Tetrabenazine, a monoamine-depleting drug used in the treatment of hyperkinetic movement disorders

Tetrabenazine, a monoamine-depleting drug used in the treatment of hyperkinetic movement disorders

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy Tetrabenazine, A Monoamine-Depleting Drug Used in the Treatment of Hyperkinetic Moveme...

660KB Sizes 0 Downloads 66 Views

D.R.P. Guay

The American Journal of Geriatric Pharmacotherapy

Tetrabenazine, A Monoamine-Depleting Drug Used in the Treatment of Hyperkinetic Movement Disorders David R.P. Guay, PharmD Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, and Division of Geriatrics, HealthPartners Inc., Minneapolis, Minnesota

ABSTRACT Background: Few drugs are available for the management of hyperkinetic movement disorders such as the dystonias, choreas, dyskinesias, and tics. Those that are available (primarily neuroleptics) are associated with a wide range of potentially serious adverse effects, including induction of tardive movement disorders. Tetrabenazine (TBZ) is a monoamine-depleting agent initially studied in the 1950s and currently approved by the US Food and Drug Administration for the treatment of chorea in Huntington’s disease. Objective: This article reviews the chemistry, pharmacology, pharmacokinetics, therapeutic use, tolerability, druginteraction potential, and dosing and administration of TBZ. Methods: MEDLINE was searched (1950–February 2010) for English-language articles investigating any aspect of TBZ. Search terms included tetrabenazine, Ro 1-9569, Nitoman, benzoquinolizines, s and reserpine. The reference lists of the identified articles were searched for other pertinent publications, particularly those that were not indexed in the 1950s and 1960s. Results: In the search for a chemical compound that was simpler than reserpine while preserving reserpine-like psychotropic activity, TBZ was identified in the 1950s as one member of a large group of benzoquinolizine derivatives. TBZ acts by depletion of the monoamines serotonin, norepinephrine, and dopamine in the central nervous system (CNS). It does this by reversibly inhibiting vesicle monoamine transporter type 2 and thus preventing monoamine uptake into presynaptic neurons. Clinical studies suggest that TBZ may have therapeutic applications in a wide range of hyperkinetic movement disorders. TBZ has been associated with numerous adverse effects, some of them serious and potentially fatal; these include parkinsonism, other extrapyramidal symptoms (particularly akathisia), depression and suicidality, neuroleptic malignant syndrome, and sedation. TBZ is subject to important drug–drug interactions with inhibitors and inducers of cytochrome P450 (CYP) 2D6, reserpine, and lithium. It is one of very few drugs whose dosing is based, in part, on the results of genotyping (in its case, genotyping for CYP2D6 metabolizer status). Conclusions: TBZ is a complicated drug in terms of its mechanism of action and its activities against the 3 major monoamines in the CNS, making it difficult to predict its efficacy and tolerability in patients with hyperkinetic movement disorders. It is associated with numerous adverse effects and several important drug–drug interactions. Much work remains to be done to determine the therapeutic potential of TBZ in the treatment of hyperkinetic movement disorders. (Am J Geriatr Pharmacother. 2010;8:331–373) © 2010 Excerpta Medica Inc. Key words: tetrabenazine, norepinephrine, dopamine, serotonin, movement disorders, tardive disorders, benzoquinolizine derivatives. Accepted for publication July 1, 2010. Published online August 30, 2010. ª&YDFSQUB.FEJDB*OD"MMSJHIUTSFTFSWFE

EPJKBNKPQIBSN TFFGSPOUNBUUFS

7PMVNFt/VNCFS

August 2010

331

The American Journal of Geriatric Pharmacotherapy

D.R.P. Guay

INTRODUCTION Tetrabenazine (TBZ; Ro 1-9569) was first synthesized in 1956 and was approved for the treatment of neuroses and psychoses/schizophrenia in Finland, the Netherlands, Switzerland, and the United Kingdom in 1958.1,2 It was withdrawn from the market in 1966 due to its failure as a potent neuroleptic. However, from ~1971 onward, TBZ was in widespread unapproved use for the treatment of involuntary movement disorders.1 It was not approved by the US Food and Drug Administration (FDA) until May 2008,1 and its current FDA approval is limited to the treatment of chorea in Huntington’s disease.3 This paper reviews the chemistry, pharmacology, pharmacokinetics, therapeutic use, tolerability, druginteraction potential, and dosing and administration of TBZ.

METHODS MEDLINE was searched (1950–February 2010) for English-language articles investigating any aspect of TBZ. Search terms included tetrabenazine, Ro 1-9569, Nitoman, benzoquinolizines, s and reserpine. The reference lists of identified articles were searched for other pertinent publications, particularly those that were not indexed in the 1950s and 1960s.

CHEMISTRY TBZ was initially identified as one member of a large group of 1,2,3,4,6,7-hexa-hydro-11bH-benzo[a]quinolizine derivatives that were synthesized in the search for a simpler chemical compound than reserpine that retained reserpine-like activity.2,4 These agents had ≥2 asymmetric centers, thus introducing the complexities of stereochemistry.4 The ketones, of which TBZ is an example, proved to be the most interesting of these compounds, and a variety of synthetic processes for TBZ were described between 1953 and 1961.4 The chemical structure of TBZ is shown in Figure 1. Its chemical name is ciss rac-1,3,4,6,7,11b-hexahydro9,10-dimethoxy-3-(2-methylpropyl)-2H-benzo[a] quinolizin-2-one.3 Its molecular weight is 317.43, its acid dissociation constant is 6.51, and its empiric formula is C19H27NO3.

MECHANISM OF ACTION TBZ works as a reversible inhibitor of vesicle monoamine transporter type 2 (VMAT-2), inhibiting the uptake of monoamines (serotonin, norepinephrine, and particularly dopamine) into the granular vesicles of presynaptic neurons.1 All monoamines are probably accumu-

332

lated within these vesicles by an identical mechanism, indicating the low specificity of the transporter.5 In fact, this transporter can, with similar efficiency, translocate dopamine, norepinephrine, epinephrine, serotonin, tyramine, histamine, and some nonphysiologic compounds.5 Catecholamine translocation is a form of secondary transport, being coupled to and driven by a transmembrane proton electrochemical gradient generated by inwardly directed proton pump adenosine triphosphatase acting on cytosolic adenosine triphosphate. In granules, the gradient drives uptake by activating the monoamine transporter, which is sensitive to both transmucosal pH and electrochemical gradients. The transporter catalyzes an electrodissipative catecholamine/antiport and, hence, forms a monoamine gradient.5 VMAT proteins should not be confused with plasma membrane monoamine transporters, which constitute a second monoamine uptake system present on monoaminergic cells, particularly neurons. Three classes of agents can block vesicular monoamine uptake: proton pump inhibitors, proton ionophores, and specific inhibitors of monoamine transport. TBZ (and reserpine) fall within the latter category. These agents work without altering the transmembrane pH and potential gradients of the vesicular membranes.5 Although reserpine and TBZ work by similar mechanisms, they can be readily distinguished from a mechanistic perspective.4,6 Reserpine is an irreversible inhibitor of VMAT-2, whereas TBZ is a reversible inhibitor. TBZ is 10- to 20-fold less potent than reserpine. Reserpine depletes monoamine stores to a greater extent than does TBZ (eg, reserpine depletes serotonin and norepinephrine stores to 10% of baseline, whereas TBZ depletes serotonin stores to 30%–50% of baseline and norepinephrine stores to 15%–25% of baseline). The time to peak effect is shorter and recovery of monoamine stores is faster with TBZ (16–24 hours) compared with reserpine (days–weeks).

CH3O N CH3O

CH3 CH2 O

Figure 1. Chemical structure of tetrabenazine.

CH CH3

D.R.P. Guay

PHARMACOLOGY Relatively few data are available regarding the pharmacology of TBZ in humans. Human synaptic vesicular monoamine transporter (SVMT) mRNA has been found in human brainstem (for 3.0 kilobase [kb] human SVMT) and hypothalamus (for 4.8 kb human SVMT), but it has not been found in high concentrations in human frontal cortex, parietal cortex, temporal cortex, occipital cortex, cingulate cortex, caudate, globus pallidum, thalamus, amygdala, cerebellum, or hippocampus.7 Significant human SVMT expression has also been found in human nigra compacta neurons.7 For example, high rates of specific (3H) binding of 15 nM dihydrotetrabenazine have been reported as follows in human brain: striatum caudate (766 fmoL/mg protein), striatum accumbens (751 fmoL/mg protein), striatum putamen (742 fmoL/mg protein), substantia nigra compacta (465 fmoL/mg protein), hypothalamus (245 fmoL/ mg protein), substantia nigra reticulata (141 fmoL/mg protein), external pallidum (128 fmoL/mg protein), and internal pallidum (115 fmoL/mg protein).5 All other areas exhibited <100 fmoL/mg protein-specific binding. Dihydrotetrabenazine binding to human caudate P1 and P3 membranes can be described by respective dissociation constant values of 4.12 and 4.86 nM and Bmax (total number of bound receptors) values of 482 and 356 fmoL/mg protein.7 Human SVMT exhibits nanomolar affinities for TBZ (inhibitory rate constant [K Ki] = 2.34 nM) and reserpine (K Ki = 73 nM).7 On the other hand, affinities are on the micromolar level for haloperidol (1.86 μM), serotonin (3.4 μM), mazindol (23 μM), nomifensin (416 μM), and D-amphetamine (381 μM). Affinities are on the millimolar level for dopamine, epinephrine, norepinephrine, and histamine. A controlled trial comparing TBZ, thiopropazate, and placebo in patients with Huntington’s chorea also measured cerebrospinal fluid (CSF) concentrations of monoamine metabolites.8 Administration of placebo was associated with CSF concentrations of homovanillic acid (HVA) and 5-hydroxyindole acetic acid (5-HIAA) that did not differ significantly from values in controls with no cerebral disease. Similar findings were noted in thiopropazate recipients. TBZ had no significant effect on CSF 5-HIAA concentrations, but was associated with a significant increase in mean CSF HVA concentrations (from 34.0 to 54.5 ng/mL; P < 0.001). There was no clear correlation between CSF metabolite concentrations and clinical response, suggesting that monitoring of CSF metabolite concentrations has no value in predicting the response to therapy.

The American Journal of Geriatric Pharmacotherapy

Postmortem brain tissue from patients with Huntington’s chorea who had and had not received TBZ therapy (11 and 7 patients, respectively) was analyzed for levels of dopamine, norepinephrine, serotonin, and their metabolites.9 In tissue from those who had received TBZ therapy, monoamine concentrations were reduced in all parts of the brain studied, although the reductions were significant only for dopamine in the caudate (P P < 0.01), norepinephrine in the amygdala (P P < 0.05), and norepinephrine and dopamine in the hippocampus (both, P < 0.05). Concentrations of HVA and serotonin were not significantly reduced, despite absolute serotonin reductions of 16% to 35%. Concentrations of 5-HIAA were not significantly different from baseline. The dopamine effect in the caudate supports evaluation of TBZ in the treatment of chorea, and the norepinephrine/serotonin effects in the limbic system suggest an explanation for the emergence of depression as a frequent adverse effect. The electroencephalographic (EEG) effects of TBZ during chronic therapy were quantitated in a human pharmacology study.4 EEG changes after chronic dosing included a slowing of spontaneous activity and an increase in amplitude. In patients with posttraumatic cerebral injury, TBZ was found to counteract the depressed alpha rhythm. In another EEG study, longterm administration of TBZ at oral doses of 250 to 375 mg/d (ie, much higher than would be contemplated in clinical use at present) led to a slowing of alpha rhythm, an increase in subalpha and theta activity, a reduction in fast beta frequency, and increased voltage, beginning within a few days after initiation of TBZ and peaking during the first week.2 These effects were interpreted as a manifestation of reduced central nervous system activity and vigilance. Changes in EEG parameters were varied, but patients with depression usually exhibited increased alpha activity, while those with schizophrenia had diffuse or localized slow activity of variable duration. In another EEG study, 40 patients received a single 100-mg intramuscular injection of TBZ.2 Changes in EEG parameters occurred in 29 patients (73%). These changes varied widely, depending on previous EEG patterns. EEG abnormalities were exacerbated in most patients with epilepsy.

PHARMACOKINETICS The pharmacokinetics of TBZ have not been well studied in humans. This is in part because of the regulatory environment of the late 1950s when TBZ was first being investigated for human use and because of the diff ficulty of quantitating the drug and its active α- and

333

The American Journal of Geriatric Pharmacotherapy

D.R.P. Guay

β-dihydro metabolites in human body fluid compartments at that time. In a Phase I mass-balance trial, 2 healthy volunteers received tritium-labeled TBZ 90 mg injected via intravenous bolus.4,10 The serum concentration–time profiles were consistent with a 2-compartment kinetic model; however, neither compartmental nor noncompartmental analyses were carried out. Radioactivity fell to minimum levels within 10 hours after administration. By day 3, no activity was detectable. Total radioactivity excretion was 40% in urine and 2.5% in feces by 24 hours after administration. After 48 hours, 54% of total radioactivity had been excreted. Of interest, no radioactivity was found in the CSF. As part of a pilot study of TBZ as a tracer compound for use in diagnostic medicine, the intracerebral pharmacokinetics of 11C-TBZ were evaluated by positron emission tomography in 2 healthy volunteers.11 Injection of 11C-TBZ (27.5 and 27.6 mCi) as an intravenous bolus was followed by 60 minutes of imaging (30-second to 10-minute frames). Brain uptake was rapid and substantial, similar to the uptake of 11C-flumazenil and 11C-tropanylbenzylate, ligands known to readily cross the blood–brain barrier. Egress from the brain was also rapid. The intracerebral concentration–time curves were of similar shape in the striatum, thalamus, cerebellum, and frontal cortex. No detailed pharmacokinetic analyses were performed in this study. The prehepatic systemic availability of TBZ is at least 75%.3 Administration with a high-fat/high-calorie breakfast did not affect the systemic bioavailability of TBZ, which was quantitated based on the Cmax and AUC for the α- and β-dihydrotetrabenazine metabolites.3 Quantitation of the metabolites rather than the parent compound was performed because serum concentrations of the parent compound were usually below the lower limit of detection of the assay in both single- and multipledose studies.12 After oral dosing, the serum Cmax of dihydrotetrabenazine (metabolites I and II) and desmethyldihydrotetrabenazine (metabolites VII and VIII) occurred at means of 1 to 1.5 and 2 hours, respectively.3 The principal circulating moieties in serum are the α- and β-dihydrotetrabenazine and α- and β-Odesmethyldihydrotetrabenazine metabolites (along with their glucuronide and sulfate conjugates).3 The pharmacokinetic parameters of the α- and d β-dihydrotetrabenazine metabolites exhibit stereoselectivity.12 In general, systemic exposure to the α metabolite exceeds exposure to the β metabolite.3 For example, the mean Cmax of the α metabolite has been reported to be 1.5- to 2-fold greater than that of the β metabolite,

334

whereas the corresponding increases were 1.3-fold and 1.3- to 1.4-fold for mean AUC and t1/2. However, there is substantial interpatient variability in all parameters, and the ratio of the α metabolite to the β metabolite can range from 5:1 to 8:1 in individual patients. Over a concentration range of 50 to 200 ng/mL, plasma protein binding of TBZ, α-dihydrotetrabenazine, and β-dihydrotetrabenazine ranged from 82% to 88%, 60% to 68%, and 59% to 63%, respectively.3 The steady-state pharmacokinetics of TBZ were evaluated in 4 patients receiving TBZ 12.5 to 37.5 mg TID for the treatment of tardive dyskinesia.13 The mean (SD) ratio of systemic exposure (ie, AUC) to the dihydro metabolite (sum of the α and β moieties) relative to systemic exposure to the parent compound was 148 (49) (range, 83–199). The apparent oral clearance of TBZ ranged from 252 to 1090 mL/min/kg. Plasma concentrations of the parent compound and the pooled α- and β-dihydro metabolites ranged from 0.24 to 6.22 ng/mL and 15.9 to 801 ng/mL, respectively. At concentrations of 10 and 25 ng/mL, the mean ratios of TBZ blood:plasma concentrations were 1.13 (0.11) and 1.19 (0.04), respectively. No parent compound was found in the urine of any patient. The bioavailability of TBZ in the systemic circulation was small and erratic (mean, 6.0% [2.6%]; range, 2%–8%). Pharmacokinetic studies of single intravenous and oral doses of TBZ were conducted in 7 patients with Huntington’s disease or tardive dyskinesia (Table I).14 The mean (SD) prehepatic systemic availability of TBZ was 81% (30%). However, due to extensive first-pass hepatic metabolism, the mean absolute oral bioavailability of the parent compound was only 4.9% (3.2%). The extent of metabolite accumulation (R Rm) was large (R Rm = 3.30 [2.76] after intravenous dosing, 35.0 [35.0] after oral dosing). There was great interindividual variation in almost all parameters for both the parent compound and the pooled α and β metabolites. The results of this study suggest that for therapeutic purposes, the dihydro metabolite may be preferable to TBZ due to its lesser extent of first-pass hepatic metabolism and plasma protein binding and its longer t1/2. After administration of TBZ 2 mg/kg SC followed by quantitative urine collection, 9 metabolites were found in urine (5 nonconjugated and 4 conjugated).15 Later studies found at least 19 metabolites.3 The major steps in TBZ metabolism are keto reduction at the C2 position, oxidation (to a hydroxyl group) at the 2′ position on the isobutyl side chain, and selective ether cleavage at the C9 position, followed by conjugation of the phenolics to glucuronic acid.15 The products of

D.R.P. Guay

The American Journal of Geriatric Pharmacotherapy

Table I. Pharmacokinetics of tetrabenazine (TBZ) and its dihydro metabolite (DHM; sum of the B and C moieties) after single intravenous doses of 10 mg administered over 1.5 to 3 minutes and single oral doses of 25 to 75 mg in 7 patients with Huntington’s disease or tardive dyskinesia.14 Values are mean (SD). %).

TBZ 1BSBNFUFS

*OUSBWFOPVT

0SBM

*OUSBWFOPVT

0SBM

%JTUSJCVUJPOU NJO

 



 



$- -NJO

 







VE BSFB, L

 







VE TT, L

 







.35 NJO .35TBZ .35%). .35 5m

 

– –

 

– –

–  

 

–  



."5TBZ NJO



 





CNBY OHN-



 

 

 

TNBY NJO



 

 

 

"6$ OHN-rNJO





 

 

VE BSFB WPMVNFPGEJTUSJCVUJPOVTJOHUIFBSFBNFUIPE7E TTWPMVNFPGEJTUSJCVUJPOBUTUFBEZTUBUF.35TBZNFBO 5m NFBOSFTJEFODFUJNFGPSNFUBCPMJUFBMPOF SFTJEFODFUJNFGPS5#;.35%).NFBOSFTJEFODFUJNFGPS%)..35 .35%). o.35TBZ ."5TBZNFBOBCTPSQUJPOUJNFGPS5#; 0OMZQBUJFOUTIBEFWBMVBCMFEBUBBGUFSJOUSBWFOPVTEPTJOH

keto reduction are α- and β-dihydrotetrabenazine; β-dihydrotetrabenazine and (–) α-dihydrotetrabenazine are pharmacologically inactive, whereas (+) α-dihydrotetrabenazine is an active metabolite.16 The primary entities in urine are the glucuronide conjugates. The α- and β-dihydrometabolites constitute <10% of a dose. The parent compound is not excreted in urine. Mass-balance studies in healthy volunteers found that 75% of a dose was eliminated in the urine and 7% to 16% in the feces.3 Figure 2 illustrates the proposed schema of TBZ metabolism. When metabolites I, II, and V were tested for pharmacologic activity (potentiation of ethanol narcosis), all were found to be less active than the parent compound.15 Keto reduction and O-dealkylation are mediated principally by the cytochrome P450 (CYP) 2D6 isozyme; O-dealkylation of α-dihydrotetrabenazine also involves CYP1A2.3 The t1/2 of α- and β-dihydrotetrabenazine ranges from 4 to 8 and 2 to 4 hours, respectively.3 Although the pharmacokinetics of TBZ and its metabolites have not been systematically evaluated in sub-

jects who do not express CYP2D6 (ie, poor metabolizers), it is likely that such subjects would have increased serum concentrations of α- and β-dihydrotetrabenazine, similar to concentrations in subjects coingesting a strong CYP2D6 inhibitor (eg, paroxetine).3 Thus, the prescribing information for TBZ recommends that CYP2D6 metabolizer status be determined before use of TBZ doses >50 mg/d, and those found to be poor metabolizers should not receive TBZ doses >50 mg/d.3 However, the test for CYP2D6 metabolizer status is available at few US laboratories, and the likelihood of adherence to this recommendation is low.

