826
Views
49
CrossRef citations to date
0
Altmetric
Reviews

Dopamine receptor agonists for Parkinson's disease

&

Bibliography

  • Cheon SM, Ha MS, Park MJ, Kim JW. Nonmotor symptoms of Parkinson's disease: prevalence and awareness of patients and families. Parkinsonism Relat Disord 2008;14(4):286-90
  • Gallagher DA, Lees AJ, Schrag A. What are the most important nonmotor symptoms in patients with Parkinson's disease and are we missing them? Mov Disord 2010;25(15):2493-500
  • Jellinger KA. Pathology of Parkinson's disease. Changes other than the nigrostriatal pathway. Mol Chem Neuropathol 1991;14(3):153-97
  • de Rijk MC, Tzourio C, Breteler MM, et al. Prevalence of parkinsonism and Parkinson's disease in Europe: the EUROPARKINSON Collaborative Study. European Community Concerted Action on the Epidemiology of Parkinson's disease. J Neurol Neurosurg Psychiatry 1997;62(1):10-15
  • Greenamyre JT, Hastings TG. Biomedicine. Parkinson's–divergent causes, convergent mechanisms. Science 2004;304(5674):1120-2
  • Hirsch EC, Hunot S. Neuroinflammation in Parkinson's disease: a target for neuroprotection? Lancet Neurol 2009;8(4):382-97
  • Blandini F. Neural and immune mechanisms in the pathogenesis of Parkinson's disease. J Neuroimmune Pharmacol 2013;8(1):189-201
  • Blandini F, Nappi G, Tassorelli C, Martignoni E. Functional changes of the basal ganglia circuitry in Parkinson's disease. Prog Neurobiol 2000;62(1):63-88
  • Obeso JA, Olanow CW, Nutt JG. Levodopa motor complications in Parkinson's disease. Trends Neurosci 2000;23(10 Suppl):S2-7
  • Fabbrini G, Brotchie JM, Grandas F, et al. Levodopa-induced dyskinesias. Mov Disord 2007;22(10):1379-89
  • Schrag A, Hovris A, Morley D, et al. Young- versus older-onset Parkinson's disease: impact of disease and psychosocial consequences. Mov Disord 2003;18(11):1250-6
  • Zappia M, Annesi G, Nicoletti G, et al. Sex differences in clinical and genetic determinants of levodopa peak-dose dyskinesias in Parkinson disease: an exploratory study. Arch Neurol 2005;62(4):601-5
  • Carta M, Carlsson T, Kirik D, Bjorklund A. Dopamine released from 5-HT terminals is the cause of L-DOPA-induced dyskinesia in parkinsonian rats. Brain 2007;130(Pt 7):1819-33
  • Tanaka H, Kannari K, Maeda T, et al. Role of serotonergic neurons in L-DOPA-derived extracellular dopamine in the striatum of 6-OHDA-lesioned rats. Neuroreport 1999;10(3):631-4
  • Blandini F, Armentero MT. New pharmacological avenues for the treatment of L-DOPA-induced dyskinesias in Parkinson's disease: targeting glutamate and adenosine receptors. Expert Opin Investig Drugs 2012;21(2):153-68
  • Armentero MT, Pinna A, Ferre S, et al. Past, present and future of A(2A) adenosine receptor antagonists in the therapy of Parkinson's disease. Pharmacol Ther 2011;132(3):280-99
  • Picconi B, Centonze D, Hakansson K, et al. Loss of bidirectional striatal synaptic plasticity in L-DOPA-induced dyskinesia. Nat Neurosci 2003;6(5):501-6
  • Konradi C, Westin JE, Carta M, et al. Transcriptome analysis in a rat model of L-DOPA-induced dyskinesia. Neurobiol Dis 2004;17(2):219-36
  • Gershanik O, Jenner P. Moving from continuous dopaminergic stimulation to continuous drug delivery in the treatment of Parkinson's disease. Eur J Neurol 2012;19(12):1502-8
  • Poewe W. Adjuncts to levodopa therapy: dopamine agonists. Neurology 1998;50(6 Suppl 6):S23-6
  • Group PS. Pramipexole vs levodopa as initial treatment for Parkinson's disease: a randomized controlled trial. JAMA 2000;284:1931-198
  • Hubble JP. Long-term studies of dopamine agonists. Neurology 2002;58(4 Suppl 1):S42-50
  • Pearce RK, Banerji T, Jenner P, Marsden CD. De novo administration of ropinirole and bromocriptine induces less dyskinesia than L-dopa in the MPTP-treated marmoset. Mov Disord 1998;13(2):234-41
  • Bedard PJ, Di Paolo T, Falardeau P, Boucher R. Chronic treatment with L-DOPA, but not bromocriptine induces dyskinesia in MPTP-parkinsonian monkeys. Correlation with [3H]spiperone binding. Brain Res 1986;379(2):294-9
  • Bonuccelli U, Pavese N. Dopamine agonists in the treatment of Parkinson's disease. Expert Rev Neurother 2006;6(1):81-9
  • Radad K, Gille G, Rausch WD. Short review on dopamine agonists: insight into clinical and research studies relevant to Parkinson's disease. Pharmacol Rep 2005;57(6):701-12
  • Bonuccelli U, Pavese N. Role of dopamine agonists in Parkinson's disease: an update. Expert Rev Neurother 2007;7(10):1391-9
  • Voon V, Mehta AR, Hallett M. Impulse control disorders in Parkinson's disease: recent advances. Curr Opin Neurol 2011;24(4):324-30
