Skip to main content
Top
Published in: CNS Drugs 12/2014

01-12-2014 | Review Article

Pharmacological Strategies for the Management of Levodopa-Induced Dyskinesia in Patients with Parkinson’s Disease

Authors: Eva Schaeffer, Andrea Pilotto, Daniela Berg

Published in: CNS Drugs | Issue 12/2014

Login to get access

Abstract

l-Dopa-induced dyskinesias (LID) are the most common adverse effects of long-term dopaminergic therapy in Parkinson’s disease (PD). However, the exact mechanisms underlying dyskinesia are still unclear. For a long time, nigrostriatal degeneration and pulsatile stimulation of striatal postsynaptic receptors have been highlighted as the key factors for the development of LID. In recent years, PD models have revealed a wide range of non-dopaminergic neurotransmitter systems involved in pre- and postsynaptic changes and thereby contributing to the pathophysiology of LID. In the current review, we focus on therapeutic LID targets, mainly based on agents acting on dopaminergic, glutamatergic, serotoninergic, adrenergic, and cholinergic systems. Despite a large number of clinical trials, currently only amantadine and, to a lesser extent, clozapine are being used as effective strategies in the treatment of LID in clinical settings. Thus, in the second part of the article, we review the placebo-controlled trials on LID treatment in order to disentangle the changing scenario of drug development. Promising results include the extension of l-dopa action without inducing LID of the novel monoamine oxidase B- and glutamate-release inhibitor safinamide; however, this had no obvious effect on existing LID. Others, like the metabotropic glutamate-receptor antagonist AFQ056, showed promising results in some of the studies; however, confirmation is still lacking. Thus, to date, strategies of continuous dopaminergic stimulation seem the most promising to prevent or ameliorate LID. The success of future therapeutic strategies once moderate to severe LID occur will depend on the translation from preclinical experimental models into clinical practice in a bidirectional process.
Literature
1.
go back to reference Warren Olanow C, Kieburtz K, Rascol O, Poewe W, Schapira AH, Emre M, et al. Factors predictive of the development of levodopa-induced dyskinesia and wearing-off in Parkinson’s disease. Mov Disord. 2013;28(8):1064–71. doi:10.1002/mds.25364. Warren Olanow C, Kieburtz K, Rascol O, Poewe W, Schapira AH, Emre M, et al. Factors predictive of the development of levodopa-induced dyskinesia and wearing-off in Parkinson’s disease. Mov Disord. 2013;28(8):1064–71. doi:10.​1002/​mds.​25364.
2.
go back to reference 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.PubMed 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.PubMed
3.
go back to reference Lundblad M, Andersson M, Winkler C, Kirik D, Wierup N, Cenci MA. Pharmacological validation of behavioural measures of akinesia and dyskinesia in a rat model of Parkinson’s disease. Eur J Neurosci. 2002;15(1):120–32.PubMed Lundblad M, Andersson M, Winkler C, Kirik D, Wierup N, Cenci MA. Pharmacological validation of behavioural measures of akinesia and dyskinesia in a rat model of Parkinson’s disease. Eur J Neurosci. 2002;15(1):120–32.PubMed
4.
go back to reference Lundblad M, Picconi B, Lindgren H, Cenci MA. A model of l-dopa-induced dyskinesia in 6-hydroxydopamine lesioned mice: relation to motor and cellular parameters of nigrostriatal function. Neurobiol Dis. 2004;16(1):110–23. doi:10.1016/j.nbd.2004.01.007.PubMed Lundblad M, Picconi B, Lindgren H, Cenci MA. A model of l-dopa-induced dyskinesia in 6-hydroxydopamine lesioned mice: relation to motor and cellular parameters of nigrostriatal function. Neurobiol Dis. 2004;16(1):110–23. doi:10.​1016/​j.​nbd.​2004.​01.​007.PubMed
7.
go back to reference Boyce S, Rupniak NM, Steventon MJ, Iversen SD. Characterisation of dyskinesias induced by l-dopa in MPTP-treated squirrel monkeys. Psychopharmacology. 1990;102(1):21–7.PubMed Boyce S, Rupniak NM, Steventon MJ, Iversen SD. Characterisation of dyskinesias induced by l-dopa in MPTP-treated squirrel monkeys. Psychopharmacology. 1990;102(1):21–7.PubMed
10.
go back to reference Boyce S, Rupniak NM, Steventon MJ, Iversen SD. Nigrostriatal damage is required for induction of dyskinesias by l-dopa in squirrel monkeys. Clin Neuropharmacol. 1990;13(5):448–58.PubMed Boyce S, Rupniak NM, Steventon MJ, Iversen SD. Nigrostriatal damage is required for induction of dyskinesias by l-dopa in squirrel monkeys. Clin Neuropharmacol. 1990;13(5):448–58.PubMed
11.
go back to reference Horstink MW, Zijlmans JC, Pasman JW, Berger HJ, van’t Hof MA. Severity of Parkinson’s disease is a risk factor for peak-dose dyskinesia. J Neurol Neurosurg Psychiatry. 1990;53(3):224–6. Horstink MW, Zijlmans JC, Pasman JW, Berger HJ, van’t Hof MA. Severity of Parkinson’s disease is a risk factor for peak-dose dyskinesia. J Neurol Neurosurg Psychiatry. 1990;53(3):224–6.
12.
go back to reference Jankovic J, Rajput AH, McDermott MP, Perl DP. The evolution of diagnosis in early Parkinson disease. Parkinson Study Group. Arch Neurol. 2000;57(3):369–72.PubMed Jankovic J, Rajput AH, McDermott MP, Perl DP. The evolution of diagnosis in early Parkinson disease. Parkinson Study Group. Arch Neurol. 2000;57(3):369–72.PubMed
13.
go back to reference Becker PM, Jamieson AO, Brown WD. Dopaminergic agents in restless legs syndrome and periodic limb movements of sleep: response and complications of extended treatment in 49 cases. Sleep. 1993;16(8):713–6.PubMed Becker PM, Jamieson AO, Brown WD. Dopaminergic agents in restless legs syndrome and periodic limb movements of sleep: response and complications of extended treatment in 49 cases. Sleep. 1993;16(8):713–6.PubMed
15.
go back to reference Grandas F, Galiano ML, Tabernero C. Risk factors for levodopa-induced dyskinesias in Parkinson’s disease. J Neurol. 1999;246(12):1127–33.PubMed Grandas F, Galiano ML, Tabernero C. Risk factors for levodopa-induced dyskinesias in Parkinson’s disease. J Neurol. 1999;246(12):1127–33.PubMed
16.
go back to reference Fabbrini G, Defazio G, Colosimo C, Suppa A, Bloise M, Berardelli A. Onset and spread of dyskinesias and motor symptoms in Parkinson’s disease. Mov Disord. 2009;24(14):2091–6. doi:10.1002/mds.22703.PubMed Fabbrini G, Defazio G, Colosimo C, Suppa A, Bloise M, Berardelli A. Onset and spread of dyskinesias and motor symptoms in Parkinson’s disease. Mov Disord. 2009;24(14):2091–6. doi:10.​1002/​mds.​22703.PubMed
17.
go back to reference Mones RJ, Elizan TS, Siegel GJ. Analysis of l-dopa induced dyskinesias in 51 patients with Parkinsonism. J Neurol Neurosurg Psychiatry. 1971;34(6):668–73.PubMedCentralPubMed Mones RJ, Elizan TS, Siegel GJ. Analysis of l-dopa induced dyskinesias in 51 patients with Parkinsonism. J Neurol Neurosurg Psychiatry. 1971;34(6):668–73.PubMedCentralPubMed
18.
19.
go back to reference Di Monte DA, McCormack A, Petzinger G, Janson AM, Quik M, Langston WJ. Relationship among nigrostriatal denervation, parkinsonism, and dyskinesias in the MPTP primate model. Mov Disord. 2000;15(3):459–66.PubMed Di Monte DA, McCormack A, Petzinger G, Janson AM, Quik M, Langston WJ. Relationship among nigrostriatal denervation, parkinsonism, and dyskinesias in the MPTP primate model. Mov Disord. 2000;15(3):459–66.PubMed
20.
go back to reference Schneider JS. Levodopa-induced dyskinesias in parkinsonian monkeys: relationship to extent of nigrostriatal damage. Pharmacol Biochem Behav. 1989;34(1):193–6.PubMed Schneider JS. Levodopa-induced dyskinesias in parkinsonian monkeys: relationship to extent of nigrostriatal damage. Pharmacol Biochem Behav. 1989;34(1):193–6.PubMed
21.
go back to reference Winkler C, Kirik D, Bjorklund A, Cenci MA. l-dopa-induced dyskinesia in the intrastriatal 6-hydroxydopamine model of Parkinson’s disease: relation to motor and cellular parameters of nigrostriatal function. Neurobiol Dis. 2002;10(2):165–86.PubMed Winkler C, Kirik D, Bjorklund A, Cenci MA. l-dopa-induced dyskinesia in the intrastriatal 6-hydroxydopamine model of Parkinson’s disease: relation to motor and cellular parameters of nigrostriatal function. Neurobiol Dis. 2002;10(2):165–86.PubMed
22.
go back to reference Carlsson T, Carta M, Munoz A, Mattsson B, Winkler C, Kirik D, et al. Impact of grafted serotonin and dopamine neurons on development of l-dopa-induced dyskinesias in parkinsonian rats is determined by the extent of dopamine neuron degeneration. Brain J Neurol. 2009;132(Pt 2):319–35. doi:10.1093/brain/awn305. Carlsson T, Carta M, Munoz A, Mattsson B, Winkler C, Kirik D, et al. Impact of grafted serotonin and dopamine neurons on development of l-dopa-induced dyskinesias in parkinsonian rats is determined by the extent of dopamine neuron degeneration. Brain J Neurol. 2009;132(Pt 2):319–35. doi:10.​1093/​brain/​awn305.
24.
go back to reference Miller DW, Abercrombie ED. Role of high-affinity dopamine uptake and impulse activity in the appearance of extracellular dopamine in striatum after administration of exogenous l-dopa: studies in intact and 6-hydroxydopamine-treated rats. J Neurochem. 1999;72(4):1516–22.PubMed Miller DW, Abercrombie ED. Role of high-affinity dopamine uptake and impulse activity in the appearance of extracellular dopamine in striatum after administration of exogenous l-dopa: studies in intact and 6-hydroxydopamine-treated rats. J Neurochem. 1999;72(4):1516–22.PubMed
26.
go back to reference Abercrombie ED, Bonatz AE, Zigmond MJ. Effects of l-dopa on extracellular dopamine in striatum of normal and 6-hydroxydopamine-treated rats. Brain Res. 1990;525(1):36–44.PubMed Abercrombie ED, Bonatz AE, Zigmond MJ. Effects of l-dopa on extracellular dopamine in striatum of normal and 6-hydroxydopamine-treated rats. Brain Res. 1990;525(1):36–44.PubMed
29.
go back to reference de la Fuente-Fernandez R, Sossi V, Huang Z, Furtado S, Lu JQ, Calne DB, et al. Levodopa-induced changes in synaptic dopamine levels increase with progression of Parkinson’s disease: implications for dyskinesias. Brain J Neurol. 2004;127(Pt 12):2747–54. doi:10.1093/brain/awh290. de la Fuente-Fernandez R, Sossi V, Huang Z, Furtado S, Lu JQ, Calne DB, et al. Levodopa-induced changes in synaptic dopamine levels increase with progression of Parkinson’s disease: implications for dyskinesias. Brain J Neurol. 2004;127(Pt 12):2747–54. doi:10.​1093/​brain/​awh290.
30.
31.
go back to reference Raevskii KS, Gainetdinov RR, Budygin EA, Mannisto P, Wightman M. Dopaminergic transmission in the rat striatum in vivo in conditions of pharmacological modulation. Neurosci Behav Physiol. 2002;32(2):183–8.PubMed Raevskii KS, Gainetdinov RR, Budygin EA, Mannisto P, Wightman M. Dopaminergic transmission in the rat striatum in vivo in conditions of pharmacological modulation. Neurosci Behav Physiol. 2002;32(2):183–8.PubMed
32.
go back to reference Sossi V, Dinelle K, Topping GJ, Holden JE, Doudet D, Schulzer M, et al. Dopamine transporter relation to levodopa-derived synaptic dopamine in a rat model of Parkinson’s: an in vivo imaging study. J Neurochem. 2009;109(1):85–92. doi:10.1111/j.1471-4159.2009.05904.x.PubMed Sossi V, Dinelle K, Topping GJ, Holden JE, Doudet D, Schulzer M, et al. Dopamine transporter relation to levodopa-derived synaptic dopamine in a rat model of Parkinson’s: an in vivo imaging study. J Neurochem. 2009;109(1):85–92. doi:10.​1111/​j.​1471-4159.​2009.​05904.​x.PubMed
34.
36.
37.
go back to reference Olanow CW, Obeso JA, Stocchi F. Drug insight: continuous dopaminergic stimulation in the treatment of Parkinson’s disease. Nat Clin Pract Neurol. 2006;2(7):382–92. doi:10.1038/ncpneuro0222.PubMed Olanow CW, Obeso JA, Stocchi F. Drug insight: continuous dopaminergic stimulation in the treatment of Parkinson’s disease. Nat Clin Pract Neurol. 2006;2(7):382–92. doi:10.​1038/​ncpneuro0222.PubMed
39.
go back to reference Schrag A, Quinn N. Dyskinesias and motor fluctuations in Parkinson’s disease. A community-based study. Brain J Neurol. 2000;123(Pt 11):2297–305. Schrag A, Quinn N. Dyskinesias and motor fluctuations in Parkinson’s disease. A community-based study. Brain J Neurol. 2000;123(Pt 11):2297–305.
40.
go back to reference Jenner P. Factors influencing the onset and persistence of dyskinesia in MPTP-treated primates. Ann Neurol. 2000;47(4 Suppl 1):S90–9. (Discussion S9–104). Jenner P. Factors influencing the onset and persistence of dyskinesia in MPTP-treated primates. Ann Neurol. 2000;47(4 Suppl 1):S90–9. (Discussion S9–104).
41.
go back to reference Smith LA, Tel BC, Jackson MJ, Hansard MJ, Braceras R, Bonhomme C, 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. doi:10.1002/mds.10200.PubMed Smith LA, Tel BC, Jackson MJ, Hansard MJ, Braceras R, Bonhomme C, 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. doi:10.​1002/​mds.​10200.PubMed
42.
go back to reference Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE, et al. Development of dyskinesias in a 5-year trial of ropinirole and l-dopa. Mov Disord. 2006;21(11):1844–50. doi:10.1002/mds.20988.PubMed Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE, et al. Development of dyskinesias in a 5-year trial of ropinirole and l-dopa. Mov Disord. 2006;21(11):1844–50. doi:10.​1002/​mds.​20988.PubMed
43.
go back to reference Holloway RG, Shoulson I, Fahn S, Kieburtz K, Lang A, Marek K, et al. Pramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. Arch Neurol. 2004;61(7):1044–53. doi:10.1001/archneur.61.7.1044.PubMed Holloway RG, Shoulson I, Fahn S, Kieburtz K, Lang A, Marek K, et al. Pramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. Arch Neurol. 2004;61(7):1044–53. doi:10.​1001/​archneur.​61.​7.​1044.PubMed
44.
go back to reference Smith LA, Jackson MJ, Johnston L, Kuoppamaki M, Rose S, Al-Barghouthy G, et al. Switching from levodopa to the long-acting dopamine D2/D3 agonist piribedil reduces the expression of dyskinesia while maintaining effective motor activity in MPTP-treated primates. Clin Neuropharmacol. 2006;29(3):112–25. doi:10.1097/01.WNF.0000220818.71231.DF.PubMed Smith LA, Jackson MJ, Johnston L, Kuoppamaki M, Rose S, Al-Barghouthy G, et al. Switching from levodopa to the long-acting dopamine D2/D3 agonist piribedil reduces the expression of dyskinesia while maintaining effective motor activity in MPTP-treated primates. Clin Neuropharmacol. 2006;29(3):112–25. doi:10.​1097/​01.​WNF.​0000220818.​71231.​DF.PubMed
46.
go back to reference Blanchet PJ, Calon F, Martel JC, Bedard PJ, Di Paolo T, Walters RR, et al. Continuous administration decreases and pulsatile administration increases behavioral sensitivity to a novel dopamine D2 agonist (U-91356A) in MPTP-exposed monkeys. J Pharmacol Exp Ther. 1995;272(2):854–9.PubMed Blanchet PJ, Calon F, Martel JC, Bedard PJ, Di Paolo T, Walters RR, et al. Continuous administration decreases and pulsatile administration increases behavioral sensitivity to a novel dopamine D2 agonist (U-91356A) in MPTP-exposed monkeys. J Pharmacol Exp Ther. 1995;272(2):854–9.PubMed
47.
go back to reference Stockwell KA, Scheller DK, Smith LA, Rose S, Iravani MM, Jackson MJ, 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. doi:10.1016/j.expneurol.2009.10.004.PubMed Stockwell KA, Scheller DK, Smith LA, Rose S, Iravani MM, Jackson MJ, 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. doi:10.​1016/​j.​expneurol.​2009.​10.​004.PubMed
51.
go back to reference 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.PubMed 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.PubMed
52.
go back to reference Hill MP, Brotchie JM, Crossman AR, Bezard E, Michel A, Grimee R, et al. Levetiracetam interferes with the l-dopa priming process in MPTP-lesioned drug-naive marmosets. Clin Neuropharmacol. 2004;27(4):171–7.PubMed Hill MP, Brotchie JM, Crossman AR, Bezard E, Michel A, Grimee R, et al. Levetiracetam interferes with the l-dopa priming process in MPTP-lesioned drug-naive marmosets. Clin Neuropharmacol. 2004;27(4):171–7.PubMed
53.
go back to reference 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.PubMed 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.PubMed
55.
go back to reference Montastruc JL, Rascol O, Senard JM, Rascol A. A randomised controlled study comparing bromocriptine to which levodopa was later added, with levodopa alone in previously untreated patients with Parkinson’s disease: a five year follow up. J Neurol Neurosurg Psychiatry. 1994;57(9):1034–8.PubMedCentralPubMed Montastruc JL, Rascol O, Senard JM, Rascol A. A randomised controlled study comparing bromocriptine to which levodopa was later added, with levodopa alone in previously untreated patients with Parkinson’s disease: a five year follow up. J Neurol Neurosurg Psychiatry. 1994;57(9):1034–8.PubMedCentralPubMed
56.
