Skip to main content
Top
Published in: CNS Drugs 9/2005

01-09-2005 | Leading Article

Treatment of Parkinson’s Disease

What’s on the Horizon?

Authors: Dr Stacy S. Wu, Steven J. Frucht

Published in: CNS Drugs | Issue 9/2005

Login to get access

Abstract

Few neurological diseases have received as much attention and investment in research as Parkinson’s disease. Although great strides have been made in the development of agents to treat this neurodegenerative disease, none yet address the underlying problem associated with it, the progressive loss of dopaminergic neurons. Current therapeutic strategies for Parkinson’s disease focus primarily on reducing the severity of its symptoms using dopaminergic medications. Although providing substantial benefit, these agents are burdened by adverse effects and long-term complications. This review highlights new and emerging therapies for Parkinson’s disease, categorised as symptomatic, neuroprotective and neurorestorative, although at times, this distinction is not easily made.
Novel symptomatic treatments target nondopaminergic areas in the hope of avoiding the motor complications seen with dopaminergic therapies. Two emerging treatment approaches under investigation are adenosine A2A receptor antagonists (such as istradefylline [KW-6002]) and glutamate AMPA receptor antagonists (such as talampanel [LY-300164]). In 2003, the results from two studies using istradefylline in patients with Parkinson’s disease were published, with both showing a positive benefit of the study drug when used as adjunctive therapy to levodopa. In non-human primate models of Parkinson’s disease, talampanel has been found to have antiparkinsonian effects when administered as high-dose monotherapy and antidyskinetic effects on levodopa-induced dyskinesias. NS-2330, another drug currently undergoing clinical trials, is a triple monoamine reuptake inhibitor that has therapeutic potential in both Parkinson’s and Alzheimer’s disease. A phase II proof-of-concept study is currently underway in early Parkinson’s disease. However, a recently published study in advanced Parkinson’s disease showed no therapeutic benefit of NS-2330 in this patient population.
Even more exciting are agents that have a neuroprotective or neurorestorative role. These therapies aim to prevent disease progression by targeting the mechanisms involved in the pathogenesis of Parkinson’s disease. Several lines of investigation for neuroprotective therapies have been taken, including the antioxidant coenzyme Q10 (ubidecarenone) and anti-apoptotic agents such as CEP-1347. Studies in patients with Parkinson’s disease with coenzyme Q10 have suggested that it slows down functional decline. The PRECEPT study is currently in progress to assess the neuroprotective role of CEP-1347 in the early phase of the disease.
Gene therapy is another exciting arena and includes both potentially neuroprotective and neurorestorative agents. Novel methods include subthalamic glutamic acid decarboxylase gene therapy and the use of glial cell line-derived neurotrophic factor (GDNF). Eleven of 12 patients have been enrolled in the first FDA-approved phase I subthalamic glutamic acid decarboxylase gene therapy trial for Parkinson’s disease, with currently no evidence of adverse events. GDNF delivered intracerebroventricularly was studied in a small population of patients with Parkinson’s disease, but unfortunately did not reveal positive results. Other methods of administering GDNF include direct delivery via infusions into the basal ganglia and the use of viral vectors; thus far, these approaches have shown promising results.
This is an exciting and rewarding time for research into Parkinson’s disease. With so many therapies currently under investigation, the time is ripe for the beginning of a new phase of treatment strategies.
Literature
1.
go back to reference Mouradian M. Recent advances in the genetics and pathogenesis of Parkinson’s disease. Neurology 2002; 58(2): 179–85PubMedCrossRef Mouradian M. Recent advances in the genetics and pathogenesis of Parkinson’s disease. Neurology 2002; 58(2): 179–85PubMedCrossRef
2.
go back to reference McNaught K, Mytilineou C, Jnobaptiste R, et al. Impairment of the ubiquitin-proteasome system causes dopaminergic cell death and inclusion body formation in the ventral mesencephalic cultures. J Neurochem 2002; 81: 301–6PubMedCrossRef McNaught K, Mytilineou C, Jnobaptiste R, et al. Impairment of the ubiquitin-proteasome system causes dopaminergic cell death and inclusion body formation in the ventral mesencephalic cultures. J Neurochem 2002; 81: 301–6PubMedCrossRef
3.
go back to reference Parkinson Study Group. Entacapone improves motor fluctuations in levodopa-treated Parkinson’s disease patients. Ann Neurol 1997; 42(5): 747–55CrossRef Parkinson Study Group. Entacapone improves motor fluctuations in levodopa-treated Parkinson’s disease patients. Ann Neurol 1997; 42(5): 747–55CrossRef
4.
go back to reference Rinne UK, Larsen JP, Siden A, et al. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations. Neurology 1998; 51: 1309–14PubMedCrossRef Rinne UK, Larsen JP, Siden A, et al. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations. Neurology 1998; 51: 1309–14PubMedCrossRef
5.
go back to reference Ruottinen HM, Rinne UK. A double-blind pharmacokinetic and clinical dose-response study of entacapone as an adjuvant to levodopa therapy in advanced Parkinson’s disease. Clin Neuropharmacol 1996; 19: 283–96PubMedCrossRef Ruottinen HM, Rinne UK. A double-blind pharmacokinetic and clinical dose-response study of entacapone as an adjuvant to levodopa therapy in advanced Parkinson’s disease. Clin Neuropharmacol 1996; 19: 283–96PubMedCrossRef
6.
