Vous êtes sur la page 1sur 13

CNS Drugs 2011; 25 (12): 1073-1085

ADIS DRUG PROFILE 1172-7047/11/0012-1073/$49.95/0

ª 2011 Adis Data Information BV. All rights reserved.

Tetrabenazine
For Chorea Associated with Huntington’s Disease
Lesley J. Scott
Adis, a Wolters Kluwer Business, Auckland, New Zealand

Contents
Abstract. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1073
1. Pharmacodynamic Properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1075
2. Pharmacokinetic Properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1076
3. Therapeutic Efficacy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1078
4. Tolerability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1081
5. Dosage and Administration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1083
6. Tetrabenazine: Current Status For Chorea Associated with Huntington’s Disease . . . . . . . . . . . . . . . 1083

Abstract Features and properties of tetrabenazine (TBZ;


Xenazine®)
Oral tetrabenazine is currently the only drug
approved by the US FDA for the treatment of Indication
chorea associated with Huntington’s disease (HD). For the treatment of chorea associated with Huntington’s
disease
Although the precise antichorea mechanism of ac-
Mechanism of action
tion is unknown, it most likely involves reversible
Most likely involves reversible depletion of monoamines,
depletion of monoamines, particularly dopamine, especially dopamine, from presynaptic nerve terminals via
from presynaptic terminals via inhibition of human inhibition of human vesicular monoamine transporter type 2
vesicular monoamine transporter type 2. Pharmacokinetic profile (values are for the active
In a 12-week, double-blind, placebo-controlled metabolite α-dihydrotetrabenazine [DHTBZ])
trial conducted in the US in patients with HD, oral Time to attain maximum 1–1.5 h
tetrabenazine (£100 mg/day; n = 54) was significantly plasma concentration
(p = 0.0001) more efficacious than placebo (n = 30) at Metabolism TBZ is generally undetectable
in plasma, as it undergoes
improving adjusted mean Unified HD Rating Scale rapid and extensive first-pass
(UHDRS) total maximum chorea scores (reduced metabolism by carbonyl
from baseline by 5 vs 1.5) [primary endpoint]. reductase to α-DHTBZ (active)
and β-DHTBZ (inactive)
After 12 weeks, improvements in UHDRS total α-DHTBZ and β-DHTBZ are
maximum chorea scores of >3 were achieved by primarily metabolized by
significantly (p < 0.0001) more patients in the tetra- cytochrome P450 2D6
benazine group than in the placebo group. Elimination Primarily via renal route
The antichorea efficacy of tetrabenazine was main- Elimination half-life 7 h [α-DHTBZ]; 5 h [β-DHTBZ]
tained in an 80-week extension study (n = 75), with the Dosage and administration (individualized to optimal
adjusted mean UHDRS total maximum chorea score dosage; titrated to a tolerated dosage that reduces chorea)
significantly (p < 0.001) reduced from baseline (score Initial dosage 12.5 mg taken once daily in the
morning
of 14.9) by 4.6 points (primary outcome).
In the 12-week trial and 80-week extension study, Titration dosage 25 mg/day as two divided
doses, then 37.5–50 mg/day as
treatment-emergent adverse events in the tetra- three divided doses if required
benazine group mainly occurred during the dosage- Maximum dosage 100 mg/day as three divided
titration phase, a period during which the dosage doses
was individually optimized. Most of these events Route of administration Oral
were mild to moderate and were manageable with Most common treatment-emergent adverse events
dosage adjustments or discontinuation of study drug. (incidence ≥15% and greater than in placebo group)
Drowsiness/somnolence, insomnia, fatigue, fall, depression,
agitation, anxiety
1074 Scott

O consequence of neuronal degeneration and atro-


CH3
phy, particularly in the striatum and cortex, that
results from the presence of an abnormal hun-
H3CO CH3 tingtin protein.[1,2,4-6] At the time of diagnosis,
N marked striatal and cortical atrophy are already
likely to be present, with brain abnormalities
H3CO occurring years before the clinical symptoms
Chemical structure of tetrabenazine manifest.[1]
As with many chronic terminal illnesses, HD is
associated with an increased risk of depression/
Huntington’s disease (HD) is an autosomal depressed mood.[7-9] However, depression often
dominant neurodegenerative disorder, with an esti- occurs several years before HD manifests, sug-
mated prevalence in the Western world of 4–10 gesting that it may be an integral component of
per 100 000 individuals.[1-3] In the US, an esti- HD.[7-9] As reviewed by van Duijn et al.,[9] the
mated 25 000–30 000 individuals have HD, and prevalence of depressed mood is relatively high
an estimated 150 000–250 000 individuals are at in patients with HD (ranging from 33% to 69%).
risk for the disease.[4] HD occurs in all races and For example, in a recent large cross-sectional
ethnicities,[1] and men and women are affected study in patients with HD (n = 2835), approximately
equally.[4] The typical age of onset of clinical symp- 40% of patients had symptoms of depression
toms is the third or fourth decade of life, although and more than half had sought treatment for
onset may occur at any time from infancy to the depression.[7]
ninth decade of life.[1,2,4-6] Death usually occurs Suicidal ideation and suicide are also common
within 10–20 years of disease onset.[1,2,4-6] features associated with HD, with reported sui-
HD is caused by amplification of the CAG cide rates that are 5–10 times greater than those
(glutamine) trinucleotide in the huntingtin gene, reported in the general population.[7,10] Of inter-
resulting in an abnormally long polyglutamate est, in patients with HD, suicidality may be asso-
repeat in the huntingtin protein.[1-3,5] The number ciated with impulsiveness, obsessive-compulsive
of repeats in the CAG expansion is inversely cor- behaviours and/or a variety of socioeconomic
related with the age at clinical onset of the dis- factors rather than correlating with the severity of
ease, with more repeats associated with earlier depression.[4] Studies show that suicide attempts
onset of HD (‡50 repeats is associated with ju- are not uncommon (occurring in up to 25%
venile onset).[1,2,6] Environmental influences and of patients) in patients with HD.[7,10] Suicidal
modifying genes are also likely to be involved in ideation is generally greatest around the time of
determining the age of onset.[2] The number of diagnosis of manifest HD and again when
CAG repeats is also indicative of the likelihood of patients start to lose their functional abilities and
developing HD: <36 repeats is considered normal independence.[11]
(typical range is 17–20), 36–39 repeats is con- There are currently no established disease-
sidered abnormal and individuals may or may modifying therapies that prevent or delay the pro-
not develop HD, and individuals with ‡40 repeats gression of HD, with current treatment strategies
will always develop the disease.[1,2,6] targeting the relief of symptoms, including the
The characteristic triad of clinical features of classic choreic movements, to reduce the signif-
HD are progressive motor dysfunction (such as icant impact that HD has on the patient’s daily
chorea, dystonia, bradykinesia), neuropsychiatric living activities.[4-6] The dance-like movements,
manifestations (such as depression, obsessive- known as chorea, are the most characteristic ini-
compulsive disorders, anxiety) and cognitive tial physical manifestation of HD and occur in
impairment (for example, disorganization as a approximately 90% of patients.[8,10,12,13] Chorea
result of difficulties with planning, initiating and involves irregular, abrupt, uncontrolled move-
organizing thoughts).[1,2,4-6] These features are a ments of the face, extremities and body that may