go back to reference Murray AM, Waddington JL. The interaction of clozapine with dopamine D1 versus dopamine D2 receptor-mediated function: behavioural indices. Eur J Pharmacol. 1990;186(1):79–86.PubMed Murray AM, Waddington JL. The interaction of clozapine with dopamine D1 versus dopamine D2 receptor-mediated function: behavioural indices. Eur J Pharmacol. 1990;186(1):79–86.PubMed
57.
go back to reference Maratos EC, Jackson MJ, Pearce RK, Cannizzaro C, Jenner P. Both short- and long-acting D-1/D-2 dopamine agonists induce less dyskinesia than l-dopa in the MPTP-lesioned common marmoset (Callithrix jacchus). Exp Neurol. 2003;179(1):90–102.PubMed Maratos EC, Jackson MJ, Pearce RK, Cannizzaro C, Jenner P. Both short- and long-acting D-1/D-2 dopamine agonists induce less dyskinesia than l-dopa in the MPTP-lesioned common marmoset (Callithrix jacchus). Exp Neurol. 2003;179(1):90–102.PubMed
58.
go back to reference Grondin R, Bedard PJ, Britton DR, Shiosaki K. Potential therapeutic use of the selective dopamine D1 receptor agonist, A-86929: an acute study in parkinsonian levodopa-primed monkeys. Neurology. 1997;49(2):421–6.PubMed Grondin R, Bedard PJ, Britton DR, Shiosaki K. Potential therapeutic use of the selective dopamine D1 receptor agonist, A-86929: an acute study in parkinsonian levodopa-primed monkeys. Neurology. 1997;49(2):421–6.PubMed
59.
go back to reference Pons R, Syrengelas D, Youroukos S, Orfanou I, Dinopoulos A, Cormand B, et al. Levodopa-induced dyskinesias in tyrosine hydroxylase deficiency. Mov Disord. 2013;28(8):1058–63. doi:10.1002/mds.25382.PubMed Pons R, Syrengelas D, Youroukos S, Orfanou I, Dinopoulos A, Cormand B, et al. Levodopa-induced dyskinesias in tyrosine hydroxylase deficiency. Mov Disord. 2013;28(8):1058–63. doi:10.​1002/​mds.​25382.PubMed
60.
go back to reference Hwang WJ, Calne DB, Tsui JK, de la Fuente-Fernandez R. The long-term response to levodopa in dopa-responsive dystonia. Parkinsonism Relat Disord. 2001;8(1):1–5.PubMed Hwang WJ, Calne DB, Tsui JK, de la Fuente-Fernandez R. The long-term response to levodopa in dopa-responsive dystonia. Parkinsonism Relat Disord. 2001;8(1):1–5.PubMed
61.
go back to reference Berthet A, Porras G, Doudnikoff E, Stark H, Cador M, Bezard E, et al. Pharmacological analysis demonstrates dramatic alteration of D1 dopamine receptor neuronal distribution in the rat analog of l-dopa-induced dyskinesia. J Neurosci. 2009;29(15):4829–35. doi:10.1523/JNEUROSCI.5884-08.2009.PubMed Berthet A, Porras G, Doudnikoff E, Stark H, Cador M, Bezard E, et al. Pharmacological analysis demonstrates dramatic alteration of D1 dopamine receptor neuronal distribution in the rat analog of l-dopa-induced dyskinesia. J Neurosci. 2009;29(15):4829–35. doi:10.​1523/​JNEUROSCI.​5884-08.​2009.PubMed
62.
go back to reference Oh JD, Russell DS, Vaughan CL, Chase TN. Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and l-dopa administration. Brain Res. 1998;813(1):150–9.PubMed Oh JD, Russell DS, Vaughan CL, Chase TN. Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and l-dopa administration. Brain Res. 1998;813(1):150–9.PubMed
64.
go back to reference Santini E, Valjent E, Usiello A, Carta M, Borgkvist A, Girault JA, et al. Critical involvement of cAMP/DARPP-32 and extracellular signal-regulated protein kinase signaling in l-dopa-induced dyskinesia. J Neurosci. 2007;27(26):6995–7005. doi:10.1523/JNEUROSCI.0852-07.2007.PubMed Santini E, Valjent E, Usiello A, Carta M, Borgkvist A, Girault JA, et al. Critical involvement of cAMP/DARPP-32 and extracellular signal-regulated protein kinase signaling in l-dopa-induced dyskinesia. J Neurosci. 2007;27(26):6995–7005. doi:10.​1523/​JNEUROSCI.​0852-07.​2007.PubMed
65.
go back to reference Iravani MM, Jenner P. Mechanisms underlying the onset and expression of levodopa-induced dyskinesia and their pharmacological manipulation. J Neural Transm. 2011;118(12):1661–90. doi:10.1007/s00702-011-0698-2.PubMed Iravani MM, Jenner P. Mechanisms underlying the onset and expression of levodopa-induced dyskinesia and their pharmacological manipulation. J Neural Transm. 2011;118(12):1661–90. doi:10.​1007/​s00702-011-0698-2.PubMed
66.
go back to reference Gerfen CR, Young WS 3rd. Distribution of striatonigral and striatopallidal peptidergic neurons in both patch and matrix compartments: an in situ hybridization histochemistry and fluorescent retrograde tracing study. Brain Res. 1988;460(1):161–7.PubMed Gerfen CR, Young WS 3rd. Distribution of striatonigral and striatopallidal peptidergic neurons in both patch and matrix compartments: an in situ hybridization histochemistry and fluorescent retrograde tracing study. Brain Res. 1988;460(1):161–7.PubMed
67.
go back to reference Le Moine C, Normand E, Bloch B. Phenotypical characterization of the rat striatal neurons expressing the D1 dopamine receptor gene. Proc Natl Acad Sci USA. 1991;88(10):4205–9.PubMedCentralPubMed Le Moine C, Normand E, Bloch B. Phenotypical characterization of the rat striatal neurons expressing the D1 dopamine receptor gene. Proc Natl Acad Sci USA. 1991;88(10):4205–9.PubMedCentralPubMed
69.
71.
go back to reference Aubert I, Guigoni C, Hakansson K, Li Q, Dovero S, Barthe N, et al. Increased D1 dopamine receptor signaling in levodopa-induced dyskinesia. Ann Neurol. 2005;57(1):17–26. doi:10.1002/ana.20296.PubMed Aubert I, Guigoni C, Hakansson K, Li Q, Dovero S, Barthe N, et al. Increased D1 dopamine receptor signaling in levodopa-induced dyskinesia. Ann Neurol. 2005;57(1):17–26. doi:10.​1002/​ana.​20296.PubMed
72.
go back to reference Santini E, Valjent E, Fisone G. mTORC1 signaling in Parkinson’s disease and l-dopa-induced dyskinesia: a sensitized matter. Cell Cycle. 2010;9(14):2713–8.PubMed Santini E, Valjent E, Fisone G. mTORC1 signaling in Parkinson’s disease and l-dopa-induced dyskinesia: a sensitized matter. Cell Cycle. 2010;9(14):2713–8.PubMed
75.
go back to reference Andersson M, Hilbertson A, Cenci MA. Striatal fosB expression is causally linked with l-dopa-induced abnormal involuntary movements and the associated upregulation of striatal prodynorphin mRNA in a rat model of Parkinson’s disease. Neurobiol Dis. 1999;6(6):461–74. doi:10.1006/nbdi.1999.0259.PubMed Andersson M, Hilbertson A, Cenci MA. Striatal fosB expression is causally linked with l-dopa-induced abnormal involuntary movements and the associated upregulation of striatal prodynorphin mRNA in a rat model of Parkinson’s disease. Neurobiol Dis. 1999;6(6):461–74. doi:10.​1006/​nbdi.​1999.​0259.PubMed
76.
go back to reference Gerfen CR, Miyachi S, Paletzki R, Brown P. D1 dopamine receptor supersensitivity in the dopamine-depleted striatum results from a switch in the regulation of ERK1/2/MAP kinase. J Neurosci. 2002;22(12):5042–54.PubMed Gerfen CR, Miyachi S, Paletzki R, Brown P. D1 dopamine receptor supersensitivity in the dopamine-depleted striatum results from a switch in the regulation of ERK1/2/MAP kinase. J Neurosci. 2002;22(12):5042–54.PubMed
78.
go back to reference Bezard E, Gross CE, Qin L, Gurevich VV, Benovic JL, Gurevich EV. l-dopa reverses the MPTP-induced elevation of the arrestin2 and GRK6 expression and enhanced ERK activation in monkey brain. Neurobiol Dis. 2005;18(2):323–35. doi:10.1016/j.nbd.2004.10.005.PubMed Bezard E, Gross CE, Qin L, Gurevich VV, Benovic JL, Gurevich EV. l-dopa reverses the MPTP-induced elevation of the arrestin2 and GRK6 expression and enhanced ERK activation in monkey brain. Neurobiol Dis. 2005;18(2):323–35. doi:10.​1016/​j.​nbd.​2004.​10.​005.PubMed
80.
go back to reference Chase TN, Oh JD. Striatal dopamine- and glutamate-mediated dysregulation in experimental parkinsonism. Trends Neurosci. 2000;23(10 Suppl):S86–91.PubMed Chase TN, Oh JD. Striatal dopamine- and glutamate-mediated dysregulation in experimental parkinsonism. Trends Neurosci. 2000;23(10 Suppl):S86–91.PubMed
81.
go back to reference Calabresi P, Giacomini P, Centonze D, Bernardi G. Levodopa-induced dyskinesia: a pathological form of striatal synaptic plasticity? Ann Neurol. 2000;47(4 Suppl 1):S60–8. (Discussion S8–9). Calabresi P, Giacomini P, Centonze D, Bernardi G. Levodopa-induced dyskinesia: a pathological form of striatal synaptic plasticity? Ann Neurol. 2000;47(4 Suppl 1):S60–8. (Discussion S8–9).
82.
go back to reference Calon F, Rajput AH, Hornykiewicz O, Bedard PJ, Di Paolo T. Levodopa-induced motor complications are associated with alterations of glutamate receptors in Parkinson’s disease. Neurobiol Dis. 2003;14(3):404–16.PubMed Calon F, Rajput AH, Hornykiewicz O, Bedard PJ, Di Paolo T. Levodopa-induced motor complications are associated with alterations of glutamate receptors in Parkinson’s disease. Neurobiol Dis. 2003;14(3):404–16.PubMed
83.
go back to reference Fiorentini C, Busi C, Spano P, Missale C. Role of receptor heterodimers in the development of l-dopa-induced dyskinesias in the 6-hydroxydopamine rat model of Parkinson’s disease. Parkinsonism Relat Disord. 2008;14(Suppl 2):S159–64. doi:10.1016/j.parkreldis.2008.04.022.PubMed Fiorentini C, Busi C, Spano P, Missale C. Role of receptor heterodimers in the development of l-dopa-induced dyskinesias in the 6-hydroxydopamine rat model of Parkinson’s disease. Parkinsonism Relat Disord. 2008;14(Suppl 2):S159–64. doi:10.​1016/​j.​parkreldis.​2008.​04.​022.PubMed
84.
go back to reference Fiorentini C, Rizzetti MC, Busi C, Bontempi S, Collo G, Spano P, et al. Loss of synaptic D1 dopamine/N-methyl-d-aspartate glutamate receptor complexes in l-dopa-induced dyskinesia in the rat. Mol Pharmacol. 2006;69(3):805–12. doi:10.1124/mol.105.016667.PubMed Fiorentini C, Rizzetti MC, Busi C, Bontempi S, Collo G, Spano P, et al. Loss of synaptic D1 dopamine/N-methyl-d-aspartate glutamate receptor complexes in l-dopa-induced dyskinesia in the rat. Mol Pharmacol. 2006;69(3):805–12. doi:10.​1124/​mol.​105.​016667.PubMed
85.
go back to reference Hallett PJ, Spoelgen R, Hyman BT, Standaert DG, Dunah AW. Dopamine D1 activation potentiates striatal NMDA receptors by tyrosine phosphorylation-dependent subunit trafficking. J Neurosci. 2006;26(17):4690–700. doi:10.1523/JNEUROSCI.0792-06.2006.PubMed Hallett PJ, Spoelgen R, Hyman BT, Standaert DG, Dunah AW. Dopamine D1 activation potentiates striatal NMDA receptors by tyrosine phosphorylation-dependent subunit trafficking. J Neurosci. 2006;26(17):4690–700. doi:10.​1523/​JNEUROSCI.​0792-06.​2006.PubMed
86.
go back to reference Picconi B, Paille V, Ghiglieri V, Bagetta V, Barone I, Lindgren HS, et al. l-dopa dosage is critically involved in dyskinesia via loss of synaptic depotentiation. Neurobiol Dis. 2008;29(2):327–35. doi:10.1016/j.nbd.2007.10.001.PubMed Picconi B, Paille V, Ghiglieri V, Bagetta V, Barone I, Lindgren HS, et al. l-dopa dosage is critically involved in dyskinesia via loss of synaptic depotentiation. Neurobiol Dis. 2008;29(2):327–35. doi:10.​1016/​j.​nbd.​2007.​10.​001.PubMed
87.
go back to reference Picconi B, Centonze D, Hakansson K, Bernardi G, Greengard P, Fisone G, et al. Loss of bidirectional striatal synaptic plasticity in l-dopa-induced dyskinesia. Nat Neurosci. 2003;6(5):501–6. doi:10.1038/nn1040.PubMed Picconi B, Centonze D, Hakansson K, Bernardi G, Greengard P, Fisone G, et al. Loss of bidirectional striatal synaptic plasticity in l-dopa-induced dyskinesia. Nat Neurosci. 2003;6(5):501–6. doi:10.​1038/​nn1040.PubMed
88.
go back to reference Hallett PJ, Dunah AW, Ravenscroft P, Zhou S, Bezard E, Crossman AR, et al. Alterations of striatal NMDA receptor subunits associated with the development of dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Neuropharmacology. 2005;48(4):503–16. doi:10.1016/j.neuropharm.2004.11.008.PubMed Hallett PJ, Dunah AW, Ravenscroft P, Zhou S, Bezard E, Crossman AR, et al. Alterations of striatal NMDA receptor subunits associated with the development of dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Neuropharmacology. 2005;48(4):503–16. doi:10.​1016/​j.​neuropharm.​2004.​11.​008.PubMed
89.
go back to reference Silverdale MA, Kobylecki C, Hallett PJ, Li Q, Dunah AW, Ravenscroft P, et al. Synaptic recruitment of AMPA glutamate receptor subunits in levodopa-induced dyskinesia in the MPTP-lesioned nonhuman primate. Synapse. 2010;64(2):177–80. doi:10.1002/syn.20739.PubMed Silverdale MA, Kobylecki C, Hallett PJ, Li Q, Dunah AW, Ravenscroft P, et al. Synaptic recruitment of AMPA glutamate receptor subunits in levodopa-induced dyskinesia in the MPTP-lesioned nonhuman primate. Synapse. 2010;64(2):177–80. doi:10.​1002/​syn.​20739.PubMed
90.
go back to reference Hurley MJ, Jackson MJ, Smith LA, Rose S, Jenner P. Immunoautoradiographic analysis of NMDA receptor subunits and associated postsynaptic density proteins in the brain of dyskinetic MPTP-treated common marmosets. Eur J Neurosci. 2005;21(12):3240–50. doi:10.1111/j.1460-9568.2005.04169.x.PubMed Hurley MJ, Jackson MJ, Smith LA, Rose S, Jenner P. Immunoautoradiographic analysis of NMDA receptor subunits and associated postsynaptic density proteins in the brain of dyskinetic MPTP-treated common marmosets. Eur J Neurosci. 2005;21(12):3240–50. doi:10.​1111/​j.​1460-9568.​2005.​04169.​x.PubMed
92.
go back to reference Sgambato-Faure V, Cenci MA. Glutamatergic mechanisms in the dyskinesias induced by pharmacological dopamine replacement and deep brain stimulation for the treatment of Parkinson’s disease. Prog Neurobiol. 2012;96(1):69–86. doi:10.1016/j.pneurobio.2011.10.005.PubMed Sgambato-Faure V, Cenci MA. Glutamatergic mechanisms in the dyskinesias induced by pharmacological dopamine replacement and deep brain stimulation for the treatment of Parkinson’s disease. Prog Neurobiol. 2012;96(1):69–86. doi:10.​1016/​j.​pneurobio.​2011.​10.​005.PubMed
93.
go back to reference Ahmed I, Bose SK, Pavese N, Ramlackhansingh A, Turkheimer F, Hotton G, et al. Glutamate NMDA receptor dysregulation in Parkinson’s disease with dyskinesias. Brain J Neurol. 2011;134(Pt 4):979–86. doi:10.1093/brain/awr028. Ahmed I, Bose SK, Pavese N, Ramlackhansingh A, Turkheimer F, Hotton G, et al. Glutamate NMDA receptor dysregulation in Parkinson’s disease with dyskinesias. Brain J Neurol. 2011;134(Pt 4):979–86. doi:10.​1093/​brain/​awr028.
