Skip to main content
Erschienen in: Drugs 13/2013

01.09.2013 | Review Article

Non-dopaminergic Treatments for Motor Control in Parkinson’s Disease

verfasst von: Susan H. Fox

Erschienen in: Drugs | Ausgabe 13/2013

Einloggen, um Zugang zu erhalten

Abstract

The pathological processes underlying Parkinson’s disease (PD) involve more than dopamine cell loss within the midbrain. These non-dopaminergic neurotransmitters include noradrenergic, serotonergic, glutamatergic, and cholinergic systems within cortical, brainstem and basal ganglia regions. Several non-dopaminergic treatments are now in clinical use to treat motor symptoms of PD, or are being evaluated as potential therapies. Agents for symptomatic monotherapy and as adjunct to dopaminergic therapies for motor symptoms include adenosine A2A antagonists and the mixed monoamine-B inhibitor (MAO-BI) and glutamate release agent safinamide. The largest area of potential use for non-dopaminergic drugs is as add-on therapy for motor fluctuations. Thus adenosine A2A antagonists, safinamide, and the antiepileptic agent zonisamide can extend the duration of action of levodopa. To reduce levodopa-induced dyskinesia, drugs that target overactive glutamatergic neurotransmission can be used, and include the non-selective N-methyl d-aspartate antagonist amantadine. More recently, selective metabotropic glutamate receptor (mGluR5) antagonists are being evaluated in phase II randomized controlled trials. Serotonergic agents acting as 5-HT2A/2C antagonists, such as the atypical antipsychotic clozapine, may also reduce dyskinesia. 5-HT1A agonists theoretically can reduce dyskinesia, but in practice, may also worsen PD motor symptoms, and so clinical applicability has not yet been shown. Noradrenergic α2A antagonism using fipamezole can potentially reduce dyskinesia. Several non-dopaminergic agents have also been investigated to reduce non-levodopa-responsive motor symptoms such as gait and tremor. Thus the cholinesterase inhibitor donepezil showed mild benefit in gait, while the predominantly noradrenergic re-uptake inhibitor methylphenidate had conflicting results in advanced PD subjects. Tremor in PD may respond to muscarinic M4 cholinergic antagonists (anticholinergics), but tolerability is often poor. Alternatives include β-adrenergic antagonists such as propranolol. Other options include 5-HT2A antagonists, and drugs that have mixed binding properties involving serotonin and acetylcholine, such as clozapine and the antidepressant mirtazapine, can be effective in reducing PD tremor. Many other non-dopaminergic agents are in preclinical and phase I/II early stages of study, and the reader is directed to recent reviews. While levodopa remains the most effective agent to treat motor symptoms in PD, the overall approach to using non-dopaminergic drugs in PD is to reduce reliance on levodopa and to target non-levodopa-responsive symptoms.
Literatur
1.
Zurück zum Zitat Halliday GM, Li YW, Blumbergs PC, et al. Neuropathology of immunohistochemically identified brainstem neurons in Parkinson’s disease. Ann Neurol. 1990;27:373–85.PubMedCrossRef Halliday GM, Li YW, Blumbergs PC, et al. Neuropathology of immunohistochemically identified brainstem neurons in Parkinson’s disease. Ann Neurol. 1990;27:373–85.PubMedCrossRef
2.
Zurück zum Zitat Zarow C, Lyness SA, Mortimer JA, et al. Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases. Arch Neurol. 2003;60:337–41.PubMedCrossRef Zarow C, Lyness SA, Mortimer JA, et al. Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases. Arch Neurol. 2003;60:337–41.PubMedCrossRef
3.
Zurück zum Zitat Braak H, Del Tredici K, Rüb U, et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.PubMedCrossRef Braak H, Del Tredici K, Rüb U, et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.PubMedCrossRef
4.
Zurück zum Zitat Halliday GM, Blumbergs PC, Cotton RG, et al. Loss of brainstem serotonin- and substance P-containing neurons in Parkinson’s disease. Brain Res. 1990;510(1):104–7.PubMedCrossRef Halliday GM, Blumbergs PC, Cotton RG, et al. Loss of brainstem serotonin- and substance P-containing neurons in Parkinson’s disease. Brain Res. 1990;510(1):104–7.PubMedCrossRef
5.
Zurück zum Zitat Paulus W, Jellinger K. The neuropathologic basis of different clinical subgroups of Parkinson’s disease. J Neuropathol Exp Neurol. 1991;50:743–55.PubMedCrossRef Paulus W, Jellinger K. The neuropathologic basis of different clinical subgroups of Parkinson’s disease. J Neuropathol Exp Neurol. 1991;50:743–55.PubMedCrossRef
6.
Zurück zum Zitat Kish SJ, Tong J, Hornykiewicz O, et al. Preferential loss of serotonin markers in caudate versus putamen in Parkinson’s disease. Brain. 2008;131:120–31.PubMed Kish SJ, Tong J, Hornykiewicz O, et al. Preferential loss of serotonin markers in caudate versus putamen in Parkinson’s disease. Brain. 2008;131:120–31.PubMed
7.
Zurück zum Zitat Lang AE, Obeso JA. Time to move beyond nigrostriatal dopamine deficiency in Parkinson’s disease. Ann Neurol. 2004;55:761–5.PubMedCrossRef Lang AE, Obeso JA. Time to move beyond nigrostriatal dopamine deficiency in Parkinson’s disease. Ann Neurol. 2004;55:761–5.PubMedCrossRef
8.
Zurück zum Zitat Huot P, Johnston TH, Koprich JB, et al. The pharmacology of l-DOPA-induced dyskinesia in Parkinson’s disease. Pharmacol Rev. 2013;65:1–52.CrossRef Huot P, Johnston TH, Koprich JB, et al. The pharmacology of l-DOPA-induced dyskinesia in Parkinson’s disease. Pharmacol Rev. 2013;65:1–52.CrossRef
9.
