throbber
Tetrabenazine in the treatment of Huntington’s
`disease
`
`E X P E RT O P I N I O N
`
`Paleacu Diana
`Neurology Service and Memory
`Clinic, Abarbanel Mental Health
`Center, Bat-Yam, affiliated to the
`Sackler School of Medicine, Tel Aviv
`University, Tel Aviv, Israel
`
`Correspondence: Paleacu Diana
`Neurological Service and Memory Clinic,
`Abarbanel Mental Health Center, 15
`Keren Kayemet, 59110 Bat Yam, Israel
`Tel +972 3 555 2861
`Fax +972 3 555 2621
`Email paleacu@post.tau.ac.il
`
`Abstract: Tetrabenazine (TBZ), a catecholamine-depleting agent initially developed for the
`treatment of schizophrenia, when tested for other indications, has proven to be more useful for
`the treatment of a variety of hyperkinetic movement disorders. These disorders include neuro-
`logical diseases characterized by abnormal involuntary movements such as chorea associated
`with Huntington’s disease, tics in Tourette’s syndrome, dyskinesias and dystonias in tardive
`dyskinesia, also primary dystonias and myoclonus. This review will include and discuss stud-
`ies published during the period of 1960–2006 regarding the clinical effi cacy and tolerability
`of TBZ in Huntington’s disease (HD). It will also review the chemistry, pharmacokinetics and
`dynamics of the drug and its mechanism of action compared to that of reserpine, the only similar
`compound. This review emphasizes the advantage of TBZ over dopamine-depleting compounds
`used in the treatment of chorea and reveals its clinical effi cacy and side effects.
`Keywords: tetrabenazine, Huntington’s disease, chorea
`
`Introduction
`Tetrabenazine (TBZ) was initially synthesized in the 1950s by O Schneider and A
`Brossi at the research laboratory of Hoffmann-La Roche in Basel. They created TBZ
`as an antipsychotic drug as part of their research into simpler chemical compounds
`with reserpine-like activity. TBZ was studied in a number of controlled trials in
`schizophrenia patients with equivocal results (Smith 1960; Weckorciez 1960;
`Kanjilal 1962). Later clinicians came to favor the use of dopamine receptor blocking
`drugs (DRBD), like phenothiazines, for treating psychosis given the evidence of
`better effi cacy (Ashcroft 1961). However, as with many other drugs, when TBZ was
`tested for indications other than the original ones the preliminary results provided
`support for its use in disorders characterized by abnormal hyperkinetic involuntary
`movements, drug induced or primary. During the last two decades TBZ has been
`used in a multitude of movement disorders: tardive dyskinesia (Fahn 1985; Ondo
`1999; Simpson 2000; Tarsy 2000) dystonia (Manji 1998; Raja 1998; Scott 2000)
`and tremor (Storey 1997), choreic syndromes (Gimenez-Rodan 1989; Ondo 2002),
`primary dystonia (Bartels 1984; Faulstich 1985; Manji 1998; Boghen 2000; Scott
`2000), secondary dystonia (Duran 2001) and tic disorders (Vieregge 1987; Jankovic
`1988; Scahill 2000).
`
`Chemistry
`TBZ, also named Ro 1-9569 by Hoffmann-LaRoche, Inc., is a benzoquinolizine
`derivative with the following formula, 2-oxo-3-isobutyl-9,10-dimethoxy-1,2,3,4,6,7-
`hexahydrobenzo[a]quinolizine. It is a white crystalline substance with a bitter taste. The
`molecule is shown in Figure 1. It can be isolated from alkalinized biological material
`by extraction into heptane into diluted hydrochloric acid and assayed fl uorometrically
`(Quinn 1959). Chromatographic analysis of TBZ reveals peak fl uorescence at 282 nm
`
`Neuropsychiatric Disease and Treatment 2007:3(5) 545–551
`© 2007 Dove Medical Press Limited. All rights reserved
`
`545
`
`Apotex Ex. 1017
`
`Apotex v. Auspex
`IPR2021-01507
`
`

`

`Diana
`
`(Roberts MS 1981). Based on the acid-basic transition, TBZ
`is characterized by a pKa of 6.0 (Scherman 1982).
`Pharmacodynamics
`and pharmacokinetics
`TBZ is readily absorbed from the intestinal tract. After intra-
`venous administration of TBZ tagged with tritium in humans,
`54% was excreted in urine after 48 hours (Stumpf 1960).
`TBZ enters the rabbit brain rapidly after intravenous
`administration attaining maximal levels of about 34
`micrograms per gram within 10 minutes. In other tissues
`maximal concentration is seen within 15–30 minutes, latest
`in fat tissue (with concentrations in fat tissue coming last).
