347
Views
68
CrossRef citations to date
0
Altmetric
Review

Tetrabenazine: the first approved drug for the treatment of chorea in US patients with Huntington disease

Pages 657-665 | Published online: 28 Sep 2010

Abstract

Huntington disease (HD) is a dominantly inherited progressive neurological disease characterized by chorea, an involuntary brief movement that tends to flow between body regions. HD is typically diagnosed based on clinical findings in the setting of a family history and may be confirmed with genetic testing. Predictive testing is available to those at risk, but only experienced clinicians should perform the counseling and testing. Multiple areas of the brain degenerate mainly involving the neurotransmitters dopamine, glutamate, and γ-aminobutyric acid. Although pharmacotherapies theoretically target these neurotransmitters, few well-conducted trials for symptomatic or neuroprotective interventions yielded positive results. Tetrabenazine (TBZ) is a dopamine-depleting agent that may be one of the more effective agents for reducing chorea, although it has a risk of potentially serious adverse effects. Some newer antipsychotic agents, such as olanzapine and aripiprazole, may have adequate efficacy with a more favorable adverse-effect profile than older antipsychotic agents for treating chorea and psychosis. This review will address the epidemiology and diagnosis of HD as background for understanding potential pharmacological treatment options. Because TBZ is the only US Food and Drug Administration-approved medication in the United States for HD, the focus of this review will be on its pharmacology, efficacy, safety, and practical uses. There are no current treatments to change the course of HD, but education and symptomatic therapies can be effective tools for clinicians to use with patients and families affected by HD.

Introduction

Huntington disease (HD) is a hereditary, progressive neurodegenerative disease clinically characterized by abnormal involuntary movements, behavioral disturbance, cognitive dysfunction, and psychiatric disease. The disease is caused by a CAG (glutamine) trinucleotide expansion in exon 1 of the huntingtin (htt) gene at the location 4p16.9.Citation1 The function of htt is not known, but it may be involved in internal cell signaling, maintenance of cyclic adenosine monophosphate response element binding protein, and preventing neuronal toxicity.Citation2 Early evidences suggest that the binding of the Ras homologue enriched in striatum (Rhes) protein to htt may be necessary to cause cellular toxicity.Citation3 However, why the protein causes cellular toxicity in adulthood is not well understood. Recent evidence suggests that the interaction of the group 1 metabotropic glutamate receptors and htt protein may be at the root of delayed onset.Citation4

Although there is no established treatment to delay the onset or forestall the progression of HD, symptomatic treatment of chorea may be beneficial in some individuals, as it may have a favorable effect on motor function, quality of life, and safety.Citation5Citation7

Pathologically, HD is associated with diffuse loss of neurons, particularly involving the cortex and the striatum. Medium spiny neurons in the striatum that contain γ-aminobutyric acid (GABA) and enkephalin are impacted early in the disease, and are the primary neurons targeted in HD. These neurons typically project into the lateral globus pallidus. Then, there is progression to the remainder of the basal ganglia with subsequent dissemination, including cortex and substantia nigra. There are intranuclear and cytoplasmic inclusions of the htt aggregate. Huntingtin is cross-linked with other soluble htt to form the inclusion bodies in neurons. It is not known if the accumulation of htt conglomerate results in cell death, or if the soluble form of the protein is the toxic form.Citation8,Citation9 Dopamine, glutamate, and GABA are thought to be the most involved neurotransmitters in HD and are targeted for treatment ().Citation10Citation23

Table 1 Neurotransmitters involved in the pathogenesis of Huntington disease

There are multiple theories on the pathogenesis of HD. It is likely that more than 1 process may be occurring at once, but there is evidence to support multiple individual mechanisms, including toxic neuronal aggregates, transcriptional dysregulation, excitotoxicity, mitochondrial dysfunction with altered energy metabolism, and changes in axonal transport and synaptic dysfunction ().Citation24Citation30,Citation31

Table 2 Potential pathways for pathogenesis of Huntington disease

Epidemiology

Most European populations show a prevalence rate of 4–8 cases per 100,000, and HD may be frequently seen in India and parts of Central Asia.Citation32 More recent studies in other European nations have confirmed this prevalence rate.Citation33,Citation34 HD is notably rare in Finland and Japan, but data for Eastern Asia and Africa are inadequate. Outside of Venezuela, there are little epidemiological studies of HD in Hispanic populations. A recent study found a slightly higher prevalence rate than expected and higher proportion of juvenile cases in Mexico.Citation35 There are also well-known large populations of patients with HD in Scotland and the Lake Maracaibo region of Venezuela.Citation36,Citation37 The disorder may be underestimated in the Black American population. There have been no widespread epidemiological studies of HD in the United States since the wide availability of genetic testing, but it is estimated that approximately 25,000–30,000 individuals have manifest HD and a further 150,000–250,000 individuals are at risk for HD.Citation38

Men and women are affected equally and typically become symptomatic in the third and fourth decades. The symptoms of HD can start at any age ranging from 1 to 90 years. Approximately 5%–10% of cases are classified as juvenile onset, with symptoms starting before the age of 20 years. The vast majority of juvenile cases are inherited paternally, and instead of chorea, patients may exhibit more parkinsonian signs of bradykinesia, dystonia, tremors, and cognitive deficit.Citation39 When patients exhibit more hypokinetic features (bradykinesia and dystonia) vs hyperkinetic features (chorea), they are said to have the Westphal variant of HD.

