420
Views
5
CrossRef citations to date
0
Altmetric
Review

State-of-the-art pharmacological approaches to reduce chorea in Huntington’s disease

&
Pages 1015-1024 | Received 04 Nov 2020, Accepted 12 Jan 2021, Published online: 08 Feb 2021

References

  • Huntington G. On chorea. J Neuropsychiatry Clin Neurosci. 2003;15(1):109–112.
  • Galvan L, André VM, Wang EA, et al. Functional differences between direct and indirect striatal output pathways in Huntington’s disease. J Huntingtons Dis. 2012;1(1):17–25.
  • Albin RL, Reiner A, Anderson KD, et al. Striatal and nigral neuron subpopulations in rigid Huntington’s disease: implications for the functional anatomy of chorea and rigidity-akinesia. Ann Neurol. 1990;27(4):357–365.
  • Ghosh R, Tabrizi SJ. Clinical features of Huntington’s disease. Adv Exp Med Biol. 2018;1049:1–28.
  • van de Zande NA, Massey TH, McLauchlan D, et al. Clinical characterization of dystonia in adult patients with Huntington’s disease. Eur J Neurol. 2017;24(9):1140–1147.
  • Squitieri F, Berardelli A, Nargi E, et al. Atypical movement disorders in the early stages of Huntington’s disease: clinical and genetic analysis. Clin Genet. 2000;58(1):50–56.
  • Racette BA, Perlmutter JS. Levodopa responsive parkinsonism in an adult with Huntington’s disease. J Neurol Neurosurg Psychiatry. 1998;65(4):577–579.
  • Thompson PD, Bhatia KP, Brown P, et al. Cortical myoclonus in Huntington’s disease. Mov Disord. 1994;9(6):633–641.
  • Cui SS, Ren RJ, Wang Y, et al. Tics as an initial manifestation of juvenile Huntington’s disease: case report and literature review. BMC Neurol. 2017;17(1):152.
  • Bhidayasiri R, Truong DD. Chorea and related disorders. Postgrad Med J. 2004;80(947):527–534.
  • Sherman CW, Iyer R, Abler V, et al. Perceptions of the impact of chorea on health-related quality of life in Huntington disease (HD): a qualitative analysis of individuals across the HD spectrum, family members, and clinicians. Neuropsychol Rehabil. 2020;30(6):1150–1168.
  • Thorley EM, Iyer RG, Wicks P, et al. Understanding how chorea affects health-related quality of life in Huntington disease: an online survey of patients and caregivers in the United States. Patient. 2018;11(5):547–559.
  • Sitek EJ, Thompson JC, Craufurd D, et al. Unawareness of deficits in Huntington’s disease. J Huntingtons Dis. 2014;3(2):125–135.
  • Isaacs D, Gibson JS, Stovall J, et al. The impact of anosognosia on clinical and patient-reported assessments of psychiatric symptoms in Huntington’s disease. J Huntingtons Dis. 2020;9(3):291-302.
  • Simpson JA, Lovecky D, Kogan J, et al. Survey of the Huntington’s disease patient and caregiver community reveals most impactful symptoms and treatment needs. J Huntingtons Dis. 2016;5(4):395–403.
  • Unified Huntington’s Disease Rating Scale: reliability and consistency. Huntington study group. Mov Disord. 1996 Mar;11(2):136–142.
  • 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(2):93–99.
  • Huntington Study Group. Tetrabenazine as antichorea therapy in Huntington disease: a randomized controlled trial. Neurology. 2006 Feb 14;66(3):366–372.
  • Frank S, Testa CM, Stamler D, et al. Effect of deutetrabenazine on chorea among patients with Huntington disease: a randomized clinical trial. Jama. 2016;316(1):40–50.
  • Reilmann R, Bohlen S, Kirsten F, et al. Assessment of involuntary choreatic movements in Huntington’s disease–toward objective and quantitative measures. Mov Disord. 2011;26(12):2267–2273.