go back to reference Jenner P. A2A antagonists as novel nondopaminergic therapy for motor dysfunction in PD. Neurology 2003; 61Suppl. 6: S33–8 Jenner P. A2A antagonists as novel nondopaminergic therapy for motor dysfunction in PD. Neurology 2003; 61Suppl. 6: S33–8
7.
go back to reference Mori A, Shindou T. Modulation of GABAergic transmission in the striatopallidal system by adenosine A2A antagonists. Neurology 2003; 61Suppl. 6: S44–8PubMedCrossRef Mori A, Shindou T. Modulation of GABAergic transmission in the striatopallidal system by adenosine A2A antagonists. Neurology 2003; 61Suppl. 6: S44–8PubMedCrossRef
8.
go back to reference Penney Jr J, Young A. Striatal in homogeneities and basal ganglia origin. Mov Disord 1986; 1: 3–15PubMedCrossRef Penney Jr J, Young A. Striatal in homogeneities and basal ganglia origin. Mov Disord 1986; 1: 3–15PubMedCrossRef
9.
go back to reference Bara-Jimenez W, Sherzai A, Dimitrova T, et al. Adenosine A2A receptor antagonist treatment of Parkinson’s disease. Neurology 2003; 61: 293–6PubMedCrossRef Bara-Jimenez W, Sherzai A, Dimitrova T, et al. Adenosine A2A receptor antagonist treatment of Parkinson’s disease. Neurology 2003; 61: 293–6PubMedCrossRef
10.
go back to reference Chase T, Oh J. Striatal mechanisms and pathogenesis of parkinsonian signs and motor complications. Ann Neurol 2000; 27Suppl. 1: S122–9 Chase T, Oh J. Striatal mechanisms and pathogenesis of parkinsonian signs and motor complications. Ann Neurol 2000; 27Suppl. 1: S122–9
11.
go back to reference Schiffmann S, Vanderhaeghen J. Adenosine A2A receptors regulate the gene expression of striatopallidal and striatonigral neurons. J Neurosci 1993; 13: 1080–7PubMed Schiffmann S, Vanderhaeghen J. Adenosine A2A receptors regulate the gene expression of striatopallidal and striatonigral neurons. J Neurosci 1993; 13: 1080–7PubMed
12.
go back to reference Fink J, Weaver D, Rivkees S, et al. Molecular cloning of the rat A2A adenosine receptor: selective co-expression with D2 dopamine receptors in rat striatum. Mol Brain Res 1992; 14: 186–95PubMedCrossRef Fink J, Weaver D, Rivkees S, et al. Molecular cloning of the rat A2A adenosine receptor: selective co-expression with D2 dopamine receptors in rat striatum. Mol Brain Res 1992; 14: 186–95PubMedCrossRef
13.
go back to reference Franco R, Ferre S, Agnati L, et al. Evidence for adenosine/dopamine receptor interactions: indications for heteromerization. Neuropsychopharmacology 2000; 23: S50–9PubMedCrossRef Franco R, Ferre S, Agnati L, et al. Evidence for adenosine/dopamine receptor interactions: indications for heteromerization. Neuropsychopharmacology 2000; 23: S50–9PubMedCrossRef
14.
go back to reference Feigin A. Nondopaminergic symptomatic therapies for Parkinson’s disease. Neurology 2003; 61: 286–7PubMedCrossRef Feigin A. Nondopaminergic symptomatic therapies for Parkinson’s disease. Neurology 2003; 61: 286–7PubMedCrossRef
15.
go back to reference Carta A, Pinna A, Tronci E, et al. Adenosine A2A and dopamine receptor interactions in basal ganglia of dopamine denervated rats. Neurology 2003; 61Suppl. 6: S39–42PubMedCrossRef Carta A, Pinna A, Tronci E, et al. Adenosine A2A and dopamine receptor interactions in basal ganglia of dopamine denervated rats. Neurology 2003; 61Suppl. 6: S39–42PubMedCrossRef
16.
go back to reference Kanda T, Jackson M, Smith L, et al. Adenosine A2A antagonist: a novel antiparkinsonian agent that does not provoke dyskinesia in parkinsonian monkeys. Ann Neurol 1998; 43: 507–13PubMedCrossRef Kanda T, Jackson M, Smith L, et al. Adenosine A2A antagonist: a novel antiparkinsonian agent that does not provoke dyskinesia in parkinsonian monkeys. Ann Neurol 1998; 43: 507–13PubMedCrossRef
17.
go back to reference Hauser R, Hubble J, Truong D, et al. Randomized trial of the adenosine A2A receptor antagonist istradefylline in advanced PD. Neurology 2003; 61: 297–303PubMedCrossRef Hauser R, Hubble J, Truong D, et al. Randomized trial of the adenosine A2A receptor antagonist istradefylline in advanced PD. Neurology 2003; 61: 297–303PubMedCrossRef
18.
go back to reference Ruottinen HM, Rinne UK. COMT inhibition in the treatment of Parkinson’s disease. J Neurol 1998; 245Suppl. 3: 25–34CrossRef Ruottinen HM, Rinne UK. COMT inhibition in the treatment of Parkinson’s disease. J Neurol 1998; 245Suppl. 3: 25–34CrossRef
19.