Diagnosis

HD is diagnosed based on the presence of typical motor findings as measured by the Unified Huntington’s Disease Rating Scale in the setting of a family history of the disease. There may be other manifestations of HD at the time of presentation or prior to diagnosis based on behavioral and cognitive symptoms. A DNA test showing abnormal CAG expansion in the htt gene can be used to confirm the diagnosis in symptomatic individuals. With proper genetic counseling and at the patient’s request, DNA analysis can be performed in individuals at risk for developing HD under the care of experienced clinicians. There are many reasons why patients request presymptomatic testing, including financial planning, family planning, insurance decisions, and the need to know. Some people at risk for HD are reluctant to undergo testing and are brought into the clinic by future spouse, family, or others who believe testing should be performed. Under these circumstances or if the at-risk individual is likely to harm themselves or others based on the outcome of the test, genetic testing should be reconsidered.

There are ongoing studies to examine individuals who are gene positive but not yet symptomatic by motor criteria (TRACK-HD, Neurobiological Predictors of Huntington’s Disease Trial [PREDICT-HD], Prospective Huntington at Risk Observational Study [PHAROS]).Citation40Citation43 There is also a study currently enrolling subjects with HD and their affected and unaffected family members to further understand biomarkers of disease and signs of onset (Cooperative Huntington’s Observational Research Trial [COHORT]).Citation44 The focus of the COHORT study is not neuroimaging and anatomical measures but rather clinical measures and biological samples.

Subtle motor abnormalities have been associated with a smaller striatal volume and higher probability of disease diagnosis.Citation40 Lower scores on the Hopkins Verbal Learning Test-Revised were associated with closer proximity to diagnosis and smaller striatal volumes.Citation44 Subjects with an expanded repeat and preclinical diagnosis of HD also had less accurate recognition of negative emotions.Citation45 In addition, motor exam score, striatal volume, speeded finger tapping, self-timed finger tapping, word-list learning, and odor identifications in subjects in the PREDICT-HD study were all associated with the predicted time to diagnosis. Citation46 Expansion-positive individuals reported more psychiatric symptoms (depression, anxiety, obsessive–compulsiveness) than expansion-negative individuals.Citation47 The TRACK-HD study has confirmed some and expanded upon findings in PREDICT-HD in that presymptomatic subjects had significant changes in whole-brain volume, regional grey and white matter differences, impairment in a range of motor tasks, oculomotor findings, and cognitive and neuropsychiatric dysfunctions. The various motor and nonmotor measures on the neurological examination used to diagnose and track HD are included in the Unified Huntington’s Disease Rating Scale.Citation48 The scale is divided into 6 sections: motor, cognitive, behavioral, and 3 functional scales (total functional capacity, functional checklist, and the independence scale).

Based on the guidelines published by the American College of Genetics, patients with 40 or more repeats have 100% penetrance.Citation49 In other words, if patients have 40 or more copies of the gene, they will inevitably express the disease clinically. In patients with a CAG repeat length in the range of 36–39, there is reduced penetrance with increased likelihood of expression occurring with longer length of repeats and with longer lifespan of the patient. Although there are case reports of patients who manifest HD in this range, patients with fewer than 36 repeats will generally not develop clinical disease.Citation50,Citation51 Patients with an allele repeat size of 27–35 have demonstrated meiotic instability, particularly in sperm, indicating that the following generation is at higher risk of inheriting an expanded number of repeats, increasing the risk of clinical disease. The length of repeat size correlates generally with the age of onset, but not necessarily with the severity or duration of disease.Citation52

The course of the disease is typically 15–20 years, with dementia, mutism, dystonia, and bradykinesia predominating in end-stages. Patients with more dystonia and swallowing issues may experience accelerated complications and, therefore, shorter lifespan. Chorea may become a safety issue with larger amplitude movements causing injury or poor positioning. Frequent movements may result in skin injuries, infections, or even fractures and head trauma. Cause of death is typically related to complications of immobility, such as skin breakdown, pneumonia, cardiac disease, or infection. However, 25% of patients attempt suicide, which is a cause of death in 8%–9% of patients.Citation53

Behavioral dyscontrol can be a severely disabling symptom of HD causing distress to the patient, family, and caregivers. Environmental approaches and cognitive interventions are the mainstay of treatments, but pharmacological agents can augment addressing disruptive behaviors. Depression, anxiety, aggressive, impulsive, and obsessive–compulsive behaviors are also frequently treated pharmacologically and require behavioral intervention, but caution should be used to avoid oversedation and apathy, already common in patients with HD. Although not well studied, cognitive approaches to treat behavior may be more effective than pharmacotherapy for some aspects of the disease.Citation54

Pharmacological treatment options

Many agents and surgical procedures have been evaluated in HD for their efficacy on suppressing chorea, including dopamine-depleting agents, dopamine antagonists, benzodiazepines, glutamate antagonists, acetylcholinesterase inhibitors, dopamine agonists, antiseizure medications, cannabinoids, lithium, deep brain stimulation, and fetal cell transplantation.Citation55Citation58 Pharmacological interventions typically address the hyperkinetic movement disorders that may be associated with HD, such as chorea, dystonia, ballism, myoclonus, and tics. With regards to choose the agent, providers need to consider if there will be a positive or negative impact of the agent on psychiatric issues associated with HD, such as irritability, depression, anxiety, mania, apathy, obsessive–compulsive disorder, or cognitive decline associated with HD. Adjunctive therapies, alternative and complementary therapies, behavioral plans, and cognitive interventions also may play a role in addressing the symptoms of HD and need to be considered when choosing medications.

