813
Views
38
CrossRef citations to date
0
Altmetric
Review

Therapeutic strategies to prevent and manage dyskinesias in Parkinson’s disease

, MD & , MD

Bibliography

  • Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov Disord 2001;16(3):448-58
  • Fahn S. The spectrum of levodopa-induced dyskinesias. Ann Neurol 2000;47(4 Suppl 1):S2-11
  • Di Monte DA, McCormack A, Petzinger G, et al. Relationship among nigrostriatal denervation, parkinsonism, and dyskinesias in the MPTP primate model. Mov Disord 2000;15(3):459-66
  • Paillé V, Brachet P, Damier P. Role of nigral lesion in the genesis of dyskinesias in a rat model of Parkinson’s disease. Neuroreport 2004;15(3):561-4
  • Lyons KE, Hubbie JP, Troster AI, et al. Gender differences in Parkinson’s disease. Clin Neuropharmacol 1998;21(2):118-21
  • Richmann CG, Zapf A, Brunner E, et al. Dopaminergic treatment is associated with decreased body weight in patients with Parkinson’s disease and dyskinesias. Eur J Neurol 2009;16(8):895-901
  • Ku S, Glass G. Age of Parkinson’s disease onset as a predictor for the development of dyskinesia. Mov Disord 2010;25(9):1177-82
  • Quinn N, Critchely P, Marsden CD. Young onset Parkinson’s disease. Mov Disord 1987;2(2):73-91
  • Zhang YH, Tang BS, Song CY, et al. The relationship between the phenotype of Parkinson’s disease and levodopa-induced dyskinesia. Neurosci Lett 2013;556:109-12
  • Pechevis M, Clarke CE, Vieregge P, et al. Direct and indirect costsof Parkinson’s disease and L-dopa induced dyskinesias: a prospectiveEuropean study. Parkinsonism Relat Disord 2001;7(Suppl):106
  • Schrag A, Quinn N. Dyskinesias and motor fluctuations in Parkinson’s disease. A community-based study. Brain 2000;123:2297-305
  • Fahn S, Oakes D, Shoulson I, et al. Does levodopa slow or hasten the rate of progression of Parkinson disease? The results of the Elldopa study. N Eng J Med 2004;351(24):2498-508
  • Calabresi P, Di Filippo M, Ghiglieri V, et al. Molecular mechanisms underlying levodopa-induced dyskinesia. Mov Disord 2008;23(Suppl 3):S570-9
  • Ballard PA, Tetrud JW, Langston JW. Permanent human parkinsonism due to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): seven cases. Neurology 1985;35(7):949-56
  • Cilia R, Akpalu A, Cham M, et al. Parkinson’s disease in sub Saharan Africa: step-by-step into the challenge. Neurodegen Dis Manage 2011;1:193-202
  • Nutt JG. Levodopa-induced dyskinesia: review, observations and speculations. Neurology 1990;40:340-5
  • De la Fuente-Fernandez R, Sossi V, Huang Z, et al. Levodopa-induced changes in synaptic dopamine levels increase with progression of Parkinson’s disease: implications for dyskinesias. Brain 2004;127:2747-54
  • Jenner P. Avoidance of dyskinesia: preclinical evidence for continuous dopaminergic stimulation. Neurology 2004;62(Suppl 1):S47-55
  • Cenci MA, Konradi C. Maladaptive striatal plasticity in L-DOPA-induced dyskinesia. Prog Brain Res 2010;183:209-33
  • Pisani A, Shen J. Levodopa-induceddyskinesia and striatalsignalingpathwaysProc Natl Acad Sci USA. 2009;106(9):2973-4
  • Calon F, Rajput AH, Hornykiewicz O, et al. Levodopa-induced motor complications are associated with alterations of glutamate receptors in Parkinson’s disease. Neurobiol Dis 2003;14(3):404-6
  • Chase TN, Engber TM, Mouradian MM. Contribution of dopaminergic and glutamatergic mechanisms to the pathogenesis of motor response complications in Parkinson’s disease. Adv Neurol 1996;69:497-501
  • Arai R, Karasawa N, Geffard M, et al. Immunohistochemical evidence that central serotonin neurons produce dopamine from exogenous L-DOPA in the rat, with reference to the involvement of aromatic L-amino acid decarboxylase. Brain Res 1994;667(2):295-9
