348
Views
8
CrossRef citations to date
0
Altmetric
Review

Emerging drugs for the treatment of L-DOPA-induced dyskinesia: an update

, &
Pages 131-144 | Received 26 Mar 2020, Accepted 29 Apr 2020, Published online: 22 May 2020

References

  • Poewe W, Seppi K, Tanner CM, et al. Parkinson disease. Nat Rev Dis Prim. 2017;3:1–21.
  • Mann DMA, Yates PO. Pathogenesis of Parkinson’s disease. Arch Neurol. 1982;39:545–549.
  • Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov Disord. 2001;16:448–458.
  • Hely MA, Morris JGL, Reid WGJ, et al. Sydney Multicenter Study of Parkinson’s disease: non-L-dopa-responsive problems dominate at 15 years. Mov Disord. 2005;20:190–199.
  • Espay AJ, Morgante F, Merola A, et al. Levodopa-induced dyskinesia in Parkinson disease: current and evolving concepts. AnnNeurol. 2018;84:797–811.
  • Warren Olanow C, Kieburtz K, Rascol O, et al. Factors predictive of the development of Levodopa-induced dyskinesia and wearing-off in Parkinson’s disease. Mov Disord. 2013;28:1064–1071.
  • Müller T, Woitalla D, Saft C, et al. Levodopa in plasma correlates with body weight of parkinsonian patients. Park Relat Disord. 2000;6:171–173.
  • Marras C, Beck JC, Bower JH, et al. Prevalence of Parkinson’s disease across North America. npj Park Dis. 2018;4:1–7.
  • Péchevis M, Clarke CE, Vieregge P, et al. Effects of dyskinesias in Parkinson’s disease on quality of life and health-related costs: A prospective European study. Eur J Neurol. 2005;12:956–963.
  • Suh DC, Pahwa R, Mallya U. Treatment patterns and associated costs with Parkinson’s disease levodopa induced dyskinesia. J Neurol Sci. 2012;319:24–31.
  • Marras C, Lang A, Krahn M, et al. Quality of life in early Parkinson’s disease: impact of dyskinesia and motor fluctuations. Mov Disord. 2004;19:22–28.
  • Hechtner MC, Vogt T, Zöllner Y, et al. Quality of life in Parkinson’s disease patients with motor fluctuations and dyskinesias in five European countries. Park Relat Disord. 2014;20:969–974.
  • Zach M, Friedman A, Sławek J, et al. Quality of life in polish patients with long-lasting Parkinson’s disease. Mov Disord. 2004;19:667–672.
  • Thanvi B, Lo N, Robinson T. Levodopa-induced dyskinesia in Parkinson’s disease: clinical features, pathogenesis, prevention and treatment. Postgrad Med J. 2007;83:384–388.
  • Scott NW, Macleod AD, Counsell CE. Motor complications in an incident Parkinson’s disease cohort. Eur J Neurol. 2016;23:304–312.
  • Fahn S. Parkinson disease, the effect of levodopa, and the ELLDOPA trial. Arch Neurol. 1999;56:529–535.
  • Verschuur CVM, Suwijn SR, Boel JA, et al. Randomized delayed-start trial of levodopa in Parkinson’s disease. N Engl J Med. 2019;380:315–324.
  • Holloway RG. Pramipexole vs levodopa as initial treatment for Parkinson disease: A 4-year randomized controlled trial. Arch Neurol. 2004;61:1044–1053.
  • Watts RL, Lyons KE, Pahwa R, et al. Onset of dyskinesia with adjunct ropinirole prolonged-release or additional levodopa in early Parkinson’s disease. Mov Disord. 2010;25:858–866.
  • Hauser RA, Schapira AHV, Barone P, et al. Long-term safety and sustained efficacy of extended-release pramipexole in early and advanced Parkinson’s disease. Eur J Neurol. 2014;21:736–743.
  • Lees AJ, Katzenschlager R, Head J, et al. Ten-year follow-up of three different initial treatments in de-novo PD: A randomized trial. Neurology. 2001;57:1687–1694.
  • Crispo JAG, Fortin Y, Thibault DP, et al. Trends in inpatient antiparkinson drug use in the USA, 2001-2012. Eur J Clin Pharmacol. 2015;71:1011–1019.
  • Dubaz OM, Wu S, Cubillos F, et al. Changes in prescribing practices of dopaminergic medications in individuals with Parkinson’s disease by expert care centers from 2010 to 2017: the Parkinson’s foundation quality improvement initiative. Mov Disord Clin Pract. 2019;6:687–692.
  • Gray R, Ives N, Rick C, et al. Long-term effectiveness of dopamine agonists and monoamine oxidase B inhibitors compared with levodopa as initial treatment for Parkinson’s disease (PD MED): A large, open-label, pragmatic randomised trial. Lancet. 2014;384:1196–1205.