Special Populations There have been no formal studies of the pharmacokinetics of TBZ in pediatric or geriatric populations, specific racial/ethnic groups, or patients with renal impairment.3 In terms of gender effects, the mean Tmax of α- and β-dihydrotetrabenazine has been reported to be similar in men and women.12 However, the mean Cmax of the β metabolite was 1.25- to 1.3-fold greater

335

The American Journal of Geriatric Pharmacotherapy

D.R.P. Guay

CYP2D6

Glucuronidation/ sulfation

Metabolite VI C9-demethylation

Tetrabenazine CYP2D6

Metabolite I C2-ketoreduction (isomer 1)

CYP2D6

Metabolite VII C9-demethylation

CYP2D6

Metabolite III C2´-hydroxylation (isomers 1 and 2)

Glucuronidation/ sulfation Glucuronidation/ sulfation

Metabolite II C2-ketoreduction (isomer 2)

CYP2D6 CYP2D6

Metabolite IX C9-demethylation

Metabolite VIII C9-demethylation Glucuronidation/ sulfation

Glucuronidation/ sulfation

Metabolite IV C2´-hydroxylation (isomer 1)

Metabolite V C2´-hydroxylation (isomer 2)

Glucuronidation/ sulfation

Figure 2. Major metabolic pathways of tetrabenazine. CYP = cytochrome P450.

in women than in men, whereas the mean Cmax of the α metabolite did not differ between women and men. The mean AUC of the β metabolite was 1.5- to 2-fold greater in women than in men, whereas the mean AUC of the α metabolite did not differ between women and men. The mean t1/2 of the β metabolite was slightly longer in women (4–6 hours) compared with men (3–4.7 hours), whereas the mean t1/2 of the α metabolite did not differ between women and men. The clinical relevance of these gender differences in the pharmacokinetics of TBZ is not known. The pharmacokinetics of TBZ and its dihydro metabolites were compared in 12 subjects with mild (Child-Pugh class A) and moderate (Child-Pugh class B) hepatic impairment and 12 age-matched healthy control subjects.3 All received TBZ as a single 25-mg dose. In the patients with hepatic disease, serum concentrations of TBZ were greater than or equal to those of α-dihydrotetrabenazine. In the group with hepatic impairment, the Cmax was 7- to 190-fold higher than detectable serum concentrations in the control group. The mean t1/2 of TBZ was increased to ~17.5 hours (usual value, 6.5 hours). Exposure to α- and βdihydrotetrabenazine was increased by 30% to 39% compared with exposure in control subjects. The mean Tmax of the dihydrotetrabenazine metabolites was

336

slightly increased in the group with hepatic impairment compared with control subjects (1.75 and 1.0 hour, respectively). The mean t1/2 of both dihydrotetrabenazine metabolites was increased compared with that in control subjects (α metabolite: 10 vs 4–8 hours, β metabolite: 8 vs 2–4 hours).

Drug–Drug Interactions In vitro studies found that TBZ and its dihydro metabolites did not significantly inhibit CYP2D6, 1A2, 2C8, 2C9, 2C19, 2E1, or 3A.3 In addition, they did not significantly induce CYP1A2, 3A4, 2B6, 2C8, 2C9, or 2C19. Neither TBZ nor its dihydro metabolites appear likely to be substrates for or inhibitors of P-glycoprotein at clinically relevant concentrations in vivo. The interaction between a single dose of TBZ 50 mg and multiple-dose paroxetine 20 mg/d was evaluated in 25 healthy volunteers.3 Pretreatment with paroxetine (a strong inhibitor of CYP2D6) was associated with mean 30% and 3-fold increases in the αdihydrotetrabenazine Cmax and AUC, respectively, and mean 2.4- and 9-fold increases in β-dihydrotetrabenazine Cmax and AUC. The t1/2 of both dihydrotetrabenazine moieties was increased to ~14 hours (usual values for the α- and β-moieties, 4–8 and 2–4 hours, respectively).

D.R.P. Guay

Because reserpine and TBZ have similar pharmacologic properties, reserpine should not be used as an antihypertensive when TBZ is to be used.3 The pharmacologic properties of neuroleptics (traditional or atypical) and metoclopramide also are similar to those of TBZ; concurrent use with TBZ is likely to exaggerate sedative, parkinsonian, and extrapyramidal adverse effects, as well as to increase the risk of neuroleptic malignant syndrome. Concurrent use of TBZ with monoamine oxidase inhibitors should be avoided. Based on observations from rat and mouse studies suggesting that lithium may inhibit the biochemical and behavioral effects of TBZ,17,18 caution is warranted when considering combined use of these agents.

THERAPEUTIC USE TBZ was initially investigated as a potentially more eff ficacious, better tolerated treatment for schizophrenia than reserpine in >20 open-label and comparative trials,2,19 and it was subsequently approved for this indication. Its popularity waned fairly quickly for reasons that are unclear, although its neuroleptic potency was moderate, at best. TBZ is currently approved for use in the treatment of a wide range of hyperkinetic movement disorders, varying from country to country. In the United States, it is approved only for the treatment of chorea associated with Huntington’s disease, as only efficacy data for this indication were submitted in the New Drug Application for TBZ. However, TBZ is used off label for many other muscle disorders.

Case Studies and Series The identified case studies and series included 51 patients, 60% of them female (based on data for 50 patients), with an overall mean age of 56.9 years (range, 10–92 years) (based on data for 43 patients).20–49 Of these 51 patients, 8 had hemiballismus with/without hemichorea, 26 had tardive disorders, and 17 had miscellaneous disorders. The mean ages of these groups were 61.7 years (range, 18–92 years) (based on data available for 7 patients), 58.2 years (range, 21–77 years) (based on data for 24 patients), and 51.6 years (range, 10–81 years) (based on data for 12 patients), respectively. The corresponding proportions of women were 86% (based on data for 7 patients), 62%, and 47%. A total of 18 adverse events (AEs) were reported: 3 cases of bradycardia; 2 cases each of rigidity, drowsiness, and restlessness; and 1 case each of depression, cognitive impairment, hypomimia, parkinsonism, rash, myalgias, tremor, insomnia, and gait disturbance.

The American Journal of Geriatric Pharmacotherapy

Retrospective Trials The retrospective trials of the efficacy and tolerability of TBZ identified by the literature search are summarized in Table II.50–59 Of the 1142 patients included in the retrospective trials, 806 (71%) had complete cessation of clinical signs and symptoms, an excellent response, very good improvement, or marked improvement (terms differed by study); 55 (5%) had mild improvement or a slight response; and 281 (25%) had a doubtful response, no response, worsening, or no improvement. The clinical response to TBZ appeared to vary by disorder. At least moderate clinical improvement or response occurred in 100% of patients with chorea (n = 103), tics (n = 75), Tourette’s syndrome (n = 27), myoclonus (n = 23), facial dyskinesia/dystonia (n = 7), and non– drug-related buccolingualmasticatory syndrome (n = 5); 99% of patients with dystonia (n = 163); 95% of patients with tardive disorder (n = 311); 80% of patients with hemiballismus (n = 5); 69% of patients with Huntington’s chorea (n = 85); 26% of patients with Meige’s syndrome (n = 31); and 0% of patients with parkinsonian tremor (n = 6) or intention tremor (n = 4). A total of 502 AEs occurred in the retrospective trials. The most frequently occurring AEs were drowsiness/ fatigue (30%), parkinsonism (27%), depression (14%), insomnia (9%), akathisia (8%), nervousness/anxiety (8%), and nausea/vomiting (4%). All other AEs occurred at a frequency of ≤3%.

Open-Label Trials A total of 881 patients participated in the open-label trials of TBZ monotherapy (16 trials) and combination therapy (9 trials) (Table III).60–84 Because it is difficult to draw conclusions regarding the efficacy of TBZ from the trials of combination therapy, which varied widely in design and quality, the 9 trials of combination therapy (represented by 10 publications) are not discussed here. Summary data from the 16 trials of TBZ monotherapy (represented by 15 publications) follow. A total of 628 patients participated in the open-label trials of TBZ monotherapy (range per trial, 5–217), representing a wide variety of hyperkinetic movement disorders. These included chorea in Huntington’s disease (n = 217), tardive dyskinesia (n = 188), Tourette’s syndrome (n = 26), generalized dystonias (n = 19), cranial dystonias (n = 57), tardive dystonia (n = 15), facial dystonias (n = 25), hemidystonia (n = 3), spontaneous oral dyskinesias (n = 6), olivocerebellar atrophy (n = 2), dystonia musculorum deformans (n = 6), spasmodic torticollis (n = 6), choreoathetosis (n = 2), Lesch-

337

338

)$ O

OPOoESVHSFMBUFE #-.TZOESPNF O

PUIFS IZQFSLJOFTJBT O

$IPSFB O

IFNJCBMMJTNVT O

EZTUPOJB O

JOUFOUJPO USFNPS O

QBSLJOTPOJBO USFNPS O

%BMCZ

1PQVMBUJPO

1BLLFOCFSH

"VUIPST

5#;oNHEPWFSTFWFSBM oXLQFSJPET BMUFSOBUJOHXJUI QIFOPCBSCJUBMPSBOUJQBSLJOTPOJBO ESVHT RVBTJDPOUSPMMFEEFTJHO XJUIPVUQMBDFCPPSCMJOEJOH

5#;oNHEGPSNP

3FHJNFO

$IPSFB  IBEBHPPE SFTQPOTF DPNQMFUFTVQQSFTTJPO PGBCOPSNBMNPWFNFOUT

  IBEBNPEFSBUFSFTQPOTF oSFEVDUJPOJO BCOPSNBMNPWFNFOUT

)FNJCBMMJTNVT  IBE HPPESFTQPOTF   XBT VODIBOHFE %ZTUPOJB  IBEB NPEFSBUFSFTQPOTF    XBTVODIBOHFE *OUFOUJPOUSFNPS  XFSF VODIBOHFE   IBE XPSTFOJOHPGUSFNPST

)$(PPEFGGFDU DPOTJEFSBCMF EFDSFBTFJOBCOPSNBM NPWFNFOUT XJUIPVUDPNQMFUF DFTTBUJPO JO 

 NPEFSBUFFGGFDUJO 

EPVCUGVMFGGFDUJO 

 BOEOPFGGFDUJO 

#-.TZOESPNFDPNQMFUFDFTTBUJPO PGBCOPSNBMNPWFNFOUTJO 

HPPEFGGFDUJO 

BOENPEFSBUFFGGFDUJO 

0UIFSIZQFSLJOFTJBTDPNQMFUF DFTTBUJPOJO   QBUJFOU IBEIFNJCBMMJTNVT

HPPEFGGFDU JO 

BOENPEFSBUF FGGFDUJO 

3FTQPOTFEFWFMPQFERVJDLMZ XJUIJOoE BOEEJTBQQFBSFE BTSBQJEMZXIFO5#;XBT XJUIESBXO

&GGJDBDZ0VUDPNFT

DPOUJOVFE

"&TXFSFSFQPSUFEJOQBUJFOUT 

JODMVEJOHGBUJHVFJO IZQFSIJESPTJTJO BOEDPOGVTJPO  QIPUPQIPCJB BOEBNFOPSSIFBJO FBDI"&TEJTBQQFBSFEPOSFEVDUJPO PG5#;EPTFCZoNHE

"&TJODMVEFEESPXTJOFTTJOBiGFXw QBUJFOUTBOEQBSLJOTPOJTNJOQB UJFOUQBSLJOTPOJTNEJTBQQFBSFE XIFO5#;EPTFXBTEFDSFBTFEGSPN UPNHE

5PMFSBCJMJUZ0VUDPNFT

Table II. Efficacy and tolerability data from retrospective trials of tetrabenazine (TBZ) in patients with hyperkinetic movement disorders.

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

)$ O 5% O DIPSFBPG )$NFBO5#;EPTF NHE VOLOPXODBVTFBOEIFNJCBMMJTNVT SBOHF oNHE GPS OFBDI 4ZEFOIBNTDIPSFB oE TFOJMFDIPSFB DIPSFPBUIFUPTJT  0UIFSEJBHOPTFTNFBO5#;EPTF  PSPMJOHVBMEZTLJOFTJB QTFVEPCVMCBS NHE SBOHF oNHE

QBMTZ BOENPUPSIZQFSBDUJWJUZ GPSoE OFBDI QBUJFOUTXJUI)$ XFSFUSFBUNFOUOBJWF

,JOHTUPO

5#;oNHEGPSXLoZ

3FHJNFO

)$ O

DIPSFBEVFUPTUSPLF O

1PQVMBUJPO

"TUJOBOE (VNQFSU

%BMCZ DPOU

"VUIPST

Table II (continued).

)$.BSLFEJNQSPWFNFOUJO  

NPEFSBUF JNQSPWFNFOUJO 

TMJHIUJNQSPWFNFOUJO 

OPDIBOHFJO 

5%.BSLFEJNQSPWFNFOUJO  

TMJHIUJNQSPWFNFOU JO 

0UIFSEJBHOPTFT.BSLFE JNQSPWFNFOUJO  BOE TMJHIUJNQSPWFNFOUJO 

1BUJFOUT  XFSFiNVDI CFUUFSw OPDIPSFJGPSN NPWFNFOUT

  XFSF iCFUUFSw TPNFDIPSFJGPSN NPWFNFOUT

  XFSFiUIF TBNFw OPJNQSPWFNFOU

1BSLJOTPOJBOUSFNPS   XFSFVODIBOHFE   IBE XPSTFOJOHPGUSFNPST

&GGJDBDZ0VUDPNFT

DPOUJOVFE

%FBUIT VOSFMBUFEUP5#;UIFSBQZ  QBUJFOUTEJTDPOUJOVFEEVFUP"&T JSSJUBCJMJUZBOEEFDSFBTFJOQPMZNPSQI DPVOUJOFBDI BOEEVFUPMBDLPG FGGFDU"&TJODMVEFENJMEEFQSFTTJPO BOEQBSLJOTPOJTNJOQBUJFOUT  ESPXTJOFTTJOQBUJFOUT BOEOBVTFB JSSJUBCJMJUZ OBVTFBBOEMFUIBSHZ BOE CSBEZDBSEJBJOQBUJFOUFBDI

1BUJFOUT  EJTDPOUJOVFE5#; EVFUP"&T IBMMVDJOBUJPOT TVJDJEBM EFQSFTTJPO BOEXJUIESBXBM EFQSFTTJPOJOFBDI

5PMFSBCJMJUZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

339

340

+BOLPWJDBOE 'PSE

"VUIPST

1PQVMBUJPO

1BUJFOUTXJUICMFQIBSPTQBTN PSPGBDJBMDFSWJDBMEZTUPOJBT PS CPUI .FJHFTTZOESPNF

Table II (continued). &GGJDBDZ0VUDPNFT

1BUJFOUTSFDFJWFE5#;BTB 5#;NBSLFEJNQSPWFNFOU TFDPOEMJOFBHFOUBGUFSUIF NP JO  GBJMVSFPGDMPOB[FQBN GPS NPOPUIFSBQZ DPNCJOBUJPO CMFQIBSPTQBTN PS UIFSBQZ

NJMEJNQSPWFNFOU USJIFYZQIFOJEZM GPS NP JO  PSPNBOEJCVMBSEZTUPOJBT  NPOPUIFSBQZ DPNCJOBUJPO SFDFJWFE5#;NPOPUIFSBQZ UIFSBQZ

OPJNQSPWFNFOU BOESFDFJWFEDPNCJOBUJPO XPSTFOJOHJO BMM SFHJNFOT5#; MJUIJVN O

SFDFJWJOHNPOPUIFSBQZ

5#; MJUIJVN USJIFYZQIFOJEZM "SFTQFDUJWF    O

5#; USJIFYZQIFOJEZM BOEPG5#; MJUIJVN O O

BOE5#; DMPOB[FQBN  

USJIFYZQIFOJEZM O

 MJUIJVN O

DMPOB[FQBN O

BOE 1BUJFOUTSFDFJWFEOPO5#; DBSCJEPQBMFWPEPQB O

DPNCJOBUJPOT USFBUNFOUXJUI SFDJQJFOUTFYIJCJUFEBNJMEPS DPNCJOBUJPOXBTBMMPXFE

USBOTJFOUFGGFDUDPSSFTQPOEJOH 1BUJFOUTXFSFDPODVSSFOUMZ QSPQPSUJPOTXJUINBSLFEPS QBSUJDJQBUJOHJOB%# $0USJBMPG MBTUJOHJNQSPWFNFOUXFSF  TBZ    BOE CFDBVTFESVHDPNCJOBUJPOT XFSFVTFEJOBSFTQFDUJWF     BOEPGUIFTFQBUJFOUT <PGQBUJFOUT> JUJT EJGGJDVMUUPBTDSJCFUIFSBQFVUJD FGGFDUTUPJOEJWJEVBMBHFOUT

3FHJNFO

DPOUJOVFE

/P"&TSFQPSUFEJO5#;SFDJQJFOUT

5PMFSBCJMJUZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

3FWJFXPGDBTFTPG5%TVDDFTTGVMMZ 5#;NH5*% XJUIUJUSBUJPO USFBUFEXJUI5#; GFNBMFBHF CBTFEPOSFTQPOTFNFBOEBJMZ SBOHF oZ

EPTF NH SBOHF oNH

1BUJFOUT XJUIEZTUPOJBT .FBO 4% 5#;EPTF  QSFEPNJOBOUMZDSBOJBM   NHE SBOHF o HFOFSBMJ[FE  NHE NFBOEVSBUJPOPG QSFEPNJOBOUMZDFSWJDBM