Zurück zum Zitat Bezard E, Brotchie JM, Gross CE. Pathophysiology of levodopa-induced dyskinesia: potential for new therapies. Nat Rev Neurosci. 2001;2:577–88.PubMedCrossRef Bezard E, Brotchie JM, Gross CE. Pathophysiology of levodopa-induced dyskinesia: potential for new therapies. Nat Rev Neurosci. 2001;2:577–88.PubMedCrossRef
10.
Zurück zum Zitat Brotchie JM. Nondopaminergic mechanisms in levodopa-induced dyskinesia. Mov Disord. 2005;20:919–31.PubMedCrossRef Brotchie JM. Nondopaminergic mechanisms in levodopa-induced dyskinesia. Mov Disord. 2005;20:919–31.PubMedCrossRef
11.
Zurück zum Zitat Huot P, Fox SH. Nondopaminergic treatments for Parkinson’s disease. Neurodegener Dis Manag. 2011;6:491–512.CrossRef Huot P, Fox SH. Nondopaminergic treatments for Parkinson’s disease. Neurodegener Dis Manag. 2011;6:491–512.CrossRef
12.
Zurück zum Zitat Kalia L, Brotchie JM, Fox SH. Non dopaminergic therapies for PD motor symptoms; update on recent clinical trials. Mov Disord. 2013;28(2):131–44.PubMedCrossRef Kalia L, Brotchie JM, Fox SH. Non dopaminergic therapies for PD motor symptoms; update on recent clinical trials. Mov Disord. 2013;28(2):131–44.PubMedCrossRef
13.
Zurück zum Zitat Gomez-Mancilla B, Bedard PJ. Effect of nondopaminergic drugs on l-dopa-induced dyskinesias in MPTP-treated monkeys. Clin Neuropharmacol. 1993;16:418–27.PubMedCrossRef Gomez-Mancilla B, Bedard PJ. Effect of nondopaminergic drugs on l-dopa-induced dyskinesias in MPTP-treated monkeys. Clin Neuropharmacol. 1993;16:418–27.PubMedCrossRef
14.
Zurück zum Zitat Fox SH, Brotchie JM, Lang AE. Non-dopaminergic treatments in development for Parkinson’s disease. Lancet Neurol. 2008;7:927–38.PubMedCrossRef Fox SH, Brotchie JM, Lang AE. Non-dopaminergic treatments in development for Parkinson’s disease. Lancet Neurol. 2008;7:927–38.PubMedCrossRef
15.
Zurück zum Zitat Schwarzschild MA, Agnati L, Fuxe K, et al. Targeting adenosine A2A receptors in Parkinson’s disease. Trends Neurosci. 2006;29:647–54.PubMedCrossRef Schwarzschild MA, Agnati L, Fuxe K, et al. Targeting adenosine A2A receptors in Parkinson’s disease. Trends Neurosci. 2006;29:647–54.PubMedCrossRef
16.
Zurück zum Zitat Nash JE, Brotchie JM. A common signaling pathway for striatal NMDA and adenosine A2a receptors: implications for the treatment of Parkinson’s disease. J Neurosci. 2000;20:7782–9.PubMed Nash JE, Brotchie JM. A common signaling pathway for striatal NMDA and adenosine A2a receptors: implications for the treatment of Parkinson’s disease. J Neurosci. 2000;20:7782–9.PubMed
17.
Zurück zum Zitat Kanda T, Jackson MJ, Smith LA, et al. Adenosine A2A antagonist: a novel antiparkinsonian agent that does not provoke dyskinesia in parkinsonian monkeys. Ann Neurol. 1998;43:507–13.PubMedCrossRef Kanda T, Jackson MJ, Smith LA, et al. Adenosine A2A antagonist: a novel antiparkinsonian agent that does not provoke dyskinesia in parkinsonian monkeys. Ann Neurol. 1998;43:507–13.PubMedCrossRef
18.
Zurück zum Zitat Fernandez HH, Greeley DR, Zweig RM, et al. Istradefylline as monotherapy for Parkinson disease: results of the 6002-US-051 trial. Parkinsonism Relat Disord. 2010;16:16–20.PubMedCrossRef Fernandez HH, Greeley DR, Zweig RM, et al. Istradefylline as monotherapy for Parkinson disease: results of the 6002-US-051 trial. Parkinsonism Relat Disord. 2010;16:16–20.PubMedCrossRef
19.
Zurück zum Zitat Merck. A phase 3, double-blind, double-dummy, placebo- and active-controlled dose-range-finding efficacy and safety study of preladenant in subjects with early Parkinson’s disease. ClinicalTrials.gov (Internet). Bethesda: National Library of Medicine (US); 2000. http://clinicaltrials.gov/show/NCT01155479. Cited 13 May 2013. Merck. A phase 3, double-blind, double-dummy, placebo- and active-controlled dose-range-finding efficacy and safety study of preladenant in subjects with early Parkinson’s disease. ClinicalTrials.gov (Internet). Bethesda: National Library of Medicine (US); 2000. http://​clinicaltrials.​gov/​show/​NCT01155479. Cited 13 May 2013.
20.
Zurück zum Zitat Stocchi F, Borgohain R, Onofrj M, 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.PubMedCrossRef Stocchi F, Borgohain R, Onofrj M, 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.PubMedCrossRef
21.