  • de Boo G, Tibben A, Hermans J, et al. Subtle involuntary movements are not reliable indicators of incipient Huntington’s disease. Mov Disord. 1998;13(1):96–99.
  • Mestre TA, Forjaz MJ, Mahlknecht P, et al. Rating scales for motor symptoms and signs in Huntington’s disease: critique and recommendations. Mov Disord Clin Pract. 2018;5(2):111–117.
  • Johnson EB, Rees EM, Labuschagne I, et al. The impact of occipital lobe cortical thickness on cognitive task performance: an investigation in Huntington’s Disease. Neuropsychologia. 2015;79(Pt A):138–146.
  • Tabrizi SJ, Reilmann R, Roos RA, et al. Potential endpoints for clinical trials in premanifest and early Huntington’s disease in the TRACK-HD study: analysis of 24 month observational data. Lancet Neurol. 2012;11(1):42–53.
  • Rosenblatt A, Ranen NG, Nance MA, et al. A physician’s guide to the management of Huntington’s disease. New York: Huntington’s Disease Society of America; 1999.
  • Fish DR, Sawyers D, Allen PJ, et al. The effect of sleep on the Dyskinetic movements of Parkinson’s Disease, Gilles de la Tourette Syndrome, Huntington’s disease, and Torsion Dystonia. Arch Neurol. 1991;48(2):210–214.
  • Hurelbrink CB, Lewis SJ, Barker RA. The use of the Actiwatch-Neurologica system to objectively assess the involuntary movements and sleep-wake activity in patients with mild-moderate Huntington’s disease. J Neurol. 2005;252(6):642–647.
  • Ranjan S, Kohler S, Harrison MB, et al. Nocturnal post-arousal chorea and repetitive ballistic movement in Huntington’s disease. Mov Disord Clin Pract. 2016;3(2):200–202.
  • Goodman AO, Rogers L, Pilsworth S, et al. Asymptomatic sleep abnormalities are a common early feature in patients with Huntington’s disease. Curr Neurol Neurosci Rep. 2011;11(2):211–217.
  • Andrzejewski KL, Dowling AV, Stamler D, et al. Wearable sensors in Huntington disease: a pilot study. J Huntingtons Dis. 2016;5(2):199–206.
  • Gordon MF, Grachev ID, Mazeh I, et al. Quantification of motor function in Huntington disease patients using wearable sensor devices. Digital Biomarkers. 2019;3(3):103–115.
  • Adams JL, Dinesh K, Xiong M, et al. Multiple wearable sensors in Parkinson and Huntington disease individuals: a pilot study in clinic and at home. Digital Biomarkers. 2017;1(1):52–63.
  • Waugh JL, Miller VS, Chudnow RS, et al. Juvenile Huntington disease exacerbated by methylphenidate: case report. J Child Neurol. 2008;23(7):807–809.
  • Hamilton CJ, Timmer TK, Munjal RC, et al. Worsening choreoathetosis in Huntington’s disease with Fluoxetine, Lisdexamfetamine, and Melatonin: a case report. Innov Clin Neurosci. 2018;15(7–8):27–31.
  • Schultz JL, Kamholz JA, Moser DJ, et al. Substance abuse may hasten motor onset of Huntington disease: evaluating the Enroll-HD database. Neurology. 2017;88(9):909–915.
  • Glidden AM, Luebbe EA, Elson MJ, et al. Patient-reported impact of symptoms in Huntington disease: PRISM-HD. Neurology. 2020;94(19):e2045-e2053.
  • Harris KL, Kuan WL, Mason SL, et al. Antidopaminergic treatment is associated with reduced chorea and irritability but impaired cognition in Huntington’s disease (Enroll-HD). J Neurol Neurosurg Psychiatry. 2020;91(6):622-630.
  • Diehl SK, Mefferd AS, Lin YC, et al. Motor speech patterns in Huntington disease. Neurology. 2019;93(22):e2042–e52.
