618
Views
8
CrossRef citations to date
0
Altmetric
Drug Evaluation

Pridopidine for the treatment of Huntington’s disease

Pages 485-492 | Received 07 Dec 2015, Accepted 09 Feb 2016, Published online: 10 Mar 2016

Bibliography

  • 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;72:971–83.
  • Pringsheim T, Wiltshire K, et al. The incidence and prevalence of Huntington’s disease: a systematic review and meta-analysis. Move Disorders: Official Journal Move Disord Soc. 2012;27:1083–1091.
  • Walker FO. Huntington’s disease. Semin Neurol. 2007;27:143–50.
  • Ross CA, Pantelyat A, Kogan J, et al. Determinants of functional disability in Huntington’s disease: role of cognitive and motor dysfunction. Move Disorders: Official Journal Move Disord Soc. 2014;29:1351–8.
  • Dumas EM, Van Den Bogaard SJ, Middelkoop HA, et al. A review of cognition in Huntington’s disease. Front Bioscience. 2013;5:1–18.
  • Craufurd D, Thompson JC, Snowden JS. Behavioral changes in Huntington disease. Neuropsychiatry Neuropsychol Behav Neurol. 2001;14:219–226.
  • Nehl C, Paulsen JS. Huntington study group. Cognitive and psychiatric aspects of Huntington disease contribute to functional capacity. J Nerv Ment Dis. 2004;192:72–74.
  • Quarrell O, O’Donovan KL, Bandmann O, et al. The prevalence of juvenile Huntington’s disease: A review of the literature and meta-analysis. Plos Currents. 2012;4:e4f8606b742ef3.
  • Ross CA, Shoulson I. Huntington disease: pathogenesis, biomarkers, and approaches to experimental therapeutics. Parkinsonism Relat Disord. 2009;15(Suppl 3):S135–8.
  • Schwab LC, Garas SN, Drouin-Ouellet J, et al. Dopamine and Huntington’s disease. Expert Rev Neurother. 2015;15:445–58.
  • Klawans HL, Goetz CG, Paulson GW, et al. Levodopa and presymptomatic detection of Huntington’s disease–eight-year follow-up. N Engl J Med. 1980;302:1090.
  • Klawans HL Jr., Goodman RM, Paulson GW, et al. Levodopa in the presymptomatic diagnosis of Huntington’s chorea. Lancet. 1972;2:49.
  • Rabinovich-Guilatt L, Schultz A, Halabi A, et al.. The effect of mild and moderate renal impairment on the pharmacokintetics of pridopidine,a new drug for Huntington’s disease. Br J Pharmacol. 2015;81(2):246–255.
  • Dunkel P, Chai CL, Sperlagh B, et al. Clinical utility of neuroprotective agents in neurodegenerative diseases: current status of drug development for Alzheimer’s, Parkinson’s and Huntington’s diseases, and amyotrophic lateral sclerosis. Expert Opin Investig Drugs. 2012;21:1267–1308.
  • 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;79:597–603.
  • Bonelli RM, Wenning GK. Pharmacological management of Huntington’s disease: an evidence-based review. Curr Pharm Des. 2006;12:2701–2720.
  • Mestre TA, Ferreira JJ. An evidence-based approach in the treatment of Huntington’s disease. Parkinsonism Relat Disord. 2012;18:316–320.
  • Killoran A, Biglan KM. Current therapeutic options for Huntington’s disease: good clinical practice versus evidence-based approaches? Move Disorders: Official Journal Move Disord Soc. 2014;29:1404–1413.
  • Reilmann R. Pharmacological treatment of chorea in Huntington’s disease-good clinical practice versus evidence-based guideline. Move Disorders: Official Journal Move Disord Soc. 2013;28:1030–1033.
  • Zielonka D, Mielcarek M, Landwehrmeyer GB. Update on Huntington’s disease: advances in care and emerging therapeutic options. Parkinsonism Relat Disord. 2015;21:169–178.
  • 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 Currents. 2011;3:RRN1261.
  • Burgunder JM, Guttman M, Perlman S, et al. An international survey-based algorithm for the pharmacologic treatment of chorea in Huntington’s disease. Plos Currents. 2011;3:RRN1260.
  • 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 Currents. 2011;3:RRN1259.
  • Robertson L, Santini H, O’Donovan KL, et al. Current pharmacological management in juvenile Huntington’s disease. Plos Currents. 2012;4:RRN1304.
  • Huntington Study G. Tetrabenazine as antichorea therapy in Huntington disease: a randomized controlled trial. Neurology. 2006;66:366–372.
  • Mahant N, McCusker EA, Byth K, et al. Huntington’s disease: clinical correlates of disability and progression. Neurology. 2003;61:1085–1092.
  • Bruck W, Wegner C. Insight into the mechanism of laquinimod action. J Neurol Sci. 2011;306:173–179.