XJUI UIFSBQZ   NP SBOHF  5% XJUI5%ZTU XJUI oNP

5PVSFUUFTTZOESPNF XJUI)$  5#;XBTDPNCJOFEXJUIMJUIJVN XJUINZPDMPOVT XJUI JOQBUJFOUT 

PUIFSEJBHOPTFT

8BUTPOFUBM

+BOLPWJDBOE #FBDI

.BYJNVNEBJMZEPTFPG5#; oNH NFBO NH

1BUJFOUTXJUI5%XJUIXJUIPVU UBSEJWFBLBUIJTJB

'BIO

3FHJNFO

1PQVMBUJPO

"VUIPST

Table II (continued). 5PMFSBCJMJUZ0VUDPNFT

DPOUJOVFE

&YDFMMFOUDMJOJDBMSFTQPOTF 5#;XBTEJTDPOUJOVFEJO DPNQPTJUFPGQBUJFOU DBSFHJWFS QBUJFOUT 

EVFUP"&T TQPVTF BOESBUFSBTTFTTNFOUT JO EVFUPMBDLPGFGGJDBDZ EVF PGQBUJFOUTXJUIEZTUPOJBT  UPTQPOUBOFPVTSFTPMVUJPOPG XJUI5% XJUI5%ZTU  NPWFNFOUEJTPSEFS EVFUP XJUI5PVSFUUFTTZOESPNF EFBUI VOSFMBUFEUP5#;

GPS XJUI)$ XJUI USBWFMGJOBODJBMSFBTPOT GPS NZPDMPOVT BOEXJUIPUIFS NJTDFMMBOFPVTSFBTPOT BOEEVFUP EJBHOPTFTQBUJFOUTXJUI TVQFSJPSSFTVMUTXJUICPUVMJOVN 5PVSFUUFTTZOESPNFIBEQPPSFTU UPYJO SFTQPOTFSBUFTPGFYDFMMFOU "&TPDDVSSFEJOPG5#; DMJOJDBMSFTQPOTFJOUIPTFXJUI SFDJQJFOUT BHHSFTTJWFEPTFUJUSBUJPO JEJPQBUIJDEZTUPOJBTEJEOPU XBTFNQMPZFEUPNBYJNJ[F EJGGFSTVCTUBOUJBMMZCZUZQFPG SFTQPOTF NBKPS"&TJODMVEFE EZTUPOJB ESPXTJOFTTGBUJHVFJO

4DPSFTPOQPJOUSBUJOHTDBMF  "&TDPOTJEFSFEiOPUFXPSUIZwXFSF OPOF NJOJNBM NJME  ESPPMJOHJOQBUJFOUTBOE NPEFSBUF TFWFSF EFDSFBTFE QBSLJOTPOJTNJO GSPNCBTFMJOFCZQPJOUTJO QBUJFOUT QPJOUTJOQBUJFOUT QPJOUTJOQBUJFOUT BOEQPJOU JOQBUJFOUT

1BUJFOUTIBENBSLFEJNQSPWFNFOU  1BSLJOTPOJTNBQQFBSFEJOQBUJFOUT IBENPEFSBUFJNQSPWFNFOU 

IBEOPJNQSPWFNFOUOP QBUJFOUDPVMECFUBQFSFEPGG5#; BOESFNBJOGSFFPGBMMUBSEJWF TZNQUPNT

&GGJDBDZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

341

342

.FBO 4% 5#;EPTF    NHE NFEJBO NHESBOHF  oNHE NFBOEVSBUJPOPG UIFSBQZ NP

1BUJFOUT  FYIJCJUFEDMJOJDBM JNQSPWFNFOUNFBO$(*$TDPSF  NJMEJNQSPWFNFOU IBE $(*$TDPSF  WFSZHPPE JNQSPWFNFOU PGUIFTF SFTQPOEFST IBEDIPSFB IBEGBDJBMEZTUPOJBTPSEZTLJOFTJBT FBDIIBEUSVODBMEZTUPOJBTBOE UJDT FBDIIBE5%BOEUBSEJWF BLBUIJTJBUIPTFXJUIGBDJBM EZTLJOFTJBTEZTUPOJBTIBE TJHOJGJDBOUMZHSFBUFSJNQSPWFNFOU WTUIPTFXJUIUSVODBMEZTLJOFTJBT EZTUPOJBT P UIPTFXJUI DIPSFBIBETJHOJGJDBOUMZHSFBUFS JNQSPWFNFOUWTUIPTFXJUI EZTUPOJBT P  OPTJHOJGJDBOU EJGGFSFODFJOSFTQPOTFCZTFY

1BUJFOUT XJUIGBDJBM EZTLJOFTJBTPSEZTUPOJBT XJUI USVODBMEZTUPOJBT XJUI DIPSFB XJUI5PVSFUUFT TZOESPNF XJUI5% XJUI UBSEJWFBLBUIJTJB

&GGJDBDZ0VUDPNFT

1BMFBDVFUBM

3FHJNFO *OUIFQBUJFOUTJOXIPNUIF FGGFDUPGMJUIJVNDPVMECF BTTFTTFE   SFQPSUFE BEEJUJPOBMCFOFGJU 1BUJFOUTWPMVOUBSJMZ EJTDPOUJOVFE5#;BGUFSNPPG QFBLCFOFGJU XJUISFMBQTF PDDVSSJOHJO 

SFRVJSJOH SFTVNQUJPOPG5#;UIFSBQZ

1PQVMBUJPO

+BOLPWJDBOE #FBDI DPOU

"VUIPST

Table II (continued).

DPOUJOVFE

5#;XBTEJTDPOUJOVFECZPG QBUJFOUT EVFUPXPSTFOJOH  EVFUPMBDLPGCFOFGJU EVFUP "&T QBUJFOUTEJFE VOSFMBUFEUP 5#; "&TPDDVSSFEJOQBUJFOUT   TPNOPMFODF XFBLOFTT BOE BQBUIZJOQBSLJOTPOJTNJO EFQSFTTJPOBOEBDVUFBLBUIJTJBJO FBDIEZTQOFBBOEQTZDIPHFOJDHBJU EJTUVSCBODFJOFBDI UIFDBTFTPG EFQSFTTJPOXFSFUSFBUFEXJUI443*T

QBSLJOTPOJTNJO EFQSFTTJPOJO  JOTPNOJBJO OFSWPVTOFTT BOYJFUZBOEBLBUIJTJBJOFBDI BOEOBVTFBWPNJUJOHJONPTUPG UIFTF"&TXFSFEPTFSFMBUFEBOE EFDSFBTFEPSDFBTFEXJUIEPTF BEKVTUNFOUPWFSBMM"&SBUF EFDSFBTFEXIFO5#;EPTFXBT SFEVDFEJODPNQBSFEXJUI QSFSBUF WT SFTQFDUJWFMZ

5PMFSBCJMJUZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

1BUJFOUT XJUI5%  XJUIEZTUPOJBT XJUIDIPSFB XJUIUJDT XJUINZPDMPOVT TPNFQBUJFOUTIBENPSFUIBO POFEJBHOPTJT NPTUEJTPSEFST XFSFPGNPEFSBUFUPTFWFSFJOUFOTJUZ NFBO 4% BHFBUPOTFUPGEJTPSEFS    ZNFBOBHFBUJOJUJBUJPOPG 5#;   Z

,FOOFZFU al .FBO 4% 5#;EPTFBUGJOBMTUVEZ WJTJU   NHESFRVJSFE EPTFNHENFBOEVSBUJPOPG USFBUNFOU   Z NBYJNVN Z

3FHJNFO "UUIFFOEPGFWBMVBUJPO +BOVBSZ 

PGQBUJFOUTXJUI5%  XJUIDIPSFB XJUIEZTUPOJBT XJUIUJDT BOEXJUI NZPDMPOVTXFSFTUJMMUBLJOH5#;     BOEPG UIFSFTQFDUJWFHSPVQTIBEBDMJOJDBM SFTQPOTFPG NBSLFESFEVDUJPO JOBCOPSNBMNPWFNFOUT FYDFMMFOU JNQSPWFNFOUJOGVODUJPO PS  NPEFSBUFSFEVDUJPOJOBCOPSNBM NPWFNFOUT WFSZHPPE JNQSPWFNFOUJOGVODUJPO  USFBUNFOUXBTUFNQPSBSJMZ TVTQFOEFE oE JOQBUJFOUT PGQBUJFOUTIBEXPSTFOJOHPG UIFJSEJTPSEFS MFBEJOHUP EJTDPOUJOVBUJPOPG5#; P  WTUIPTFXJUIOPXPSTFOJOHPO UFNQPSBSZTVTQFOTJPO 

&GGJDBDZ0VUDPNFT

"&TPDDVSSFEJOPGQBUJFOUT ESPXTJOFTTJO QBSLJOTPOJTNJO  EFQSFTTJPOBOEBLBUIJTJBJO FBDI OBVTFBWPNJUJOHJO BOYJFUZ JO BMM"&TXFSFEPTFEFQFOEFOU BOEBCBUFEXJUIEPTFSFEVDUJPO QBSLJOTPOJTNXBTUSFBUFEXJUI BNBOUBEJOF MFWPEPQB BOEEPQBNJOF BHPOJTUT XJUIWBSJBCMFTVDDFTT "&QSPGJMFJOUIFQBUJFOUTXJUI MPOHFTUEVSBUJPOTPGGPMMPXVQXBT TJNJMBSUPUIBUJOFOUJSFTUVEZ QPQVMBUJPOBNPOHBMM"&TSFQPSUFE JOUIFTFQBUJFOUT POMZBHFXBTB TJHOJGJDBOUQSFEJDUPSPGQBSLJOTPOJTN P

5PMFSBCJMJUZ0VUDPNFT

)$)VOUJOHUPOTDIPSFB#-.CVDDPMJOHVBMNBTUJDBUPSZ"&TBEWFSTFFWFOUT5%UBSEJWFEZTLJOFTJB%#EPVCMFCMJOE$0DSPTTPWFS5%ZTUUBSEJWFEZTUPOJB$(*$$MJOJDBM(MPCBM*NQSFTTJPO PG$IBOHF443*TTFMFDUJWFTFSPUPOJOSFVQUBLFJOIJCJUPST

1PQVMBUJPO

"VUIPST

Table II (continued).

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

343

344

4XBTIFUBM61 &WBMVBUPSCMJOEFE

'PHBOE1BLLFOCFSH60

"VUIPST

3FHJNFO

&GGJDBDZ0VUDPNFT

1BUJFOUT XJUIEZTUPOJB NVTDVMPSVNEFGPSNBOT XJUITQBTNPEJDUPSUJDPMMJT  FBDIXJUICJMBUFSBMBUIFUPTJT BOE)$ BOEFBDIXJUI QFSJOBUBMIZQPYJBXJUIBUIFUPTJT  IFNJCBMMJTNVT QSPHSFTTJWF BUIFUPJEEZTUPOJB IZQPYJD FODFQIBMPQBUIZBUIFUPTJT  BOELFSOJDUFSVTBUIFUPTJT

5#;NH#*%JOJUJBMMZ JODSFBTFEJONHE JODSFNFOUTRE .BYJNVN5#;EPTF  oNHE NFBO  NHE FGGFDUJWF5#; EPTF oNHE NFBO NHE EVSBUJPOPG UIFSBQZ o XL

&JHIUQBUJFOUT  SFTQPOEFE XJUIEZTUPOJBNVTDVMPSVN EFGPSNBOT XJUI)$ FBDI XJUITQBTNPEJDUPSUJDPMMJT CJMBUFSBMBUIFUPTJT QFSJOBUBM IZQPYJBXJUIBUIFUPTJT BOE IFNJCBMMJTNVT IBEOP SFTQPOTFXJUIEZTUPOJB NVTDVMPSVNEFGPSNBOT XPSTFOFEEVSJOHUIFSBQZ

1BUJFOUT XJUI)$  5#;oNHE )$BMNPTUDPNQMFUF XJUIOPOoESVHSFMBUFE#-. QJNP[JEFoNHEGPSoNP EJTBQQFBSBODFPG TZOESPNF DPNCJOBUJPOUIFSBQZXBTVTFE IZQFSLJOFTJBTJO   CFDBVTFQJNP[JEFNPOPUIFSBQZ NPEFSBUFFGGFDUXJUI PCWJPVTEFDSFBTFJO IBECFFOBTTPDJBUFEXJUI EFWFMPQNFOUPGUPMFSBODF BCOPSNBMNPWFNFOUTJO XJUIJOoXLPGJOJUJBUJPOJO  

QSFWJPVTQJMPUUSJBM

#-.TZOESPNFBMNPTU DPNQMFUFEJTBQQFBSBODFPG IZQFSLJOFTJBTJO   NPEFSBUFFGGFDUXJUIPCWJPVT EFDSFBTFJOBCOPSNBM NPWFNFOUTJO 

5IFSBQFVUJDFGGFDUEFWFMPQFE SBQJEMZ PWFSoE BOE EJTBQQFBSFEKVTUBTSBQJEMZ XIFOESVHTXFSF EJTDPOUJOVFE

1PQVMBUJPO

DPOUJOVFE

"&TJODMVEFETFWFSFESPXTJOFTT JOQBUJFOUT JOTPNOJBJO  EFQSFTTJPOJO BOEGFFMJOHTPG VOSFBMJUZBOENZPDMPOVTJO FBDIUPMFSBODFEFWFMPQFEJO PGUIF  XJUIJNQSPWFNFOU FBDIXJUIQFSJOBUBMIZQPYJB XJUIBUIFUPTJT EZTUPOJB NVTDVMPSVNEFGPSNBOT BOE TQBTNPEJDUPSUJDPMMJT

%SPXTJOFTTSFQPSUFEJOiBGFXw QBUJFOUTQBSLJOTPOJTN EFWFMPQFE CVUTZNQUPNT SFNJUUFEXJUIEPTFSFEVDUJPO

5PMFSBCJMJUZ0VUDPNFT

Table III. Efficacy and tolerability data from open-label trials of tetrabenazine (TBZ) in patients with hyperkinetic movement disorders.

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

.D-FMMBO

4XBTI61 FUBM DPOU

CO

"VUIPST

Table III (continued).

5#;NH5*%YE UJUSBUFEPWFSE

"NBOUBEJOFNH5*%Y E UJUSBUFEPWFSE

1BUJFOUTSFDFJWFE5#;GJSTU BOESFDFJWFEBNBOUBEJOF GJSTU

3FHJNFO

1BUJFOUT  JNQSPWFE XJUI5#; XJUIBMNPTU DPNQMFUFTVQQSFTTJPOPG BCOPSNBMNPWFNFOUTJO     IBEOP JNQSPWFNFOUBNBOUBEJOF IBEOPFGGFDUPOBCOPSNBM NPWFNFOUTPSNFOUBMTUBUVT

&GGJDBDZ0VUDPNFT

1BUJFOUT PGUIFN 1BUJFOUTDPOUJOVFE 5#;NH#*%JOJUJBMMZ  JOQBUJFOUT

XJUI)$ XJUI JODSFBTFECZNHERoE USFBUNFOUGPSȑXL DIPSFBEVFUPDFSFCSPWBTDVMBS VOUJMBQQFBSBODFPGUPYJDJUZ )$$PNQMFUFPSBMNPTU PSDPNQMFUFTVQQSFTTJPOPG DPNQMFUFTVQQSFTTJPOPG EJTFBTF XJUIDIPSFBEVFUP QPTUFODFQIBMJUJDQBSLJOTPOJTN BCOPSNBMNPWFNFOUT NPWFNFOUTJO 

 XJUIDIPSFBEVFUPlEPQB .BJOUFOBODFEPTFJOUIPTF TVQQSFTTJPOPGNPWFNFOUT JOJEJPQBUIJDQBSLJOTPOJTN  USFBUFEGPSȑXL o XJUIJODSFBTFEGVODUJPOJO 

OPDIBOHFJO 

XJUIDIPSFBEVFUP NHE NFBO NHE  JOUSBDSBOJBMUVCFSDVMPNB EVSBUJPOPGUIFSBQZJOUIPTF 7BSJPVTDIPSFBUZQFT XJUIQPTUUSBVNBUJDDIPSFB USFBUFEGPSȑXL oNP $PNQMFUFPSBMNPTUDPNQMFUF XJUIIFNJCBMMJTNVT XJUI NFBO NP

TVQQSFTTJPOPGNPWFNFOUTJO FTTFOUJBMUSFNPS XJUI  

TVQQSFTTJPOPG IFSFEJUBSZBUBYJB BOEXJUI NPWFNFOUTXJUIJODSFBTFE PMJWPQPOUPDFSFCFMMBSBUSPQIZ GVODUJPOJO 

OP DIBOHFJO 

BOE XPSTFOFENPWFNFOUTJO  

.JTDFMMBOFPVT1BUJFOUXJUI IFNJCBMMJTNVTIBEDPNQMFUF PSBMNPTUDPNQMFUF TVQQSFTTJPOPGNPWFNFOUT QBUJFOUTXJUIIFSFEJUBSZ BUBYJBIBETVQQSFTTJPOPG NPWFNFOUTXJUIJODSFBTFE GVODUJPOJOSFNBJOJOH QBUJFOUT IBEOPDIBOHFBOE IBEXPSTFOFENPWFNFOUT

1BUJFOUTXJUIMPOHTUBOEJOH HC

1PQVMBUJPO

DPOUJOVFE

%JTDPOUJOVBUJPOT EFBUI EVFUP XPSTFOJOHPGNPWFNFOUT GFMU ESVHXBTJOFGGFDUJWF EVFUP TFMGQPJTPOJOH EVFUP"&T  "&TBTTPDJBUFEXJUI EJTDPOUJOVBUJPOXFSFESPXTJOFTT DBTFT

EFQSFTTJPO 

BLBUIJTJB BOEDPOGVTJPO FBDI

BOE BUBYJB BHJUBUJPO OBVTFB BOE SFEVDFEUPMFSBODFUPCFFS FBDI PWFSBMM UIFOVNCFSTPG QBUJFOUTBGGFDUFEBOETFSJPVTMZ BGGFDUFECZ"&TXFSFESPXTJOFTT BOE SFTQFDUJWFMZ

EFQSFTTJPO BOE

BLBUIJTJB BOE

 BUBYJB BOE

DPOGVTJPO BOE

OBVTFB BOE

 QBSLJOTPOJTN BOE

IJDDVQT BOE

BOESFEVDFECFFS DPOTVNQUJPO BOE POMZ QBUJFOUTIBEOP"&T

%VSJOH5#;UIFSBQZ ESPXTJOFTT BOEDPOGVTJPOXFSFSFQPSUFEJO QBUJFOUTFBDIOP"&TSFQPSUFE XJUIBNBOUBEJOF

5PMFSBCJMJUZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

345

346

,B[BNBUTVSJFUBM &WBMVBUPSCMJOEFE

"VUIPST

Table III (continued).

1BUJFOUTXJUI5%

1PQVMBUJPO 5#;NHEYE JODSFBTFE UPNHEYEBOE UIFOUPNHEYE POMZUIPTFOPUSFTQPOEJOH UPNHEIBEJODSFBTFUP NHE

5#;QFSJPEXBTQSFDFEFECZ XLCBTFMJOFQMBDFCPQFSJPE GPMMPXFECZXLQMBDFCP QFSJPE OFVSPMFQUJD XJUIESBXBM

3FHJNFO "GUFSXLPG5#;UIFSBQZ  QBUJFOUT  IBEUPUBM EJTBQQFBSBODFPGEZTLJOFUJD NPWFNFOUT   IBEB oEFDSFBTF    IBEBoEFDSFBTF    IBEBo EFDSFBTF BOE  IBEB oEFDSFBTFPSXPSTFOJOH PWTGJSTUQMBDFCP QFSJPE BGUFSXLPG5#; UIFSBQZ DPSSFTQPOEJOHWBMVFT XFSF 

 

 

  

BOE   P WTGJSTUQMBDFCPQFSJPE BGUFS XLPG5#;UIFSBQZ DPSSFTQPOEJOHWBMVFTXFSF  

 

 

 

 BOE   QBUJFOUTXFSF SFDFJWJOHNHE  P WTGJSTUQMBDFCPQFSJPE JOUFOTJUZ PGNPWFNFOUTBMTPEFDSFBTFE JONPTUQBUJFOUTCZXLBGUFS 5#;EJTDPOUJOVBUJPO EZTLJOFUJD NPWFNFOUTIBEBMNPTU SFUVSOFEUPCBTFMJOFMFWFMTOP DIBOHFTJOQBUJFOUCFIBWJPST EVSJOHTUVEZ

&GGJDBDZ0VUDPNFT

DPOUJOVFE

%JTDPOUJOVBUJPOT BMMEVSJOH XFFLT PG5#;QFSJPE

 EVFUPEJTDIBSHF EVFUPB QTZDIPUJDFYBDFSCBUJPO BOE EVFUP"&T"&MFBEJOHUP EJTDPOUJOVBUJPOXBTTFWFSF NBMBJTF O OPPUIFS iTJHOJGJDBOUw"&TSFQPSUFE

5PMFSBCJMJUZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

1BLLFOCFSHBOE'PH

"VUIPST

Table III (continued).