go back to reference Kanda T, Jackson M, Smith L, et al. Combined use of the adenosine A2a antagonist KW-6002 with L-dopa or with selective D1 or D2 dopamine agonists increases antiparkinsonian activity but not dyskinesia in MPTP-treated monkeys. Exp Neurol 2000; 162: 321–7PubMedCrossRef Kanda T, Jackson M, Smith L, et al. Combined use of the adenosine A2a antagonist KW-6002 with L-dopa or with selective D1 or D2 dopamine agonists increases antiparkinsonian activity but not dyskinesia in MPTP-treated monkeys. Exp Neurol 2000; 162: 321–7PubMedCrossRef
20.
go back to reference Grondin R, Bedard PJ, Tahar AH, et al. Antiparkinsonian effect of a new selective adenosine A2A receptor antagonist in MPTP-treated monkeys. Neurology 1999; 52: 1673–7PubMedCrossRef Grondin R, Bedard PJ, Tahar AH, et al. Antiparkinsonian effect of a new selective adenosine A2A receptor antagonist in MPTP-treated monkeys. Neurology 1999; 52: 1673–7PubMedCrossRef
21.
go back to reference Frucht S, Greene P, Fahn S. Sleep episodes in Parkinson’s disease: a wake-up call. Mov Disord 2000; 15: 601–3PubMedCrossRef Frucht S, Greene P, Fahn S. Sleep episodes in Parkinson’s disease: a wake-up call. Mov Disord 2000; 15: 601–3PubMedCrossRef
22.
go back to reference Frucht S, Rogers J, Greene P, et al. Falling asleep at the wheel: motor vehicle mishaps in persons taking pramipexole and ropinirole. Neurology 1999; 52: 1908–10PubMedCrossRef Frucht S, Rogers J, Greene P, et al. Falling asleep at the wheel: motor vehicle mishaps in persons taking pramipexole and ropinirole. Neurology 1999; 52: 1908–10PubMedCrossRef
23.
go back to reference Paus S, Brecht H, Köster J, et al. Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s disease. Mov Disord 2003; 18: 659–67PubMedCrossRef Paus S, Brecht H, Köster J, et al. Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s disease. Mov Disord 2003; 18: 659–67PubMedCrossRef
24.
go back to reference Shaunak S, Wilkins A, Pillig J, et al. Pericardial, retroperitoneal, and pleural fibrosis induced by pergolide. J Neurol Neurosurg Psychiatry 1999; 66: 79–81PubMedCrossRef Shaunak S, Wilkins A, Pillig J, et al. Pericardial, retroperitoneal, and pleural fibrosis induced by pergolide. J Neurol Neurosurg Psychiatry 1999; 66: 79–81PubMedCrossRef
25.
go back to reference Schwarzschild M, Xu K, Oztas E, et al. Neuroprotection by caffeine and more specific A2A receptor antagonists in animal models of Parkinson’s disease. Neurology 2003; 61Suppl. 6: S55–61PubMedCrossRef Schwarzschild M, Xu K, Oztas E, et al. Neuroprotection by caffeine and more specific A2A receptor antagonists in animal models of Parkinson’s disease. Neurology 2003; 61Suppl. 6: S55–61PubMedCrossRef
26.
go back to reference Pedata F, Pugliese A, Melani A, et al. A2A receptors in neuroprotection of dopaminergic neurons. Neurology 2003; 61Suppl. 6: S49–50PubMedCrossRef Pedata F, Pugliese A, Melani A, et al. A2A receptors in neuroprotection of dopaminergic neurons. Neurology 2003; 61Suppl. 6: S49–50PubMedCrossRef
27.
go back to reference Hernàn M, Takkouche B, Caamaño-Isorna F, et al. A meta-analysis of coffee drinking, cigarette smoking, and the risk of Parkinson’s disease. Ann Neurol 2002; 52: 276–84PubMedCrossRef Hernàn M, Takkouche B, Caamaño-Isorna F, et al. A meta-analysis of coffee drinking, cigarette smoking, and the risk of Parkinson’s disease. Ann Neurol 2002; 52: 276–84PubMedCrossRef
28.
go back to reference Fall P, Fredrikson M, Axelson O, et al. Nutritional and occupational factors influencing the risk of Parkinson’s disease: a case-controlled study in southeastern Sweden. Mov Disord 1999; 14: 28–37PubMedCrossRef Fall P, Fredrikson M, Axelson O, et al. Nutritional and occupational factors influencing the risk of Parkinson’s disease: a case-controlled study in southeastern Sweden. Mov Disord 1999; 14: 28–37PubMedCrossRef
29.
go back to reference Ascherio A, Chen H. Caffeinated clues from epidemiology of Parkinson’s disease. Neurology 2001; 61Suppl. 6: S51–4 Ascherio A, Chen H. Caffeinated clues from epidemiology of Parkinson’s disease. Neurology 2001; 61Suppl. 6: S51–4
30.
go back to reference Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease. The Parkinson Study Group. N Engl J Med 1993; 328(3): 176–83CrossRef Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease. The Parkinson Study Group. N Engl J Med 1993; 328(3): 176–83CrossRef
31.