Several excellent reviews have summarized the symptomatic treatment of HD.Citation55Citation67 Overall, there is not enough evidence available to guide long-term symptomatic treatment in HD, and double-blind and long-term studies assessing various treatment strategies in HD are needed.Citation61 A Cochrane review of studies for the symptomatic treatment of HD examined 22 trials that involved 1,254 different participants.Citation62 Nine trials had a crossover design and 13 were conducted in parallel. The studies examined were of relatively short duration, ranging from 2 to 80 weeks. The number of trials examining various pharmacological interventions included antidopaminergic drugs (n = 5), glutamate receptor antagonists (n = 5), and energy metabolites (n = 5). Based on available evidence, the authors of the Cochrane review concluded that only tetrabenazine (TBZ) showed clear efficacy for the control of chorea, but “no statement can be made regarding the best medical practice for the control of motor and non-motor symptoms in HD”.

Tetrabenazine

TBZ is the only US Food and Drug Administration (FDA)-approved drug for HD, indicated for the treatment of chorea associated with HD. In addition to the United States and Canada, TBZ is marketed in Australia, Denmark, France, Germany, Ireland, Israel, Italy, New Zealand, Portugal, Spain, Switzerland, and United Kingdom. An excellent review of the chemistry, pharmacodynamics, pharmacokinetics, and mechanism of action is available in a previous issue of this journal.Citation68 By reversibly inhibiting the central vesicular monoamine transporter type 2 (VMAT2), TBZ more selectively depletes dopamine than norepinephrine.Citation69,Citation70 The highest binding density for TBZ is in the caudate nucleus, putamen, and nucleus accumbens, areas known to bear the brunt of pathology in HD.Citation71,Citation72 VMAT2 binding and monoamine depletion by TBZ are reversible, last hours, and are not modified by long-term treatment.Citation73,Citation74 These features of the drug differentiate it from the other dopamine-depleting agent, reserpine. Reserpine binds to both VMAT1 and VMAT2. While VMAT2 is located solely in the central nervous system, VMAT1 is localized to the peripheral nervous system, accounting for some of the peripheral adverse effects, such as orthostatic hypotension and diarrhea. In contrast to TBZ, reserpine binds irreversibly to VMAT1 and VMAT2, making the duration of action considerably longer (hours vs days). In addition to TBZ, the 2 active metabolites, α- and β-dihydrotetrabenazine, have longer half-lives and are more highly bound to proteins than the parent compound.Citation75Citation77

The efficacy of TBZ as an antichoreic drug was convincingly demonstrated in a double-blind, placebo-controlled trial conducted by the Huntington Study Group (HSG).Citation78 Subjects were randomized to receive either TBZ (n = 54) or placebo (n = 30). TBZ was titrated weekly in 12.5 mg increments to a maximum of 100 mg/d or to the development of intolerable adverse effects. The primary efficacy outcome was the change from baseline in the total maximal chorea score of the Unified Huntington’s Disease Rating Scale.Citation48 On this scale, chorea is graded from 0 to 4 (with 0 representing no chorea) for 7 body regions for a range in total scores from 0 to 28. Compared with baseline, TBZ treatment resulted in a reduction of 5.0 units in chorea compared with a reduction of 1.5 units in the placebo group. About 50% of TBZ-treated subjects had a 6-point or greater improvement compared with 7% of placebo recipients. There is also evidence to suggest continuous long-term efficacy and tolerability of TBZ in patients with HD.Citation79Citation82

In the same study, the adverse events that occurred significantly more frequently in the TBZ group included drowsiness or somnolence, insomnia, depressed mood, agitation, akathisia, and hyperkinesia. However, by the conclusion of the maintenance phase, when subjects were presumably on optimal dosage, there were no significant differences between TBZ and placebo. Among subjects completing the study, the most common adverse event at the end of TBZ exposure was fatigue, reported by 7 subjects on TBZ (14.3%) and 2 on placebo (6.9%). There was 1 suicide in the double-blind study in a subject taking TBZ. Depression is common in HD and can be exacerbated by TBZ. However, attempted or completed suicides in HD do not necessarily correlate with severity of depression and may be related to associated impulsiveness, obsessive–compulsive behavior, and a variety of socioeconomic factors. Nevertheless, all patients taking TBZ need to be monitored for signs of depression and suicidal ideation. Cognition in the TBZ group, as assessed by the Unified Huntington’s Disease Rating Scale, did not differ from that in the placebo group statistically, although both groups declined as expected over the course of the study. There were no HD-related quality-of-life measures included in the published TBZ studies.

The same group of study participants was given the opportunity to participate in an open-label extension study.Citation82 Seventy-five subjects were elected to participate initially, and at the end of the 3 phases of the extension study, 45 subjects had completed 80 additional weeks of treatment with TBZ. In the extension study, no new adverse effects were reported, and the drug was well tolerated, despite the attrition rate in the study (mostly for logistical reasons). There are a number of older open-label studies, primarily out of the Baylor College of Medicine database.Citation80 The side effects and doses reported were fairly consistent with the double-blind study with the exception of insomnia. Chronic doses of the Baylor group were comparable with that in the long-term HSG study of 62.5-mg total daily dose.