`The half life in various tissues (when given intravenously
`to rabbits) ranges from 0.9–2.7 hours, with the longest
`half-life being in fat tissue. After 24 hours, TBZ will have
`disappeared from most tissues, brain tissue included (Quinn
`1959). TBZ has a relatively low bioavailability, 0.049
`± 0.032 (Roberts 1986).
`TBZ is metabolized into two compounds: α- and
`β-dihydrotetrabenazine (DTBZ); α-DTBZ being the
`active compound, whereas β-DTBZ is biochemically inert.
`α- DTBZ has a high bioavailability because it is less protein-
`bound (44%–59%) compared to TBZ (83%–88%) (Roberts
`1986). α-DTBZ has higher plasma levels than TBZ and
`the half-life is longer, 10 hours versus 6 hours (in humans)
`(Roberts 1986). Therefore it has to be administered two to
`three times a day.
`Acute toxicity in mice LD 50 is found to be 150 mg/kg
`by intravenous injection, 400 mg/kg by subcutaneous
`injection and about 550 mg/kg by oral administration,
`being about ten times less toxic than reserpine, another
`cathecolamine depletor). Toxic doses produce spasms with
`respiratory inhibition and opisthotonus. Chronic toxicity
`observed in rats given a daily dose of 8–15 mg/kg daily
`
`in their food for 13 weeks was well tolerated and growth
`was little infl uenced and neither blood nor organ exami-
`nations showed any abnormality (Lingierde 1963). TBZ
`crosses the placenta but no case of teratogenicity has been
`reported in humans. It is also excreted in breast milk and
`therefore breastfeeding should be avoided while taking
`TBZ (Roberts 1986).
`
`Mechanism of action
`The mechanism of action of TBZ is well known (Pettibone
`et al 1984). It acts mainly as a reversible high affi nity inhibitor
`of monoamine uptake into granular vesicles of presynaptic
`neurons and secondary depletion at low doses, as well as a
`weak D2 postsynaptic receptor blocker in high doses (Reches
`1983) TBZ depletes all three monoamines, but particularly
`dopamine (Pletscher et al 1962). One in vivo study of rats
`showed that TBZ decreased dopamine levels by 40%, sero-
`tonin by 44%, and norepinephrine by 41% in the brain (Lane
`et al 1976).
`The effect of a seven-day repetitive administration of
`TBZ in rats was investigated by examining the locomotive
`behavior and histomorphological fi ndings of substantia nigra
`in rats. These studies compared the effect of a single dose of
`TBZ which caused decrease of voluntary movements and no
`histological changes versus repetitive administration of TBZ
`which demonstrated irreversible and signifi cant changes
`in spontaneous locomotion as well as histological changes
`in the neurons of the substantia nigra pars compacta. The
`results suggest that this could be a new and useful model
`for the behavioral characteristics and anatomical pathol-
`ogy of Parkinson’s disease as one of the oxidative stress
`models induced by abnormal dopamine metabolism (Satou
`et al 2001).
`In an autopsy study of Huntington’s disease (HD)
`patients, those patients who had received TBZ displayed a
`
`CH3O
`
`CH3O
`
`N
`
`CH2CH(CH3)2
`
`O
`Figure 1 Tetrabenazine, a benzoquinolizine derivative with the chemical name, 2-oxo-3-isobutyl-9,10-dimethoxy-1,2,3,4,6,7-hexahydrobenzo[a]quinolizine.
`
`546
`
`Neuropsychiatric Disease and Treatment 2007:3(5)
`
`Apotex Ex. 1017
`
`

`

`greater overall depletion of monoamines than patients not
`exposed to TBZ, primarily in the caudate, but also, to a lesser
`degree, in the amygdala, hippocampus, and temporal lobe
`(Pearson and Reynolds 1988). PET studies w/11-C-raclopride
`show a 28% reduction in striatal binding after TBZ.
`α- DTBZ has been shown to inhibit monoamine uptake
`driven by a transmembrane proton elecrochemical gradi-
`ent generated by an ATP-ase proton pump using a special
`transporter (VMAT). In rat fi broblasts the central neural
`transporter VMAT2 is inhibited by TBZ, but the periph-
`eral endocrine specifi c VMAT1 is not (Masuo et al 1990;
`Erickson et al 1996). VMAT2 is a large protein with 12
`transmembrane helices encoded by the VMAT2 gene
`localized to chromosome 10q25. VMAT2 is expressed
`primarily in the brain. VMAT1 is encoded by a gene on
`chromosome 8p21.3 and is expressed in the periphery
`(Gonzalez et al 1994).