  • Marras C, McDermott MP, Rochon PA, et al. Survival in Parkinson disease: thirteen-year follow-up of the DATATOP cohort. Neurology. 2005;64:87–93.
  • Rascol O, Fitzer-Attas CJ, Hauser R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease (the ADAGIO study): prespecified and post-hoc analyses of the need for additional therapies, changes in UPDRS scores, and non-motor outcomes. Lancet Neurol. 2011;10:415–423.
  • Rascol O, Hauser RA, Stocchi F, et al. Long-term effects of rasagiline and the natural history of treated Parkinson’s disease. Mov Disord. 2016;31:1489–1496.
  • Smith LA, Jackson MJ, Hansard MJ, et al. Effect of pulsatile administration of levodopa on dyskinesia induction in drug-naïve MPTP-treated common marmosets: effect of dose, frequency of administration, and brain exposure. Mov Disord. 2003;18:487–495.
  • Fox SH, Katzenschlager R, Lim SY, et al. International Parkinson and movement disorder society evidence-based medicine review: update on treatments for the motor symptoms of Parkinson’s disease. Mov Disord. 2018;33:1248–1266.
  • Stocchi F, Rascol O, Kieburtz K, et al. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: the STRIDE-PD study. Ann Neurol. 2010;68:18–27.
  • Muller T. Entacapone. Expert Opin Drug Metab Toxicol. 2010;6:983–993.
  • Antonini A, Fung VSC, Boyd JT, et al. Effect of levodopa-carbidopa intestinal gel on dyskinesia in advanced Parkinson’s disease patients. Mov Disord. 2016;31:530–537.
  • Marano M, Naranian T, di Biase L, et al. Complex dyskinesias in Parkinson patients on levodopa/carbidopa intestinal gel. Park Relat Disord. 2019;69:140–146.
  • Millan MJ, Maiofiss L, Cussac D, et al. Differential actions of antiparkinson agents at multiple classes of monoaminergic receptor. I. A multivariate analysis of the binding profiles of 14 drugs at 21 native and cloned human receptor subtypes. J Pharmacol Exp Ther. 2002;303:791–804.
  • Katzenschlager R, Hughes A, Evans A, et al. Continuous subcutaneous apomorphine therapy improves dyskinesias in Parkinson’s disease: A prospective study using single-dose challenges. Mov Disord. 2005;20:151–157.
  • García Ruiz PJ, Ignacio ÁS, Pensado BA, et al. Efficacy of long-term continuous subcutaneous apomorphine infusion in advanced Parkinson’s disease with motor fluctuations: A multicenter study. Mov Disord. 2008;23:1130–1136.
  • Stocchi F, Vacca L, De Pandis MF, et al. Subcutaneous continuous apomorphine infusion in fluctuating patients with Parkinson’s disease: long-term results. Neurol Sci. 2001;22:93–94.
  • Katzenschlager R, Poewe W, Rascol O, et al. Apomorphine subcutaneous infusion in patients with Parkinson’s disease with persistent motor fluctuations (TOLEDO): a multicentre, double-blind, randomised, placebo-controlled trial. Lancet Neurol. 2018;17:749–759.
  • Odekerken VJJ, van Laar T, Staal MJ, et al. Subthalamic nucleus versus globus pallidus bilateral deep brain stimulation for advanced Parkinson’s disease (NSTAPS study): A randomised controlled trial. Lancet Neurol. 2013;12:37–44.
  • Odekerken VJJ. GPi vs STN deep brain stimulation for Parkinson disease: three-year follow-up. Neurology. 2016;87:745–746.
  • Figee M, De Koning P, Klaassen S, et al. Deep brain stimulation induces striatal dopamine release in obsessive-compulsive disorder. Biol Psychiatry [Internet]. 2014;75:647–652.
  • Buhmann C, Huckhagel T, Engel K, et al. Adverse events in deep brain stimulation: A retrospective long-term analysis of neurological, psychiatric and other occurrences. PLoS One. 2017;12:1–21.
  • Hubsher G, Haider M, Okun MS. Amantadine: the journey from fighting flu to treating Parkinson disease. Neurology. 2012;78:1096–1099.
  • Müller T, Möhr JD. Recent clinical advances in pharmacotherapy for levodopa-induced dyskinesia. Drugs [Internet]. 2019;79:1367–1374.
  • Pahwa R, Tanner CM, Hauser RA, et al. Amantadine extended release for levodopa-induced dyskinesia in Parkinson’s disease (EASED Study). Mov Disord. 2015;30:788–795.
  • Paik JKS. Amantadine extended-release (GOCOVRITM): a review in levodopa-induced dyskinesia in Parkinson’s disease. CNS Drugs. 2018;32:797–806.