94.
go back to reference Robelet S, Melon C, Guillet B, Salin P, Kerkerian-Le Goff L. Chronic l-dopa treatment increases extracellular glutamate levels and GLT1 expression in the basal ganglia in a rat model of Parkinson’s disease. Eur J Neurosci. 2004;20(5):1255–66. doi:10.1111/j.1460-9568.2004.03591.x.PubMed Robelet S, Melon C, Guillet B, Salin P, Kerkerian-Le Goff L. Chronic l-dopa treatment increases extracellular glutamate levels and GLT1 expression in the basal ganglia in a rat model of Parkinson’s disease. Eur J Neurosci. 2004;20(5):1255–66. doi:10.​1111/​j.​1460-9568.​2004.​03591.​x.PubMed
96.
go back to reference Luginger E, Wenning GK, Bosch S, Poewe W. Beneficial effects of amantadine on l-dopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2000;15(5):873–8.PubMed Luginger E, Wenning GK, Bosch S, Poewe W. Beneficial effects of amantadine on l-dopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2000;15(5):873–8.PubMed
97.
go back to reference Snow BJ, Macdonald L, McAuley D, Wallis W. The effect of amantadine on levodopa-induced dyskinesias in Parkinson’s disease: a double-blind, placebo-controlled study. Clin Neuropharmacol. 2000;23(2):82–5.PubMed Snow BJ, Macdonald L, McAuley D, Wallis W. The effect of amantadine on levodopa-induced dyskinesias in Parkinson’s disease: a double-blind, placebo-controlled study. Clin Neuropharmacol. 2000;23(2):82–5.PubMed
98.
go back to reference Metman LV, Del Dotto P, LePoole K, Konitsiotis S, Fang J, Chase TN. Amantadine for levodopa-induced dyskinesias: a 1-year follow-up study. Arch Neurol. 1999;56(11):1383–6.PubMed Metman LV, Del Dotto P, LePoole K, Konitsiotis S, Fang J, Chase TN. Amantadine for levodopa-induced dyskinesias: a 1-year follow-up study. Arch Neurol. 1999;56(11):1383–6.PubMed
99.
go back to reference Wolf E, Seppi K, Katzenschlager R, Hochschorner G, Ransmayr G, Schwingenschuh P, et al. Long-term antidyskinetic efficacy of amantadine in Parkinson’s disease. Mov Disord. 2010;25(10):1357–63. doi:10.1002/mds.23034.PubMed Wolf E, Seppi K, Katzenschlager R, Hochschorner G, Ransmayr G, Schwingenschuh P, et al. Long-term antidyskinetic efficacy of amantadine in Parkinson’s disease. Mov Disord. 2010;25(10):1357–63. doi:10.​1002/​mds.​23034.PubMed
100.
go back to reference Stocchi F, Rascol O, Destee A, Hattori N, Hauser RA, Lang AE, et al. AFQ056 in Parkinson patients with levodopa-induced dyskinesia: 13-week, randomized, dose-finding study. Mov Disord. 2013;28(13):1838–46. doi:10.1002/mds.25561.PubMed Stocchi F, Rascol O, Destee A, Hattori N, Hauser RA, Lang AE, et al. AFQ056 in Parkinson patients with levodopa-induced dyskinesia: 13-week, randomized, dose-finding study. Mov Disord. 2013;28(13):1838–46. doi:10.​1002/​mds.​25561.PubMed
101.
go back to reference Berg D, Godau J, Trenkwalder C, Eggert K, Csoti I, Storch A, et al. AFQ056 treatment of levodopa-induced dyskinesias: results of 2 randomized controlled trials. Mov Disord. 2011;26(7):1243–50. doi:10.1002/mds.23616.PubMed Berg D, Godau J, Trenkwalder C, Eggert K, Csoti I, Storch A, et al. AFQ056 treatment of levodopa-induced dyskinesias: results of 2 randomized controlled trials. Mov Disord. 2011;26(7):1243–50. doi:10.​1002/​mds.​23616.PubMed
102.
go back to reference Rylander D, Recchia A, Mela F, Dekundy A, Danysz W, Cenci MA. 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(1):227–35. doi:10.1124/jpet.108.150425.PubMedCentralPubMed Rylander D, Recchia A, Mela F, Dekundy A, Danysz W, Cenci MA. 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(1):227–35. doi:10.​1124/​jpet.​108.​150425.PubMedCentralPubMed
103.
go back to reference 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 J Neurol. 2007;130(Pt 7):1819–33. doi:10.1093/brain/awm082. 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 J Neurol. 2007;130(Pt 7):1819–33. doi:10.​1093/​brain/​awm082.
104.
105.
go back to reference Arai R, Karasawa N, Geffard M, Nagatsu I. l-dopa is converted to dopamine in serotonergic fibers of the striatum of the rat: a double-labeling immunofluorescence study. Neurosci Lett. 1995;195(3):195–8.PubMed Arai R, Karasawa N, Geffard M, Nagatsu I. l-dopa is converted to dopamine in serotonergic fibers of the striatum of the rat: a double-labeling immunofluorescence study. Neurosci Lett. 1995;195(3):195–8.PubMed
106.
go back to reference Arai R, Karasawa N, Geffard M, Nagatsu T, Nagatsu I. 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.PubMed Arai R, Karasawa N, Geffard M, Nagatsu T, Nagatsu I. 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.PubMed
107.
108.
go back to reference Maeda T, Nagata K, Yoshida Y, Kannari K. Serotonergic hyperinnervation into the dopaminergic denervated striatum compensates for dopamine conversion from exogenously administered l-dopa. Brain Res. 2005;1046(1–2):230–3. doi:10.1016/j.brainres.2005.04.019.PubMed Maeda T, Nagata K, Yoshida Y, Kannari K. Serotonergic hyperinnervation into the dopaminergic denervated striatum compensates for dopamine conversion from exogenously administered l-dopa. Brain Res. 2005;1046(1–2):230–3. doi:10.​1016/​j.​brainres.​2005.​04.​019.PubMed
109.
go back to reference Yamada H, Aimi Y, Nagatsu I, Taki K, Kudo M, Arai R. Immunohistochemical detection of l-dopa-derived dopamine within serotonergic fibers in the striatum and the substantia nigra pars reticulata in Parkinsonian model rats. Neurosci Res. 2007;59(1):1–7. doi:10.1016/j.neures.2007.05.002.PubMed Yamada H, Aimi Y, Nagatsu I, Taki K, Kudo M, Arai R. Immunohistochemical detection of l-dopa-derived dopamine within serotonergic fibers in the striatum and the substantia nigra pars reticulata in Parkinsonian model rats. Neurosci Res. 2007;59(1):1–7. doi:10.​1016/​j.​neures.​2007.​05.​002.PubMed
110.
go back to reference Carta M, Carlsson T, Munoz A, Kirik D, Bjorklund A. Role of serotonin neurons in the induction of levodopa- and graft-induced dyskinesias in Parkinson’s disease. Mov Disord. 2010;25(Suppl 1):S174–9. doi:10.1002/mds.22792.PubMed Carta M, Carlsson T, Munoz A, Kirik D, Bjorklund A. Role of serotonin neurons in the induction of levodopa- and graft-induced dyskinesias in Parkinson’s disease. Mov Disord. 2010;25(Suppl 1):S174–9. doi:10.​1002/​mds.​22792.PubMed
112.
go back to reference Lopez A, Munoz A, Guerra MJ, Labandeira-Garcia JL. Mechanisms of the effects of exogenous levodopa on the dopamine-denervated striatum. Neuroscience. 2001;103(3):639–51.PubMed Lopez A, Munoz A, Guerra MJ, Labandeira-Garcia JL. Mechanisms of the effects of exogenous levodopa on the dopamine-denervated striatum. Neuroscience. 2001;103(3):639–51.PubMed
113.
go back to reference Tanaka H, Kannari K, Maeda T, Tomiyama M, Suda T, Matsunaga M. Role of serotonergic neurons in l-dopa-derived extracellular dopamine in the striatum of 6-OHDA-lesioned rats. Neuroreport. 1999;10(3):631–4.PubMed Tanaka H, Kannari K, Maeda T, Tomiyama M, Suda T, Matsunaga M. Role of serotonergic neurons in l-dopa-derived extracellular dopamine in the striatum of 6-OHDA-lesioned rats. Neuroreport. 1999;10(3):631–4.PubMed
114.
go back to reference Eskow KL, Dupre KB, Barnum CJ, Dickinson SO, Park JY, Bishop C. The role of the dorsal raphe nucleus in the development, expression, and treatment of l-dopa-induced dyskinesia in hemiparkinsonian rats. Synapse. 2009;63(7):610–20. doi:10.1002/syn.20630.PubMedCentralPubMed Eskow KL, Dupre KB, Barnum CJ, Dickinson SO, Park JY, Bishop C. The role of the dorsal raphe nucleus in the development, expression, and treatment of l-dopa-induced dyskinesia in hemiparkinsonian rats. Synapse. 2009;63(7):610–20. doi:10.​1002/​syn.​20630.PubMedCentralPubMed
115.
116.
go back to reference Lindgren HS, Andersson DR, Lagerkvist S, Nissbrandt H, Cenci MA. 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(6):1465–76. doi:10.1111/j.1471-4159.2009.06556.x.PubMed Lindgren HS, Andersson DR, Lagerkvist S, Nissbrandt H, Cenci MA. 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(6):1465–76. doi:10.​1111/​j.​1471-4159.​2009.​06556.​x.PubMed
117.
go back to reference Kannari K, Yamato H, Shen H, Tomiyama M, Suda T, Matsunaga M. Activation of 5-HT(1A) but not 5-HT(1B) receptors attenuates an increase in extracellular dopamine derived from exogenously administered l-dopa in the striatum with nigrostriatal denervation. J Neurochem. 2001;76(5):1346–53.PubMed Kannari K, Yamato H, Shen H, Tomiyama M, Suda T, Matsunaga M. Activation of 5-HT(1A) but not 5-HT(1B) receptors attenuates an increase in extracellular dopamine derived from exogenously administered l-dopa in the striatum with nigrostriatal denervation. J Neurochem. 2001;76(5):1346–53.PubMed
118.
go back to reference Munoz A, Carlsson T, Tronci E, Kirik D, Bjorklund A, Carta M. Serotonin neuron-dependent and -independent reduction of dyskinesia by 5-HT1A and 5-HT1B receptor agonists in the rat Parkinson model. Exp Neurol. 2009;219(1):298–307. doi:10.1016/j.expneurol.2009.05.033.PubMed Munoz A, Carlsson T, Tronci E, Kirik D, Bjorklund A, Carta M. Serotonin neuron-dependent and -independent reduction of dyskinesia by 5-HT1A and 5-HT1B receptor agonists in the rat Parkinson model. Exp Neurol. 2009;219(1):298–307. doi:10.​1016/​j.​expneurol.​2009.​05.​033.PubMed
119.
go back to reference Munoz A, Li Q, Gardoni F, Marcello E, Qin C, Carlsson T, et al. Combined 5-HT1A and 5-HT1B receptor agonists for the treatment of l-dopa-induced dyskinesia. Brain J Neurol. 2008;131(Pt 12):3380–94. doi:10.1093/brain/awn235. Munoz A, Li Q, Gardoni F, Marcello E, Qin C, Carlsson T, et al. Combined 5-HT1A and 5-HT1B receptor agonists for the treatment of l-dopa-induced dyskinesia. Brain J Neurol. 2008;131(Pt 12):3380–94. doi:10.​1093/​brain/​awn235.
120.
121.
go back to reference Navailles S, Bioulac B, Gross C, De Deurwaerdere P. Serotonergic neurons mediate ectopic release of dopamine induced by l-dopa in a rat model of Parkinson’s disease. Neurobiol Dis. 2010;38(1):136–43. doi:10.1016/j.nbd.2010.01.012.PubMed Navailles S, Bioulac B, Gross C, De Deurwaerdere P. Serotonergic neurons mediate ectopic release of dopamine induced by l-dopa in a rat model of Parkinson’s disease. Neurobiol Dis. 2010;38(1):136–43. doi:10.​1016/​j.​nbd.​2010.​01.​012.PubMed
122.
go back to reference Politis M, Wu K, Loane C, Quinn NP, Brooks DJ, Rehncrona S, et al. Serotonergic neurons mediate dyskinesia side effects in Parkinson’s patients with neural transplants. Sci Transl Med. 2010;2(38):38ra46. doi:10.1126/scitranslmed.3000976. Politis M, Wu K, Loane C, Quinn NP, Brooks DJ, Rehncrona S, et al. Serotonergic neurons mediate dyskinesia side effects in Parkinson’s patients with neural transplants. Sci Transl Med. 2010;2(38):38ra46. doi:10.​1126/​scitranslmed.​3000976.
123.
go back to reference Politis M, Oertel WH, Wu K, Quinn NP, Pogarell O, Brooks DJ, et al. Graft-induced dyskinesias in Parkinson’s disease: High striatal serotonin/dopamine transporter ratio. Mov Disord. 2011;26(11):1997–2003. doi:10.1002/mds.23743.PubMed Politis M, Oertel WH, Wu K, Quinn NP, Pogarell O, Brooks DJ, et al. Graft-induced dyskinesias in Parkinson’s disease: High striatal serotonin/dopamine transporter ratio. Mov Disord. 2011;26(11):1997–2003. doi:10.​1002/​mds.​23743.PubMed
124.
go back to reference Hagell P, Piccini P, Bjorklund A, Brundin P, Rehncrona S, Widner H, et al. Dyskinesias following neural transplantation in Parkinson’s disease. Nat Neurosci. 2002;5(7):627–8. doi:10.1038/nn863.PubMed Hagell P, Piccini P, Bjorklund A, Brundin P, Rehncrona S, Widner H, et al. Dyskinesias following neural transplantation in Parkinson’s disease. Nat Neurosci. 2002;5(7):627–8. doi:10.​1038/​nn863.PubMed
125.
go back to reference Zeng BY, Iravani MM, Jackson MJ, Rose S, Parent A, Jenner P. Morphological changes in serotoninergic neurites in the striatum and globus pallidus in levodopa primed MPTP treated common marmosets with dyskinesia. Neurobiol Dis. 2010;40(3):599–607. doi:10.1016/j.nbd.2010.08.004.PubMed Zeng BY, Iravani MM, Jackson MJ, Rose S, Parent A, Jenner P. Morphological changes in serotoninergic neurites in the striatum and globus pallidus in levodopa primed MPTP treated common marmosets with dyskinesia. Neurobiol Dis. 2010;40(3):599–607. doi:10.​1016/​j.​nbd.​2010.​08.​004.PubMed
126.
go back to reference Rylander D, Parent M, O’Sullivan SS, Dovero S, Lees AJ, Bezard E, et al. Maladaptive plasticity of serotonin axon terminals in levodopa-induced dyskinesia. Ann Neurol. 2010;68(5):619–28. doi:10.1002/ana.22097.PubMed Rylander D, Parent M, O’Sullivan SS, Dovero S, Lees AJ, Bezard E, et al. Maladaptive plasticity of serotonin axon terminals in levodopa-induced dyskinesia. Ann Neurol. 2010;68(5):619–28. doi:10.​1002/​ana.​22097.PubMed
127.
go back to reference Guerra MJ, Liste I, Labandeira-Garcia JL. Effects of lesions of the nigrostriatal pathway and of nigral grafts on striatal serotonergic innervation in adult rats. Neuroreport. 1997;8(16):3485–8.PubMed Guerra MJ, Liste I, Labandeira-Garcia JL. Effects of lesions of the nigrostriatal pathway and of nigral grafts on striatal serotonergic innervation in adult rats. Neuroreport. 1997;8(16):3485–8.PubMed
128.
go back to reference Fornai F, di Poggio AB, Pellegrini A, Ruggieri S, Paparelli A. Noradrenaline in Parkinson’s disease: from disease progression to current therapeutics. Curr Med Chem. 2007;14(22):2330–4.PubMed Fornai F, di Poggio AB, Pellegrini A, Ruggieri S, Paparelli A. Noradrenaline in Parkinson’s disease: from disease progression to current therapeutics. Curr Med Chem. 2007;14(22):2330–4.PubMed
129.
go back to reference Grenhoff J, Nisell M, Ferre S, Aston-Jones G, Svensson TH. Noradrenergic modulation of midbrain dopamine cell firing elicited by stimulation of the locus coeruleus in the rat. J Neural Transm Gen Sect. 1993;93(1):11–25.PubMed Grenhoff J, Nisell M, Ferre S, Aston-Jones G, Svensson TH. Noradrenergic modulation of midbrain dopamine cell firing elicited by stimulation of the locus coeruleus in the rat. J Neural Transm Gen Sect. 1993;93(1):11–25.PubMed
130.
go back to reference Bucheler MM, Hadamek K, Hein L. Two alpha(2)-adrenergic receptor subtypes, alpha(2A) and alpha(2C), inhibit transmitter release in the brain of gene-targeted mice. Neuroscience. 2002;109(4):819–26.PubMed Bucheler MM, Hadamek K, Hein L. Two alpha(2)-adrenergic receptor subtypes, alpha(2A) and alpha(2C), inhibit transmitter release in the brain of gene-targeted mice. Neuroscience. 2002;109(4):819–26.PubMed
131.
go back to reference Foote SL, Bloom FE, Aston-Jones G. Nucleus locus ceruleus: new evidence of anatomical and physiological specificity. Physiol Rev. 1983;63(3):844–914.PubMed Foote SL, Bloom FE, Aston-Jones G. Nucleus locus ceruleus: new evidence of anatomical and physiological specificity. Physiol Rev. 1983;63(3):844–914.PubMed
132.
go back to reference Jones BE, Yang TZ. The efferent projections from the reticular formation and the locus coeruleus studied by anterograde and retrograde axonal transport in the rat. J Comp Neurol. 1985;242(1):56–92. doi:10.1002/cne.902420105.PubMed Jones BE, Yang TZ. The efferent projections from the reticular formation and the locus coeruleus studied by anterograde and retrograde axonal transport in the rat. J Comp Neurol. 1985;242(1):56–92. doi:10.​1002/​cne.​902420105.PubMed
133.
go back to reference Gaspar P, Stepniewska I, Kaas JH. Topography and collateralization of the dopaminergic projections to motor and lateral prefrontal cortex in owl monkeys. J Comp Neurol. 1992;325(1):1–21. doi:10.1002/cne.903250102.PubMed Gaspar P, Stepniewska I, Kaas JH. Topography and collateralization of the dopaminergic projections to motor and lateral prefrontal cortex in owl monkeys. J Comp Neurol. 1992;325(1):1–21. doi:10.​1002/​cne.​903250102.PubMed
134.
go back to reference Ordway GA, Jaconetta SM, Halaris AE. Characterization of subtypes of alpha-2 adrenoceptors in the human brain. J Pharmacol Exp Ther. 1993;264(2):967–76.PubMed Ordway GA, Jaconetta SM, Halaris AE. Characterization of subtypes of alpha-2 adrenoceptors in the human brain. J Pharmacol Exp Ther. 1993;264(2):967–76.PubMed
136.
go back to reference Hill MP, Brotchie JM. The adrenergic receptor agonist, clonidine, potentiates the anti-parkinsonian action of the selective kappa-opioid receptor agonist, enadoline, in the monoamine-depleted rat. Br J Pharmacol. 1999;128(7):1577–85. doi:10.1038/sj.bjp.0702943.PubMedCentralPubMed Hill MP, Brotchie JM. The adrenergic receptor agonist, clonidine, potentiates the anti-parkinsonian action of the selective kappa-opioid receptor agonist, enadoline, in the monoamine-depleted rat. Br J Pharmacol. 1999;128(7):1577–85. doi:10.​1038/​sj.​bjp.​0702943.PubMedCentralPubMed
137.
go back to reference Zhang W, Ordway GA. The alpha2C-adrenoceptor modulates GABA release in mouse striatum. Brain Res Mol Brain Res. 2003;112(1–2):24–32.PubMed Zhang W, Ordway GA. The alpha2C-adrenoceptor modulates GABA release in mouse striatum. Brain Res Mol Brain Res. 2003;112(1–2):24–32.PubMed
138.