1BUJFOUTXJUIPSBMEZTLJOFTJBT BOEXJUIIZQFSLJOFTJBTPG FYUSFNJUJFT

1PQVMBUJPO

&GGJDBDZ0VUDPNFT

5PMFSBCJMJUZ0VUDPNFT

DPOUJOVFE

5#;oNHE NFBO "&TJODMVEFEQBSLJOTPOJTNBOE 5#;NPOPUIFSBQZ NHE XJUIXJUIPVU i*NNFEJBUFSFTQPOTFwXBT TJBMPSSIFB FBDI

QJNP[JEFoNHE NFBO  UPUBMEJTBQQFBSBODFPG NHE  DPNCJOBUJPO EZTLJOFTJBTJO   HFOFSBMMZVTFEPOMZXIFOPS BOEPCWJPVTSFEVDUJPOJO CPUINPOPUIFSBQJFTGBJMFE    BGUFSNP  NFBOEVSBUJPOPG5#; DPOUJOVFEUPUBM NPOPUIFSBQZ NP SBOHF EJTBQQFBSBODFPGEZTLJOFTJBT oNP NFBOEVSBUJPOPG JO 

DPOUJOVFE DPNCJOBUJPOUIFSBQZ NP PCWJPVTSFEVDUJPOJO SBOHF oNP



SFEVDUJPOEPVCUGVM JO 

OPDIBOHF GSPNCBTFMJOFJO 

 BOEEBUBOPUBWBJMBCMFJO  

$PNCJOBUJPOUIFSBQZ i*NNFEJBUFSFTQPOTFwXBT UPUBMEJTBQQFBSBODFJO  BOEOPDIBOHFJO  BGUFSNP DPOUJOVFE UPUBMEJTBQQFBSBODFJO 

PCWJPVTSFEVDUJPO JO 

SFEVDUJPO EPVCUGVMJO 

OP DIBOHFGSPNCBTFMJOFJO 

BOEEBUBOPUBWBJMBCMF JO  CPUIJOTUBODFT PGOPDIBOHFJOWPMWFEUIF QBUJFOUXJUIIZQFSLJOFTJBTPG FYUSFNJUJFT

3FHJNFO

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

347

348

1BUJFOUTXJUI5PVSFUUFT TZOESPNF

1BUJFOUTXJUI)$

.D"SUIVSFUBM66 CO

1PQVMBUJPO

4XFFUFUBM CO

"VUIPST

Table III (continued).

8Lo5#;NHEJOJUJBMMZ JODSFBTFECZNHEREUP NBYJNVNPGNHE  QJNP[JEFNHEJOJUJBMMZ  JODSFBTFECZNHEREUP NBYJNVNPGNHE 8LoOP5#; QJNP[JEF NHE QMBDFCP 8LoOP5#; QJNP[JEF EJTDPOUJOVFE TFDPOE QMBDFCPHJWFO 8Lo5#;NHE O PSNHE O   QJNP[JEFNHE O PS NHE O

5#;HJWFO2*%UPȐNHE O  NBYJNVN5#;EPTF NHEJOQBUJFOUTBOE NHEJOQBUJFOUT EVSBUJPOPG5#;UIFSBQZ oE NFBO E

".15HJWFO2*% HSBEVBMMZ JODSFBTFEUPȐNHE O  NBYJNVN".15EPTF NHEJOQBUJFOU  NHEJO BOE NHEJOEVSBUJPO PG".15UIFSBQZoE NFBO E

".15QFSJPEQSFDFEFE5#; QFSJPEJOBMMQBUJFOUT XJUI QIBTFTTFQBSBUFECZ XBTIPVUQFSJPEPGoE

3FHJNFO

5PMFSBCJMJUZ0VUDPNFT

"MMQBUJFOUTIBEJOJUJBMiSBQJE BOEDPOTJEFSBCMFwSFEVDUJPO JOOVNCFSPGBCOPSNBM NPWFNFOUTBUSFTU XIJDIXBT NBYJNBMBUXLoBOE UIFSFBGUFSSFHSFTTFEUPXBSE CBTFMJOFJOEJWJEVBMQBUJFOUT IBEFBSMZTVQQSFTTJPOUP  BOEPGDPOUSPM  SFHSFTTJOHUP  BOE PGDPOUSPMBUXL JF PGG 5#;GPSXL PGGQJNP[JEFGPS XL DPNCJOBUJPOPG5#; QJNP[JEFXBTNPTUCFOFGJDJBM BMUIPVHISFHSFTTJPOCFHBO EVSJOHUIFSBQZQJNP[JEF BMPOFIBEBTNBMMTVQQSFTTJWF FGGFDUJO  QBUJFOUT

DPOUJOVFE

1BUJFOUIBEBHHSFTTJPOJO XLo XIJDISFRVJSFE SFJOUSPEVDUJPOPG5#;GPSXL BOEEFQSFTTJPOJOXLo XIJDISFRVJSFEIBMPQFSJEPM QBUJFOUXBTXJUIESBXOJOXL EVFUPEFQSFTTJPO

"&TXJUI5#;JODMVEFE 5#;QBUJFOUT   EFQSFTTJPOJOQBUJFOUT JNQSPWFEiSFNBSLBCMZwBOE TFEBUJPOJO BOEBLJOFTJB    SFDFJWFEOP SFTUMFTTMFHT MFUIBSHZ PDVMPHZSJD CFOFGJU DSJTJT BLBUIJTJB BOESFTUMFTTOFTT ".15QBUJFOUT  IBE JOFBDIBMMQBUJFOUTIBEȑ"& NBSLFEJNQSPWFNFOU  "&TMFBEJOHUPEJTDPOUJOVBUJPO   SFDFJWFETPNF PG".15JODMVEFEBHJUBUJPO CFOFGJU   SFDFJWFE BOECSFBUIMFTTOFTTPUIFS"&T OPCFOFGJU BOE   XJUI".15JODMVEFE XJUIESFX DSZTUBMMVSJBBOEBLJOFTJBJO QBUJFOUTFBDI MFUIBSHZJO FOVSFTJTBOEBLBUIJTJBJO FBDI BOETJBMPSSIFB  JOTPNOJB EZTVSJB SFTUMFTTMFHT PDVMPHZSJDDSJTJT BOEEZTBSUISJB JOFBDI

&GGJDBDZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

1BUJFOUTXJUI)$

1BUJFOUT XJUI)$ XJUI )$XJUIPVUQPTJUJWFGBNJMZ IJTUPSZ XJUIDIPSFPBUIFUPTJT  XJUITQBTNPEJDUPSUJDPMMJT XJUIEZTUPOJBNVTDVMPSVN EFGPSNBOT XJUI PMJWPQPOUPDFSFCFMMBSBUSPQIZ XJUITQBTNPEJDDPOUSBDUJPO PGBCEPNJOBMXBMM

5PHMJBFUBM68

1PQVMBUJPO

)VBOHFUBM

"VUIPST

Table III (continued).

5#;NH#*%JOJUJBMMZ JODSFBTFECZNHEBUXFFLMZ JOUFSWBMTNBYJNVNEBJMZEPTF oNH NFBOEBJMZEPTF NH EVSBUJPOPGUIFSBQZ XLoNP NFBO NP

5#;NHEJOJUJBMMZ JODSFBTFE CZNHERoEUP NBYJNVNPGNHEEPTF DPVMECFSFEVDFEUPNHE JOUIFDBTFPG"&TNFBO NBYJNVNEBJMZEPTF NH SBOHF oNHE

3FHJNFO

)$$PNQMFUFDFTTBUJPOPGBMM BCOPSNBMNPWFNFOUTJO  

NPEFSBUF JNQSPWFNFOUJO 

 OPJNQSPWFNFOUJO 

)$XJUIPVUQPTJUJWFGBNJMZ IJTUPSZNPEFSBUF JNQSPWFNFOUJO 

BOEOPJNQSPWFNFOUJO  

.JTDFMMBOFPVTEJTPSEFSTOP JNQSPWFNFOUJO 

1BUJFOUT  JNQSPWFE BOE  EJEOPU QBUJFOUT  XIP JNQSPWFEXFSFDPOTJEFSFEUP IBWFNBSLFEJNQSPWFNFOU FWFOXIFOUIFSFXBT JNQSPWFNFOUJOUSVODBMBOE MJNCDIPSFJGPSNNPWFNFOUT TPNFQBUJFOUTIBEMJUUMF SFEVDUJPOJOMJOHVBMEZTLJOFTJBT

&GGJDBDZ0VUDPNFT

DPOUJOVFE

1BUJFOUXJUIESFXEVFUP"&T BLBUIJTJBBOEJOTPNOJB "&T PDDVSSFEJOQBUJFOUT ESPXTJOFTT JO JOTPNOJBBOEBLBUIJTJBJO FBDI TXFBUJOH QBSFTUIFTJBTPG UPOHVF BOEUSBOTJFOUGFWFSJO FBDI

%JTDPOUJOVBUJPOTEVFUP"&TJO QBUJFOUT ESPXTJOFTT QPTUVSBM IZQPUFOTJPO BOEBUBYJBJO QPTUVSBMIZQPUFOTJPOBOE CSPODIPQOFVNPOJBJO "&T JODMVEFEEZTQIBHJBBOEQPTUVSBM IZQPUFOTJPO DBTFTFBDI

ESPXTJOFTTBOEEFQSFTTJPO DBTFTFBDI

BOEEZTBSUISJB BLBUIJTJB BUBYJB BOE CSPODIPQOFVNPOJB DBTF FBDI BGUFSUIFDFTTBUJPOPG DIPSFJGPSNNPWFNFOUTJO QBUJFOU BEZTUPOJDQPTUVSF EFWFMPQFEPOXBMLJOH

5PMFSBCJMJUZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

349

350

-BOHBOE.BSTEFO

"VUIPST

Table III (continued).

5#;OBJWFQBUJFOUT  XJUIEZTUPOJBTBOEXJUI DIPSFB<XJUI)$>

1PQVMBUJPO

&GGJDBDZ0VUDPNFT

1IBTF 5#;OBJWF 5#; 1IBTFQBUJFOUT  IBE NH#*% JODSFBTFECZ NBSLFECFOFGJU   IBE NHEREUPNBYJNVN NPEFSBUFCFOFGJU  

PGNHEYXL IBENJMECFOFGJU  

1IBTF FYDMVEJOHUIPTFXIP IBEMJUUMFPSOPDIBOHF  

XFSF5#;JOUPMFSBOU CFDBNFXPSTF BOE 

PSEJTDPOUJOVFEBGUFSQIBTF XFSFVOBCMFUPUPMFSBUF "EEFE".15 USFBUNFOUJOUIPTFBCMFUP NHE JODSFBTFECZ UPMFSBUF5#; NFBOGJOBM NHERXLUPNBYJNVN EBJMZEPTFTXFSFNH PGHE NH2*%  SBOHF oNH JOUIPTF XJUIEZTUPOJBTBOENH EFQFOEJOHPOSFTQPOTF ".15DPVMECFDIBOHFEUP SBOHF oNH JOUIPTF 20%TDIFEVMF 5#;EPTF XJUIDIPSFB P 

DPVMECFSFEVDFE PSEPTFTPG 1IBTFBOEQBUJFOUT XJUIEZTUPOJBTBOEDIPSFB CPUIESVHTDPVMECFBEKVTUFE 1IBTF UIPTFXJUI SFTQFDUJWFMZ XFSFUPSFDFJWF JNQSPWFNFOUJONPWFNFOUT ".15XJUIXJUIPVU5#; BOEPSGVODUJPOBOEUPMFSBCMF CVUQBUJFOUT  XFSF "&T 5#;DPOUJOVFEBUTBNF VOBCMFUPUPMFSBUF".15 EPTF".15BOEQMBDFCPY MPOHFOPVHIUPCFFWBMVBUFE XLFBDI JO%# $0GBTIJPO PGQBUJFOUTXJUIEZTUPOJBT TFQBSBUFECZXLXBTIPVU SFDFJWJOH".15 XJUI QFSJPE XJUIPVU5#;

  

3FHJNFO

DPOUJOVFE

ȑ.PEFSBUFTFWFSF"&PDDVSSFE JOPGQBUJFOUTUSFBUFEXJUI 5#;BOEPGQBUJFOUTUSFBUFE XJUI".15 XJUIXJUIPVU5#;  NPTUDPNNPO"&TJOUIPTFXIP SFDFJWFE5#;XFSFESPXTJOFTT BOEBLBUIJTJB FBDI

EFQSFTTJPO 

BOEIZQFSTBMJWBUJPO   NPTUDPNNPO"&TJOUIPTFXIP SFDFJWFE".15 XJUIXJUIPVU 5#; XFSFESPXTJOFTT 

BLBUIJTJB 

IZQFSTBMJWBUJPO 

BOEEFQSFTTJPOBOE QBSLJOTPOJTN FBDI

5PMFSBCJMJUZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

-BOHBOE.BSTEFO DPOU

"VUIPST

Table III (continued). 1PQVMBUJPO

3FHJNFO FYIJCJUFEBNPEFTUSFTQPOTF PONPWFNFOUTDPSFT DPNQBSFEXJUI1IBTF   FYIJCJUFEMJUUMFPSOP CFOFGJU BOE   XPSTFOFEPGQBUJFOUTXJUI DIPSFBSFDFJWJOH".15   FYIJCJUFENJMECFOFGJU PONPWFNFOUTDPSFT  

FYIJCJUFENPEFSBUFCFOFGJU BOE  FYIJCJUFEMJUUMFPS OPCFOFGJUJOUSPEVDUJPOPG ".15OFDFTTJUBUFE SFEVDUJPOPG5#;EPTF CZB NFBOPGBOENHEJO QBUJFOUTXJUIEZTUPOJBTBOE DIPSFB SFTQFDUJWFMZ

1IBTF1BUJFOUTSFDFJWJOH ".15BMPOF SFDFJWJOH ".15 5#;QMBDFCP TVCTUJUVUJPOMFEUPOP EFUFSJPSBUJPOJONPUPS TZNQUPNTJOQBUJFOU XJUIEZTUPOJBT TMJHIU EFUFSJPSBUJPOJOQBUJFOU XJUIEZTUPOJBT BOE TJHOJGJDBOUEFUFSJPSBUJPOJO QBUJFOUT   XJUI EZTUPOJBT XJUIDIPSFB

&GGJDBDZ0VUDPNFT

DPOUJOVFE

5PMFSBCJMJUZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

351

352

+BOLPWJDFUBM

"VUIPST

Table III (continued).

1BUJFOUTXJUI5PVSFUUFT TZOESPNF

1PQVMBUJPO .FBO5#;EPTF NHE SBOHF ¦NHE  EVSBUJPOPGUIFSBQZ oNP NFBO NP

3FHJNFO 1BUJFOUT  IBENBSLFE BOEMBTUJOHJNQSPWFNFOUJO UJDTBOECFIBWJPSBMEZTGVODUJPO   IBEPOMZNJMEPS USBOTJFOUJNQSPWFNFOU    IBEOPJNQSPWFNFOUJOUIF QBUJFOUTXIPIBEVOEFSHPOF TMFFQTUVEJFTCFGPSFUSFBUNFOU BOEBGUFSoNPPOBTUBCMF EPTFPG5#; 5#;XBTBTTPDJBUFE XJUIBSFEVDFEOVNCFSPGUJDT QFSSFDPSEJOHTFTTJPO NFBO GSPNUP PȐ

FMJNJOBUJPOPGTUBHFFWFOUT P 

FMJNJOBUJPOPGTMFFQ BQOFBJOQBUJFOUT 

BOE SFEVDUJPOJOUPUBMTMFFQUJNF P

XJUIPVUBTJHOJGJDBOU DIBOHFJOQSPQPSUJPOPG3&. TMFFQ

&GGJDBDZ0VUDPNFT

DPOUJOVFE

%SPXTJOFTTJOQBUJFOUT OFSWPVTOFTTBOEEFQSFTTJPOJO FBDI BOEQBSLJOTPOJTNBOE PDVMPHZSJDDSJTJTJOFBDI

5PMFSBCJMJUZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

'BIO

"VUIPST

Table III (continued).

1BUJFOUTXJUIUSBEJUJPOBM OFVSPMFQUJDoBTTPDJBUFE5%

1PQVMBUJPO .BYJNVN5#;EPTF o NHEEVSBUJPOPG UIFSBQZ ȑZ

3FHJNFO

5PMFSBCJMJUZ0VUDPNFT

DPOUJOVFE

1BUJFOUIBEBMNPTUDPNQMFUF 1BSLJOTPOJTNJO5#;SFDJQJFOUT TZNQUPNSFMJFGXJUI5#; 

NPOPUIFSBQZ NBYJNVN EPTF NHE QBUJFOUXIP IBEEFWFMPQFE"&TEVSJOH USFBUNFOUXJUISFTFSQJOFBOE TXJUDIFEUP5#;NPOPUIFSBQZ IBENBSLFETZNQUPNSFMJFG QBUJFOUXIPIBEBSFUVSOPG TZNQUPNTEVSJOHUIFSBQZXJUI SFTFSQJOF ".15BOE TXJUDIFEUP5#;NPOPUIFSBQZ IBEBMNPTUDPNQMFUFTZNQUPN SFMJFG NBYJNVNEPTF NHE JOQBUJFOUTXIP DPNNFODFEUSFBUNFOUXJUI 5#;BMPOF NBYJNVNEPTFT  BOENHE CVUSFRVJSFE UIFBEEJUJPOPG".15  BOENHE

UIFQBUJFOU XIPTXJUDIFEUPDPNCJOBUJPO UIFSBQZEVFUPUIFNPEFTU FGGJDBDZPG5#;BMPOFIBE BMNPTUDPNQMFUFTZNQUPN SFMJFGBOEUIFQBUJFOUXIP TXJUDIFEUPDPNCJOBUJPO UIFSBQZEVFUPOPSFTQPOTFUP 5#;BMPOFIBEOPSFTQPOTFUP DPNCJOBUJPOUIFSBQZ

&GGJDBDZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

353

354

.BSTEFOFUBM

"VUIPST

Table III (continued).