  • Tanner CM, Pahwa R, Hauser RA, et al. EASE LID 2: A 2-year open-label trial of gocovri (Amantadine) extended release for dyskinesia in Parkinson’s disease. J Parkinsons Dis. 2020;10:543-558.
  • Fox SH, Brotchie JM. Viewpoint: developing drugs for levodopa-induced dyskinesia in PD: lessons learnt, what does the future hold? Eur J Neurosci. 2019;49:399–409.
  • Bastide MF, Meer WG, Picconi B, et al. Pathophysiology of L-dopa-induced motor and non-motor complications in Parkinson’s disease. Prog Neurobiol. 2015;132:96–168.
  • Bezard E, Brotchie JM, Gross CE. Pathophysiology of levodopa-induced dyskinesia: potential for new therapies. Nat Rev Neurosci. 2001;2:577–588.
  • Cedarbaum JM. Clinical pharmacokinetics of anti-parkinsonian drugs. Clin Pharmacokinet. 1987;13:141–178.
  • Gancher ST, Nutt JG, Woodward WR. Peripheral pharmacokinetics of levodopa in untreated, stable, and fluctuating parkinsonian patients. Neurology. 1987;37:940–944.
  • Huot P, Johnston TH, Koprich JB, et al. The pharmacology of L-DOPA-induced dyskinesia in Parkinson’s disease. Pharmacol Rev. 2013;65:171–222.
  • Dragašević-Mišković N, Petrović I, Stanković I, et al. Chemical management of levodopa-induced dyskinesia in Parkinson’s disease patients. Expert Opin Pharmacother. 2019;20:219–230.
  • Sebastianutto I, Cenci MA. mGlu receptors in the treatment of Parkinson’s disease and L-DOPA-induced dyskinesia. Curr Opin Pharmacol [Internet]. 2018;38:81–89.
  • Vegas-Suarez S, Paredes-Rodriguez E, Aristieta A, et al. Dysfunction of serotonergic neurons in Parkinson’s disease and dyskinesia [Internet]. 1st ed. International Review Neurobiology Elsevier Inc. Cambridge, MA, USA; 2019. DOI:10.1016/bs.irn.2019.06.013.
  • Picconi B, Hernández LF, Obeso JA, et al. Motor complications in Parkinson’s disease: striatal molecular and electrophysiological mechanisms of dyskinesias. Mov Disord. 2018;33:867–876.
  • Finlay C, Duty S. Therapeutic potential of targeting glutamate receptors in Parkinson’s disease. J Neural Transm. 2014;121:861–880.
  • Tahar AH, Grégoire L, Darré A, et al. Effect of a selective glutamate antagonist on L-dopa-induced dyskinesias in drug-naive parkinsonian monkeys. Neurobiol Dis. 2004;15:171–176.
  • Morin N, Grégoire L, Morissette M, et al. MPEP, an mGlu5 receptor antagonist, reduces the development of l-DOPA-induced motor complications in de novo parkinsonian monkeys: biochemical correlates. Neuropharmacology. 2013;66:355–364.
  • Morin N, Morissette M, Grégoire L, et al. mGlu5, Dopamine D 2 and Adenosine A 2A Receptors in L-DOPA-induced Dyskinesias. Curr Neuropharmacol. 2016;14:481–493.
  • Kim A, Kim YE, Yun JY, et al. Amantadine and the risk of dyskinesia in patients with early Parkinson’s disease: an open-label, pragmatic trial. J Mov Disord. 2018;11:65–71.
  • Alagarsamy S, Marino MJ, Rouse ST, et al. Activation of NMDA receptors reverses desensitization of mGluR5 in native and recombinant systems. Nat Neurosci. 1999;2:234–240.
  • Mela F, Marti M, Dekundy A, et al. Antagonism of metabotropic glutamate receptor type 5 attenuates L-DOPA-induced dyskinesia and its molecular and neurochemical correlates in a rat model of Parkinson’s disease. J Neurochem. 2007;101:483–497.
  • Ouattara B, Hoyer D, Grégoire L, et al. Changes of AMPA receptors in MPTP monkeys with levodopa-induced dyskinesias. Neuroscience. 2010;167:1160–1167.
  • Ouattara B, Belkhir S, Morissette M, et al. Implication of NMDA receptors in the antidyskinetic activity of cabergoline, CI-1041, and Ro 61-8048 in MPTP monkeys with levodopa-induced dyskinesias. J Mol Neurosci. 2009;38:128–142.
  • Rylander D, Recchia A, Mela F, et al. Pharmacological modulation of glutamate transmission in a rat model of L-DOPA-induced dyskinesia: effects on motor behavior and striatal nuclear signaling. J Pharmacol Exp Ther. 2009;330:227–235.
  • Johnston TH, Fox SH, McIldowie MJ, et al. Reduction of L-DOPA-induced dyskinesia by the selective metabotropic glutamate receptor 5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson’s disease. J Pharmacol Exp Ther. 2010;333:865–873.