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
Tetrabenazine: Adis Drug Profile 1075

appear dance-like because of the jerky, twisting 1. Pharmacodynamic Properties


motions. Although initially chorea may be quite
mild, consisting of general restlessness or clum-  Although the precise mechanism of action
siness, as the disease progresses these movements involved in the antichorea effects of tetrabena-
often make it very difficult to perform tasks of zine is unknown, these effects are thought to
daily living such as getting dressed. Ultimately, be mediated via reversible depletion of mono-
chorea may interfere with voluntary movements amines from presynaptic vesicles, especially do-
and thereby impact on activities like walking, pamine. Tetrabenazine is a selective, reversible, high
talking, eating and bladder control. However, affinity (inhibitory constant [Ki] »100 nmol/L[15])
there is a poor correlation between the extent of inhibitor of VMAT2 (the neuronal isoform of
chorea and disease severity.[8,10,12,13] VMAT).[16-27] Inhibition of VMAT2 by tetra-
Current pharmacotherapies used in the treat- benazine reduces the uptake of monoamines into
ment of chorea associated with HD include neu- presynaptic vesicles and thereby depletes concen-
roleptic agents (atypical neuroleptic agents such trations of monoamines, particularly dopamine,
as olanzapine, aripiprazole and risperidone or as observed in preclinical[16-25] and human post-
older neuroleptics such as haloperidol) and the mortem[26,27] studies.
vesicular monoamine transporter (VMAT) type 2  The major active circulating metabolite of tetra-
(VMAT2) inhibitor tetrabenazine (Xenazine), benazine, a-dihydrotetrabenazine (see section 2),
with VMAT2 inhibition resulting in depletion of also inhibited VMAT2 and reduced monoamine
monoamines, especially dopamine, in presynap- uptake into presynaptic vesicles in in vitro[20] and
tic vesicles.[4-6] In a recent Cochrane review of 22 animal studies.[18,19] In rats, a-dihydrotetrabenazine
trials evaluating various pharmacotherapies for was as effective or more effective than tetrabenazine
the management of chorea associated with HD, in terms of its effects on monoamine depletion.[18]
including anti-dopaminergic agents, glutamate re- Of the monoamines, dopamine concentrations de-
ceptor antagonists and energy metabolites, tetra- creased the most and serotonin concentrations de-
benazine treatment provided the best clinical creased the least, with monoamine concentrations in
dence for efficacy in the control of chorea.[14] To the brain returning to control concentrations within
date, tetrabenazine is the only drug that is ap- 12 hours. Serum and brain concentrations of tetra-
proved by the US FDA for the treatment of benazine and a-dihydrotetrabenazine peaked at
chorea associated with HD. This review focuses 30 minutes post-dose, which correlated with the
on the use of oral tetrabenazine at dosages re- time to peak depletion of monoamines.[18]
commended in the US for the treatment of chorea  In vitro studies indicated that tetrabenazine
associated with HD (see section 5);[15] discussion only inhibited monoamine transport by neuronal
of its use in patient populations with other forms VMAT2, whereas reserpine inhibited both neu-
of dyskinesia and of studies in which dosages ronal VMAT2 and peripheral, endocrine-specific
greater than those recommended were used is be- VMAT type 1 (VMAT1) resulting in both central
yond the scope of this review. and peripheral depletion of monoamines.[16,19,21,28]
Medical literature (including published and This lack of inhibition of VMAT1 by tetrabena-
unpublished data) on the use of tetrabenazine in zine may, at least in part, explain its lower pro-
Huntington’s disease was identified by searching pensity than reserpine to cause hypotension and
databases since 1996 (including MEDLINE, EM- gastrointestinal adverse effects such as epigastric
BASE), bibliographies from published literature, pain and diarrhoea.[4,29]
clinical trial registries/databases and web sites (in-  Tetrabenazine also differs from reserpine in
cluding those of regional regulatory agencies and the chemical structure (tetrabenazine lacks the indole
manufacturer). Additional information (including ring of reserpine), reversible rather than irrevers-
contributory unpublished data) was also requested ible inhibition of VMAT2, binding at intravesicular
from the drug company. Searches were last updated sites rather than cytoplasmic sites and its shorter
15 November 2011. duration of action (hours vs days).[16,18,26,29]

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
1076 Scott

 In in vitro studies, tetrabenazine exhibited a irrespective of whether tetrabenazine treatment


low affinity for dopamine D2 receptors (Ki = 2100 was initiated in younger (2 months of age) or
nmol/L).[30,31] Since the affinity of tetrabenazine older mice (6 months of age).[33,34]
at D2 receptors is approximately 1000-fold lower  Single doses of tetrabenazine 25 and 50 mg
than that for VMAT2, it appears unlikely that the increased the maximum time-matched placebo-
clinical antichorea effects of tetrabenazine are adjusted change from baseline in corrected QT
mediated via this mechanism.[29] (QTc) interval by 3.6 and 7.7 msec in healthy adult
 In postmortem brain tissue from patients with volunteers in a double-blind, Thorough QTc study,
HD, tetrabenazine-treated patients (n = 11) had as reported in the US FDA briefing document.[35]
significantly lower concentrations of dopamine To date, studies evaluating the effects of greater
in the caudate (1113 vs 3281 ng/g tissue [reduced doses of tetrabenazine or its metabolites on the
by 67%]; p < 0.01) and in the hippocampus (9.61 QTc interval have not been conducted.[15,35] Since
vs 22.2 ng/g tissue [reduced by 57%]; p < 0.05) tetrabenazine treatment causes small increases in
than patients who had not received tetrabenazine the QTc interval, the use of tetrabenazine in com-
(n = 7).[27] There were no significant between- bination with drugs that are known to prolong
group differences in dopamine concentrations in the QTc interval and in patients with congenital
the amygdala or temporal cortex. The significant long QT syndrome or with a history of cardiac
reduction of dopamine concentrations in the arrhythmias should be avoided because of poten-
caudate probably explains the amelioration of tial adverse events, such as torsade de pointes-
chorea observed with tetrabenazine treatment, as type ventricular arrhythmias.[15]
chorea is thought to be related to hyperdopami-  Since tetrabenazine or its metabolites bind to
nergic activity. Dopamine concentrations in post- melanin-containing tissues (assessed in animal
mortem brain tissue of tetrabenazine-treated studies), they may accumulate in these tissues over
patients reflected those observed in a matched time and potentially be associated with toxicity
control group.[27] in these tissues (reported in the US manufacturer’s
 In this study,[27] there were also significantly prescribing information).[15] The clinical rele-
(p < 0.05) lower concentrations of norepinephrine vance of the binding of tetrabenazine to melanin-
(noradrenaline) in the amygdala (47.1 vs 105 ng/g containing tissues is unknown. Ophthalmological
tissue in untreated patients [reduced by 55%]) and monitoring in humans was inadequate to exclude
hippocampus (14.1 vs 47.4 ng/g tissue [reduced by the possibility of injury occurring after long-term
70%]) in tetrabenazine-treated patients, but no exposure to tetrabenazine.[15]
significant between-group differences in norepi-
nephrine concentrations in the temporal cortex. 2. Pharmacokinetic Properties
There were no between-group differences in terms
of serotonin concentrations in any of these re- There is a relative paucity of published data
gions of the CNS. relating to the pharmacokinetics of oral tetra-
 In an observational study in ten patients with benazine in humans,[36-38] with most data derived
HD, a single dose of tetrabenazine improved from the US manufacturer’s prescribing informa-
chorea for a mean of 5.4 hours (minimum of tion[15] and the FDA briefing document for tetra-
3.2 hours and maximum of 8.1 hours), as assessed benazine.[35]
using the Unified HD Rating Scale (UHDRS)  After oral administration, tetrabenazine is
total chorea score.[32] See section 3 for discussion rapidly absorbed in the gastrointestinal tract, with
of the antichorea effects of tetrabenazine treat- the extent of absorption being at least 75% of the
ment in patients with HD participating in short- administered dosage.[15]
and long-term clinical studies.  After single tetrabenazine doses of 12.5–50 mg,
 In a transgenic mouse model of HD (YAC128 plasma concentrations of tetrabenazine are gener-
mice), tetrabenazine treatment alleviated the motor ally below the limit of detection because of rapid
deficits and reduced striatal neuronal cell loss, and extensive first-pass metabolism by carbonyl

