine hydrochloride and neuroleptics in Neurochir Suppl. 2007;97:207–14.
the treatment of tardive dyskinesia. J Clin Psychopharmacol. 145. Meng D-W, Liu H-G, Yang A-C, Zhang K, Zhang J-G. Long-
1997;17:88–91. term effects of subthalamic nucleus deep brain stimulation in
132. Pappa S, Tsouli S, Apostolou G, Mavreas V, Konitsiotis S. tardive dystonia. Chin Med J (Engl). 2016;129:1257–8.
Effects of amantadine on tardive dyskinesia: a randomized, 146. Yasui-Furukori N, Nakamura K, Katagai H, Kaneko S. The
double-blind, placebo-controlled study. Clin Neuropharmacol. effects of electroconvulsive therapy on tardive dystonia or
2010;33:271–5. dyskinesia induced by psychotropic medication: a retrospective
133. Thaker GK, Nguyen JA, Strauss ME, Jacobson R, Kaup BA, study. Neuropsychiatr Dis. Treat. 2014;10:1209–12.
Tamminga CA. Clonazepam treatment of tardive dyskinesia: a 147. Peng L-Y, Lee Y, Lin P-Y. Electroconvulsive therapy for a
practical GABAmimetic strategy. Am J Psychiatry. patient with persistent tardive dyskinesia. J ECT.
1990;147:445–51. 2013;29:e52–4.
134. Zheng W, Xiang Y-Q, Ng C, Ungvari G, Chiu H, Xiang Y-T. 148. Manteghi A, Hojjat SK, Javanbakht A. Remission of tardive
Extract of Ginkgo biloba for tardive dyskinesia: meta-analysis dystonia with electroconvulsive therapy. J Clin Psychopharma-
of randomized controlled trials. Pharmacopsychiatry. col. 2009;29:314–5.
2016;49:107–11.

Anda mungkin juga menyukai