Zurück zum Zitat Barone P, Fernandez H, Ferreira J, et al. Safinamide as an add-on therapy to a stable dose of a single dopamine agonist: results from a randomized, placebo-controlled, 24-week multicenter trial in early idiopathic Parkinson disease (PD) patients (MOTION Study). In: Abstract 65th Annual American Academy of Neurology poster 1.061; 2013. Barone P, Fernandez H, Ferreira J, et al. Safinamide as an add-on therapy to a stable dose of a single dopamine agonist: results from a randomized, placebo-controlled, 24-week multicenter trial in early idiopathic Parkinson disease (PD) patients (MOTION Study). In: Abstract 65th Annual American Academy of Neurology poster 1.061; 2013.
22.
Zurück zum Zitat Schapira AH, Fox S, Hauser R, et al., on behalf of the SETTLE Investigators. Safinamide add on to l-dopa: a randomized, placebo-controlled, 24-week global trial in patients with Parkinson’s disease (PD) and motor fluctuations (SETTLE). In: Abstract 65th Annual American Academy of Neurology poster 1.062; 2013. Schapira AH, Fox S, Hauser R, et al., on behalf of the SETTLE Investigators. Safinamide add on to l-dopa: a randomized, placebo-controlled, 24-week global trial in patients with Parkinson’s disease (PD) and motor fluctuations (SETTLE). In: Abstract 65th Annual American Academy of Neurology poster 1.062; 2013.
23.
Zurück zum Zitat Murata M, Hasegawa K, Kanazawa I, Japan Zonisamide on PD Study Group. Zonisamide improves motor function in Parkinson disease: a randomized, double-blind study. Neurology. 2007;68(1):45–50. Murata M, Hasegawa K, Kanazawa I, Japan Zonisamide on PD Study Group. Zonisamide improves motor function in Parkinson disease: a randomized, double-blind study. Neurology. 2007;68(1):45–50.
24.
Zurück zum Zitat Bara-Jimenez W, Sherzai A, Dimitrova T, et al. Adenosine A(2A) receptor antagonist treatment of Parkinson’s disease. Neurology. 2003;61(3):293–6.PubMedCrossRef Bara-Jimenez W, Sherzai A, Dimitrova T, et al. Adenosine A(2A) receptor antagonist treatment of Parkinson’s disease. Neurology. 2003;61(3):293–6.PubMedCrossRef
25.
Zurück zum Zitat Hauser RA, Hubble JP, Truong DD. Randomized trial of the adenosine A(2A) receptor antagonist istradefylline in advanced PD. Neurology. 2003;61(3):297–303.PubMedCrossRef Hauser RA, Hubble JP, Truong DD. Randomized trial of the adenosine A(2A) receptor antagonist istradefylline in advanced PD. Neurology. 2003;61(3):297–303.PubMedCrossRef
26.
Zurück zum Zitat Stacy M, Silver D, Mendis T, et al. A 12-week, placebo-controlled study (6002-US-006) of istradefylline in Parkinson disease. Neurology. 2008;70(23):2233–40.PubMedCrossRef Stacy M, Silver D, Mendis T, et al. A 12-week, placebo-controlled study (6002-US-006) of istradefylline in Parkinson disease. Neurology. 2008;70(23):2233–40.PubMedCrossRef
27.
Zurück zum Zitat LeWitt PA, Guttman M, Tetrud JW, et al. Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces “off” time in Parkinson’s disease: a double-blind, randomized, multicenter clinical trial (6002-US-005). Ann Neurol. 2008;63(3):295–302.PubMedCrossRef LeWitt PA, Guttman M, Tetrud JW, et al. Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces “off” time in Parkinson’s disease: a double-blind, randomized, multicenter clinical trial (6002-US-005). Ann Neurol. 2008;63(3):295–302.PubMedCrossRef
28.
Zurück zum Zitat Mizuno Y, Hasegawa K, Kondo T, et al. Clinical efficacy of istradefylline (KW-6002) in Parkinson’s disease: a randomized, controlled study. Mov Disord. 2010;25(10):1437–43.PubMedCrossRef Mizuno Y, Hasegawa K, Kondo T, et al. Clinical efficacy of istradefylline (KW-6002) in Parkinson’s disease: a randomized, controlled study. Mov Disord. 2010;25(10):1437–43.PubMedCrossRef
29.
Zurück zum Zitat Hauser RA, Shulman LM, Trugman JM, et al. Study of istradefylline in patients with Parkinson’s disease on levodopa with motor fluctuations. Mov Disord. 2008;23(15):2177–85.PubMedCrossRef Hauser RA, Shulman LM, Trugman JM, et al. Study of istradefylline in patients with Parkinson’s disease on levodopa with motor fluctuations. Mov Disord. 2008;23(15):2177–85.PubMedCrossRef
30.
Zurück zum Zitat Mizuno Y, Kondo T, the Japanese Istradefylline Study Group. Adenosine A2A receptor antagonist istradefylline reduces daily off time in Parkinson’s disease. Mov Disord. 2013. doi:10.1002/mds.25418. Mizuno Y, Kondo T, the Japanese Istradefylline Study Group. Adenosine A2A receptor antagonist istradefylline reduces daily off time in Parkinson’s disease. Mov Disord. 2013. doi:10.​1002/​mds.​25418.
31.
Zurück zum Zitat Hauser RA, Cantillon M, Pourcher E, et al. Preladenant in patients with Parkinson’s disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol. 2011;10(3):221–9.PubMedCrossRef Hauser RA, Cantillon M, Pourcher E, et al. Preladenant in patients with Parkinson’s disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol. 2011;10(3):221–9.PubMedCrossRef
32.
Zurück zum Zitat Merck. A phase 3, 12-week, double-blind, double-dummy, placebo- and active-controlled efficacy and safety study of preladenant in subjects with moderate to severe Parkinson’s disease. ClinicalTrials.gov (Internet). Bethesda: National Library of Medicine (US); 2000. http://clinicaltrials.gov/show/NCT01155466. Cited 13 May 2013. Merck. A phase 3, 12-week, double-blind, double-dummy, placebo- and active-controlled efficacy and safety study of preladenant in subjects with moderate to severe Parkinson’s disease. ClinicalTrials.gov (Internet). Bethesda: National Library of Medicine (US); 2000. http://​clinicaltrials.​gov/​show/​NCT01155466. Cited 13 May 2013.