The safety of TBZ was evaluated in these studies, and in an additional study, the safety of sudden withdrawal after long-term use was assessed. The side effects are consistent among studies, and once the drug is titrated slowly to effect, the drug is well tolerated. Potential side effects include akathisia, depression, dizziness, fatigue, or parkinsonism (). During titration, patients may experience insomnia, somnolence, or gastrointestinal distress. If at any time side effects do occur, the dose can be lowered to the previously well-tolerated dose. Since the half-life is short, side effects tend to wane quickly. Withdrawal of TBZ results in a recurrence of chorea, not worse than before starting the drug.Citation83 There are no particular characteristics that may predispose patients to experience 1 or more side effects, but patients with a history of depression should be more closely monitored for changes in mood or impulsivity. Although it is listed in the product information sheet, TBZ does not cause and in fact may be an excellent treatment for tardive dyskinesia.

Table 3 Treatment-emergent side effects to monitor for when using tetrabenazine

Other antichorea medications

Other medications that are commonly considered when treating chorea include dopamine antagonists, benzodiazepines, and glutamate antagonists. Dopamine antagonists (neuroleptics) are perhaps the most commonly considered agents in the management of chorea and psychosis in patients with HD, but few double-blind, placebo-controlled studies evaluating the efficacy and safety of these agents have been published.Citation84Citation86 None of the typical neuroleptics have been found to be effective in reducing chorea in placebo-controlled trials. However, in a study of haloperidol in 10 subjects, oral doses of 1.5–10 mg/d corresponded with at least a 30% reduction in chorea compared with baseline.Citation87 The quantity and quality of these efficacy data need to be taken into account when considering the risks of using typical neuroleptics, particularly tardive dyskinesia. Apathy and akathisia, other potential adverse effects of the dopamine receptor blockers, can be particularly problematic in patients with HD, as they may not have the insight to recognize these problems or may wrongly attribute the symptoms to HD.

Owing to their presumed better tolerability, atypical antipsychotics have recently been evaluated in HD. Olanzapine has been used in small open-label studies to treat the motor symptoms of HD.Citation88Citation92 The range of effect on chorea has been 0%–66% reduction. There are no clinical trials of risperidone for HD, but a few reports suggest a positive effect on the disease with a tolerable adverse-effect profile.Citation93Citation97 Quetiapine has been tried in multiple, small, uncontrolled, nonrandomized trials for HD with some success on both motor and psychiatric symptoms of HD.Citation97Citation99 Clozapine was studied in patients with HD up to a dose of 150 mg/d for up to 31 days, but many adverse events, such as drowsiness, fatigue, anticholinergic symptoms, and walking difficulties, occurred without beneficial effects.Citation100 The newer atypical agent with multiple mechanisms of action, aripiprazole, has been found to be beneficial in a few small trials with a reduction in chorea equivalent to that with TBZ.Citation101Citation103 Although aripiprazole may have fewer adverse effects on mood than TBZ, it is associated with tardive dyskinesia, similar to other typical and atypical neuroleptics.

The N-methyl D-aspartate-antagonist amantadine has been shown in controlled trials to significantly reduce chorea in patients with HD.Citation104 Doses in the range of 400 mg/d or higher may be required for symptomatic benefit, but even in doses used to treat influenza, amantadine may increase irritability and aggressiveness.Citation105 Because riluzole retards striatal glutamate release and the pathological consequences in neurotoxic animal models of HD, multiple large trials have been conducted to determine if there is a possible neuroprotective effect. Riluzole was found to reduce chorea at a dose of 200 mg/d, but not 100 mg/d.Citation106,Citation107 Benzodiazepines are also frequently used clinically in patients with HD to treat anxiety and chorea, and there is limited evidence to suggest that higher doses of clonazepam (up to 5.5 mg/d) may be needed to suppress chorea.Citation108 At this dose, sedation becomes a potential adverse effect. In a few short-term studies (hours), there is evidence that dopamine agonists may reduce the motor signs of HD.Citation109Citation111 Although the pharmacological rationale for using dopamine agonists in the treatment of chorea is not clear, presumably they act by activating the presynaptic dopamine receptors leading to decreased dopamine turnover. More likely, however, is that the observed symptomatic effects are related to nonspecific or sedating effects.

For patients with the akinetic form of HD (Westphal variant), antiparkinsonian medications, such as levodopa, dopamine agonists, and amantadine, may be beneficial.Citation112Citation115 Botulinum toxin injections can also be considered for focal dystonia associated with HD.

Practical uses of TBZ

TBZ is formulated in 12.5- and 25-mg tablets. The medication is not available at local pharmacies but must be ordered through a central distribution center. Prescription information can be found through the company’s Web site, including prescribing information and obtaining the prescription form.Citation116 Clinicians should note on the form that patients (or their guardian) must sign the release of information for the pharmacy to be able to communicate with the patient’s insurance company.

