Está en la página 1de 12

Drugs 2010; 70 (5): 561-571

REVIEW ARTICLE 0012-6667/10/0005-0561/$55.55/0

ª 2010 Adis Data Information BV. All rights reserved.

Advances in the Pharmacological


Management of Huntington’s Disease
Samuel Frank1 and Joseph Jankovic2
1 Boston University School of Medicine, Boston, Massachusetts, USA
2 Baylor College of Medicine, Houston, Texas, USA

Contents
Abstract. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 561
1. Pathogenesis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 562
2. Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 562
3. Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 563
4. Pharmacological Treatment Options . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 564
4.1 Tetrabenazine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 565
4.2 Antipsychotics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 565
4.3 Other Antichorea Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 566
5. Potential Neuroprotective and Experimental Strategies. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 566
6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 568

Abstract There is inevitable physical, cognitive and behavioural decline in Hun-


tington’s disease (HD), a dominantly inherited progressive neurological
disorder. The hallmark of the disease is chorea, an involuntary brief move-
ment that tends to flow between body regions. HD is diagnosed clinically with
genetic confirmation. Predictive testing is available; however, it should be
undertaken with caution in patients at risk for the disease but without clinical
disease expression. Ongoing observational trials have identified not only
early subtle motor signs, but also striatal volume, verbal memory and olfac-
tion as possible early manifestations of clinical disease. Multiple areas of
the brain degenerate, with dopamine, glutamate and GABA being the pre-
dominant neurotransmitters affected in HD. Although many pharmaco-
therapies have been evaluated targeting these neurotransmitters, few well
conducted trials for symptomatic or neuroprotective interventions have
yielded positive results. Tetrabenazine is one of the better studied and more
effective agents for reducing chorea, although with a risk of potentially ser-
ious adverse effects. Newer antipsychotic agents such as olanzapine and
aripiprazole may have adequate efficacy with a more favourable adverse-effect
profile than older antipsychotics for treating chorea and psychosis. In this
review, the pathogenesis, epidemiology and diagnosis of HD are discussed as
background for understanding potential pharmacological treatment options.
Potential strategies to delay the progression of HD that have been studied
and are planned for the future are summarized. Although there is no current
562 Frank & Jankovic

method to change the course of this devastating disease, education and


symptomatic therapies are effective tools available to clinicians and the fa-
milies affected by HD.

Huntington’s disease (HD) is a hereditary, and the striatum. Medium spiny neurons in the
progressive neurodegenerative disease clinically striatum that contain GABA and enkephalin are
characterized by abnormal involuntary move- affected early in the disease, and are the primary
ments, behavioural disturbance, cognitive dys- neurons targeted in HD. These neurons typically
function and psychiatric disease. The disease is project to the lateral globus pallidus. There is
caused by a CAG (glutamine) trinucleotide ex- then progression to the remainder of the basal
pansion in exon 1 of the huntingtin gene (HTT) at ganglia with subsequent dissemination, including
the location 4p16.9.[1] The function of huntingtin the cortex and substantia nigra. There are intra-
protein (HTT) is not known, but it may be in- nuclear and cytoplasmic inclusions of the ag-
volved in internal cell signalling, maintenance of gregate HTT. Huntingtin is cross-linked with
cyclic adenosine monophosphate response ele- other soluble HTT to form the inclusion bodies in
ment binding (CREB) protein and preventing neurons. It is not known if the accumulation of
neuronal toxicity.[2] There is early evidence to HTT conglomerate results in cell death or if the
suggest that the binding of the protein Rhes (Ras soluble form of the protein is the toxic form.[7,8]
homologue enriched in striatum) to huntingtin Although dopamine, glutamate and GABA
may be necessary to cause cellular toxicity.[3] are thought to be most affected in HD and
Although there is no established treatment to are targeted for treatment, multiple neuro-
delay the onset or forestall the progression of transmitters and their receptors are involved in
HD, symptomatic treatment of chorea may be various regions in the HD brain (table I).[9-22]
beneficial in some individuals, as it may have a There is typically a sequence and pattern of re-
favourable effect on motor function, quality of ceptor changes that emerges with degeneration in
life and safety.[4-6] Since pathogenesis-targeted HD, beginning with cannabinoid and adenosine
therapy is a desirable goal, we first briefly review A2a receptor binding in the striatum and globus
current hypotheses about the pathogenesis of pallidus externa (GPe). There is also increased
HD, but the chief focus is on symptomatic and GABAA receptor binding in the GPe.
experimental treatment strategies. As the disease progresses, there is typically loss
For this review, the search strategy began with a of dopamine D1 receptors in the striatum as well as
PubMed search with the term Huntington’s disease cannabinoid and D1 receptors in the substantia
(limits of Huntington’s disease in the title/abstract nigra, eventually involving the rest of the basal
field and English, human, clinical trials) that yield- ganglia.[9] The preferential loss of enkephalinergic
ed 175 original articles (search last run on 15 striatal neurons correlates with the appearance
December 2009). Additional trials of an older of chorea.[10,11] Loss of substance P/dynorphin
nature were identified using the same search re- neurons correlates with the increase in dystonia in
strictions with the outdated keyword Huntington’s the later stages of disease.[12]
chorea (65 original articles). Further ongoing trials
were identified using www.clinicaltrials.gov and 2. Epidemiology
the search term Huntington’s disease in the condi-
tion field, yielding 47 results. Most European populations show a pre-
valence rate of 4–8 per 100 000, and HD may be
1. Pathogenesis as frequently seen in India and parts of central
Asia.[23] More recent studies in other European
Pathologically, HD is associated with diffuse nations have confirmed this prevalence rate.[24,25]
loss of neurons, particularly involving the cortex HD is notably rare in Finland and Japan, but

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
Pharmacological Management of Huntington’s Disease 563