33.
Zurück zum Zitat Biotie. A double-blind, randomized, placebo-controlled study of the safety and efficacy of SYN115 as adjunctive therapy in levodopa-treated Parkinson’s subjects with end of dose wearing off. ClinicalTrials.gov (Internet). Bethesda: National Library of Medicine (US); 2000. http://clinicaltrials.gov/show/NCT01283594. Cited 23 May 2013. Biotie. A double-blind, randomized, placebo-controlled study of the safety and efficacy of SYN115 as adjunctive therapy in levodopa-treated Parkinson’s subjects with end of dose wearing off. ClinicalTrials.gov (Internet). Bethesda: National Library of Medicine (US); 2000. http://​clinicaltrials.​gov/​show/​NCT01283594. Cited 23 May 2013.
34.
Zurück zum Zitat Fox SH, Lang AE, Brotchie JM. Translation of non-dopaminergic treatments for levodopa-induced dyskinesia from MPTP-lesioned nonhuman primates to phase IIa clinical studies: keys to success and roads to failure. Mov Disord. 2006;21:1578–94.PubMedCrossRef Fox SH, Lang AE, Brotchie JM. Translation of non-dopaminergic treatments for levodopa-induced dyskinesia from MPTP-lesioned nonhuman primates to phase IIa clinical studies: keys to success and roads to failure. Mov Disord. 2006;21:1578–94.PubMedCrossRef
35.
Zurück zum Zitat Calabresi P, Giacomini P, Centonze D, et al. Levodopa-induced dyskinesia: a pathological form of striatal synaptic plasticity? Ann Neurol. 2000;47:60–8. Calabresi P, Giacomini P, Centonze D, et al. Levodopa-induced dyskinesia: a pathological form of striatal synaptic plasticity? Ann Neurol. 2000;47:60–8.
36.
Zurück zum Zitat Conn PJ, Battaglia G, Marino MJ, et al. Metabotropic glutamate receptors in the basal ganglia motor circuit. Nat Rev Neurosci. 2005;6(10):787–98.PubMedCrossRef Conn PJ, Battaglia G, Marino MJ, et al. Metabotropic glutamate receptors in the basal ganglia motor circuit. Nat Rev Neurosci. 2005;6(10):787–98.PubMedCrossRef
37.
Zurück zum Zitat 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.PubMedCrossRef 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.PubMedCrossRef
38.
Zurück zum Zitat Fox SH, Katzenschlager R, Lim SY, et al. 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.PubMedCrossRef Fox SH, Katzenschlager R, Lim SY, et al. 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.PubMedCrossRef
39.
Zurück zum Zitat Wolf E, Seppi K, Katzenschlager R, et al. Long-term antidyskinetic efficacy of amantadine in Parkinson’s disease. Mov Disord. 2010;25:1357–63.PubMedCrossRef Wolf E, Seppi K, Katzenschlager R, et al. Long-term antidyskinetic efficacy of amantadine in Parkinson’s disease. Mov Disord. 2010;25:1357–63.PubMedCrossRef
40.
Zurück zum Zitat Adamas Pharmaceuticals, Inc. Extended Release Amantadine Safety and Efficacy Study in Levodopa-Induced Dyskinesia (EASED Study). ClinicalTrials.gov (Internet). Bethesda: National Library of Medicine (US); 2000. http://clinicaltrials.gov/show/NCT01397422. Cited 9 May 2013. Adamas Pharmaceuticals, Inc. Extended Release Amantadine Safety and Efficacy Study in Levodopa-Induced Dyskinesia (EASED Study). ClinicalTrials.gov (Internet). Bethesda: National Library of Medicine (US); 2000. http://​clinicaltrials.​gov/​show/​NCT01397422. Cited 9 May 2013.
41.
Zurück zum Zitat Parkinson Study Group. Evaluation of dyskinesias in a pilot, randomized, placebo-controlled trial of remacemide in advanced Parkinson disease. Arch Neurol. 2001;58:1660–8.CrossRef Parkinson Study Group. Evaluation of dyskinesias in a pilot, randomized, placebo-controlled trial of remacemide in advanced Parkinson disease. Arch Neurol. 2001;58:1660–8.CrossRef
42.
Zurück zum Zitat Lees A, Fahn S, Eggert KM, 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.PubMedCrossRef Lees A, Fahn S, Eggert KM, 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.PubMedCrossRef
43.
Zurück zum Zitat Johnston TH, Brotchie JM. Drugs in development for Parkinson’s disease. Curr Opin Investig Drugs. 2004;5(7):720–6.PubMed Johnston TH, Brotchie JM. Drugs in development for Parkinson’s disease. Curr Opin Investig Drugs. 2004;5(7):720–6.PubMed
44.
Zurück zum Zitat Rascol O, Fox SH, Gasparini F, et al. Use of metabotropic glutamate 5 receptor antagonists for treatment of levodopa-induced dyskinesias. Mov Disord (in press). Rascol O, Fox SH, Gasparini F, et al. Use of metabotropic glutamate 5 receptor antagonists for treatment of levodopa-induced dyskinesias. Mov Disord (in press).
45.
Zurück zum Zitat Berg D, Godau J, Trenkwalder C, et al. AFQ056 treatment of levodopa-induced dyskinesias: results of 2 randomized controlled trials. Mov Disord. 2011;26(7):1243–50.PubMedCrossRef Berg D, Godau J, Trenkwalder C, et al. AFQ056 treatment of levodopa-induced dyskinesias: results of 2 randomized controlled trials. Mov Disord. 2011;26(7):1243–50.PubMedCrossRef
46.