  • Sid-Otmane L, Hamadjida A, Nuara SG, et al. Selective metabotropic glutamate receptor 2 positive allosteric modulation alleviates L-DOPA-induced psychosis-like behaviours and dyskinesia in the MPTP-lesioned marmoset. Eur J Pharmacol. 2020;873:172957.
  • Trenkwalder C, Stocchi F, Poewe W, et al. Mavoglurant in Parkinson’s patients with l-Dopa-induced dyskinesias: two randomized phase 2 studies. Mov Disord. 2016;31:1054–1058.
  • Charvin D. mGlu4 allosteric modulation for treating Parkinson’s disease. Neuropharmacology [Internet]. 2018;135:308–315.
  • Rovira X, Malhaire F, Scholler P, et al. Overlapping binding sites drive allosteric agonism and positive cooperativity in type 4 metabotropic glutamate receptors. Faseb J. 2015;29:116–130.
  • Danysz W, Zajaczkowski W, Parsons CG. Modulation of learning processes by ionotropic glutamate receptor ligands. Behav Pharmacol. 1995;6:455–474.
  • Therapeutics V. VistaGen announces positive preclinical data supporting AV-101’s potential for treating levodopa-induced dyskinesia in patients with Parkinson’s disease, without the psychological side effects and safety concerns of amantadine [Internet]. [cited 2020 Mar 18]. Available from: https://www.vistagen.com/news-media/press-releases/detail/122/vistagen-announces-positive-preclinical-data-supporting
  • Carta M, Carlsson T, Kirik D, et al. Dopamine released from 5-HT terminals is the cause of L-DOPA-induced dyskinesia in parkinsonian rats. Brain. 2007;130:1819–1833.
  • Carta M, Carlsson T, Muñoz A, et al. Serotonin-dopamine interaction in the induction and maintenance of L-DOPA-induced dyskinesias. Prog Brain Res. 2008;172:465–478.
  • Nevalainen N, Af Bjerkén S, Lundblad M, et al. Dopamine release from serotonergic nerve fibers is reduced in L -DOPA-induced dyskinesia. J Neurochem. 2011;118:12–23.
  • Cenci MA, Lundblad M. Post- versus presynaptic plasticity in L-DOPA-induced dyskinesia. J Neurochem. 2006;99:381–392.
  • Huot P, Fox SH. AN-T and JMB. anatomically selective serotonergic type 1A and serotonergic type 2A therapies for Parkinson’s disease: an approach to reducing dyskinesia without exacerbating Parkinsonism? J Pharmacol Exp Ther. 2011;339:2–8.
  • Eskow KL, Dupre KB, Barnum CJ, et al. The role of the dorsal raphe nucleus in the development, expression, and treatment of L-dopa-induced dyskinesia in hemiparkinsonian rats. Synapse. 2009;63:610–620.
  • Lindgren HS, Andersson DR, Lagerkvist S, et al. L-DOPA-induced dopamine efflux in the striatum and the substantia nigra in a rat model of Parkinson’s disease: temporal and quantitative relationship to the expression of dyskinesia. J Neurochem. 2010;112:1465–1476.
  • Muñoz A, Li Q, Gardoni F, et al. Combined 5-HT1A and 5-HT1B receptor agonists for the treatment of L-DOPA-induced dyskinesia. Brain. 2008;131:3380–3394.
  • Goetz CG, Laska E, Hicking C, et al. Placebo influences on dyskinesia in Parkinson’s disease. Mov Disord. 2008;23:700–707.
  • Grégoire L, Samadi P, Graham J, et al. Low doses of sarizotan reduce dyskinesias and maintain antiparkinsonian efficacy of l-Dopa in parkinsonian monkeys. Park Relat Disord. 2009;15:445–452.
  • Goetz CG, Damier P, Hicking C, et al. Sarizotan as a treatment for dyskinesias in Parkinson’s disease: A double-blind placebo-controlled trial. Mov Disord. 2007;22:179–186.
  • Dupre KB, Ostock CY, Eskow Jaunarajs KL, et al. Local modulation of striatal glutamate efflux by serotonin 1A receptor stimulation in dyskinetic, hemiparkinsonian rats. Exp Neurol. 2011;229:288–299.
  • Chen L, Togasaki DM, Langston JW, et al. Enhanced striatal opioid receptor-mediated G-protein activation in L-dopa-treated dyskinetic monkeys. Neuroscience. 2005;132:409–420.
  • Henry B, Fox SH, Crossman AR, et al. μ- and δ-Opioid receptor antagonists reduce levodopa-induced dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Exp Neurol. 2001;171:139–146.