`The dual effects of TBZ are thought to be responsible both
`for its therapeutic effects as well as its side effects.
`Tetrabenazine versus reserpine
`The pharmacologic agent most similar to TBZ is reserpine
`(R). Both TBZ and R seem to reduce cells’ capacity to store
`monoamines, therefore causing a depletion of the brain stor-
`age of these amines and in the case of Reserpine a depletion
`of the amines in the peripheral sites. Both drugs act centrally
`on VMAT2, but reserpine also inhibits VMAT1 peripher-
`ally, which may explain the higher frequency of hypotension
`and gastrointestinal side effects it causes. While reserpine
`binds irreversibly to both VMAT’s, TBZ binds reversibly
`only to VMAT2. TBZ displays a much shorter half life than
`reserpine (hours as opposed to days) and has a more rapid
`onset of action. This confers an advantage on TBZ in the
`clinical setting as its effi cacy can be assessed more rapidly
`and its side effects abate more rapidly upon discontinuation
`of drug usage. Differences summarized in Table 1.
`
`Table 1 Difference between tetrabenazine and reserpine
`
`Tetrabenazine
`Reserpine
`Monoamine
`Central through
`Central and peripheral
`depletion
`VMAT2
`through VMAT1 and
`
`
`VMAT2
`Binding
`Reversible
`Non-reversible
`Post-synaptic
`YES
`NO
`effects
`(weak D2 blocker
`
`at high dose)
`T1/2
`10 hrs
`Side effects
`NO
`
`
`
`
`
`Several days
`YES
`(gastrointestinal and
`hypotension)
`
`Tetrabenazine in Huntington’s disease
`
`Huntington’s disease
`HD is a genetic neurological disease, which manifests a triad
`of psychiatric, cognitive and movement disorders. HD is
`inherited in an autosomal dominant fashion with complete
`penetrance. The prevalence rate in the United States has been
`estimated at 5–10 per 100,000 (Jancovic et al 1995). Genetic
`studies of families with a very high prevalence and incidence
`of HD from the Lake Maracaibo area in Venezuela led to the
`discovery of an unstable trinucleotide (CAG) repeat present
`in the gene on chromosome 4. The number of CAG repeats in
`normal subjects is up to 29 repeats, while the presence of 36
`or more CAG repeats ensures the development of HD (The
`Huntington’s Disease Collaborative Research Group 1993).
`The phenomena of anticipation and paternal imprinting have
`been well described in HD. The disease often presents with
`psychiatric problems or one type of hyperkinetic move-
`ment disorder, usually chorea. HD patients slowly progress
`over 15–20 years to a bedridden state. In advanced stages
`functional disability develops with dysphagia, dysarthria,
`prominent chorea with motor impersistence and in advanced
`stages a hypokinetic rigid state accompanied by dementia,
`depression and psychosis. HD represents the neurological
`disease with the highest rates of depression and suicide.
`Unfortunately there is no treatment that can cure or slow
`the course of the disease. Only modest symptomatic treatment
`options exist for those suffering from HD, mostly focused
`on ameliorating depression, psychosis, and chorea. Many
`therapeutical modalities, most of them including dopamine
`receptor blockers or dopamine depletors have been evalu-
`ated over the years, most of them in open label studies or
`presented as case reports. Few randomized double-blind
`studies with TBZ have been done in HD. (Mc Lellan et al
`1974; Sajatovic et al 1991; Van Vugt et al 1997; Huntington
`Study Group 2006).
`Huntington’s disease
`and tetrabenazine treatment
`One of the main reasons for using TBZ instead of dopamine
`receptor blockers is its relative safety, exemplifi ed by the
`fact that TBZ has never been documented as having caused
`tardive dyskinesia (TD). This is a major advantage of TBZ
`over the typical neuroleptics, since between 25%–40% of
`those chronically treated with DRBD eventually develop
`TD (Smith and Baldessarini 1980).
`The fi rst reports of the therapeutic potential of TBZ in
`patients with HD were published in the 1960’s (Brandrup
`1960; Sattes 1960; Sattes and Hase 1964). Since the
`1970’s, numerous other clinical trials with relatively
`
`Neuropsychiatric Disease and Treatment 2007:3(5)
`
`547
`
`Apotex Ex. 1017
`
`

`

`Diana
`
`small numbers of patients have demonstrated the ben-
`efi cial effects of TBZ for patients with chorea (Fog and
`Pakkenberg 1970; Gilligan et al 1972; Swash et al 1972;
`Huang 1976; Toglia et al 1978; Kingston 1979; Asher and
`Aminoff 1979; Jancovic 1982, 1983; Jancovic and Orman
`1988; Gimenez-Roldan and Mateo 1989; Jancovic and
`Beach 1997; Ondo et al 2002; Paleacu et al 2004; Kenney
`and Jancovic 2006).