TBZ should be started at 12.5 mg/d, and every week, the dose should be increased by 12.5 mg/d, distributing the drug in 3-times daily dosing. Once patients reach twice-daily dose of 12.5 mg and daily dose of 25 mg (total daily dose of 50 mg), as per the prescribing information, “before patients are given a daily dose of greater than 50 mg, they should be tested for the CYP2D6 gene to determine whether they are poor, extensive, or intermediate metabolizers”. This test can be costly (up to $3,000) and may or may not be covered by insurance. In addition, the drug is titrated to clinical effect, and the genotyping has no impact on the clinical assessment of the patient on the drug. Ultimately, it is up to the discretion of the provider with the patient to determine if this test should be performed.

The goal of the effect of TBZ is to reduce chorea to a point that is acceptable to the patient. The goal should not be to eliminate chorea completely or to reduce chorea to a level that is acceptable to the caregiver or even physician treating the patient. The most common scenario is the titration of the drug to a side effect and then reducing the daily dose to a tolerated dose. Because HD is a progressive disease that changes over time, the dose of TBZ needs to be reassessed frequently (every 2–3 months). TBZ interacts with other CYP2D6 inhibitors, such as paroxetine. There is no evidence if TBZ can be used safely or effectively with neuroleptics or other commonly used medications for chorea.

Conclusion

Until clear neuroprotective strategies are found, clinicians can address the symptoms of patients with this devastating disease. The most and best evidence for treating troublesome chorea leads toward TBZ, but other abnormal involuntary movements, cognition, affective disorders, and behavioral dyscontrol all need to be considered when choosing therapies for patients. Further studies to determine interactions and combination therapy of TBZ need to be completed, as most patients need multiple medications to adequately address symptoms. Other issues related to HD also need to be studied more closely when treating patient with TBZ. Although there is one FDA-approved option at this time and other medications hopefully soon to be available, there are not enough effective, safe interventions that can be offered to our patients and their families with this invariably fatal disease.

Disclosure

The author reports no conflicts of interest in this work.

