245
Views
3
CrossRef citations to date
0
Altmetric
Review

Therapeutic advances in Huntington’s disease

, , &
Pages 809-821 | Received 02 Mar 2016, Accepted 27 May 2016, Published online: 13 Jun 2016

References

  • A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. The Huntington’s Disease Collaborative Research Group. Cell. 1993;72(6):971–983.
  • Legleiter J, Mitchell E, Lotz GP, et al. Mutant huntingtin fragments form oligomers in a polyglutamine length-dependent manner in vitro and in vivo. J Biol Chem. 2010;285(19):14777–14790.
  • Cha J-HJ. Transcriptional signatures in Huntington’s disease. Prog Neurobiol. 2007;83(4):228–248.
  • Heng MY, Detloff PJ, Wang PL, et al. In vivo evidence for NMDA receptor-mediated excitotoxicity in a murine genetic model of Huntington disease. J Neurosci. 2009;29(10):3200–3205.
  • Browne SE, Bowling AC, MacGarvey U, et al. Oxidative damage and metabolic dysfunction in Huntington’s disease: selective vulnerability of the basal ganglia. Ann Neurol. 1997;41(5):646–653.
  • Zuccato C, Cattaneo E. Role of brain-derived neurotrophic factor in Huntington’s disease. Prog Neurobiol. 2007;81(5–6):294–330.
  • Smith GA, Rocha EM, McLean JR, et al. Progressive axonal transport and synaptic protein changes correlate with behavioral and neuropathological abnormalities in the heterozygous Q175 KI mouse model of Huntington’s disease. Hum Mol Genet. 2014;23(17):4510–4527.
  • Cattaneo E, Rigamonti D, Goffredo D, et al. Loss of normal huntingtin function: new developments in Huntington’s disease research. Trends Neurosci. 2001;24(3):182–188.
  • Nithianantharajah J, Hannan AJ. Dysregulation of synaptic proteins, dendritic spine abnormalities and pathological plasticity of synapses as experience-dependent mediators of cognitive and psychiatric symptoms in Huntington’s disease. Neuroscience. 2013;251:66–74.
  • 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 U S A. 2004;101(10):3498–3503.
  • Sanberg PR, Fibiger HC, Mark RF. Body weight and dietary factors in Huntington’s disease patients compared with matched controls. Med J Aust. 1981;1(8):407–409.
  • Kirkwood SC, Su JL, Conneally P, et al. Progression of symptoms in the early and middle stages of Huntington disease. Arch Neurol. 2001;58(2):273–278.
  • Snowden JS, Craufurd D, Griffiths HL, et al. Awareness of involuntary movements in Huntington disease. Arch Neurol. 1998;55(6):801–805.
  • Sitek EJ, Soltan W, Wieczorek D, et al. Self-awareness of memory function in Parkinson’s disease in relation to mood and symptom severity. Aging Ment Health. 2011;15(2):150–156.
  • Jankovic J, Roos RAC. Chorea associated with Huntington’s disease: to treat or not to treat? Mov Disord. 2014;29(11):1414–1418.
  • Ross CA, Pantelyat A, Kogan J, et al. Determinants of functional disability in Huntington’s disease: role of cognitive and motor dysfunction. Mov Disord. 2014;29(11):1351–1358.
  • Scherman D, Jaudon P, Henry JP. Characterization of the monoamine carrier of chromaffin granule membrane by binding of [2-3H]dihydrotetrabenazine. Proc Natl Acad Sci U S A. 1983;80(2):584–588.
  • Pettibone DJ, Pflueger AB, Totaro JA. Tetrabenazine-induced depletion of brain monoamines: mechanism by which desmethylimipramine protects cortical norepinephrine. Eur J Pharmacol. 1984;102(3–4):431–436.
  • Darchen F, Masuo Y, Vial M, et al. Quantitative autoradiography of the rat brain vesicular monoamine transporter using the binding of [3H]dihydrotetrabenazine and 7-amino-8-[125I]iodoketanserin. Neuroscience. 1989;33(2):341–349.
  • Pearson SJ, Reynolds GP. Depletion of monoamine transmitters by tetrabenazine in brain tissue in Huntington’s disease. Neuropharmacology. 1988;27(7):717–719.