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
Tetrabenazine: Adis Drug Profile 1077

reductase to the major circulating metabolites, metabolism of a-dihydrotetrabenazine and b-


a-dihydrotetrabenazine (biologically active) and dihydrotetrabenazine.[15]
b-dihydrotetrabenazine (inactive).[15,36,37] Peak  In healthy volunteers, metabolites of tetrabe-
plasma concentrations (Cmax) of a-dihydrotetra- nazine were primarily eliminated via the renal
benazine and b-dihydrotetrabenazine are attained route, with approximately 75% of the dosage ex-
within 1–1.5 hours post-dose and that of their creted in the urine and 7–16% of in the faeces.[15]
major metabolite, O-dealkyated-dihydrotetra- Unchanged tetrabenazine has not been found in
benazine, is attained approximately 2 hours post- human urine; a-dihydrotetrabenazine and b-dihy-
dose. drotetrabenazine detected in the urine accounted
 Across a dosage range of 37.5–112.5 mg/day, for less than 10% of the administered dose. The
tetrabenazine and dihydrotetrabenazine displayed majority of metabolites in the urine consisted of
linear pharmacokinetics in four patients with tar- sulfate and glucuronide conjugates of dihydrote-
dive dyskinesia.[36] Patients exhibit a high degree trabenazine or products of oxidative metabolism.[15]
of interindividual variability in the distribution,  The respective elimination half-lives (t½) of
metabolism (conversion to dihydrotetrabenazine) a-dihydrotetrabenazine and b-dihydrotetrabena-
and clearance of tetrabenazine and dihydrotetra- zine are 7 and 5 hours.[15]
benazine,[37] with the manufacturer’s prescribing
information[15] indicating that the dosage should In Special Patient Populations
be optimized on an individual basis (section 5).
 There were no clinically relevant effects of  There is no apparent effect of the sex of an
food on the bioavailability of tetrabenazine;[15,37] individual on the pharmacokinetics of a-dihy-
thus, the drug may be taken without regard to drotetrabenazine and b-dihydrotetrabenazine.[15]
food.[15] The pharmacokinetics of tetrabenazine and its pri-
 Positron emission tomography scan studies in mary metabolites have not been studied in paedi-
humans showed that radioactivity is rapidly dis- atric patients or in patients with renal impairment,
tributed into brain tissue following an intrave- nor have they been formally studied in geriatric pa-
nous injection of radiolabelled tetrabenazine, with tients or in patients of different races.[15]
the greatest binding occurring in the striatum  Since tetrabenazine is extensively metabolized
and the least binding in the cortex.[15] At human in the liver, exposure to the drug is increased in
plasma concentrations of 50–200 ng/mL, in vitro patients with hepatic impairment.[15] There was a
protein binding of tetrabenazine, a-dihydrotetra- marked decrease in the metabolism of tetrabena-
benazine and b-dihydrotetrabenazine ranged zine in patients with mild to moderate hepatic
from 82% to 85%, 60% to 68% and 59% to 63%, impairment (Child-Pugh scores 5–9; n = 12) com-
respectively.[15,37] pared with sex-matched volunteers with normal
 After oral administration, tetrabenazine is hepatic function, with plasma concentrations of
rapidly and extensively metabolized to at least tetrabenazine being similar to those of a-dihy-
19 metabolites, including initial metabolism by drotetrabenazine in patients with hepatic impair-
carbonyl reductase (mainly localized in the liver) ment. Mean Cmax values for tetrabenazine were
to a-dihydrotetrabenazine and b-dihydrotetrabe- approximately 7- to 190-fold greater in patients
nazine.[15,36,37] These major metabolites are then with hepatic impairment than those in healthy
converted to O-deaklyated-dihydrotetrabenazine, volunteers, with t½ values increased to approxi-
principally by cytochrome P450 (CYP) 2D6, with mately 17.5 hours in patients with hepatic impair-
some contribution from CYP1A2 in the case of ment. Hence, tetrabenazine is contraindicated in
a-dihydrotetrabenazine. Subsequently, the major patients with hepatic impairment.[15]
metabolites are converted to sulfate and glu-  Since a-dihydrotetrabenazine and b-dihydro-
curonide conjugates.[15] In vitro studies indicate tetrabenazine are principally metabolized by the
that CYP1A2, CYP2A6, CYP2C9, CYP2C19 genetically polymorphic CYP2D6 pathway, it is
and CYP2E1 do not play a major role in the likely that exposure to these major metabolites of

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
1078 Scott

tetrabenazine will be increased in individuals who affect on the bioavailability of digoxin (a sub-
are classified as poor metabolizers (PM) com- strate for P-glycoprotein).[15]
pared with those classified as extensive meta-
bolizers (EM).[15] The increase in exposure to 3. Therapeutic Efficacy
a-dihydrotetrabenazine and b-dihydrotetrabenazine
would be expected to be similar to that observed The efficacy of oral tetrabenazine has been
in patients receiving tetrabenazine concomitantly evaluated in adult patients with HD in short-term
with a strong CYP2D6 inhibitor such as parox- (<12 months’ duration)[39-44] and long-term (‡12
etine (also see discussion below on potential drug months’ duration)[45-48] studies. Discussion focuses
interactions), although the clinical relevance of on the pivotal, 12-week, randomized, double-blind,
this remains to be fully elucidated.[15,35] placebo-controlled, multicentre trial[39] conducted
in the US and an open-label extension phase[45] of
Potential For Drug Interactions this study, both of which are fully published.
 Coadministration of tetrabenazine with a
Short-Term Studies
strong CYP2D6 inhibitor (such as paroxetine,
fluoxetine, quinidine) may increase exposure to Key trial design and study details, including the
a-dihydrotetrabenazine and b-dihydrotetrabena- dosage regimen and efficacy outcome measures,
zine; caution is advised when coadministering these for the 12-week, double-blind study are sum-
agents and the dosage of tetrabenazine should be marized in table I.[39] There were generally no
halved.[15] Concomitant administration of a single significant between-group differences in baseline
dose of tetrabenazine 50 mg following 10 days demographics and HD history (including age,
of paroxetine 20 mg/day resulted in an increase HD duration, CAG repeat number and history of
in Cmax and area under the plasma concentration- depression [table I]), with the exception of base-
time curve values for a-dihydrotetrabenazine of ap- line scores for UHDRS symbol digit (18.1 in the
proximately 30% and 3-fold, with respective values tetrabenazine group vs 24.4 in the placebo group;
for b-dihydrotetrabenazine increasing 2.4- and 9-fold. p = 0.018) and Functional Impact Scale (1.3 vs 0.4;
The effects of moderate or weak CYP2D6 inhib- p = 0.035) which favoured the placebo group. Pre-
itors (such as duloxetine, amiodarone, terbinafine, dictably, the number of CAG repeats was inversely
sertraline) have not been evaluated.[15] correlated (p < 0.0001) with the age at disease onset.
 Based on in vitro studies, a clinically relevant  After 12 weeks’ treatment, tetrabenazine (£100
interaction between tetrabenazine and other CYP mg/day) was significantly more efficacious than
inhibitors (except those inhibiting CYP2D6) placebo at improving chorea, as measured by ad-
appears unlikely.[15] In these studies, tetrabena- justed mean UHDRS total maximum chorea scores
zine, a-dihydrotetrabenazine and b-dihydrotetra- (primary outcome) [figure 1].[39] The adjusted treat-
benazine resulted in no clinically significant inhib- ment effect size for improvement in chorea severity
ition of CYP2D6, CYP1A2, CYP2C8, CYP2C9, was -3.5 (95% CI -5.2, -1.9), with an average 23.5%
CYP2C19, CYP2E1 or CYP3A isoenzymes; their reduction from baseline in chorea severity. Improve-
effect on the CYP2B6 isoenzyme has not been ments in chorea with tetrabenazine treatment
evaluated.[15] Similarly, it appears unlikely that they occurred irrespective of age, sex of the patient,
result in any clinically significant induction of CAG repeat number, sex of affected parent, baseline
CYP1A2, CYP3A4, CYP2B6, CYP2C8, CYP2C9 Clinical Global Impression (CGI) severity score or
or CYP2C19 isoenzymes.[15] baseline UHDRS total maximum chorea score.
 Tetrabenazine, a-dihydrotetrabenazine and  More than two-thirds (69%) of patients receiv-
b-dihydrotetrabenazine are also unlikely to be a ing tetrabenazine had an improvement in UHDRS
substrate for or inhibitor of P-glycoprotein, based total maximum chorea score of >3 compared with
on in vitro studies.[15] In healthy volunteers, tetra- 20% of patients in the placebo group (odds ratio
benazine 25 mg twice daily for 3 days had no 9.9; 95% CI 3.2, 29.9; p < 0.0001).[39]

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
Tetrabenazine: Adis Drug Profile 1079

Table I. Key patient characteristics and trial design details for the double-blind, placebo (PL)-controlled, multicenter trial in patients (pts) with
Huntington’s disease (HD)[39]
Inclusion criteria HD as confirmed by presence of chorea, a family history and an expanded CAG repeat (n ‡ 37);
independently ambulatory; TFC score at screening of >5; TMC scorea of ‡10; pts who had
previously received DOP D2 receptor blockers were enrolled if they had discontinued this
treatment ‡4 wk before enrolment; pts continued existing antidepressant or benzodiazepine
drugs provided they had been taking a stable dosage for ‡8 wk prior to enrolment
Exclusion criteria Presence of disabling depression, dysphagia or dysarthria; prior treatment with TBZ or current
treatment with DOP-depleting drugs, DOP D2 receptor blockers, selective and nonselective
MAOIs, levodopa, DOP agonists, amantadine or memantine
Patient characteristics at baseline (TBZ Age 49.4 y,b 48.8b y [25–77 y]; CAG repeats 44.9,b 44.3b [39–54 repeats]; disease duration
group, PL group) [range] 8.7 y,b 7.5b y [1.6–25.6 y]; history of depression 63%, 47% of pts; female 61%, 63% of pts
Treatment regimen Pts were randomized to TBZ (n = 54) or PL (n = 30). During wk 0–7 (titration phase), the dosage
was titrated to up to a maximum of 8 tablets/day (TBZ 12.5 mg/tablet or PL tablet identical in
appearance) taken as 3 divided doses, with the dosage individualized to the optimum dosage
based on efficacy and tolerabilityc; during wk 7–12 (maintenance phase), the dosage remained
constant unless reduced because of intolerable adverse effects. After wk 12, there was a 1 wk
wash-out period
Optimized dosage range At wk 12, 27 of 49 pts (55%) in the TBZ group and 4 of 29 pts (14%) in the PL group were taking
less than the maximum dosage
Primary efficacy outcome The difference between the baseline TMC score and the average of the wk 9 and 12 scores,
with analyses conducted in the intention-to-treatd population; if either wk 9 or 12 data were
missing, the one available score was imputed for missing data; if neither wk 9 nor wk 12 data
were available, the last observation carried forward method was used to impute missing data
Secondary efficacy outcomes The study prespecified four secondary endpointse (mean change from baseline in CGI-GI,
UHDRS total motor score, UHDRS functional checklist and UHDRS gait score) to be analysed
in a hierarchical manner until there was no statistical between-group difference (p ‡ 0.05).
Thereafter, endpoints were considered to be exploratory endpoints
a Sum of maximum chorea score for facial, buccal-oral-lingual, trunk and each extremity from the motor subscale of the UHDRS. Chorea is
graded for each of these seven body regions on a scale of 0 (no chorea) to 4, giving a total score of 0–28.[49]
b Mean value.
c To reduce intolerable adverse effects, the dosage was reduced to the pt’s previously well tolerated level or lower if necessary.
d All randomized pts.
e Analysed using wk 9 and 12 data, as per the primary outcome measure, except CGI-GI data which only utilized wk 12 scores.
CAG = trinucleotide encoding glutamine; CGI-GI = Clinical Global Impression-Global Improvement; DOP = dopamine; MAOIs = monoamine
oxidase inhibitors; TBZ = tetrabenazine; TFC = total functional capacity; TMC = total maximum chorea; UHDRS = Unified HD Rating Scale.

 Tetrabenazine treatment was also more mum global improvement (defined as a CGI-GI
efficacious than placebo in terms of improve- score of £3) at 12 weeks (69% vs 24%; p = 0.0001),
ments in the adjusted mean CGI-Global Improve- with 45% and 7% of patients showing a greater
ment (GI) scores (figure 1), which was the first than minimum improvement (p < 0.001).[39]
outcome in the prespecified hierarchical plan of  As per the hierarchical plan for analyses of
analyses for secondary outcomes (see table I for secondary outcomes, since the adjusted mean
further details).[39] The adjusted mean treatment changes from baseline in UHDRS total motor
effect size for the CGI-GI score at 12 weeks was -0.7 scores failed to reach statistical significance
(95% CI -1.3, -0.2), representing an improvement in (mean treatment effect size -3.3; 95% CI -7.0,
favour of tetrabenazine treatment. These improve- 0.3) [figure 1], all other efficacy endpoints were
ments in CGI-GI scores significantly (p £ 0.0001) considered exploratory.[39]
correlated with improvements in UHDRS total  For exploratory endpoints, significant between-
maximum chorea and total motor scores. group differences in favour of placebo treatment
 Significantly more tetrabenazine recipients were observed for adjusted mean changes in
than placebo recipients showed at least a mini- UHDRS functional checklist scores (-0.8 in the

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
1080 Scott

Primary Secondary TBZ Long-Term Studies


Adjusted mean score/change from baseline

6 outcome outcomes PL

4
*
The long-term efficacy of tetrabenazine has
been evaluated in the extension phase[45] of the
2
12-week, double-blind trial,[39] and in observa-
0 tional[46] and retrospective[44,48] studies.
Of the 84 participants in the 12-week, double-
−2
blind study,[39] 75 met the criteria for enrolment
−4 in the 80-week, open-label extension phase.[45]
The criteria for inclusion in the extension study
−6 **
were that patients had completed the initial
−8 12-week study and had not had a severe adverse
UHDRS TMC CGI-GI UHDRS total motor
event during this 12-week period. The extension
Fig. 1. Comparative efficacy of oral tetrabenazine (TBZ) in adult study consisted of a 24-week phase (a 12-week
patients (pts) with Huntington’s disease. Results from a 12-week,
randomized, double-blind, multicenter trial in which pts received TBZ dose-titration phase followed by a 12-week main-
(n = 54; maximum dosage £100 mg/day) or placebo (PL; n = 30) for tenance phase; n = 75 enrolled patients), a 24-week
12 weeks (see table I for dosage and design details).[39] Adjusted (for
site and baseline scores) mean changes from baseline in UHDRS
extension phase (total of 48 weeks of treatment;
TMC and total motor scores (a decrease in score indicates an im- n = 58) and a final extension through to week 80
provement) to study end, and the mean CGI-GI score (a score of (n = 46), with a 1-week wash-out between each of
<4 = an improvement, 4 = no change and >4 = worsening) at study
end, as analysed in the intention-to-treat population. All scores were these three phases. During the extension study,
rated by the investigator. Baseline scores in the TBZ and PL groups the dosage of tetrabenazine was individually op-
were: UHDRS TMC 14.7 and 15.2; UHDRS total motor 47.0 and
44.8. CGI-GI = Clinical Global Impression-Global Improvement; timized over a 12-week period, with the dosage
TMC = total maximum chorea; UHDRS = Unified Huntington’s Dis- adjusted every 3–7 days up to a maximum of
ease Rating Scale; * p = 0.007, ** p = 0.0001 vs PL.
200 mg/day.[45]
 Of note, more than half of the patients
remained on the same tetrabenazine dosage from
tetrabenazine group vs 0.4 in the placebo group; 24 weeks onwards (70% of patients between weeks
p = 0.02; lower scores represent a worsening), Ep- 24 and 48, and 59% between weeks 48 and 80).[45]
worth sleepiness scores (1.5 vs -0.3; p = 0.02; greater Of the 44 patients with complete tetrabenazine
scores represent more sleepiness) and Stroop test dosage data at 80 weeks, including three patients
word reading scores (-4.8 vs 1.8; p = 0.01; lower receiving no study drug, 55% were receiving a
scores mean a deterioration).[39] There were no dosage of 37.5 or 50 mg/day. Moreover, the dos-
significant between-group differences in Stroop age of tetrabenazine remained relatively constant
test colour naming or interference scores and following the dosage-titration phase; at week 24,
no clinically relevant changes in UHDRS gait the mean tetrabenazine dosage in patients who
scores. remained on study drug was 74.2 mg/day (n = 66),
 At the end of the 1-week wash-out period at 48 weeks it was 71.5 mg/day (n = 54) and at
(weeks 12–13 of the study), there was no occur- 80 weeks it was 63.4 mg/day (n = 41).
rence of rebound chorea, with no statistically  The efficacy of tetrabenazine during long-term
significant differences between the tetrabenazine treatment was maintained during the 80-week
and placebo groups for changes in motor, cog- extension study.[45] There was a significant
nitive, behavioural or global measures of illness (p < 0.0001) reduction from baseline (that is, the
severity during this period.[39] As might be pre- start of the extension study; score 14.9) in
dicted, compared with placebo recipients, tetra- adjusted mean UHDRS total maximum chorea
benazine-treated patients showed a significant score with tetrabenazine treatment (reduced by
worsening of chorea severity during the wash-out 4.6 units; primary outcome). Respective reduc-
phase (adjusted treatment effect size of 4.4 tions from baseline in this score at weeks 24 and
UHDRS; 95% CI 2.8, 6.0; p < 0.0001). 48 were 5.8 and 5.6 (both p < 0.05).

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
Tetrabenazine: Adis Drug Profile 1081

 At week 81, after discontinuation of the study giver detected any signs of depression and no
drug for 1 week, no rebound chorea was observed.[45] abnormalities were detected on the patient’s
The adjusted mean UHDRS total maximum cho- Hamilton Depression Scale (HAM-D) score at
rea score increased by 5.3 from the 80-week score that point in time.
(p < 0.001), with no significant difference between  During this study, five patients discontinued
this score at week 81 and that at baseline.[45] tetrabenazine treatment; four because of serious
 Of the 45 patients who completed 80 weeks of adverse events and one because of akathisia.[39]
treatment, approximately one-third (36%) expe- Serious adverse events included an intracerebral
rienced an improvement in adjusted mean CGI haemorrhage following a fall, hospitalization for
scores from baseline, 9% of patients experienced restlessness that resolved within 48 hours of dos-
a worsening and 55% showed no change in their age reduction (2 weeks later the patient developed
score (secondary outcome).[45] At week 80 there depressive symptoms, irritability and suicidal
was no significant change from baseline in UHDRS ideation) and breast cancer (the patient had
total motor scores. a breast lump prior to screening). There were no
 Secondary efficacy outcomes assessing cogni- occurrences of serious adverse events in the
tive and functional abilities generally showed a placebo group.[39]
deterioration during the 80-week study.[45] Thus,  Dosage titration of tetrabenazine was discontin-
there was a significant (p < 0.05) deterioration from ued or the dosage reduced in 52% of patients
baseline in scores for the following cognitive because of the occurrence of one or more adverse
measures: verbal fluency (17.2 at baseline vs 13.6 events.[15] Dosage-limiting events included sedation
at 80 weeks), symbol digit (19.3 vs 14.5), Stroop (13 patients), akathisia (4), parkinsonism (2), depres-
colour naming (42.6 vs 31.6), Stroop words (49.8 sion as description of mood (3) and others (3).[39]
vs 40.8) and Stroop interference (23.2 vs 18.2).  Treatment-emergent adverse events in the
Similarly, for functional measures, there was a tetrabenazine group mainly occurred during
significant (p < 0.05) worsening of scores from the dosage-titration phase, a period during which
baseline for UHDRS functional checklist (18.1 vs the dosage was optimized based, for the most
15.5), UHDRS independence (75.7 vs 68.7) and part, on tolerability.[39] By the end of the main-
total functional capacity (7.6 vs 5.6). tenance phase, when presumably patients were
 Results from this extension study are supported on an optimal individualized dosage, there were
by observational[46] (n = 29 with HD; average treat- no significant differences between the tetrabena-
ment duration »29 months) and retrospective zine and placebo groups in relation to the
studies (n = 24[48] and 68;[44] treatment duration frequency of specific adverse events that had not
»1–53 months[48] and a median of 34 months[44]). been reported at baseline.[39]
 In the 12-week double-blind trial, 91% of
4. Tolerability patients in the tetrabenazine group experienced
at least one adverse event versus 70% in the
This section focuses on tolerability data from placebo group (p = 0.01), most of which were mild
the 12-week placebo-controlled trial[39] and its to moderate and manageable with dosage adjust-
extension study (see section 3 for dosage and de- ments or discontinuation of study drug.[39] The
sign details). Adverse events were coded using most common (incidence ‡15% and greater in
WHO preferred terms. Supplemental data are the tetrabenazine group than in the placebo group)
derived from the US manufacturer’s prescribing treatment-emergent adverse events occurring during
information[15] and FDA briefing document[35] the 12-week trial were drowsiness/somnolence,
for tetrabenazine. insomnia, fatigue, fall, depression, agitation and
 In the 12-week, placebo-controlled study, one anxiety (figure 2).[39] Drowsiness/somnolence and
death (a suicide) occurred in the tetrabenazine insomnia occurred with a significantly greater
group.[39] At the visit 2 weeks prior to the suicide, incidence in the tetrabenazine group than in the
neither the investigator nor the patient’s care- placebo group (figure 2). Of note, depression

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
1082 Scott

refers to a description of mood rather than a for- AST and ALT concentrations in patients receiv-
mal diagnosis. ing tetrabenazine compared with those receiving
 The FDA briefing document for tetrabena- placebo.[15] In the extension study (£80 weeks’
zine[35] indicated that not all adverse events may treatment), although three patients (4%) expe-
have been captured during the 12-week double- rienced an isolated increase in AST concentration
blind study.[39] In the tetrabenazine group, the of ‡3 times the upper limit of normal, no patients
FDA analysis identified an additional three cases experienced clinical liver dysfunction.[45]
of parkinsonism, two cases of akathisia, three  Tetrabenazine treatment had no clinically
cases of restlessness that could have been cases of relevant effects on blood pressure, pulse and body-
akathisia, two cases of depression and one case of weight in controlled clinical trials.[15]
dysphagia.[35] No additional adverse events were  As discussed earlier, HD is associated with an
identified in the placebo arm. The incidences of increased risk of depression and suicidal ideation
the most common adverse events, as reported in the or behaviour. Tetrabenazine may potentially
briefing document,[35] were sedation (32% in tetra- increase these risks,[15] because along with deple-
benazine group vs 3% in placebo group), fatigue tion of dopamine it also causes depletion of sero-
(22% vs 13%), insomnia (22% vs 0%), akathisia/ tonin and norepinephrine (section 1).
restlessness (19% vs 0%), depression (19% vs 0%),  In the 12-week double-blind study, 15% of pa-
falls/traumatic injury (19% vs 13%), anxiety (15% tients receiving tetrabenazine had an adverse event
vs 3%) and parkinsonism (15% vs 0%). of depression (figure 2).[39] During this study, both
 No clinically significant changes in laboratory tetrabenazine-treated patients and placebo recip-
parameters were reported in patients receiving ients experienced an improvement in HAM-D
tetrabenazine in clinical trials.[15] In controlled scores, with these improvements from baseline
clinical trials, there were small increases in mean being more marked in the placebo group than in
the tetrabenazine group (-2.4 vs -0.7; p = 0.003).
 The use of antidepressants by patients at base-
TBZ
URTI line (56% of patients) or a history of depression
PL
Purpura (67%) were not associated with the development
Nausea
of depressed mood during tetrabenazine treat-
ment, based on a post hoc analysis of data from
Anxiety
48 participants in the 12-week study (abstract
Agitation
θ presentation).[50] Depressed mood was defined as
Depression the incident development of the adverse event of
θ
depressed mood (occurred in 10% of patients)
Fall
or at least a 2-point worsening on the depressed
Fatigue mood item of the UHDRS (occurred in 8% of
Insomnia
θ
* patients).
Drowsiness/ *
 In a retrospective analysis of 518 patients with
somnolence various dyskinesias, including chorea (31% of
0 5 10 15 20 25 30 35 patients), 78 patients (15%) treated with tetrabe-
Percentage of patients
nazine developed depression for the first time or
Fig. 2. Tolerability profile of oral tetrabenazine (TBZ) in adult had an exacerbation of their pre-existing depres-
patients with Huntington’s disease. Treatment-emergent adverse
events occurring in ‡10% of patients receiving TBZ (n = 54; max-
sion.[51] In this study, of the 246 patients without
imum dosage £100 mg/day) and with a greater frequency in the TBZ a history of depression prior to tetrabenazine treat-
group than in the placebo (PL; n = 30) group in a 12-week, random- ment, 11.4% were newly diagnosed with depression
ized, double-blind, multicentre trial discussed in section 3 (see table I
for dosage and design details).[39] Coding was based on the adverse during tetrabenazine treatment. Over the entire
event log using WHO preferred terms. Depression applied to a de- cohort, the mean duration of tetrabenazine treat-
scription of mood, rather than a formal diagnosis. URTI = upper res-
piratory tract infection; y indicates a frequency of 0%; * p = 0.006 ment was 29.7 months and the mean daily dosage
vs PL. was 53.0 mg/day.[51]

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
Tetrabenazine: Adis Drug Profile 1083

 In studies evaluating the use of tetrabenazine 5. Dosage and Administration


for the treatment of chorea associated with HD
(n = 187), one patient committed suicide, one at- Oral tetrabenazine is approved in the US[15] for
tempted suicide and six reported suicidal ideations.[15] the treatment of chorea associated with HD and is
approved in several other countries (including the
UK, Canada, France, Germany) for the treatment
Long-Term Treatment
of various movement disorders, including HD.[4]
 In the long-term extension study (up to 80 Careful individualization of the dosage of tetra-
weeks’ treatment) of the 12-week trial (n = 75 benazine is required.[15] The starting dosage
participants), the most commonly reported treat- should be 12.5 mg per day given once in the
ment-related adverse events (excluding those of mild morning. After 1 week, the dosage should be in-
severity) in the 44 patients who completed creased to 25 mg per day given as 12.5 mg twice a
the 80-week study were sedation/somnolence day. Tetrabenazine should be titrated up slowly
(18 patients), depressed mood (17), anxiety (13), at weekly intervals by 12.5 mg daily, to allow the
insomnia (10) and akathisia (9).[45] Three patients identification of a tolerated dosage that reduces
withdrew from the study because of adverse events, chorea. Dosages of 37.5–50 mg/day should be
including vocal tics, depression and delusions, given as three divided doses, with the maximum
that were attributed to the study drug by inves- recommended single dose being 25 mg. Patients
tigators. One patient died of breast cancer during requiring dosages above 50 mg/day should be
the study. CYP2D6 genotyped to determine whether they
 As was the case during the initial 12-week are PM or EM (see section 2). The maximum
study, the frequency of treatment-emergent ad- dosage for PM is 50 mg/day, with a maximum
verse events was greater during the dosage- single dose of 25 mg. For EM and intermediate
titration phase than during the maintenance metabolizers, the maximum dosage is 100 mg/day,
phase, with 39 patients experiencing at least with a maximum single dose of 37.5 mg.[15]
one adverse event during the dosage-titration Tetrabenazine is contraindicated for patients
phase versus 20 patients during the maintenance who are actively suicidal or who have depression
phase (p < 0.001).[45] Somnolence (occurred in that is untreated or undertreated.[15] It is also con-
36 patients during the titration phase vs 11 traindicated for patients with hepatic impairment
patients during the maintenance phase; p < 0.0001), and in those taking monoamine oxidase inhibitors
insomnia (14 vs 2; p < 0.003) and diarrhoea (5 vs 1; (MAOIs) or reserpine (see section 2). Tetrabena-
p < 0.05) generally resolved during the maintenance zine should also not be taken within 14 days of
phase.[45] discontinuing MAOIs or within 20 days of dis-
 From baseline to week 80, there were signif- continuing reserpine (since reserpine binds irrevers-
icant (p £ 0.002) increases in the mean UHDRS ibly to VMAT2 and thus has a prolonged effect on
parkinsonism score (by 2.1) and mean UHDRS monoamine concentrations; see section 1).[15]
dysphagia scores (by 0.4), although there was no Local prescribing information should be
significant change from baseline in the Unified consulted for comprehensive prescribing details,
Parkinson’s Disease Rating Scale dysarthria score.[45] including precautions, special warnings, contra-
There were no changes form baseline to week 80 in indications and use in special patient populations.
HAM-D or Barnes Akathisia Rating Scale (BARS)
scores. 6. Tetrabenazine: Current Status For
 During the extension study, ten patients (13.3%) Chorea Associated with Huntington’s
developed mild or moderate akathisia and a Disease
further three (4%) patients had mild or moderate
akathisia at baseline.[45] One patient experienced Oral tetrabenazine is the only drug approved
marked or severe akathisia (defined as a BARS by the FDA for the treatment of chorea in
score of 4 or 5). patients with HD. In a 12-week, double-blind

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
1084 Scott

trial in 84 patients with HD, oral tetrabenazine 8. Kirkwood SC, Su JL, Conneally M, et al. Progression of
(£100 mg/day) significantly improved chorea se- symptoms in the early and middle stages of Huntington
disease. Arch Neurol 2001; 58 (2): 273-8
verity compared with placebo, as assessed by the 9. van Duijn E, Kingma EM, van der Mast RC. Psychopa-
adjusted mean change from baseline in UHDRS thology in verified Huntington’s disease gene carriers.
total maximum chorea score. The beneficial ef- J Neuropsychiatry Clin Neurosci 2007; 19 (4): 441-8
fects of tetrabenazine treatment on chorea sever- 10. Walker FO. Huntington’s disease. Lancet 2007; 369:
218-28
ity were sustained in an 80-week extension study.
11. Paulsen JS, Hoth KF, Nehl C, et al. Critical periods of sui-
The most common treatment-emergent adverse cide risk in Huntington’s disease. Am J Psychiatry 2005;
events that occurred in the tetrabenazine groups 162 (4): 725-31
in these studies were drowsiness/somnolence, in- 12. Setter SM, Neumiller JJ, Dobbins EK, et al. Treatment of
chorea associated with Huntington’s disease: focus on tetra-
somnia, fatigue, fall, depression, agitation and benazine. Consult Pharm 2009; 24 (7): 524-37
anxiety. Most of these events were mild to mod- 13. Poon LH, Kang GA, Lee AJ. Role of tetrabenazine for
erate, manageable with dosage adjustments or dis- Huntington’s disease-associated chorea. Ann Pharma-
continuation of study drug, and mainly occurred cother 2010; 44 (6): 1080-9
during the dosage-titration phase, a period during 14. Mestre T, Ferreira J, Coelho MM, et al. Therapeutic inter-
ventions for symptomatic treatment in Huntington’s disease
which the dosage was individually optimized. (review). Cochrane Database Syst Rev 2009; 3: CD006456
15. Lundbeck Inc. US prescribing information: Xenazine (tetra-
benazine) tablets [online]. Available from URL: http://
Acknowledgments and Disclosures www.lundbeckinc.com/USA/products/CNS/xenazine/usa_
xen_pi.pdf [Accessed 2011 Jul 20]
The manuscript was reviewed by: A. Antonini, Parkinson 16. Quinn GP, Shore PA, Brodie BB. Biochemical and phar-
Institute and Neuroradiology, Istituti Clinici di Perfeziona- macological studies of RO I-9569 (tetrabenazine), a non-
mento, Milan, Italy; J.J. Chen, Loma Linda University, indole tranquilizing agent with reserpine-like effects.
Movement Disorders Center, Schools of Medicine and Phar- J Pharmacol Exp Ther 1959; 127: 103-9
macy, Loma Linda, CA, USA; S. Mehta, Medical College of 17. Pettibone DJ, Totaro JA, Pflueger AB. Tetrabenazine-
Georgia, Department of Neurology, Augusta, GA, USA. induced depletion of brain monoamines: characterization
The preparation of this review was not supported by any and interaction with selected antidepressants. Eur J Phar-
external funding. During the peer review process, the manu- macol 1984; 102 (3-4): 425-30
facturer of the agent under review was also offered an op- 18. Mehvar R, Jamali F. Concentration-effect relationships of
portunity to comment on this article. Changes resulting from tetrabenazine and dihydrotetrabenazine in the rat. J Pharm
comments received were made by the author on the basis of Sci 1987; 76 (6): 461-5
scientific and editorial merit. 19. Scherman D. Dihydrotetrabenazine binding and mono-
amine uptake in mouse brain regions. J Neurochem 1986;
47: 331-9
References 20. Scherman D, Jaudon P, Henry JP. Characterization of the
1. Krobitsch S, Kazantsev AG. Huntington’s disease: from monoamine carrier of chromaffin granule membrane by
molecular basis to therapeutic advances. Int J Biochem Cell binding of [2-3H]dihydrotetrabenazine. Proc Natl Acad Sci
Biol 2011; 43 (1): 20-4 U S A 1983; 80: 584-8
2. Ross CA, Tabrizi SJ. Huntington’s disease: from molecular 21. Erickson JD, Eiden LE, Schafer MK, et al. Reserpine- and
pathogenesis to clinical treatment. Lancet Neurol 2011; 10 tetrabenazine-sensitive transport of 3H-histamine by the
(1): 83-98 neuronal isoform of the vesicular monoamine transporter.
3. Huntington’s Disease Society of America. Lifting the veil J Mol Neurosci 1995; 6 (4): 277-87
of Huntington’s disease recommendations to the field 22. Lane JD, Smith JE, Shea PA, et al. Neurochemical changes
from the Huntington’s Disease Peer Workgroup [online]. following the administration of depleters of biogenic
Available from URL: http://www.hdsa.org/images/con monoamines. Life Sci 1976; 19 (11): 1663-7
tent/1/1/11709.pdf [Accessed 2011 Aug 4] 23. Christmas AJ, Coulson CJ, Maxwell DR, et al. A compar-
4. Frank S, Jankovic J. Advances in the pharmacological man- ison of the pharmacological and biochemical properties
agement of Huntington’s disease. Drugs 2010; 70 (5): 561-71 of substrate-selective monoamine oxidase inhibitors. \Br J
5. de Tommaso M, Serpino C, Sciruicchio V. Management of Pharmac 1972; 45: 490-503
Huntington’s disease: role of tetrabenazine. Ther Clin Risk 24. Bagchi SP. Differential interactions of phencyclidine with
Manag 2011; 7: 123-9 tetrabenazine and reserpine affecting intraneuronal dopa-
6. Novak MJU, Tabrizi SJ. Huntington’s disease. BMJ 2010; mine. Biochem Pharmacol 1983; 32 (19): 2851-6
340: c3109 25. Pettibone DJ, Pflueger AB, Totaro JA. Tetrabenazine-induced
7. Paulsen JS, Nehl C, Ferneyhough K, et al. Depression and depletion of brain monoamines: mechanisms by which des-
stages of Huntington’s disease. J Neuropsychiatry Clin methylimipramine protects cortical norepinephrine. Eur J
Neurosci 2005; 17 (4): 496-502 Pharmacol 1984; 102 (3-4): 431-6

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)
Tetrabenazine: Adis Drug Profile 1085

26. Thibaut F, Faucheux BA, Marquez J, et al. Regional dis- 39. Huntington Study Group. Tetrabenazine as antichorea
tribution of monoamine vesicular uptake sites in the mes- therapy in Huntington disease: a randomized controlled
encephalon of control subjects and patients with Parkinson’s trial. Neurology 2006; 66 (3): 366-72
disease: a postmortem study using tritiated tetrabenazine. 40. Ferrara JM, Mostile G, Hunter C, et al. Effect of tetrabenazine
Brain Res 1995; 692: 233-43 on motor function in patients with Huntington disease [ab-
27. Pearson SJ, Reynolds GP. Depletion of monoamine trans- stract no. P05.021]. 62nd Annual Meeting of the American
mitters by tetrabenazine in brain tissue in Huntington’s Academy of Neurology; 2010 Sep 10–17; Toronto (ON),
disease. Neuropharmacology 1988; 27 (7): 717-9 41. Ondo WG, Tintner R, Thomas M, et al. Tetrabenazine
28. Tomlinson DR. The mode of action of tetrabenazine on treatment for Huntington’s disease-associated chorea. Clin
peripheral noradrenergic nerves. Br J Pharmac 1977; 61: Neuropharmacol 2002; 25 (6): 300-2
339-44 42. Brusa L, Orlacchio A, Moschella V, et al. Treatment of the
29. Kenney C, Jankovic J. Tetrabenazine in the treatment of symptoms of Huntington’s disease: preliminary results
hyperkinetic movement disorders. Expert Rev Neurotherline comparing aripiprazole and tetrabenazine. Mov Disord
2006; 6 (1): 7-17 2009; 24 (1): 126-9
30. Reches A, Burke RE, Kuhn CM, et al. Tetrabenazine, an 43. McLellan DL, Chalmers RJ, Johnson RH. A double-blind
amine-depleting drug, also blocks dopamine receptors in trial of tetrabenazine, thiopropazate, and placebo in
rat brain. J Pharmacol Exp Ther 1983; 225 (3): 515-21 patients with chorea. Lancet 1974; 1 (7848): 104-7
31. Login IS, Cronin MJ, MacLeod RM. Tetrabenazine has 44. Fasano A, Cadeddu F, Guidubaldi A, et al. The long-term
properties of a dopamine receptor antagonist. Ann Neurol effect of tetrabenazine in the management of Huntington
1982; 12: 257-62 disease. Clin Neuropharmacol 2008; 31 (6): 313-8
32. Kenney C, Hunter C, Davidson A, et al. Short-term effects 45. Frank S. Tetrabenazine as anti-chorea therapy in Huntington
of tetrabenazine on chorea associated with Huntington’s disease: an open-label continuation study. Huntington
disease. Mov Disord 2007; 22 (1): 10-3 Study Group/TETRA-HD Investigators. [Erratum appears
33. Wang H, Chen X, Li Y, et al. Tetrabenazine is neuropro- in BMC Neurol 2011; 11: 18]. BMC Neurol 2009; 9: 62
tective in Huntington’s disease mice. Mol Neurodegener- 46. Jankovic J, Beach J. Long-term effects of tetrabenazine in hy-
ation 2010; 5 (18) perkinetic movement disorders. Neurology 1997; 48 (2): 358-62
34. Tang T-S, Chen X, Liu J, et al. Dopaminergic signaling and 47. Frank S, Ondo W, Fahn S, et al. A study of chorea after
striatal neurodegeneration in Huntington’s disease. tetrabenazine withdrawal in patients with Huntington dis-
J Neurosci 2007; 27 (30): 7899-910 ease. Clin Neuropharmacol 2008; 31 (3): 127-33
35. US FDA Peripheral and Central Nervous Systems Advisory 48. Kingston D. Tetrabenazine for involuntary movement dis-
Committee. Briefing Memo for December 6, 2007 PCNS orders. Med J Aust 1979; 1 (13): 628-30
AC meeting to discuss NDA 21-894, for the use of Xena- 49. Huntington Study Group. Unified Huntington’s Disease
zine (tetrabenazine) in the treatment of the chorea of Rating Scale: reliability and consistency. Mov Disord 1996;
Huntington’s Disease (HD) [online]. Available from URL: 11 (2): 136-42
http://www.fda.gov/ohrms/dockets/ac/07/briefing/2007-432
50. Biglan K, Eberly S, Auinger P, et al. Depressed mood, anti-
8b1-01-FDA.pdf [Accessed 2011 Nov 7]
depressants, and tetrabenazine: results from the TETRA-
36. Mehvar R, Jamali F, Watson MW, et al. Pharmacokinetics HD study [abstract]. World Congress of Huntington’s
of tetrabenazine and its major metabolite in man and rat: Disease; 2011 Sep 11–14; Melbourne (VIC)
bioavailability and dose dependency studies. Drug Metab
51. Kenney C, Hunter C, Mejia N, et al. Is history of depression
Dispos 1987; 15 (2): 250-5
a contraindication to treatment with tetrabenazine? Clin
37. Roberts MS, McLean S, Millingen KS, et al. The phar- Neuropharm 2006; 29 (5): 259-64
macokinetics of tetrabenazine and its hydroxy metabolite
in patients treated for involuntary movement disorders.
Eur J Clin Pharmacol 1986; 29 (6): 703-8 Correspondence: Lesley J. Scott, Adis, a Wolters Kluwer
38. Schwartz DE, Bruderer H, Rieder J, et al. Metabolic studies Business, 41 Centorian Drive, Private Bag 65901, Mairangi
of tetrabenazine, a psychotropic drug in animals and man. Bay, North Shore 0754, Auckland, New Zealand.
Biochem Pharmacol 1966; 15 (5): 645-55 E-mail: demail@adis.co.nz

ª 2011 Adis Data Information BV. All rights reserved. CNS Drugs 2011; 25 (12)

Vous aimerez peut-être aussi