References

  • The Huntington’s Disease Collaborative Research GroupA novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomesCell19937269719838458085
  • NuciforaFCJrSasakiMPetersMFInterference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicityScience200129155122423242811264541
  • SubramaniamSSixtKMBarrowRSnyderSHRhes, a striatal specific protein, mediates mutant-huntingtin cytotoxicityScience200932459321327133019498170
  • RibeiroFMPaquetMFerreiraLTMetabotropic glutamate receptor-mediated cell signaling pathways are altered in a mouse model of Huntington’s diseaseJ Neurosci201030131632420053912
  • AndrichJSaftCOstholtNMüllerTComplex movement behavior and progression of Huntington’s diseaseNeurosci Lett2007416327227417321683
  • FrankSMarshallFPlumbSFunctional decline due to chorea in Huntington’s diseaseNeurology200462Suppl 5A204
  • VerhagenMLMorrisMJFarmerCHuntington’s disease: a randomized, controlled trial using the NMDA-antagonist amantadineNeurology200259569469912221159
  • BatesGHuntingtin aggregation and toxicity in Huntington’s diseaseLancet200336193691642164412747895
  • ArrasateMMitraSSchweitzerESSegalMRFinkbeinerSInclusion body formation reduces levels of mutant huntingtin and the risk of neuronal deathNature2004431701080581015483602
  • GlassMDragunowMFaullRLThe pattern of neurodegeneration in Huntington’s disease: a comparative study of cannabinoid, dopamine, adenosine and GABA(A) receptor alterations in the human basal ganglia in Huntington’s diseaseNeuroscience200097350551910828533
  • AlbinRLReinerAAndersonKDPreferential loss of striato-external pallidal projection neurons in presymptomatic Huntington’s diseaseAnn Neurol19923144254301375014
  • ReinerAAlbinRLAndersonKDD’AmatoCJPenneyJBYoungABDifferential loss of striatal projection neurons in Huntington diseaseProc Natl Acad Sci U S A19888515573357372456581
  • MitchellIJCooperAJGriffithsMRThe selective vulnerability of striatopallidal neuronsProg Neurobiol199959669171910845758
  • AntoniniALeendersKLSpiegelRStriatal glucose metabolism and dopamine D2 receptor binding in asymptomatic gene carriers and patients with Huntington’s diseaseBrain1996119Pt 6208520959010012
  • AugoodSJFaullRLEmsonPCDopamine D1 and D2 receptor gene expression in the striatum in Huntington’s diseaseAnn Neurol19974222152219266732
  • FeiginATangCMaYThalamic metabolism and symptom onset in preclinical Huntington’s diseaseBrain2007130Pt 112858286717893097
  • AndrewsTCWeeksRATurjanskiNHuntington’s disease progression. PET and clinical observationsBrain1999122Pt 122353236310581228
  • WeeksRAPicciniPHardingAEStriatal D1 and D2 dopamine receptor loss in asymptomatic mutation carriers of Huntington’s diseaseAnn Neurol199640149548687191
  • ChaJHKosinskiCMKernerJAAltered brain neurotransmitter receptors in transgenic mice expressing a portion of an abnormal human huntington disease geneProc Natl Acad Sci U S A19989511648064859600992
  • PerryTLHansenSKlosterMHuntington’s chorea. Deficiency of gamma-aminobutyric acid in brainN Engl J Med197328873373424345566
  • PaulsenJSFunctional imaging in Huntington’s diseaseExp Neurol2009216227227719171138
  • Gourfinkel-AnIParainKHartmannAChanges in GAD67 mRNA expression evidenced by in situ hybridization in the brain of R6/2 transgenic miceJ Neurochem20038661369137812950446
  • NicniocaillBHaraldssonBHanssonOAltered striatal amino acid neurotransmitter release monitored using microdialysis in R6/1 Huntington transgenic miceEur J Neurosci200113120621011135020
  • LiSHLiXJHuntingtin-protein interactions and the pathogenesis of Huntington’s diseaseTrends Genet200420314615415036808
  • BenceNFSampatRMKopitoRRImpairment of the ubiquitin-proteasome system by protein aggregationScience200129255211552155511375494
  • RavikumarBDudenRRubinszteinDCAggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagyHum Mol Genet20021191107111711978769
  • ChaJHTranscriptional dysregulation in Huntington’s diseaseTrends Neurosci200023938739210941183
  • CepedaCHurstRSCalvertCRTransient and progressive electrophysiological alterations in the corticostriatal pathway in a mouse model of Huntington’s diseaseJ Neurosci200323396196912574425
  • LudolphACHeFSpencerPSHammerstadJSabriM3-Nitropropionic acid-exogenous animal neurotoxin and possible human striatal toxinCan J Neurol Sci19911844924981782616
  • GunawardenaSGoldsteinLSPolyglutamine diseases and transport problems: deadly traffic jams on neuronal highwaysArch Neurol2005621465115642849
  • BrowneSEBealMFThe energetics of Huntington’s diseaseNeurochem Res200429353154615038601
  • HarperPSThe epidemiology of Huntington’s diseaseHum Genet19928943653761535611
  • MorrisonPJJohnstonWPNevinNCThe epidemiology of Huntington’s disease in Northern IrelandJ Med Genet19953275245307562964
  • PeterlinBKobalJTeranNEpidemiology of Huntington’s disease in SloveniaActa Neurol Scand2009119637137518976322
  • AlonsoMEOchoaABollMCClinical and genetic characteristics of Mexican Huntington’s disease patientsMov Disord200924132012201519672992
  • SimpsonSAJohnstonAWThe prevalence and patterns of care of Huntington’s chorea in GrampianBr J Psychiatry19891557998042533515
  • PenneyJBJrYoungABShoulsonIHuntington’s disease in Venezuela: 7 years of follow-up on symptomatic and asymptomatic individualsMov Disord19905293992139171
  • HarperPSThe epidemiology of Huntington’s diseaseBatesGHarperPJonesLHuntington’s disease3rd edOxfordMonographs on Medical Genetics2002
  • RibaiPNguyenKHahn-BarmaVPsychiatric and cognitive difficulties as indicators of juvenile huntington disease onset in 29 patientsArch Neurol200764681381917562929
  • BiglanKMRossCALangbehnDRPREDICT-HD Investigators of the Huntington Study GroupMotor abnormalities in premanifest persons with Huntington’s disease: the PREDICT-HD studyMov Disord200924121763177219562761