  • Lane JD, Smith JE, Shea PA, et al. Neurochemical changes following the administration of depleters of biogenic monoamines. Life Sci. 1976;19(11):1663–1667.
  • Huntington Study Group. Tetrabenazine as antichorea therapy in Huntington disease: a randomized controlled trial. Neurology. 2006;66(3):366–372.
  • Frank S, Ondo W, Fahn S, et al. A study of chorea after tetrabenazine withdrawal in patients with Huntington disease. Clin Neuropharmacol. 2008;31(3):127–133.
  • Paleacu D. Tetrabenazine in the treatment of Huntington’s disease. Neuropsychiatr Dis Treat. 2007;3(5):545–551.
  • Jankovic J, Beach J. Long-term effects of tetrabenazine in hyperkinetic movement disorders. Neurology. 1997;48(2):358–362.
  • Jankovic J. Treatment of hyperkinetic movement disorders with tetrabenazine: a double-blind crossover study. Ann Neurol. 1982;11(1):41–47.
  • Kingston D. Tetrabenazine for involuntary movement disorders. Med J Aust. 1979;1(13):628–630.
  • Ondo WG, Tintner R, Thomas M, et al. Tetrabenazine treatment for Huntington’s disease-associated chorea. Clin Neuropharmacol. 2002;25(6):300–302.
  • Paleacu D, Giladi N, Moore O, et al. Tetrabenazine treatment in movement disorders. Clin Neuropharmacol. 2004;27(5):230–233.
  • LeWitt PA. Tardive dyskinesia caused by tetrabenazine. Clin Neuropharmacol. 2013;36(3):92–93.
  • Smith JM, Baldessarini RJ. Changes in prevalence, severity, and recovery in tardive dyskinesia with age. Arch Gen Psychiatry. 1980;37(12):1368–1373.
  • Guay DR. Tetrabenazine, a monoamine-depleting drug used in the treatment of hyperkinetic movement disorders. Am J Geriatr Pharmacother. 2010;8(4):331–373.
  • Burgunder JM, 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.
  • Dallocchio C, Buffa C, Tinelli C, et al. Effectiveness of risperidone in Huntington chorea patients. J Clin Psychopharmacol. 1999;19(1):101–103.
  • Orth M, Handley OJ, Schwenke C, et al. Observing Huntington’s disease: the European Huntington’s disease network’s registry. PLoS Curr. 2010;2:RRN1184.
  • 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.
  • 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, Tribl GG, et al. High-dose olanzapine in Huntington’s disease. Int Clin Psychopharmacol. 2002;17(2):91–93.
  • Paleacu D, Anca M, Giladi N. Olanzapine in Huntington’s disease. Acta Neurol Scand. 2002;105(6):441–444.
  • 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.
  • 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.
  • Madhusoodanan S, Brecher M, Brenner R, et al. Risperidone in the treatment of elderly patients with psychotic disorders. Am J Geriatr Psychiatry. 1999;7(2):132–138.
  • 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.
  • 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.
  • 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.
  • Bonelli RM, Niederwieser G. Quetiapine in Huntington’s disease: a first case report. J Neurol. 2002;249(8):1114–1115.
  • van Vugt JP, Siesling S, Vergeer M, et al. Clozapine versus placebo in Huntington’s disease: a double blind randomised comparative study. J Neurol Neurosurg Psychiatry. 1997;63(1):35–39.
  • Mestre T, Ferreira J, Coelho MM, et al. Therapeutic interventions for symptomatic treatment in Huntington’s disease. Cochrane Database Syst Rev. 2009;3:CD006456.
  • Armstrong MJ, Miyasaki JM. 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(6):597–603.
  • Bonelli RM, Wenning GK. Pharmacological management of Huntington’s disease: an evidence-based review. Curr Pharm Des. 2006;12(21):2701–2720.
  • O’Suilleabhain P, Dewey RB Jr. A randomized trial of amantadine in Huntington disease. Arch Neurol. 2003;60(7):996–998.
  • Heckmann JM, Legg P, Sklar D, et al. IV amantadine improves chorea in Huntington’s disease: an acute randomized, controlled study. Neurology. 2004;63(3):597–598; author reply −8.