Zurück zum Zitat Tison F, Durif F, Ferrand C, et al. Safety, tolerability and anti-dyskinetic efficacy of dipraglurant, a novel mGluR5 negative allosteric modulator (NAM) in Parkinson’s disease (PD) patients with levodopa-induced dyskinesia (LID). In: Abstract 65th Annual American Academy of Neurology poster 23.004; Mar 2013. Tison F, Durif F, Ferrand C, et al. Safety, tolerability and anti-dyskinetic efficacy of dipraglurant, a novel mGluR5 negative allosteric modulator (NAM) in Parkinson’s disease (PD) patients with levodopa-induced dyskinesia (LID). In: Abstract 65th Annual American Academy of Neurology poster 23.004; Mar 2013.
47.
Zurück zum Zitat Mavridis M, Degryse AD, Lategan AJ, et al. Effects of locus coeruleus lesions on parkinsonian signs, striatal dopamine and substantia nigra cell loss after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in monkeys: a possible role for the locus coeruleus in the progression of Parkinson’s disease. Neuroscience. 1991;41(2–3):507–23.PubMedCrossRef Mavridis M, Degryse AD, Lategan AJ, et al. Effects of locus coeruleus lesions on parkinsonian signs, striatal dopamine and substantia nigra cell loss after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in monkeys: a possible role for the locus coeruleus in the progression of Parkinson’s disease. Neuroscience. 1991;41(2–3):507–23.PubMedCrossRef
48.
Zurück zum Zitat 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.PubMedCrossRef 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.PubMedCrossRef
49.
Zurück zum Zitat Henry B, Fox SH, Peggs D, et al. 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;14:744–53.PubMedCrossRef Henry B, Fox SH, Peggs D, et al. 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;14:744–53.PubMedCrossRef
50.
Zurück zum Zitat Barnum CJ, Bhide N, Lindenbach D, et al. Effects of noradrenergic denervation on l-DOPA-induced dyskinesia and its treatment by α- and β-adrenergic receptor antagonists in hemiparkinsonian rats. Pharmacol Biochem Behav. 2012;100(3):607–15.PubMedCentralPubMedCrossRef Barnum CJ, Bhide N, Lindenbach D, et al. Effects of noradrenergic denervation on l-DOPA-induced dyskinesia and its treatment by α- and β-adrenergic receptor antagonists in hemiparkinsonian rats. Pharmacol Biochem Behav. 2012;100(3):607–15.PubMedCentralPubMedCrossRef
51.
Zurück zum Zitat Lewitt PA, Hauser RA, Lu M, et al. Randomized clinical trial of fipamezole for dyskinesia in Parkinson disease (FJORD study). Neurology. 2012;79(2):163–9.PubMedCrossRef Lewitt PA, Hauser RA, Lu M, et al. Randomized clinical trial of fipamezole for dyskinesia in Parkinson disease (FJORD study). Neurology. 2012;79(2):163–9.PubMedCrossRef
52.
Zurück zum Zitat Carta M, Carlsson T, Kirik D, Bjorklund A. Dopamine released from 5-HT terminals is the cause of l-DOPA-induced dyskinesia in parkinsonian rats. Brain. 2007;130:1819–33.PubMedCrossRef Carta M, Carlsson T, Kirik D, Bjorklund A. Dopamine released from 5-HT terminals is the cause of l-DOPA-induced dyskinesia in parkinsonian rats. Brain. 2007;130:1819–33.PubMedCrossRef
53.
Zurück zum Zitat Munoz A, Li Q, Gardoni F, et al. Combined 5-HT1A and 5-HT1B receptor agonists for the treatment of l-DOPA-induced dyskinesia. Brain. 2008;131:3380–94.PubMedCrossRef Munoz A, Li Q, Gardoni F, et al. Combined 5-HT1A and 5-HT1B receptor agonists for the treatment of l-DOPA-induced dyskinesia. Brain. 2008;131:3380–94.PubMedCrossRef
54.
55.
Zurück zum Zitat Goetz CG, Damier P, Hicking C, et al. Sarizotan as a treatment for dyskinesias in Parkinson’s disease: a double-blind placebo-controlled trial. Mov Disord. 2007;22:179–86.PubMedCrossRef Goetz CG, Damier P, Hicking C, et al. Sarizotan as a treatment for dyskinesias in Parkinson’s disease: a double-blind placebo-controlled trial. Mov Disord. 2007;22:179–86.PubMedCrossRef
57.
Zurück zum Zitat Sage JJ, Hauser RA, Cordon ME, Gonzalez MA, Tani Y, Koyamam M, Apfel SC, Reed RF, Okamato M, Gertbner JM. Pilot study of the efficacy and safety of piclozotan in Parkinson’s disease patients with l-dopa induced motor complications. Mov Disord. 2009;24(Suppl 1):S277. Sage JJ, Hauser RA, Cordon ME, Gonzalez MA, Tani Y, Koyamam M, Apfel SC, Reed RF, Okamato M, Gertbner JM. Pilot study of the efficacy and safety of piclozotan in Parkinson’s disease patients with l-dopa induced motor complications. Mov Disord. 2009;24(Suppl 1):S277.
58.
Zurück zum Zitat Rascol O, Bronzova J, Hauser RA, et al. Pardoprunox as adjunct therapy to levodopa in patients with Parkinson’s disease experiencing motor fluctuations: results of a double-blind, randomized, placebo-controlled, trial. Parkinsonism Relat Disord. 2012;18(4):370–6.PubMedCrossRef Rascol O, Bronzova J, Hauser RA, et al. Pardoprunox as adjunct therapy to levodopa in patients with Parkinson’s disease experiencing motor fluctuations: results of a double-blind, randomized, placebo-controlled, trial. Parkinsonism Relat Disord. 2012;18(4):370–6.PubMedCrossRef
59.