`TBZ was approved for the treatment of chorea in the
`United Kingdom in 1971. It is also available in Canada and
`Australia, as well as in several European countries. TBZ
`is still unavailable in the United States, though it has been
`obtained by selected physicians via the Notice of Claimed
`Investigational Exemption for a New Drug (IND). However,
`in spite of its low availability, several long-term studies
`of TBZ in movement disorders, including HD, have been
`reported from the US.
`There have been fi ve major studies, which assessed the
`long-term effi cacy of TBZ in chorea patients, most of them
`with HD. The most recent, a phase-III study assessing the
`safety, effi cacy, and dose-tolerability of TBZ for amelio-
`rating chorea in 84 patients with HD, was published by
`the Huntington Study Group (HSG) in 2006. HD patients
`were randomized to placebo (n = 30) or TBZ (n = 54) up to
`100 mg/day at a titration rate of 12.5 mg per week. Based
`on the chorea score of the Unifi ed Huntington Disease
`Rating Scale (UHDRS), TBZ signifi cantly reduced the
`chorea score by 5.0 points compared to 1.5 for placebo
`(p < 0.0001). Likewise, the CGIC showed signifi cantly
`higher improvement rates in patients treated with TBZ
`in comparison to those given placebo. There were fi ve
`withdrawals in the TBZ treatment group and fi ve serious
`adverse events (SAE) including one suicide, which was felt
`to be due to situational depression rather than TBZ-induced
`depression. There were no SAE in the placebo group.
`A retrospective chart analysis of 76 hyperkinetic patients
`in a pediatric population treated at Baylor College of Medi-
`cine from 1996–2004 revealed signifi cant improvement in
`chorea in 89% of the patients (Vuong et al 2004).
`In a study comprising 118 patients with hyperkinetic
`movement disorders (28 diagnosed with HD), patients
`were assessed by phone interview using the Clinical Global
`Impression of Change (CGIC) and the overall effi cacy of
`treatment was shown via a composite score made up of the
`patient’s score and the caregiver’s score. Signifi cant improve-
`ment of hyperkinesias was seen in 61% of patients, with
`a subgroup of subjects with chorea and facial dyskinesias
`responding most favorably (Paleacu et al 2004).
`
`In another small study of 19 HD patients treated with
`TBZ, the follow-up was carried out in a prospective fashion.
`The evaluation was done using the Abnormal Involuntary
`Movement Scale (AIMS), which was done by two separate
`investigators blinded to the drug administered. Eighteen
`patients completed and these patients were rated at 3.3
`months at a fi nal mean dose of 62.5 ± 37.4 mg/day. Fifteen
`patients scored better than before treatment, the scores of one
`remained the same and the scores of two others worsened
`(Ondo et al 2002).
`The fi rst published study assessing the long-term effi cacy
`of TBZ in 400 hyperkinetic movement disorders patients
`selected from a large group of 526 included 29 HD patients.
`These patients improved by 82.8% on a scale of 1 to 5 (where
`1 = improvement, 4 = no response and 5 = worsening). The
`average treatment duration was 28 ± 31.1 months (Jancovic
`and Beach 1997).
`Kingston summarized the experience of 40 patients
`who had been receiving TBZ for almost 7 years for sev-
`eral movement disorders including chorea. 75% of these
`patients experienced marked or moderate improvement
`(Kingstone 1979).
`A single-blind crossover study with a pretreatment phase,
`active drug, followed by placebo in 26 patients with HD,
`TD, and dystonia found that 54% experienced marked or
`moderate improvement of chorea with TBZ for the 3-week
`duration of the study (Asher and Aminoff 1979). Many
`studies that included a mixed group of movement disorders
`mention that the patients with chorea and TD responded
`better than patients with dystonia or tics.
`A double-blind prospective crossover study of 20 patients
`assessed TBZ versus placebo for its effect on a variety of
`hyperkinetic movement disorders (Jancovic 1982). TBZ was
`found to improve the hyperkinesia score more than placebo
`to a statistically signifi cant degree. Patients noted func-
`tional improvement, but this endpoint was not assessed in a
`quantitative manner. An open-label follow up of these same
`patients found that 62% of patients initially enrolled in the
`double-blind crossover study continued to display moderate
`improvement of the movement disorder 6–18 months later
`(Jancovic 1983). Major studies reporting on TBZ effi cacy
`on chorea are summarized in Table 2.