  • Sung VW, Iyer RG, Gandhi SK, et al. Physician perceptions of pharmacologic treatment options for chorea associated with Huntington disease in the United States. Curr Med Res Opin. 2018;34(4):643–648.
  • Priller J, Ecker D, Landwehrmeyer B, et al. Europe-wide assessment of current medication choices in Huntington’s disease. Mov Disord. 2008;23(12):1788.
  • Videnovic A. Treatment of huntington disease. Curr Treat Options Neurol. 2013;15(4):424–438.
  • McCue RE, Waheed R, Urcuyo L, et al. Comparative effectiveness of second-generation antipsychotics and haloperidol in acute schizophrenia. Br J Psychiatry. 2006;189(5):433–440.
  • Martino D, Karnik V, Osland S, et al. Movement disorders associated with antipsychotic medication in people with Schizophrenia: an overview of Cochrane reviews and meta-analysis. Can J Psychiatry. 2018;63(11):706743718777392.
  • Quinn N, Marsden CD. A double blind trial of sulpiride in Huntington’s disease and tardive dyskinesia. J Neurol Neurosurg Psychiatry. 1984;47(8):844–847.
  • Deroover J, Baro F, Bourguignon RP, et al. Tiapride versus placebo: a double-blind comparative study in the management of Huntington’s chorea. Curr Med Res Opin. 1984;9(5):329–338.
  • Girotti F, Carella F, Scigliano G, et al. Effect of neuroleptic treatment on involuntary movements and motor performances in Huntington’s disease. J Neurol Neurosurg Psychiatry. 1984;47(8):848–852.
  • Desamericq G, Dolbeau G, Verny C, et al. Effectiveness of anti-psychotics and related drugs in the Huntington French-speaking group cohort. PLoS One. 2014;9(1):e85430.
  • Cankurtaran ES, Ozalp E, Soygur H, et al. Clinical experience with risperidone and memantine in the treatment of Huntington’s disease. J Natl Med Assoc. 2006;98(8):1353–1355.
  • Dallocchio C, Buffa C, Tinelli C, et al. Effectiveness of risperidone in Huntington chorea patients. J Clin Psychopharmacol. 1999;19(1):101–103.
  • Duff K, Beglinger LJ, O’Rourke ME, et al. Risperidone and the treatment of psychiatric, motor, and cognitive symptoms in Huntington’s disease. Ann Clin Psychiatry. 2008;20(1):1–3.
  • Erdemoglu AK, Boratav C. Risperidone in chorea and psychosis of Huntington’s disease. Eur J Neurol. 2002;9(2):182–183.
  • Johnston TG. Risperidone long-acting injection and Huntington’s disease: case series with significant psychiatric and behavioural symptoms. Int Clin Psychopharmacol. 2011;26(2):114–119.
  • Meco G, Bonifati V, Alessandri A, et al. Risperidone in Huntington's disease. Human Psychopharmacology: Clinical and Experimental. 1995;10(4):353–354.
  • Parsa MA, Szigethy E, Voci JM, et al. Risperidone in treatment of choreoathetosis of Huntington’s disease. J Clin Psychopharmacol. 1997;17(2):134–135.
  • Reveley MA, Dursun SM, Andrews H. A comparative trial use of sulpiride and risperidone in Huntington’s disease: a pilot study. J Psychopharmacol. 1996;10(2):162–165.
  • Bogelman G, Hirschmann S, Modai I. Olanzapine and Huntington’s disease. J Clin Psychopharmacol. 2001;21(2):245–246.
  • Bonelli RM, Niederwieser G, Tribl GG, et al. High-dose olanzapine in Huntington’s disease. Int Clin Psychopharmacol. 2002;17(2):91–93.
  • Bonelli RM, Mahnert FA, Niederwieser G. Olanzapine for Huntington’s disease: an open label study. Clin Neuropharmacol. 2002;25(5):263–265.