  • Weintraub D, Koester J, Potenza MN, et al. Impulse control disorders in Parkinson disease: a cross-sectional study of 3090 patients. Arch Neurol 2010;67(5):589-95
  • Weintraub D, Potenza MN. Impulse control disorders in Parkinson's disease. Curr Neurol Neurosci Rep 2006;6(4):302-6
  • Weintraub D, Nirenberg MJ. Impulse control and related disorders in Parkinson's disease. Neurodegener Dis 2013;11(2):63-71
  • Lee JY, Kim JM, Kim JW, et al. Association between the dose of dopaminergic medication and the behavioral disturbances in Parkinson disease. Parkinsonism Relat Disord 2010;16(3):202-7
  • Poletti M, Logi C, Lucetti C, et al. A single-center, cross-sectional prevalence study of impulse control disorders in Parkinson disease: association with dopaminergic drugs. J Clin Psychopharmacol 2013;33(5):691-4
  • Cilia R, van Eimeren T. Impulse control disorders in Parkinson's disease: seeking a roadmap toward a better understanding. Brain Struct Funct 2011;216(4):289-99
  • Katzenschlager R, Head J, Schrag A, et al. Fourteen-year final report of the randomized PDRG-UK trial comparing three initial treatments in PD. Neurology 2008;71(7):474-80
  • Talati R, Baker WL, Patel AA, et al. Adding a dopamine agonist to preexisting levodopa therapy vs. levodopa therapy alone in advanced Parkinson's disease: a meta analysis. Int J Clin Pract 2009;63(4):613-23
  • Calne DB, Teychenne PF, Claveria LE, et al. Bromocriptine in Parkinsonism. Br Med J 1974;4(5942):442-4
  • Antonini A, Poewe W. Fibrotic heart-valve reactions to dopamine-agonist treatment in Parkinson's disease. Lancet Neurol 2007;6(9):826-9
  • Dhawan V, Medcalf P, Stegie F, et al. Retrospective evaluation of cardio-pulmonary fibrotic side effects in symptomatic patients from a group of 234 Parkinson's disease patients treated with cabergoline. J Neural Transm 2005;112(5):661-8
  • Danoff SK, Grasso ME, Terry PB, Flynn JA. Pleuropulmonary disease due to pergolide use for restless legs syndrome. Chest 2001;120(1):313-16
  • Oechsner M, Groenke L, Mueller D. Pleural fibrosis associated with dihydroergocryptine treatment. Acta Neurol Scand 2000;101(4):283-5
  • Grunblatt E, Mandel S, Gassen M, Youdim MB. Potent neuroprotective and antioxidant activity of apomorphine in MPTP and 6-hydroxydopamine induced neurotoxicity. J Neural Transm Suppl 1999;55:57-70
  • Gassen M, Glinka Y, Pinchasi B, Youdim MB. Apomorphine is a highly potent free radical scavenger in rat brain mitochondrial fraction. Eur J Pharmacol 1996;308(2):219-25
  • Guo H, Tang Z, Yu Y, et al. Apomorphine induces trophic factors that support fetal rat mesencephalic dopaminergic neurons in cultures. Eur J Neurosci 2002;16(10):1861-70
  • Yuan H, Sarre S, Ebinger G, Michotte Y. Neuroprotective and neurotrophic effect of apomorphine in the striatal 6-OHDA-lesion rat model of Parkinson's disease. Brain Res 2004;1026(1):95-107
  • Battaglia G, Busceti CL, Cuomo L, et al. Continuous subcutaneous infusion of apomorphine rescues nigro-striatal dopaminergic terminals following MPTP injection in mice. Neuropharmacology 2002;42(3):367-73
  • Schwab RS, Amador LV, Lettvin JY. Apomorphine in Parkinson's disease. Trans Am Neurol Assoc 1951;56:251-3
  • Cotzias GC, Papavasiliou PS, Fehling C, et al. Similarities between neurologic effects of L-dipa and of apomorphine. N Engl J Med 1970;282(1):31-3
  • Corsini GU, Del Zompo M, Gessa GL, Mangoni A. Therapeutic efficacy of apomorphine combined with an extracerebral inhibitor of dopamine receptors in Parkinson's disease. Lancet 1979;1(8123):954-6
  • Ostergaard L, Werdelin L, Odin P, et al. Pen injected apomorphine against off phenomena in late Parkinson's disease: a double blind, placebo controlled study. J Neurol Neurosurg Psychiatry 1995;58(6):681-7
  • Dewey RB Jr, Hutton JT, LeWitt PA, Factor SA. A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch Neurol 2001;58(9):1385-92
  • Pfeiffer RF, Gutmann L, Hull KL Jr, et al. Continued efficacy and safety of subcutaneous apomorphine in patients with advanced Parkinson's disease. Parkinsonism Relat Disord 2007;13(2):93-100
  • Pietz K, Hagell P, Odin P. Subcutaneous apomorphine in late stage Parkinson's disease: a long term follow up. J Neurol Neurosurg Psychiatry 1998;65(5):709-16
  • Hughes AJ, Bishop S, Kleedorfer B, et al. Subcutaneous apomorphine in Parkinson's disease: response to chronic administration for up to five years. Mov Disord 1993;8(2):165-70