  • Koprich JB, Fox SH, Johnston TH, et al. The selective mu-opioid receptor antagonist adl5510 reduces levodopa-induced dyskinesia without affecting antiparkinsonian action in mptp-lesioned macaque model of Parkinson’s disease. Mov Disord. 2011;26:1225–1233.
  • Potts LF, Park ES, Woo J, et al. Dual κ-agonist/μ-antagonist opioid receptor modulation reduces L-dopa induced dyskinesia and corrects dysregulated striatal changes in the non-human primate model of Parkinson’s disease. Ann Neurol. 2018;77:930–941.
  • Calabresi P, Di Filippo M, Ghiglieri V, et al. Levodopa-induced dyskinesias in patients with Parkinson’s disease: filling the bench-to-bedside gap. Lancet Neurol. 2010;9:1106–1117.
  • Heiman M, Heilbut A, Francardo V, et al. Molecular adaptations of striatal spiny projection neurons during levodopa-induced dyskinesia. Proc Natl Acad Sci U S A. 2014;111:4578–4583.
  • Lanza K, Chemakin K, Lefkowitz S, et al. Reciprocal cross-sensitization of D1 and D3 receptors following pharmacological stimulation in the hemiparkinsonian rat. Psychopharmacology (Berl). 2020;237:155–165.
  • Bézard E, Ferry S, Mach U, et al. Attenuation of levodopa-induced dyskinesia by normalizing dopamine D3 receptor function. Nat Med. 2003;9:762–767.
  • Hsu A, Togasaki DM, Bezard E, et al. Effect of the D3 dopamine receptor partial agonist BP897 [N-[4-(4-(2-methoxyphenyl)piperazinyl)butyl]-2-naphthamide] on L-3,4-dihydroxyphenylalanine-induced dyskinesias and parkinsonism in squirrel monkeys. J Pharmacol Exp Ther. 2004;311:770–777.
  • Visanji NP, Fox SH, Johnston T, et al. Dopamine D3 receptor stimulation underlies the development of L-DOPA-induced dyskinesia in animal models of Parkinson’s disease. Neurobiol Dis. 2009;35:184–192.
  • Huot P, Fox SH, Brotchie JM. Dopamine reuptake inhibitors in Parkinson’s disease: A review of nonhuman primate studies and clinical trials. J Pharmacol Exp Ther. 2016;357:562–569.
  • Huot P, Johnston TH, Koprich JB, et al. L-745, 870 reduces L -DOPA-induced dyskinesia in the 1-methyl- Parkinson ’ s disease. J Pharmacol Exp Ther. 2012;342:576–585.
  • Huot P, Johnston TH, Lewis KD, et al. UWA-121, a mixed dopamine and serotonin re-uptake inhibitor, enhances l-DOPA anti-parkinsonian action without worsening dyskinesia or psychosis-like behaviours in the MPTP-lesioned common marmoset. Neuropharmacology. 2014;82:76–87.
  • Johnston TH, Millar Z, Huot P, et al. A novel MDMA analogue, UWA-101, that lacks psychoactivity and cytotoxicity, enhances l -DOPA benefit in parkinsonian primates. Faseb J. 2012;26:2154–2163.
  • Giladi N, Caraco Y, Gurevich T, et al. ND0612, a novel liquid formulation of levodopa/carbidopa for subcutaneous infusion in patients with Parkinson’s disease achieves stable levodopa plasma levels when administered in low and high doses (S4.002). Neurology. 2017;88:S4.002.
  • NCT04006210. A clinical trial investigating the efficacy, safety and tolerability of continuous subcutaneous ND0612 infusion in comparison to oral IR-LD/CD in subjects with Parkinson’s disease experiencing motor fluctuations (BouNDless). [Internet]. [cited 2020 Apr 27]. Available from: https://clinicaltrials.gov/ct2/show/NCT04006210
  • Erdö SL, Kiss BRB. Inhibition of dopamine uptake by a new psychostimulant mesocarb (Sydnocarb®). Pol J Pharmacol Pharm. 1981;33:141–147.
  • CN105209445B. The method for treating dyskinesia and associated conditions [Internet]. [cited 2020 Mar 17]. Available from: https://patents.google.com/patent/CN105209445B/en
  • Melior Pharmaceuticals. Mesocarb [Internet]. [cited 2020 Mar 16]. Available from: http://www.meliorpharmaceuticals.com/mesocarb.html
  • Svenningsson P, Johansson A, Nyholm D, et al. Safety and tolerability of IRL790 in Parkinson’s disease with levodopa-induced dyskinesia—a phase 1b trial. npj Park Dis. 2018;4:1–5.
  • NCT03368170. Efficacy and tolerability of IRL790 in Parkinson’s disease dyskinesia [Internet]. [cited 2020 Apr 27]. Available from: https://clinicaltrials.gov/ct2/show/NCT03368170
  • Borgohain R, Szasz J, Stanzione P, et al. Two-Year, randomized, controlled study of safinamide as add-on to levodopa in mid to late Parkinson’s disease. Mov Disord. 2014;29:1273–1280.