  • 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.
  • Armstrong MJ, Miyasaki JM. 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(6):597–603.
  • Landwehrmeyer GB, Dubois B, de Yebenes JG, et al. Riluzole in Huntington’s disease: a 3-year, randomized controlled study. Ann Neurol. 2007;62(3):262–272.
  • Huntington Study Group. Dosage effects of riluzole in Huntington’s disease: a multicenter placebo-controlled study. Neurology. 2003;61(11):1551–1556.
  • Lacomblez L, Bensimon G, Leigh PN, et al. Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Amyotrophic lateral sclerosis/riluzole study group II. Lancet. 1996;347(9013):1425–1431.
  • Vaddadi KS, Soosai E, Chiu E, et al. A randomised, placebo-controlled, double blind study of treatment of Huntington’s disease with unsaturated fatty acids. Neuroreport. 2002;13(1):29–33.
  • Mestre T, Ferreira J, Coelho MM, et al. Therapeutic interventions for disease progression in Huntington’s disease. Cochrane Database Syst Rev. 2009;3:CD006455.
  • Curtis A, Mitchell I, Patel S, et al. A pilot study using nabilone for symptomatic treatment in Huntington’s disease. Mov Disord. 2009;24(15):2254–2259.
  • Gatto EM, Uribe Roca C, Raina G, et al. Vascular hemichorea/hemiballism and topiramate. Mov Disord. 2004;19(7):836–838.
  • 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.
  • Puri BK, Leavitt BR, Hayden MR, et al. Ethyl-EPA in Huntington disease: a double-blind, randomized, placebo-controlled trial. Neurology. 2005;65(2):286–292.
  • Huntington Study Group TREND-HD Investigators. Randomized controlled trial of ethyl-eicosapentaenoic acid in Huntington disease: the TREND-HD study. Arch Neurol. 2008;65(12):1582–1589.
  • Huntington Study Group. Minocycline safety and tolerability in Huntington disease. Neurology. 2004;63(3):547–549.
  • Mateo D, Gimenez-Roldan S. [The effect of piracetam on involuntary movements in Huntington’s disease. A double-blind, placebo-controlled study]. Neurologia. 1996;11(1):16–19.
  • Hersch SM, Gevorkian S, Marder K, et al. Creatine in Huntington disease is safe, tolerable, bioavailable in brain and reduces serum 8OH2ʹdG. Neurology. 2006;66(2):250–252.
  • A randomized, placebo-controlled trial of coenzyme Q10. and remacemide in Huntington’s disease. Neurology. 2001;57(3):397–404.
  • Stocchi F, Carta A, Berardelli A, et al. Effects of terguride in patients with Huntington’s disease. Clin Neuropharmacol. 1989;12(5):435–439.
  • Yero T, Rey JA. Tetrabenazine (xenazine), an FDA-approved treatment option for Huntington’s disease-related chorea. P T. 2008;33(12):690–694.
  • Frank S. Treatment of Huntington’s disease. Neurotherapeutics. 2014;11(1):153–160.
  • 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.
  • Reilmann R. Pharmacological treatment of chorea in Huntington’s disease-good clinical practice versus evidence-based guideline. Mov Disord. 2013;28(8):1030–1033.
  • Bonelli RM, Niederwieser G, Diez J, et al. Pramipexole ameliorates neurologic and psychiatric symptoms in a Westphal variant of Huntington’s disease. Clin Neuropharmacol. 2002;25(1):58–60.
  • 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.
  • Low PA, Allsop JL, Halmagyi GM. Huntington’s chorea: the rigid form (Westphal variant) treated with levodopa. Med J Aust. 1974;1(11):393–394.
  • Racette BA, Perlmutter JS. Levodopa responsive parkinsonism in an adult with Huntington’s disease. J Neurol Neurosurg Psychiatry. 1998;65(4):577–579.
  • Carella F, Scaioli V, Ciano C, et al. Adult onset myoclonic Huntington’s disease. Mov Disord. 1993;8(2):201–205.
  • Anderson KE, Marder KS. An overview of psychiatric symptoms in Huntington’s disease. Curr Psychiatry Rep. 2001;3(5):379–388.