go back to reference Ward C. Does selegiline delay progression of Parkinson’s disease? A critical re-evaluation of the DATATOP study. J Neurol Neurosurg Psychiatry 1994; 57: 217–20PubMedCrossRef Ward C. Does selegiline delay progression of Parkinson’s disease? A critical re-evaluation of the DATATOP study. J Neurol Neurosurg Psychiatry 1994; 57: 217–20PubMedCrossRef
32.
go back to reference Fahn S. Controversies in the therapy of Parkinson’s disease. Adv Neurol 1996; 69: 477–86PubMed Fahn S. Controversies in the therapy of Parkinson’s disease. Adv Neurol 1996; 69: 477–86PubMed
33.
go back to reference Chase TN, Oh JD, Blanchet PJ. Neostriatal mechanisms in Parkinson’s disease. Neurology 1998; 51(2 Suppl. 2): S30–5PubMedCrossRef Chase TN, Oh JD, Blanchet PJ. Neostriatal mechanisms in Parkinson’s disease. Neurology 1998; 51(2 Suppl. 2): S30–5PubMedCrossRef
34.
go back to reference Chase TN, Engber TM, Mouradian MM. Contribution of dopaminergic and glutamatergic mechanisms to the pathogenesis of motor response complications in Parkinson’s disease. Adv Neurol 1996; 69: 497–501PubMed Chase TN, Engber TM, Mouradian MM. Contribution of dopaminergic and glutamatergic mechanisms to the pathogenesis of motor response complications in Parkinson’s disease. Adv Neurol 1996; 69: 497–501PubMed
35.
go back to reference Calon F, Rajput AH, Hornykiewicz O, et al. Levodopa-induced motor complications are associated with alterations of glutamate receptors in Parkinson’s disease. Neurobiol Dis 2003; 14(3): 404–16PubMedCrossRef Calon F, Rajput AH, Hornykiewicz O, et al. Levodopa-induced motor complications are associated with alterations of glutamate receptors in Parkinson’s disease. Neurobiol Dis 2003; 14(3): 404–16PubMedCrossRef
36.
go back to reference Hadj Tahar A, Gregoire L, Darre A, et al. Effect of a selective glutamate antagonist on L-dopa-induced dyskinesias in drug-naive parkinsonian monkeys. Neurobiol Dis 2004; 15(2): 171–6PubMedCrossRef Hadj Tahar A, Gregoire L, Darre A, et al. Effect of a selective glutamate antagonist on L-dopa-induced dyskinesias in drug-naive parkinsonian monkeys. Neurobiol Dis 2004; 15(2): 171–6PubMedCrossRef
37.
go back to reference Merims D, Ziv I, Sherki Y, et al. The role of glutamatergic transmission in the pathogenesis of levodopa-induced dyskinesias: potential therapeutic approaches. Neurol Neurochir Pol 2001; 35Suppl. 3: 65–8PubMed Merims D, Ziv I, Sherki Y, et al. The role of glutamatergic transmission in the pathogenesis of levodopa-induced dyskinesias: potential therapeutic approaches. Neurol Neurochir Pol 2001; 35Suppl. 3: 65–8PubMed
38.
go back to reference Baas H. Dyskinesia in Parkinson’s disease: pathophysiology and clinical risk factors. J Neurol 2000; 247Suppl. 4: IV/12–6 Baas H. Dyskinesia in Parkinson’s disease: pathophysiology and clinical risk factors. J Neurol 2000; 247Suppl. 4: IV/12–6
39.
go back to reference Oh JD, Russell DS, Vaughan CL, et al. Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and L-DOPA administration. Brain Res 1998; 813(1): 150–9PubMedCrossRef Oh JD, Russell DS, Vaughan CL, et al. Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and L-DOPA administration. Brain Res 1998; 813(1): 150–9PubMedCrossRef
40.
go back to reference Oh JD, Vaughan CL, Chase TN. Effect of dopamine denervation and dopamine agonist administration on serine phosphorylation of striatal NMDA receptor subunits. Brain Res 1999; 821(2): 433–42PubMedCrossRef Oh JD, Vaughan CL, Chase TN. Effect of dopamine denervation and dopamine agonist administration on serine phosphorylation of striatal NMDA receptor subunits. Brain Res 1999; 821(2): 433–42PubMedCrossRef
41.
go back to reference Chase TN. Levodopa therapy: consequences of the nonphysiologic replacement of dopamine. Neurology 1998; 50(5 Suppl. 5): S17–25PubMedCrossRef Chase TN. Levodopa therapy: consequences of the nonphysiologic replacement of dopamine. Neurology 1998; 50(5 Suppl. 5): S17–25PubMedCrossRef
42.
go back to reference Blanchet PJ, Konitsiotis S, Chase TN. Amantadine reduces levodopa-induced dyskinesias in parkinsonian monkeys. Mov Disord 1998; 13(5): 798–802PubMedCrossRef Blanchet PJ, Konitsiotis S, Chase TN. Amantadine reduces levodopa-induced dyskinesias in parkinsonian monkeys. Mov Disord 1998; 13(5): 798–802PubMedCrossRef
43.