  • Frankel JP, Lees AJ, Kempster PA, Stern GM. Subcutaneous apomorphine in the treatment of Parkinson's disease. J Neurol Neurosurg Psychiatry 1990;53(2):96-101
  • 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
  • Antonini A, Isaias IU, Rodolfi G, et al. A 5-year prospective assessment of advanced Parkinson disease patients treated with subcutaneous apomorphine infusion or deep brain stimulation. J Neurol 2011;258(4):579-85
  • Dewey RB Jr, Maraganore DM, Ahlskog JE, Matsumoto JY. A double-blind, placebo-controlled study of intranasal apomorphine spray as a rescue agent for off-states in Parkinson's disease. Mov Disord 1998;13(5):782-7
  • Grosset KA, Malek N, Morgan F, Grosset DG. Inhaled dry powder apomorphine (VR040) for ‘off’ periods in Parkinson's disease: an in-clinic double-blind dose ranging study. Acta Neurol Scand 2013;128(3):166-71
  • Grosset KA, Malek N, Morgan F, Grosset DG. Phase IIa randomized double-blind, placebo-controlled study of inhaled apomorphine as acute challenge for rescuing ‘off' periods in patients with established Parkinson's disease. Eur J Neurol 2013;20(11):1445-50
  • Grosset KA, Malek N, Morgan F, Grosset DG. Inhaled apomorphine in patients with ‘on-off' fluctuations: a randomized, double-blind, placebo-controlled, clinic and home based, parallel-group study. J Parkinsons Dis 2013;3(1):31-7
  • Ondo W, Hunter C, Almaguer M, Jankovic J. A novel sublingual apomorphine treatment for patients with fluctuating Parkinson's disease. Mov Disord 1999;14(4):664-8
  • Ondo W, Hunter C, Almaguer M, et al. Efficacy and tolerability of a novel sublingual apomorphine preparation in patients with fluctuating Parkinson's disease. Clin Neuropharmacol 1999;22(1):1-4
  • van Laar T, Neef C, Danhof M, et al. A new sublingual formulation of apomorphine in the treatment of patients with Parkinson's disease. Mov Disord 1996;11(6):633-8
  • van Laar T, Jansen EN, Neef C, et al. Pharmacokinetics and clinical efficacy of rectal apomorphine in patients with Parkinson's disease: a study of five different suppositories. Mov Disord 1995;10(4):433-9
  • Edwards LL, Quigley EM, Harned RK, et al. Defecatory function in Parkinson's disease: response to apomorphine. Ann Neurol 1993;33(5):490-3
  • Dellapina E, Gerdelat-Mas A, Ory-Magne F, et al. Apomorphine effect on pain threshold in Parkinson's disease: a clinical and positron emission tomography study. Mov Disord 2011;26(1):153-7
  • Turle-Lorenzo N, Maurin B, Puma C, et al. The dopamine agonist piribedil with L-DOPA improves attentional dysfunction: relevance for Parkinson's disease. J Pharmacol Exp Ther 2006;319(2):914-23
  • Lane EL, Dunnett SB. Pre-treatment with dopamine agonists influence L-dopa mediated rotations without affecting abnormal involuntary movements in the 6-OHDA lesioned rat. Behav Brain Res 2010;213(1):66-72
  • Gerlach M, Halley P, Riederer P, van den Buuse M. The effect of piribedil on L-DOPA-induced dyskinesias in a rat model of Parkinson's disease: differential role of alpha(2) adrenergic mechanisms. J Neural Transm 2013;120(1):31-6
  • Smith LA, Tel BC, Jackson MJ, et al. Repeated administration of piribedil induces less dyskinesia than L-dopa in MPTP-treated common marmosets: a behavioural and biochemical investigation. Mov Disord 2002;17(5):887-901
  • Smith L, De Salvia M, Jenner P, Marsden CD. An appraisal of the antiparkinsonian activity of piribedil in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated common marmosets. Mov Disord 1996;11(2):125-35
  • Smith LA, Jackson MG, Bonhomme C, et al. Transdermal administration of piribedil reverses MPTP-induced motor deficits in the common marmoset. Clin Neuropharmacol 2000;23(3):133-42
  • Brocco M, Dekeyne A, Papp M, Millan MJ. Antidepressant-like properties of the anti-Parkinson agent, piribedil, in rodents: mediation by dopamine D2 receptors. Behav Pharmacol 2006;17(7):559-72
  • Marighetto A, Valerio S, Philippin JN, et al. Comparative effects of the dopaminergic agonists piribedil and bromocriptine in three different memory paradigms in rodents. J Psychopharmacol 2008;22(5):511-21
  • Rascol O, Dubois B, Caldas AC, et al. Early piribedil monotherapy of Parkinson's disease: a planned seven-month report of the REGAIN study. Mov Disord 2006;21(12):2110-15
  • Ziegler M, Castro-Caldas A, Del Signore S, Rascol O. Efficacy of piribedil as early combination to levodopa in patients with stable Parkinson's disease: a 6-month, randomized, placebo-controlled study. Mov Disord 2003;18(4):418-25