  • Carlsson T, Carta M, Winkler C, et al. Serotonin neuron transplants exacerbate L-DOPA-induced dyskinesias in a rat model of Parkinson’s disease. J Neuro sci 2007;27(30):8011-22
  • Querejeta E, Martínez-Romero B, Miranda JE, et al. Modulation of the striato-pallidal pathway by adenosineA2areceptors depends on dopaminergic striatal input. Brain Res 2010;1349:137-42
  • Fox SH, Henry B, Hill MP, et al. Neural mechanisms underlying peak-dose dyskinesia induced by levodopa and apomorphine are distinct: evidence from the effects of the alpha(2) adrenoceptor antagonist idazoxan. Mov Disord 2001;16(4):642-50
  • Rascol O, Goetz C, Koller W, et al. Treatment interventions for Parkinson’s disease: an evidence-based assessment. Lancet 2002;359(9317):1589-98
  • Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. 065 Study Group. N Engl J Med 2000;342(20):1481-91
  • Holloway RG, Shoulson I, Fahn S, et al. Pramipexole vs levodopaas initial treatment for Parkinson’s disease: a 4-yearrandomized controlled trial. Arch Neurol 2004;61(7):1044-53
  • Olanow CW, Fahn S, Muenter M, et al. A multi-center, double-blind, placebo controlled trial of pergolide as an adjunct to Sinemet in Parkinson’s disease. Mov Disord 1994;9(1):40-7
  • Bracco F, Battaglia A, Chouza C, et al. PKDS009 study group. The long acting DA cabergoline in early Parkinson’s disease: final results of five year, levodopa controlled study. CNS Drugs 2004;18(11):733-46
  • Jankovic J, Watts RL, Martin W, et al. Transdermal rotigotine: double-blind, placebo-controlled trial in Parkinson disease. Arch Neurol 2007;64(5):676-82
  • Poewe WH, Rascol O, Quinn N, et al. Efficacy of pramipexole and transdermal rotigotine in advanced Parkinson’s disease: a double-blind, double-dummy, randomised controlled trial. Lancet Neurol 2007;6(6):513-20
  • Giladi N, Boroojerdi B, Surmann E. The safety and tolerability of rotigotine transdermal system over a 6-year period in patients with early-stage Parkinson’s disease. J Neural Transm 2013;120(9):1321-9
  • Pahwa R, Stacy MA, Factor SA, et al. EASE-PD. Ropinirole 24-hour prolonged release: randomized. controlled study in advanced Parkinson disease Neurology 2007;68(14):1108-15
  • Zhou CQ, Lou JH, Zhang YP, et al. Long-acting versus standard non-ergot dopamine agonists in Parkinson’s disease: a meta-analysis of randomized controlled trials. CNS Neurosci Ther 2014;20(4):368-76
  • Malik P, Andersen MB, Peacock L. The effects of dopamine D3 agonists and antagonists in a nonhuman primate model of tardive dyskinesia. Pharmacol Biochem Behav 2004;78(4):805-10
  • Bézard E, Ferry S, Mach U, et al. Attenuation of levodopa induced dyskinesia by normalizing dopamine D3 receptor function. Nat Med 2003;9(6):762-7
  • Ohno H, Nakajima M, Fujioka S, et al. Overnight switching from ergot-derived dopamine agonists to pramipexole in patients with Parkinson’s disease: an open preliminary trial in Japan. J ClinNeurosci 2009;16(6):790-2
  • Stowe RL, Ives NJ, Clarke C, et al. Dopamine agonist therapy in early Parkinson’s disease. Cochrane Database Syst Rev 2008(2):CD006564
  • Lees AJ, Katzenschlager R, Head J, et al. onbehalf of the Parkinson’s Disease Research Group of the UK. Ten-year follow-up of three different initial treatments in de-novo PD: a randomized trial. Neurology 2001;57(9):1687-94
  • Lieberman A, Olanow CW, Sethi K, et al. A multi-center double blind placebo-controlled trial of ropinirole as an adjunct to L-dopa in the treatment of Parkinson’s disease patients with motor fluctuations. Neurology 1998;51(4):1057-62