Zurück zum Zitat Kapur S, Seeman P. Does fast dissociation from the dopamine d(2) receptor explain the action of atypical antipsychotics? A new hypothesis. Am J Psychiatry. 2001;158(3):360–9.PubMedCrossRef Kapur S, Seeman P. Does fast dissociation from the dopamine d(2) receptor explain the action of atypical antipsychotics? A new hypothesis. Am J Psychiatry. 2001;158(3):360–9.PubMedCrossRef
60.
Zurück zum Zitat Durif F, Debilly B, Galitzky M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology. 2004;62(3):381–8.PubMedCrossRef Durif F, Debilly B, Galitzky M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology. 2004;62(3):381–8.PubMedCrossRef
61.
Zurück zum Zitat Katzenschlager R, Manson AJ, Evans A, et al. 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, et al. 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
62.
Zurück zum Zitat Hill MP, Bezard E, McGuire SG, et al. Novel antiepileptic drug levetiracetam decreases dyskinesia elicited by l-dopa and ropinirole in the MPTP-lesioned marmoset. Mov Disord. 2003;18(11):1301–5.PubMedCrossRef Hill MP, Bezard E, McGuire SG, et al. Novel antiepileptic drug levetiracetam decreases dyskinesia elicited by l-dopa and ropinirole in the MPTP-lesioned marmoset. Mov Disord. 2003;18(11):1301–5.PubMedCrossRef
63.
Zurück zum Zitat Bezard E, Hill MP, Crossman AR, et al. Levetiracetam improves choreic levodopa-induced dyskinesia in the MPTP-treated macaque. Eur J Pharmacol. 2004;485(1–3):159–64.PubMedCrossRef Bezard E, Hill MP, Crossman AR, et al. Levetiracetam improves choreic levodopa-induced dyskinesia in the MPTP-treated macaque. Eur J Pharmacol. 2004;485(1–3):159–64.PubMedCrossRef
64.
Zurück zum Zitat Hill MP, Ravenscroft P, Bezard E, et al. Levetiracetam potentiates the antidyskinetic action of amantadine in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primate model of Parkinson’s disease. J Pharmacol Exp Ther. 2004;310(1):386–94.PubMedCrossRef Hill MP, Ravenscroft P, Bezard E, et al. Levetiracetam potentiates the antidyskinetic action of amantadine in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primate model of Parkinson’s disease. J Pharmacol Exp Ther. 2004;310(1):386–94.PubMedCrossRef
65.
Zurück zum Zitat Wolz M, Löhle M, Strecker K, 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.PubMedCrossRef Wolz M, Löhle M, Strecker K, 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.PubMedCrossRef
66.
Zurück zum Zitat Stathis P, Konitsiotis S, Tagaris G, et al. Levetiracetam for the management of levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2011;26(2):264–70.PubMedCrossRef Stathis P, Konitsiotis S, Tagaris G, et al. Levetiracetam for the management of levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2011;26(2):264–70.PubMedCrossRef
67.
Zurück zum Zitat Wong KK, Alty JE, Goy AG, et al. A randomized, double-blind, placebo-controlled trial of levetiracetam for dyskinesia in Parkinson’s disease. Mov Disord. 2011;26(8):1552–5.PubMedCrossRef Wong KK, Alty JE, Goy AG, et al. A randomized, double-blind, placebo-controlled trial of levetiracetam for dyskinesia in Parkinson’s disease. Mov Disord. 2011;26(8):1552–5.PubMedCrossRef
68.
Zurück zum Zitat Lyons KE, Pahwa R. Efficacy and tolerability of levetiracetam in Parkinson disease patients with levodopa-induced dyskinesia. Clin Neuropharmacol. 2006;29(3):148–53.PubMedCrossRef Lyons KE, Pahwa R. Efficacy and tolerability of levetiracetam in Parkinson disease patients with levodopa-induced dyskinesia. Clin Neuropharmacol. 2006;29(3):148–53.PubMedCrossRef
69.
Zurück zum Zitat Zesiewicz TA, Sullivan KL, Maldonado JL, et al. Open-label pilot study of levetiracetam (Keppra) for the treatment of levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2005;20(9):1205–9.PubMedCrossRef Zesiewicz TA, Sullivan KL, Maldonado JL, et al. Open-label pilot study of levetiracetam (Keppra) for the treatment of levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2005;20(9):1205–9.PubMedCrossRef
70.
Zurück zum Zitat Hely MA, Reid WG, Adena MA, et al. The Sydney multicenter study of Parkinson’s disease: the inevitability of dementia at 20 years. Mov Disord. 2008;23(6):837–44.PubMedCrossRef Hely MA, Reid WG, Adena MA, et al. The Sydney multicenter study of Parkinson’s disease: the inevitability of dementia at 20 years. Mov Disord. 2008;23(6):837–44.PubMedCrossRef
71.
Zurück zum Zitat Balaban CD. Neural substrates linking balance control and anxiety. Physiol Behav. 2002;77:469–75.PubMedCrossRef Balaban CD. Neural substrates linking balance control and anxiety. Physiol Behav. 2002;77:469–75.PubMedCrossRef
72.
Zurück zum Zitat Tohgi H, Abe T, Takahashi S. The effects of l-threo-3,4-dihydroxyphenylserine on the total norepinephrine and dopamine concentrations in the cerebrospinal fluid and freezing gait in parkinsonian patients. J Neural Transm Park Dis Dement Sect. 1993;5:27–34.PubMedCrossRef Tohgi H, Abe T, Takahashi S. The effects of l-threo-3,4-dihydroxyphenylserine on the total norepinephrine and dopamine concentrations in the cerebrospinal fluid and freezing gait in parkinsonian patients. J Neural Transm Park Dis Dement Sect. 1993;5:27–34.PubMedCrossRef
73.