1BUJFOUTXJUITFWFSFBYJBM UPSTJPOEZTUPOJBT

1PQVMBUJPO *OJUJBM5#;EPTFT o NHE O BOEo NHE O JOUIF QBUJFOUTDPOUJOVJOH5#; NFBOEPTFXBTNHE XFSFSFDFJWJOHȐNHE

 UBLFOGPSBNFBOPGNP SBOHF oNP

"EEFEQJNP[JEFNHE O

JODSFBTFECZNHE RXLVOUJMDPOUSPMPGEZTUPOJBT PSPDDVSSFODFPG VOBDDFQUBCMFQBSLJOTPOJTN NFBOEPTF NHE SBOHF oNHE

UBLFO GPSBNFBOPGNP SBOHF oNP

"UUIJTQPJOU BO BOUJDIPMJOFSHJDXBTBEEFE UP5#;NPOPUIFSBQZ O PS5#; QJNP[JEF UIFSBQZ O XJUIVQXBSE UJUSBUJPOVOUJMDPOUSPMPG QBSLJOTPOJBO"&TJG EZTUPOJBTDPOUJOVFEUPCF VODPOUSPMMFE QJNP[JEF BOEPSBOUJDIPMJOFSHJDEPTFT DPVMECFGVSUIFSUJUSBUFE

3FHJNFO 0OUIFJSGJOBMSFHJNFOT QBUJFOUT  IBEESBNBUJD JNQSPWFNFOU   IBE NBSLFEJNQSPWFNFOU    IBENPEFSBUF JNQSPWFNFOU   IBE TMJHIUJNQSPWFNFOU BOE  IBEOPJNQSPWFNFOU

&GGJDBDZ0VUDPNFT

DPOUJOVFE

*OJUJBMMZIJHI5#;EPTFTXFSF BTTPDJBUFEXJUIVOBDDFQUBCMF QSFWBMFODFPGEFQSFTTJPO EVFUP XIJDIQBUJFOUTEJTDPOUJOVFE 5#;BOESFRVJSFEEPTFSFEVDUJPO JOUIFQBUJFOUTXIPDPOUJOVFE UIFSBQZMPOHUFSN "&TJODMVEFE QBUJFOUT  XJUIFWJEFOU QBSLJOTPOJTN   XJUI BLBUIJTJB   XJUIPUIFS BOUJDIPMJOFSHJD"&T BOE   XJUITPNFESPXTJOFTTBMMXFSF BCMFUPDPOUJOVFUSFBUNFOU EFTQJUF"&T

5PMFSBCJMJUZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

1BUJFOUTXJUITQPOUBOFPVT PSBMEZTLJOFTJBT

1BUJFOUTXJUIUSBEJUJPOBM OFVSPMFQUJDoBTTPDJBUFE5%ZTU

,BOHFUBM 

1PQVMBUJPO

#BSUFMTBOE;FMMFS

"VUIPST

Table III (continued).

5#;oNHE NFBO  NHE

.FBOEBJMZ5#;EPTFTXFSF NH O

NH O

BOENH O NFBO 4% EVSBUJPOPGUIFSBQZ   NP

3FHJNFO

0WFSBMM   5#; SFDJQJFOUTIBEBiGBWPSBCMFw SFTQPOTFJOUIFQBUJFOUT SFDFJWJOH5#;NPOPUIFSBQZ   IBEBiGBWPSBCMFw SFTQPOTFJOUIFQBUJFOUTJO XIPN5#;XBTBEEFEUP QSFWJPVTMZVOTVDDFTTGVMUIFSBQZ   IBEBiGBWPSBCMFw SFTQPOTFJOUIFQBUJFOUT  XIPSFDFJWFE5#;BOE SFTFSQJOFNPOPUIFSBQZBU EJGGFSFOUUJNFT    JNQSPWFEPOCPUIESVHT   EJEOPUSFTQPOEUPFJUIFS   JNQSPWFEPO5#;CVU EJEOPUSFTQPOEIBE TJHOJGJDBOU"&TXJUISFTFSQJOF BOE  JNQSPWFEPO SFTFSQJOFCVUEJEOPUSFTQPOE IBETJHOJGJDBOU"&TXJUI5#;

%ZTLJOFTJBTXFSFiESBNBUJDBMMZw JNQSPWFEJOQBUJFOUT   BOE  IBEHPPEUP TBUJTGBDUPSZJNQSPWFNFOU HPPEDPSSFTQPOEFODFCFUXFFO JNQSPWFNFOUTJOWJEFPUBQF &.(EBUBBOEDMJOJDBM JNQSFTTJPOEBUB

&GGJDBDZ0VUDPNFT

DPOUJOVFE

"&TJOQBUJFOUT  XFSF TFSJPVTFOPVHIUPMJNJU5#; EPTBHF"&TJODMVEFE QBSLJOTPOJTNBOEEFQSFTTJPO FWFOUTFBDI

MFUIBSHZ 

BOE IBMMVDJOBUJPOT DPOGVTJPO EJ[[JOFTT WPNJUJOH VOJMBUFSBMMFHUSFNPS FBDI JOUIFQBUJFOUTXJUIB IJTUPSZPGEFQSFTTJPO NBYJNVN 5#;EPTF NHE

   IBESFMBQTFTPGEFQSFTTJPOBU5#; EPTFTPG  BOENHE QBUJFOUEFWFMPQFEBOBDVUF EZTUPOJDSFBDUJPO

5JSFEOFTTBOEQPTUVSBM PSUIPTUBUJDIZQPUFOTJPO O BOENJMESJHJEJUZ O BQQFBSFEFBSMZ XJUIPOMZ UJSFEOFTTBOEQPTUVSBM PSUIPTUBUJDIZQPUFOTJPO EJTBQQFBSJOHBGUFSoXL

5PMFSBCJMJUZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

355

356

+BOLPWJDBOE0SNBO

"VUIPST

Table III (continued).

1BUJFOUT XJUI5%  XJUI)$ XJUI5PVSFUUFT TZOESPNF XJUIHFOFSBMJ[FE EZTUPOJBT XJUIDSBOJBM EZTUPOJBT XJUI5%ZTU  XJUIGPDBMEZTUPOJBT XJUI FTTFOUJBMCMFQIBSPTQBTN  XJUIIFNJEZTUPOJB

1PQVMBUJPO %BJMZ5#;EPTFTBOE EVSBUJPOT‰5%oNH NFBO NH GPSo NP NFBO NP )$ oNH NFBO NH

GPSoNP NFBO NP  5PVSFUUFTTZOESPNFo NH NFBO NH GPS oNP NFBO NP  HFOFSBMJ[FEEZTUPOJBTo NH NFBO NH GPS oNP NFBO NP  DSBOJBMEZTUPOJBToNH NFBO NH GPSo NP NFBO NP  5%ZTUoNH NFBO  NH GPSoNP NFBO NP GPDBM EZTUPOJBToNH NFBO NH GPSoNP NFBO NP

3FHJNFO 0WFSBMMQPQVMBUJPO  BOEIBENBSLFE  NPEFSBUF BOENJME JNQSPWFNFOU SFTQFDUJWFMZ XIJMFIBEOPDIBOHF BOEXPSTFOFE 5%$PSSFTQPOEJOH QSPQPSUJPOTXJUINBSLFE NPEFSBUF BOENJME JNQSPWFNFOUBOEOP JNQSPWFNFOUXPSTFOJOH XFSF   BOE )$IBENPEFSBUF JNQSPWFNFOUBOEIBE NJMEJNQSPWFNFOU 5PVSFUUFTTZOESPNF    BOEIBENJME NPEFSBUF BOENBSLFE JNQSPWFNFOUBOEOP JNQSPWFNFOUXPSTFOJOH  SFTQFDUJWFMZ 0WFSBMMEZTUPOJBQPQVMBUJPO DPSSFTQPOEJOHQSPQPSUJPOT XJUINBSLFE NPEFSBUF BOE NJMEJNQSPWFNFOUBOEOP JNQSPWFNFOUXPSTFOJOH XFSF   BOE

&GGJDBDZ0VUDPNFT

DPOUJOVFE

1BSLJOTPOJTNJO ESPXTJOFTT PSGBUJHVFJO EFQSFTTJPOJO  OFSWPVTOFTTPSBOYJFUZJO  JOTPNOJBBOESFTUMFTTOFTT BLBUIJTJBJOFBDI BOE PUIFS"&TJOȐFBDINPTU"&T XFSFEPTFSFMBUFEBOEJNQSPWFE XJUIEPTFSFEVDUJPOOPBQQBSFOU EJGGFSFODFTJOUZQFTSBUFTPG"&T JOQBUJFOUTXJUIEJGGFSFOU NPWFNFOUEJTPSEFST

5PMFSBCJMJUZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

+BOLPWJDFUBM CO

"VUIPST

Table III (continued).

1BUJFOUTXJUI-FTDI/ZIBO TZOESPNF

1PQVMBUJPO 4FRVFOUJBMUSFBUNFOUXJUI ESVHT 5#; GMVQIFOB[JOF DBSCJEPQBMFWPEPQB BOE CSPNPDSJQUJOF

XJUIHSBEVBM EPTFJODSFBTFTUPNBYJNVN SFTQPOTFPSJOUPMFSBCMF"&T

3FHJNFO 4FMGNVUJMBUPSZPS BVUPBHHSFTTJWFCFIBWJPST QBUJFOUT  IBE NPEFSBUFJNQSPWFNFOU JNQBJSNFOUOPXPOMZ TMJHIUMZEJTBCMJOH XJUI5#;    IBEOPDIBOHF BOE   XPSTFOFE )ZQFSLJOFUJDNPWFNFOU EJTPSEFSTQBUJFOUT   IBENPEFSBUFJNQSPWFNFOU JNQBJSNFOUOPXPOMZ TMJHIUMZEJTBCMJOH XJUI5#;    IBETMJHIU JNQSPWFNFOU JNQBJSNFOU EFGJOJUFMZJOUFSGFSJOHXJUI GVODUJPOJOH

BOE 

IBEOPDIBOHF

&GGJDBDZ0VUDPNFT

DPOUJOVFE

)JHIJODJEFODFPG"&T TPNFPG UIFNJOUPMFSBCMF

5PMFSBCJMJUZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

357

358

4UBDZFUBM

"VUIPST

Table III (continued).

1BUJFOUTXJUI5%  XJUITUFSFPUZQZ XJUI EZTUPOJBT XJUIBLBUIJTJB XJUIUSFNPS XJUIDIPSFB XJUINZPDMPOVT IBE BDPNCJOBUJPOPGEJTPSEFST IBEEZTUPOJBBMPOF  IBETUFSFPUZQZBMPOF

1PQVMBUJPO 5#; EPTFOPUTQFDJGJFE GPS NFBOEVSBUJPOPGNPJO TUFSFPUZQZHSPVQBOENP JOEZTUPOJBHSPVQ

3FHJNFO .FBOJNQSPWFNFOUTPO QPJOUTDBMF XJUIBTDPSFPG SFQSFTFOUJOHNPEFSBUF JNQSPWFNFOUJOJOUFOTJUZ BNQMJUVEFPGBCOPSNBM NPWFNFOUTBOENPEFSBUF GVODUJPOBMJNQSPWFNFOU ‰ JOUIPTFXJUITUFSFPUZQZ BMPOFJODPNCJOBUJPO JO UIPTFXJUIEZTUPOJBTJO DPNCJOBUJPO JOUIPTFXJUI EZTUPOJBTBMPOF BOEJO UIPTFXJUIBLBUIJTJBPWFSBMM NFBOJNQSPWFNFOU PUIFS PVUDPNFEBUBBWBJMBCMFGPSPOMZ QBUJFOUTXIPSFDFJWFE SFTFSQJOFCFGPSF5#;BOE DPNQMFUFEUIF5#;USJBM PG XIPN  IBENBSLFE JNQSPWFNFOUXJUI5#;    IBENPEFSBUF JNQSPWFNFOU BOE 

 IBEMJUUMFPSOPSFTQPOTF

&GGJDBDZ0VUDPNFT

DPOUJOVFE

"&TPDDVSSFEJOPGQBUJFOUT  JODMVEJOHEPTFSFMBUFE QBSLJOTPOJTNJOEFQSFTTJPO  EZTQIBHJB IBMMVDJOBUJPOT BOE ESPXTJOFTTJOFBDIBMM"&T SFTPMWFEXJUI5#;EPTFSFEVDUJPO PSEJTDPOUJOVBUJPO

5PMFSBCJMJUZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

1BUJFOUTXJUI5% SFMBUFE UPNFUPDMPQSBNJEFJO BNPYBQJOFJO GJSTUHFOFSBUJPO OFVSPMFQUJDTJO BOE UIJFUIZMQFSB[JOFJO

1BUJFOUTXJUI)$

0OEPFUBM80

1PQVMBUJPO

0OEPFUBM

"VUIPST

Table III (continued).

5#;NH#*%JOJUJBMMZ  UJUSBUFEJOXFFLMZJODSFNFOUT UPNBYJNVNPGNH5*% GJOBMNFBO 4% 5#;EPTF   NHE SBOHF o NHE NFBOEVSBUJPO   NP SBOHF oNP

5#;NH#*%JOJUJBMMZ  UJUSBUFEJOXFFLMZJODSFNFOUT UPNBYJNVNPGNH5*% NFBO 4% GJOBMEPTF   NHENFBOEVSBUJPO    XL

3FHJNFO

1BUJFOUXBTMPTUUPGPMMPXVQ QBUJFOUTTFMGBTTFTTNFOUT JOEJDBUFENBSLFEJNQSPWFNFOU JO 

NPEFSBUF JNQSPWFNFOUJO 

 NJMEJNQSPWFNFOUJO 

BOEOPDIBOHFJO  CMJOEFEBTTFTTNFOUPG WJEFPUBQFTJOEJDBUFEUIBU   IBEJNQSPWFE XJUI5#;   XFSF CFUUFSCFGPSF5#; BOE  XBTVODIBOHFE P  P NFBO 4% "*.4NPUPSTDPSFT JNQSPWFEGSPN  UP    P BMMQBUJFOUT DPOUJOVFE5#;BGUFSDPNQMFUJPO PGTUVEZ

.FBOJNQSPWFNFOUPO "*.4NPUPSTVCTFUTDPSFT GSPNNFBO<4%>PG<> UP<> P   NFBOJNQSPWFNFOUPO TVCKFDUJWFTDPSFT GSPN <>UP<> P  QBUJFOUTTFMGBTTFTTNFOUT JOEJDBUFENBSLFE JNQSPWFNFOUJO 

 NPEFSBUFJNQSPWFNFOUJO  

BOENJME JNQSPWFNFOUJO 

&GGJDBDZ0VUDPNFT

DPOUJOVFE

"&TJODMVEFEBLBUIJTJB DBTFT  BOEJOTPNOJB DPOTUJQBUJPO  EFQSFTTJPO ESPPMJOH BOETVCKFDUJWF XFBLOFTT DBTFFBDI OP DPNQMBJOUTPGQBSLJOTPOJTNBMM "&TXFSFNJMEJOJOUFOTJUZ FYDFQU DBTFPGBLBUIJTJB XIJDI SFTQPOEFEUPEPTFSFEVDUJPO

1BUJFOUEJTDPOUJOVFEEVFUP TFEBUJPOBGUFSENJME QBSLPOTPOJTNBOETFEBUJPOXFSF OPUFEJOQBUJFOUT  FBDI

5PMFSBCJMJUZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

359

360

1BUJFOUTXJUI)$

1BUJFOUT XJUITJNQMF 5PVSFUUFTTZOESPNF XJUI GVMMCMPXO5PVSFUUFTTZOESPNF XJUI 5PVSFUUFTTZOESPNF "%)% PS0$%

1PSUBFUBM

1PQVMBUJPO

,FOOFZFUBM81

"VUIPST

Table III (continued).

5#;BENJOJTUFSFEVQUP5*% BTBEKVODUJWFUIFSBQZ 

PSNPOPUIFSBQZ  UPUBM EBJMZEPTFSBOHF oNH NFBOEPTF NHENFBO EVSBUJPO NP

.FBO 4% 5#;EPTF   NHE SBOHF o NHE FWFOJOH5#;EPTF UBLFOBUIPNF OFYUNPSOJOH EPTFIFMEGPSBENJOJTUSBUJPO BUDMJOJD6)%34NPUPSTDPSFT CFGPSFBOEBGUFSNPSOJOHEPTF XFSFGPMMPXFEVOUJMDIPSFJD NPWFNFOUTTVCTJEFEBOE TVCTFRVFOUMZSFUVSOFE

3FHJNFO

$(*$‰TJNQMF5PVSFUUFT TZOESPNFQBUJFOUT  JNQSPWFE  

XFSFVODIBOHFE BOE  XPSTFOFEGVMMCMPXO 5PVSFUUFTTZOESPNF  JNQSPWFE  

XFSFVODIBOHFE BOE  XPSTFOFE5PVSFUUFT TZOESPNF "%)%PS0$%   JNQSPWFE   XFSFVODIBOHFE BOE   XPSTFOFEPWFSBMM JNQSPWFE XFSF VODIBOHFE BOEXPSTFOFE

.FBO 4% 6)%34DIPSFB TDPSFTEFDSFBTFECZQPJOUT  NFBOEVSBUJPOPG FGGFDU   I SBOHF  oI

&GGJDBDZ0VUDPNFT

DPOUJOVFE

4JNQMF5PVSFUUFTTZOESPNF ESPXTJOFTTJOQBUJFOUTBOE EFQSFTTJPO BTUIFOJB BOEBLBUIJTJB JOFBDIGVMMCMPXO5PVSFUUFT TZOESPNFESPXTJOFTTJOQB UJFOUTBOEEFQSFTTJPOBOEBTUIFOJB JOFBDI5PVSFUUFTTZOESPNF  "%)%PS0$%ESPXTJOFTTJO QBUJFOUT EFQSFTTJPOJO  BTUIFOJBJO BLBUIJTJBJO BOE QBSLJOTPOJTNJOPWFSBMMEPTF EFQFOEFOUESPXTJOFTTJO EFQSFTTJPOJO NBSLFE BTUIFOJBJO BLBUIJTJBJO  BOEQBSLJOTPOJTNJO

/P"&TSFQPSUFE

5PMFSBCJMJUZ0VUDPNFT

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

'BTBOPFUBM

"VUIPST

Table III (continued).