  • Southwell AL, Franciosi S, Villanueva EB, et al. Anti-semaphorin 4D immunotherapy ameliorates neuropathology and some cognitive impairment in the YAC128 mouse model of Huntington disease. Neurobiol Dis. 2015;76:46–56.
  • Klivenyi P, Ferrante RJ, Gardian G, et al. Increased survival and neuroprotective effects of BN82451 in a transgenic mouse model of Huntington’s disease. J Neurochem. 2003;86:267–272.
  • Adanyeguh IM, Rinaldi D, Henry PG, et al. Triheptanoin improves brain energy metabolism in patients with Huntington disease. Neurology. 2015;84:490–495.
  • Fabio KM, Guillon CD, Lu SF, et al. Pharmacokinetics and metabolism of SRX246: a potent and selective vasopressin 1a antagonist. J Pharm Sci. 2013;102:2033–2043.
  • Kara E, Lin H, Svensson K, et al. Analysis of the actions of the novel dopamine receptor-directed compounds (S)-OSU6162 and ACR16 at the D2 dopamine receptor. Br J Pharmacol. 2010;161:1343–1350.
  • Miki Y, Tanji K, Mori F, et al. Sigma-1 receptor is involved in degradation of intranuclear inclusions in a cellular model of Huntington’s disease. Neurobiol Dis. 2015;74:25–31.
  • Squitieri F, Di Pardo A, Favellato M, et al.. Pridopidine, a dopamine stabilizer, improves motor performance and shows neuroprotective effects in Huntington disease R6/2 mouse model. J Cell Mol Med. 2015;19(11):2540–2548.
  • Dyhring T, Nielsen EO, Sonesson C, et al. The dopaminergic stabilizers pridopidine (ACR16) and (-)-OSU6162 display dopamine D(2) receptor antagonism and fast receptor dissociation properties. Eur J Pharmacol. 2010;628:19–26.
  • Natesan S, Svensson KA, Reckless GE, et al. The dopamine stabilizers (S)-(-)-(3-methanesulfonyl-phenyl)-1-propyl-piperidine [(-)-OSU6162] and 4-(3-methanesulfonylphenyl)-1-propyl-piperidine (ACR16) show high in vivo D2 receptor occupancy, antipsychotic-like efficacy, and low potential for motor side effects in the rat.. J Pharm Exp Ther. 2006;318:810–818.
  • Huntington Study Group. A randomized, double-blind, placebo-controlled trial of pridopidine in Huntington’s disease. Move Disorders: Official Journal Move Disord Soc. 2013;28:1407–15.
  • Ponten H, Kullingsjo J, Lagerkvist S, et al. In vivo pharmacology of the dopaminergic stabilizer pridopidine. Eur J Pharmacol. 2010;644:88–95.
  • Pettersson F, Ponten H, Waters N, et al. Synthesis and evaluation of a set of 4-phenylpiperidines and 4-phenylpiperazines as D2 receptor ligands and the discovery of the dopaminergic stabilizer 4-[3-(methylsulfonyl)phenyl]-1-propylpiperidine (huntexil, pridopidine, ACR16). J Med Chem. 2010;53:2510–20.
  • Sahlholm K, Sijbesma JW, Maas B, et al. Pridopidine selectively occupies sigma-1 rather than dopamine D2 receptors at behaviorally active doses. Psychopharmacology (Berl). 2015;232:3443–53.
  • Krog PL, Osterberg O, Drewes PG, et al. Pharmacokinetic and tolerability profiles of pridopidine in healthy-volunteer poor and extensive CYP2D6 metabolizers, following single and multiple dosing. Eur J Drug Metab Pharmacokinet. 2013;38:43–51.
  • Hellden A, Panagiotidis G, Johansson P, et al. The dopaminergic stabilizer pridopidine is to a major extent N-depropylated by CYP2D6 in humans. Eur J Clin Pharmacol. 2012;68:1281–1286.
  • Nakamura K, Goto F, Ray WA, et al. Interethnic differences in genetic polymorphism of debrisoquin and mephenytoin hydroxylation between Japanese and Caucasian populations. Clin Pharmacol Ther. 1985;38:402–408.
  • Lundin A, Dietrichs E, Haghighi S, et al. Efficacy and safety of the dopaminergic stabilizer Pridopidine (ACR16) in patients with Huntington’s disease. Clin Neuropharmacol. 2010;33:260–4.
  • 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;10:1049–57.
  • Landwehrmeyer B, Marder K, Biilmann Ronn B, et al. Effects of the dopaminergic stabilizer pridopidine on motor symptoms in Huntington’s disease: a meta-analysis [Abstract]. Clin Genet. 2011;80:48.
  • Tanji H, Gruber-Baldini AL, Anderson KE, et al. A comparative study of physical performance measures in Parkinson’s disease. Move Disorders: Official Journal Move Disord Soc. 2008;23:1897–1905.
  • Squitieri F, Landwehrmeyer B, Reilmann R, et al. One-year safety and tolerability profile of pridopidine in patients with Huntington disease. Neurology. 2013;80:1086–1094.

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.