go back to reference Alachkar A, Brotchie JM, Jones OT. Changes in the mRNA levels of alpha2A and alpha2C adrenergic receptors in rat models of Parkinson’s disease and l-dopa-induced dyskinesia. J Mol Neurosci. 2012;46(1):145–52. doi:10.1007/s12031-011-9539-x.PubMed Alachkar A, Brotchie JM, Jones OT. Changes in the mRNA levels of alpha2A and alpha2C adrenergic receptors in rat models of Parkinson’s disease and l-dopa-induced dyskinesia. J Mol Neurosci. 2012;46(1):145–52. doi:10.​1007/​s12031-011-9539-x.PubMed
139.
go back to reference Barnum CJ, Bhide N, Lindenbach D, Surrena MA, Goldenberg AA, Tignor S, et al. Effects of noradrenergic denervation on l-dopa-induced dyskinesia and its treatment by alpha- and beta-adrenergic receptor antagonists in hemiparkinsonian rats. Pharmacol Biochem Behav. 2012;100(3):607–15. doi:10.1016/j.pbb.2011.09.009.PubMedCentralPubMed Barnum CJ, Bhide N, Lindenbach D, Surrena MA, Goldenberg AA, Tignor S, et al. Effects of noradrenergic denervation on l-dopa-induced dyskinesia and its treatment by alpha- and beta-adrenergic receptor antagonists in hemiparkinsonian rats. Pharmacol Biochem Behav. 2012;100(3):607–15. doi:10.​1016/​j.​pbb.​2011.​09.​009.PubMedCentralPubMed
140.
go back to reference Fulceri F, Biagioni F, Ferrucci M, Lazzeri G, Bartalucci A, Galli V, et al. Abnormal involuntary movements (AIMs) following pulsatile dopaminergic stimulation: severe deterioration and morphological correlates following the loss of locus coeruleus neurons. Brain Res. 2007;1135(1):219–29. doi:10.1016/j.brainres.2006.12.030.PubMed Fulceri F, Biagioni F, Ferrucci M, Lazzeri G, Bartalucci A, Galli V, et al. Abnormal involuntary movements (AIMs) following pulsatile dopaminergic stimulation: severe deterioration and morphological correlates following the loss of locus coeruleus neurons. Brain Res. 2007;1135(1):219–29. doi:10.​1016/​j.​brainres.​2006.​12.​030.PubMed
141.
go back to reference Piccini P, Weeks RA, Brooks DJ. Alterations in opioid receptor binding in Parkinson’s disease patients with levodopa-induced dyskinesias. Ann Neurol. 1997;42(5):720–6. doi:10.1002/ana.410420508.PubMed Piccini P, Weeks RA, Brooks DJ. Alterations in opioid receptor binding in Parkinson’s disease patients with levodopa-induced dyskinesias. Ann Neurol. 1997;42(5):720–6. doi:10.​1002/​ana.​410420508.PubMed
142.
go back to reference Cohen RM, Carson RE, Aigner TG, Doudet DJ. Opiate receptor avidity is reduced in non-motor impaired MPTP-lesioned rhesus monkeys. Brain Res. 1998;806(2):292–6.PubMed Cohen RM, Carson RE, Aigner TG, Doudet DJ. Opiate receptor avidity is reduced in non-motor impaired MPTP-lesioned rhesus monkeys. Brain Res. 1998;806(2):292–6.PubMed
143.
go back to reference Aubert I, Guigoni C, Li Q, Dovero S, Bioulac BH, Gross CE, et al. Enhanced preproenkephalin-B-derived opioid transmission in striatum and subthalamic nucleus converges upon globus pallidus internalis in l-3,4-dihydroxyphenylalanine-induced dyskinesia. Biol Psychiatry. 2007;61(7):836–44. doi:10.1016/j.biopsych.2006.06.038.PubMed Aubert I, Guigoni C, Li Q, Dovero S, Bioulac BH, Gross CE, et al. Enhanced preproenkephalin-B-derived opioid transmission in striatum and subthalamic nucleus converges upon globus pallidus internalis in l-3,4-dihydroxyphenylalanine-induced dyskinesia. Biol Psychiatry. 2007;61(7):836–44. doi:10.​1016/​j.​biopsych.​2006.​06.​038.PubMed
144.
go back to reference Gerdeman G, Lovinger DM. CB1 cannabinoid receptor inhibits synaptic release of glutamate in rat dorsolateral striatum. J Neurophysiol. 2001;85(1):468–71.PubMed Gerdeman G, Lovinger DM. CB1 cannabinoid receptor inhibits synaptic release of glutamate in rat dorsolateral striatum. J Neurophysiol. 2001;85(1):468–71.PubMed
145.
go back to reference Mailleux P, Parmentier M, Vanderhaeghen JJ. Distribution of cannabinoid receptor messenger RNA in the human brain: an in situ hybridization histochemistry with oligonucleotides. Neurosci Lett. 1992;143(1–2):200–4.PubMed Mailleux P, Parmentier M, Vanderhaeghen JJ. Distribution of cannabinoid receptor messenger RNA in the human brain: an in situ hybridization histochemistry with oligonucleotides. Neurosci Lett. 1992;143(1–2):200–4.PubMed
146.
go back to reference Wang Y, Zhang QJ, Wang HS, Wang T, Liu J. Genome-wide microarray analysis identifies a potential role for striatal retrograde endocannabinoid signaling in the pathogenesis of experimental l-dopa-induced dyskinesia. Synapse. 2014;. doi:10.1002/syn.21740. Wang Y, Zhang QJ, Wang HS, Wang T, Liu J. Genome-wide microarray analysis identifies a potential role for striatal retrograde endocannabinoid signaling in the pathogenesis of experimental l-dopa-induced dyskinesia. Synapse. 2014;. doi:10.​1002/​syn.​21740.
147.
go back to reference Ferre S, Popoli P, Gimenez-Llort L, Rimondini R, Muller CE, Stromberg I, et al. Adenosine/dopamine interaction: implications for the treatment of Parkinson’s disease. Parkinsonism Relat Disord. 2001;7(3):235–41.PubMed Ferre S, Popoli P, Gimenez-Llort L, Rimondini R, Muller CE, Stromberg I, et al. Adenosine/dopamine interaction: implications for the treatment of Parkinson’s disease. Parkinsonism Relat Disord. 2001;7(3):235–41.PubMed
148.
go back to reference Kase H. New aspects of physiological and pathophysiological functions of adenosine A2A receptor in basal ganglia. Biosci Biotechnol Biochem. 2001;65(7):1447–57.PubMed Kase H. New aspects of physiological and pathophysiological functions of adenosine A2A receptor in basal ganglia. Biosci Biotechnol Biochem. 2001;65(7):1447–57.PubMed
149.
go back to reference Calon F, Dridi M, Hornykiewicz O, Bedard PJ, Rajput AH, Di Paolo T. Increased adenosine A2A receptors in the brain of Parkinson’s disease patients with dyskinesias. Brain J Neurol. 2004;127(Pt 5):1075–84. doi:10.1093/brain/awh128. Calon F, Dridi M, Hornykiewicz O, Bedard PJ, Rajput AH, Di Paolo T. Increased adenosine A2A receptors in the brain of Parkinson’s disease patients with dyskinesias. Brain J Neurol. 2004;127(Pt 5):1075–84. doi:10.​1093/​brain/​awh128.
150.
go back to reference Xiao D, Bastia E, Xu YH, Benn CL, Cha JH, Peterson TS, et al. Forebrain adenosine A2A receptors contribute to l-3,4-dihydroxyphenylalanine-induced dyskinesia in hemiparkinsonian mice. J Neurosci. 2006;26(52):13548–55. doi:10.1523/JNEUROSCI.3554-06.2006.PubMed Xiao D, Bastia E, Xu YH, Benn CL, Cha JH, Peterson TS, et al. Forebrain adenosine A2A receptors contribute to l-3,4-dihydroxyphenylalanine-induced dyskinesia in hemiparkinsonian mice. J Neurosci. 2006;26(52):13548–55. doi:10.​1523/​JNEUROSCI.​3554-06.​2006.PubMed
151.
go back to reference Koranda JL, Cone JJ, McGehee DS, Roitman MF, Beeler JA, Zhuang X. Nicotinic receptors regulate the dynamic range of dopamine release in vivo. J Neurophysiol. 2014;111(1):103–11. doi:10.1152/jn.00269.2013.PubMed Koranda JL, Cone JJ, McGehee DS, Roitman MF, Beeler JA, Zhuang X. Nicotinic receptors regulate the dynamic range of dopamine release in vivo. J Neurophysiol. 2014;111(1):103–11. doi:10.​1152/​jn.​00269.​2013.PubMed
152.
go back to reference Perez XA, O’Leary KT, Parameswaran N, McIntosh JM, Quik M. Prominent role of alpha3/alpha6beta2* nAChRs in regulating evoked dopamine release in primate putamen: effect of long-term nicotine treatment. Mol Pharmacol. 2009;75(4):938–46. doi:10.1124/mol.108.053801.PubMedCentralPubMed Perez XA, O’Leary KT, Parameswaran N, McIntosh JM, Quik M. Prominent role of alpha3/alpha6beta2* nAChRs in regulating evoked dopamine release in primate putamen: effect of long-term nicotine treatment. Mol Pharmacol. 2009;75(4):938–46. doi:10.​1124/​mol.​108.​053801.PubMedCentralPubMed
153.
go back to reference Garcao P, Szabo EC, Wopereis S, Castro AA, Tome AR, Prediger RD, et al. Functional interaction between pre-synaptic alpha6beta2-containing nicotinic and adenosine A2A receptors in the control of dopamine release in the rat striatum. Br J Pharmacol. 2013;169(7):1600–11. doi:10.1111/bph.12234.PubMedCentralPubMed Garcao P, Szabo EC, Wopereis S, Castro AA, Tome AR, Prediger RD, et al. Functional interaction between pre-synaptic alpha6beta2-containing nicotinic and adenosine A2A receptors in the control of dopamine release in the rat striatum. Br J Pharmacol. 2013;169(7):1600–11. doi:10.​1111/​bph.​12234.PubMedCentralPubMed
154.
go back to reference Moreno E, Hoffmann H, Gonzalez-Sepulveda M, Navarro G, Casado V, Cortes A, et al. Dopamine D1-histamine H3 receptor heteromers provide a selective link to MAPK signaling in GABAergic neurons of the direct striatal pathway. J Biol Chem. 2011;286(7):5846–54. doi:10.1074/jbc.M110.161489.PubMedCentralPubMed Moreno E, Hoffmann H, Gonzalez-Sepulveda M, Navarro G, Casado V, Cortes A, et al. Dopamine D1-histamine H3 receptor heteromers provide a selective link to MAPK signaling in GABAergic neurons of the direct striatal pathway. J Biol Chem. 2011;286(7):5846–54. doi:10.​1074/​jbc.​M110.​161489.PubMedCentralPubMed
155.
go back to reference Prast H, Tran MH, Fischer H, Kraus M, Lamberti C, Grass K, et al. Histaminergic neurons modulate acetylcholine release in the ventral striatum: role of H3 histamine receptors. Naunyn Schmiedebergs Arch Pharmacol. 1999;360(5):558–64.PubMed Prast H, Tran MH, Fischer H, Kraus M, Lamberti C, Grass K, et al. Histaminergic neurons modulate acetylcholine release in the ventral striatum: role of H3 histamine receptors. Naunyn Schmiedebergs Arch Pharmacol. 1999;360(5):558–64.PubMed
156.
go back to reference Molina-Hernandez A, Nunez A, Arias-Montano JA. Histamine H3-receptor activation inhibits dopamine synthesis in rat striatum. Neuroreport. 2000;11(1):163–6.PubMed Molina-Hernandez A, Nunez A, Arias-Montano JA. Histamine H3-receptor activation inhibits dopamine synthesis in rat striatum. Neuroreport. 2000;11(1):163–6.PubMed
157.
go back to reference Gomez-Ramirez J, Johnston TH, Visanji NP, Fox SH, Brotchie JM. Histamine H3 receptor agonists reduce l-dopa-induced chorea, but not dystonia, in the MPTP-lesioned nonhuman primate model of Parkinson’s disease. Mov Disord. 2006;21(6):839–46. doi:10.1002/mds.20828.PubMed Gomez-Ramirez J, Johnston TH, Visanji NP, Fox SH, Brotchie JM. Histamine H3 receptor agonists reduce l-dopa-induced chorea, but not dystonia, in the MPTP-lesioned nonhuman primate model of Parkinson’s disease. Mov Disord. 2006;21(6):839–46. doi:10.​1002/​mds.​20828.PubMed
158.
go back to reference Lindgren HS, Ohlin KE, Cenci MA. Differential involvement of D1 and D2 dopamine receptors in l-dopa-induced angiogenic activity in a rat model of Parkinson’s disease. Neuropsychopharmacology. 2009;34(12):2477–88. doi:10.1038/npp.2009.74.PubMed Lindgren HS, Ohlin KE, Cenci MA. Differential involvement of D1 and D2 dopamine receptors in l-dopa-induced angiogenic activity in a rat model of Parkinson’s disease. Neuropsychopharmacology. 2009;34(12):2477–88. doi:10.​1038/​npp.​2009.​74.PubMed
159.
go back to reference Westin JE, Lindgren HS, Gardi J, Nyengaard JR, Brundin P, Mohapel P, et al. Endothelial proliferation and increased blood-brain barrier permeability in the basal ganglia in a rat model of 3,4-dihydroxyphenyl-l-alanine-induced dyskinesia. J Neurosci. 2006;26(37):9448–61. doi:10.1523/JNEUROSCI.0944-06.2006.PubMed Westin JE, Lindgren HS, Gardi J, Nyengaard JR, Brundin P, Mohapel P, et al. Endothelial proliferation and increased blood-brain barrier permeability in the basal ganglia in a rat model of 3,4-dihydroxyphenyl-l-alanine-induced dyskinesia. J Neurosci. 2006;26(37):9448–61. doi:10.​1523/​JNEUROSCI.​0944-06.​2006.PubMed
160.
go back to reference Ohlin KE, Francardo V, Lindgren HS, Sillivan SE, O’Sullivan SS, Luksik AS, et al. Vascular endothelial growth factor is upregulated by l-dopa in the parkinsonian brain: implications for the development of dyskinesia. Brain J Neurol. 2011;134(Pt 8):2339–57. doi:10.1093/brain/awr165. Ohlin KE, Francardo V, Lindgren HS, Sillivan SE, O’Sullivan SS, Luksik AS, et al. Vascular endothelial growth factor is upregulated by l-dopa in the parkinsonian brain: implications for the development of dyskinesia. Brain J Neurol. 2011;134(Pt 8):2339–57. doi:10.​1093/​brain/​awr165.
163.
go back to reference Fahn S. A new look at levodopa based on the ELLDOPA study. J Neural Transm Suppl. 2006;70:419–26.PubMed Fahn S. A new look at levodopa based on the ELLDOPA study. J Neural Transm Suppl. 2006;70:419–26.PubMed
164.
go back to reference Fahn S, Oakes D, Shoulson I, Kieburtz K, Rudolph A, Lang A, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med. 2004;351(24):2498–508. doi:10.1056/NEJMoa033447.PubMed Fahn S, Oakes D, Shoulson I, Kieburtz K, Rudolph A, Lang A, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med. 2004;351(24):2498–508. doi:10.​1056/​NEJMoa033447.PubMed
165.
go back to reference Sage JI, Mark MH. Comparison of controlled-release Sinemet (CR4) and standard Sinemet (25 mg/100 mg) in advanced Parkinson’s disease: a double-blind, crossover study. Clin Neuropharmacol. 1988;11(2):174–9.PubMed Sage JI, Mark MH. Comparison of controlled-release Sinemet (CR4) and standard Sinemet (25 mg/100 mg) in advanced Parkinson’s disease: a double-blind, crossover study. Clin Neuropharmacol. 1988;11(2):174–9.PubMed
166.
go back to reference Ahlskog JE, Muenter MD, McManis PG, Bell GN, Bailey PA. Controlled-release Sinemet (CR-4): a double-blind crossover study in patients with fluctuating Parkinson’s disease. Mayo Clin Proc. 1988;63(9):876–86.PubMed Ahlskog JE, Muenter MD, McManis PG, Bell GN, Bailey PA. Controlled-release Sinemet (CR-4): a double-blind crossover study in patients with fluctuating Parkinson’s disease. Mayo Clin Proc. 1988;63(9):876–86.PubMed
167.
go back to reference Hutton JT, Morris JL, Bush DF, Smith ME, Liss CL, Reines S. Multicenter controlled study of Sinemet CR vs Sinemet (25/100) in advanced Parkinson’s disease. Neurology. 1989;39(11 Suppl 2):67–72. (Discussion 3). Hutton JT, Morris JL, Bush DF, Smith ME, Liss CL, Reines S. Multicenter controlled study of Sinemet CR vs Sinemet (25/100) in advanced Parkinson’s disease. Neurology. 1989;39(11 Suppl 2):67–72. (Discussion 3).