Table I. Receptors involved in Huntington’s disease[9-22] nosis. Recent evidence has been published ex-
Receptor Location Stage of disease amining those who are gene positive but not yet
Adenosine A2a Striatum, Gpe Preclinical to advanced symptomatic by motor criteria (TRACK-HD,
Cannabinoid Striatum, GPe Preclinical to advanced Neurobiological Predictors of Huntington’s
Dopamine D1 Striatum, substantia Clinical diagnosis to Disease Trial [PREDICT-HD] and Prospective
nigra advanced Huntington at Risk Observational Study
Dopamine D2 Caudate, putamen Prodromal [PHAROS]).[29-31] There is also a study currently
Dynorphin Striatum Emergence of dystonia prospectively enrolling subjects with HD and their
Enkephalin Striatum Emergence of chorea affected and unaffected family members to further
GABA Striatum Advanced understand biomarkers of disease and signs of
Glutamate Cortical Preclinical to advanced onset (Cooperative Huntington’s Observational
Substance P Striatum Emergence of dystonia Research Trial [COHORT]).[32] The focus of the
GPe = globus pallidus externa. COHORT study is not neuroimaging, but rather
clinical measures and biological samples.
Subtle motor abnormalities have been asso-
data for eastern Asia and Africa are inadequate. ciated with a smaller striatal volume and higher
There are also well known large populations of probability of disease diagnosis.[29] Lower scores
patients with HD in Scotland and the Lake on the Hopkins Verbal Learning Test-Revised
Maracaibo region of Venezuela.[26,27] The dis- were associated with closer proximity to diag-
order may be underestimated in the Black nosis and smaller striatal volumes.[33] Subjects
American population. There have been no wide- with an expanded repeat and preclinical diag-
spread epidemiological studies of HD in the US nosis of HD also had less accurate recognition of
since the wide availability of genetic testing, but it negative emotions.[34] In addition, motor ex-
is estimated that approximately 25 000–30 000 amination score, striatal volume, speeded finger
individuals have manifest HD and a further tapping, self-timed finger tapping, word list
150 000–250 000 individuals are at risk for HD. learning and odour identification in subjects in
Men and women are affected equally and typi- the PREDICT-HD study were all associated with
cally become symptomatic in the third and the predicted time to diagnosis.[35] Expansion-
fourth decades. The symptoms of HD can start at positive individuals also reported more psychiatric
any age ranging from 1 to 90 years. Approxi- symptoms (depression, anxiety, obsessive-compul-
mately 5–10% of cases are classified as juvenile siveness) than expansion-negative individuals.[36]
onset, with symptoms starting before the age of The TRACK-HD study has confirmed some and
20 years. The vast majority of juvenile cases are expanded upon findings in PREDICT-HD in that
inherited paternally, and in place of chorea, pa- presymptomatic subjects had significant changes in
tients may exhibit more parkinsonian signs of whole-brain volume, regional grey and white mat-
bradykinesia, dystonia, tremors and a cognitive ter differences, impairment in a range of motor
deficit.[28] When patients exhibit more hypo- tasks, oculomotor findings, and cognitive and
kinetic features, they are said to have the Westphal neuropsychiatric dysfunction.[30] The various mo-
variant of HD. tor and non-motor measures on the neurological
examination used to diagnose and track HD are
included in the Unified Huntington Disease Rating
3. Diagnosis Scale (UHDRS).[37] The scale is divided into six
sections: motor, cognitive, behavioural and three
HD is diagnosed based on the combination of functional scales (total functional capacity, func-
motor, behavioural and cognitive symptoms in tional checklist and the independence scale).
the setting of a family history of the disease. A Based on the guidelines published by the
DNA test showing abnormal CAG expansion in American College of Genetics, patients with 40 or
the HTT gene can be used to confirm the diag- more repeats have 100% penetrance.[38] In other

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
564 Frank & Jankovic

words, if patients have 40 or more copies of the attempt suicide, which is a cause of death in 8–9%
gene, they will inevitably express the disease of patients.[42]
clinically. In patients with a CAG repeat length in Behavioural dyscontrol can be a severely dis-
the 36–39 range, there is reduced penetrance with abling symptom of HD, causing distress to the
increased likelihood of expression occurring with patient, family and caregivers. Environmental
longer length of repeats and with longer lifespan approaches and cognitive interventions are the
of the patient. Patients with fewer than 36 repeats mainstay of treatments, but pharmacological
will generally not develop clinical disease, al- agents can augment addressing disruptive beha-
though there are case reports of patients who viours. Depression, anxiety, aggressive, impulsive
manifest HD in this range.[39,40] Patients with an and obsessive-compulsive behaviours are also
allele repeat size of 27–35 have demonstrated frequently treated pharmacologically and require
meiotic instability, particularly in sperm, indi- behavioural intervention, but caution should be
cating that the following generation is at higher used to avoid oversedation and apathy, already
risk of inheriting an expanded number of repeats, common in HD patients.
increasing the risk of clinical disease. The length
of repeat size correlates generally with the age of 4. Pharmacological Treatment Options
onset, but not necessarily with the severity or
duration of disease (figure 1).[41] Many agents and surgical procedures have
The course of the disease is typically 15–20 been evaluated in HD for their antichoreic effi-
years, with dementia, mutism, dystonia and bra- cacy, including dopamine-depleting agents, do-
dykinesia predominating in end stages. Patients pamine antagonists, benzodiazepines, glutamate
with more dystonia and swallowing issues may antagonists, acetylcholinesterase inhibitors,
experience accelerated complications and, there- dopamine agonists, antiseizure medications, can-
fore, shorter lifespan. Chorea may become a nabinoids, lithium, deep brain stimulation and
safety issue with larger amplitude movements fetal cell transplantation.[43-46] Pharmacological
causing injury or malpositioning, and frequent interventions typically address the hyperkinetic
movements resulting in skin injuries, infections or movement disorders (chorea, dystonia, ballism,
even fractures and head trauma. Cause of death is myoclonus and tics), but may also affect psy-
typically related to complications of immobility chiatric issues (irritability, depression, anxiety,
such as skin breakdown, pneumonia, cardiac mania, apathy, obsessive-compulsive disorder) or
disease or infection. However, 25% of patients cognitive decline associated with HD. Adjunctive
therapies, alternative and complementary thera-
pies, behavioural plans and cognitive interven-
tions also play a critical role in addressing the
symptoms of HD.
Age at onset of symptoms (y)

90
Several excellent guidelines on the sympto-
matic treatment of HD have been published.[43-53]
Overall, there is far too little evidence available to
guide long-term symptomatic treatment in HD,
40 and double-blind and long-term studies assessing
various treatment strategies in HD are needed.[49]
A Cochrane review examined 22 trials (1254
1
participants); 9 trials had a crossover design and
20 44 100
13 were conducted in parallel. The studies ex-
CAGn
amined were of relatively short duration, ranging
Fig. 1. On average, the larger the repeat size of CAG sequence from 2 to 80 weeks. Various pharmacological
inherited, the younger the age of onset of Huntington’s disease (HD).
The median number of repeats (CAGn) inherited for HD is 44, with a interventions were studied including anti-
mean age at onset of about 40 years. dopaminergic drugs (five studies), glutamate