  • Castro-Caldas A, Delwaide P, Jost W, et al. The Parkinson-Control study: a 1-year randomized, double-blind trial comparing piribedil (150 mg/day) with bromocriptine (25 mg/day) in early combination with levodopa in Parkinson's disease. Mov Disord 2006;21(4):500-9
  • Suwantamee J, Nidhinandana S, Srisuwananukorn S, et al. Efficacy and safety of piribedil in early combination with L-dopa in the treatment of Parkinson's disease: a 6-month open study. J Med Assoc Thai 2004;87(11):1293-300
  • Rascol O, Azulay JP, Blin O, et al. Orodispersible sublingual piribedil to abort OFF episodes: a single dose placebo-controlled, randomized, double-blind, cross-over study. Mov Disord 2010;25(3):368-76
  • Peretti G. Clinical evaluation of 30 mg cinnoxicam in patients with osteoarthrosis. Double blind controlled trial. Minerva Med 1996;87(4):155-60
  • Schuck S, Bentue-Ferrer D, Kleinermans D, et al. Psychomotor and cognitive effects of piribedil, a dopamine agonist, in young healthy volunteers. Fundam Clin Pharmacol 2002;16(1):57-65
  • Thobois S, Lhommee E, Klinger H, et al. Parkinsonian apathy responds to dopaminergic stimulation of D2/D3 receptors with piribedil. Brain 2013;136(Pt 5):1568-77
  • Montastruc JL, Ziegler M, Rascol O, Malbezin M. A randomized, double-blind study of a skin patch of a dopaminergic agonist, piribedil, in Parkinson's disease. Mov Disord 1999;14(2):336-41
  • Prikhojan A, Brannan T, Yahr MD. Comparative effects of repeated administration of dopamine agonists on circling behavior in rats. J Neural Transm 2000;107(10):1159-64
  • Larramendy C, Taravini IR, Saborido MD, et al. Cabergoline and pramipexole fail to modify already established dyskinesias in an animal model of parkinsonism. Behav Brain Res 2008;194(1):44-51
  • Papathanou M, Rose S, McCreary A, Jenner P. Induction and expression of abnormal involuntary movements is related to the duration of dopaminergic stimulation in 6-OHDA-lesioned rats. Eur J Neurosci 2011;33(12):2247-54
  • Domino EF, Ni L, Zhang H, et al. Effects of pramipexole on contraversive rotation and functional motor impairments in 1-methyl-4-phenyl1,2,3, 6-tetrahydropyridine-induced chronic hemiparkinsonian monkeys. J Pharmacol Exp Ther 1998;287(3):983-7
  • Mierau J, Schingnitz G. Biochemical and pharmacological studies on pramipexole, a potent and selective dopamine D2 receptor agonist. Eur J Pharmacol 1992;215(2-3):161-70
  • Tayarani-Binazir KA, Jackson MJ, Rose S, et al. Pramipexole combined with levodopa improves motor function but reduces dyskinesia in MPTP-treated common marmosets. Mov Disord 2010;25(3):377-84
  • Asanuma M, Miyazaki I, Diaz-Corrales FJ, et al. Pramipexole has ameliorating effects on levodopa-induced abnormal dopamine turnover in parkinsonian striatum and quenching effects on dopamine-semiquinone generated in vitro. Neurol Res 2005;27(5):533-9
  • Ferger B, Buck K, Shimasaki M, et al. Continuous dopaminergic stimulation by pramipexole is effective to treat early morning akinesia in animal models of Parkinson's disease: a pharmacokinetic-pharmacodynamic study using in vivo microdialysis in rats. Synapse 2010;64(7):533-41
  • Tokunaga N, Choudhury ME, Nishikawa N, et al. Pramipexole upregulates dopamine receptor D(2) and D(3) expression in rat striatum. J Pharmacol Sci 2012;120(2):133-7
  • Riddle JL, Rokosik SL, Napier TC. Pramipexole- and methamphetamine-induced reward-mediated behavior in a rodent model of Parkinson's disease and controls. Behav Brain Res 2012;233(1):15-23
  • Rokosik SL, Napier TC. Pramipexole-induced increased probabilistic discounting: comparison between a rodent model of Parkinson's disease and controls. Neuropsychopharmacology 2012;37(6):1397-408
  • Madden GJ, Johnson PS, Brewer AT, et al. Effects of pramipexole on impulsive choice in male wistar rats. Exp Clin Psychopharmacol 2010;18(3):267-76
  • Abramova NA, Cassarino DS, Khan SM, et al. Inhibition by R(+) or S(-) pramipexole of caspase activation and cell death induced by methylpyridinium ion or beta amyloid peptide in SH-SY5Y neuroblastoma. J Neurosci Res 2002;67(4):494-500
  • Kakimura J, Kitamura Y, Takata K, et al. Release and aggregation of cytochrome c and alpha-synuclein are inhibited by the antiparkinsonian drugs, talipexole and pramipexole. Eur J Pharmacol 2001;417(1-2):59-67
  • Chau KY, Cooper JM, Schapira AH. Pramipexole reduces phosphorylation of alpha-synuclein at serine-129. J Mol Neurosci 2013;51(2):573-80
  • Anderson DW, Neavin T, Smith JA, Schneider JS. Neuroprotective effects of pramipexole in young and aged MPTP-treated mice. Brain Res 2001;905(1-2):44-53