`
`Dosing issues
`TBZ is usually initiated at a dose of 12.5 mg twice a day
`and is slowly titrated in two or three divided doses up to
`150–200 mg/day in increments of 25 mg/week. In studies
`and in daily practice, given the short half-life, dosing TID is
`
`548
`
`Neuropsychiatric Disease and Treatment 2007:3(5)
`
`Apotex Ex. 1017
`
`

`

`Tetrabenazine in Huntington’s disease
`
`5 point reduction in HD
`compared to1.5 in placebo
`patients
`(p < 0.0001)
`89% of the patients improved
`
`Table 2 Major studies in which chorea, including HD, patients were treated with TBZ showing treatment outcome
`Authors and year
`Number of
`Outcome measures
`Outcome
`of publication
`patients
`
`Huntington study
`84 HD patients
`Reduction in chorea
`group 2006
`
`score of the UHDRS
`
`
`
`
`
`
`Vuong et al 2004
`76 pediatric
`Chorea improvement by
`
`patients
`CGIC
`Paleacu et al 2004
`118 patients with
`CGIC
`
`hyperkinetic
`
`
`movement
`
`
`disorders, 28 HD
`
`Ondo et al 2002
`19 HD patients
`AIMS
`
`
`
`Jancovic and
`400 hyperkinetic
`Modifi ed CGIC
`Beach 1997
`patients, 29 HD
`(from 1–5)
`
`
`
`Jancovic 1982
`20 hyperkinetic
`Functional
`
`patients
`
`Asher and
`26 patients with
`Clinical
`Aminoff 1979
`chorea, TD, tics
`
`Kingstone 1979
`40 hyperkinetic
`Clinical
`
`patients
`
`
`61% of patients improved
`(chorea improved in19/28
`patients)
`
`79% of patients improved
`(15/19)
`Overall improved by 82.8%
`97% of chorea patients
`improved (28/29 )
`62% of patients improved
`
`54% of chorea patients
`improved
`75% marked to moderate
`improvement
`
`Abbreviation: UHDRS, unifi ed Huntington disease rating scale; CGIC, clinical global impression of change; AIMS, abnormal involuntary movement scale
`
`sometimes necessary. The dose escalation is stopped when
`the patient experiences a clear therapeutic effect or intoler-
`able side effects. This technique might indeed create a bias
`towards an increase in side effects in many clinical studies on
`TBZ as most studies are designed to increase the dose until
`intolerable side effects are noted and then the dose is slightly
`decreased to alleviate the side effects. Most drug-related side
`effects can be alleviated by lowering the dose. Overdose in
`a case of self-poisoning with tetrabenazine was described
`in a 27-year-old female without any signifi cant sequelae,
`except for sedation, after taking approximately 1 gram of
`TBZ (Kidd et al 1972).
`
`Tolerability and side effects
`The most common immediate side effects include drowsi-
`ness/sedation, weakness, parkinsonism, depression and
`acute akathisia, all of which are reversible with decreased
`dosing (Jancovic 1997; Paleacu 2004). Several studies have
`observed that younger patients tolerate TBZ better than the
`elderly. It is also notable that side effects vary slightly across
`different age groups: while younger patients showed a trend
`to experience more insomnia and depression, older patients
`seemed more likely to develop parkinsonism (Hunter 2002;
`Paleacu 2004). Other rare side effects include: insomnia,
`nervousness/anxiety, nausea and vomiting, tremor, memory
`
`problems, confusion, “trance-like/zombie”, orthostatic hypo-
`tension, balance and gait diffi culties, dizziness, diarrhea,
`headaches, hallucinations, paresthesias, pharyngeal spasm
`and pain, blurred vision, panic attacks, paranoia (Jancovic
`and Beach 1997).
`Changes in clinico-chemical tests during 12 months
`on TBZ were always minor, nearly always unsystematic
`and, on the whole, tended more towards a normalization of
`values. Even taking into account the limitations inherent in
`an uncontrolled trial, the conclusion was reached that long-
`term treatment with TBZ seems to be quite safe (Jancovic
`and Beach 1997). The extrapyramidal side effects of TBZ
`include parkinsonism, acute dystonia and akathisia and rarely
`neuroleptic malignant syndrome (NMS). It is particularly
`notable than not one single case of tardive dyskinesia has
`been reported with TBZ; it is this fact which confers its great
`advantage over dopamine receptor blockers. Concurrent use
`of antipsychotics with tetrabenazine can induce parkinsonism
`in HD patients (Moss and Stewart 1986) or acute dystonic
`reactions (Schott et al 1989). In one of the NMS cases,
`factors potentiating NMS included a high dosage of tetra-
`benazine exceeding the accepted therapeutic range together
`with co-medication with the dopamine-synthesis inhibitor
`alpha-methylparatyrosine, while in an other case, abrupt
`introduction of the drug and discontinuation of concomitant
`
`Neuropsychiatric Disease and Treatment 2007:3(5)
`
`549
`
`Apotex Ex. 1017
`
`

`

`Diana
`
`neuroleptics may have contributed to this adverse reaction.