168.
go back to reference Jankovic J, Schwartz K, Vander Linden C. Comparison of Sinemet CR4 and standard Sinemet: double blind and long-term open trial in parkinsonian patients with fluctuations. Mov Disord. 1989;4(4):303–9. doi:10.1002/mds.870040403.PubMed Jankovic J, Schwartz K, Vander Linden C. Comparison of Sinemet CR4 and standard Sinemet: double blind and long-term open trial in parkinsonian patients with fluctuations. Mov Disord. 1989;4(4):303–9. doi:10.​1002/​mds.​870040403.PubMed
169.
go back to reference Lieberman A, Gopinathan G, Miller E, Neophytides A, Baumann G, Chin L. Randomized double-blind cross-over study of Sinemet-controlled release (CR4 50/200) versus Sinemet 25/100 in Parkinson’s disease. Eur Neurol. 1990;30(2):75–8.PubMed Lieberman A, Gopinathan G, Miller E, Neophytides A, Baumann G, Chin L. Randomized double-blind cross-over study of Sinemet-controlled release (CR4 50/200) versus Sinemet 25/100 in Parkinson’s disease. Eur Neurol. 1990;30(2):75–8.PubMed
170.
go back to reference Wolters EC, Horstink MW, Roos RA, Jansen EN. Clinical efficacy of Sinemet CR 50/200 versus Sinemet 25/100 in patients with fluctuating Parkinson’s disease. An open, and a double-blind, double-dummy, multicenter treatment evaluation. The Dutch Sinemet CR Study Group. Clin Neurol Neurosurg. 1992;94(3):205–11.PubMed Wolters EC, Horstink MW, Roos RA, Jansen EN. Clinical efficacy of Sinemet CR 50/200 versus Sinemet 25/100 in patients with fluctuating Parkinson’s disease. An open, and a double-blind, double-dummy, multicenter treatment evaluation. The Dutch Sinemet CR Study Group. Clin Neurol Neurosurg. 1992;94(3):205–11.PubMed
171.
go back to reference Dupont E, Andersen A, Boas J, Boisen E, Borgmann R, Helgetveit AC, et al. Sustained-release Madopar HBS compared with standard Madopar in the long-term treatment of de novo parkinsonian patients. Acta Neurol Scand. 1996;93(1):14–20.PubMed Dupont E, Andersen A, Boas J, Boisen E, Borgmann R, Helgetveit AC, et al. Sustained-release Madopar HBS compared with standard Madopar in the long-term treatment of de novo parkinsonian patients. Acta Neurol Scand. 1996;93(1):14–20.PubMed
172.
go back to reference Block G, Liss C, Reines S, Irr J, Nibbelink D. Comparison of immediate-release and controlled release carbidopa/levodopa in Parkinson’s disease. A multicenter 5-year study. The CR First Study Group. Eur Neurol. 1997;37(1):23–7.PubMed Block G, Liss C, Reines S, Irr J, Nibbelink D. Comparison of immediate-release and controlled release carbidopa/levodopa in Parkinson’s disease. A multicenter 5-year study. The CR First Study Group. Eur Neurol. 1997;37(1):23–7.PubMed
173.
go back to reference Koller WC, Hutton JT, Tolosa E, Capilldeo R. Immediate-release and controlled-release carbidopa/levodopa in PD: a 5-year randomized multicenter study. Carbidopa/Levodopa Study Group. Neurology. 1999;53(5):1012–9.PubMed Koller WC, Hutton JT, Tolosa E, Capilldeo R. Immediate-release and controlled-release carbidopa/levodopa in PD: a 5-year randomized multicenter study. Carbidopa/Levodopa Study Group. Neurology. 1999;53(5):1012–9.PubMed
174.
go back to reference Stocchi F, Fabbri L, Vecsei L, Krygowska-Wajs A, Monici Preti PA, Ruggieri SA. Clinical efficacy of a single afternoon dose of effervescent levodopa-carbidopa preparation (CHF 1512) in fluctuating Parkinson disease. Clin Neuropharmacol. 2007;30(1):18–24. doi:10.1097/01.WNF.0000236762.77913.C6. Stocchi F, Fabbri L, Vecsei L, Krygowska-Wajs A, Monici Preti PA, Ruggieri SA. Clinical efficacy of a single afternoon dose of effervescent levodopa-carbidopa preparation (CHF 1512) in fluctuating Parkinson disease. Clin Neuropharmacol. 2007;30(1):18–24. doi:10.​1097/​01.​WNF.​0000236762.​77913.​C6.
175.
go back to reference Stocchi F, Zappia M, Dall’Armi V, Kulisevsky J, Lamberti P, Obeso JA. Melevodopa/carbidopa effervescent formulation in the treatment of motor fluctuations in advanced Parkinson’s disease. Mov Disord. 2010;25(12):1881–7. doi:10.1002/mds.23206.PubMed Stocchi F, Zappia M, Dall’Armi V, Kulisevsky J, Lamberti P, Obeso JA. Melevodopa/carbidopa effervescent formulation in the treatment of motor fluctuations in advanced Parkinson’s disease. Mov Disord. 2010;25(12):1881–7. doi:10.​1002/​mds.​23206.PubMed
176.
go back to reference Djaldetti R, Inzelberg R, Giladi N, Korczyn AD, Peretz-Aharon Y, Rabey MJ, et al. Oral solution of levodopa ethylester for treatment of response fluctuations in patients with advanced Parkinson’s disease. Mov Disord. 2002;17(2):297–302.PubMed Djaldetti R, Inzelberg R, Giladi N, Korczyn AD, Peretz-Aharon Y, Rabey MJ, et al. Oral solution of levodopa ethylester for treatment of response fluctuations in patients with advanced Parkinson’s disease. Mov Disord. 2002;17(2):297–302.PubMed
177.
go back to reference Metman LV, Hoff J, Mouradian MM, Chase TN. Fluctuations in plasma levodopa and motor responses with liquid and tablet levodopa/carbidopa. Mov Disord. 1994;9(4):463–5. doi:10.1002/mds.870090416.PubMed Metman LV, Hoff J, Mouradian MM, Chase TN. Fluctuations in plasma levodopa and motor responses with liquid and tablet levodopa/carbidopa. Mov Disord. 1994;9(4):463–5. doi:10.​1002/​mds.​870090416.PubMed
178.
go back to reference Kurth MC, Tetrud JW, Tanner CM, Irwin I, Stebbins GT, Goetz CG, et al. Double-blind, placebo-controlled, crossover study of duodenal infusion of levodopa/carbidopa in Parkinson’s disease patients with ‘on-off’ fluctuations. Neurology. 1993;43(9):1698–703.PubMed Kurth MC, Tetrud JW, Tanner CM, Irwin I, Stebbins GT, Goetz CG, et al. Double-blind, placebo-controlled, crossover study of duodenal infusion of levodopa/carbidopa in Parkinson’s disease patients with ‘on-off’ fluctuations. Neurology. 1993;43(9):1698–703.PubMed
180.
go back to reference Nilsson D, Nyholm D, Aquilonius SM. Duodenal levodopa infusion in Parkinson’s disease—long-term experience. Acta Neurol Scand. 2001;104(6):343–8.PubMed Nilsson D, Nyholm D, Aquilonius SM. Duodenal levodopa infusion in Parkinson’s disease—long-term experience. Acta Neurol Scand. 2001;104(6):343–8.PubMed
181.
go back to reference Eggert K, Schrader C, Hahn M, Stamelou M, Russmann A, Dengler R, et al. Continuous jejunal levodopa infusion in patients with advanced parkinson disease: practical aspects and outcome of motor and non-motor complications. Clin Neuropharmacol. 2008;31(3):151–66. doi:10.1097/wnf.0b013e31814b113e.PubMed Eggert K, Schrader C, Hahn M, Stamelou M, Russmann A, Dengler R, et al. Continuous jejunal levodopa infusion in patients with advanced parkinson disease: practical aspects and outcome of motor and non-motor complications. Clin Neuropharmacol. 2008;31(3):151–66. doi:10.​1097/​wnf.​0b013e31814b113e​.PubMed
182.
go back to reference 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. doi:10.1002/mds.10281.PubMed 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. doi:10.​1002/​mds.​10281.PubMed
183.
go back to reference Katzenschlager R, Hughes A, Evans A, Manson AJ, Hoffman M, Swinn L, et al. Continuous subcutaneous apomorphine therapy improves dyskinesias in Parkinson’s disease: a prospective study using single-dose challenges. Mov Disord. 2005;20(2):151–7. doi:10.1002/mds.20276.PubMed Katzenschlager R, Hughes A, Evans A, Manson AJ, Hoffman M, Swinn L, et al. Continuous subcutaneous apomorphine therapy improves dyskinesias in Parkinson’s disease: a prospective study using single-dose challenges. Mov Disord. 2005;20(2):151–7. doi:10.​1002/​mds.​20276.PubMed
184.
go back to reference Garcia Ruiz PJ, Sesar Ignacio A, Ares Pensado B, Castro Garcia A, Alonso Frech F, Alvarez Lopez M, 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(8):1130–6. doi:10.1002/mds.22063. Garcia Ruiz PJ, Sesar Ignacio A, Ares Pensado B, Castro Garcia A, Alonso Frech F, Alvarez Lopez M, 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(8):1130–6. doi:10.​1002/​mds.​22063.
185.
go back to reference Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. 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. doi:10.1056/NEJM200005183422004.PubMed Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. 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. doi:10.​1056/​NEJM200005183422​004.PubMed
186.
go back to reference Watts RL, Lyons KE, Pahwa R, Sethi K, Stern M, Hauser RA, 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. doi:10.1002/mds.22890.PubMed Watts RL, Lyons KE, Pahwa R, Sethi K, Stern M, Hauser RA, 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. doi:10.​1002/​mds.​22890.PubMed
187.
go back to reference Bracco F, Battaglia A, Chouza C, Dupont E, Gershanik O, Marti Masso JF, et al. The long-acting dopamine receptor agonist cabergoline in early Parkinson’s disease: final results of a 5-year, double-blind, levodopa-controlled study. CNS Drugs. 2004;18(11):733–46. Bracco F, Battaglia A, Chouza C, Dupont E, Gershanik O, Marti Masso JF, et al. The long-acting dopamine receptor agonist cabergoline in early Parkinson’s disease: final results of a 5-year, double-blind, levodopa-controlled study. CNS Drugs. 2004;18(11):733–46.
188.
go back to reference Fox SH, Katzenschlager R, Lim SY, Ravina B, Seppi K, Coelho M, et al. The movement disorder society evidence-based medicine review update: treatments for the motor symptoms of Parkinson’s disease. Mov Disord. 2011;26(Suppl 3):S2–41. doi:10.1002/mds.23829.PubMed Fox SH, Katzenschlager R, Lim SY, Ravina B, Seppi K, Coelho M, et al. The movement disorder society evidence-based medicine review update: treatments for the motor symptoms of Parkinson’s disease. Mov Disord. 2011;26(Suppl 3):S2–41. doi:10.​1002/​mds.​23829.PubMed
189.
go back to reference Stowe R, Ives N, Clarke CE, Handley K, Furmston A, Deane K, et al. Meta-analysis of the comparative efficacy and safety of adjuvant treatment to levodopa in later Parkinson’s disease. Mov Disord. 2011;26(4):587–98. doi:10.1002/mds.23517.PubMed Stowe R, Ives N, Clarke CE, Handley K, Furmston A, Deane K, et al. Meta-analysis of the comparative efficacy and safety of adjuvant treatment to levodopa in later Parkinson’s disease. Mov Disord. 2011;26(4):587–98. doi:10.​1002/​mds.​23517.PubMed
190.
go back to reference Tayarani-Binazir K, Jackson MJ, Rose S, McCreary AC, Jenner P. 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. doi:10.1016/j.expneurol.2010.09.007.PubMed Tayarani-Binazir K, Jackson MJ, Rose S, McCreary AC, Jenner P. 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. doi:10.​1016/​j.​expneurol.​2010.​09.​007.PubMed
191.
go back to reference Johnston LC, Jackson MJ, Rose S, McCreary AC, Jenner P. Pardoprunox reverses motor deficits but induces only mild dyskinesia in MPTP-treated common marmosets. Mov Disord. 2010;25(13):2059–66. doi:10.1002/mds.23249.PubMed Johnston LC, Jackson MJ, Rose S, McCreary AC, Jenner P. Pardoprunox reverses motor deficits but induces only mild dyskinesia in MPTP-treated common marmosets. Mov Disord. 2010;25(13):2059–66. doi:10.​1002/​mds.​23249.PubMed
192.
go back to reference Bronzova J, Sampaio C, Hauser RA, Lang AE, Rascol O, Theeuwes A, et al. Double-blind study of pardoprunox, a new partial dopamine agonist, in early Parkinson’s disease. Mov Disord. 2010;25(6):738–46. doi:10.1002/mds.22948.PubMed Bronzova J, Sampaio C, Hauser RA, Lang AE, Rascol O, Theeuwes A, et al. Double-blind study of pardoprunox, a new partial dopamine agonist, in early Parkinson’s disease. Mov Disord. 2010;25(6):738–46. doi:10.​1002/​mds.​22948.PubMed
193.
go back to reference Sampaio C, Bronzova J, Hauser RA, Lang AE, Rascol O, van de Witte SV, et al. Pardoprunox in early Parkinson’s disease: results from 2 large, randomized double-blind trials. Mov Disord. 2011;26(8):1464–76. doi:10.1002/mds.23590.PubMed Sampaio C, Bronzova J, Hauser RA, Lang AE, Rascol O, van de Witte SV, et al. Pardoprunox in early Parkinson’s disease: results from 2 large, randomized double-blind trials. Mov Disord. 2011;26(8):1464–76. doi:10.​1002/​mds.​23590.PubMed
194.
go back to reference Rascol O, Bronzova J, Hauser RA, Lang AE, Sampaio C, Theeuwes A, 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. doi:10.1016/j.parkreldis.2011.12.006.PubMed Rascol O, Bronzova J, Hauser RA, Lang AE, Sampaio C, Theeuwes A, 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. doi:10.​1016/​j.​parkreldis.​2011.​12.​006.PubMed
195.
go back to reference Stocchi F, Ruggieri S, Vacca L, Olanow CW. Prospective randomized trial of lisuride infusion versus oral levodopa in patients with Parkinson’s disease. Brain J Neurol. 2002;125(Pt 9):2058–66. Stocchi F, Ruggieri S, Vacca L, Olanow CW. Prospective randomized trial of lisuride infusion versus oral levodopa in patients with Parkinson’s disease. Brain J Neurol. 2002;125(Pt 9):2058–66.
196.
go back to reference 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):241–8. doi:10.1001/archneur.62.2.241. 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):241–8. doi:10.​1001/​archneur.​62.​2.​241.
197.
go back to reference Shoulson I, Oakes D, Fahn S, Lang A, Langston JW, LeWitt P, et al. Impact of sustained deprenyl (selegiline) in levodopa-treated Parkinson’s disease: a randomized placebo-controlled extension of the deprenyl and tocopherol antioxidative therapy of parkinsonism trial. Ann Neurol. 2002;51(5):604–12. doi:10.1002/ana.10191.PubMed Shoulson I, Oakes D, Fahn S, Lang A, Langston JW, LeWitt P, et al. Impact of sustained deprenyl (selegiline) in levodopa-treated Parkinson’s disease: a randomized placebo-controlled extension of the deprenyl and tocopherol antioxidative therapy of parkinsonism trial. Ann Neurol. 2002;51(5):604–12. doi:10.​1002/​ana.​10191.PubMed
200.
go back to reference Waters CH, Sethi KD, Hauser RA, Molho E, Bertoni JM. Zydis selegiline reduces off time in Parkinson’s disease patients with motor fluctuations: a 3-month, randomized, placebo-controlled study. Mov Disord. 2004;19(4):426–32. doi:10.1002/mds.20036.PubMed Waters CH, Sethi KD, Hauser RA, Molho E, Bertoni JM. Zydis selegiline reduces off time in Parkinson’s disease patients with motor fluctuations: a 3-month, randomized, placebo-controlled study. Mov Disord. 2004;19(4):426–32. doi:10.​1002/​mds.​20036.PubMed
201.
202.
203.
go back to reference Gregoire L, Morin N, Ouattara B, Gasparini F, Bilbe G, Johns D, et al. The acute antiparkinsonian and antidyskinetic effect of AFQ056, a novel metabotropic glutamate receptor type 5 antagonist, in l-dopa-treated parkinsonian monkeys. Parkinsonism Relat Disord. 2011;17(4):270–6. doi:10.1016/j.parkreldis.2011.01.008.PubMed Gregoire L, Morin N, Ouattara B, Gasparini F, Bilbe G, Johns D, et al. The acute antiparkinsonian and antidyskinetic effect of AFQ056, a novel metabotropic glutamate receptor type 5 antagonist, in l-dopa-treated parkinsonian monkeys. Parkinsonism Relat Disord. 2011;17(4):270–6. doi:10.​1016/​j.​parkreldis.​2011.​01.​008.PubMed
205.
go back to reference Stocchi F, Borgohain R, Onofrj M, Schapira AH, Bhatt M, Lucini V, et al. A randomized, double-blind, placebo-controlled trial of safinamide as add-on therapy in early Parkinson’s disease patients. Mov Disord. 2012;27(1):106–12. doi:10.1002/mds.23954.PubMed Stocchi F, Borgohain R, Onofrj M, Schapira AH, Bhatt M, Lucini V, et al. A randomized, double-blind, placebo-controlled trial of safinamide as add-on therapy in early Parkinson’s disease patients. Mov Disord. 2012;27(1):106–12. doi:10.​1002/​mds.​23954.PubMed
206.
207.
go back to reference Verhagen Metman L, Del Dotto P, van den Munckhof P, Fang J, Mouradian MM, Chase TN. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology. 1998;50(5):1323–6. Verhagen Metman L, Del Dotto P, van den Munckhof P, Fang J, Mouradian MM, Chase TN. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology. 1998;50(5):1323–6.
208.
go back to reference Del Dotto P, Pavese N, Gambaccini G, Bernardini S, Metman LV, Chase TN, et al. Intravenous amantadine improves levadopa-induced dyskinesias: an acute double-blind placebo-controlled study. Mov Disord. 2001;16(3):515–20.PubMed Del Dotto P, Pavese N, Gambaccini G, Bernardini S, Metman LV, Chase TN, et al. Intravenous amantadine improves levadopa-induced dyskinesias: an acute double-blind placebo-controlled study. Mov Disord. 2001;16(3):515–20.PubMed
209.
go back to reference da Silva-Junior FP, Braga-Neto P, Sueli Monte F, de Bruin VM. Amantadine reduces the duration of levodopa-induced dyskinesia: a randomized, double-blind, placebo-controlled study. Parkinsonism Relat Disord. 2005;11(7):449–52. doi:10.1016/j.parkreldis.2005.05.008. da Silva-Junior FP, Braga-Neto P, Sueli Monte F, de Bruin VM. Amantadine reduces the duration of levodopa-induced dyskinesia: a randomized, double-blind, placebo-controlled study. Parkinsonism Relat Disord. 2005;11(7):449–52. doi:10.​1016/​j.​parkreldis.​2005.​05.​008.