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
Pharmacological Management of Huntington’s Disease 565

receptor antagonists (five studies) and energy 100 mg/day or the development of intolerable
metabolites (five studies). The authors concluded adverse effects. The primary efficacy outcome
that based on available evidence, only tetra- was the change from baseline in the total maximal
benazine showed clear efficacy for the control of chorea score of the UHDRS.[37] On this scale,
chorea, but ‘‘no statement can be made regarding chorea is graded from 0 to 4 (with 0 representing
the best medical practice for the control of motor no chorea) for seven body regions for a range in
and non-motor symptoms in HD.’’ total scores from 0 to 28. Compared with base-
line, treatment with tetrabenazine resulted in a
4.1 Tetrabenazine reduction of 5.0 units in chorea compared with a
1.5-unit reduction for those assigned to the pla-
Tetrabenazine is the only US FDA-approved cebo group. About 50% of tetrabenazine-treated
drug for HD and is indicated for the treatment subjects had a 6-point or greater improvement
of chorea associated with HD. In addition to the compared with 7% of placebo recipients. There is
US and Canada, tetrabenazine is marketed in also evidence to suggest continued long-term effi-
Australia, Denmark, France, Germany, Ireland, cacy and tolerability of tetrabenazine in HD.[64-67]
Israel, Italy, New Zealand, Portugal, Spain, In the same study, the adverse events that oc-
Switzerland and the UK. By reversibly inhibiting curred significantly more frequently in the tetra-
the central vesicular monoamine transporter type benazine group included drowsiness/somnolence,
2 (VMAT2), tetrabenazine depletes dopamine insomnia, depressed mood, agitation, akathisia
more selectively than it depletes noradrenaline and hyperkinesia. However, by the conclusion of
(norepinephrine).[54,55] The highest binding dens- the maintenance phase, when subjects were pre-
ity for tetrabenazine is in the caudate nucleus, sumably on optimal dosage, there were no sig-
putamen and nucleus accumbens, areas known to nificant differences between tetrabenazine and
bear the brunt of pathology in HD.[56,57] VMAT2 placebo. Among subjects completing the study,
binding and monoamine depletion by tetra- the most common adverse event at the end of
benazine is reversible, lasts hours and is not tetrabenazine exposure was fatigue, reported by
modified by long-term treatment.[58,59] These seven subjects on tetrabenazine (14.3%) and two
features of the drug differentiate it from the other on placebo (6.9%). There was one suicide in the
dopamine-depleting agent reserpine that binds double-blind study in a subject taking tetra-
not only to VMAT2 but also to VMAT1, which is benazine. Depression is common in HD and can
localized to the peripheral nervous system. The be exacerbated by tetrabenazine. However, sui-
peripheral binding accounts for some of the cidality in HD does not necessarily correlate with
peripheral adverse effects of reserpine, such as severity of depression, and may also be related to
orthostatic hypotension and diarrhoea. In con- associated impulsiveness, obsessive-compulsive
trast to tetrabenazine, reserpine binds to VMAT1 behaviour and a variety of socioeconomic
and VMAT2 irreversibly, making the duration of factors. Nevertheless, all patients taking tetra-
action considerably longer (hours vs days). In benazine need to be monitored for signs of de-
addition to tetrabenazine, the two active meta- pression and suicidality. Cognition, as assessed
bolites, a- and b-dihydrotetrabenazine, have by the UHDRS, did not differ from that in the
longer half-lives and are more highly bound to placebo group statistically, although both groups
proteins than the parent compound.[60-62] declined as expected over the course of the study.
The efficacy of tetrabenazine as an antichoreic There were no HD-related quality-of-life measures
drug was convincingly demonstrated in a double- included in the published tetrabenazine studies.
blind, placebo-controlled trial conducted by the
Huntington Study Group.[63] Subjects were ran- 4.2 Antipsychotics
domized to receive either tetrabenazine (n = 54)
or placebo (n = 30). tetrabenazine was titrated Dopamine receptor blocking agents (anti-
weekly in 12.5 mg increments to a maximum of psychotics) are commonly considered in the

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
566 Frank & Jankovic

management of chorea and psychosis in HD, chorea in HD.[3,88] Dosages in the range of
but, surprisingly, few double-blind, placebo- 400 mg/day or higher may be required for symp-
controlled studies evaluating the efficacy and tomatic benefit, but even in dosages used to treat
safety of these agents have been published.[68-70] influenza, amantadine may increase irritability
None of the typical antipsychotic agents have and aggressiveness.[89] Because riluzole retards
been found to be effective in reducing chorea in striatal glutamate release and the pathological
placebo-controlled trials. However, in one study consequences in neurotoxic animal models of
of haloperidol in ten subjects, oral doses of HD, multiple large trials have been conducted to
1.5–10 mg/day corresponded with at least a 30% determine if there is a possible neuroprotective
reduction in chorea compared with baseline.[71] effect. Riluzole was found to reduce chorea at a
The quantity and quality of these efficacy data daily dose of 200 mg but not 100 mg.[90,91] Benzo-
need to be taken into account when considering diazepines are also frequently used clinically in
the risks of using typical antipsychotic agents, HD to treat anxiety as well as chorea and there is
particularly tardive dyskinesia. Apathy and aka- limited evidence to suggest that higher dosages of
thisia, other potential adverse effects of the clonazepam (up to 5.5 mg/day) may be needed to
dopamine receptor blockers, can be particularly suppress chorea.[92] At this dosage, sedation
problematic in patients with HD, as they may not becomes a potential adverse effect. In a few short-
have the insight to recognize these problems or term studies (hours), there is evidence that do-
may wrongly attribute the symptoms to HD. pamine agonists may reduce the motor signs of
Because of their presumed better tolerability, HD.[93-95] Although the pharmacological ratio-
atypical antipsychotics have been evaluated in HD nale for using dopamine agonists in the treatment
more recently. Olanzapine has been used in small of chorea is not clear, presumably they act by
open-label studies to treat the motor symptoms of activating the presynaptic dopamine receptors
HD.[72-76] The range of effect on chorea has been leading to decreased dopamine turnover. How-
0–66% reduction. There are no clinical trials of ever, it is more likely that the observed sympto-
risperidone for HD, but a few reports suggest matic effects are related to nonspecific or sedating
a positive effect on the disease with a tolerable effects.
adverse-effect profile.[77-80] Quetiapine has been tried For patients with the akinetic form of HD,
in multiple, small, uncontrolled, nonrandomized such as the Westphal variant, antiparkinsonian
trials for HD with some success on both motor and medications such as levodopa, dopamine ago-
psychiatric symptoms of HD.[81-83] Clozapine was nists and amantadine may be beneficial.[96-99]
studied in patients with HD up to a dose of Botulinum toxin injections can be also used for
150 mg/day for up to 31 days, but many adverse focal dystonia associated with HD, including
events such as drowsiness, fatigue, anticholinergic oromandibular dystonia.[100]
symptoms and walking difficulties occurred with-
out beneficial effects.[84] The newer atypical agent 5. Potential Neuroprotective and
with multiple mechanisms of action, aripiprazole, Experimental Strategies
has been found to be beneficial in a few small trials
with a reduction in chorea equivalent to that with No known therapies slow the progression of
tetrabenazine.[85-87] Although aripiprazole may symptoms or change the course of HD. However,
have fewer adverse effects on mood than tetra- there is suggestive evidence using various agents
benazine, it is associated with tardive dyskinesia, that some compounds may extend life in animal
similar to other typical and atypical antipsychotics. models.[101,102] There are some indications from
early human clinical trials that antioxidants and
4.3 Other Antichorea Agents mitochondrial enhancers such as ubidecarenone,
creatine or ethyl-eicosapentaenoic acid (ethyl-
The NMDA-antagonist amantadine has been EPA) may favourably influence the course of
shown in controlled trials to significantly reduce disease.[103,104] Other agents hold theoretical