`Uneventful recovery occurred in both cases without the need
`for drugs specifi cally enhancing dopaminergic transmission,
`while rechallenge by tetrabenazine with conventional doses
`and slow upward titration was not followed by recurrence of
`the NMS (Burke et al 1981; Mateo et al 1992; Osseman et
`al 1996). Other serious adverse events included pneumonia
`(Shoulson 1981) severe dysphagia (Jancovic and Beach
`1997), and suicide (Gimenez-Roldan 1989). While it is well
`known that depression is extremely prevalent in HD and that
`suicide rates for this population are among the highest of
`those for all neurological diseases, TBZ, while capable of
`potentiating depression, did not seem to be directly involved
`in the suicide cases or in any other fatalities which occurred
`during treatment.
`
`Conclusion
`TBZ, a dopamine-depleting drug with a unique pharmaco-
`logic mechanism, has been found to be safe and effi cacious
`for the treatment of a variety of hyperkinetic movement
`disorders, including HD. In the absence of modern disease
`modifying treatments for HD, symptomatic drug treatments
`such as TBZ remain important. These drugs, ideally, should
`be effi cacious and devoid of debilitating side-effects.
`Though most published studies are not double-blind,
`placebo-controlled trials, the sustained long-term benefi t of
`patients speaks in favor of TBZ’s clinical effi cacy in HD.
`Furthermore, TBZ does not cause TD, which is its major
`advantage over dopamine receptor blockers. It also has gener-
`ally well tolerated side effects that are usually dose-related
`and therefore resolvable by decreasing doses or employing
`slow titration. Potentially fatal side effects such as dysphagia
`and depression are rare, though their existence mandates
`close monitoring of patients.
`Based on his observations and previous open studies
`demonstrating TBZ efficacy in chorea Mc Lellan may
`have rightfully affi rmed that “tetrabenazine is the drug of
`fi rst choice for the suppression of chorea in patients with
`Huntington’s disease” (McLellan 1974).
`
`References
`
`Ashcroft GW, Macdougall EJ, Barker PA. 1961. A comparison of tetra-
`benazine and chlorpromazine in chronic schizophrenia. J Ment Sci,
`107:287–93.
`Asher SW, Aminoff MJ. 1981. Tetrabenazine and movement disorders.
`Neurology, 31:1051–4.
`Bartels M, Zeller E. 1984. Tetrabenazine (Nitoman) therapy of chronic
`spontaneous oral dyskinesia. A video- and EMG-controlled study. Eur
`Arch Psychiatry Neurol Sci, 234:172–4.
`Boghen DR, Lesser RL. 2000. Blepharospasm and hemifacial spasm. Curr
`Treat Options Neurol, 2:393–400.
`
`Brandrup E. 1960. Reserpin og tetrabenacin ved chorea Huntington. Nordisk
`Medicin, 4:968–9.
`Burke RE, Fahn S, Mayeux R, et al. 1981. Neuroleptic malignant syndrome
`caused by dopamine-depleting drugs in a patient with Huntington
`disease Neurology, 31:1022–5.
`Duran E, Chacon JR. 2001. Spasmodic torticollis and vertebral hemangioma.
`Rev Neurol, 32:60–2.
`Erickson JD, Schafer MK, Bonner TI, et al. 1996. Distinct pharmacological
`properties and distribution in neurons and endocrine cells of two iso-
`forms of the human vesicular monoamine transporter. Proc Natl Acad
`Sci USA, 93:5166–71.
`Fahn S. 1985. A therapeutic approach to tardive dyskinesia. J Clin Psy-
`chiatry, 46:19–24.
`Faulstich ME, Carnrike CL Jr, Williamson DA. 1985. Blepharospasm and
`Meige syndrome: a review of diagnostic, aetiological and treatment
`approaches. Psychosom Res, 29:89–94.
`Fog R, Pakkenberg H. 1970. Combined nitoman-pimozide treatment of
`Huntington’s chorea and other hyperkinetic syndromes. Acta Neurol
`Scand, 46:249–51.