Zurück zum Zitat Fukada K, Endo T, Yokoe M, et al. l-Threo-3,4-dihydroxyphenylserine (l-DOPS) co-administered with entacapone improves freezing of gait in Parkinson’s disease. Med Hypotheses. 2013;80(2):209–12.PubMedCrossRef Fukada K, Endo T, Yokoe M, et al. l-Threo-3,4-dihydroxyphenylserine (l-DOPS) co-administered with entacapone improves freezing of gait in Parkinson’s disease. Med Hypotheses. 2013;80(2):209–12.PubMedCrossRef
74.
Zurück zum Zitat Devilbiss DM, Berridge CW. Low-dose methylphenidate actions on tonic and phasic locus coeruleus discharge. J Pharmacol Exp Ther. 2006;319:1327–35.PubMedCrossRef Devilbiss DM, Berridge CW. Low-dose methylphenidate actions on tonic and phasic locus coeruleus discharge. J Pharmacol Exp Ther. 2006;319:1327–35.PubMedCrossRef
75.
Zurück zum Zitat Volkow ND, Wang G, Fowler JS, et al. Therapeutic doses of oral methylphenidate significantly increase extracellular dopamine in the human brain. J Neurosci. 2001;21(2):RC121.PubMed Volkow ND, Wang G, Fowler JS, et al. Therapeutic doses of oral methylphenidate significantly increase extracellular dopamine in the human brain. J Neurosci. 2001;21(2):RC121.PubMed
76.
Zurück zum Zitat Espay AJ, Dwivedi AK, Payne M, et al. Methylphenidate for gait impairment in Parkinson disease: a randomized clinical trial. Neurology. 2011;76(14):1256–62.PubMedCentralPubMedCrossRef Espay AJ, Dwivedi AK, Payne M, et al. Methylphenidate for gait impairment in Parkinson disease: a randomized clinical trial. Neurology. 2011;76(14):1256–62.PubMedCentralPubMedCrossRef
77.
Zurück zum Zitat Moreau C, Delval A, Defebvre L, et al. Methylphenidate for gait hypokinesia and freezing in patients with Parkinson’s disease undergoing subthalamic stimulation: a multicentre, parallel, randomised, placebo-controlled trial. Lancet Neurol. 2012;11(7):589–96.PubMedCrossRef Moreau C, Delval A, Defebvre L, et al. Methylphenidate for gait hypokinesia and freezing in patients with Parkinson’s disease undergoing subthalamic stimulation: a multicentre, parallel, randomised, placebo-controlled trial. Lancet Neurol. 2012;11(7):589–96.PubMedCrossRef
78.
Zurück zum Zitat Bohnen NI, Müller ML, Koeppe RA, et al. History of falls in Parkinson disease is associated with reduced cholinergic activity. Neurology. 2009;73(20):1670–6.PubMedCentralPubMedCrossRef Bohnen NI, Müller ML, Koeppe RA, et al. History of falls in Parkinson disease is associated with reduced cholinergic activity. Neurology. 2009;73(20):1670–6.PubMedCentralPubMedCrossRef
79.
Zurück zum Zitat Karachi C, Grabli D, Bernard FA, et al. Cholinergic mesencephalic neurons are involved in gait and postural disorders in Parkinson disease. J Clin Investig. 2010;120(8):2745–54.PubMedCentralPubMedCrossRef Karachi C, Grabli D, Bernard FA, et al. Cholinergic mesencephalic neurons are involved in gait and postural disorders in Parkinson disease. J Clin Investig. 2010;120(8):2745–54.PubMedCentralPubMedCrossRef
80.
Zurück zum Zitat Chung KA, Lobb BM, Nutt JG, et al. Effects of a central cholinesterase inhibitor on reducing falls in Parkinson disease. Neurology. 2010;75:1263–9.PubMedCentralPubMedCrossRef Chung KA, Lobb BM, Nutt JG, et al. Effects of a central cholinesterase inhibitor on reducing falls in Parkinson disease. Neurology. 2010;75:1263–9.PubMedCentralPubMedCrossRef
82.
Zurück zum Zitat DeLong MR. Primate models of movement disorders of basal ganglia origin. Trends Neurosci. 1990;13(7):281–5.PubMedCrossRef DeLong MR. Primate models of movement disorders of basal ganglia origin. Trends Neurosci. 1990;13(7):281–5.PubMedCrossRef
83.
Zurück zum Zitat Ghaemi M, Raethjen J, Hilker R, et al. Monosymptomatic resting tremor and Parkinson’s disease: a multitracer positron emission tomographic study. Mov Disord. 2002;17(4):782–8.PubMedCrossRef Ghaemi M, Raethjen J, Hilker R, et al. Monosymptomatic resting tremor and Parkinson’s disease: a multitracer positron emission tomographic study. Mov Disord. 2002;17(4):782–8.PubMedCrossRef
84.
Zurück zum Zitat Asenbaum S, Pirker W, Angelberger P, et al. [123I]beta-CIT and SPECT in essential tremor and Parkinson’s disease. J Neural Transm. 1998;105(10–12):1213–28.PubMedCrossRef Asenbaum S, Pirker W, Angelberger P, et al. [123I]beta-CIT and SPECT in essential tremor and Parkinson’s disease. J Neural Transm. 1998;105(10–12):1213–28.PubMedCrossRef
85.