go back to reference Papa SM, Chase TN. Levodopa-induced dyskinesias improved by a glutamate antagonist in Parkinsonian monkeys. Ann Neurol 1996; 39(5): 574–8PubMedCrossRef Papa SM, Chase TN. Levodopa-induced dyskinesias improved by a glutamate antagonist in Parkinsonian monkeys. Ann Neurol 1996; 39(5): 574–8PubMedCrossRef
44.
go back to reference Verhagen Metman L, Del Dotto P, van den Munckhof P, et al. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology 1998; 50(5): 1323–6PubMedCrossRef Verhagen Metman L, Del Dotto P, van den Munckhof P, et al. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology 1998; 50(5): 1323–6PubMedCrossRef
45.
go back to reference Danysz W, Parsons CG, Kornhuber J, et al. Aminoadamantanes as NMDA receptor antagonists and antiparkinsonian agents: preclinical studies. Neurosci Biobehav Rev 1997; 21(4): 455–68PubMedCrossRef Danysz W, Parsons CG, Kornhuber J, et al. Aminoadamantanes as NMDA receptor antagonists and antiparkinsonian agents: preclinical studies. Neurosci Biobehav Rev 1997; 21(4): 455–68PubMedCrossRef
46.
go back to reference Konitsiotis S, Blanchet PJ, Verhagen L, et al. AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys. Neurology 2000; 54(8): 1589–95PubMedCrossRef Konitsiotis S, Blanchet PJ, Verhagen L, et al. AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys. Neurology 2000; 54(8): 1589–95PubMedCrossRef
47.
go back to reference Löschmann PA, Wullner U, Heneka MT, et al. Differential interaction of competitive NMDA and AMPA antagonists with selective dopamine D-1 and D-2 agonists in a rat model of Parkinson’s disease. Synapse 1997; 26(4): 381–91PubMedCrossRef Löschmann PA, Wullner U, Heneka MT, et al. Differential interaction of competitive NMDA and AMPA antagonists with selective dopamine D-1 and D-2 agonists in a rat model of Parkinson’s disease. Synapse 1997; 26(4): 381–91PubMedCrossRef
48.
go back to reference Gossel M, Schmidt WJ, Loscher W, et al. Effect of coadministration of glutamate receptor antagonists and dopaminergic agonists on locomotion in monoamine-depleted rats. J Neural Transm Park Dis Dement Sect 1995; 10(1): 27–39PubMedCrossRef Gossel M, Schmidt WJ, Loscher W, et al. Effect of coadministration of glutamate receptor antagonists and dopaminergic agonists on locomotion in monoamine-depleted rats. J Neural Transm Park Dis Dement Sect 1995; 10(1): 27–39PubMedCrossRef
49.
go back to reference Löschmann PA, Lange KW, Kunow M, et al. Synergism of the AMPA-antagonist NBQX and the NMDA-antagonist CPP with L-dopa in models of Parkinson’s disease. J Neural Transm Park Dis Dement Sect 1991; 3(3): 203–13PubMedCrossRef Löschmann PA, Lange KW, Kunow M, et al. Synergism of the AMPA-antagonist NBQX and the NMDA-antagonist CPP with L-dopa in models of Parkinson’s disease. J Neural Transm Park Dis Dement Sect 1991; 3(3): 203–13PubMedCrossRef
50.
go back to reference Wachtel H, Kunow M, Loschmann PA. NBQX (6-nitrosulfamoyl-benzo-quinoxaline-dione) and CPP (3-carboxy-piperazin-propyl phosphonic acid) potentiate dopamine agonist induced rotations in substantia nigra lesioned rats. Neurosci Lett 1992; 142(2): 179–82PubMedCrossRef Wachtel H, Kunow M, Loschmann PA. NBQX (6-nitrosulfamoyl-benzo-quinoxaline-dione) and CPP (3-carboxy-piperazin-propyl phosphonic acid) potentiate dopamine agonist induced rotations in substantia nigra lesioned rats. Neurosci Lett 1992; 142(2): 179–82PubMedCrossRef
51.
go back to reference Marin C, Jimenez A, Bonastre M, et al. Non-NMDA receptor-mediated mechanisms are involved in levodopa-induced motor response alterations in Parkinsonian rats. Synapse 2000; 36(4): 267–74PubMedCrossRef Marin C, Jimenez A, Bonastre M, et al. Non-NMDA receptor-mediated mechanisms are involved in levodopa-induced motor response alterations in Parkinsonian rats. Synapse 2000; 36(4): 267–74PubMedCrossRef
52.
go back to reference Marin C, Jimenez A, Bonastre M, et al. LY293558, an AMPA glutamate receptor antagonist, prevents and reverses levodopa-induced motor alterations in Parkinsonian rats. Synapse 2001; 42(1): 40–7PubMedCrossRef Marin C, Jimenez A, Bonastre M, et al. LY293558, an AMPA glutamate receptor antagonist, prevents and reverses levodopa-induced motor alterations in Parkinsonian rats. Synapse 2001; 42(1): 40–7PubMedCrossRef
53.
go back to reference Young AB, Fagg GE. Excitatory amino acid receptors in the brain: membrane binding and receptor autoradiographic approaches. Trends Pharmacol Sci 1990; 11(3): 126–33PubMedCrossRef Young AB, Fagg GE. Excitatory amino acid receptors in the brain: membrane binding and receptor autoradiographic approaches. Trends Pharmacol Sci 1990; 11(3): 126–33PubMedCrossRef
54.