  • TabriziSJLangbehnDRLeavittBRTRACK-HD investigatorsBiological and clinical manifestations of Huntington’s disease in the longitudinal TRACK-HD study: cross-sectional analysis of baseline dataLancet Neurol20098979180119646924
  • Huntington Study Group PHAROS Investigators (Shoulson I, primary author)At risk for Huntington’s disease: The PHAROS (Prospective Huntington At Risk Observational Study) cohort enrolledArch Neurol200663799199616831969
  • Huntington Study GroupCooperative Huntington’s Observational Research Trial [ClinicalTrials.gov identifier NCT00313495]US National Institutes of Health Available from: http://www.clinicaltrials.govAccessed May 3, 2010
  • SolomonACStoutJCJohnsonSAPredict-HD Investigators of the Huntington Study GroupVerbal episodic memory declines prior to diagnosis in Huntington’s diseaseNeuropsychologia20074581767177617303196
  • JohnsonSAStoutJCSolomonACPredict-HD Investigators of the Huntington Study GroupBeyond disgust: impaired recognition of negative emotions prior to diagnosis in Huntington’s diseaseBrain2007130Pt 71732174417584778
  • PaulsenJSLangbehnDRStoutJCPredict-HD Investigators and Coordinators of the Huntington Study GroupDetection of Huntington’s disease decades before diagnosis: the Predict-HD studyJ Neurol Neurosurg Psychiatry200879887488018096682
  • DuffKPaulsenJSBeglingerLJPredict-HD Investigators of the Huntington Study GroupPsychiatric symptoms in Huntington’s disease before diagnosis: the predict-HD studyBiol Psychiatry200762121341134617481592
  • Huntington Study GroupUnified Huntington’s Disease Rating Scale: reliability and consistencyMov Disord19961121361428684382
  • The American College of Medical Genetics/American Society of Human Genetics Huntington Disease Genetic Testing Working GroupACMG/ASHG statement. Laboratory guidelines for Huntington disease genetic testingAm J Hum Genet1998625124312479545416
  • AndrichJArningLWieczorekSKrausPHGoldRSaftCHuntington’s disease as caused by 34 CAG repeatsMov Disord200823687988118307262
  • KenneyCPowellSJankovicJAutopsy-proven Huntington’s disease with 29 trinucleotide repeatsMov Disord200722112713017115386
  • SnellRGMacMillanJCCheadleJPRelationship between trinucleotide repeat expansion and phenotypic variation in Huntington’s diseaseNat Genet1993443933978401588
  • DiMLSquitieriFNapolitanoGCampanellaGTrofatterJAConneallyPMSuicide risk in Huntington’s diseaseJ Med Genet19933042932958487273
  • PollardJHurry Up and Wait: a cognitive care companion9/25/08 142 pagesLulu press
  • AdamORJankovicJSymptomatic treatment of Huntington diseaseNeurotherapeutics20085218119718394562
  • PhillipsWShannonKMBarkerRAThe current clinical management of Huntington’s diseaseMov Disord200823111491150418581443
  • RozeESaudouFCabocheJPathophysiology of Huntington’s disease: from huntingtin functions to potential treatmentsCurr Opin Neurol200821449750318607213
  • ImarisioSCarmichaelJKorolchukVHuntington’s disease: from pathology and genetics to potential therapiesBiochem J200841219120918466116
  • WalkerFOHuntington’s diseaseLancet2007369955721822817240289
  • JankovicJTreatment of hyperkinetic movement disordersLancet Neurol20098984485619679276
  • BonelliRMWenningGKPharmacological management of Huntington’s disease: an evidence-based reviewCurr Pharm Des200612212701272016842168
  • MestreTFerreiraJCoelhoMMRosaMSampaioCTherapeutic interventions for symptomatic treatment in Huntington’s diseaseCochrane Database Sys Rev20093CD006456
  • NakamuraKAminoffMJHuntington’s disease: clinical characteristics, pathogenesis and therapiesDrugs Today (Barc)20074329711617353947
  • HandleyOJNajiJJDunnettSBRosserAEPharmaceutical, cellular and genetic therapies for Huntington’s diseaseClin Sci (Lond)20061101738816336206
  • BonelliRMHofmannPA review of the treatment options for Huntington’s diseaseExpert Opin Pharmacother20045476777615102562
  • GrimbergenYARoosRATherapeutic options for Huntington’s diseaseCurr Opin Investig Drugs2003415154
  • FrankSAJankovicJAdvances in the pharmacological management of Huntington’s DiseaseDrugs201070556157120329804
  • PaleacuDTetrabenazine in the treatment of Huntington’s DiseaseNeuropsychiatr Dis Treat20073554555119381278
  • BagchiSPDifferential interactions of phencyclidine with tetrabenazine and reserpine affecting intraneuronal dopamineBiochem Pharmacol19833219285128566626259
  • PettiboneDJPfluegerABTotaroJATetrabenazine-induced depletion of brain monoamines: mechanism by which desmethylimipramine protects cortical norepinephrineEur J Pharmacol198410234431436
  • MehvarRJamaliFConcentration-effect relationships of tetrabenazine and dihydrotetrabenazine in the ratJ Pharm Sci19877664614653625491
  • ThibautFFaucheuxBAMarquezJRegional distribution of monoamine vesicular uptake sites in the mesencephalon of control subjects and patients with Parkinson’s disease: a postmortem study using tritiated tetrabenazineBrain Res1995692122332438548292
  • KenneyCHunterCDavidsonAJankovicJShort-term effects of tetrabenazine on chorea associated with Huntington’s diseaseMov Disord2007221101317078062
  • SchermanDHenryJPReserpine binding to bovine chromaffin granule membranes. Characterization and comparison with dihydrotetrabenazine bindingMol Pharmacol19842511131226708929
  • MehvarRJamaliFWatsonMWSkeltonDPharmacokinetics of tetrabenazine and its major metabolite in man and rat. Bioavailability and dose dependency studiesDrug Metab Dispos19871522502552882986
  • RobertsMSWatsonHMMcLeanSMillingenKSDetermination of therapeutic plasma concentrations of tetrabenazine and an active metabolite by high-performance liquid chromatographyJ Chromatogr198122611751826459332