1BUJFOUTXJUI)$

1PQVMBUJPO .FBO 4% 5#;EPTF   NHE SBOHF o NHE BUGJSTUGPMMPXVQWJTJU NFBO <>NP BOE  NHE SBOHF o NHE BUNPTUSFDFOU GPMMPXVQWJTJU NFBO  <>NP NFBONBYJNVN EPTF   NHE SBOHF  oNHE NFBOEVSBUJPO PGUIFSBQZ   NP SBOHF oNP

3FHJNFO "UUJNFPGEBUBDPMMFDUJPO  QBUJFOUT  XFSFTUJMM CFJOHUSFBUFEBUGJSTUGPMMPXVQ WJTJU OPTJHOJGJDBOUDIBOHFGSPN CBTFMJOFJONFBO6)%34NPUPS TDPSFTPSEZTUPOJBTVCTDPSFT CVUDIPSFBTVCTDPSFTXFSF TJHOJGJDBOUMZEFDSFBTFECZB NFBOPG P Ȑ BU NPTUSFDFOUGPMMPXVQWJTJU  OPTJHOJGJDBOUDIBOHFGSPN CBTFMJOFJONFBODIPSFBBOE EZTUPOJBTVCTDPSFT BOENFBO 6)%34NPUPSTDPSFTXFSF TJHOJGJDBOUMZJODSFBTFECZB NFBOPG P   JNQSPWFNFOUJODIPSFB EFDSFBTFEGSPNBNFBOPG UPCFUXFFOUIF GPMMPXVQWJTJUT P

 EFTQJUFNFBOEPTFJODSFBTFPG  P  QBUJFOUT   IBEOPJNQSPWFNFOUJODIPSFB BOEEJTDPOUJOVFEFBSMZPOMZ QSFEJDUPSPGCFUUFSSFTQPOTF UP5#;XBTPMEFSBHFBUPOTFU PGEJTFBTF rSBOHFGPSGJSTUBOE r MBTUFWBMVBUJPOTBOENBYJNVN FGGFDUBDIJFWFE oBMM DPSSFMBUJPOT P

&GGJDBDZ0VUDPNFT

DPOUJOVFE

8JUIESBXBMTEVFUP"&T XJUI SBQJEXPSTFOJOHPGQTZDIJBUSJD JMMOFTTBOEXJUIEJTBCMJOH BTUIFOJB QBUJFOUT  IBE ȑ"& JODMVEJOHESPXTJOFTTJO  EFQSFTTJPOJO EJBSSIFB DPOTUJQBUJPO OBVTFB WPNJUJOH  BOEBCEPNJOBMQBJOJOFBDI XPSTFOJOHPGQBSLJOTPOJTNJO  YFSPTUPNJBJO BOEIZQPUFOTJPO JOOPEPTFSFMBUJPOTIJQOPUFE GPSBOZ"&OFDFTTBSZDPODPNJUBOU USFBUNFOUTXFSFBOUJQBSLJOTPOJBO BHFOUTJOQBUJFOUT 

 CFO[PEJB[FQJOFTJO 

UZQJDBMOFVSPMFQUJDTJO 

 RVFUJBQJOFPSDMP[BQJOFJO 

BOEBOUJEFQSFTTBOUTJO 

5PMFSBCJMJUZ0VUDPNFT

D.R.P. Guay The American Journal of Geriatric Pharmacotherapy

361

362

1BUJFOUTXJUI)$

1PQVMBUJPO 5#;JOJUJBUFEBUNHE UJUSBUFEVQXBSEBUFOEPGFBDI XLJOJODSFNFOUTPGNHE GPSBUPUBMEBJMZEPTFPGȐNH

PSNHE GPSUPUBMEBJMZEPTF PGNH UPNBYJNVN UPMFSBUFEEPTFPSNBYJNVN PGNHENFBOEBJMZ5#; EPTFBUXL NH SBOHF  oNH EVSBUJPOPG USFBUNFOU VQUPXL

3FHJNFO 1BUJFOUT  DPNQMFUFE XLPGUSFBUNFOUNFBO 4%

EFDSFBTFGSPNCBTFMJOFJO 6)%34UPUBMNBYJNBMDIPSFB TDPSFPG  VOJUT P   DIPSFBSFFNFSHFEXL BGUFSXBTIPVU XJUIJODSFBTF JOUPUBMNBYJNBMDIPSFBTDPSF PG  VOJUT P  JO UIFQBUJFOUTXJUIDPNQMFUF EBUB OPTJHOJGJDBOUEJGGFSFODF JOUPUBMNBYJNBMDIPSFBTDPSF BUXLWTCBTFMJOF <> BOE<> SFTQFDUJWFMZ CZ XL $(*$TDPSFJODSFBTFECZ   VOJUTGSPNCBTFMJOF P  PGDPNQMFUFST  JNQSPWFEGSPNCBTFMJOF    XPSTFOFE BOE 

IBEOPDIBOHFBNPOH DPNQMFUFST UIFSFXFSF TJHOJGJDBOUEFDSFBTFTJO DPHOJUJWFNFBTVSFT WFSCBM GMVFODZTZNCPMEJHJUNPEBMJUJFT UFTUBOE4USPPQXPSESFBEJOH DPMPS BOEJOUFSGFSFODFUFTUT  BOEGVODUJPOBMNFBTVSFT GVODUJPOBMDIFDLMJTU JOEFQFOEFODFTDBMF UPUBM GVODUJPOBMDBQBDJUZ  BMM PP 

&GGJDBDZ0VUDPNFT 8JUIESBXBMEVFUPEFBUI VOSFMBUFEUP5#;

EVFUP"&T BQQFBSBODFPGWPDBMUJDT EFQSFTTJPO EFMVTJPOT FMFWBUFE CJMJSVCJO JODSFBTFEUSBOTBNJOBTFT  BLBUIJTJB

BOEEVFUPQSPUPDPM FSSPST XJUIESBXBMPGDPOTFOU OPODPNQMJBODF BOEMPTTUP GPMMPXVQ"&TXFSFSFQPSUFE JOQBUJFOUTUSFBUNFOUFNFSHFOU "&TPDDVSSJOHJOȑPGQBUJFOUT XFSFTFEBUJPO QBUJFOUT

EFQSFTTFENPPE <XJUIB IJTUPSZPGEFQSFTTJPO>

BOYJFUZ 

JOTPNOJB 

BLBUIJTJB 

GBUJHVF 

BHJUBUJPO 

GBMM 

 BOEEZTQIBHJBEZTUPOJB FBDI  OVNCFSPGQBUJFOUTXJUIPOFPS NPSF"&TXBTIJHIFSJOUJUSBUJPO QIBTFUIBOJONBJOUFOBODFQIBTF WT SFTQFDUJWFMZP   DPSSFTQPOEJOHOVNCFSTXFSF WTGPSTPNOPMFODF P 

WTGPSJOTPNOJB P 

BOEWTGPSEJBSSIFB P   GSPNCBTFMJOFUPXL NFBO 4%  6)%34QBSLJOTPOJTNTDPSF JODSFBTFECZ  VOJUT P =  BOE61%34EZTBSUISJB TDPSFJODSFBTFECZ  VOJU P 

5PMFSBCJMJUZ0VUDPNFT

)$)VOUJOHUPOTDIPSFB#-.CVDDPMJOHVBMNBTUJDBUPSZ"&TBEWFSTFFWFOUT$0DSPTTPWFS5%UBSEJWFEZTLJOFTJB".15α-methyl-para-tyrosine; DB = double blind; REM = rapid eye movement; EMG = electromyelogram; TDyst = tardive dystonia; AIMS = Abnormal Involuntary Movement Scale; UHDRS = Unified Huntington’s Disease Rating Scale; ADHD = attentiondeficit/hyperactivity disorder; OCD = obsessive-compulsive disorder; CGIC = Clinical Global Impression of Change.

'SBOLFUBM

"VUIPST

Table III (continued).

The American Journal of Geriatric Pharmacotherapy D.R.P. Guay

D.R.P. Guay

Nyhan syndrome (n = 5), bilateral athetosis (n = 2), hemiballismus (n = 2), chorea due to cerebrovascular disease (n = 4), chorea due to postencephalitic parkinsonism (n = 2), chorea due to L-dopa treatment of Parkinson’s disease (n = 3), chorea due to tuberculoma (n = 1), posttraumatic chorea (n = 2), essential tremor (n = 3), hereditary ataxia (n = 2), perinatal hypoxia (n = 1), progressive athetoid dystonia (n = 1), hypoxic encephalopathy/athetosis (n = 1), and kernicterus/ athetosis (n = 1). Positive clinical responses were noted in 72 of 109 patients (66%) with Huntington’s chorea,61,62,67,68,73,80,84 77 of 88 patients (88%) with tardive dyskinesia,63,76,79 and 23 of 26 patients (88%) with Tourette’s syndrome.70,73 Rates of positive response in patients with spontaneous oral dyskinesias73 and Lesch-Nyhan syndrome77 were 100% (6 of 6 patients) and 80% (4 of 5 patients), respectively. In some cases, the numbers of patients with each disorder cited here differ from those in the previous paragraph due to the use of a nonclinical definition of positive response in several of the studies.78,81,83 When response data for the patients with Huntington’s disease are removed from the data sets from 3 of the studies,61,62,76 it is possible to calculate pooled response rates for less-common hyperkinetic disorders, such as other choreas, hemiballismus, and the dystonias. In these 3 modified data sets, rates of response to TBZ were 40% (6 of 15 patients),61 32% (6 of 19),62 and 24% (30 of 124).76 Across studies, the most frequently reported AEs were parkinsonism (22%), drowsiness (20%), depression (14%), confusion (13%), postural hypotension (12%), anxiety (10%), akathisia (9%), insomnia (8%), and dysphagia (8%). McArthur et al66 reported the development of tolerance with continuous TBZ exposure in 3 patients with Huntington’s chorea treated with the combination of TBZ and pimozide. In this trial, combination therapy initially was associated with a “rapid and considerable” reduction in the number of abnormal movements. The response was maximal at 3 to 4 weeks after the initiation of TBZ and regressed toward baseline over the subsequent 3 to 4 weeks. Fasano et al83 reported similar findings in a trial of TBZ monotherapy in 68 patients with Huntington’s chorea. A 13% reduction in efficacy, as measured using the Unified Huntington’s Disease Rating Scale (UHDRS) chorea subscale, occurred between follow-up visit 1 (at a mean [SD] of 10 [8] months after initiation of TBZ) and follow-up visit 2 (at 34 [25] months) (P < 0.01). This occurred despite a mean 63% dose increase between the 2 followup visits (from 35 [15] to 58 [15] mg/d; P < 0.01).

The American Journal of Geriatric Pharmacotherapy

Fasano et al83 also investigated possible predictors of TBZ-associated improvement in chorea. Drug dose, sex, age at the time of TBZ treatment, and treatment duration were not significantly associated with outcomes. However, older age at the onset of disease predicted better outcomes (rr = 0.33, 0.33, and 0.38 for the first and last evaluations and the maximal effect achieved, respectively; P < 0.01).

Controlled Trials There have been 10 controlled trials of TBZ,8,85–93 the majority of them conducted many years ago. The major outcome measures used in these trials included the UHDRS, the Unified Parkinson’s Disease Rating Scale (UPDRS), and the Clinical Global Impression (CGI) scale.

Crossover Trials In a randomized, double-blind, active- and placebocontrolled crossover trial, Godwin-Austen and Clark85 compared TBZ (initiated at 25 mg BID for 3 days, followed by 25 mg QID for 4 days), diazepam (initiated at 1 mg BID for 3 days, followed by 1 mg QID for 4 days), and placebo for 7 days in the treatment of phenothiazine-induced tardive dyskinesia in 6 elderly (age range, 60–85 years) women who were long-stay residents of a psychiatric hospital. There was no washout period between phases. Three assessors (2 of clinical symptoms, 1 of videotapes made on the last day of each phase) provided blinded input on patients’ responses. Details of the 4-point response scale were not provided, although lower scores indicated greater improvement. With TBZ, the 6 patients had score reductions of 1.5, 2, 4, 4, 4, and 6 points after 1 week; the mean point tally fell from 6.4 at baseline to 2.8 points (56%). With diazepam, the 6 patients had score reductions of 1, 1, 2, 2.5, 3, and 3.5 points after 1 week, and the mean point tally fell from 6.4 to 4.3 points (33%). After 1 week of placebo, 3 patients had score reductions of 0.5, 2, and 1.5 points; 2 had no change; and 1 had a 1-point increase. The mean point tally was reduced from 6.4 to 5.9 (8%). No statistical analysis was provided. AEs were poorly described and quantitated. The authors reported that “most” patients experienced drowsiness during the TBZ phase that was no greater than that during the diazepam phase. Gilligan et al86 conducted a randomized, placebocontrolled, double-blind crossover trial of TBZ 150 mg/d in patients with a variety of abnormal movement disorders. Eighteen of the 27 enrolled patients (67%) completed the trial (6 with Huntington’s chorea, 9 with athetosis, 1 with bilateral facial spasms, 1 with chorei-

363

The American Journal of Geriatric Pharmacotherapy

D.R.P. Guay

form movements of the right arm, and 1 with chorea due to cerebral atherosclerosis). The treatment phases were 4 weeks each, with no washout between phases. Three blinded evaluators watched videotapes made before the study and at the end of each phase, and were asked to provide an opinion about the phase in which patients received TBZ. If they perceived no pattern of change during the 2 study phases, they could state that they had no opinion. For the 18 patients who completed the study, the evaluators had an opinion in 14 cases and were correct for 11 (79%) of them; they had no opinion in 4 cases. Thus, clinical improvement due to TBZ was noted in 11 of 18 patients (61%); however, it was the impression of the blinded evaluators that the degree of improvement was moderate in 3 of these 11 patients (27%) and mild in 8 (73%). When asked about their preference for the study regimens, 3 patients preferred TBZ, 6 preferred placebo, and 9 had no preference. Patients’ spontaneously volunteered reasons for their preferences appeared to be concerned more with the absence of drowsiness than with any change in motor control. Drowsiness (17 events), hypersalivation (2), and agitation (2) were reported as AEs. Both instances of hypersalivation occurred during the TBZ phase, and both instances of agitation occurred during the placebo phase. Drowsiness was reported by 3 patients during both phases. The placebo phase occurred first in 2 of these patients, so the double-reporting of this AE cannot be ascribed to persistence from the TBZ phase into the placebo phase in the absence of a washout between phases. Eleven patients reported drowsiness during only one of the study phases; in 8 cases, this coincided with the TBZ phase. Clearly, drowsiness is a frequent and important AE. In the randomized, double-blind, 3-way crossover trial by McLellan et al,8 TBZ, the phenothiazine thiopropazate, and placebo were compared in 10 patients with chorea (9 due to Huntington’s disease, 1 due to cerebral palsy). TBZ was titrated upward from 50 to 200 mg/d in increments of 25 mg every 2 days; thiopropazate was titrated upward from 5 to 30 mg/d in increments of 5 mg every 2 days. The total duration of therapy was 6 weeks, with no washout between phases. TBZ therapy was associated with marked improvement (“virtually complete suppression of the chorea”) in 5 patients (50%), moderate improvement in 2 patients (20%), slight improvement in 1 patient (10%), and no improvement in 1 patient (10%). One patient developed severe parkinsonism at a TBZ dose of 25 mg/d and was withdrawn from the trial. Thiopropazate therapy was associated with considerable improvement in

364

1 patient (10%), slight improvement in 3 patients (30%), and no improvement in 6 patients (60%). Placebo was associated with slight improvement in 3 patients (30%), no improvement in 6 patients (60%), and worsening in 1 patient (10%). No statistical analysis was provided. Eight blinded physicians evaluated videotapes from each study phase to assign chorea scores. Mean (SEM) pretreatment, TBZ, thiopropazate, and placebo scores were 2.06 (0.10), 1.24 (0.06), 1.53 (0.12), and 1.72 (0.17), respectively (P P < 0.001, pretreatment vs TBZ; P < 0.01, pretreatment vs thiopropazate). Scores for TBZ and thiopropazate were not compared statistically. During the TBZ, thiopropazate, and placebo phases, there were a total of 18 AEs in 8 patients, 7 AEs in 6 patients, and 10 AEs in 4 patients, respectively. Manual dexterity was tested as a marker of improvement in choreiform movements. The mean times to completion of the dexterity test with each hand in normal control subjects and TBZ, thiopropazate, and placebo recipients were 47.0, 74.1, 81.7, and 82.6 seconds, respectively. The corresponding mean times to write a standard sentence with the dominant hand were 18.0, 41.2, 45.6, and 61.2 seconds (statistical analysis not provided). In a randomized, double-blind, crossover trial, Asher and Aminofff87 compared TBZ and placebo in 32 patients with a variety of abnormal movement disorders (9 with Huntington’s chorea, 12 with tardive dyskinesia, and 11 with segmental or generalized dystonias). TBZ was initiated at 25 mg BID, followed by upward titration in daily increments of 25 mg every 3 days until the maximal therapeutic response was attained, AEs limited further dose escalation, or the maximum allowable dose of 200 mg/d was reached. The duration of the study was 6 weeks, with no washout between phases. Among the 26 patients (81%) who completed the trial, 15 (58%) exhibited clinical improvement, 10 (38%) had no change from baseline, and 1 (4%) exhibited worsening. Overall, 68% of patients had at least moderate clinical improvement. Patients were not evaluated in the same manner during the placebo phase as during the TBZ phase; it appears that placebo was used solely to determine whether clinical improvement/worsening could be attributed to TBZ. Sixteen patients (62%) chose to continue TBZ after the end of the study—7 of 8 (88%) with Huntington’s chorea, 7 of 10 (70%) with tardive dyskinesia, and 2 of 8 (25%) with dystonia. Of the 7 early withdrawals from the study, 6 (86%) occurred during the TBZ phase. AEs were cited by patients in the context of study withdrawal in only 3 cases (43%). Drowsiness, hypersaliva-

D.R.P. Guay

tion, parkinsonism, and depression were noted in 30%, 12%, 9%, and 6% of patients, respectively. Twenty patients with a variety of abnormal movement disorders (6 with Meige’s syndrome, 4 with tardive dyskinesia, 3 with adult-onset dystonia, 2 with dystonic choreoathetosis, 4 with various rarer dystonias, and 1 with Huntington’s disease) participated in a randomized, double-blind, crossover trial by Jankovic.88,89 TBZ was initiated at 25 mg/d and was titrated upward in 25-mg/d increments to 200 mg/d, in the absence of intolerable AEs. Patients were hospitalized at the beginning of each phase (mean of 10 days in first phase and 7 days in second phase). The duration of the treatment phases was not fixed, but was reported to be ~6 weeks each (range, 1–7 weeks). The treatment response was measured using an unvalidated hyperkinesia rating scale, as well as being evaluated by blinded assessment of videotapes made while patients performed standard tests. Overall, there was a significant reduction in the number of abnormal movements during the TBZ phase compared with the placebo phase (P P < 0.005). The responses differed by disorder. All 4 patients with tardive dyskinesia improved during the TBZ phase. Among the 6 patients with Meige’s syndrome, 4 had what was described as “definite” improvement during TBZ therapy and 1 each had no change or worsened due to emergence of parkinsonism and study withdrawal. Among the 9 patients with dystonic disorders, all exhibited some degree of improvement, while the patient with Huntington’s disease was “markedly” improved. Overall, 13 of 19 patients (68%) had at least moderate improvement (defined as a reduction of ≥25% in number of abnormal movements) during TBZ therapy. AEs were reported in 13 of 19 patients (68%). There were a total of 32 AEs, the most frequent being drowsiness (5), hypersalivation (4), insomnia and tremor (3 each), and fatigue and parkinsonism (2 each). Brusa et al90 conducted a randomized, evaluatorblinded, placebo- and active-controlled crossover study comparing 6 months’ treatment with TBZ and the neuroleptic aripiprazole in patients with chorea associated with Huntington’s disease. Both drugs were slowly titrated to the maximum tolerated doses (mean [SD], 95.83 [33.20] and 10.76 [4.91] mg/d, respectively) and continued at those doses for the remainder of the 3-month treatment period. There was no washout phase between treatment periods. Both drugs were associated with significant reductions from baseline in the UHDRS chorea item score in the “off” condition (ie, after drug[s] for chorea were withheld for ≥3 weeks);

The American Journal of Geriatric Pharmacotherapy

from a baseline value of 10 (1.58), the chorea item score decreased to 4.6 (1.67) with TBZ and to 4.8 (1.30) with aripiprazole (both, P < 0.01). No significant drug effect was noted on scores on the UHDRS parkinsonism item or Mini–Mental State Examination score. However, TBZ therapy was associated with significant sedative effects, with scores on the Epworth Sleepiness Scale increasing from a baseline value of 8.8 (0.83) to 11.2 (1.30) (P < 0.01). Aripiprazole was not associated with a significant change from baseline in Epworth Sleepiness Scale score, and the score with aripiprazole (9.0 [1.00]) was significantly lower than that with TBZ (P < 0.01). TBZ was also associated with a significant increase from baseline in scores on the Hamilton Rating Scale for Depression (P < 0.01); the difference between aripiprazole and TBZ was statistically significant (P < 0.01).