go back to reference Albin RL, Makowiec RL, Hollingsworth ZR, et al. Excitatory amino acid binding sites in the basal ganglia of the rat: a quantitative autoradiographic study. Neuroscience 1992; 46(1): 35–48PubMedCrossRef Albin RL, Makowiec RL, Hollingsworth ZR, et al. Excitatory amino acid binding sites in the basal ganglia of the rat: a quantitative autoradiographic study. Neuroscience 1992; 46(1): 35–48PubMedCrossRef
55.
go back to reference Bernard V, Somogyi P, Bolam JP. Cellular, subcellular, and subsynaptic distribution of AMPA-type glutamate receptor subunits in the neostriatum of the rat. J Neurosci 1997; 17(2): 819–33PubMed Bernard V, Somogyi P, Bolam JP. Cellular, subcellular, and subsynaptic distribution of AMPA-type glutamate receptor subunits in the neostriatum of the rat. J Neurosci 1997; 17(2): 819–33PubMed
56.
go back to reference Ozawa S, Kamiya H, Tsuzuki K. Glutamate receptors in the mammalian central nervous system. Prog Neurobiol 1998; 54(5): 581–618PubMedCrossRef Ozawa S, Kamiya H, Tsuzuki K. Glutamate receptors in the mammalian central nervous system. Prog Neurobiol 1998; 54(5): 581–618PubMedCrossRef
57.
go back to reference Carvalho AL, Kameyama K, Huganir RL. Characterization of phosphorylation sites on the glutamate receptor 4 subunit of the AMPA receptors. J Neurosci 1999; 19(12): 4748–54PubMed Carvalho AL, Kameyama K, Huganir RL. Characterization of phosphorylation sites on the glutamate receptor 4 subunit of the AMPA receptors. J Neurosci 1999; 19(12): 4748–54PubMed
58.
go back to reference Derkach V, Barria A, Soderling TR. Ca2+/calmodulin-kinase II enhances channel conductance of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate type glutamate receptors. Proc Natl Acad Sci U S A 1999; 96(6): 3269–74PubMedCrossRef Derkach V, Barria A, Soderling TR. Ca2+/calmodulin-kinase II enhances channel conductance of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate type glutamate receptors. Proc Natl Acad Sci U S A 1999; 96(6): 3269–74PubMedCrossRef
59.
go back to reference Sheardown MJ, Nielsen EO, Hansen AJ, et al. 2,3-Dihydroxy-6-nitro-7-sulfamoyl-benzo (F)quinoxaline: a neuroprotectant for cerebral ischemia. Science 1990; 247(4942): 571–4PubMedCrossRef Sheardown MJ, Nielsen EO, Hansen AJ, et al. 2,3-Dihydroxy-6-nitro-7-sulfamoyl-benzo (F)quinoxaline: a neuroprotectant for cerebral ischemia. Science 1990; 247(4942): 571–4PubMedCrossRef
60.
go back to reference Johnson SA, Luu NT, Herbst TA, et al. Synergistic interactions between ampakines and antipsychotic drugs. J Pharmacol Exp Ther 1999; 289(1): 392–7PubMed Johnson SA, Luu NT, Herbst TA, et al. Synergistic interactions between ampakines and antipsychotic drugs. J Pharmacol Exp Ther 1999; 289(1): 392–7PubMed
61.
go back to reference Lodge D, Bond A, O’Neill MJ, et al. Stereoselective effects-of 2,3-benzodiazepines in vivo: electrophysiology and neuroprotection studies. Neuropharmacology 1996; 35(12): 1681–8PubMedCrossRef Lodge D, Bond A, O’Neill MJ, et al. Stereoselective effects-of 2,3-benzodiazepines in vivo: electrophysiology and neuroprotection studies. Neuropharmacology 1996; 35(12): 1681–8PubMedCrossRef
62.
go back to reference Bleakman D, Ballyk BA, Schoepp DD, et al. Activity of 2,3-benzodiazepines at native rat and recombinant human glutamate receptors in vitro: stereospecificity and selectivity profiles. Neuropharmacology 1996; 35(12): 1689–702PubMedCrossRef Bleakman D, Ballyk BA, Schoepp DD, et al. Activity of 2,3-benzodiazepines at native rat and recombinant human glutamate receptors in vitro: stereospecificity and selectivity profiles. Neuropharmacology 1996; 35(12): 1689–702PubMedCrossRef
63.
go back to reference Bara-Jiminez W, Dimitrova T, Sherzai A, et al. Effect of monoamine reuptake inhibitor NS 2330 in advanced Parkinson’s Disease. Mv Disord 2004; 19(10): 1183–6CrossRef Bara-Jiminez W, Dimitrova T, Sherzai A, et al. Effect of monoamine reuptake inhibitor NS 2330 in advanced Parkinson’s Disease. Mv Disord 2004; 19(10): 1183–6CrossRef
64.
go back to reference Wesnes K, Preskorn S, Friesen S, et al. NS2330 enhances cognitive function in normal volunteers and volunteers with possible Alzheimer’s disease. Clin Pharmacol Ther 2002; 71(2): 7 Wesnes K, Preskorn S, Friesen S, et al. NS2330 enhances cognitive function in normal volunteers and volunteers with possible Alzheimer’s disease. Clin Pharmacol Ther 2002; 71(2): 7
65.