`Gilligan BS, Wodak J, Veale 1972. Tetrabenazine in the treatment of
`extrapyramidal dyskinesias. Med J Aust, 2:1054–6.
`Gimenez-Roldan S, Mateo D. 1989. Huntington disease: tetrabenazine
`compared to haloperidol in the reduction of involuntary movements
`Neurologia, 4:282–7.
`Gonzalez AM, Walther D, Pazos A, et al. 1994. Synaptic vesicular mono-
`amine transporter expression: distribution and pharmacologic profi le.
`Mol Brain Res, 22:219–26.
`Huang CY, McLeod JG, Holland et al. 1976. Tetrabenazine in the treatment
`of Huntington’s chorea. Med J Aust, 1:583–4.
`Hunter C, Wang A, Vuong K. 2002. Age-related tolerability of tetrabenazine.
`Mov Disord, 17(Suppl 5):S41.
`Huntington Study Group. 2006. Tetrabenazine as an antichorea therapy
`in Huntington’s disease: a random controlled trial Neurology,
`66:366–72.
`Jankovic J. 1982. Treatment of hyperkinetic movement disorders with tetra-
`benazine: a double-blind crossover study. Ann Neurol, 11:41–7.
`Jankovic J. 1983. Tetrabenazine in the treatment of hyperkinetic movement
`disorders. Adv Neurol, 37:277–89.
`Jankovic J, Beach J. 1997. Long-term effects of tetrabenazine in hyperkinetic
`movement disorders. Neurology, 48:358–62.
`Jankovic J, Beach J, Ashizawa T. 1995. Emotional and functional impact
`of DNA testing on patients with symptoms of Huntington’s disease. J
`Med Genet, 32:516–18.
`Jankovic J, Glaze DG, Frost JD Jr. 1984. Effect of tetrabenazine on
`tics and sleep of Gilles de la Tourette’s syndrome Neurology,
`34:688–92.
`Jankovic J, Orman J. 1988. Tetrabenazine therapy of dystonia, chorea, tics,
`and other dyskinesias. Neurology, 38:391–4.
`Jankovic J, Rohaidy H. 1987. Motor, behavioral and pharmacologic fi ndings
`in Tourette’s syndrome. Can J Neurol Sci, 14(3 Suppl):541–6.
`Jimenez-Jimenez FJ, Garcia-Ruiz PJ. 2001. Pharmacological options for
`the treatment of Tourette’s disorder. Drugs, 61:2207–20.
`Kanjilal GC, Matheson B. 1962. A trial of tetrabenazine (Nituman) in dis-
`turbed mentally subnormal patients. J Ment Sci, 108:225–8.
`Kenney C, Jankovic J. 2006. Tetrabenazine in the treatment of hyperkinetic
`movement disorders. Expert Rev Neurother, 6:7–17.
`Kidd DW, McLellan DL. 1972. Self-poisoning with tetrabenazine. Br J
`Clin Pract, 26:179–80.
`Kingston D. 1979. Tetrabenazine for involuntary movement disorders.
`Med J Aus,1:628–30.
`Lane JD, Smith JE, Shea PA. 1976. Neurochemical changes following
`the administration of depleters of biogenic monoamines. Life Sci,
`19:1663–7.
`Lingjerde O. 1963. Tetrabenazine (Nitoman) in the treatment of psychoses.
`Acta Psychiatrica Scand, Suppl 170:158–209.
`Manji H, Howard RS, Miller DH, et al. 1998. Status dystonicus: the syn-
`drome and its management. Brain, 121:243–52.
`
`550
`
`Neuropsychiatric Disease and Treatment 2007:3(5)
`
`Apotex Ex. 1017
`
`

`

`Masuo Y, Pélaprat D, Scherman D, et al. 1990. [3H]Dihydrotetrabenazine,
`a new marker for the visualization of dopaminergic denervation in the
`rat striatum Neurosci Lett, 114:45–50.
`Mateo D, Munoz-Blanco JL, Gimenez-Rodan S. 1992. Neuroleptic
`malignant syndrome related to tetrabenazine introduction and halo-
`peridol discontinuation in Huntington’s disease. Clin Neuropharmacol,
`15:63–8.
`Mc Lellan DL, Chalmers RJ, Johnson RH. 1974. A double blind trial of
`tetrabenazine, thiopropazate and placebo in patients with chorea, The
`Lancet, 26:104–7.
`Mikkelsen BO. 1983. Tolerance of tetrabenazine during long-term treatment.
`Acta Neurol Scand, 68:57–60.