  • Ho AK, Gilbert AS, Mason SL, et al. Health-related quality of life in Huntington’s disease: which factors matter most? Mov Disord. 2009;24(4):574–578.
  • 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;3:RRN1259.
  • Hassler F, Reis O. Pharmacotherapy of disruptive behavior in mentally retarded subjects: a review of the current literature. Dev Disabil Res Rev. 2010;16(3):265–272.
  • 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;3:RRN1261.
  • Ranen NG, Peyser CE, Folstein SE. ECT as a treatment for depression in Huntington’s disease. J Neuropsychiatry Clin Neurosci. 1994;6(2):154–159.
  • van Duijn E, Roos RA, Smarius LJ, et al. [Electroconvulsive therapy in patients with Huntington’s disease and depression]. Ned Tijdschr Geneeskd. 2005;149(39):2141–2144.
  • Gliatto MF, Rai AK. Evaluation and treatment of patients with suicidal ideation. Am Fam Physician. 1999;59(6):1500–1506.
  • Foerde K, Braun EK, Shohamy D. A trade-off between feedback-based learning and episodic memory for feedback events: evidence from Parkinson’s disease. Neurodegener Dis. 2013;11(2):93–101.
  • Fernandez HH, Friedman JH, Grace J, et al. Donepezil for Huntington’s disease. Mov Disord. 2000;15(1):173–176.
  • Rot U, Kobal J, Sever A, et al. Rivastigmine in the treatment of Huntington’s disease. Eur J Neurol. 2002;9(6):689–690.
  • Petrikis P, Andreou C, Piachas A, et al. Treatment of Huntington’s disease with galantamine. Int Clin Psychopharmacol. 2004;19(1):49–50.
  • de Tommaso M, Specchio N, Sciruicchio V, et al. Effects of rivastigmine on motor and cognitive impairment in Huntington’s disease. Mov Disord. 2004;19(12):1516–1518.
  • Cubo E, Shannon KM, Tracy D, et al. Effect of donepezil on motor and cognitive function in Huntington disease. Neurology. 2006;67(7):1268–1271.
  • Blackwell AD, Paterson NS, Barker RA, et al. The effects of modafinil on mood and cognition in Huntington’s disease. Psychopharmacology (Berl). 2008;199(1):29–36.
  • Andrews SC, Dominguez JF, Mercieca EC, et al. Cognitive interventions to enhance neural compensation in Huntington’s disease. Neurodegener Dis Manag. 2015;5(2):155–164.
  • Thompson JA, Cruickshank TM, Penailillo LE, et al. The effects of multidisciplinary rehabilitation in patients with early-to-middle-stage Huntington’s disease: a pilot study. Eur J Neurol. 2013;20(9):1325–1329.
  • Zinzi P, Salmaso D, De Grandis R, et al. Effects of an intensive rehabilitation programme on patients with Huntington’s disease: a pilot study. Clin Rehabil. 2007;21(7):603–613.
  • Leng TR, Woodward MJ, Stokes MJ, et al. Effects of multisensory stimulation in people with Huntington’s disease: a randomized controlled pilot study. Clin Rehabil. 2003;17(1):30–41.
  • Mo C, Hannan AJ, Renoir T. Environmental factors as modulators of neurodegeneration: insights from gene-environment interactions in Huntington’s disease. Neurosci Biobehav Rev. 2015;52:178–192.
  • Bohlen S, Ekwall C, Hellstrom K, et al. Physical therapy in Huntington’s disease–toward objective assessments? Eur J Neurol. 2013;20(2):389–393.
  • Schilling G, Savonenko AV, Coonfield ML, et al. Environmental, pharmacological, and genetic modulation of the HD phenotype in transgenic mice. Exp Neurol. 2004;187(1):137–149.
  • Lopez-Sendon JL, Royuela A, Trigo P, et al. What is the impact of education on Huntington’s disease? Mov Disord. 2011;26(8):1489–1495.
  • Trembath MK, Horton ZA, Tippett L, et al. A retrospective study of the impact of lifestyle on age at onset of Huntington disease. Mov Disord. 2010;25(10):1444–1450.