  • Iravani MM, Haddon CO, Cooper JM, et al. Pramipexole protects against MPTP toxicity in non-human primates. J Neurochem 2006;96(5):1315-21
  • Shin JY, Park HJ, Ahn YH, Lee PH. Neuroprotective effect of L-dopa on dopaminergic neurons is comparable to pramipexol in MPTP-treated animal model of Parkinson's disease: a direct comparison study. J Neurochem 2009;111(4):1042-50
  • Inden M, Kitamura Y, Tamaki A, et al. Neuroprotective effect of the antiparkinsonian drug pramipexole against nigrostriatal dopaminergic degeneration in rotenone-treated mice. Neurochem Int 2009;55(8):760-7
  • Li C, Guo Y, Xie W, et al. Neuroprotection of pramipexole in UPS impairment induced animal model of Parkinson's disease. Neurochem Res 2010;35(10):1546-56
  • Iravani MM, Sadeghian M, Leung CC, et al. Continuous subcutaneous infusion of pramipexole protects against lipopolysaccharide-induced dopaminergic cell death without affecting the inflammatory response. Exp Neurol 2008;212(2):522-31
  • Hubble JP, Koller WC, Cutler NR, et al. Pramipexole in patients with early Parkinson's disease. Clin Neuropharmacol 1995;18(4):338-47
  • Shannon KM, Bennett JP Jr, Friedman JH. Efficacy of pramipexole, a novel dopamine agonist, as monotherapy in mild to moderate Parkinson's disease. The Pramipexole Study Group. Neurology 1997;49(3):724-8
  • Kieburtz K. Twice-daily, low-dose pramipexole in early Parkinson's disease: a randomized, placebo-controlled trial. Mov Disord 2011;26(1):37-44
  • Pogarell O, Gasser T, van Hilten JJ, et al. Pramipexole in patients with Parkinson's disease and marked drug resistant tremor: a randomised, double blind, placebo controlled multicentre study. J Neurol Neurosurg Psychiatry 2002;72(6):713-20
  • Mizuno Y, Yanagisawa N, Kuno S, et al. Randomized, double-blind study of pramipexole with placebo and bromocriptine in advanced Parkinson's disease. Mov Disord 2003;18(10):1149-56
  • Wong KS, Lu CS, Shan DE, et al. Efficacy, safety, and tolerability of pramipexole in untreated and levodopa-treated patients with Parkinson's disease. J Neurol Sci 2003;216(1):81-7
  • Moller JC, Oertel WH, Koster J, et al. Long-term efficacy and safety of pramipexole in advanced Parkinson's disease: results from a European multicenter trial. Mov Disord 2005;20(5):602-10
  • 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
  • Group PS. Long-term effect of initiating pramipexole vs levodopa in early Parkinson disease. Arch Neurol 2009;66(5):563-70
  • Group PS. Dopamine transporter brain imaging to assess effect of pramipexole vs levodopa on Parkinson's disease progression. JAMA 2002;287:1653-61
  • Utsumi H, Okuma Y, Kano O, et al. Evaluation of the efficacy of pramipexole for treating levodopa-induced dyskinesia in patients with Parkinson's disease. Intern Med 2013;52(3):325-32
  • Rektorova I, Rektor I, Bares M, et al. Pramipexole and pergolide in the treatment of depression in Parkinson's disease: a national multicentre prospective randomized study. Eur J Neurol 2003;10(4):399-406
  • Harada T, Ishizaki F, Horie N, et al. New dopamine agonist pramipexole improves parkinsonism and depression in Parkinson's disease. Hiroshima J Med Sci 2011;60(4):79-82
  • Morita A, Okuma Y, Kamei S, et al. Pramipexole reduces the prevalence of fatigue in patients with Parkinson's disease. Intern Med 2011;50(19):2163-8
  • Depboylu C, Maurer L, Matusch A, et al. Effect of long-term treatment with pramipexole or levodopa on presynaptic markers assessed by longitudinal [(123)I]FP-CIT SPECT and histochemistry. Neuroimage 2013;79:191-200
  • Schapira AH, McDermott MP, Barone P, et al. Pramipexole in patients with early Parkinson's disease (PROUD): a randomised delayed-start trial. Lancet Neurol 2013;12(8):747-55
  • Schapira AH, Barone P, Hauser RA, et al. Patient-reported convenience of once-daily versus three-times-daily dosing during long-term studies of pramipexole in early and advanced Parkinson's disease. Eur J Neurol 2013;20(1):50-6
  • Hametner EM, Seppi K, Poewe W. Pramipexole extended release in Parkinson's disease. Expert Rev Neurother 2011;11(9):1229-34
  • Poewe W, Rascol O, Barone P, et al. Extended-release pramipexole in early Parkinson disease: a 33-week randomized controlled trial. Neurology 2011;77(8):759-66
  • Schapira AH, Barone P, Hauser RA, et al. Extended-release pramipexole in advanced Parkinson disease: a randomized controlled trial. Neurology 2011;77(8):767-74