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
Pharmacological Management of Huntington’s Disease 567

promise such as those that affect adenosine or designed to examine for delay of progression of
cannabinoid receptors.[105-107] disease dopamine stabilizers (OSU6162 and
In a Cochrane review of the literature evalu- ACR16), ethyl-EPA and phenylbutyrate.[111-113]
ating neuroprotective strategies, eight random- Other recently completed trials have confirmed
ized, double-blind, placebo-controlled clinical suspicion of futility, such as with minocycline.[114]
trials were identified that included 1366 sub- Past trials of individual agents have failed, but the
jects.[108] Subjects were treated for a mean 52 synergy of the combination of ubidecarenone and
(range 30–144) weeks. Multiple mechanisms of creatine may have promise.[115] Lithium has been
action and interventions were summarized, includ- found to induce autophagy via inositol mono-
ing vitamin E, idebenone, baclofen, lamotrigine, phosphatase (IMPase) inhibition, a process in-
creatine, ubidecarenone/remacemide, ethyl-EPA dependent of the mammalian target of rapamycin
and riluzole. The conclusion from this meta- (mTOR)-induced autophagy, but combining it
analysis was that no compounds studied thus far with sirolimus (rapamycin) may be particularly
provided neuroprotection from advancement of useful in the treatment of neurodegenerative dis-
the disease, but interventions were generally safe eases, such as HD.[116] Fluoxetine, one of the se-
and well tolerated. All studies to date have used lective serotonin reuptake inhibitors, has been
clinical scales to measure progression of disease, also shown to stimulate CREB, increase produc-
and future studies will probably need to use a tion of brain-derived neurotrophic factor,
more sensitive biomarker for disease onset and enhance glycogenolysis, block voltage-gated cal-
progression. cium and sodium channels, and decrease the
If therapies are developed to delay the pro- conductance of mitochondrial voltage-dependent
gression or prevent the onset of disease, the in- anion channels, and thus it may potentially exert
tervention will likely need to be initiated as early a neuroprotective effect in HD.[117] There are
in the course of the disease as possible. There is an ongoing trials of citalopram, memantine, ato-
ongoing clinical trial to examine if high doses of moxetine and ramelteon, medications currently
creatine can be safely tolerated in gene-positive, available that have promise for symptomatic
asymptomatic patients.[109] However, there were treatment as well as neuroprotection. A new drug
significant delays initiating this study owing to in the antihistamine class, dimebolin, has been
the regulatory issues of giving an intervention to under development for the past few years for the
a population without a disease. Although the re- cognitive aspects of HD.[118] Dimebolin is well
search participants have an expanded mutation tolerated in short-term studies and will need to be
and will inevitably develop clinical symptoms, at studied not only for its effect on cognition, but
the time of enrolment they are without disease. also for its potential effects on motor and non-
Administering relatively high, unstudied doses of motor symptoms and progression of disease.[119]
creatine created issues in study design. If future Future studies may also include older drugs de-
interventions are going to address the underlying signed for another purpose such as methazola-
pathology of cell death before the onset of clinical mide, since it crosses the blood-brain barrier and
disease, the FDA will need to find a way to make is an inhibitor of cytochrome c release.[120] The
it easier for drug companies to initiate and eval- future also holds promise for the use of neurturin,
uate therapy in delayed-onset genetic disorders anti-huntingtin antibodies, RNA interference or
such as HD. Eventually, this compound may be inhibition of cleavage at the caspase-6 site.[121]
used in clinical trials to determine if disease onset Finally, as suggested by recent findings,[3] Rhes-
can be delayed or if the disease course can be elicited disaggregation of mutated HTT may
modified by taking high doses of creatine. account for the localized neuropathology of HD
Future neuroprotection strategies are likely to in the striatum and to a lesser extent in the cortex;
be built from previously published trials and drugs that block the binding of Rhes (and other
concepts.[110] There are recently completed trials interacting proteins) to mutated HTT may exert
with early evidence of promise for larger trials neuroprotective effects in HD.[122]