Zurück zum Zitat Benamer HT, Oertel WH, Patterson J, et al. Prospective study of presynaptic dopaminergic imaging in patients with mild parkinsonism and tremor disorders: part 1. Baseline and 3-month observations. Mov Disord. 2003;18(9):977–84.PubMedCrossRef Benamer HT, Oertel WH, Patterson J, et al. Prospective study of presynaptic dopaminergic imaging in patients with mild parkinsonism and tremor disorders: part 1. Baseline and 3-month observations. Mov Disord. 2003;18(9):977–84.PubMedCrossRef
86.
Zurück zum Zitat Pirker W. Correlation of dopamine transporter imaging with parkinsonian motor handicap: how close is it? Mov Disord. 2003;18(Suppl 7):S43–51.PubMedCrossRef Pirker W. Correlation of dopamine transporter imaging with parkinsonian motor handicap: how close is it? Mov Disord. 2003;18(Suppl 7):S43–51.PubMedCrossRef
87.
Zurück zum Zitat Collins-Praino LE, Paul NE, Rychalsky KL, et al. Pharmacological and physiological characterization of the tremulous jaw movement model of parkinsonian tremor: potential insights into the pathophysiology of tremor. Front Syst Neurosci. 2011;5:49.PubMedCentralPubMedCrossRef Collins-Praino LE, Paul NE, Rychalsky KL, et al. Pharmacological and physiological characterization of the tremulous jaw movement model of parkinsonian tremor: potential insights into the pathophysiology of tremor. Front Syst Neurosci. 2011;5:49.PubMedCentralPubMedCrossRef
88.
Zurück zum Zitat Doder M, Rabiner EA, Turjanski N, et al. Tremor in Parkinson’s disease and serotonergic dysfunction: an 11C-WAY 100635 PET study. Neurology. 2003;60(4):601–5.PubMedCrossRef Doder M, Rabiner EA, Turjanski N, et al. Tremor in Parkinson’s disease and serotonergic dysfunction: an 11C-WAY 100635 PET study. Neurology. 2003;60(4):601–5.PubMedCrossRef
89.
Zurück zum Zitat Loane C, Wu K, Bain P, et al. Serotonergic loss in motor circuitries correlates with severity of action-postural tremor in PD. Neurology. 2013;80:1850–5.PubMedCentralPubMedCrossRef Loane C, Wu K, Bain P, et al. Serotonergic loss in motor circuitries correlates with severity of action-postural tremor in PD. Neurology. 2013;80:1850–5.PubMedCentralPubMedCrossRef
90.
Zurück zum Zitat Carlson BB, Wisniecki A, Salamone JD. Local injections of the 5-hydroxytryptamine antagonist mianserin into substantia nigra pars reticulata block tremulous jaw movements in rats: studies with a putative model of parkinsonian tremor. Psychopharmacology (Berl). 2003;165(3):229–37. Carlson BB, Wisniecki A, Salamone JD. Local injections of the 5-hydroxytryptamine antagonist mianserin into substantia nigra pars reticulata block tremulous jaw movements in rats: studies with a putative model of parkinsonian tremor. Psychopharmacology (Berl). 2003;165(3):229–37.
91.
Zurück zum Zitat Friedman JH, Koller WC, Lannon MC, et al. Benztropine versus clozapine for the treatment of tremor in Parkinson’s disease. Neurology. 1997;48(4):1077–81.PubMedCrossRef Friedman JH, Koller WC, Lannon MC, et al. Benztropine versus clozapine for the treatment of tremor in Parkinson’s disease. Neurology. 1997;48(4):1077–81.PubMedCrossRef
92.
Zurück zum Zitat Trosch RM, Friedman JH, Lannon MC, et al. Clozapine use in Parkinson’s disease: a retrospective analysis of a large multicentered clinical experience. Mov Disord. 1998;13(3):377–82.PubMedCrossRef Trosch RM, Friedman JH, Lannon MC, et al. Clozapine use in Parkinson’s disease: a retrospective analysis of a large multicentered clinical experience. Mov Disord. 1998;13(3):377–82.PubMedCrossRef
93.
Zurück zum Zitat Bonuccelli U, Ceravolo R, Salvetti S, et al. Clozapine in Parkinson’s disease tremor. Effects of acute and chronic administration. Neurology. 1997;49(6):1587–90.PubMedCrossRef Bonuccelli U, Ceravolo R, Salvetti S, et al. Clozapine in Parkinson’s disease tremor. Effects of acute and chronic administration. Neurology. 1997;49(6):1587–90.PubMedCrossRef
94.
Zurück zum Zitat Gordon PH, Pullman SL, Louis ED, et al. Mirtazapine in Parkinsonian tremor. Parkinsonism Relat Disord. 2002;9(2):125–6.PubMedCrossRef Gordon PH, Pullman SL, Louis ED, et al. Mirtazapine in Parkinsonian tremor. Parkinsonism Relat Disord. 2002;9(2):125–6.PubMedCrossRef
95.
Zurück zum Zitat Crosby NJ, Deane KH, Clarke CE. Beta-blocker therapy for tremor in Parkinson’s disease. Cochrane Database Syst Rev. 2003;(1):CD003361. Crosby NJ, Deane KH, Clarke CE. Beta-blocker therapy for tremor in Parkinson’s disease. Cochrane Database Syst Rev. 2003;(1):CD003361.
Metadaten
Titel
Non-dopaminergic Treatments for Motor Control in Parkinson’s Disease
verfasst von
Susan H. Fox
Publikationsdatum
01.09.2013
Verlag
Springer International Publishing
Erschienen in
Drugs / Ausgabe 13/2013
Print ISSN: 0012-6667
Elektronische ISSN: 1179-1950
DOI
https://doi.org/10.1007/s40265-013-0105-4

Weitere Artikel der Ausgabe 13/2013

Drugs 13/2013 Zur Ausgabe