  • Huntington Study Group HART Investigators. A randomized, double-blind, placebo-controlled trial of pridopidine in Huntington’s disease. Mov Disord. 2013;28(10):1407–1415.
  • 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.
  • Sampaio C, Borowsky B, Reilmann R. Clinical trials in Huntington’s disease: interventions in early clinical development and newer methodological approaches. Mov Disord. 2014;29(11):1419–1428.
  • Reilmann R, Rouzade-Dominguez ML, Saft C, et al. A randomized, placebo-controlled trial of AFQ056 for the treatment of chorea in Huntington’s disease. Mov Disord. 2015;30(3):427–431.
  • Chappie T, Humphrey J, Menniti F, et al. PDE10A inhibitors: an assessment of the current CNS drug discovery landscape. Curr Opin Drug Discov Devel. 2009;12(4):458–467.
  • Starr PA, Kang GA, Heath S, et al. Pallidal neuronal discharge in Huntington’s disease: support for selective loss of striatal cells originating the indirect pathway. Exp Neurol. 2008;211(1):227–233.
  • Tang JKH, Moro E, Lozano AM, et al. Firing rates of pallidal neurons are similar in Huntington’s and Parkinson’s disease patients. Exp Brain Res. 2005;166(2):230–236.
  • Biolsi B, Cif L, Fertit HE, et al. Long-term follow-up of Huntington disease treated by bilateral deep brain stimulation of the internal globus pallidus. J Neurosurg. 2008;109(1):130–132.
  • Cislaghi G, Capiluppi E, Saleh C, et al. Bilateral globus pallidus stimulation in Westphal variant of Huntington disease. Neuromodulation. 2014;17(5):502–505.
  • Fasano A, Mazzone P, Piano C, et al. GPi-DBS in Huntington’s disease: results on motor function and cognition in a 72-year-old case. Mov Disord. 2008;23(9):1289–1292.
  • Garcia-Ruiz PJ, Ayerbe J, del Val J, et al. Deep brain stimulation in disabling involuntary vocalization associated with Huntington’s disease. Parkinsonism Relat Disord. 2012;18(6):803–804.
  • 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.
  • Gruber D, Kuhn AA, Schoenecker T, et al. Quadruple deep brain stimulation in Huntington’s disease, targeting pallidum and subthalamic nucleus: case report and review of the literature. J Neural Transm (Vienna). 2014;121(10):1303–1312.
  • Hebb MO, Garcia R, Gaudet P, et al. Bilateral stimulation of the globus pallidus internus to treat choreathetosis in Huntington’s disease: technical case report. Neurosurgery. 2006;58(2):E383; discussion E.
  • Huys D, Bartsch C, Poppe P, et al. Management and outcome of pallidal deep brain stimulation in severe Huntington’s disease. Fortschr Neurol Psychiatr. 2013;81(4):202–205.
  • Kang GA, Heath S, Rothlind J, et al. Long-term follow-up of pallidal deep brain stimulation in two cases of Huntington’s disease. J Neurol Neurosurg Psychiatry. 2011;82(3):272–277.
  • Moro E, Lang AE, Strafella AP, et al. Bilateral globus pallidus stimulation for Huntington’s disease. Ann Neurol. 2004;56(2):290–294.
  • Spielberger S, Hotter A, Wolf E, et al. Deep brain stimulation in Huntington’s disease: a 4-year follow-up case report. Mov Disord. 2012;27(6):806-7;author reply 7-8.
  • Velez-Lago FM, Thompson A, Oyama G, et al. Differential and better response to deep brain stimulation of chorea compared to dystonia in Huntington’s disease. Stereotact Funct Neurosurg. 2013;91(2):129–133.
  • Lopez-Sendon Moreno JL, Garcia-Caldentey J, Regidor I, et al. A 5-year follow-up of deep brain stimulation in Huntington’s disease. Parkinsonism Relat Disord. 2014;20(2):260–261.
  • Zeef DH, Vlamings R, Lim LW, et al. Motor and non-motor behaviour in experimental Huntington’s disease. Behav Brain Res. 2012;226(2):435–439.
  • Smith KM, Spindler MA. Uncommon applications of deep brain stimulation in hyperkinetic movement disorders. Tremor Other Hyperkinet Mov (N Y). 2015;5:278.