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
568 Frank & Jankovic

6. Conclusions 8. Arrasate M, Mitra S, Schweitzer ES, et al. Inclusion body


formation reduces levels of mutant huntingtin and the risk
of neuronal death. Nature 2004; 431 (7010): 805-10
Until clear neuroprotective strategies are
9. Glass M, Dragunow M, Faull RL. The pattern of neuro-
found and tested, clinicians will continue to treat degeneration in Huntington’s disease: a comparative
patients symptomatically. There is best evidence study of cannabinoid, dopamine, adenosine and
for treating the chorea with tetrabenazine, but GABA(A) receptor alterations in the human basal ganglia
in Huntington’s disease. Neuroscience 2000; 97 (3): 505-19
other abnormal involuntary movements, cogni-
10. Albin RL, Reiner A, Anderson KD, et al. Preferential loss
tion, affective disorders and behavioural dys- of striato-external pallidal projection neurons in pre-
control all need to be considered when choosing symptomatic Huntington’s disease. Ann Neurol 1992;
therapies for patients. There is less evidence for 31 (4): 425-30
11. Reiner A, Albin RL, Anderson KD, et al. Differential loss
use of therapies in HD than for other neuro- of striatal projection neurons in Huntington disease. Proc
degenerative diseases such as Parkinson’s disease, Natl Acad Sci U S A 1988; 85 (15): 5733-7
but well designed clinical trials continue to be 12. Mitchell IJ, Cooper AJ, Griffiths MR. The selective vul-
developed and implemented. There are not nerability of striatopallidal neurons. Prog Neurobiol
1999; 59 (6): 691-719
enough effective, safe interventions that can be
13. Antonini A, Leenders KL, Spiegel R, et al. Striatal glucose
offered to our patients and their families with this metabolism and dopamine D2 receptor binding in
invariably fatal disease, but there are multiple asymptomatic gene carriers and patients with Hunting-
compounds currently under investigation. ton’s disease. Brain 1996; 119 (Pt 6): 2085-95
14. Augood SJ, Faull RL, Emson PC. Dopamine D1 and D2
receptor gene expression in the striatum in Huntington’s
disease. Ann Neurol 1997; 42 (2): 215-21
Acknowledgements 15. Feigin A, Tang C, Ma Y, et al. Thalamic metabolism and
symptom onset in preclinical Huntington’s disease. Brain
Dr Samuel Frank has received consulting fees from 2007; 130 (Pt 11): 2858-67
Lundbeck and Allergan. Dr Joseph Jankovic is a consultant/ 16. Andrews TC, Weeks RA, Turjanski N, et al. Huntington’s
advisory committee member for Allergan, Inc., Michael J. Fox disease progression. PET and clinical observations. Brain
Foundation for Parkinson Research, Merz Pharmaceuticals, 1999; 122 (Pt 12): 2353-63
Lundbeck Inc. and Teva. No sources of funding were used to 17. Weeks RA, Piccini P, Harding AE, et al. Striatal D1 and
assist in the preparation of this review. D2 dopamine receptor loss in asymptomatic mutation
carriers of Huntington’s disease. Ann Neurol 1996; 40 (1):
49-54
18. Cha JH, Kosinski CM, Kerner JA, et al. Altered brain
neurotransmitter receptors in transgenic mice expressing a
References portion of an abnormal human huntington disease gene.
1. The Huntington’s Disease Collaborative Research Group. Proc Natl Acad Sci U S A 1998; 95 (11): 6480-5
A novel gene containing a trinucleotide repeat that is ex- 19. Perry TL, Hansen S, Kloster M. Huntington’s chorea.
panded and unstable on Huntington’s disease chromo- Deficiency of gamma-aminobutyric acid in brain. N Engl
somes. Cell 1993; 72 (6): 971-83 J Med 1973; 288 (7): 337-42
2. Nucifora Jr FC, Sasaki M, Peters MF, et al. Interference by 20. Paulsen JS. Functional imaging in Huntington’s disease.
huntingtin and atrophin-1 with cbp-mediated transcrip- Exp Neurol 2009; 216 (2): 272-7
tion leading to cellular toxicity. Science 2001; 291 (5512): 21. Gourfinkel-An I, Parain K, Hartmann A, et al. Changes in
2423-8 GAD67 mRNA expression evidenced by in situ hybridi-
3. Subramaniam S, Sixt KM, Barrow R, et al. Rhes, a striatal zation in the brain of R6/2 transgenic mice. J Neuro-
specific protein, mediates mutant-huntingtin cytotoxicity. chem 2003; 86 (6): 1369-78
Science 2009; 324 (5932): 1327-30 22. Nicniocaill B, Haraldsson B, Hansson O, et al. Altered
4. Andrich J, Saft C, Ostholt N, et al. Complex movement striatal amino acid neurotransmitter release monitored
behaviour and progression of Huntington’s disease. using microdialysis in R6/1 Huntington transgenic mice.
Neurosci Lett 2007; 416 (3): 272-4 Eur J Neurosci 2001; 13 (1): 206-10
5. Frank S, Marshall F, Plumb S, et al. Functional decline due 23. Harper PS. The epidemiology of Huntington’s disease.
to chorea in Huntington’s disease. Neurology 2004; 62 Hum Genet 1992; 89 (4): 365-76
Suppl. 5: A204 24. Morrison PJ, Johnston WP, Nevin NC. The epidemiology
6. Verhagen ML, Morris MJ, Farmer C, et al. Huntington’s of Huntington’s disease in Northern Ireland. J Med Genet
disease: a randomized, controlled trial using the NMDA- 1995; 32 (7): 524-30
antagonist amantadine. Neurology 2002; 59 (5): 694-9 25. Peterlin B, Kobal J, Teran N, et al. Epidemiology of Hun-
7. Bates G. Huntingtin aggregation and toxicity in Hunting- tington’s disease in Slovenia. Acta Neurol Scand 2009;
ton’s disease. Lancet 2003; 361 (9369): 1642-4 119 (6): 371-5

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
Pharmacological Management of Huntington’s Disease 569

26. Simpson SA, Johnston AW. The prevalence and patterns of 40. Kenney C, Powell S, Jankovic J. Autopsy-proven
care of Huntington’s chorea in Grampian. Br J Psychiatry Huntington’s disease with 29 trinucleotide repeats. Mov
1989; 155: 799-804 Disord 2007; 22 (1): 127-30
27. Penney Jr JB, Young AB, Shoulson I, et al. Huntington’s 41. Snell RG, MacMillan JC, Cheadle JP, et al. Relationship
disease in Venezuela: 7 years of follow-up on symptomatic between trinucleotide repeat expansion and phenotypic
and asymptomatic individuals. Mov Disord 1990; 5 (2): variation in Huntington’s disease. Nat Genet 1993; 4 (4):
93-9 393-7
28. Ribai P, Nguyen K, Hahn-Barma V, et al. Psychiatric 42. Di ML, Squitieri F, Napolitano G, et al. Suicide risk in
and cognitive difficulties as indicators of juvenile hun- Huntington’s disease. J Med Genet 1993; 30 (4): 293-5
tington disease onset in 29 patients. Arch Neurol 2007; 64 43. Adam OR, Jankovic J. Symptomatic treatment of
(6): 813-9 Huntington disease. Neurotherapeutics 2008; 5 (2): 181-97
29. Biglan KM, Ross CA, Langbehn DR, et al., PREDICT- 44. Phillips W, Shannon KM, Barker RA. The current clinical
HD Investigators of the Huntington Study Group. Motor management of Huntington’s disease. Mov Disord 2008;
abnormalities in premanifest persons with Huntington’s 23 (11): 1491-504
disease: the PREDICT-HD study. Mov Disord 2009 Sep 45. Roze E, Saudou F, Caboche J. Pathophysiology of Hun-
15; 24 (12): 1763-72 tington’s disease: from huntingtin functions to potential
30. Tabrizi SJ, Langbehn DR, Leavitt BR, et al., TRACK-HD treatments. Curr Opin Neurol 2008; 21 (4): 497-503
investigators. Biological and clinical manifestations of 46. Imarisio S, Carmichael J, Korolchuk V, et al. Huntington’s
Huntington’s disease in the longitudinal TRACK-HD disease: from pathology and genetics to potential thera-
study: cross-sectional analysis of baseline data. Lancet pies. Biochem J 2008; 412: 191-209
Neurol 2009 Sep; 8 (9): 791-801
47. Walker FO. Huntington’s disease. Lancet 2007; 369 (9557):
31. Huntington Study Group PHAROS Investigators (Shoul- 218-28
son I, primary author). At risk for Huntington’s disease:
48. Jankovic J. Treatment of hyperkinetic movement dis-
the PHAROS (Prospective Huntington at risk observa-
orders. Lancet Neurol 2009; 8 (9): 844-56
tional study) cohort enrolled. Arch Neurol 2006; 63: 991-8
49. Bonelli RM, Wenning GK. Pharmacological management
32. Huntington Study Group. Cooperative Huntington’s Ob-
of Huntington’s disease: an evidence-based review. Curr
servational Research Trial [ClinicalTrials.gov identifier
Pharm Des 2006; 12 (21): 2701-20
NCT00313495]. US National Institutes of Health. Clinical
Trials.gov [online]. Available from URL: http://www.clini 50. Nakamura K, Aminoff MJ. Huntington’s disease: clinical
caltrials.gov [Accessed 2010 Jan 26] characteristics, pathogenesis and therapies. Drugs Today
(Barc) 2007; 43 (2): 97-116
33. Solomon AC, Stout JC, Johnson SA, et al., Predict-HD in-
vestigators of the Huntington Study Group. Verbal episodic 51. Handley OJ, Naji JJ, Dunnett SB, et al. Pharmaceutical,
memory declines prior to diagnosis in Huntington’s disease. cellular and genetic therapies for Huntington’s disease.
Neuropsychologia 2007 Apr 9; 45 (8): 1767-76 Clin Sci (Lond) 2006; 110 (1): 73-88
34. Johnson SA, Stout JC, Solomon AC, et al., Predict-HD 52. Bonelli RM, Hofmann P. A review of the treatment options
Investigators of the Huntington Study Group. Beyond for Huntington’s disease. Expert Opin Pharmacother
disgust: impaired recognition of negative emotions prior 2004; 5 (4): 767-76
to diagnosis in Huntington’s disease. Brain 2007 Jul; 130 53. Grimbergen YA, Roos RA. Therapeutic options for Hun-
(Pt 7): 1732-44 tington’s disease. Curr Opin Investig Drugs 2003; 4 (1):
35. Paulsen JS, Langbehn DR, Stout JC, et al., Predict-HD 51-4
Investigators and Coordinators of the Huntington Study 54. Bagchi SP. Differential interactions of phencyclidine
Group. Detection of Huntington’s disease decades before with tetrabenazine and reserpine affecting intraneuronal
diagnosis: the Predict-HD study. J Neurol Neurosurg dopamine. Biochem Pharmacol 1983; 32 (19): 2851-6
Psychiatry 2008 Aug; 79 (8): 874-80 55. Pettibone DJ, Pflueger AB, Totaro JA. Tetrabenazine-
36. Duff K, Paulsen JS, Beglinger LJ, et al., Predict-HD induced depletion of brain monoamines: mechanism by
Investigators of the Huntington Study Group. Psychiatric which desmethylimipramine protects cortical norepine-
symptoms in Huntington’s disease before diagnosis: the phrine. Eur J Pharmacol 1984; 102 (3-4): 431-6
Predict-HD study. Biol Psychiatry 2007 Dec 15; 62 (12): 56. Mehvar R, Jamali F. Concentration-effect relationships
1341-6 of tetrabenazine and dihydrotetrabenazine in the rat.
37. Huntington Study Group. Unified Huntington’s Disease J Pharm Sci 1987; 76 (6): 461-5
Rating Scale: reliability and consistency. Mov Disord 57. Thibaut F, Faucheux BA, Marquez J, et al. Regional dis-
1996; 11 (2): 136-42 tribution of monoamine vesicular uptake sites in the
38. The American College of Medical Genetics/American mesencephalon of control subjects and patients with
Society of Human Genetics Huntington Disease Genetic Parkinson’s disease: a postmortem study using tritiated
Testing Working Group. ACMG/ASHG statement. tetrabenazine. Brain Res 1995; 692 (1-2): 233-43
Laboratory guidelines for Huntington disease genetic 58. Kenney C, Hunter C, Davidson A, et al. Short-term effects
testing. Am J Hum Genet 1998; 62 (5): 1243-7 of tetrabenazine on chorea associated with Huntington’s
39. Andrich J, Arning L, Wieczorek S, et al. Huntington’s disease. Mov Disord 2007; 22 (1): 10-3
disease as caused by 34 CAG repeats. Mov Disord 2008; 59. Scherman D, Henry JP. Reserpine binding to bovine
23 (6): 879-81 chromaffin granule membranes. Characterization and

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
570 Frank & Jankovic

comparison with dihydrotetrabenazine binding. Mol of Huntington’s disease. J Natl Med Assoc 2006; 98 (8):
Pharmacol 1984; 25 (1): 113-22 1353-5
60. Mehvar R, Jamali F, Watson MW, et al. Pharmacokinetics 78. Erdemoglu AK, Boratav C. Risperidone in chorea and
of tetrabenazine and its major metabolite in man and rat. psychosis of Huntington’s disease. Eur J Neurol 2002;
Bioavailability and dose dependency studies. Drug Metab 9 (2): 182-3
Dispos 1987; 15 (2): 250-5 79. Madhusoodanan S, Brenner R. Use of risperidone in psy-
61. Roberts MS, Watson HM, McLean S, et al. Determination chosis associated with Huntington’s disease. Am J Geriatr
of therapeutic plasma concentrations of tetrabenazine and Psychiatry 1998; 6 (4): 347-9
an active metabolite by high-performance liquid chro- 80. Parsa MA, Szigethy E, Voci JM, et al. Risperidone in
matography. J Chromatogr 1981; 226 (1): 175-82 treatment of choreoathetosis of Huntington’s disease.
62. Roberts MS, McLean S, Millingen KS, et al. The phar- J Clin Psychopharmacol 1997; 17 (2): 134-5
macokinetics of tetrabenazine and its hydroxy metabolite 81. Alpay M, Koroshetz WJ. Quetiapine in the treatment of
in patients treated for involuntary movement disorders.
behavioral disturbances in patients with Huntington’s
Eur J Clin Pharmacol 1986; 29 (6): 703-8
disease. Psychosomatics 2006; 47 (1): 70-2
63. Huntington Study Group. Tetrabenazine as antichorea
82. Seitz DP, Millson RC. Quetiapine in the management of
therapy in Huntington disease: a randomized controlled
psychosis secondary to Huntington’s disease: a case re-
trial. Neurology 2006; 66 (3): 366-72
port. Can J Psychiatry 2004; 49 (6): 413
64. Jankovic J, Beach J. Long-term effects of tetrabenazine in
83. Bonelli RM, Niederwieser G. Quetiapine in Huntington’s
hyperkinetic movement disorders. Neurology 1997; 48 (2):
disease: a first case report. J Neurol 2002; 249 (8): 1114-5
358-62
65. Kenney C, Hunter C, Jankovic J. Long-term tolerability of 84. van Vugt JP, Siesling S, Vergeer M, et al. Clozapine versus
tetrabenazine in the treatment of hyperkinetic movement placebo in Huntington’s disease: a double blind random-
disorders. Mov Disord 2007; 22 (2): 193-7 ised comparative study. J Neurol Neurosurg Psychiatry
1997; 63 (1): 35-9
66. Fasano A, Cadeddu F, Guidubaldi A, et al. The long-term
effect of tetrabenazine in the management of Huntington 85. Ciammola A, Sassone J, Colciago C, et al. Aripiprazole in
disease. Clin Neuropharmacol 2008; 31 (6): 313-8 the treatment of Huntington’s disease: a case series.
Neuropsychiatr Dis Treat 2009; 5: 1-4
67. Huntington Study Group/Tetra-HD Investigators. Tetra-
benazine as anti-chorea therapy in Huntington disease: an 86. Brusa L, Orlacchio A, Moschella V, et al. Treatment of the
open-label continuation study. BMC Neurol 2009 Dec 18; symptoms of Huntington’s disease: preliminary results
9: 62 comparing aripiprazole and tetrabenazine. Mov Disord
2009; 24 (1): 126-9
68. Quinn N, Marsden CD. A double blind trial of sulpiride in
Huntington’s disease and tardive dyskinesia. J Neurol 87. Lin WC, Chou YH. Aripiprazole effects on psychosis
Neurosurg Psychiatry 1984; 47 (8): 844-7 and chorea in a patient with Huntington’s disease. Am J
Psychiatry 2008; 165 (9): 1207-8
69. Deroover J, Baro F, Bourguignon RP, et al. Tiapride
versus placebo: a double-blind comparative study in the 88. Lucetti C, Gambaccini G, Bernardini S, et al. Amantadine
management of Huntington’s chorea. Curr Med Res Opin in Huntington’s disease: open-label video-blinded study.
1984; 9 (5): 329-38 Neurol Sci 2002; 23 Suppl. 2: S83-4
70. Leonard DP, Kidson MA, Brown JG, et al. A double blind 89. Stewart JT. Adverse behavioral effects of amantadine
trial of lithium carbonate and haloperidol in Huntington’s therapy in Huntington’s disease. South Med J 1987; 80
chorea. Aust N Z J Psychiatry 1975; 9 (2): 115-8 (10): 1324-5
71. Barr AN, Fischer JH, Koller WC, et al. Serum haloperidol 90. Dosage effects of riluzole in Huntington’s disease: a
concentration and choreiform movements in Hunting- multicenter placebo-controlled study. Neurology 2003;
ton’s disease. Neurology 1988; 38 (1): 84-8 61 (11): 1551-6
72. Bonelli RM, Mahnert FA, Niederwieser G. Olanzapine for 91. Landwehrmeyer GB, Dubois B, de Yebenes JG, et al.
Huntington’s disease: an open label study. Clin Neuro- Riluzole in Huntington’s disease: a 3-year, randomized
pharmacol 2002; 25 (5): 263-5 controlled study. Ann Neurol 2007; 62 (3): 262-72
73. Bonelli RM, Niederwieser G, Tribl GG, et al. High-dose 92. Peiris JB, Boralessa H, Lionel ND. Clonazepam in the
olanzapine in Huntington’s disease. Int Clin Psycho- treatment of choreiform activity. Med J Aust 1976; 1 (8):
pharmacol 2002; 17 (2): 91-3 225-7
74. Paleacu D, Anca M, Giladi N. Olanzapine in Huntington’s 93. Frattola L, Albizzati MG, Alemani A, et al. Acute treat-
disease. Acta Neurol Scand 2002; 105 (6): 441-4 ment of Huntington’s chorea with lisuride. J Neurol Sci
75. Squitieri F, Cannella M, Piorcellini A, et al. Short-term 1983; 59 (2): 247-53
effects of olanzapine in Huntington disease. Neuro- 94. Vitale C, Marconi S, Di ML, et al. Short-term continuous
psychiatry Neuropsychol Behav Neurol 2001; 14 (1): infusion of apomorphine hydrochloride for treatment of
69-72 Huntington’s chorea: a double blind, randomized cross-
76. Dipple HC. The use of olanzapine for movement disorder over trial. Mov Disord 2007; 22 (16): 2359-64
in Huntington’s disease: a first case report. J Neurol 95. Corsini GU, Onali P, Masala C, et al. Apomorphine hydro-
Neurosurg Psychiatry 1999; 67 (1): 123-4 chloride-induced improvement in Huntington’s chorea:
77. Cankurtaran ES, Ozalp E, Soygur H, et al. Clinical ex- stimulation of dopamine receptor. Arch Neurol 1978;
perience with risperidone and memantine in the treatment 35 (1): 27-30

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
Pharmacological Management of Huntington’s Disease 571

96. Low PA, Allsop JL, Halmagyi GM. Huntington’s chorea: 111. Tedroff J, Ekesbo A, Sonesson C, et al. Long-lasting im-
the rigid form (Westphal variant) treated with levodopa. provement following (-)-OSU6162 in a patient with
Med J Aust 1974; 1 (11): 393-4 Huntington’s disease. Neurology 1999; 53 (7): 1605-6
97. Low PA, Allsop JL. Huntington’s chorea–the rigid form 112. Huntington Study Group TREND-HD Investigators.
(Westphal variant) treated with l-DOPA: a case report. Randomized controlled trial of ethyl-eicosapentaenoic
Proc Aust Assoc Neurol 1973; 10 (0): 45-6 acid in Huntington disease: the TREND-HD study. Arch
98. Magnet MK, Bonelli RM, Kapfhammer HP. Amantadine Neurol 2008; 65 (12): 1582-9
in the akinetic-rigid variant of Huntington’s disease. Ann 113. Huntington Study Group. PHEND-HD: a safety,
Pharmacother 2004; 38 (7-8): 1194-6 tolerability, and biomarker study of phenylbutyrate in
99. Bonelli RM, Niederwieser G, Diez J, et al. Pramipexole symptomatic HD [abstract]. Neurotherapeutics 2008;
ameliorates neurologic and psychiatric symptoms in a 5 (2): 363
Westphal variant of Huntington’s disease. Clin Neuro- 114. Huntington Study Group. A phase 2 trial of minocycline
pharmacol 2002; 25 (1): 58-60 in Huntington’s disease [abstract]. Mov Disord 2009;
100. Tan EK, Jankovic J, Ondo W. Bruxism in Huntington’s 24 Suppl. 1: S164
disease. Mov Disord 2000; 15 (1): 171-3 115. Yang L, Calingasan NY, Wille EJ, et al. Combination
101. Beal MF, Ferrante RJ. Experimental therapeutics in therapy with coenzyme Q10 and creatine produces ad-
transgenic mouse models of Huntington’s disease. Nat ditive neuroprotective effects in models of Parkinson’s
Rev Neurosci 2004; 5 (5): 373-84 and Huntington’s diseases. J Neurochem 2009; 109 (5):
1427-39
102. Hersch SM, Ferrante RJ. Translating therapies for Hun-
tington’s disease from genetic animal models to clinical 116. Pan T, Kondo S, Le W, et al. The role of autophagy-
trials. NeuroRx 2004; 1 (3): 298-306 lysosome pathway in neurodegeneration associated with
Parkinson’s disease. Brain 2008; 131 (Pt 8): 1969-78
103. Hersch SM, Gevorkian S, Marder K, et al. Creatine in Hun-
tington disease is safe, tolerable, bioavailable in brain and 117. Mostert JP, Koch MW, Heerings M, et al. Therapeutic
reduces serum 8OH2’dG. Neurology 2006; 66 (2): 250-2 potential of fluoxetine in neurological disorders. CNS
Neurosci Ther 2008; 14 (2): 153-64
104. Huntington Study Group. A randomized, placebo-
controlled trial of coenzyme Q10 and remacemide in 118. US National Institutes of Health. ClinicalTrials.gov [on-
Huntington’s disease. Neurology 2001; 57 (3): 397-404 line]. Available from URL: http://www.clinicaltrials.gov
[Accessed 2010 Jan 26]
105. Curtis A, Rickards H. Nabilone could treat chorea and
irritability in Huntington’s disease. J Neuropsychiatry 119. Kieburtz K, McDermott MP, Voss TS, et al. A randomized,
Clin Neurosci 2006; 18 (4): 553-4 placebo-controlled trial of latrepirdine in Huntington
disease. Arch Neurol 2010; 67 (2): 154-60
106. Consroe P, Laguna J, Allender J, et al. Controlled clinical
trial of cannabidiol in Huntington’s disease. Pharmacol 120. Wang X, Zhu S, Pei Z, et al. Inhibitors of cytochrome
Biochem Behav 1991; 40 (3): 701-8 c release with therapeutic potential for Huntington’s
disease. J Neurosci 2008; 28 (38): 9473-85
107. Muller-Vahl KR, Schneider U, Emrich HM. Nabilone in-
creases choreatic movements in Huntington’s disease. 121. Graham RK, Deng Y, Slow EJ, et al. Cleavage at the
Mov Disord 1999; 14 (6): 1038-40 caspase-6 site is required for neuronal dysfunction and
degeneration due to mutant huntingtin. Cell 2006; 125 (6):
108. Mestre T, Ferreira J, Coelho MM, et al. Therapeutic 1179-91
interventions for disease progression in Huntington’s dis-
ease. Cochrane Database Syst Rev 2009; (3): CD006455 122. Kaltenbach LS, Romero E, Becklin RR, et al. Huntingtin
interacting proteins are genetic modifiers of neurodegen-
109. Massachusetts General Hospital. Creatine Safety and Tol- eration. PLoS Genet 2007 May 11; 3 (5): e82
erability in Premanifest HD [ClinicalTrials.gov identifier
NCT00592995]. US National Institutes of Health. Clin-
ical Trials.gov [online]. Available from URL: http://
www.clinicaltrials.gov [Accessed 2010 Jan 26] Correspondence: Dr Samuel Frank, 72 East Concord St,
110. Hersch SM, Rosas HD. Neuroprotection for Huntington’s Collamore 329, Boston, MA 02118, USA.
disease: ready, set, slow. Neurotherapeutics 2008; 5 (2): 226-36 E-mail: samfrank@bu.edu

ª 2010 Adis Data Information BV. All rights reserved. Drugs 2010; 70 (5)
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

También podría gustarte