go back to reference Thatte U. NS-2330 (Neurosearch). Curr Opin Investig Drugs 2001; 2(11): 1592–4PubMed Thatte U. NS-2330 (Neurosearch). Curr Opin Investig Drugs 2001; 2(11): 1592–4PubMed
67.
go back to reference Tran M, Mitchell T, Kennedy D, et al. Role of coenzyme Q10 in chronic heart failure, angina, and hypertension. Pharmacotherapy 2001; 7: 797–806CrossRef Tran M, Mitchell T, Kennedy D, et al. Role of coenzyme Q10 in chronic heart failure, angina, and hypertension. Pharmacotherapy 2001; 7: 797–806CrossRef
68.
go back to reference Huntington Study Group. A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington’s disease. Neurology 2001; 57(3): 397–404 Huntington Study Group. A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington’s disease. Neurology 2001; 57(3): 397–404
69.
go back to reference Przedborski S, Jackson-Lewis V. Mechanisms of MPTP toxicity. Mov Disord 1998; 13Suppl. 1: 35–8PubMed Przedborski S, Jackson-Lewis V. Mechanisms of MPTP toxicity. Mov Disord 1998; 13Suppl. 1: 35–8PubMed
70.
go back to reference Krige D, Carroll M, Cooper J, et al. Platelet mitochondrial dysfunction in Parkinson’s disease. Ann Neurol 1992; 32: 782–8PubMedCrossRef Krige D, Carroll M, Cooper J, et al. Platelet mitochondrial dysfunction in Parkinson’s disease. Ann Neurol 1992; 32: 782–8PubMedCrossRef
71.
go back to reference Parker Jr W, Boyson S, Parks J. Abnormalities of the electron transport chain in idiopathic Parkinson’s disease. Ann Neurol 1989; 26: 719–23PubMedCrossRef Parker Jr W, Boyson S, Parks J. Abnormalities of the electron transport chain in idiopathic Parkinson’s disease. Ann Neurol 1989; 26: 719–23PubMedCrossRef
72.
go back to reference Turner C, Schapira A. Mitochondrial dysfunction in neurodegenerative disorders and ageing. Adv Exp Med Biol 2001; 487: 229–51PubMedCrossRef Turner C, Schapira A. Mitochondrial dysfunction in neurodegenerative disorders and ageing. Adv Exp Med Biol 2001; 487: 229–51PubMedCrossRef
73.
go back to reference Müller T, Büttner T, Gholipour A, et al. Coenzyme Q10 supplementation provides mild symptomatic benefit in patients with Parkinson’s disease. Neurosci Lett 2003; 341: 201–4PubMedCrossRef Müller T, Büttner T, Gholipour A, et al. Coenzyme Q10 supplementation provides mild symptomatic benefit in patients with Parkinson’s disease. Neurosci Lett 2003; 341: 201–4PubMedCrossRef
74.
go back to reference Beal MF, Matthews R, Tieleman A, et al. Coenzyme Q10 attenuates the l-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced loss of striatal dopamine and dopaminergic axons in aged mice. Brain Res 1998; 783: 109–14PubMedCrossRef Beal MF, Matthews R, Tieleman A, et al. Coenzyme Q10 attenuates the l-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced loss of striatal dopamine and dopaminergic axons in aged mice. Brain Res 1998; 783: 109–14PubMedCrossRef
75.
go back to reference Matthews R, Yang L, Browne S, et al. Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc Natl Acad Sci U S A 1998; 95: 8892–7PubMedCrossRef Matthews R, Yang L, Browne S, et al. Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc Natl Acad Sci U S A 1998; 95: 8892–7PubMedCrossRef
76.
go back to reference Shults C, Beal M, Fontaine S, et al. Absorption, tolerability, and effects on mitochondrial activity of oral coenzyme Q10 in parkinsonian patients. Neurology 1998; 50: 793–5PubMedCrossRef Shults C, Beal M, Fontaine S, et al. Absorption, tolerability, and effects on mitochondrial activity of oral coenzyme Q10 in parkinsonian patients. Neurology 1998; 50: 793–5PubMedCrossRef
77.
go back to reference Shults C, Oakes D, Kieburtz K, et al. Effects of coenzyme Q10 in early Parkinson disease. Arch Neurol 2002; 59: 1541–50PubMedCrossRef Shults C, Oakes D, Kieburtz K, et al. Effects of coenzyme Q10 in early Parkinson disease. Arch Neurol 2002; 59: 1541–50PubMedCrossRef
78.
go back to reference Hirsch EC, Hunot S, Faucheux B, et al. Dopaminergic neurons degenerate by apoptosis in Parkinson’s disease. Mov Disord 1999; 14(2): 383–5PubMedCrossRef Hirsch EC, Hunot S, Faucheux B, et al. Dopaminergic neurons degenerate by apoptosis in Parkinson’s disease. Mov Disord 1999; 14(2): 383–5PubMedCrossRef
79.