  • Gibrat C, Cicchetti F. Potential of cystamine and cysteamine in the treatment of neurodegenerative diseases. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(2):380–389.
  • Borrell-Pages M, Canals JM, Cordelieres FP, et al. Cystamine and cysteamine increase brain levels of BDNF in Huntington disease via HSJ1b and transglutaminase. J Clin Invest. 2006;116(5):1410–1424.
  • Besouw M, Masereeuw R, van den Heuvel L, et al. Cysteamine: an old drug with new potential. Drug Discov Today. 2013;18(15–16):785–792.
  • Smith MR, Syed A, Lukacsovich T, et al. A potent and selective Sirtuin 1 inhibitor alleviates pathology in multiple animal and cell models of Huntington’s disease. Hum Mol Genet. 2014;23(11):2995–3007.
  • Huntington Study Group Reach2HD Investigators. Safety, tolerability, and efficacy of PBT2 in Huntington’s disease: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2015;14(1):39–47.
  • Bjorkqvist M, Wild EJ, Thiele J, et al. A novel pathogenic pathway of immune activation detectable before clinical onset in Huntington’s disease. J Exp Med. 2008;205(8):1869–1877.
  • Sagredo O, Pazos MR, Valdeolivas S, et al. Cannabinoids: novel medicines for the treatment of Huntington’s disease. Recent Pat CNS Drug Discov. 2012;7(1):41–48.
  • Lopez-Sendon Moreno JL, Garcia Caldentey J, Trigo Cubillo P, et al. A double-blind, randomized, cross-over, placebo-controlled, pilot trial with Sativex in Huntington’s disease. J Neurol. 2016. [Epub ahead of print].
  • Bruck W, Pfortner R, Pham T, et al. Reduced astrocytic NF-κB activation by laquinimod protects from cuprizone-induced demyelination. Acta Neuropathol. 2012;124(3):411–424.
  • Ross CA, Akimov SS. Human-induced pluripotent stem cells: potential for neurodegenerative diseases. Hum Mol Genet. 2014;23(R1):R17–R26.
  • Smith DK, He M, Zhang CL, et al. The therapeutic potential of cell identity reprogramming for the treatment of aging-related neurodegenerative disorders. Prog Neurobiol. 2016. [Epub ahead of print].
  • Wild EJ, Tabrizi SJ. Targets for future clinical trials in Huntington’s disease: what’s in the pipeline? Mov Disord. 2014;29(11):1434–1445.
  • Harper SQ, Staber PD, He X, et al. RNA interference improves motor and neuropathological abnormalities in a Huntington’s disease mouse model. Proc Natl Acad Sci U S A. 2005;102(16):5820–5825.
  • Stanek LM, Sardi SP, Mastis B, et al. Silencing mutant huntingtin by adeno-associated virus-mediated RNA interference ameliorates disease manifestations in the YAC128 mouse model of Huntington’s disease. Hum Gene Ther. 2014;25(5):461–474.
  • Carroll JB, Warby SC, Southwell AL, et al. Potent and selective antisense oligonucleotides targeting single-nucleotide polymorphisms in the Huntington disease gene/allele-specific silencing of mutant huntingtin. Mol Ther. 2011;19(12):2178–2185.
  • Nasir J, Floresco SB, O’Kusky JR, et al. Targeted disruption of the Huntington’s disease gene results in embryonic lethality and behavioral and morphological changes in heterozygotes. Cell. 1995;81(5):811–823.
  • Miller TM, Pestronk A, David W, et al. An antisense oligonucleotide against SOD1 delivered intrathecally for patients with SOD1 familial amyotrophic lateral sclerosis: a phase 1, randomised, first-in-man study. Lancet Neurol. 2013;12(5):435–442.
  • Chiriboga CA, Swoboda KJ, Darras BT, et al. Results from a phase 1 study of nusinersen (ISIS-SMNRx) in children with spinal muscular atrophy. Neurology. 2016;86(10):890–897.
  • Banez-Coronel M, Ayhan F, Tarabochia AD, et al. RAN translation in Huntington disease. Neuron. 2015;88(4):667–677.

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.