`Moss JH, Stewart DE. 1986. Iatrogenic parkinsonism in Huntington’s chorea.
`Can J Psychiatry, 31:865–6.
`Ondo WG, Hanna PA, Jankovic J. 1999. Tetrabenazine treatment for tar-
`dive dyskinesia: assessment by randomized videotape protocol. Am. J
`Psychiatry, 156:1279–81.
`Ondo WG, Tintner R, Thomas M, et al. 2002. Tetrabenazine Treatment
`for Huntington’s Disease-Associated Chorea. Clin Neuropharmacol,
`25:300–2.
`Ossemann M, Sindic CJ, Laterre C. 1996.Tetrabenazine as a cause of neu-
`roleptic malignant syndrome. Mov Disord, 11:95.
`Paleacu D, Giladi N, Moore O, et al. 2004. Tetrabenazine treatment in
`movement disorders. Clin Neuropharmacol, 27:230–3.
`Pearson SJ, Reynolds G P. 1988. Depletion of monoamine transmitters by
`tetrabenazine in brain tissue in Huntington’s disease. Neuropharma-
`cology, 27:717–19.
`Pettibone DJ, Totaro JA, Pfl ueger AB. 1984. Tetrabenazine-induced deple-
`tion of brain monoamines: characterization and interaction with selected
`antidepressants. Eur J Pharmacol, 20:425–30.
`Pletscher A, Brossi A, Gey K. 1962. Benzoquinolizine derivatives: a new
`class of monoamine decreasing drugs with psychotropic action. Inter-
`national Review of Neurobiology, 4:275–306.
`Quinn GP, Shore PA, Brodie BB. 1959. Biochemical and pharmacological
`studies of RO 1-9569 (tetrabenazine), a nonindole tranquilizing agent
`with reserpine-like effects. J Pharmacol Exp Ther, 127:103–9.
`Raja M. 1998. Managing antipsychotic-induced acute and tardive dystonia.
`Drug Saf, 9:57–72.
`Reches A, et al. 1983. Tetrabenazine, an amine-depleting drug, also blocks
`dopamine receptors in rat brain. J Pharmacol Exp Ther, 225:515–21.
`Roberts MS, McLean S, Millingen KS, et al. 1986. The pharmacokinetics of
`tetrabenazine and its hydroxy metabolite in patients treated for involun-
`tary movement disorders. Eur J Clin Pharmacol, 29:703–8.
`Roberts MS, Watson HM, Mc Lean, et al. 1981. Determination of therapeutic
`plasma concentrations of tetrabenazine and an active metabolite by high-
`performance liquid chromatography. J Chromatogr, 226:175–82.
`
`Tetrabenazine in Huntington’s disease
`
`Sajatovic M, Verbanac P, Ramirez F, et al. 1991. Clozapine treatment of
`psychiatric syndromes resistant to neuroleptic treatment in patients with
`Huntington’s disease. Neurology, 41:156–8.
`Satou T, Anderson AJ, Itoh T, et al. 2001. Repetitive administration of
`tetrabenazine induces irreversible changes in locomotion and morphol-
`ogy of the substantia nigra in rats. Exp Toxicol Pathol, 53:303–8.
`Sattes H. 1960. The treatment of chorea minor with the monoamine liberator
`“Nitoman.” Psychiatr Neurol Neurochirurg (Basel), 140:13.
`Sattes H, Hase E, 1964. Die Behandherd Extrapyramidaler Hypokinesien
`unto besonderer Beriicksichtigung der chorea Huntington. Psychiatr
`Neurol Neurochirurg (Aust), 67:289.
`Scahill L, Chappell PB, King RA, et al. 2000. Pharmacologic treatment of
`tic disorders. Child Adolesc Psychiatr Clin N Am, 9:99–117.
`Scherman D, Henry JP. 1982. Acido-basic properties of the catecholamine
`uptake inhibitors tetrabenazine and dihydrotetrabenazine. Biochimie,
`64:915–21.
`Schott K, Ried S, Stevens I, et al. 1989. Neuroleptically induced dystonia
`in Huntington’s disease: a case report. Eur Neurol, 29:39–40.
`Scott BL. 2000. Evaluation and treatment of dystonia. South Med J,
`93:746–51.
`Shoulson I, Goldblatt D. 1981.Huntington’s disease (HD): effect of
`tetrabenazine and antipsychotic drugs on motoric features. Neurology,
`31:79.
`Simpson GM. 2000. The treatment of tardive dyskinesia and tardive dys-
`tonia. J Clin Psychiatry, 61(Suppl 4):39–44.
`Smi

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket