Skip to main content
Log in

Treatment of Huntington’s Disease

  • Review
  • Published:
Neurotherapeutics

Abstract

Huntington’s 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 family members 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 interventions have yielded positive results and current treatments have focused on the motor aspects of HD. Tetrabenazine 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 neuroleptic agents, such as olanzapine and aripiprazole, may have adequate efficacy with a more favorable adverse effect profile than older neuroleptic agents for treating chorea and psychosis. 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.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. The Huntington’s Disease Collaborative Research Group. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell 1993;72:971–983.

    Google Scholar 

  2. Nucifora FC Jr, Sasaki M, Peters MF, et al. Interference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicity. Science 2001;291:2423–2428.

    CAS  PubMed  Google Scholar 

  3. Subramaniam S, Sixt KM, Barrow R, Snyder SH. Rhes, a striatal specific protein, mediates mutant-huntingtin cytotoxicity. Science 2009;324:1327–1330.

    CAS  PubMed Central  PubMed  Google Scholar 

  4. Ribeiro FM, Paquet M, Ferreira LT, et al. Metabotropic glutamate receptor-mediated cell signaling pathways are altered in a mouse model of Huntington's disease. J Neurosci 2010;30:316–324.

    CAS  PubMed  Google Scholar 

  5. Andrich J, Saft C, Ostholt N, Müller T. Complex movement behavior and progression of Huntington’s disease. Neurosci Lett 2007;416:272–274.

    CAS  PubMed  Google Scholar 

  6. Frank S, Marshall F, Plumb S, et al. Functional decline due to chorea in Huntington’s disease. Neurology 2004;62(Suppl. 5):A204.

    Google Scholar 

  7. Verhagen ML, Morris MJ, Farmer C, et al. Huntington’s disease: a randomized, controlled trial using the NMDA-antagonist amantadine. Neurology 2002;59:694–699.

    Google Scholar 

  8. Bates G. Huntingtin aggregation and toxicity in Huntington’s disease. Lancet 2003;361:1642–1644.

    CAS  PubMed  Google Scholar 

  9. Arrasate M, Mitra S, Schweitzer ES, Segal MR, Finkbeiner S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature 2004;431:805–810.

    CAS  PubMed  Google Scholar 

  10. Glass M, Dragunow M, Faull RL. The 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 disease. Neuroscience 2000;97:505–519.

    CAS  PubMed  Google Scholar 

  11. Albin RL, Reiner A, Anderson KD, et al. Preferential loss of striato-external pallidal projection neurons in presymptomatic Huntington’s disease. Ann Neurol 1992;31:425–430.

    CAS  PubMed  Google Scholar 

  12. Reiner A, Albin RL, Anderson KD, D'Amato CJ, Penney JB, Young AB. Differential loss of striatal projection neurons in Huntington disease. Proc Natl Acad Sci U S A 1988;85:5733–5737.

    CAS  PubMed Central  PubMed  Google Scholar 

  13. Mitchell IJ, Cooper AJ, Griffiths MR. The selective vulnerability of striatopallidal neurons. Prog Neurobiol 1999;59:691–719.

    CAS  PubMed  Google Scholar 

  14. Antonini A, Leenders KL, Spiegel R, et al. Striatal glucose metabolism and dopamine D2 receptor binding in asymptomatic gene carriers and patients with Huntington’s disease. Brain 1996;119:2085–2095.

    PubMed  Google Scholar 

  15. Augood SJ, Faull RL, Emson PC. Dopamine D1 and D2 receptor gene expression in the striatum in Huntington’s disease. Ann Neurol 1997;42:215–221.

    CAS  PubMed  Google Scholar 

  16. Feigin A, Tang C, Ma Y, et al. Thalamic metabolism and symptom onset in preclinical Huntington’s disease. Brain 2007;130:2858–2867.

    CAS  PubMed  Google Scholar 

  17. Andrews TC, Weeks RA, Turjanski N, et al. Huntington’s disease progression. PET and clinical observations. Brain 1999;122:2353–2363.

    Google Scholar 

  18. Weeks RA, Piccini P, Harding AE, et al. Striatal D1 and D2 dopamine receptor loss in asymptomatic mutation carriers of Huntington’s disease. Ann Neurol 1996;40:49–54.

    CAS  PubMed  Google Scholar 

  19. Cha JH, Kosinski CM, Kerner JA, et al. Altered brain neurotransmitter receptors in transgenic mice expressing a portion of an abnormal human huntington disease gene. Proc Natl Acad Sci U S A 1998;95:6480–6485.

    CAS  PubMed Central  PubMed  Google Scholar 

  20. Perry TL, Hansen S, Kloster M. Huntington’s chorea. Deficiency of gamma-aminobutyric acid in brain. N Engl J Med 1973;288:337–342.

    CAS  PubMed  Google Scholar 

  21. Paulsen JS. Functional imaging in Huntington’s disease. Exp Neurol 2009;216:272–277.

    PubMed  Google Scholar 

  22. Gourfinkel-An I, Parain K, Hartmann A, et al. Changes in GAD67 mRNA expression evidenced by in situ hybridization in the brain of R6/2 transgenic mice. J Neurochem 2003;86:1369–1378.

    CAS  PubMed  Google Scholar 

  23. Nicniocaill B, Haraldsson B, Hansson O, et al. Altered striatal amino acid neurotransmitter release monitored using microdialysis in R6/1 Huntington transgenic mice. Eur J Neurosci 2001;13:206–210.

    CAS  PubMed  Google Scholar 

  24. Li SH, Li XJ. Huntingtin-protein interactions and the pathogenesis of Huntington’s disease. Trends Genet 2004;20:146–154.

    PubMed  Google Scholar 

  25. Bence NF, Sampat RM, Kopito RR. Impairment of the ubiquitin-proteasome system by protein aggregation. Science 2001;292:1552–1555.

    CAS  PubMed  Google Scholar 

  26. Ravikumar B, Duden R, Rubinsztein DC. Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Hum Mol Genet 2002;11:1107–1117.

    CAS  PubMed  Google Scholar 

  27. Cha JH. Transcriptional dysregulation in Huntington’s disease. Trends Neurosci 2000;23:387–392.

    CAS  PubMed  Google Scholar 

  28. Cepeda C, Hurst RS, Calvert CR, et al. Transient and progressive electrophysiological alterations in the corticostriatal pathway in a mouse model of Huntington’s disease. J Neurosci 2003;23:961–969.

    CAS  PubMed  Google Scholar 

  29. Ludolph AC, He F, Spencer PS, Hammerstad J, Sabri M. 3-Nitropropionic acid-exogenous animal neurotoxin and possible human striatal toxin. Can J Neurol Sci 1991;18:492–498.

    CAS  PubMed  Google Scholar 

  30. Gunawardena S, Goldstein LS. Polyglutamine diseases and transport problems: deadly traffic jams on neuronal highways. Arch Neurol 2005;62:46–51.

    PubMed  Google Scholar 

  31. Harper PS. The epidemiology of Huntington’s disease. Hum Genet 1992;89:365–376.

    CAS  PubMed  Google Scholar 

  32. Morrison PJ, Johnston WP, Nevin NC. The epidemiology of Huntington’s disease in Northern Ireland. J Med Genet 1995;32:524–530.

    CAS  PubMed  Google Scholar 

  33. Peterlin B, Kobal J, Teran N, et al. Epidemiology of Huntington’s disease in Slovenia. Acta Neurol Scand 2009;119:371–375.

    CAS  PubMed  Google Scholar 

  34. Alonso ME, Ochoa A, Boll MC, et al. Clinical and genetic characteristics of Mexican Huntington's disease patients. Mov Disord 2009;24:2012–2015.

    PubMed  Google Scholar 

  35. Simpson SA, Johnston AW. The prevalence and patterns of care of Huntington’s chorea in Grampian. Br J Psychiatry 1989;155:799–804.

    CAS  PubMed  Google Scholar 

  36. Penney JB Jr, Young AB, Shoulson I, et al. Huntington’s disease in Venezuela: 7 years of follow-up on symptomatic and asymptomatic individuals. Mov Disord 1990;5:93–99.

    PubMed  Google Scholar 

  37. Harper PS. The epidemiology of Huntington’s disease. In: Bates G, Harper P, Jones L, eds. Huntington’s disease. 3rd ed. Oxford Monographs on Medical Genetics, 2002.

  38. Ribai P, Nguyen K, Hahn-Barma V, et al. Psychiatric and cognitive difficulties as indicators of juvenile huntington disease onset in 29 patients. Arch Neurol 2007;64:813–819.

    PubMed  Google Scholar 

  39. Di ML, Squitieri F, Napolitano G, Campanella G, Trofatter JA, Conneally PM. Suicide risk in Huntington’s disease. J Med Genet 1993;30:293–295.

    Google Scholar 

  40. Pollard J. Hurry up and wait: a cognitive care companion. 9/25/08. XX, Lulu Press, US, 2008. http://www.lulu.com/shop/james-pollard/hurry-up-and-wait/paperback/product-3610588.html#productDetails

  41. Cubo E, Shannon KM, Tracy D, et al. Effect of donepezil on motor and cognitive function in Huntington disease. Neurology 2006;67:1268–1271.

    CAS  PubMed  Google Scholar 

  42. de Tommaso M, Difruscolo O, Sciruicchio V, Specchio N, Livrea P. Two years’ follow-up of rivastigmine treatment in Huntington disease. Clin Neuropharmacol 2007;30:43–46.

    PubMed  Google Scholar 

  43. Beglinger LJ, Adams WH, Paulson H, et al. Randomized controlled trial of atomoxetine for cognitive dysfunction in early Huntington disease. J Clin Psychopharmacol 2009;29:484–487.

    CAS  PubMed Central  PubMed  Google Scholar 

  44. Paulsen Cognitive impairment in Huntington disease: diagnosis and treatment. Curr Neurol Neurosci Rep 2011;11:474–483.

  45. Pidgeon C, Rickards H. The pathophysiology and pharmacological treatment of Huntington disease. Behav Neurol 2013;26:245–253.

    PubMed  Google Scholar 

  46. Armstrong MJ, Miyasaki JM; American Academy of Neurology. Evidence-based guideline: pharmacologic treatment of chorea in Huntington disease: report of the guideline development subcommittee of the American Academy of Neurology. Neurology 2012;79:597–603.

    Google Scholar 

  47. Paleacu D. Tetrabenazine in the treatment of Huntington’s Disease. Neuropsychiatr Dis Treat 2007;3:545–551.

    CAS  PubMed Central  PubMed  Google Scholar 

  48. Bagchi SP. Differential interactions of phencyclidine with tetrabenazine and reserpine affecting intraneuronal dopamine. Biochem Pharmacol 1983;32:2851–2856.

    CAS  PubMed  Google Scholar 

  49. Adam OR, Jankovic J. Symptomatic treatment of Huntington disease. Neurotherapeutics 2008;5:181–197.

    CAS  PubMed  Google Scholar 

  50. Phillips W, Shannon KM, Barker RA. The current clinical management of Huntington’s disease. Mov Disord 2008;23:1491–1504.

    PubMed  Google Scholar 

  51. Roze E, Saudou F, Caboche J. Pathophysiology of Huntington’s disease: from huntingtin functions to potential treatments. Curr Opin Neurol 2008;21:497–503.

    CAS  PubMed  Google Scholar 

  52. Imarisio S, Carmichael J, Korolchuk V, et al. Huntington’s disease: from pathology and genetics to potential therapies. Biochem J 2008;412:191–209.

    CAS  PubMed  Google Scholar 

  53. Walker FO. Huntington’s disease. Lancet 2007;369:218–228.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  55. Bonelli RM, Wenning GK. Pharmacological management of Huntington’s disease: an evidence-based review. Curr Pharm Des 2006;12:2701–2720.

    CAS  PubMed  Google Scholar 

  56. Mestre T, Ferreira J, Coelho MM, Rosa M, Sampaio C. Therapeutic interventions for symptomatic treatment in Huntington’s disease. Cochrane Database Sys Rev 2009:CD006456.

  57. Nakamura K, Aminoff MJ. Huntington’s disease: clinical characteristics, pathogenesis and therapies. Drugs Today (Barc) 2007;43:97–116.

    CAS  Google Scholar 

  58. Handley OJ, Naji JJ, Dunnett SB, Rosser AE. Pharmaceutical, cellular and genetic therapies for Huntington’s disease. Clin Sci (Lond) 2006;110:73–88.

    CAS  Google Scholar 

  59. Bonelli RM, Hofmann P. A review of the treatment options for Huntington’s disease. Expert Opin Pharmacother 2004;5:767–776.

    CAS  PubMed  Google Scholar 

  60. Grimbergen YA, Roos RA. Therapeutic options for Huntington’s disease. Curr Opin Investig Drugs 2003;4:51–54.

    CAS  PubMed  Google Scholar 

  61. Frank SA, Jankovic J. Advances in the pharmacological management of Huntington’s Disease. Drugs 2010;70:561–571.

    CAS  PubMed  Google Scholar 

  62. Jankovic J, Clarence-Smith K. Tetrabenazine for the treatment of chorea and other hyperkinetic movement disorders. Expert Rev Neurother 2011;11:1509–1523.

    CAS  PubMed  Google Scholar 

  63. Pettibone DJ, Pflueger AB, Totaro JA. Tetrabenazine-induced depletion of brain monoamines: mechanism by which desmethylimipramine protects cortical norepinephrine. Eur J Pharmacol 1984;102:431–436.

    CAS  PubMed  Google Scholar 

  64. Mehvar R, Jamali F. Concentration-effect relationships of tetrabenazine and dihydrotetrabenazine in the rat. J Pharm Sci 1987;76:461–465.

    CAS  PubMed  Google Scholar 

  65. Thibaut F, Faucheux BA, Marquez J, et al. Regional distribution of monoamine vesicular uptake sites in the mesencephalon of control subjects and patients with Parkinson’s disease: a postmortem study using tritiated tetrabenazine. Brain Res 1995;692:233–243.

    CAS  PubMed  Google Scholar 

  66. Kenney C, Hunter C, Davidson A, Jankovic J. Short-term effects of tetrabenazine on chorea associated with Huntington’s disease. Mov Disord 2007;22:10–13.

    PubMed  Google Scholar 

  67. Scherman D, Henry JP. Reserpine binding to bovine chromaffin granule membranes. Characterization and comparison with dihydrotetrabenazine binding. Mol Pharmacol 1984;25:113–122.

    CAS  PubMed  Google Scholar 

  68. Huntington Study Group. Tetrabenazine as antichorea therapy in Huntington disease: a randomized controlled trial. Neurology 2006;66:366–372.

    Google Scholar 

  69. Jankovic J, Beach J. Long-term effects of tetrabenazine in hyperkinetic movement disorders. Neurology 1997;48:358–362.

    CAS  PubMed  Google Scholar 

  70. Kenney C, Hunter C, Jankovic J. Long-term tolerability of tetrabenazine in the treatment of hyperkinetic movement disorders. Mov Disord 2007;22:193–197.

    PubMed  Google Scholar 

  71. Fasano A, Cadeddu F, Guidubaldi A, et al. The long-term effect of tetrabenazine in the management of Huntington disease. Clin Neuropharmacol 2008;31:313–318.

    CAS  PubMed  Google Scholar 

  72. Frank S. Tetrabenazine as anti-chorea therapy in Huntington disease: an open-label continuation study. Huntington Study Group/TETRA-HD Investigators. BMC Neurol 2009;9:62.

    Google Scholar 

  73. Xenazine. Product information, September 2012. http://www.lundbeck.com/upload/us/files/pdf/Products/Xenazine_PI_US_EN.pdf

  74. Mehanna R, Hunter C, Davidson A, Jimenez-Shahed J, Jankovic J. Analysis of CYP2D6 genotype and response to tetrabenazine. Mov Disord 2013;28:210–215.

    CAS  PubMed  Google Scholar 

  75. Quinn N, Marsden CD. A double blind trial of sulpiride in Huntington’s disease and tardive dyskinesia. J Neurol Neurosurg Psychiatry 1984;47:844–847.

    CAS  PubMed  Google Scholar 

  76. Deroover J, Baro F, Bourguignon RP, Smets P. Tiapride versus placebo: a double-blind comparative study in the management of Huntington’s chorea. Curr Med Res Opin 1984;9:329–338.

    CAS  PubMed  Google Scholar 

  77. Leonard DP, Kidson MA, Brown JG, Shannon PJ, Taryan S. A double blind trial of lithium carbonate and haloperidol in Huntington’s chorea. Aust N Z J Psychiatry 1975;9:115–118.

    CAS  PubMed  Google Scholar 

  78. Barr AN, Fischer JH, Koller WC, Spunt AL, Singhal A. Serum haloperidol concentration and choreiform movements in Huntington’s disease. Neurology 1988;38:84–88.

    CAS  PubMed  Google Scholar 

  79. Bonelli RM, Mahnert FA, Niederwieser G. Olanzapine for Huntington’s disease: an open label study. Clin Neuropharmacol 2002;25:263–265.

    CAS  PubMed  Google Scholar 

  80. Bonelli RM, Niederwieser G, Tribl GG, Költringer P. High-dose olanzapine in Huntington’s disease. Int Clin Psychopharmacol 2002;17:91–93.

    CAS  PubMed  Google Scholar 

  81. Paleacu D, Anca M, Giladi N. Olanzapine in Huntington’s disease. Acta Neurol Scand 2002;105:441–444.

    CAS  PubMed  Google Scholar 

  82. Squitieri F, Cannella M, Piorcellini A, Brusa L, Simonelli M, Ruggieri S. Short-term effects of olanzapine in Huntington disease. Neuropsychiatry Neuropsychol Behav Neurol 2001;14:69–72.

    CAS  PubMed  Google Scholar 

  83. Dipple HC. The use of olanzapine for movement disorder in Huntington’s disease: a first case report. J Neurol Neurosurg Psychiatry 1999;67:123–124.

    CAS  PubMed  Google Scholar 

  84. Cankurtaran ES, Ozalp E, Soygur H, Cakir A. Clinical experience with risperidone and memantine in the treatment of Huntington’s disease. J Natl Med Assoc. 2006;98:1353–1355.

    PubMed  Google Scholar 

  85. Erdemoglu AK, Boratav C. Risperidone in chorea and psychosis of Huntington’s disease. Eur J Neurol 2002;9:182–183.

    PubMed  Google Scholar 

  86. Madhusoodanan S, Brenner R. Use of risperidone in psychosis associated with Huntington’s disease. Am J Geriatr Psychiatry 1998;6:347–349.

    CAS  PubMed  Google Scholar 

  87. Parsa MA, Szigethy E, Voci JM, Meltzer HY. Risperidone in treatment of choreoathetosis of Huntington’s disease. J Clin Psychopharmacol 1997;17:134–135.

    CAS  PubMed  Google Scholar 

  88. Alpay M, Koroshetz WJ. Quetiapine in the treatment of behavioral disturbances in patients with Huntington’s disease. Psychosomatics 2006;47:70–72.

    PubMed  Google Scholar 

  89. Seitz DP, Millson RC. Quetiapine in the management of psychosis secondary to Huntington’s disease: a case report. Can J Psychiatry 2004;49:413.

    PubMed  Google Scholar 

  90. Bonelli RM, Niederwieser G. Quetiapine in Huntington’s disease: a first case report. J Neurol 2002;249:1114–1115.

    PubMed  Google Scholar 

  91. van Vugt JP, Siesling S, Vergeer M, van der Velde EA, Roos RA. Clozapine versus placebo in Huntington’s disease: a double blind randomised comparative study. J Neurol Neurosurg Psychiatry 1997;63:35–39.

    PubMed  Google Scholar 

  92. Ciammola A, Sassone J, Colciago C, et al. Aripiprazole in the treatment of Huntington’s disease: a case series. Neuropsychiatr Dis Treat 2009;5:1–4.

    CAS  PubMed Central  PubMed  Google Scholar 

  93. Brusa L, Orlacchio A, Moschella V, Iani C, Bernardi G, Mercuri NB. Treatment of the symptoms of Huntington’s disease: preliminary results comparing aripiprazole and tetrabenazine. Mov Disord 2009;24:126–129.

    PubMed  Google Scholar 

  94. Lin WC, Chou YH. Aripiprazole effects on psychosis and chorea in a patient with Huntington’s disease. Am J Psychiatry 2008;165:1207–1208.

    PubMed  Google Scholar 

  95. Lucetti C, Gambaccini G, Bernardini S, et al. Amantadine in Huntington’s disease: open-label video-blinded study. Neurol Sci 2002;23(Suppl. 2):S83-S84.

    PubMed  Google Scholar 

  96. Stewart JT. Adverse behavioral effects of amantadine therapy in Huntington’s disease. South Med J 1987;80:1324–1325.

    CAS  PubMed  Google Scholar 

  97. Huntington Study Group. Dosage effects of riluzole in Huntington’s disease: a multicenter placebo-controlled study. Neurology 2003;61:1551–1556.

    Google Scholar 

  98. Landwehrmeyer GB, Dubois B, de Yebenes JG, et al. Riluzole in Huntington’s disease: a 3-year, randomized controlled study. Ann Neurol 2007;62:262–272.

    CAS  PubMed  Google Scholar 

  99. Peiris JB, Boralessa H, Lionel ND. Clonazepam in the treatment of choreiform activity. Med J Aust 1976;1:225–227.

    CAS  PubMed  Google Scholar 

  100. Frattola L, Albizzati MG, Alemani A, Bassi S, Ferrarese C, Trabucchi M. Acute treatment of Huntington’s chorea with lisuride. J Neurol Sci 1983;59:247–253.

    CAS  PubMed  Google Scholar 

  101. Vitale C, Marconi S, Di ML, et al. Short-term continuous infusion of apomorphine hydrochloride for treatment of Huntington’s chorea: a double blind, randomized cross-over trial. Mov Disord 2007;22:2359–2364.

    PubMed  Google Scholar 

  102. Corsini GU, Onali P, Masala C, Cianchetti C, Mangoni A, Gessa G. Apomorphine hydrochloride-induced improvement in Huntington’s chorea: stimulation of dopamine receptor. Arch Neurol 1978;35:27–30.

    CAS  PubMed  Google Scholar 

  103. Low PA, Allsop JL, Halmagyi GM. Huntington’s chorea: the rigid form (Westphal variant) treated with levodopa. Med J Aust 1974;1:393–394.

    CAS  PubMed  Google Scholar 

  104. Low PA, Allsop JL. Huntington’s chorea—the rigid form (Westphal variant) treated with l-DOPA: a case report. Proc Aust Assoc Neurol 1973;10:45–46.

    CAS  PubMed  Google Scholar 

  105. Magnet MK, Bonelli RM, Kapfhammer HP. Amantadine in the akinetic-rigid variant of Huntington’s disease. Ann Pharmacother 2004;38:1194–1196.

    PubMed  Google Scholar 

  106. Bonelli RM, Niederwieser G, Diez J, Gruber A, Költringer P. Pramipexole ameliorates neurologic and psychiatric symptoms in a Westphal variant of Huntington’s disease. Clin Neuropharmacol 2002;25:58–60.

    PubMed  Google Scholar 

  107. Groves M, van Duijn E, Anderson K, et al. An international survey-based algorithm for the pharmacologic treatment of irritability in Huntington's disease. PLoS Curr 2011;30:3.

    Google Scholar 

  108. Anderson K, Craufurd D, Edmondson MC, et al. An international survey-based algorithm for the pharmacologic treatment of obsessive-compulsive behaviors in Huntington's disease. PLoS Curr 2011;20:3.

    Google Scholar 

  109. www.clinicaltrials.gov. Accessed August 9, 2013.

  110. Kieburtz K, McDermott MP, Voss TS, et al. A randomized, placebo-controlled trial of latrepirdine in Huntington disease. Arch Neurol 2010;67:154–160.

    PubMed  Google Scholar 

  111. Huntington Study Group TREND-HD Investigators. Randomized controlled trial of ethyl-eicosapentaenoic acid in Huntington disease: the TREND-HD study. Arch Neurol 2008;65:1582–1589.

    Google Scholar 

  112. Curtis A, Rickards H. Nabilone could treat chorea and irritability in Huntington's disease. J Neuropsychiatry Clin Neurosci 2006;18:553–554.

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Samuel Frank.

Electronic supplementary material

Below is the link to the electronic supplementary material.

ESM 1

(PDF 1210 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Frank, S. Treatment of Huntington’s Disease. Neurotherapeutics 11, 153–160 (2014). https://doi.org/10.1007/s13311-013-0244-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13311-013-0244-z

Key Words

Navigation