  • Bonelli RM, Niederwieser G, Diez J, et al. Riluzole and olanzapine in Huntington’s disease. Eur J Neurol. 2002;9(2):183–184.
  • Dipple HC. The use of olanzapine for movement disorder in Huntington’s disease: a first case report. J Neurol Neurosurg Psychiatry. 1999;67(1):123–124.
  • Grove VE Jr., Quintanilla J, DeVaney GT. Improvement of Huntington’s disease with olanzapine and valproate. N Engl J Med. 2000;343(13):973–974.
  • Laks J, Rocha M, Capitao C, et al. Functional and motor response to low dose olanzapine in Huntington’s disease: case report. Arq Neuropsiquiatr. 2004;62(4):1092–1094.
  • Paleacu D, Anca M, Giladi N. Olanzapine in Huntington’s disease. Acta Neurol Scand. 2002;105(6):441–444.
  • Skocic M, Dujmovic J, Jevtovic S, et al. Premorbid combat related ptsd in Huntington’s disease - Case report. Psychiatr Danub. 2010;22(2):286–288.
  • Squitieri F, Cannella M, Porcellini A, et al. Short-term effects of olanzapine in Huntington disease. Neuropsychiatry Neuropsychol Behav Neurol. 2001;14(1):69–72.
  • Brusa L, Orlacchio A, Moschella V, et al. Treatment of the symptoms of Huntington’s disease: preliminary results comparing aripiprazole and tetrabenazine. Mov Disord. 2009;24(1):126–129.
  • 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.
  • Edlinger M, Seppi K, Fleischhacker W, et al. Treatment of psychotic and behavioral symptoms with clozapine, aripiprazole, and reboxetine in a patient with Huntington’s disease. Int Clin Psychopharmacol. 2013;28(4):214–216.
  • Lin WC, Chou YH. Aripiprazole effects on psychosis and chorea in a patient with Huntington’s disease. Am J Psychiatry. 2008;165(9):1207–1208.
  • Oulis P, Mourikis I, Konstantakopoulos G, et al. Aripiprazole in the treatment of olanzapine-resistant psychotic and motor symptoms of Huntington’s disease. J Neuropsychiatry Clin Neurosci. 2010;22(3):352c.e4–52.e5.
  • Yavuz KF, Ulusoy S, Alniak I. Aripiprazole treatment for choreoathetoid and psychotic symptoms of Huntington’s disease. J Neuropsychiatry Clin Neurosci. 2013;25(2):E31.
  • Ding J, Gadit AM. Psychosis with Huntington’s disease: role of antipsychotic medications. BMJ Case Rep. 2014;2014:bcr2013202625.
  • Madhusoodanan S, Brenner R. Use of risperidone in psychosis associated with Huntington’s disease. Am J Geriatr Psychiatry England. 1998;347–349.
  • Xu C, Yogaratnam J, Tan N, et al. Psychosis, treatment emergent extrapyramidal events, and subsequent onset of Huntington’s disease: a case report and review of the literature. Clin Psychopharmacol Neurosci. 2016;14(3):302–304.
  • Li P, Snyder GL, Vanover KE. Dopamine targeting drugs for the treatment of Schizophrenia: past, present and future. Curr Top Med Chem. 2016;16(29):3385–3403.
  • Arciniegas DB. Psychosis. Continuum (Minneap Minn). 2015;21(3Behavioral Neurology and Neuropsychiatry):715–736.
  • Morigaki R, Goto S. Striatal Vulnerability in Huntington’s Disease: neuroprotection Versus Neurotoxicity. Brain Sci. 2017;7(6):63.
  • American Diabetes Association. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27(2):596–601.
  • Fang F, Sun H, Wang Z, et al. Antipsychotic drug-induced somnolence: incidence, mechanisms, and management. CNS Drugs. 2016;30(9):845–867.
  • Howard AK, Krishnamoorthy A, Leavitt BR, et al. Treatment of Huntington disease and Comorbid Trichotillomania with aripiprazole. J Neuropsychiatry Clin Neurosci. 2015;27(3):e211–2.