  • Pinter MM, Pogarell O, Oertel WH. Efficacy, safety, and tolerance of the non-ergoline dopamine agonist pramipexole in the treatment of advanced Parkinson’s disease: a double blind,placebo controlled, randomised, multicentre study. J Neurol Neurosurg Psychiatry 1999;66(4):436-41
  • Calandrella D, Antonini A. Pulsatile or continuous dopaminomimetic strategies in Parkinson’s disease. Parkinsonism Relat Disord 2012;18(Suppl 1):S120-2
  • Deleu D, Hanssens Y, Northway MG. Subcutaneous apomorphine : an evidence-based review of its use in Parkinson’s disease. Drugs Aging 2004;21(11):687-709
  • Colzi AK, Turner K, Lees AJ. Continuous subcutaneous waking day apomorphine in the long term treatment of levodopa induced interdosedyskinesias in Parkinson’s disease. J Neurol Neurosurg Psychiatry 1998;64(5):573-5
  • Manson AJ, Turner K, Lees AJ. Apomorphine monotherapy in the treatment of refractory motor complications of Parkinson’s disease: long-term follow-up study of 64 patients. Mov Disord 2002;17(6):1235-41
  • De Gaspari D, Siri C, Landi A, et al. Clinical and neuropsychological follow up at 12 months in patients with complicated Parkinson’s disease treated with subcutaneous apomorphine infusion or deep brain stimulation of the subthalamic nucleus. J Neurol Neurosurg Psychiatry 2006;77(4):450-3
  • García Ruiz PJ, Sesar Ignacio A, Ares Pensado B, et al. Efficacy of long-term continuous subcutaneous apomorphine infusion in advanced Parkinson’s disease with motor fluctuations: a multicenterstudy. Mov Disord 2008;23(8):1130-6
  • Nyholm D, Nilsson Remahl AI, Dizdar N, et al. Duodenal levodopainfusion monotherapy vs oral polypharmacy in advanced Parkinson disease. Neurology 2005;64(2):216-23
  • Antonini A, Odin P, Lopiano L, et al. Effect and safety of duodenal levodopa infusion in advanced Parkinson’s disease: a retrospective multicenter outcome assessment in patient routine care. J Neural Transm 2013;120(11):1553-8
  • Olanow CW, Kieburtz K, Odin P, et al. Continuousintrajejunalinfusion of levodopa-carbidopa intestinal gel for patients with advanced Parkinson’s disease: a randomised, controlled, double-blind, double-dummy study. Lancet Neurol 2014;13(2):141-9
  • Zibetti M, Merola A, Artusi CA, et al. Levodopa/carbidopa intestinal gel infusion in advanced Parkinson’s disease: a 7-year experience. Eur J Neurol 2014;21(2):312-18
  • LeWitt PA, Nelson MV, Berchou RC, et al. Controlled-release carbidopa/levodopa (Sinemet 50/200 CR4): clinical and pharmacokinetic studies. Neurology 1989;39(11 Suppl 2):45-53
  • Yeh KC, August TF, Bush DF, et al. Pharmacokinetics and bioavailability of Sinemet CR: a summary of human studies. Neurology 1989;39(11 Suppl 2):25-38
  • Olanow CW, Kieburtz K, Rascol O, et al. Stalevo Reduction in Dyskinesia Evaluation in Parkinson’s Disease (STRIDE-PD) Investigators. Mov Disord 2013
  • Hauser RA, Hsu A, Kell S, et al. Extended-release carbidopa-levodopa (IPX066) compared with immediate-release carbidopa-levodopa in patients with Parkinson’s disease and motor fluctuations: a phase 3 randomised, double-blind trial. Lancet Neurol 2013;12(4):346-56
  • Guigoni C, Aubert I, Li Q, et al. Pathogenesis of levodopa-induced dyskinesia: focus on D1 and D3 dopamine receptors. Parkinsonism Relat Disord 2005;11:S25-9
  • Meco G, Stirpe P, Edito F, et al. Aripiprazole in L-dopa-induced dyskinesias: a one-year open-label pilot study. J Neural Transm 2009;116(7):881-4
  • Sweeney EB, Lawlor BA. Case series: extrapyramidal symptoms associated with use of aripiprazole in older adults. Int J Geriatr Psychiatry 2013;28(11):1208-10