211.
go back to reference Thomas A, Iacono D, Luciano AL, Armellino K, Di Iorio A, Onofrj M. Duration of amantadine benefit on dyskinesia of severe Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2004;75(1):141–3.PubMedCentralPubMed Thomas A, Iacono D, Luciano AL, Armellino K, Di Iorio A, Onofrj M. Duration of amantadine benefit on dyskinesia of severe Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2004;75(1):141–3.PubMedCentralPubMed
212.
go back to reference Ory-Magne F, Corvol JC, Azulay JP, Bonnet AM, Brefel-Courbon C, Damier P, et al. Withdrawing amantadine in dyskinetic patients with Parkinson disease: the AMANDYSK trial. Neurology. 2014;82(4):300–7. doi:10.1212/WNL.0000000000000050.PubMed Ory-Magne F, Corvol JC, Azulay JP, Bonnet AM, Brefel-Courbon C, Damier P, et al. Withdrawing amantadine in dyskinetic patients with Parkinson disease: the AMANDYSK trial. Neurology. 2014;82(4):300–7. doi:10.​1212/​WNL.​0000000000000050​.PubMed
213.
go back to reference Hely MA, Morris JG, Reid WG, Trafficante R. Sydney Multicenter Study of Parkinson’s disease: non-l-dopa-responsive problems dominate at 15 years. Mov Disord. 2005;20(2):190–9. doi:10.1002/mds.20324.PubMed Hely MA, Morris JG, Reid WG, Trafficante R. Sydney Multicenter Study of Parkinson’s disease: non-l-dopa-responsive problems dominate at 15 years. Mov Disord. 2005;20(2):190–9. doi:10.​1002/​mds.​20324.PubMed
214.
go back to reference Rascol O, Brooks DJ, Melamed E, Oertel W, Poewe W, Stocchi F, et al. Rasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, lasting effect in adjunct therapy with rasagiline given once daily, study): a randomised, double-blind, parallel-group trial. Lancet. 2005;365(9463):947–54. doi:10.1016/S0140-6736(05)71083-7.PubMed Rascol O, Brooks DJ, Melamed E, Oertel W, Poewe W, Stocchi F, et al. Rasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, lasting effect in adjunct therapy with rasagiline given once daily, study): a randomised, double-blind, parallel-group trial. Lancet. 2005;365(9463):947–54. doi:10.​1016/​S0140-6736(05)71083-7.PubMed
215.
go back to reference Factor SA, Molho ES, Brown DL. Acute delirium after withdrawal of amantadine in Parkinson’s disease. Neurology. 1998;50(5):1456–8.PubMed Factor SA, Molho ES, Brown DL. Acute delirium after withdrawal of amantadine in Parkinson’s disease. Neurology. 1998;50(5):1456–8.PubMed
216.
217.
go back to reference Inzelberg R, Bonuccelli U, Schechtman E, Miniowich A, Strugatsky R, Ceravolo R, et al. Association between amantadine and the onset of dementia in Parkinson’s disease. Mov Disord. 2006;21(9):1375–9. doi:10.1002/mds.20968.PubMed Inzelberg R, Bonuccelli U, Schechtman E, Miniowich A, Strugatsky R, Ceravolo R, et al. Association between amantadine and the onset of dementia in Parkinson’s disease. Mov Disord. 2006;21(9):1375–9. doi:10.​1002/​mds.​20968.PubMed
218.
go back to reference Jahangirvand A, Rajput A. Early use of amantadine to prevent or delay onset of levodopa-induced dyskinesia in Parkinson’s disease. Mov Disord. 2013;28(Suppl 1):S207. Jahangirvand A, Rajput A. Early use of amantadine to prevent or delay onset of levodopa-induced dyskinesia in Parkinson’s disease. Mov Disord. 2013;28(Suppl 1):S207.
219.
go back to reference Pahwa RTC, Hauser RA. Randomized trial of extended release amantadine in Parkinson’s disease patients with l-dopa-induced dyskinesia (EASED study). Mov Disord. 2013;28(Suppl 1):S158. Pahwa RTC, Hauser RA. Randomized trial of extended release amantadine in Parkinson’s disease patients with l-dopa-induced dyskinesia (EASED study). Mov Disord. 2013;28(Suppl 1):S158.
220.
go back to reference Hanagasi HA, Kaptanoglu G, Sahin HA, Emre M. The use of NMDA antagonist memantine in drug-resistant dyskinesias resulting from l-dopa. Mov Disord. 2000;15(5):1016–7.PubMed Hanagasi HA, Kaptanoglu G, Sahin HA, Emre M. The use of NMDA antagonist memantine in drug-resistant dyskinesias resulting from l-dopa. Mov Disord. 2000;15(5):1016–7.PubMed
221.
go back to reference Varanese S, Howard J, Di Rocco A. NMDA antagonist memantine improves levodopa-induced dyskinesias and “on-off” phenomena in Parkinson’s disease. Mov Disord. 2010;25(4):508–10. doi:10.1002/mds.22917.PubMed Varanese S, Howard J, Di Rocco A. NMDA antagonist memantine improves levodopa-induced dyskinesias and “on-off” phenomena in Parkinson’s disease. Mov Disord. 2010;25(4):508–10. doi:10.​1002/​mds.​22917.PubMed
222.
go back to reference Moreau C, Delval A, Tiffreau V, Defebvre L, Dujardin K, Duhamel A, 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. doi:10.1136/jnnp-2012-303182.PubMedCentralPubMed Moreau C, Delval A, Tiffreau V, Defebvre L, Dujardin K, Duhamel A, 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. doi:10.​1136/​jnnp-2012-303182.PubMedCentralPubMed
223.
go back to reference Merello M, Nouzeilles MI, Cammarota A, Leiguarda R. Effect of memantine (NMDA antagonist) on Parkinson’s disease: a double-blind crossover randomized study. Clin Neuropharmacol. 1999;22(5):273–6.PubMed Merello M, Nouzeilles MI, Cammarota A, Leiguarda R. Effect of memantine (NMDA antagonist) on Parkinson’s disease: a double-blind crossover randomized study. Clin Neuropharmacol. 1999;22(5):273–6.PubMed
224.
go back to reference Emre M, Tsolaki M, Bonuccelli U, Destee A, Tolosa E, Kutzelnigg A, et al. Memantine for patients with Parkinson’s disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010;9(10):969–77. doi:10.1016/S1474-4422(10)70194-0.PubMed Emre M, Tsolaki M, Bonuccelli U, Destee A, Tolosa E, Kutzelnigg A, et al. Memantine for patients with Parkinson’s disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010;9(10):969–77. doi:10.​1016/​S1474-4422(10)70194-0.PubMed
225.
go back to reference Verhagen Metman L, Del Dotto P, Natte R, van den Munckhof P, Chase TN. Dextromethorphan improves levodopa-induced dyskinesias in Parkinson’s disease. Neurology. 1998;51(1):203–6. Verhagen Metman L, Del Dotto P, Natte R, van den Munckhof P, Chase TN. Dextromethorphan improves levodopa-induced dyskinesias in Parkinson’s disease. Neurology. 1998;51(1):203–6.
226.
go back to reference Braz CA, Borges V, Ferraz HB. Effect of riluzole on dyskinesia and duration of the on state in Parkinson disease patients: a double-blind, placebo-controlled pilot study. Clin Neuropharmacol. 2004;27(1):25–9.PubMed Braz CA, Borges V, Ferraz HB. Effect of riluzole on dyskinesia and duration of the on state in Parkinson disease patients: a double-blind, placebo-controlled pilot study. Clin Neuropharmacol. 2004;27(1):25–9.PubMed
227.
go back to reference Wessell RH, Ahmed SM, Menniti FS, Dunbar GL, Chase TN, Oh JD. NR2B selective NMDA receptor antagonist CP-101,606 prevents levodopa-induced motor response alterations in hemi-parkinsonian rats. Neuropharmacology. 2004;47(2):184–94. doi:10.1016/j.neuropharm.2004.03.011.PubMed Wessell RH, Ahmed SM, Menniti FS, Dunbar GL, Chase TN, Oh JD. NR2B selective NMDA receptor antagonist CP-101,606 prevents levodopa-induced motor response alterations in hemi-parkinsonian rats. Neuropharmacology. 2004;47(2):184–94. doi:10.​1016/​j.​neuropharm.​2004.​03.​011.PubMed
228.
go back to reference Morissette M, Dridi M, Calon F, Hadj Tahar A, Meltzer LT, Bedard PJ, et al. Prevention of levodopa-induced dyskinesias by a selective NR1A/2B N-methyl-d-aspartate receptor antagonist in parkinsonian monkeys: implication of preproenkephalin. Mov Disord. 2006;21(1):9–17. doi:10.1002/mds.20654. Morissette M, Dridi M, Calon F, Hadj Tahar A, Meltzer LT, Bedard PJ, et al. Prevention of levodopa-induced dyskinesias by a selective NR1A/2B N-methyl-d-aspartate receptor antagonist in parkinsonian monkeys: implication of preproenkephalin. Mov Disord. 2006;21(1):9–17. doi:10.​1002/​mds.​20654.
229.
go back to reference Tamim MK, Samadi P, Morissette M, Gregoire L, Ouattara B, Levesque D, et al. Effect of non-dopaminergic drug treatment on Levodopa induced dyskinesias in MPTP monkeys: common implication of striatal neuropeptides. Neuropharmacology. 2010;58(1):286–96. doi:10.1016/j.neuropharm.2009.06.030.PubMed Tamim MK, Samadi P, Morissette M, Gregoire L, Ouattara B, Levesque D, et al. Effect of non-dopaminergic drug treatment on Levodopa induced dyskinesias in MPTP monkeys: common implication of striatal neuropeptides. Neuropharmacology. 2010;58(1):286–96. doi:10.​1016/​j.​neuropharm.​2009.​06.​030.PubMed
230.
go back to reference Loftis JM, Janowsky A. The N-methyl-d-aspartate receptor subunit NR2B: localization, functional properties, regulation, and clinical implications. Pharmacol Ther. 2003;97(1):55–85.PubMed Loftis JM, Janowsky A. The N-methyl-d-aspartate receptor subunit NR2B: localization, functional properties, regulation, and clinical implications. Pharmacol Ther. 2003;97(1):55–85.PubMed
231.
go back to reference Nutt JG, Gunzler SA, Kirchhoff T, Hogarth P, Weaver JL, Krams M, et al. Effects of a NR2B selective NMDA glutamate antagonist, CP-101,606, on dyskinesia and parkinsonism. Mov Disord. 2008;23(13):1860–6. doi:10.1002/mds.22169.PubMedCentralPubMed Nutt JG, Gunzler SA, Kirchhoff T, Hogarth P, Weaver JL, Krams M, et al. Effects of a NR2B selective NMDA glutamate antagonist, CP-101,606, on dyskinesia and parkinsonism. Mov Disord. 2008;23(13):1860–6. doi:10.​1002/​mds.​22169.PubMedCentralPubMed
232.
go back to reference Duty S. Targeting glutamate receptors to tackle the pathogenesis, clinical symptoms and levodopa-induced dyskinesia associated with Parkinson’s disease. CNS Drugs. 2012;26(12):1017–32. doi:10.1007/s40263-012-0016-z.PubMed Duty S. Targeting glutamate receptors to tackle the pathogenesis, clinical symptoms and levodopa-induced dyskinesia associated with Parkinson’s disease. CNS Drugs. 2012;26(12):1017–32. doi:10.​1007/​s40263-012-0016-z.PubMed
234.
go back to reference Mela F, Marti M, Dekundy A, Danysz W, Morari M, Cenci MA. 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(2):483–97. doi:10.1111/j.1471-4159.2007.04456.x.PubMed Mela F, Marti M, Dekundy A, Danysz W, Morari M, Cenci MA. 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(2):483–97. doi:10.​1111/​j.​1471-4159.​2007.​04456.​x.PubMed
236.
go back to reference Johnston TH, Fox SH, McIldowie MJ, Piggott MJ, Brotchie JM. 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(3):865–73. doi:10.1124/jpet.110.166629.PubMed Johnston TH, Fox SH, McIldowie MJ, Piggott MJ, Brotchie JM. 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(3):865–73. doi:10.​1124/​jpet.​110.​166629.PubMed
237.
go back to reference Tison F DF, Christophe J. Safety, tolerability and anti-dyskinetic efficacy of dipraglurant, a novel mGluR5 negative allosteric modulator (NAM) in Parkinson’s disease (PD) patients with l-dopa-induced dyskinesia. Poster presented at: 65th American Academy of Neurology Annual Meeting, San Diego; 2013. Tison F DF, Christophe J. Safety, tolerability and anti-dyskinetic efficacy of dipraglurant, a novel mGluR5 negative allosteric modulator (NAM) in Parkinson’s disease (PD) patients with l-dopa-induced dyskinesia. Poster presented at: 65th American Academy of Neurology Annual Meeting, San Diego; 2013.
238.
go back to reference Konitsiotis S, Blanchet PJ, Verhagen L, Lamers E, Chase TN. AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys. Neurology. 2000;54(8):1589–95.PubMed Konitsiotis S, Blanchet PJ, Verhagen L, Lamers E, Chase TN. AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys. Neurology. 2000;54(8):1589–95.PubMed
239.
go back to reference Klockgether T, Turski L, Honore T, Zhang ZM, Gash DM, Kurlan R, et al. The AMPA receptor antagonist NBQX has antiparkinsonian effects in monoamine-depleted rats and MPTP-treated monkeys. Ann Neurol. 1991;30(5):717–23. doi:10.1002/ana.410300513.PubMed Klockgether T, Turski L, Honore T, Zhang ZM, Gash DM, Kurlan R, et al. The AMPA receptor antagonist NBQX has antiparkinsonian effects in monoamine-depleted rats and MPTP-treated monkeys. Ann Neurol. 1991;30(5):717–23. doi:10.​1002/​ana.​410300513.PubMed
240.
go back to reference Marin C, Jimenez A, Bonastre M, Vila M, Agid Y, Hirsch EC, et al. LY293558, an AMPA glutamate receptor antagonist, prevents and reverses levodopa-induced motor alterations in parkinsonian rats. Synapse. 2001;42(1):40–7. doi:10.1002/syn.1097.PubMed Marin C, Jimenez A, Bonastre M, Vila M, Agid Y, Hirsch EC, et al. LY293558, an AMPA glutamate receptor antagonist, prevents and reverses levodopa-induced motor alterations in parkinsonian rats. Synapse. 2001;42(1):40–7. doi:10.​1002/​syn.​1097.PubMed
241.
go back to reference Lees A, Fahn S, Eggert KM, Jankovic J, Lang A, Micheli F, et al. Perampanel, an AMPA antagonist, found to have no benefit in reducing “off” time in Parkinson’s disease. Mov Disord. 2012;27(2):284–8. doi:10.1002/mds.23983.PubMed Lees A, Fahn S, Eggert KM, Jankovic J, Lang A, Micheli F, et al. Perampanel, an AMPA antagonist, found to have no benefit in reducing “off” time in Parkinson’s disease. Mov Disord. 2012;27(2):284–8. doi:10.​1002/​mds.​23983.PubMed
242.
go back to reference Rascol O, Barone P, Behari M, Emre M, Giladi N, Olanow CW, et al. Perampanel in Parkinson disease fluctuations: a double-blind randomized trial with placebo and entacapone. Clin Neuropharmacol. 2012;35(1):15–20. doi:10.1097/WNF.0b013e318241520b.PubMed Rascol O, Barone P, Behari M, Emre M, Giladi N, Olanow CW, et al. Perampanel in Parkinson disease fluctuations: a double-blind randomized trial with placebo and entacapone. Clin Neuropharmacol. 2012;35(1):15–20. doi:10.​1097/​WNF.​0b013e318241520b​.PubMed
243.
go back to reference Calabresi P, Di Filippo M, Ghiglieri V, Tambasco N, Picconi B. Levodopa-induced dyskinesias in patients with Parkinson’s disease: filling the bench-to-bedside gap. Lancet Neurol. 2010;9(11):1106–17. doi:10.1016/S1474-4422(10)70218-0.PubMed Calabresi P, Di Filippo M, Ghiglieri V, Tambasco N, Picconi B. Levodopa-induced dyskinesias in patients with Parkinson’s disease: filling the bench-to-bedside gap. Lancet Neurol. 2010;9(11):1106–17. doi:10.​1016/​S1474-4422(10)70218-0.PubMed
244.
245.
go back to reference Price PA, Parkes JD, Marsden CD. Sodium valproate in the treatment of levodopa-induced dyskinesia. J Neurol Neurosurg Psychiatry. 1978;41(8):702–6.PubMedCentralPubMed Price PA, Parkes JD, Marsden CD. Sodium valproate in the treatment of levodopa-induced dyskinesia. J Neurol Neurosurg Psychiatry. 1978;41(8):702–6.PubMedCentralPubMed
246.
go back to reference Van Blercom N, Lasa A, Verger K, Masramon X, Sastre VM, Linazasoro G. Effects of gabapentin on the motor response to levodopa: a double-blind, placebo-controlled, crossover study in patients with complicated Parkinson disease. Clin Neuropharmacol. 2004;27(3):124–8.PubMed Van Blercom N, Lasa A, Verger K, Masramon X, Sastre VM, Linazasoro G. Effects of gabapentin on the motor response to levodopa: a double-blind, placebo-controlled, crossover study in patients with complicated Parkinson disease. Clin Neuropharmacol. 2004;27(3):124–8.PubMed
248.
249.
go back to reference Stathis P, Konitsiotis S, Tagaris G, Peterson D. Levetiracetam for the management of levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2011;26(2):264–70. doi:10.1002/mds.23355.PubMed Stathis P, Konitsiotis S, Tagaris G, Peterson D. Levetiracetam for the management of levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2011;26(2):264–70. doi:10.​1002/​mds.​23355.PubMed
250.
go back to reference Wong KK, Alty JE, Goy AG, Raghav S, Reutens DC, Kempster PA. A randomized, double-blind, placebo-controlled trial of levetiracetam for dyskinesia in Parkinson’s disease. Mov Disord. 2011;26(8):1552–5. doi:10.1002/mds.23687.PubMed Wong KK, Alty JE, Goy AG, Raghav S, Reutens DC, Kempster PA. A randomized, double-blind, placebo-controlled trial of levetiracetam for dyskinesia in Parkinson’s disease. Mov Disord. 2011;26(8):1552–5. doi:10.​1002/​mds.​23687.PubMed
251.
go back to reference Wolz M, Lohle M, Strecker K, Schwanebeck U, Schneider C, Reichmann H, et al. Levetiracetam for levodopa-induced dyskinesia in Parkinson’s disease: a randomized, double-blind, placebo-controlled trial. J Neural Transm. 2010;117(11):1279–86. doi:10.1007/s00702-010-0472-x.PubMed Wolz M, Lohle M, Strecker K, Schwanebeck U, Schneider C, Reichmann H, et al. Levetiracetam for levodopa-induced dyskinesia in Parkinson’s disease: a randomized, double-blind, placebo-controlled trial. J Neural Transm. 2010;117(11):1279–86. doi:10.​1007/​s00702-010-0472-x.PubMed
252.