  • Lazowski LK, Townsend B, Hawken ER, et al. Sleep architecture and cognitive changes in olanzapine-treated patients with depression: a double blind randomized placebo controlled trial. BMC Psychiatry. 2014;17(14):202.
  • Pae CU, Serretti A, Patkar AA, et al. Aripiprazole in the treatment of depressive and anxiety disorders: a review of current evidence. CNS Drugs. 2008;22(5):367–388.
  • Van Vugt JPP, Siesling S, Vergeer M, et al. Clozapine versus placebo in Huntington’s disease: a double blind randomised comparative study. J Neurol Neurosurg. 1997;63(1):35–39.
  • Caine ED, Polinsky RJ, Kartzinel R, et al. The trial use of clozapine for abnormal involuntary movement disorders. Am J Psychiatry. 1979;136(3):317-320.
  • Alpay M, Koroshetz WJ. Quetiapine in the treatment of behavioral disturbances in patients with Huntington’s disease. Psychosomatics. 2006;47(1):70–72.
  • Bonelli RM, Niederwieser G. Quetiapine in Huntington’s disease: a first case report. J Neurol. 2002;249(8):1114–1115.
  • Seitz DP, Millson RC. Quetiapine in the management of psychosis secondary to huntington’s disease: a case report. Can J Psychiatry. 2004;49(6):413.
  • Koch J, Shi WX, Dashtipour K. VMAT2 inhibitors for the treatment of hyperkinetic movement disorders. Pharmacol Ther. 2020;212:107580.
  • Schuldiner S, Liu Y, Edwards RH. Reserpine binding to a vesicular amine transporter expressed in Chinese hamster ovary fibroblasts. J Biol Chem. 1993;268(1):29–34.
  • Schultz JL, Killoran A, Nopoulos PC, et al. Evaluating depression and suicidality in tetrabenazine users with Huntington disease. Neurology. 2018;91(3):e202–e07.
  • Dorsey ER, Brocht AF, Nichols PE, et al. Depressed mood and suicidality in individuals exposed to tetrabenazine in a large Huntington disease observational study. J Huntingtons Dis. 2013;2(4):509–515.
  • Xenazine. Package insert. Deerfield, Il: Lundbeck; 2019.
  • AUSTEDO- deutetrabenazine tablet, coated [package insert]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/208082s000lbl.pdf
  • Claassen DO, Carroll B, De Boer LM, et al. Indirect tolerability comparison of Deutetrabenazine and Tetrabenazine for Huntington disease. J Clin Mov Disord. 2017;4(1):3.
  • Ingrezza. Package insert. San Diego, CA: Neurocrine Biosciences, Inc; 2019.
  • Rodrigues FB, Duarte GS, Costa J, et al. Tetrabenazine Versus Deutetrabenazine for Huntington’s Disease: twins or Distant Cousins? Mov Disord Clin Pract. 2017;4(4):582–585.
  • Frank S, Stamler D, Kayson E, et al. Safety of converting from tetrabenazine to deutetrabenazine for the treatment of Chorea. JAMA Neurol. 2017;74(8):977–982.
  • Thompson JC, Harris J, Sollom AC, et al. Longitudinal evaluation of neuropsychiatric symptoms in Huntington’s disease. J Neuropsychiatry Clin Neurosci. 2012;24(1):53–60.
  • Burgunder J-M, Guttman M, Perlman S, et al. An international survey-based algorithm for the pharmacologic treatment of chorea in Huntington’s disease. PLoS Curr. 2011;3:RRN1260.
  • Zesiewicz TA, Sullivan KL, Hauser RA, et al. Open-label pilot study of levetiracetam (Keppra) for the treatment of chorea in Huntington’s disease. Mov Disord. 2006;21(11):1998–2001.