  • Schapira AHV, Fox SH, Hauser RA, et al. Assessment of safety and efficacy of safinamide as a levodopa adjunct in patients with Parkinson disease and motor fluctuations a randomized clinical trial. JAMA Neurol. 2017;74:216–224.
  • Borgohain R, Szasz J, Stanzione P, et al. Randomized trial of safinamide add-on to levodopa in Parkinson’s disease with motor fluctuations. Mov Disord. 2014;29:229–237.
  • Cattaneo C, La Ferla R, Bonizzoni E, et al. Long-term effects of safinamide on dyskinesia in mid- to late-stage Parkinson’s disease: A post-hoc analysis. J Parkinsons Dis. 2015;5:475–481.
  • NCT03987750. Safinamide for Levodopa-induced Dyskinesia (PD-LID) [Internet]. [cited 2020 Apr 27]. Available from: https://clinicaltrials.gov/ct2/show/NCT03987750?recrs=ab&cond=Safinamide&draw=2&rank=2
  • Bezard E, Pioli EY, Li Q, et al. The mGluR5 negative allosteric modulator dipraglurant reduces dyskinesia in the MPTP macaque model. Mov Disord. 2014;29:1074–1079.
  • Tison F, Keywood C, Wakefield M, et al. A phase 2A trial of the novel mGluR5-negative allosteric modulator dipraglurant for levodopa-induced dyskinesia in Parkinson’s disease. Mov Disord. 2016;31:1373–1380.
  • Addex therapeutics. DIPRAGLURANT-IR FOR PD-LID [Internet]. [cited 2020 Mar 17]. Available from: https://www.addextherapeutics.com/en/pipeline/researches/dipraglurant-pd-lid/
  • Colpaert F. 5-HT1A receptor activation: new molecular and neuroadaptive mechanisms of pain relief. Curr Opin Investig DRUGS. 2006;7:40–47.
  • Colpaert FC, Tarayre JP, Koek W, et al. Large-amplitude 5-HT1A receptor activation: A new mechanism of profound, central analgesia. Neuropharmacology. 2002;43:945–958.
  • Iderberg H, McCreary AC, Varney MA, et al. NLX-112, a novel 5-HT1A receptor agonist for the treatment of l-DOPA-induced dyskinesia: behavioral and neurochemical profile in rat. Exp Neurol. 2015;271:335–350.
  • Biotech Parkinson’s Virtual. FDA approves Neurolixis IND application for clinical trial with NLX-112 in Parkinson’s disease [Internet]. [cited 2020 Mar 17]. Available from: https://www.parkinsonsvirtualbiotech.co.uk/single-post/2019/03/12/FDA-approves-Neurolixis-IND-application-for-clinical-trial-with-NLX-112-in-Parkinson’s-disease
  • Eison AS, Eison MS, Stanleyt M, et al. Serotonergic mechanisms in the behavioral effects of buspirone and gepirone. Pharmacol Biochem Behav. 1986;24:701–707.
  • Lechin F, Van Der Dijs B, Pardey-Maldonado B, et al. Effects of amantadine on circulating neurotransmitters in healthy subjects. J Neural Transm. 2010;117:293–299.
  • Eskow KL, Gupta V, Alam S, et al. The partial 5-HT1A agonist buspirone reduces the expression and development of l-DOPA-induced dyskinesia in rats and improves l-DOPA efficacy. Pharmacol Biochem Behav. 2007;87:306–314.
  • Bonifati V, Fabrizio E, Cipriani R, et al. Buspirone in levodopa-induced dyskinesias. Clin Neuropharmacol. 1994;17:73–82.
  • Kleedorfer B, Lees AJ, Stern GM. Buspirone in the treatment of levodopa induced dyskinesias [10]. J Neurol Neurosurg Psychiatry. 1991;54:376–377.
  • Politis M, Wu K, Loane C, et al. Serotonergic mechanisms responsible for levodopa-induced dyskinesias in Parkinson’s disease patients. J Clin Invest. 2014;124:1340–1349.