253.
go back to reference Dupre KB, Ostock CY, Eskow Jaunarajs KL, Button T, Savage LM, Wolf W, et al. Local modulation of striatal glutamate efflux by serotonin 1A receptor stimulation in dyskinetic, hemiparkinsonian rats. Exp Neurol. 2011;229(2):288–99. doi:10.1016/j.expneurol.2011.02.012. Dupre KB, Ostock CY, Eskow Jaunarajs KL, Button T, Savage LM, Wolf W, et al. Local modulation of striatal glutamate efflux by serotonin 1A receptor stimulation in dyskinetic, hemiparkinsonian rats. Exp Neurol. 2011;229(2):288–99. doi:10.​1016/​j.​expneurol.​2011.​02.​012.
254.
go back to reference Olanow CW, Damier P, Goetz CG, Mueller T, Nutt J, Rascol O, et al. Multicenter, open-label, trial of sarizotan in Parkinson disease patients with levodopa-induced dyskinesias (the SPLENDID Study). Clin Neuropharmacol. 2004;27(2):58–62.PubMed Olanow CW, Damier P, Goetz CG, Mueller T, Nutt J, Rascol O, et al. Multicenter, open-label, trial of sarizotan in Parkinson disease patients with levodopa-induced dyskinesias (the SPLENDID Study). Clin Neuropharmacol. 2004;27(2):58–62.PubMed
255.
go back to reference Kannari K, Kurahashi K, Tomiyama M, Maeda T, Arai A, Baba M, et al. Tandospirone citrate, a selective 5-HT1A agonist, alleviates l-dopa-induced dyskinesia in patients with Parkinson’s disease. No To Shinkei. 2002;54(2):133–7.PubMed Kannari K, Kurahashi K, Tomiyama M, Maeda T, Arai A, Baba M, et al. Tandospirone citrate, a selective 5-HT1A agonist, alleviates l-dopa-induced dyskinesia in patients with Parkinson’s disease. No To Shinkei. 2002;54(2):133–7.PubMed
256.
go back to reference Rascol ODP, Goetz CG. A large phase III study to evaluate the safety and efficacy of sarizotan in the treatment of l-dopa-induced dyskinesia associated with Parkinson’s disease: the Paddy-1 study. Mov Disord. 2006;21(Suppl 15):S492–3. Rascol ODP, Goetz CG. A large phase III study to evaluate the safety and efficacy of sarizotan in the treatment of l-dopa-induced dyskinesia associated with Parkinson’s disease: the Paddy-1 study. Mov Disord. 2006;21(Suppl 15):S492–3.
257.
go back to reference Müller TOC, Nutt J. The PADDY-2 study: the evaluation of sarizotan for treatment-associated dyskinesia in Parkinson’s disease patients. Mov Disord. 2006;21(Suppl 15):S591. Müller TOC, Nutt J. The PADDY-2 study: the evaluation of sarizotan for treatment-associated dyskinesia in Parkinson’s disease patients. Mov Disord. 2006;21(Suppl 15):S591.
258.
go back to reference Goetz CG, Damier P, Hicking C, Laska E, Muller T, Olanow CW, 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. doi:10.1002/mds.21226.PubMed Goetz CG, Damier P, Hicking C, Laska E, Muller T, Olanow CW, 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. doi:10.​1002/​mds.​21226.PubMed
259.
go back to reference Tani Y, Ogata A, Koyama M, Inoue T. Effects of piclozotan (SUN N4057), a partial serotonin 1A receptor agonist, on motor complications induced by repeated administration of levodopa in parkinsonian rats. Eur J Pharmacol. 2010;649(1–3):218–23. doi:10.1016/j.ejphar.2010.09.013.PubMed Tani Y, Ogata A, Koyama M, Inoue T. Effects of piclozotan (SUN N4057), a partial serotonin 1A receptor agonist, on motor complications induced by repeated administration of levodopa in parkinsonian rats. Eur J Pharmacol. 2010;649(1–3):218–23. doi:10.​1016/​j.​ejphar.​2010.​09.​013.PubMed
260.
go back to reference Bezard E, Tronci E, Pioli EY, Li Q, Porras G, Bjorklund A, et al. Study of the antidyskinetic effect of eltoprazine in animal models of levodopa-induced dyskinesia. Mov Disord. 2013;28(8):1088–96. doi:10.1002/mds.25366.PubMed Bezard E, Tronci E, Pioli EY, Li Q, Porras G, Bjorklund A, et al. Study of the antidyskinetic effect of eltoprazine in animal models of levodopa-induced dyskinesia. Mov Disord. 2013;28(8):1088–96. doi:10.​1002/​mds.​25366.PubMed
261.
go back to reference Visanji NP, Gomez-Ramirez J, Johnston TH, Pires D, Voon V, Brotchie JM, et al. Pharmacological characterization of psychosis-like behavior in the MPTP-lesioned nonhuman primate model of Parkinson’s disease. Mov Disord. 2006;21(11):1879–91. doi:10.1002/mds.21073.PubMed Visanji NP, Gomez-Ramirez J, Johnston TH, Pires D, Voon V, Brotchie JM, et al. Pharmacological characterization of psychosis-like behavior in the MPTP-lesioned nonhuman primate model of Parkinson’s disease. Mov Disord. 2006;21(11):1879–91. doi:10.​1002/​mds.​21073.PubMed
262.
go back to reference Reddy S, Factor SA, Molho ES, Feustel PJ. The effect of quetiapine on psychosis and motor function in parkinsonian patients with and without dementia. Mov Disord. 2002;17(4):676–81. doi:10.1002/mds.10176.PubMed Reddy S, Factor SA, Molho ES, Feustel PJ. The effect of quetiapine on psychosis and motor function in parkinsonian patients with and without dementia. Mov Disord. 2002;17(4):676–81. doi:10.​1002/​mds.​10176.PubMed
263.
go back to reference Katzenschlager R, Manson AJ, Evans A, Watt H, Lees AJ. Low dose quetiapine for drug induced dyskinesias in Parkinson’s disease: a double blind cross over study. J Neurol Neurosurg Psychiatry. 2004;75(2):295–7.PubMedCentralPubMed Katzenschlager R, Manson AJ, Evans A, Watt H, Lees AJ. Low dose quetiapine for drug induced dyskinesias in Parkinson’s disease: a double blind cross over study. J Neurol Neurosurg Psychiatry. 2004;75(2):295–7.PubMedCentralPubMed
264.
go back to reference Durif F, Debilly B, Galitzky M, Morand D, Viallet F, Borg M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology. 2004;62(3):381–8.PubMed Durif F, Debilly B, Galitzky M, Morand D, Viallet F, Borg M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology. 2004;62(3):381–8.PubMed
265.
266.
go back to reference Johnston TH, Fox SH, Piggott MJ, Savola JM, Brotchie JM. The alpha(2) adrenergic antagonist fipamezole improves quality of levodopa action in Parkinsonian primates. Mov Disord. 2010;25(13):2084–93. doi:10.1002/mds.23172.PubMed Johnston TH, Fox SH, Piggott MJ, Savola JM, Brotchie JM. The alpha(2) adrenergic antagonist fipamezole improves quality of levodopa action in Parkinsonian primates. Mov Disord. 2010;25(13):2084–93. doi:10.​1002/​mds.​23172.PubMed
267.
go back to reference Savola JM, Hill M, Engstrom M, Merivuori H, Wurster S, McGuire SG, et al. Fipamezole (JP-1730) is a potent alpha2 adrenergic receptor antagonist that reduces levodopa-induced dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Mov Disord. 2003;18(8):872–83. doi:10.1002/mds.10464.PubMed Savola JM, Hill M, Engstrom M, Merivuori H, Wurster S, McGuire SG, et al. Fipamezole (JP-1730) is a potent alpha2 adrenergic receptor antagonist that reduces levodopa-induced dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Mov Disord. 2003;18(8):872–83. doi:10.​1002/​mds.​10464.PubMed
268.
go back to reference Henry B, Fox SH, Peggs D, Crossman AR, Brotchie JM. The alpha2-adrenergic receptor antagonist idazoxan reduces dyskinesia and enhances anti-parkinsonian actions of l-dopa in the MPTP-lesioned primate model of Parkinson’s disease. Mov Disord. 1999;14(5):744–53.PubMed Henry B, Fox SH, Peggs D, Crossman AR, Brotchie JM. The alpha2-adrenergic receptor antagonist idazoxan reduces dyskinesia and enhances anti-parkinsonian actions of l-dopa in the MPTP-lesioned primate model of Parkinson’s disease. Mov Disord. 1999;14(5):744–53.PubMed
269.
go back to reference Manson AJ, Iakovidou E, Lees AJ. Idazoxan is ineffective for levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2000;15(2):336–7.PubMed Manson AJ, Iakovidou E, Lees AJ. Idazoxan is ineffective for levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2000;15(2):336–7.PubMed
270.
go back to reference Rascol O, Arnulf I, Peyro-Saint Paul H, Brefel-Courbon C, Vidailhet M, Thalamas C, 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. Rascol O, Arnulf I, Peyro-Saint Paul H, Brefel-Courbon C, Vidailhet M, Thalamas C, 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.
271.
272.
go back to reference Henry B, Fox SH, Crossman AR, Brotchie JM. Mu- and delta-opioid receptor antagonists reduce levodopa-induced dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Exp Neurol. 2001;171(1):139–46. doi:10.1006/exnr.2001.7727.PubMed Henry B, Fox SH, Crossman AR, Brotchie JM. Mu- and delta-opioid receptor antagonists reduce levodopa-induced dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Exp Neurol. 2001;171(1):139–46. doi:10.​1006/​exnr.​2001.​7727.PubMed
273.
go back to reference Rascol O, Fabre N, Blin O, Poulik J, Sabatini U, Senard JM, et al. Naltrexone, an opiate antagonist, fails to modify motor symptoms in patients with Parkinson’s disease. Mov Disord. 1994;9(4):437–40. doi:10.1002/mds.870090410.PubMed Rascol O, Fabre N, Blin O, Poulik J, Sabatini U, Senard JM, et al. Naltrexone, an opiate antagonist, fails to modify motor symptoms in patients with Parkinson’s disease. Mov Disord. 1994;9(4):437–40. doi:10.​1002/​mds.​870090410.PubMed
274.
go back to reference Manson AJ, Katzenschlager R, Hobart J, Lees AJ. High dose naltrexone for dyskinesias induced by levodopa. J Neurol Neurosurg Psychiatry. 2001;70(4):554–6.PubMedCentralPubMed Manson AJ, Katzenschlager R, Hobart J, Lees AJ. High dose naltrexone for dyskinesias induced by levodopa. J Neurol Neurosurg Psychiatry. 2001;70(4):554–6.PubMedCentralPubMed
275.
go back to reference Fox S, Silverdale M, Kellett M, Davies R, Steiger M, Fletcher N, et al. Non-subtype-selective opioid receptor antagonism in treatment of levodopa-induced motor complications in Parkinson’s disease. Mov Disord. 2004;19(5):554–60. doi:10.1002/mds.10693.PubMed Fox S, Silverdale M, Kellett M, Davies R, Steiger M, Fletcher N, et al. Non-subtype-selective opioid receptor antagonism in treatment of levodopa-induced motor complications in Parkinson’s disease. Mov Disord. 2004;19(5):554–60. doi:10.​1002/​mds.​10693.PubMed
276.
go back to reference Di Marzo V, Hill MP, Bisogno T, Crossman AR, Brotchie JM. Enhanced levels of endogenous cannabinoids in the globus pallidus are associated with a reduction in movement in an animal model of Parkinson’s disease. FASEB J. 2000;14(10):1432–8.PubMed Di Marzo V, Hill MP, Bisogno T, Crossman AR, Brotchie JM. Enhanced levels of endogenous cannabinoids in the globus pallidus are associated with a reduction in movement in an animal model of Parkinson’s disease. FASEB J. 2000;14(10):1432–8.PubMed
277.
go back to reference Fox SH, Henry B, Hill M, Crossman A, Brotchie J. Stimulation of cannabinoid receptors reduces levodopa-induced dyskinesia in the MPTP-lesioned nonhuman primate model of Parkinson’s disease. Mov Disord. 2002;17(6):1180–7. doi:10.1002/mds.10289.PubMed Fox SH, Henry B, Hill M, Crossman A, Brotchie J. Stimulation of cannabinoid receptors reduces levodopa-induced dyskinesia in the MPTP-lesioned nonhuman primate model of Parkinson’s disease. Mov Disord. 2002;17(6):1180–7. doi:10.​1002/​mds.​10289.PubMed
278.
go back to reference Venderova K, Ruzicka E, Vorisek V, Visnovsky P. Survey on cannabis use in Parkinson’s disease: subjective improvement of motor symptoms. Mov Disord. 2004;19(9):1102–6. doi:10.1002/mds.20111.PubMed Venderova K, Ruzicka E, Vorisek V, Visnovsky P. Survey on cannabis use in Parkinson’s disease: subjective improvement of motor symptoms. Mov Disord. 2004;19(9):1102–6. doi:10.​1002/​mds.​20111.PubMed
279.
go back to reference Sieradzan KA, Fox SH, Hill M, Dick JP, Crossman AR, Brotchie JM. Cannabinoids reduce levodopa-induced dyskinesia in Parkinson’s disease: a pilot study. Neurology. 2001;57(11):2108–11.PubMed Sieradzan KA, Fox SH, Hill M, Dick JP, Crossman AR, Brotchie JM. Cannabinoids reduce levodopa-induced dyskinesia in Parkinson’s disease: a pilot study. Neurology. 2001;57(11):2108–11.PubMed
280.
go back to reference Carroll CB, Bain PG, Teare L, Liu X, Joint C, Wroath C, et al. Cannabis for dyskinesia in Parkinson disease: a randomized double-blind crossover study. Neurology. 2004;63(7):1245–50.PubMed Carroll CB, Bain PG, Teare L, Liu X, Joint C, Wroath C, et al. Cannabis for dyskinesia in Parkinson disease: a randomized double-blind crossover study. Neurology. 2004;63(7):1245–50.PubMed
282.
284.
go back to reference Mori A, Shindou T. Modulation of GABAergic transmission in the striatopallidal system by adenosine A2A receptors: a potential mechanism for the antiparkinsonian effects of A2A antagonists. Neurology. 2003;61(11 Suppl 6):S44–8.PubMed Mori A, Shindou T. Modulation of GABAergic transmission in the striatopallidal system by adenosine A2A receptors: a potential mechanism for the antiparkinsonian effects of A2A antagonists. Neurology. 2003;61(11 Suppl 6):S44–8.PubMed
285.
go back to reference Carta AR, Pinna A, Cauli O, Morelli M. Differential regulation of GAD67, enkephalin and dynorphin mRNAs by chronic-intermittent l-dopa and A2A receptor blockade plus l-dopa in dopamine-denervated rats. Synapse. 2002;44(3):166–74. doi:10.1002/syn.10066.PubMed Carta AR, Pinna A, Cauli O, Morelli M. Differential regulation of GAD67, enkephalin and dynorphin mRNAs by chronic-intermittent l-dopa and A2A receptor blockade plus l-dopa in dopamine-denervated rats. Synapse. 2002;44(3):166–74. doi:10.​1002/​syn.​10066.PubMed
287.
288.
go back to reference Hauser RA, Cantillon M, Pourcher E, Micheli F, Mok V, Onofrj M, 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. doi:10.1016/S1474-4422(11)70012-6.PubMed Hauser RA, Cantillon M, Pourcher E, Micheli F, Mok V, Onofrj M, 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. doi:10.​1016/​S1474-4422(11)70012-6.PubMed
289.
go back to reference Factor SA, Wolski K, Togasaki DM, Huyck S, Cantillon M, Ho TW, et al. Long-term safety and efficacy of preladenant in subjects with fluctuating Parkinson’s disease. Mov Disord. 2013;28(6):817–20. doi:10.1002/mds.25395.PubMed Factor SA, Wolski K, Togasaki DM, Huyck S, Cantillon M, Ho TW, et al. Long-term safety and efficacy of preladenant in subjects with fluctuating Parkinson’s disease. Mov Disord. 2013;28(6):817–20. doi:10.​1002/​mds.​25395.PubMed
290.
go back to reference Hauser RA, Olanow C, Kieburtz K, Neale A, Resburg C, Maya U, Bandaket S. A phase 2, placebo-controlled, randomized, double-blind trial of tozadenant (SYN-115) in patients with Parkinson’s disease with wearing-off fluctuations on l-dopa. Mov Disord. 2013;28(Suppl 1):S158. Hauser RA, Olanow C, Kieburtz K, Neale A, Resburg C, Maya U, Bandaket S. A phase 2, placebo-controlled, randomized, double-blind trial of tozadenant (SYN-115) in patients with Parkinson’s disease with wearing-off fluctuations on l-dopa. Mov Disord. 2013;28(Suppl 1):S158.