  • Mizuno Y, Yamamoto M, Kuno S, et al. Efficacy and safety of extended- versus immediate-release pramipexole in Japanese patients with advanced and L-dopa-undertreated Parkinson disease: a double-blind, randomized trial. Clin Neuropharmacol 2012;35(4):174-81
  • Fukuzaki K, Kamenosono T, Kitazumi K, Nagata R. Effects of ropinirole on motor behavior in MPTP-treated common marmosets. Pharmacol Biochem Behav 2000;67(1):121-9
  • Stephenson DT, Meglasson MD, Connell MA, et al. The effects of a selective dopamine D2 receptor agonist on behavioral and pathological outcome in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated squirrel monkeys. J Pharmacol Exp Ther 2005;314(3):1257-66
  • Maratos EC, Jackson MJ, Pearce RK, Jenner P. Antiparkinsonian activity and dyskinesia risk of ropinirole and L-DOPA combination therapy in drug naive MPTP-lesioned common marmosets (Callithrix jacchus). Mov Disord 2001;16(4):631-41
  • Carta AR, Frau L, Pinna A, et al. Behavioral and biochemical correlates of the dyskinetic potential of dopaminergic agonists in the 6-OHDA lesioned rat. Synapse 2008;62(7):524-33
  • Stockwell KA, Virley DJ, Perren M, et al. Continuous delivery of ropinirole reverses motor deficits without dyskinesia induction in MPTP-treated common marmosets. Exp Neurol 2008;211(1):172-9
  • Rogers DC, Costall B, Domeney AM, et al. Anxiolytic profile of ropinirole in the rat, mouse and common marmoset. Psychopharmacology (Berl) 2000;151(1):91-7
  • Michinaga S, Hisatsune A, Isohama Y, Katsuki H. An anti-Parkinson drug ropinirole depletes orexin from rat hypothalamic slice culture. Neurosci Res 2010;68(4):315-21
  • Nair VD, Olanow CW. Differential modulation of Akt/glycogen synthase kinase-3beta pathway regulates apoptotic and cytoprotective signaling responses. J Biol Chem 2008;283(22):15469-78
  • Du F, Li R, Huang Y, et al. Dopamine D3 receptor-preferring agonists induce neurotrophic effects on mesencephalic dopamine neurons. Eur J Neurosci 2005;22(10):2422-30
  • Presgraves SP, Borwege S, Millan MJ, Joyce JN. Involvement of dopamine D(2)/D(3) receptors and BDNF in the neuroprotective effects of S32504 and pramipexole against 1-methyl-4-phenylpyridinium in terminally differentiated SH-SY5Y cells. Exp Neurol 2004;190(1):157-70
  • Chen S, Zhang X, Yang D, et al. D2/D3 receptor agonist ropinirole protects dopaminergic cell line against rotenone-induced apoptosis through inhibition of caspase- and JNK-dependent pathways. FEBS Lett 2008;582(5):603-10
  • Iida M, Miyazaki I, Tanaka K, et al. Dopamine D2 receptor-mediated antioxidant and neuroprotective effects of ropinirole, a dopamine agonist. Brain Res 1999;838(1-2):51-9
  • Park G, Park YJ, Yang HO, Oh MS. Ropinirole protects against 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced neurotoxicity in mice via anti-apoptotic mechanism. Pharmacol Biochem Behav 2013;104:163-8
  • 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. 056 Study Group. N Engl J Med 2000;342(20):1484-91
  • Adler CH, Sethi KD, Hauser RA, et al. Ropinirole for the treatment of early Parkinson's disease. The Ropinirole Study Group. Neurology 1997;49(2):393-9
  • Brooks DJ, Abbott RJ, Lees AJ, et al. A placebo-controlled evaluation of ropinirole, a novel D2 agonist, as sole dopaminergic therapy in Parkinson's disease. Clin Neuropharmacol 1998;21(2):101-7
  • Rascol O, Brooks DJ, Brunt ER, et al. Ropinirole in the treatment of early Parkinson's disease: a 6-month interim report of a 5-year levodopa-controlled study. 056 Study Group. Mov Disord 1998;13(1):39-45
  • Hauser RA, Rascol O, Korczyn AD, et al. Ten-year follow-up of Parkinson's disease patients randomized to initial therapy with ropinirole or levodopa. Mov Disord 2007;22(16):2409-17
  • Singer C, Lamb J, Ellis A, Layton G. A comparison of sumanirole versus placebo or ropinirole for the treatment of patients with early Parkinson's disease. Mov Disord 2007;22(4):476-82
  • Giladi N, Boroojerdi B, Korczyn AD, et al. Rotigotine transdermal patch in early Parkinson's disease: a randomized, double-blind, controlled study versus placebo and ropinirole. Mov Disord 2007;22(16):2398-404
  • Tessa C, Diciotti S, Lucetti C, et al. fMRI changes in cortical activation during task performance with the unaffected hand partially reverse after ropinirole treatment in de novo Parkinson's disease. Parkinsonism Relat Disord 2013;19(2):265-8
  • Lieberman A, Olanow CW, Sethi K, et al. A multicenter trial of ropinirole as adjunct treatment for Parkinson's disease. Ropinirole Study Group. Neurology 1998;51(4):1057-62