  • NCT02617017. Buspirone treatment of iatrogenic dyskinesias in advanced Parkinson’ Disease (BUSPARK) [Internet]. [cited 2020 Mar 17]. Available from: https://clinicaltrials.gov/ct2/show/NCT02617017
  • NCT02589340. Buspirone, in combination with amantadine, for the treatment of levodopa-induced dyskinesia (BUS-PD) [Internet]. [cited 2020 Mar 18]. Available from: https://clinicaltrials.gov/ct2/show/NCT02589340
  • NCT02439203. Efficacy and safety of JM-010 in PD with Levodopa-Induced Dyskinesia (LID) [Internet]. [cited 2020 Mar 18]. Available from: https://clinicaltrials.gov/ct2/show/NCT02439203
  • Contera Pharma. European clinical phase 2 study of JM-010 to treat dyskinesia in Parkinson’s disease has been started [Internet]. [cited 2020 Mar 18]. Available from: http://www.conterapharma.com/page/news.html
  • NCT03956979. A study in Parkinson’s disease in paTients with mOderate to seveRe dyskInesiA (ASTORIA) [Internet]. [cited 2020 Apr 27]. Available from: https://clinicaltrials.gov/ct2/show/NCT03956979
  • Svenningsson P, Rosenblad C, Arvidsson KAE, et al. Eltoprazine counteracts l-DOPA-induced dyskinesias in Parkinson’s disease: A dose-finding study. Brain. 2015;138:963–973.
  • NCT02439125. A study of efficacy and safety of eltoprazine HCl for treating levodopa-induced dyskinesia in Parkinson’s Disease patients [Internet]. [cited 2020 Apr 27]. Available from: https://clinicaltrials.gov/ct2/show/NCT02439125
  • Kemp JA, Foster AC, Leeson PD, et al. 7-Chlorokynurenic acid is a selective antagonist at the glycine modulatory site of the N-methyl-D-aspartate receptor complex. Proc Natl Acad Sci U S A. 1988;85:6547–6550.
  • NCT04147949. AV-101 (L-4-chlorokynurenine) in Parkinson’s disease subjects with levodopa-induced dyskinesia. [cited 2020 Apr 27]. Available from: https://clinicaltrials.gov/ct2/show/NCT04147949
  • Trevi Therapeutics. A pipeline in a product: nalbuphine ER [Internet]. [cited 2020 Apr 27]. Available from: https://www.trevitherapeutics.com/pipeline/
  • Waters S, Tedroff J, Ponten H, et al. Pridopidine: overview of pharmacology and rationale for its use in Huntington’s disease. J Huntingtons Dis. 2018;7:1–16.
  • Johnston TH, Lacoste AMB, Visanji NP, et al. Repurposing drugs to treat L-DOPA-induced dyskinesia in Parkinson’s disease. Neuropharmacology. 2019;147:11–27.
  • Ponten H, Kullingsjö J, Sonesson C, et al. The dopaminergic stabilizer pridopidine decreases expression of l-DOPA-induced locomotor sensitisation in the rat unilateral 6-OHDA model. Eur J Pharmacol. 2013;698:278–285.
  • NCT03922711. A study to assess the safety and effectiveness of pridopidine compared to placebo in the treatment of levodopa-induced dyskinesia in patients with Parkinson’s disease [Internet]. [cited 2020 Apr 27]. Available from: https://clinicaltrials.gov/ct2/show/NCT03922711
  • Biton V. Clinical pharmacology and mechanism of action of zonisamide. Clin Neuropharmacol. 2007;30:230–240.
  • Murata M, Hasegawa K, Kanazawa I, et al. Zonisamide improves wearing-off in Parkinson’s disease: A randomized, double-blind study. Mov Disord. 2015;30:1343–1350.
  • Oki M, Kaneko S, Morise S, et al. Zonisamide ameliorates levodopa-induced dyskinesia and reduces expression of striatal genes in Parkinson model rats. Neurosci Res. 2017;122:45–50.
  • Sano H, Nambu A. The effects of zonisamide on L-DOPA–induced dyskinesia in Parkinson’s disease model mice. Neurochem Int. 2019;124:171–180.
  • NCT03034538. An open-label tolerability and exploratory efficacy study of zonisamide for dyskinesias in Parkinson’s disease [Internet]. [cited 2020 Mar 18]. Available from: https://clinicaltrials.gov/ct2/show/NCT03034538
  • Neuringer M. The essentiality of N-3 fatty acids for the development and function of the retina and brain. Annu Rev Nutr. 1988;8:517–541.
  • Gómez-Soler M, Cordobilla B, Morató X, et al. Triglyceride form of docosahexaenoic acid mediates neuroprotection in experimental parkinsonism. Front Neurosci. 2018;12:1–11.
  • Mata A, Liu S, Sjöberg M, et al. Docosahexaenoic acid, a ligand for the retinoid X receptor in mouse brain. Science. 2000;290:2140–2144.
  • Éthier I, Kagechika H, Shudo K, et al. Docosahexaenoic acid reduces haloperidol-induced dyskinesias in mice: involvement of Nur77 and retinoid receptors. Biol Psychiatry. 2004;56:522–526.
  • Samadi P, Grégoire L, Rouillard C, et al. Docosahexaenoic acid reduces levodopa-induced dyskinesias in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine monkeys. Ann Neurol. 2006;59:282–288.