291.
go back to reference Bara-Jimenez W, Dimitrova TD, Sherzai A, Aksu M, Chase TN. Glutamate release inhibition ineffective in levodopa-induced motor complications. Mov Disord. 2006;21(9):1380–3. doi:10.1002/mds.20976.PubMed Bara-Jimenez W, Dimitrova TD, Sherzai A, Aksu M, Chase TN. Glutamate release inhibition ineffective in levodopa-induced motor complications. Mov Disord. 2006;21(9):1380–3. doi:10.​1002/​mds.​20976.PubMed
292.
go back to reference Zhou FM, Liang Y, Dani JA. Endogenous nicotinic cholinergic activity regulates dopamine release in the striatum. Nat Neurosci. 2001;4(12):1224–9. doi:10.1038/nn769.PubMed Zhou FM, Liang Y, Dani JA. Endogenous nicotinic cholinergic activity regulates dopamine release in the striatum. Nat Neurosci. 2001;4(12):1224–9. doi:10.​1038/​nn769.PubMed
293.
go back to reference Zhang D, Bordia T, McGregor M, McIntosh JM, Decker MW, Quik M. ABT-089 and ABT-894 reduce levodopa-induced dyskinesias in a monkey model of Parkinson’s disease. Mov Disord. 2014;29(4):508–17. doi:10.1002/mds.25817.PubMed Zhang D, Bordia T, McGregor M, McIntosh JM, Decker MW, Quik M. ABT-089 and ABT-894 reduce levodopa-induced dyskinesias in a monkey model of Parkinson’s disease. Mov Disord. 2014;29(4):508–17. doi:10.​1002/​mds.​25817.PubMed
294.
go back to reference Xie CL, Pan JL, Zhang SF, Gan J, Liu ZG. Effect of nicotine on l-dopa-induced dyskinesia in animal models of Parkinson’s disease: a systematic review and meta-analysis. Neurol Sci. 2014;35(5):653–62. doi:10.1007/s10072-014-1652-5.PubMed Xie CL, Pan JL, Zhang SF, Gan J, Liu ZG. Effect of nicotine on l-dopa-induced dyskinesia in animal models of Parkinson’s disease: a systematic review and meta-analysis. Neurol Sci. 2014;35(5):653–62. doi:10.​1007/​s10072-014-1652-5.PubMed
297.
go back to reference Mango D, Bonito-Oliva A, Ledonne A, Cappellacci L, Petrelli R, Nistico R, et al. Adenosine A1 receptor stimulation reduces D1 receptor-mediated GABAergic transmission from striato-nigral terminals and attenuates l-dopa-induced dyskinesia in dopamine-denervated mice. Exp Neurol. 2014;. doi:10.1016/j.expneurol.2014.08.022.PubMed Mango D, Bonito-Oliva A, Ledonne A, Cappellacci L, Petrelli R, Nistico R, et al. Adenosine A1 receptor stimulation reduces D1 receptor-mediated GABAergic transmission from striato-nigral terminals and attenuates l-dopa-induced dyskinesia in dopamine-denervated mice. Exp Neurol. 2014;. doi:10.​1016/​j.​expneurol.​2014.​08.​022.PubMed
298.
go back to reference Park HY, Kang YM, Kang Y, Park TS, Ryu YK, Hwang JH, et al. Inhibition of adenylyl cyclase type 5 prevents l-dopa-induced dyskinesia in an animal model of Parkinson’s disease. J Neurosci. 2014;34(35):11744–53. doi:10.1523/JNEUROSCI.0864-14.2014.PubMed Park HY, Kang YM, Kang Y, Park TS, Ryu YK, Hwang JH, et al. Inhibition of adenylyl cyclase type 5 prevents l-dopa-induced dyskinesia in an animal model of Parkinson’s disease. J Neurosci. 2014;34(35):11744–53. doi:10.​1523/​JNEUROSCI.​0864-14.​2014.PubMed
300.
go back to reference Foltynie T, Cheeran B, Williams-Gray CH, Edwards MJ, Schneider SA, Weinberger D, et al. BDNF val66met influences time to onset of levodopa induced dyskinesia in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2009;80(2):141–4. doi:10.1136/jnnp.2008.154294.PubMed Foltynie T, Cheeran B, Williams-Gray CH, Edwards MJ, Schneider SA, Weinberger D, et al. BDNF val66met influences time to onset of levodopa induced dyskinesia in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2009;80(2):141–4. doi:10.​1136/​jnnp.​2008.​154294.PubMed
301.
go back to reference Zappia M, Annesi G, Nicoletti G, Arabia G, Annesi F, Messina D, 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. doi:10.1001/archneur.62.4.601.PubMed Zappia M, Annesi G, Nicoletti G, Arabia G, Annesi F, Messina D, 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. doi:10.​1001/​archneur.​62.​4.​601.PubMed
302.
go back to reference Goetz CG, Stebbins GT, Chung KA, Hauser RA, Miyasaki JM, Nicholas AP, et al. Which dyskinesia scale best detects treatment response? Mov Disord. 2013;28(3):341–6. doi:10.1002/mds.25321.PubMed Goetz CG, Stebbins GT, Chung KA, Hauser RA, Miyasaki JM, Nicholas AP, et al. Which dyskinesia scale best detects treatment response? Mov Disord. 2013;28(3):341–6. doi:10.​1002/​mds.​25321.PubMed
303.
go back to reference Pietracupa S, Fasano A, Fabbrini G, Sarchioto M, Bloise M, Latorre A, et al. Poor self-awareness of levodopa-induced dyskinesias in Parkinson’s disease: clinical features and mechanisms. Parkinsonism Relat Disord. 2013;19(11):1004–8. doi:10.1016/j.parkreldis.2013.07.002.PubMed Pietracupa S, Fasano A, Fabbrini G, Sarchioto M, Bloise M, Latorre A, et al. Poor self-awareness of levodopa-induced dyskinesias in Parkinson’s disease: clinical features and mechanisms. Parkinsonism Relat Disord. 2013;19(11):1004–8. doi:10.​1016/​j.​parkreldis.​2013.​07.​002.PubMed
304.
go back to reference Manson AJ, Brown P, O’Sullivan JD, Asselman P, Buckwell D, Lees AJ. An ambulatory dyskinesia monitor. J Neurol Neurosurg Psychiatry. 2000;68(2):196–201.PubMedCentralPubMed Manson AJ, Brown P, O’Sullivan JD, Asselman P, Buckwell D, Lees AJ. An ambulatory dyskinesia monitor. J Neurol Neurosurg Psychiatry. 2000;68(2):196–201.PubMedCentralPubMed
305.
go back to reference Maetzler W, Domingos J, Srulijes K, Ferreira JJ, Bloem BR. Quantitative wearable sensors for objective assessment of Parkinson’s disease. Mov Disord. 2013;28(12):1628–37. doi:10.1002/mds.25628.PubMed Maetzler W, Domingos J, Srulijes K, Ferreira JJ, Bloem BR. Quantitative wearable sensors for objective assessment of Parkinson’s disease. Mov Disord. 2013;28(12):1628–37. doi:10.​1002/​mds.​25628.PubMed
306.
go back to reference No authors listed. A comparison of Madopar CR and standard Madopar in the treatment of nocturnal and early-morning disability in Parkinson’s disease. The UK Madopar CR Study Group. Clin Neuropharmacol. 1989;12(6):498–505. No authors listed. A comparison of Madopar CR and standard Madopar in the treatment of nocturnal and early-morning disability in Parkinson’s disease. The UK Madopar CR Study Group. Clin Neuropharmacol. 1989;12(6):498–505.
307.
go back to reference Moller JC, Oertel WH, Koster J, Pezzoli G, Provinciali L. 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. doi:10.1002/mds.20397.PubMed Moller JC, Oertel WH, Koster J, Pezzoli G, Provinciali L. 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. doi:10.​1002/​mds.​20397.PubMed
308.
go back to reference Poewe WH, Rascol O, Quinn N, Tolosa E, Oertel WH, Martignoni E, 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. doi:10.1016/S1474-4422(07)70108-4.PubMed Poewe WH, Rascol O, Quinn N, Tolosa E, Oertel WH, Martignoni E, 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. doi:10.​1016/​S1474-4422(07)70108-4.PubMed
309.
go back to reference Mizuno Y, Abe T, Hasegawa K, Kuno S, Kondo T, Yamamoto M, 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. doi:10.1002/mds.21313.PubMed Mizuno Y, Abe T, Hasegawa K, Kuno S, Kondo T, Yamamoto M, 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. doi:10.​1002/​mds.​21313.PubMed
310.
go back to reference 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. doi:10.1002/mds.21191.PubMed 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. doi:10.​1002/​mds.​21191.PubMed
313.
go back to reference Borgohain R, Szasz J, Stanzione P, Meshram C, Bhatt MH, Chirilineau D, et al. Two-year, randomized, controlled study of safinamide as add-on to levodopa in mid to late Parkinson’s disease. Mov Disord. 2014;. doi:10.1002/mds.25961. Borgohain R, Szasz J, Stanzione P, Meshram C, Bhatt MH, Chirilineau D, et al. Two-year, randomized, controlled study of safinamide as add-on to levodopa in mid to late Parkinson’s disease. Mov Disord. 2014;. doi:10.​1002/​mds.​25961.
314.
go back to reference Borgohain R, Szasz J, Stanzione P, Meshram C, Bhatt M, Chirilineau D, et al. Randomized trial of safinamide add-on to levodopa in Parkinson’s disease with motor fluctuations. Mov Disord. 2014;29(2):229–37. doi:10.1002/mds.25751.PubMed Borgohain R, Szasz J, Stanzione P, Meshram C, Bhatt M, Chirilineau D, et al. Randomized trial of safinamide add-on to levodopa in Parkinson’s disease with motor fluctuations. Mov Disord. 2014;29(2):229–37. doi:10.​1002/​mds.​25751.PubMed
315.
go back to reference Poewe WH, Deuschl G, Gordin A, Kultalahti ER, Leinonen M. Efficacy and safety of entacapone in Parkinson’s disease patients with suboptimal levodopa response: a 6-month randomized placebo-controlled double-blind study in Germany and Austria (Celomen Study). Acta Neurol Scand. 2002;105(4):245–55.PubMed Poewe WH, Deuschl G, Gordin A, Kultalahti ER, Leinonen M. Efficacy and safety of entacapone in Parkinson’s disease patients with suboptimal levodopa response: a 6-month randomized placebo-controlled double-blind study in Germany and Austria (Celomen Study). Acta Neurol Scand. 2002;105(4):245–55.PubMed
316.
go back to reference Fenelon G, Gimenez-Roldan S, Montastruc JL, Bermejo F, Durif F, Bourdeix I, et al. Efficacy and tolerability of entacapone in patients with Parkinson’s disease treated with levodopa plus a dopamine agonist and experiencing wearing-off motor fluctuations. A randomized, double-blind, multicentre study. J Neural Transm. 2003;110(3):239–51. doi:10.1007/s00702-002-0799-z.PubMed Fenelon G, Gimenez-Roldan S, Montastruc JL, Bermejo F, Durif F, Bourdeix I, et al. Efficacy and tolerability of entacapone in patients with Parkinson’s disease treated with levodopa plus a dopamine agonist and experiencing wearing-off motor fluctuations. A randomized, double-blind, multicentre study. J Neural Transm. 2003;110(3):239–51. doi:10.​1007/​s00702-002-0799-z.PubMed
317.
go back to reference Brooks DJ, Sagar H. Entacapone is beneficial in both fluctuating and non-fluctuating patients with Parkinson’s disease: a randomised, placebo controlled, double blind, six month study. J Neurol Neurosurg Psychiatry. 2003;74(8):1071–9.PubMedCentralPubMed Brooks DJ, Sagar H. Entacapone is beneficial in both fluctuating and non-fluctuating patients with Parkinson’s disease: a randomised, placebo controlled, double blind, six month study. J Neurol Neurosurg Psychiatry. 2003;74(8):1071–9.PubMedCentralPubMed
318.
go back to reference Reichmann H, Boas J, Macmahon D, Myllyla V, Hakala A, Reinikainen K. Efficacy of combining levodopa with entacapone on quality of life and activities of daily living in patients experiencing wearing-off type fluctuations. Acta Neurol Scand. 2005;111(1):21–8. doi:10.1111/j.1600-0404.2004.00363.x.PubMed Reichmann H, Boas J, Macmahon D, Myllyla V, Hakala A, Reinikainen K. Efficacy of combining levodopa with entacapone on quality of life and activities of daily living in patients experiencing wearing-off type fluctuations. Acta Neurol Scand. 2005;111(1):21–8. doi:10.​1111/​j.​1600-0404.​2004.​00363.​x.PubMed
319.
go back to reference Mizuno Y, Kanazawa I, Kuno S, Yanagisawa N, Yamamoto M, Kondo T. Placebo-controlled, double-blind dose-finding study of entacapone in fluctuating parkinsonian patients. Mov Disord. 2007;22(1):75–80. doi:10.1002/mds.21218.PubMed Mizuno Y, Kanazawa I, Kuno S, Yanagisawa N, Yamamoto M, Kondo T. Placebo-controlled, double-blind dose-finding study of entacapone in fluctuating parkinsonian patients. Mov Disord. 2007;22(1):75–80. doi:10.​1002/​mds.​21218.PubMed
320.
go back to reference Baas H, Beiske AG, Ghika J, Jackson M, Oertel WH, Poewe W, et al. Catechol-O-methyltransferase inhibition with tolcapone reduces the “wearing off” phenomenon and levodopa requirements in fluctuating parkinsonian patients. J Neurol Neurosurg Psychiatry. 1997;63(4):421–8.PubMedCentralPubMed Baas H, Beiske AG, Ghika J, Jackson M, Oertel WH, Poewe W, et al. Catechol-O-methyltransferase inhibition with tolcapone reduces the “wearing off” phenomenon and levodopa requirements in fluctuating parkinsonian patients. J Neurol Neurosurg Psychiatry. 1997;63(4):421–8.PubMedCentralPubMed
321.
go back to reference Waters CH, Kurth M, Bailey P, Shulman LM, LeWitt P, Dorflinger E, et al. Tolcapone in stable Parkinson’s disease: efficacy and safety of long-term treatment. The Tolcapone Stable Study Group. Neurology. 1997;49(3):665–71.PubMed Waters CH, Kurth M, Bailey P, Shulman LM, LeWitt P, Dorflinger E, et al. Tolcapone in stable Parkinson’s disease: efficacy and safety of long-term treatment. The Tolcapone Stable Study Group. Neurology. 1997;49(3):665–71.PubMed
322.
go back to reference Rajput AH, Martin W, Saint-Hilaire MH, Dorflinger E, Pedder S. Tolcapone improves motor function in parkinsonian patients with the “wearing-off” phenomenon: a double-blind, placebo-controlled, multicenter trial. Neurology. 1997;49(4):1066–71.PubMed Rajput AH, Martin W, Saint-Hilaire MH, Dorflinger E, Pedder S. Tolcapone improves motor function in parkinsonian patients with the “wearing-off” phenomenon: a double-blind, placebo-controlled, multicenter trial. Neurology. 1997;49(4):1066–71.PubMed
323.
go back to reference Parkinson Study Group. Evaluation of dyskinesias in a pilot, randomized, placebo-controlled trial of remacemide in advanced Parkinson disease. Arch Neurol. 2001;58(10):1660–8. Parkinson Study Group. Evaluation of dyskinesias in a pilot, randomized, placebo-controlled trial of remacemide in advanced Parkinson disease. Arch Neurol. 2001;58(10):1660–8.
324.
go back to reference Eggert K, Squillacote D, Barone P, Dodel R, Katzenschlager R, Emre M, et al. Safety and efficacy of perampanel in advanced Parkinson’s disease: a randomized, placebo-controlled study. Mov Disord. 2010;25(7):896–905. doi:10.1002/mds.22974.PubMed Eggert K, Squillacote D, Barone P, Dodel R, Katzenschlager R, Emre M, et al. Safety and efficacy of perampanel in advanced Parkinson’s disease: a randomized, placebo-controlled study. Mov Disord. 2010;25(7):896–905. doi:10.​1002/​mds.​22974.PubMed
326.
go back to reference Dimitrova TDB-JW, Savola J-M. Alpha-2 adrenergic antagonist effects in Parkinson’s disease. Mov Disord. 2009;19(Suppl 9):S222. Dimitrova TDB-JW, Savola J-M. Alpha-2 adrenergic antagonist effects in Parkinson’s disease. Mov Disord. 2009;19(Suppl 9):S222.
327.
go back to reference Bara-Jimenez W, Sherzai A, Dimitrova T, Favit A, Bibbiani F, Gillespie M, et al. Adenosine A(2A) receptor antagonist treatment of Parkinson’s disease. Neurology. 2003;61(3):293–6.PubMed Bara-Jimenez W, Sherzai A, Dimitrova T, Favit A, Bibbiani F, Gillespie M, et al. Adenosine A(2A) receptor antagonist treatment of Parkinson’s disease. Neurology. 2003;61(3):293–6.PubMed
328.
go back to reference LeWitt PA. “Off” time reduction from adjunctive use of istradefylline (KW-6002) in levodopa-treated patients with advanced Parkinson’s disease. Mov Disord. 2004;19(Suppl 9):S222. LeWitt PA. “Off” time reduction from adjunctive use of istradefylline (KW-6002) in levodopa-treated patients with advanced Parkinson’s disease. Mov Disord. 2004;19(Suppl 9):S222.
330.
go back to reference Hauser RA, Shulman LM, Trugman JM, Roberts JW, Mori A, Ballerini R, et al. Study of istradefylline in patients with Parkinson’s disease on levodopa with motor fluctuations. Mov Disord. 2008;23(15):2177–85. doi:10.1002/mds.22095.PubMed Hauser RA, Shulman LM, Trugman JM, Roberts JW, Mori A, Ballerini R, et al. Study of istradefylline in patients with Parkinson’s disease on levodopa with motor fluctuations. Mov Disord. 2008;23(15):2177–85. doi:10.​1002/​mds.​22095.PubMed
331.
go back to reference LeWitt PA, Guttman M, Tetrud JW, Tuite PJ, Mori A, Chaikin P, 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. doi:10.1002/ana.21315.PubMed LeWitt PA, Guttman M, Tetrud JW, Tuite PJ, Mori A, Chaikin P, 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. doi:10.​1002/​ana.​21315.PubMed
333.
go back to reference Pourcher E, Fernandez HH, Stacy M, Mori A, Ballerini R, Chaikin P. 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. doi:10.1016/j.parkreldis.2011.09.023.PubMed Pourcher E, Fernandez HH, Stacy M, Mori A, Ballerini R, Chaikin P. 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. doi:10.​1016/​j.​parkreldis.​2011.​09.​023.PubMed
334.
go back to reference Hauser RA, Olanow CW, Kieburtz KD, Pourcher E, Docu-Axelerad A, Lew M et al. Tozadenant (SYN115) in patients with Parkinson’s disease who have motor fluctuations on levodopa: a phase 2b, double-blind, randomised trial. Lancet Neurol. 2014;13(8):767–76. doi:10.1016/S1474-4422(14)70148-6. Hauser RA, Olanow CW, Kieburtz KD, Pourcher E, Docu-Axelerad A, Lew M et al. Tozadenant (SYN115) in patients with Parkinson’s disease who have motor fluctuations on levodopa: a phase 2b, double-blind, randomised trial. Lancet Neurol. 2014;13(8):767–76. doi:10.​1016/​S1474-4422(14)70148-6.
Metadata
Title
Pharmacological Strategies for the Management of Levodopa-Induced Dyskinesia in Patients with Parkinson’s Disease
Authors
Eva Schaeffer
Andrea Pilotto
Daniela Berg
Publication date
01-12-2014
Publisher
Springer International Publishing
Published in
CNS Drugs / Issue 12/2014
Print ISSN: 1172-7047
Electronic ISSN: 1179-1934
DOI
https://doi.org/10.1007/s40263-014-0205-z

Other articles of this Issue 12/2014

CNS Drugs 12/2014 Go to the issue