Noncrossover Trials Kazamatsuri et al91 conducted a 19-week, randomized, evaluator-blinded, active-controlled trial of TBZ and haloperidol in 13 patients with the typical abnormal buccolingualmasticatory movements associated with neuroleptic-induced tardive dyskinesia. TBZ was initiated at 50 mg BID and increased to 100 mg BID at week 15; haloperidol was initiated at 4 mg BID and increased to 8 mg BID at week 15. Placebo was substituted for active treatment at week 19 (end of the study). In the 6 TBZ recipients, 2 patients had a complete cessation of abnormal movements at the end of week 2, and 4 had no response. The mean frequency of oral dyskinesias decreased from 25.7 per minute at baseline to 12.8 per minute at the end of week 2 and to 16.8 per minute at the end of week 6. The TBZ response was maintained at approximately the same level from the end of week 6 through the end of week 18. Withdrawal of TBZ led to a rapid reemergence of dyskinesias at baseline levels of severity in all patients. TBZ was described as having significantly reduced the frequency of dyskinesias over time relative to baseline (P < 0.05), although no specific data were reported. In the 7 haloperidol recipients, 5 patients exhibited a complete cessation of oral dyskinesias at the end of week 2, and 2 had a marked reduction in the frequency of dyskinesias. Dyskinesia frequency decreased from a mean of 34.9 per minute at baseline to a mean of 2.4 per minute at the end of week 2. At the end of week 4, 4 haloperidol recipients had a complete cessation of dyskinesias and 3 had a reemergence of dyskinesias to nearly baseline frequencies (mean dyskinesia frequency in all 7 haloperidol recipients was 18.0 per minute). At

365

The American Journal of Geriatric Pharmacotherapy

D.R.P. Guay

the end of week 8, 2 haloperidol recipients had complete cessation of dyskinesias and 5 had continued dyskinesias (P < 0.01 for the latter), albeit at a lower frequency than at baseline (mean dyskinesia frequency in all 7 haloperidol recipients was 24.1 per minute). At the end of week 17 (ie, 2 weeks after doubling the haloperidol dose), 3 haloperidol recipients had a considerable reduction in dyskinesias, 2 had no change, and 2 could not tolerate the dose escalation and had to be withdrawn from therapy; the mean dyskinesia frequency was 21.8 per minute, still significantly less frequent than at baseline (34.9 per minute) (P P < 0.01). At the end of week 19 (when placebo replaced active drug), oral dyskinesias rapidly reappeared at baseline levels of severity. Haloperidol was said to significantly reduce the frequency of dyskinesias over time relative to baseline (P < 0.05), although again, no specific data were reported. Neither drug had a noticeable effect on patients’ behaviors. Reversible extrapyramidal symptoms appeared to be negatively correlated with changes in the frequency of oral dyskinesias with haloperidol (r = 0.6292; P < 0.02). However, only 40% of the variance in the data could be explained by this relationship. The same correlation was not significant for TBZ (r = 0.4149). The authors reported a lack of “significant” AEs with TBZ, whereas 2 haloperidol recipients developed severe malaise after dose escalation to 16 mg/d, necessitating their withdrawal from the study. In a 12-week, randomized, double-blind trial, the Huntington Study Group compared TBZ (n = 50) and placebo (n = 34) in patients with Huntington’s chorea.92 TBZ was initiated at 12.5 mg on day 1, increased to 12.5 mg BID on days 2 through 7, and increased by 12.5 mg/d at weekly intervals to a maximum of 100 mg/d (given in 3 divided doses). TBZ was associated with significant reductions from baseline in mean total maximum UHDRS chorea scores compared with placebo (–5.0 vs –1.5, respectively; P < 0.001) and a significant improvement in absolute CGI scores (to 3.0 vs 3.7; P = 0.007). More than minimal clinical improvement in chorea scores occurred in 45.1% of TBZ recipients and 6.9% of placebo recipients (P P < 0.001). At least minimal clinical improvement occurred in 69% and 24% of the respective groups (P P < 0.001). There were no significant differences between groups in UHDRS motor scores, color-naming test scores, or interference test scores. Placebo was significantly better than TBZ in terms of scores on the following measures: UHDRS functional checklist (P P = 0.02), 17-item Hamilton Rating Scale for Depression (P P = 0.003), Epworth Sleepiness Scale (P P = 0.02), and Stroop word-reading test (P =

366

0.01). Taken in order, these results suggest that TBZ may have clinically relevant negative effects on performance of activities of daily living, mood, wakefulness, and cognition, respectively. TBZ had no effect on scores on the following measures: UHDRS gait assessment, UHDRS parkinsonism assessment, Functional Impact Scale, UHDRS independence scale, Barnes Akathisia Scale, UHDRS behavioral assessment, and UPDRS swallowing/speech scores. When TBZ was discontinued, the resulting chorea scores were significantly worse compared with those of former placebo recipients (P P < 0.001), as were CGI scores (P = 0.01). CGI scores were at least minimally worse in 71% of TBZ recipients and 22% of placebo recipients (P P = 0.01). The proportions of patients experiencing at least one serious AE did not differ significantly between groups. However, the mean (SE) number of new AEs per patient was significantly higher in TBZ recipients compared with placebo recipients (all events: 3.8 [3.1] vs 1.5 [1.8], respectively [P P = 0.001]; excluding mild events: 1.9 [2.0] vs 0.6 [0.9] [P P = 0.001]). The proportion of patients with at least one new AE was significantly higher in TBZ recipients compared with placebo recipients (all events: 49/54 [91%] vs 21/30 [70%] [P P = 0.01]; excluding mild events: 37/54 [69%] vs 10/30 [33%] [P P = 0.002]). At week 12, more TBZ recipients required a dose reduction due to AEs compared with placebo recipients (24/54 [44%] vs 1/30 [3%]; P < 0.001). Dose-limiting AEs associated with TBZ included sedation (27%), akathisia (8%), and parkinsonism and depression (4% each). Frank et al93 conducted a randomized, double-blind trial of TBZ withdrawal (on day 1) (n = 12), partial withdrawal (on day 3) (n = 12), and no withdrawal (n = 6) in patients with Huntington’s chorea over an observation period of 5 days. Adjusted chorea scores (the primary outcome) increased by a mean of 5.3 units from day 1 to day 3 in the withdrawal group, compared with an increase of 3.0 units in the pooled partialwithdrawal and no-withdrawal groups (P P = NS). Similar results were noted when age (dichotomized at the median) was added to the statistical model. The treatment-by-age interaction was not statistically significant. Because of a significant difference in total motor scores at baseline across groups (P P = 0.006), chorea scores were adjusted post hoc for baseline motor scores; the results remained nonsignificant. Four patients continued use of a neuroleptic throughout the study (1 each taking haloperidol and fluphenazine for chorea, 1 taking fluphenazine for unknown reasons, 1 taking quetiapine for sleep). When data from these 4 patients were

D.R.P. Guay

excluded, the results were again nonsignificant. Adjustment for benzodiazepine use also had no effect. Chorea scores on day 1 and day 5 were not subjected to statistical analysis. Post hoc analysis of linear trends was positive for reemerging chorea (P P = 0.049). No significant changes were seen in total functional capacity (the secondary outcome) or in the exploratory end points, which included the UHDRS total motor score, cognitive assessment, behavioral assessment, functional assessment, independence scale, and clinical summary scores; the UPDRS dysarthria score; individual UHDRS items (gait score, verbal fluency, symbol digit modalities test, Stroop word-reading test, and Stroop interference test); and the CGI. No recognizable withdrawal syndrome (other than reemergence of chorea) was noted.

TOLERABILITY During product development, TBZ was administered to 773 unique individuals.3 Dosing and exposure varied widely, from single- and multiple-dose clinical pharmacology studies in healthy volunteers (n = 259) to open-label (n = 529) and double-blind (n = 84) clinical studies in patients. The AEs from a pivotal randomized, placebo-controlled clinical trial in patients with Huntington’s disease have been summarized in the prescribing information (Table IV).3 TBZ dose escalation was stopped or the dose reduced due to one or more AEs in 28 of 54 patients (52%). These AEs included sedation (15 cases), akathisia (7), parkinsonism (4), depression (3), anxiety (2), and fatigue and diarrhea (1 each). (The number of cases exceeds the number of patients with AEs, as patients could have had more than one AE and be counted more than once.) The overall frequency of extrapyramidal symptoms was 33%, including 10 patients (19%) with akathisia (which included the preferred terms akathisia, hyperkinesias, s and restlessness) and 8 patients (15%) with extrapyramidal events (which included the preferred terms bradykinesia, parkinsonism, extrapyramidal disorder, and hypertonia). TBZ was approved by the FDA despite its association with several serious and potentially fatal AEs. The most problematic of these are depression and suicidality. The frequency of depression in various case series and clinical studies has varied, but rates ranging from 4% to 42% (mean, 20%) have been reported. Patients developing depression/suicidality should be evaluated immediately, and a reduction in the TBZ dose and/or initiation of antidepressant therapy should be given strong consideration. Caution is necessary in selecting an antidepressant, as some antidepressants are potent inhibitors of CYP2D6 (eg, paroxetine, fluoxetine). If an antidepres-

The American Journal of Geriatric Pharmacotherapy

Table IV. Treatment-emergent adverse events occurr ring in ≥4% of the tetrabenazine group and with a higher frequency than in the placebo group in a 12-week, double-blind, placebocontrolled trial in patients with Huntington’s disease.3 Values are number (%) of patients. #PEZ4ZTUFN "EWFSTF&WFOU 1TZDIJBUSJD 4FEBUJPOTPNOPMFODF *OTPNOJB %FQSFTTJPO "OYJFUZBOYJFUZBHHSBWBUFE *SSJUBCJMJUZ %FDSFBTFEBQQFUJUF 0CTFTTJWFSFBDUJPO

5FUSBCFOB[JOF O

1MBDFCP O

 

 

 

 

 

 

 

 

o o  

 

o o

$FOUSBMBOEQFSJQIFSBMOFSWPVTTZTUFN "LBUIJTJB  

#BMBODFEJGGJDVMUZ  

1BSLJOTPOJTNCSBEZLJOFTJB  

%J[[JOFTT  

%ZTBSUISJB  

(BJUVOTUFBEZ  

)FBEBDIF  

(BTUSPJOUFTUJOBMTZTUFN /BVTFB 7PNJUJOH

o o o o o o o

 

 

 

 

 

 

 

 

 

 

o o

3FTQJSBUPSZTZTUFN 6QQFSSFTQJSBUPSZUSBDUJOGFDUJPO #SPODIJUJT 4IPSUOFTTPGCSFBUI

 

 

 

 

o o

6SJOBSZTZTUFN %ZTVSJB

 

o

#PEZBTBXIPMF 'BUJHVF 'BMM -BDFSBUJPO IFBE

&DDIZNPTJT

sant with strong CYP2D6-inhibitory activity is selected, the TBZ dose should be reduced by 50%. If depression/ suicidality does not resolve, consideration should be given to discontinuation of TBZ. Patients with a history of depression or other psychiatric disorder should be carefully monitored during TBZ therapy.

367

The American Journal of Geriatric Pharmacotherapy

D.R.P. Guay

Kenney et al94 conducted a retrospective review of the charts of 518 patients with hyperkinetic syndromes and a history of depression to examine whether TBZ treatment was associated with recurrent or worsening depression. The patients ranged in age from 3 to 87 years and had one or more hyperkinetic syndromes—chorea in 162, dyskinesias in 156, dystonias in 142, tics in 95, myoclonus in 19, and other disorders in 15. The mean (SD) daily dose of TBZ at the last visit was 61.6 (38.7) mg, and the maximum dose achieved in any patient was 300 mg. The most frequent AEs were somnolence (27.4%), depression (15.1%), parkinsonism (11.8%), and akathisia (8.9%). At baseline, 272 patients (52.5%) had a history of depression and 246 (47.5%) did not. Among those with no history of depression, 28 (11.4%) developed depression during the study; among those with a documented history of depression, 50 (18.4%) developed a clinically significant exacerbation of depression (P P = 0.03). Of interest, patients with a history of depression had greater improvement (lower ratings) in abnormal movements than did those without a history of depression (mean response ratings: 1.8 vs 2.1, respectively; P < 0.01). Sixteen patients (3.1%) discontinued TBZ due to depression, 7 (2.6%) of those with a history of depression and 9 (3.7%) of those without a history (P P = NS). These 2 groups did not differ in terms of the proportion experiencing one or more AEs leading to drug discontinuation. Based on the findings of this study, depression associated with TBZ therapy appears more likely to occur or worsen in individuals with a history of depression than in those without such history. As a result of the dopamine blockade induced by TBZ, parkinsonism is a common dose-related AE.95 TBZ-associated parkinsonism is characterized by the bradykinesia, hypertonia, and rigidity seen in the idiopathic form of the disease. The appearance of parkinsonian symptoms may necessitate reduction of the TBZ dose or, in some cases, drug discontinuation. Carbidopa/ levodopa therapy for parkinsonism may sometimes produce symptomatic benefit without causing a worsening of abnormal involuntary movements. Extrapyramidal symptoms such as akathisia are common dose-related AEs of TBZ therapy, again as a result of dopamine blockade. Restlessness and agitation may be indicators of imminent akathisia. Appearance of these symptoms frequently necessitates a reduction in the TBZ dose, and drug discontinuation may be necessary in a small number of cases. The constellation of AEs such as akathisia, parkinsonism, dysphagia, sedation, hypotension, and cognitive decline may also be a

368

manifestation of the progression of Huntington’s disease, thereby creating diagnostic ambiguity.96 Neuroleptic malignant syndrome is a potentially fatal AE associated with drugs interfering with dopaminergic neurotransmission, including TBZ.97 This AE may recur when the patient is rechallenged with the drug. Despite the breadth of potentially serious and occasionally fatal AEs associated with TBZ, relatively few AE reports have appeared in the literature over the years. There are published reports of neuroleptic malignant syndrome/hyperthermia in 5 patients, parkinsonism in 4 patients, acute dystonias in 4 patients, catastrophic (florid psychiatric) reactions in 2 patients, and depression in 1 patient.95,98–107 A multicenter trial by Mikkelsen108 evaluated the long-term tolerability of TBZ in 124 patients with a variety of hyperkinetic syndromes (primarily Huntington’s disease, abnormal buccolingualmasticatory movements, and tardive dyskinesia). Of the original 124 patients, 18 were lost to follow-up, 5 died (unrelated to therapy), 3 were considered clinical failures, 5 discontinued due to AEs, and 2 were discontinued because long-term therapy was not considered appropriate. Thus, results were available for 93 patients, 12 of them TBZ naive and 81 who had been receiving TBZ for a median of 2.3 years (maximum, 15 years). The median age was 67.3 years in the 35 males and 59.0 years in the 58 females. The median daily dose of TBZ was 70 mg both before and after the study (range, 12.5–225 mg). Observations were available for ≥12 months in 82% of patients and for <6 months in 6%. Twenty-one percent of patients had complaints that may have represented AEs, most frequently fatigue. Five of 124 enrollees (4%) discontinued due to AEs (depression in 2 and fatigue, sweating, and malaise in 1 each). No clinically important laboratory abnormalities were noted.

DOSING AND ADMINISTRATION In the United States, TBZ is available only through prescribers and pharmacists who are specially qualified to prescribe and dispense it. This is a component of the Risk Evaluation and Mitigation System required by the FDA as a condition of TBZ’s approval. A year’s supply of TBZ 50 mg/d costs ~$40,000. The manufacturer has a financial assistance program to make TBZ affordable for eligible patients.1 TBZ is available in unscored 12.5-mg tablets and scored 25-mg tablets.3 The initial dosage is 12.5 mg once daily in the morning. After a week, the dosage is increased to 12.5 mg BID. The dosage can then be titrated slowly upward in weekly increments of 12.5 mg/d. Doses

D.R.P. Guay

of 37.5 to 50 mg should be given 3 times daily. The maximum recommended single dose is 25 mg. If TBZ therapy is interrupted for >5 days, it should be retitrated, starting at 12.5 mg once daily in the morning. In the case of an interruption of <5 days, TBZ can be restarted at the previous dose without titration. Patients in whom a TBZ dose >50 mg/d is desired should first undergo genotyping for CYP2D6 metabolizer status, if the test is available.3 In CYP2D6 extensive and intermediate metabolizers requiring >50 mg/d, the dose should be titrated in weekly increments of 12.5 mg/d. All doses >50 mg/d should be given 3 times daily. The maximum recommended daily dose in these patients is 100 mg, and the maximum single dose is 37.5 mg. Dosing is similar in CYP2D6 poor metabolizers, except that the maximum recommended daily dose is 50 mg and the maximum single dose is 25 mg. The TBZ dose should be halved in patients who are about to initiate concurrent therapy with strong CYP2D6 inhibitors (eg, fluoxetine, paroxetine, quinidine).3 In patients taking strong CYP2D6 inhibitors who are about to begin concurrent TBZ therapy, the dosing recommendations for CYP2D6 poor metabolizers should be followed. The effects of moderate or weak CYP2D6 inhibitors on TBZ pharmacokinetics have not been studied. TBZ use should be avoided in patients with any degree of hepatic impairment.

FUTURE RESEARCH QUESTIONS Although TBZ is approved by the FDA for the treatment of chorea in Huntington’s disease, there have been no comparative studies with neuroleptics (particularly haloperidol), which have been the drugs of choice for this disorder. Neuroleptics and TBZ have similar AE profiles, although given the serious AEs (eg, depression/suicidality) uniquely associated with TBZ, some incremental efficacy relative to neuroleptics should be required to justify its risks and cost. Although a recent Cochrane review found that among the 3 medication classes examined (antidopaminergics [n = 5], glutamate antagonists [n = 5], and energy metabolites [n = 5]), only TBZ had clear efficacy in the treatment of chorea,109 practitioners continue to believe in the efficacy of neuroleptics for this disorder. In terms of other hyperkinetic movement disorders, TBZ has been recommended for the tardive stereotypy, tardive chorea, and tardive tremor components of tardive dyskinesia,110 although this recommendation was based on the limited evidence of efficacy from randomized controlled trials. Given the cost of the test and the shortage of clinical laboratories offering it, more research is

The American Journal of Geriatric Pharmacotherapy

needed into the risk of not following the recommendation that CYP2D6 genotyping be performed before use of a TBZ dose >50 mg/d. Furthermore, there are almost no published data on the efficacy and tolerability of TBZ use over a period of years. There is a need for investigation of the magnitude and clinical importance of TBZ-induced cognitive impairment and impairment in the performance of activities of daily living, concerns brought to light in the course of the FDA’s review of the New Drug Application for TBZ.12 In addition, pharmacokinetic and drug–drug interaction studies are necessary to refine dosing recommendations.

CONCLUSIONS Approved by the FDA for the chorea of Huntington’s disease, TBZ has also been investigated in a variety of hyperkinetic movement disorders. It is associated with numerous AEs and several important drug–drug interactions. Over time and with the conduct of additional research trials, its role in the therapeutic armamentarium will become better defined.