  • NCT01563913. Reducing dyskinesia in Parkinson’s disease with omega-3 fatty acids (RLID-PD) [Internet]. [cited 2020 Mar 18]. Available from: https://clinicaltrials.gov/ct2/show/NCT01563913
  • Ayar A, Storer C, Tatham EL, et al. Naftazone reduces glutamate cerebro spinal fluid levels in rats and glutamate release from mouse cerebellum synaptosomes. Neurosci Lett. 1999;271:171–174.
  • Corvol JC, Durif F, Meer WG, et al. Naftazone in advanced Parkinson’s disease: an acute L-DOPA challenge randomized controlled trial. Park Relat Disord. 2019;60:51–56.
  • Brotchie J, Johnston T, Fox S, et al. FP0011 extends the duration of the anti-parkinsonian actions of L-DOPA and reduces L-DOPA-induced dyskinesia in the MPTP-lesiones macaque model of Parkinson’s disease. Mov Disord. 2007;22:S96.
  • Schunack. Pharmacology of H2-receptor antagonists: an overview. J Int Med Res. 1988;17:9A–16A.
  • Mestre TA, Shah BB, Connolly BS, et al. Famotidine, a histamine H2 receptor antagonist, does not reduce levodopa-induced dyskinesia in Parkinson’s disease: a proof-of-concept study. Mov Disord Clin Pract. 2014;1:219–224.
  • Johnston TH, Van Der Meij A, Brotchie JM, et al. Effect of histamine H2 receptor antagonism on levodopa-induced dyskinesia in the MPTP-macaque model of Parkinson’s disease. Mov Disord. 2010;25:1379–1390.
  • Goetz CG, Stebbins GT, Chung KA, et al. Topiramate as an adjunct to amantadine in the treatment of dyskinesia in parkinson’s disease: A randomized, double-blind, placebo-controlled multicenter study. Mov Disord. 2017;32:1335–1336.
  • Silverdale MA, Nicholson SL, Crossman AR, et al. Topiramate reduces levodopa-induced dyskinesia in the MPTP-lesioned marmoset model of Parkinson’s disease. Mov Disord. 2005;20:403–409.
  • Kobylecki C, Hill MP, Crossman AR, et al. Synergistic antidyskinetic effects of topiramate and amantadine in animal models of Parkinson’s disease. Mov Disord. 2011;26:2354–2363.
  • Kobylecki C, Burn DJ, Kass-Iliyya L, et al. Randomized clinical trial of topiramate for levodopa-induced dyskinesia in Parkinson’s disease. Park Relat Disord. 2014;20:452–455.
  • Huot P, Sgambato-Faure V, Fox SH, et al. Serotonergic approaches in Parkinson’s disease: translational perspectives, an update. ACS Chem Neurosci. 2017;8:973–986.
  • Hung SW, Adeli GM, Arenovich T, et al. Patient perception of dyskinesia in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2010;81:1112–1115.
  • Papapetropoulos SS. Patient diaries as a clinical endpoint in Parkinson’s disease clinical trials. CNS Neurosci Ther. 2012;18:380–387.
  • Stone AA, Shiffman S, Schwartz JE, et al. Patient non-compliance with paper diaries. Br Med J. 2002;324:1193–1194.
  • Ossig C, Antonini A, Buhmann C, et al. Wearable sensor-based objective assessment of motor symptoms in Parkinson’s disease. J Neural Transm. 2016;123:57–64.
  • Lopane G, Mellone S, Chiari L, et al. Dyskinesia detection and monitoring by a single sensor in patients with Parkinson’s disease. Mov Disord. 2015;30:1267–1271.
  • Delrobaei M, Baktash N, Gilmore G, et al. Using wearable technology to generate objective Parkinson’s disease dyskinesia severity score: possibilities for home monitoring. IEEE Trans Neural Syst Rehabil Eng. 2017;25:1853–1863.
  • Quattrone A, Barbagallo G, Cerasa A, et al. Neurobiology of placebo effect in Parkinson’s disease: what we have learned and where we are going. Mov Disord. 2018;33:1213–1227.
  • Titova N, Chaudhuri KR. Personalized medicine in Parkinson’s disease: time to be precise. Mov Disord. 2017;32:1147–1154.
  • Espay AJ, Kalia LV, Gan-Or Z, et al. Disease modification and biomarker development in Parkinson disease. Neurology. 2020;94:481–494.
  • Kieburtz K, Katz R, Olanow CW. New drugs for Parkinson’s disease: the regulatory and clinical development pathways in the United States. Mov Disord. 2018;33:920–927.
  • Lundbeck. Lundbeck reports headline results from phase IIa AMBLED study of foliglurax in Parkinson’s disease [Internet]. [cited 2020 Apr 27]. Available from: https://investor.lundbeck.com/news-releases/news-release-details/lundbeck-reports-headline-results-phase-iia-ambled-study

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.