  • RobertsMSMcLeanSMillingenKSGallowayHMThe pharmacokinetics of tetrabenazine and its hydroxy metabolite in patients treated for involuntary movement disordersEur J Clin Pharmacol19862967037083709613
  • Huntington Study GroupTetrabenazine as antichorea therapy in Huntington disease: a randomized controlled trialNeurology200666336637216476934
  • JankovicJBeachJLong-term effects of tetrabenazine in hyperkinetic movement disordersNeurology19974823583629040721
  • KenneyCHunterCJankovicJLong-term tolerability of tetrabenazine in the treatment of hyperkinetic movement disordersMov Disord200722219319717133512
  • FasanoACadedduFGuidubaldiAThe long-term effect of tetrabenazine in the management of Huntington diseaseClin Neuropharmacol200831631331819050408
  • FrankSTetrabenazine as anti-chorea therapy in Huntington disease: an open-label continuation study. Huntington Study Group/TETRA-HD InvestigatorsBMC Neurol200996220021666
  • FrankSOndoWFahnSA study of chorea after tetrabenazine withdrawal in patients with Huntington’s diseaseClin Neuropharmacol200831312713318520979
  • QuinnNMarsdenCDA double blind trial of sulpiride in Huntington’s disease and tardive dyskinesiaJ Neurol Neurosurg Psychiatry19844788448476236286
  • DerooverJBaroFBourguignonRPSmetsPTiapride versus placebo: a double-blind comparative study in the management of Huntington’s choreaCurr Med Res Opin1984953293386241563
  • LeonardDPKidsonMABrownJGShannonPJTaryanSA double blind trial of lithium carbonate and haloperidol in Huntington’s choreaAust N Z J Psychiatry197592115118125578
  • BarrANFischerJHKollerWCSpuntALSinghalASerum haloperidol concentration and choreiform movements in Huntington’s diseaseNeurology198838184882962009
  • BonelliRMMahnertFANiederwieserGOlanzapine for Huntington’s disease: an open label studyClin Neuropharmacol200225526326512410058
  • BonelliRMNiederwieserGTriblGGKöltringerPHigh-dose olanzapine in Huntington’s diseaseInt Clin Psychopharmacol2002172919311890191
  • PaleacuDAncaMGiladiNOlanzapine in Huntington’s diseaseActa Neurol Scand2002105644144412027832
  • SquitieriFCannellaMPiorcelliniABrusaLSimonelliMRuggieriSShort-term effects of olanzapine in Huntington diseaseNeuropsychiatry Neuropsychol Behav Neurol2001141697211234911
  • DippleHCThe use of olanzapine for movement disorder in Huntington’s disease: a first case reportJ Neurol Neurosurg Psychiatry199967112312410454874
  • CankurtaranESOzalpESoygurHCakirAClinical experience with risperidone and memantine in the treatment of Huntington’s diseaseJ Natl Med Assoc20069881353135516916137
  • ErdemogluAKBoratavCRisperidone in chorea and psychosis of Huntington’s diseaseEur J Neurol20029218218311882064
  • MadhusoodananSBrennerRUse of risperidone in psychosis associated with Huntington’s diseaseAm J Geriatr Psychiatry1998643473499793585
  • ParsaMASzigethyEVociJMMeltzerHYRisperidone in treatment of choreoathetosis of Huntington’s diseaseJ Clin Psychopharmacol199717213413510950488
  • AlpayMKoroshetzWJQuetiapine in the treatment of behavioral disturbances in patients with Huntington’s diseasePsychosomatics2006471707216384811
  • SeitzDPMillsonRCQuetiapine in the management of psychosis secondary to Huntington’s disease: a case reportCan J Psychiatry200449641315283541
  • BonelliRMNiederwieserGQuetiapine in Huntington’s disease: a first case reportJ Neurol200224981114111512420714
  • van VugtJPSieslingSVergeerMvan der VeldeEARoosRAClozapine versus placebo in Huntington’s disease: a double blind randomised comparative studyJ Neurol Neurosurg Psychiatry199763135399221965
  • CiammolaASassoneJColciagoCAripiprazole in the treatment of Huntington’s disease: a case seriesNeuropsychiatr Dis Treat200951419557093
  • BrusaLOrlacchioAMoschellaVIaniCBernardiGMercuriNBTreatment of the symptoms of Huntington’s disease: preliminary results comparing aripiprazole and tetrabenazineMov Disord200924112612919170197
  • LinWCChouYHAripiprazole effects on psychosis and chorea in a patient with Huntington’s diseaseAm J Psychiatry200816591207120818765501
  • LucettiCGambacciniGBernardiniSAmantadine in Huntington’s disease: open-label video-blinded studyNeurol Sci200223Suppl 2S83S8412548355
  • StewartJTAdverse behavioral effects of amantadine therapy in Huntington’s diseaseSouth Med J19878010132413252958943
  • Huntington Study GroupDosage effects of riluzole in Huntington’s disease: a multicenter placebo-controlled studyNeurology200361111551155614663041
  • LandwehrmeyerGBDuboisBde YebenesJGRiluzole in Huntington’s disease: a 3-year, randomized controlled studyAnn Neurol200762326227217702031
  • PeirisJBBoralessaHLionelNDClonazepam in the treatment of choreiform activityMed J Aust197618225227131236
  • FrattolaLAlbizzatiMGAlemaniABassiSFerrareseCTrabucchiMAcute treatment of Huntington’s chorea with lisurideJ Neurol Sci19835922472536222164
  • VitaleCMarconiSDiMLShort-term continuous infusion of apomorphine hydrochloride for treatment of Huntington’s chorea: a double blind, randomized cross-over trialMov Disord200722162359236417894335
  • CorsiniGUOnaliPMasalaCCianchettiCMangoniAGessaGApomorphine hydrochloride-induced improvement in Huntington’s chorea: stimulation of dopamine receptorArch Neurol19783512730145840
  • LowPAAllsopJLHalmagyiGMHuntington’s chorea: the rigid form (Westphal variant) treated with levodopaMed J Aust19741113933944275254
  • LowPAAllsopJLHuntington’s chorea – the rigid form (Westphal variant) treated with l-DOPA: a case reportProc Aust Assoc Neurol197310045464275960
  • MagnetMKBonelliRMKapfhammerHPAmantadine in the akinetic-rigid variant of Huntington’s diseaseAnn Pharmacother2004387811941196
  • BonelliRMNiederwieserGDiezJGruberAKöltringerPPramipexole ameliorates neurologic and psychiatric symptoms in a Westphal variant of Huntington’s diseaseClin Neuropharmacol2002251586011852299
  • http://www.xenazineusa.comAccessed 3 May 2010