ACKNOWLEDGMENT The author has indicated that he has no conflicts of interest with regard to the content of this article.

REFERENCES 1. Tetrabenazine (Xenazine) for Huntington’s chorea. Med Lett Drugs Ther. 2009;51:7–8. 2. Lingjaerde O. Tetrabenazine (Nitoman) in the treatment of psychoses. With a discussion on the central mode of action of tetrabenazine and reserpine. Acta Psychiatr Scand. 1963;39(Suppl 170):1–109. 3. Tetrabenazine (Xenazine tablets) [package insert]. Mississauga, Canada: Biovail Corporation; May 2008. 4. Pletscher A, Brossi A, Gey KF. Benzoquinolizine derivatives: A new class of monoamine decreasing drugs with psychotropic action. Int Rev Neurobiol. 1962;4:275– 306. 5. Henry JP, Scherman D. Radioligands of the vesicular monoamine transporter and their use as markers of monoamine storage vesicles. Biochem Pharmacol. 1989; 38:2395–2404. 6. Peter D, Vu T, Edwards RH. Chimeric vesicular monoamine transporters identify structural domains that influence substrate affinity and sensitivity to tetrabenazine. J Biol Chem. 1996;271:2979–2986. 7. Gonzalez AM, Walther D, Pazos A, Uhl GR. Synaptic vesicular monoamine transporter expression: Distribution and pharmacologic profile. Brain Res Mol Brain Res. 1994;22:219–226.

369

The American Journal of Geriatric Pharmacotherapy

D.R.P. Guay

8. McLellan DL, Chalmers RJ, Johnson RH. A doubleblind trial of tetrabenazine, thiopropazate, and placebo in patients with chorea. Lancet. 1974;1:104–107. 9. Pearson SJ, Reynolds GP. Depletion of monoamine transmitters by tetrabenazine in brain tissue in Huntington’s disease. Neuropharmacology. 1988;27:717–719. 10. Stumpf W, Graul EH, Hundeshagen H. Experimentelle Untersuchungen zur Pharmakologie des mit 3H-markierten tetra-benazines. Arzneim Forsch. 1961;11:47–49. 11. Kilbourn MR, DaSilva JN, Frey KA, et al. In vivo imaging of vesicular monoamine transporters in human brain using [11C]tetrabenazine and positron emission tomography. J Neurochem. 1993;60:2315–2318. 12. US Food and Drug Administration, Center for Drug Evaluation and Research. New Drug Application No. 21894: Tetrabenazine. Medical and Clinical Pharmacology reviews. 13. Mehvar R, Jamali F, Watson MW, Skelton D. Pharmacokinetics of tetrabenazine and its major metabolite in man and rat. Bioavailability and dose dependency studies. Drug Metab Dispos. 1987;15:250–255. 14. Roberts MS, McLean S, Millingen KS, Galloway HM. The pharmacokinetics of tetrabenazine and its hydroxyl metabolite in patients treated for involuntary movement disorders. Eur J Clin Pharmacol. 1986;29:703–708. 15. Schwartz DE, Bruderer H, Rieder J, Brossi A. Metabolic studies of tetrabenazine, a psychotropic drug in animals and man. Biochem Pharmacol. 1966;15:645–655. 16. Kilbourn MR, Lee LC, Heeg MJ, Jewett DM. Absolute configuration of (+)-alpha-dihydrotetrabenazine, an active metabolite of tetrabenazine. Chirality. 1997;9: 59–62. 17. Reches A, Hassan MN, Jackson VR, Fahn S. Lithium attenuates dopamine depleting effects of reserpine and tetrabenazine but not that of alpha methyl-p-tyrosine. Life Sci. 1983;33:157–160. 18. Furukawa T, Ushizima I, Ono N. Modifications by lithium of behavioral responses to methamphetamine and tetrabenazine. Psychopharmacologia. 1975;42:243–248. 19. Malik A, Balkoski V. Neurotransmitter depleter tetrabenazine; potential candidate for schizophrenia treatment? Schizophr Res. 2007;96:267–268. 20. Pearce J. Reversal of hemiballismus by tetrabenazine. JAMA. 1972;219:1345. Letter. 21. Riley D, Lang AE. Hemiballism in multiple sclerosis. Mov Disord. 1988;3:88–94. 22. Martí-Massó JF, Obeso JA. Coprolalia associated with hemiballismus: Response to tetrabenazine. Clin Neuropharmacol. 1985;8:189–190. 23. Geoghegan MF, Khaw CH, McCormick PA. Hemiballismus. Ir Med JJ. 2002;95:123–124.

370

24. Dewey RB Jr, Jankovic J. Hemiballism-hemichorea. Clinical and pharmacologic findings in 21 patients. Arch Neurol. 1989;46:862–867. 25. Laing RW, Howell SJ. Acute bilateral ballism in a patient with intravascular dissemination of gastric carcinoma. Neuropathol Appl Neurobiol. 1992;18:201–205. 26. Sitburana O, Ondo WG. Tetrabenazine for hyperglycemicinduced hemichorea-hemiballismus. Mov Disord. 2006; 21:2023–2025. 27. Shanahan P, Hutchinson M, Bohan A, et al. Hemichorea, moya-moya, and ulcerative colitis. Mov Disord. 2001; 16:570–572. 28. Brandrup E. Tetrabenacine treatment in persisting dyskinesia caused by psychopharmaca. Am J Psychiatry. 1961; 118:551–552. 29. MacCallum WA. Tetrabenazine for extra-pyramidal movement disorders. Br Med JJ. 1970;1:760. 30. Srinivasan K, Mouli KS, Viegas B, et al. Metoclopramide induced tardive dyskinesia. J Indian Med Assoc. 1991; 89:260–261. 31. Fève A, Angelard B, Lacau St Guily J. Laryngeal tardive dyskinesia. J Neurol. 1995;242:455–459. 32. Koch HJ, Szecsey A, Vogel M, Fischer-Barnicol D. Successful therapy of tardive dyskinesia in a 71-year-old woman with a combination of tetrabenazine, olanzapine and tiapride. Int J Clin Pract. 2003;57:147–149. 33. Stacy M, Jankovic J. Tardive tremor. Mov Disord. 1992; 7:53–57. 34. Storey E, Lloyd J. Tardive tremor. Mov Disord. 1997; 12:808–810. 35. Singh SK, Jankovic J. Tardive dystonia in patients with Tourette’s syndrome. Mov Disord. 1988;3:274– 280. 36. Rauchverger B, Isakov V, Jabarin M. Olanzapine-induced tardive dystonia successfully treated by tetrabenazine. J Neuropsychiatry Clin Neurosci. 2007;19:484–485. 37. Kalian M, Lerner V, Goldman M. Atypical variants of tardive dyskinesia, treated by a combination of clozapine with propranolol and clozapine with tetrabenazine. J Nerv Ment Dis. 1993;181:649–651. 38. FitzGerald PM, Jankovic J. Tardive oculogyric crises. Neurology. 1989;39:1434–1437. 39. Soutar CO. Tetrabenazine for Huntington’s chorea. Br Med JJ. 1970;4:55. 40. Hoehn MM, Cherington M. Spinal myoclonus. Neurology. 1977;27:942–946. 41. Jankovic J, Pardo R. Segmental myoclonus. Clinical and pharmacologic study. Arch Neurol. 1986;43:1025– 1031. 42. Hawkes CH, Nourse CH. Tetrabenazine in Sydenham’s chorea. Br Med JJ. 1977;1:1391–1392.

D.R.P. Guay

43. Vecht CJ, Meerwaldt JD, Lees AJ, et al. What is it? Case 2, 1991: Unusual tremor, myoclonus, and a limping gait. Mov Disord. 1991;6:371–375. 44. Linazasoro G, Urtasun M, Poza JJ, et al. Generalized chorea induced by nonketotic hyperglycemia. Mov Disord. 1993;8:119–120. 45. Papapetropoulos S, Singer C. Management of the extrapyramidal syndrome in chronic acquired hepatocerebral degeneration (CAHD). Mov Disord. 2005;20:1088– 1089. 46. Lagueny A, Burbaud P, Le Masson G, et al. Involvement of respiratory muscles in adult-onset dystonia: A clinical and electrophysiological study. Mov Disord. 1995;10: 708–713. 47. Vieregge P. Tetrabenazine in the treatment of senile vocal tics. J Neurol. 1987;235:126–127. 48. Alentorn A, Palasí A, Campdelacreu J, et al. Pelvic dyskinesia with an outstanding response to tetrabenazine. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:847–848. 49. Papapetropoulos S, Singer C. Eating dysfunction associated with oromandibular dystonia: Clinical characteristics and treatment considerations. Head Face Med. 2006;2:47. 50. Pakkenberg H. The effect of tetrabenazine in some hyperkinetic syndromes. Acta Neurol Scand. 1968;44:391– 393. 51. Dalby MA. Effect of tetrabenazine on extrapyramidal movement disorders. Br Med JJ. 1969;2:422–423. 52. Astin KJ, Gumpert EW. Letter: Tetrabenazine in chorea. Lancet. 1974;1:512. 53. Kingston D. Tetrabenazine for involuntary movement disorders. Med J Aust. 1979;1:628–630. 54. Jankovic J, Ford J. Blepharospasm and orofacial-cervical dystonia: Clinical and pharmacological findings in 100 patients. Ann Neurol. 1983;13:402–411. 55. Fahn S. A therapeutic approach to tardive dyskinesia. J Clin Psychiatry. 1985;46:19–24. 56. Watson MW, Skelton D, Jamali F. Treatment of tardive dyskinesia: Preliminary report on use of tetrabenazine. Can J Psychiatry. 1988;33:11–13. 57. Jankovic J, Beach J. Long-term effects of tetrabenazine in hyperkinetic movement disorders. Neurology. 1997;48: 358–362. 58. Paleacu D, Giladi N, Moore O, et al. Tetrabenazine treatment in movement disorders. Clin Neuropharmacol. 2004;27:230–233. 59. Kenney C, Hunter C, Jankovic J. Long-term tolerability of tetrabenazine in the treatment of hyperkinetic movement disorders. Mov Disord. 2007;22:193–197. 60. Fog R, Pakkenberg H. Combined nitoman-pimozide treatment of Huntington’s chorea and other hyperkinetic syndromes. Acta Neurol Scand. 1970;46:249–251.

The American Journal of Geriatric Pharmacotherapy

61. Swash M, Roberts AH, Zakko H, Heathfield KW. Treatment of involuntary movement disorders with tetrabenazine. J Neurol Neurosurg Psychiatry. 1972;35:186– 191. 62. McLellan DL. The suppression of involuntary movements with tetrabenazine. Scott Med JJ. 1972;17:367– 370. 63. Kazamatsuri H, Chien C, Cole JO. Treatment of tardive dyskinesia. I. Clinical efficacy of a dopamine-depleting agent, tetrabenazine. Arch Gen Psychiatry. 1972;27:95– 99. 64. Pakkenberg H, Fog R. Spontaneous oral dyskinesia. Results of treatment with tetrabenazine, pimozide, or both. Arch Neurol. 1974;31:352–353. 65. Sweet RD, Bruun R, Shapiro E, Shapiro AK. Presynaptic catecholamine antagonists as treatment for Tourette syndrome. Effects of alpha methyl para tyrosine and tetrabenazine. Arch Gen Psychiatry. 1974;31:857–861. 66. McArthur AW, Pollock M, Smidt NA. Combined therapy with tetrabenazine and pimozide in Huntington’s chorea: Pilot study. N Z Med JJ. 1976;83:114–116. 67. Huang CY, McLeod JG, Holland RT, Elliot C. Tetrabenazine in the treatment of Huntington’s chorea. Med J Aust. 1976;1:583–584. 68. Toglia JU, McGlamery M, Sambandham RR. Tetrabenazine in the treatment of Huntington’s chorea and other hyperkinetic movement disorders. J Clin Psychiatry. 1978;39:81–87. 69. Lang AE, Marsden CD. Alpha methylparatyrosine and tetrabenazine in movement disorders. Clin Neuropharmacol. 1982;5:375–387. 70. Jankovic J, Glaze DG, Frost JD Jr. Effect of tetrabenazine on tics and sleep of Gilles de la Tourette’s syndrome. Neurology. 1984;34:688–692. 71. Fahn S. Long-term treatment of tardive dyskinesia with presynaptically acting dopamine-depleting agents. Adv Neurol. 1983;37:267–276. 72. Marsden CD, Marion MH, Quinn N. The treatment of severe dystonia in children and adults. J Neurol Neurosurg Psychiatry. 1984;47:1166–1173. 73. Bartels M, Zeller E. Tetrabenazine (Nitoman) therapy of chronic spontaneous oral dyskinesia. A video- and EMGcontrolled study. Eur Arch Psychiatry Neurol Sci. 1984; 234:172–174. 74. Kang UJ, Burke RE, Fahn S. Natural history and treatment of tardive dystonia. Mov Disord. 1986;1:193–208. 75. Kang UJ, Burke RE, Fahn S. Tardive dystonia. Adv Neurol. 1988;50:415–429. 76. Jankovic J, Orman J. Tetrabenazine therapy of dystonia, chorea, tics, and other dyskinesias. Neurology. 1988;38: 391–394.

371

The American Journal of Geriatric Pharmacotherapy

D.R.P. Guay

77. Jankovic J, Caskey TC, Stout JT, Butler IJ. Lesch-Nyhan syndrome: A study of motor behavior and cerebrospinal fluid neurotransmitters. Ann Neurol. 1988;23:466–469. 78. Stacy M, Cardoso F, Jankovic J. Tardive stereotypy and other movement disorders in tardive dyskinesias. Neurology. 1993;43:937–941. 79. Ondo WG, Hanna PA, Jankovic J. Tetrabenazine treatment for tardive dyskinesia: Assessment by randomized videotape protocol. Am J Psychiatry. 1999;156:1279–1281. 80. Ondo WG, Tintner R, Thomas M, Jankovic J. Tetrabenazine treatment for Huntington’s disease-associated chorea. Clin Neuropharmacol. 2002;25:300–302. 81. Kenney C, Hunter C, Davidson A, Jankovic J. Shortterm effects of tetrabenazine on chorea associated with Huntington’s disease. Mov Disord. 2007;22:10–13. 82. Porta M, Sassi M, Cavallazzi M, et al. Tourette’s syndrome and role of tetrabenazine: Review and personal experience. Clin Drug Investig. 2008;28:443–459. 83. Fasano A, Cadeddu F, Guidubaldi A, et al. The longterm effect of tetrabenazine in the management of Huntington disease. Clin Neuropharmacol. l 2008;31:313–318. 84. Frank S, for the Huntington Study Group/TETRA-HD Investigators. Tetrabenazine as anti-chorea therapy in Huntington disease: An open-label continuation study. BMC Neurol. 2009;9:62. 85. Godwin-Austen RB, Clark T. Persistent phenothiazine dyskinesia treated with tetrabenazine. Br Med JJ. 1971;4: 25–26. 86. Gilligan BS, Wodak J, Veale JL, Munro OR. Tetrabenazine in the treatment of extrapyramidal dyskinesias. Med J Aust. 1972;2:1054–1056. 87. Asher SW, Aminoff MJ. Tetrabenazine and movement disorders. Neurology. 1981;31:1051–1054. 88. Jankovic J. Treatment of hyperkinetic movement disorders with tetrabenazine: A double-blind crossover study. Ann Neurol. 1982;11:41–47. 89. Jankovic J. Tetrabenazine in the treatment of hyperkinetic movement disorders. Adv Neurol. 1983;37:277–289. 90. Brusa L, Orlacchio A, Moschella V, et al. Treatment of the symptoms of Huntington’s disease: Preliminary results comparing aripiprazole and tetrabenazine. Mov Disord. 2009;24:126–129. 91. Kazamatsuri H, Chien CP, Cole JO. Long-term treatment of tardive dyskinesia with haloperidol and tetrabenazine. Am J Psychiatry. 1973;130:479–483. 92. Huntington Study Group. Tetrabenazine as antichorea therapy in Huntington disease: A randomized controlled trial. Neurology. 2006;66:366–372. 93. Frank S, Ondo W, Fahn S, et al. A study of chorea after tetrabenazine withdrawal in patients with Huntington disease. Clin Neuropharmacol. 2008;31:127–133.

372

94. Kenney C, Hunter C, Mejia A, Jankovic J. Is history of depression a contraindication to treatment with tetrabenazine? Clin Neuropharmacol. 2006;29:259– 264. 95. Giladi N, Melamed E. Levodopa therapy can ameliorate tetrabenazine-induced parkinsonism. Mov Disord. 1999; 14:158–159. 96. Novak MJ, Tabrizi SJ. Huntington’s disease. BMJ. 2010;340:C3109. 97. Ables AZ, Nagubilli R. Prevention, recognition, and management of serotonin syndrome. Am Fam Physician. 2010;81:1139–1142. 98. Petzinger GM, Bressman SB. A case of tetrabenazineinduced neuroleptic malignant syndrome after prolonged treatment. Mov Disord. 1997;12:246–248. 99. Ossemann M, Sindic CJ, Laterre C. Tetrabenazine as a cause of neuroleptic malignant syndrome. Mov Disord. 1996;11:95. 100. Kulisevsky J, Lima de Freitas M, Barraquer-Bordas L. Bilateral akinesia in drug-induced parkinsonism after a subthalamic lesion. Mov Disord. 1994;9:111–112. 101. Mateo D, Muñoz-Blanco JL, Giménez-Roldán S. Neuroleptic malignant syndrome related to tetrabenazine introduction and haloperidol discontinuation in Huntington’s disease. Clin Neuropharmacol. 1992;15:63– 68. 102. Moss JH, Stewart DE. Iatrogenic parkinsonism in Huntington’s chorea. Can J Psychiatry. 1986;31:865– 866. 103. Burke RE, Fahn S, Mayeux R, et al. Neuroleptic malignant syndrome caused by dopamine-depleting drugs in a patient with Huntington disease. Neurology. 1981; 31:1022–1025. 104. Bruneau MA, Lespérance P, Chouinard S. Catastrophic reactions induced by tetrabenazine. Can J Psychiatry. 2002;47:683. 105. Schreiber W, Krieg JC, Eichhorn T. Reversal of tetrabenazine induced depression by selective noradrenaline (norepinephrine) reuptake inhibition. J Neurol Neurosurg Psychiatry. 1999;67:550. 106. Stevens E, Roman A, Houa M, et al. Severe hyperthermia during tetrabenazine therapy for tardive dyskinesia. Intensive Care Med. 1998;24:369–371. 107. Burke RE, Reches A, Traub MM, et al. Tetrabenazine induces acute dystonic reactions. Ann Neurol. 1985; 17:200–202. 108. Mikkelsen BO. Tolerance of tetrabenazine during long-term treatment. Acta Neurol Scand. 1983;68:57– 60. 109. Mestre T, Ferreira J, Coelho MM, et al. Therapeutic interventions for symptomatic treatment in Hunting-

D.R.P. Guay

ton’s disease. Cochrane Database Syst Rev. 2009;(3): CD006456.

The American Journal of Geriatric Pharmacotherapy

110. Jankovic J. Treatment of hyperkinetic movement disorders. Lancet Neurol. 2009;8:844–856.

Address correspondence to: David R.P. Guay, PharmD, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 7-148 Weaver-Densford Hall, 308 Harvard Street SE, Minneapolis, MN 55455. E-mail: [email protected]

373