  • Verhagen Metman L, Morris MJ, Farmer C, et al. Huntington’s disease: a randomized, controlled trial using the NMDA-antagonist amantadine. Neurology. 2002;59(5):694–699.
  • O’Suilleabhain P, Dewey, Jr RB Jr. A randomized trial of amantadine in Huntington disease. Arch Neurol. 2003;60(7):996–998.
  • de Yebenes JG, Landwehrmeyer B, Squitieri F, et al. Pridopidine for the treatment of motor function in patients with Huntington’s disease (MermaiHD): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2011;10(12):1049–1057.
  • Huntington Study Group HI. A randomized, double‐blind, placebo‐controlled trial of pridopidine in Huntington’s disease. Mov Disord. 2013;28(10):1407–1415.
  • Reilmann R, McGarry A, Grachev ID, et al. Safety and efficacy of pridopidine in patients with Huntington’s disease (PRIDE-HD): a phase 2, randomised, placebo-controlled, multicentre, dose-ranging study. Lancet Neurol. 2019;18(2):165–176.
  • McGarry A, Auinger P, Kieburtz K, et al. Additional safety and exploratory efficacy data at 48 and 60 months from open-HART, an open-label extension study of pridopidine in Huntington disease. J Huntingtons Dis. 2020;9(2):173–184.
  • Ondo WG, Mejia NI, Hunter CB. A pilot study of the clinical efficacy and safety of memantine for Huntington’s disease. Parkinsonism relat disord. 2007;13(7):453–454
  • Zittel S, Tadic V, Moll CKE, et al. Prospective evaluation of Globus pallidus internus deep brain stimulation in Huntington’s disease. Parkinsonism Relat Disord. 2018;51:96–100.
  • Delorme C, Rogers A, Lau B, et al. Deep brain stimulation of the internal pallidum in Huntington’s disease patients: clinical outcome and neuronal firing patterns. J Neurol. 2016;263(2):290–298.
  • Gonzalez V, Cif L, Biolsi B, et al. Deep brain stimulation for Huntington’s disease: long-term results of a prospective open-label study. J Neurosurg. 2014;121(1):114–122.
  • Wojtecki L, Groiss SJ, Ferrea S, et al. A prospective pilot trial for Pallidal deep brain stimulation in Huntington’s disease. Front Neurol. 2015;6:177.
  • Byars JA, Beglinger LJ, Moser DJ, et al. Substance abuse may be a risk factor for earlier onset of Huntington disease. J Neurol. 2012;259(9):1824–1831.
  • Wexler NS, Lorimer J, Porter J, et al. Venezuelan kindreds reveal that genetic and environmental factors modulate Huntington’s disease age of onset. Proc Natl Acad Sci USA. 2004;101(10):3498–3503.
  • Myers RH, Sax DS, Koroshetz WJ, et al. Factors associated with slow progression in Huntington’s disease. Arch Neurol. 1991;48(8):800–804.
  • Busse M, Quinn L, Debono K, et al. A randomized feasibility study of a 12-week community-based exercise program for people with Huntington’s disease. J Neurol Phys Ther. 2013;37(4):149–158.
  • Frese S, Petersen JA, Ligon-Auer M, et al. Exercise effects in Huntington disease. J Neurol. 2017;264(1):32–39.
  • Khalil H, Quinn L, van Deursen R, et al. What effect does a structured home-based exercise programme have on people with Huntington’s disease? A randomized, controlled pilot study. Clin Rehabil. 2013;27(7):646–658.
  • Quinn L, Hamana K, Kelson M, et al. A randomized, controlled trial of a multi-modal exercise intervention in Huntington’s disease. Parkinsonism Relat Disord. 2016;31:46–52.
  • for FCC, Research CE. Report to the President and the Congress In: Services DoHaH, ed. 2009.
  • Gibson JS, Ridner SH, Dietrich MS, et al. Measuring functional status in Huntington’s disease. Mov Disord. 2020;27. doi: 10.1002/mds.28363.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.