  • Rascol O, Lees AJ, Senard JM, et al. Ropinirole in the treatment of levodopa-induced motor fluctuations in patients with Parkinson's disease. Clin Neuropharmacol 1996;19(3):234-45
  • Barone P, Lamb J, Ellis A, Clarke Z. Sumanirole versus placebo or ropinirole for the adjunctive treatment of patients with advanced Parkinson's disease. Mov Disord 2007;22(4):483-9
  • Mizuno Y, Abe T, Hasegawa K, et al. Ropinirole is effective on motor function when used as an adjunct to levodopa in Parkinson's disease: STRONG study. Mov Disord 2007;22(13):1860-5
  • Onofrj M, Bonanni L, Thomas A. An expert opinion on safinamide in Parkinson's disease. Expert Opin Investig Drugs 2008;17(7):1115-25
  • Ray Chaudhuri K, Martinez-Martin P, Rolfe KA, et al. Improvements in nocturnal symptoms with ropinirole prolonged release in patients with advanced Parkinson's disease. Eur J Neurol 2012;19(1):105-13
  • 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(7):858-66
  • Hauser RA, Reichmann H, Lew M, et al. Long-term, open-label study of once-daily ropinirole prolonged release in early Parkinson's disease. Int J Neurosci 2011;121(5):246-53
  • Stocchi F, Giorgi L, Hunter B, Schapira AH. Prepared: comparison of prolonged and immediate release ropinirole in advanced Parkinson's disease. Mov Disord 2011;26(7):1259-65
  • Pardeshi CV, Belgamwar VS, Tekade AR, Surana SJ. Novel surface modified polymer-lipid hybrid nanoparticles as intranasal carriers for ropinirole hydrochloride: in vitro, ex vivo and in vivo pharmacodynamic evaluation. J Mater Sci Mater Med 2013;24(9):2101-15
  • De Caro V, Giandalia G, Siragusa MG, et al. New prospective in treatment of Parkinson's disease: studies on permeation of ropinirole through buccal mucosa. Int J Pharm 2012;429(1-2):78-83
  • Rektorova I, Balaz M, Svatova J, et al. Effects of ropinirole on nonmotor symptoms of Parkinson disease: a prospective multicenter study. Clin Neuropharmacol 2008;31(5):261-6
  • Rakshi JS, Pavese N, Uema T, et al. A comparison of the progression of early Parkinson's disease in patients started on ropinirole or L-dopa: an 18F-dopa PET study. J Neural Transm 2002;109(12):1433-43
  • Whone AL, Watts RL, Stoessl AJ, et al. Slower progression of Parkinson's disease with ropinirole versus levodopa: the REAL-PET study. Ann Neurol 2003;54(1):93-101
  • Loschmann PA, Chong PN, Nomoto M, et al. Stereoselective reversal of MPTP-induced parkinsonism in the marmoset after dermal application of N-0437. Eur J Pharmacol 1989;166(3):373-80
  • Stockwell KA, Scheller D, Rose S, et al. Continuous administration of rotigotine to MPTP-treated common marmosets enhances anti-parkinsonian activity and reduces dyskinesia induction. Exp Neurol 2009;219(2):533-42
  • Stockwell KA, Scheller DK, Smith LA, et al. Continuous rotigotine administration reduces dyskinesia resulting from pulsatile treatment with rotigotine or L-DOPA in MPTP-treated common marmosets. Exp Neurol 2010;221(1):79-85
  • Schmidt WJ, Lebsanft H, Heindl M, et al. Continuous versus pulsatile administration of rotigotine in 6-OHDA-lesioned rats: contralateral rotations and abnormal involuntary movements. J Neural Transm 2008;115(10):1385-92
  • Wang A, Wang L, Sun K, et al. Preparation of rotigotine-loaded microspheres and their combination use with L-DOPA to modify dyskinesias in 6-OHDA-lesioned rats. Pharm Res 2012;29(9):2367-76
  • Scheller D, Stichel-Gunkel C, Lubbert H, et al. Neuroprotective effects of rotigotine in the acute MPTP-lesioned mouse model of Parkinson's disease. Neurosci Lett 2008;432(1):30-4
  • Scheller D, Chan P, Li Q, et al. Rotigotine treatment partially protects from MPTP toxicity in a progressive macaque model of Parkinson's disease. Exp Neurol 2007;203(2):415-22
  • Perez-Lloret S, Rey MV, Ratti PL, Rascol O. Rotigotine transdermal patch for the treatment of Parkinson's Disease. Fundam Clin Pharmacol 2013;27(1):81-95
  • LeWitt PA, Lyons KE, Pahwa R. Advanced Parkinson disease treated with rotigotine transdermal system: PREFER Study. Neurology 2007;68(16):1262-7
  • LeWitt PA, Boroojerdi B, Surmann E, Poewe W. Rotigotine transdermal system for long-term treatment of patients with advanced Parkinson's disease: results of two open-label extension studies, CLEOPATRA-PD and PREFER. J Neural Transm 2013;120(7):1069-81
  • Trenkwalder C, Kies B, Rudzinska M, et al. Rotigotine effects on early morning motor function and sleep in Parkinson's disease: a double-blind, randomized, placebo-controlled study (RECOVER). Mov Disord 2011;26(1):90-9