  • Johnston LC, Jackson MJ, Rose S, et al. Pardoprunox reverses motor deficits but induces only mild dyskinesia in MPTP-treated common marmosets. Mov Disord 2010;25(13):2059-66
  • Rascol O, Bronzova J, Hauser RA, et al. Pardoprunox as adjunct therapy to levodopa in patients with Parkinson’s disease experiencing motor fluctuations: results of a double-blind, randomized, placebo-controlled, trial. Parkinsonism Relat Disord 2012;18(4):370-6
  • Da Silva-Júnior FP, Braga-Neto P, Sueli Monte F, et al. Amantadine reduces the duration of levodopa-induced dyskinesia: a randomized, double-blind, placebo-controlled study. Parkinsonism Relat Disord 2005;11(7):449-52
  • Thomas A, Iacono D, Luciano AL, et al. Duration of amantadine benefit on dyskinesia of severe Parkinson disease. J Neurol Neurosurg Psychiatry 2004;75(1):141-3
  • Wolf E, Seppi K, Katzenschlager R, et al. Long-term antidyskinetic efficacy of amantadine in Parkinson’s disease. Mov Disord 2010;25(10):1357-63
  • Ory-Magne F, Corvol JC, Azulay JP, et al. Withdrawing amantadine in dyskinetic patients with Parkinson disease: the AMANDYSK trial. Neurology 2014;82(4):300-7
  • Moreau C, Delval A, Tiffreau V, et al. Memantine for axial signs in Parkinson’s disease: a randomised, double-blind, placebo-controlled pilot study. J Neurol Neurosurg Psychiatry 2013;84(5):552-5
  • Stocchi F, Rascol O, Destee A, et al. AFQ056 in Parkinson patients with levodopa-induced dyskinesia: 13-week, randomized, dose-finding study. Mov Disord 2013;28(13):1838-46
  • Rascol O, Fox S, Gasparini F, et al. Use of metabotropic glutamate 5-receptor antagonists for treatment of levodopa-induced dyskinesias. Parkinsonism Relat Disord 2014;20(9):947-56
  • Lees A, Fahn S, Eggert KM, et al. Perampanel, an AMPA antagonist, found to have no benefit in reducing ‘‘off’’ time in Parkinson’sdisease. Mov Disord 2012;27(2):284-8
  • Bara-Jimenez W, Bibbiani F, Morris MJ, et al. Effects of serotonin 5-HT1A agonist in advanced Parkinson’s disease. Mov Disord 2005;20(8):932-6
  • Durif F, Debilly B, Galitzky M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology 2004;62(3):381-8
  • Guenette MD, Powell V, Johnston K, et al. Risk of neutropenia in a clozapine-treated elderly population. Schizophr Res 2013;148(1-3):183-5
  • 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(2):179-86
  • 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(Pt 12):3380-94
  • Bara-Jimenez W, Sherzai A, Dimitrova T, et al. Adenosine A(2A) receptor antagonist treatment of Parkinson’s disease. Neurology 2003;61(3):293-6
  • Hauser RA, Cantillon M, Pourcher E, et al. Preladenant in patients with Parkinson’s disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol 2011;10(3):221-9
  • Factor SA, Wolski K, Togasaki DM, et al. Long-term safety and efficacy of preladenant in subjects with fluctuating Parkinson’s disease. Mov Disord 2013;28(6):817-20
  • Hauser RA, Shulman LM, Trugman JM, et al. Study of istradefylline in patients with Parkinson’s disease on levodopa with motor fluctuations. Mov Disord 2008;23(15):2177-85
  • LeWitt PA, Guttman M, Tetrud JW, et al. Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces “off” time in Parkinson’s disease: a double-blind, randomized, multicenter clinical trial (6002-US-005). Ann Neurol 2008;63(3):295-302
  • Mizuno Y, Hasegawa K, Kondo T, et al. Clinical efficacy of istradefylline (KW-6002) in Parkinson’s disease: a randomized, controlled study. Mov Disord 2010;25(10):1437-43
  • Pourcher E, Fernandez HH, Stacy M, et al. Istradefylline for Parkinson’s disease patients experiencing motor fluctuations: results of the KW-6002-US-018 study. Parkinsonism Relat Disord 2012;18(2):178-84
  • Pisani V, Madeo G, Tassone A, et al. Homeostatic changes of the endocannabinoid system in Parkinson’s disease. Mov Disord 2011;26(2):216-22
  • Sieradzan KA, Fox SH, Hill M, et al. Cannabinoids reduce levodopa-induced dyskinesia in Parkinson’s disease: a pilot study. Neurology 2001;57(11):2108-11
  • González-Aparicio R, Moratalla R. Oleoylethanolamide reduces L-DOPA-induced dyskinesia via TRPV1 receptor in a mouse model of Parkinson´s disease. Neurobiol Dis 2014;62:416-25
  • Carroll CB, Bain PG, Teare L, et al. Cannabis for dyskinesia in Parkinson disease: a randomized double-blind crossover study. Neurology 2004;63(7):1245-50
  • Lewitt PA, Hauser RA, Lu M, et al. Randomized clinical trial of fipamezole for dyskinesia in Parkinson disease (FJORD study). Neurology 2012;79(2):163-9
  • Rascol O, Arnulf I, Peyro-Saint Paul H, et al. Idazoxan, an alpha-2 antagonist, and L-DOPA-induced dyskinesias in patients with Parkinson’s disease. Mov Disord 2001;16(4):708-13
  • Manson AJ, Iakovidou E, Lees AJ. Idazoxan is ineffective for levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord 2000;15(2):336-7
  • Bezard E, Hill MP, Grossman AR, et al. Levetiracetam improves choreic levodopa-induced dyskinesia in the MPTP-treated macaque. Eur J Pharmacol 2004;485(1–3):159-64
  • Zesiewicz TA, Sullivan KL, Maldonado JL, et al. Open-label pilot study of levetiracetam (Keppra) for the treatment of levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord 2005;20(9):1205-9
  • Stathis P, Konitsiotis S, Tagaris G, et al. VALID-PD Study Group. Levetiracetam for the management of levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord 2011;26(2):264-70
  • Lyons KE, Pahwa R. Efficacy and tolerability of levetiracetam in Parkinson disease patients with levodopa-induced dyskinesia. Clin Neuropharmacol 2006;29(3):148-53
  • 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(10):1379-90
  • Anderson VC, Burchiel K, Hogarth P, et al. Pallidal vs subthalamic nucleus deep brain stimulation in Parkinson’s disease. Arch Neurol 2005;62(4):554-60
  • The Deep-Brain Stimulation for Parkinson’s Disease Study Group. Deep-brain stimulation of the subthalamic nucleus or the pars interna of the globuspallidus in Parkinson’s disease. N EngI J Med 2001;345(13):956-63
  • Olanow CW, Obeso JA. Preventing levodopa-induced dyskinesia. Ann Neurol 2000;47(4 Suppl 1):167-78
  • Brodsky MA, Godbold J, Roth T, et al. Sleepiness in Parkinson’s disease: a controlled study. Mov Disord 2003;18(6):668-72
  • Driver-Dunckley E, Samanta J, Stacy M. Pathological gambling associated with dopamine agonist therapy in Parkinson’s disease. Neurology 2003;61(3):422-3
  • Nirenberg MJ, Waters C. Compulsive eating and weight gain related to dopamine agonist use. Mov Disord 2006;21:524-9
  • Cilia R, Akpalu A, Sarfo FS, et al. The modern pre-levodopa era of Parkinson’s disease: insights into motor complications from sub-Saharan Africa. Brain 2014;137(Pt 10):2731-42
  • Zanettini R, Antonini A, Gatto G, et al. Valvular heart disease and the use of dopamine agonists for Parkinson’s disease. N Engl J Med 2007;356(1):39-646
  • Antonini A, Poewe W. Fibrotic heart-valve reactions to dopamine agonist treatment in Parkinson’s disease. Lancet Neurol 2007;6(9):826-9
  • Parkinson Study Group. A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations. The PRESTO study. Arch Neurol 2005;62(2):241-3
  • Olanow CW, Goetz CG, Kordower JH, et al. A double-blind controlled trial of bilateral fetalnigral transplantation in Parkinson’s disease. Ann Neurol 2003;54(3):403-14

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.