  • Ray Chaudhuri K, Martinez-Martin P, Antonini A, et al. Rotigotine and specific non-motor symptoms of Parkinson's disease: post hoc analysis of RECOVER. Parkinsonism Relat Disord 2013;19(7):660-5
  • Boroojerdi B, Watts RL, Jankovic J, et al. The long-term impact of early vs delayed treatment with rotigotine transdermal system in patinets with early parkinson's disease. Mov Disord 2011;26(Suppl):S121
  • Jankovic J, Watts RL, Martin W, Boroojerdi B. Transdermal rotigotine: double-blind, placebo-controlled trial in Parkinson disease. Arch Neurol 2007;64(5):676-82
  • Glennon JC, Van Scharrenburg G, Ronken E, et al. In vitro characterization of SLV308 (7-[4-methyl-1-piperazinyl]-2(3H)-benzoxazolone, monohydrochloride): a novel partial dopamine D2 and D3 receptor agonist and serotonin 5-HT1A receptor agonist. Synapse 2006;60(8):599-608
  • Wolf WA. SLV-308. Solvay. Curr Opin Investig Drugs 2003;4(7):878-82
  • Betry C, Etievant A, Lambas-Senas L, et al. In vivo effects of pardoprunox (SLV308), a partial D(2)/D(3) receptor and 5-HT1A receptor agonist, on rat dopamine and serotonin neuronal activity. Synapse 2011;65(10):1042-51
  • Jones CA, Johnston LC, Jackson MJ, et al. An in vivo pharmacological evaluation of pardoprunox (SLV308)–a novel combined dopamine D(2)/D(3) receptor partial agonist and 5-HT(1A) receptor agonist with efficacy in experimental models of Parkinson's disease. Eur Neuropsychopharmacol 2010;20(8):582-93
  • 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
  • Tayarani-Binazir K, Jackson MJ, Rose S, et al. The partial dopamine agonist pardoprunox (SLV308) administered in combination with l-dopa improves efficacy and decreases dyskinesia in MPTP treated common marmosets. Exp Neurol 2010;226(2):320-7
  • Bronzova J, Sampaio C, Hauser RA, et al. Double-blind study of pardoprunox, a new partial dopamine agonist, in early Parkinson's disease. Mov Disord 2010;25(6):738-46
  • Sampaio C, Bronzova J, Hauser RA, et al. Pardoprunox in early Parkinson's disease: results from 2 large, randomized double-blind trials. Mov Disord 2011;26(8):1464-76
  • Hauser RA, Bronzova J, Sampaio C, et al. Safety and tolerability of pardoprunox, a new partial dopamine agonist, in a randomized, controlled study of patients with advanced Parkinson's disease. Eur Neurol 2009;62(1):40-8
  • 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
  • Heinrich JN, Brennan J, Lai MH, et al. Aplindore (DAB-452), a high affinity selective dopamine D2 receptor partial agonist. Eur J Pharmacol 2006;552(1-3):36-45
  • Jackson MJ, Andree TH, Hansard M, et al. The dopamine D(2) receptor partial agonist aplindore improves motor deficits in MPTP-treated common marmosets alone and combined with L-dopa. J Neural Transm 2010;117(1):55-67
  • McCall RB, Lookingland KJ, Bedard PJ, Huff RM. Sumanirole, a highly dopamine D2-selective receptor agonist: in vitro and in vivo pharmacological characterization and efficacy in animal models of Parkinson's disease. J Pharmacol Exp Ther 2005;314(3):1248-56
  • Duchane J, Jenkinson C. Changes in the quality of life resulting from treatment of persons with advanced Parkinson's disease: suminarole vs placebo2):P499
  • Duchane J, Jenkinson C. Changes in the quality of life resulting from treatment of persons with early Parkinson's disease: suminarole vs placebo. Mov Disord 2002;17(Suppl 2):P309
  • Kang JH, Irwin DJ, Chen-Plotkin AS, et al. Association of cerebrospinal fluid beta-amyloid 1-42, T-tau, P-tau181, and alpha-synuclein levels with clinical features of drug-naive patients with early parkinson disease. JAMA Neurol 2013; doi:10.1001/jamaneurol.2013.3861
  • Goetz CG, Tilley BC, Shaftman SR, et al. Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord 2008;23(15):2129-70
  • 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(2):791-804
  • Gerlach M, Double K, Arzberger T, et al. Dopamine receptor agonists in current clinical use: comparative dopamine receptor binding profiles defined in the human striatum. J Neural Transm 2003;110(10):1119-27
  • Scheller D, Ullmer C, Berkels R, et al. The in vitro receptor profile of rotigotine: a new agent for the treatment of Parkinson's disease. Naunyn Schmiedebergs Arch Pharmacol 2009;379(1):73-86
  • Newman-Tancredi A, Cussac D, Quentric Y, et al. Differential actions of antiparkinson agents at multiple classes of monoaminergic receptor. III. Agonist and antagonist properties at serotonin, 5-HT(1) and 5-HT(2), receptor subtypes. J Pharmacol Exp Ther 2002;303(2):815-22

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.