go back to reference Hartmann A, Hunot S, Michel PP, et al. Caspase-3: a vulnerability factor and final effector in apoptotic death of dopaminergic neurons in Parkinson’s disease. Proc Natl Acad Sci U S A 2000; 97(6): 2875–80PubMedCrossRef Hartmann A, Hunot S, Michel PP, et al. Caspase-3: a vulnerability factor and final effector in apoptotic death of dopaminergic neurons in Parkinson’s disease. Proc Natl Acad Sci U S A 2000; 97(6): 2875–80PubMedCrossRef
80.
go back to reference Maroney AC, Finn JP, Bozyczko-Coyne D, et al. CEP-1347 (KT7515), an inhibitor of JNK activation, rescues sympathetic neurons and neuronally differentiated PC12 cells from death evoked by three distinct insults. J Neurochem 1999; 73(5): 1901–12PubMed Maroney AC, Finn JP, Bozyczko-Coyne D, et al. CEP-1347 (KT7515), an inhibitor of JNK activation, rescues sympathetic neurons and neuronally differentiated PC12 cells from death evoked by three distinct insults. J Neurochem 1999; 73(5): 1901–12PubMed
81.
go back to reference Fanger GR, Gerwins P, Widmann C, et al. MEKKs, GCKs, MLKs, PAKs, TAKs, and tpls: upstream regulators of the c-Jun amino-terminal kinases? Curr Opin Genet Dev 1997; 7(1): 67–74PubMedCrossRef Fanger GR, Gerwins P, Widmann C, et al. MEKKs, GCKs, MLKs, PAKs, TAKs, and tpls: upstream regulators of the c-Jun amino-terminal kinases? Curr Opin Genet Dev 1997; 7(1): 67–74PubMedCrossRef
82.
go back to reference Gallo KA, Johnson GL. Mixed-lineage kinase control of JNK and p38 MAPK pathways. Nat Rev Mol Cell Biol 2002; 3(9): 663–72PubMedCrossRef Gallo KA, Johnson GL. Mixed-lineage kinase control of JNK and p38 MAPK pathways. Nat Rev Mol Cell Biol 2002; 3(9): 663–72PubMedCrossRef
83.
go back to reference Maroney AC, Finn JP, Connors TJ, et al. Cep-1347 (KT7515), a semisynthetic inhibitor of the mixed lineage kinase family. J Biol Chem 2001; 276(27): 25302–8PubMedCrossRef Maroney AC, Finn JP, Connors TJ, et al. Cep-1347 (KT7515), a semisynthetic inhibitor of the mixed lineage kinase family. J Biol Chem 2001; 276(27): 25302–8PubMedCrossRef
84.
go back to reference Murakata C, Kaneko M, Gessner G, et al. Mixed lineage kinase activity of indolocarbazole analogues. Bioorg Med Chem Lett 2002; 12(2): 147–50PubMedCrossRef Murakata C, Kaneko M, Gessner G, et al. Mixed lineage kinase activity of indolocarbazole analogues. Bioorg Med Chem Lett 2002; 12(2): 147–50PubMedCrossRef
85.
go back to reference Saporito MS, Brown EM, Miller MS, et al. CEP-1347/KT-7515, an inhibitor of c-jun N-terminal kinase activation, attenuates the 1-methyl-4-phenyl tetrahydropyridine-mediated loss of nigrostriatal dopaminergic neurons in vivo. J Pharmacol Exp Ther 1999; 288(2): 421–7PubMed Saporito MS, Brown EM, Miller MS, et al. CEP-1347/KT-7515, an inhibitor of c-jun N-terminal kinase activation, attenuates the 1-methyl-4-phenyl tetrahydropyridine-mediated loss of nigrostriatal dopaminergic neurons in vivo. J Pharmacol Exp Ther 1999; 288(2): 421–7PubMed
86.
go back to reference Saporito MS, Thomas BA, Scott RW. MPTP activates c-Jun NH (2)-terminal kinase (JNK) and its upstream regulatory kinase MKK4 in nigrostriatal neurons in vivo. J Neurochem 2000; 75(3): 1200–8PubMedCrossRef Saporito MS, Thomas BA, Scott RW. MPTP activates c-Jun NH (2)-terminal kinase (JNK) and its upstream regulatory kinase MKK4 in nigrostriatal neurons in vivo. J Neurochem 2000; 75(3): 1200–8PubMedCrossRef
87.
go back to reference Parkinson Study Group. The safety and tolerability of a mixed lineage kinase inhibitor (CEP-1347) in PD. Neurology 2004; 62(2): 330–2CrossRef Parkinson Study Group. The safety and tolerability of a mixed lineage kinase inhibitor (CEP-1347) in PD. Neurology 2004; 62(2): 330–2CrossRef
90.
go back to reference Lyons KE, Koller WC, Wilkinson SB, et al. Long term safety and efficacy of unilateral deep brain stimulation of the thalamus for parkinsonian tremor. J Neurol Neurosurg Psychiatry 2001; 71(5): 682–4PubMedCrossRef Lyons KE, Koller WC, Wilkinson SB, et al. Long term safety and efficacy of unilateral deep brain stimulation of the thalamus for parkinsonian tremor. J Neurol Neurosurg Psychiatry 2001; 71(5): 682–4PubMedCrossRef
91.
go back to reference Herzog J, Volkmann J, Krack P, et al. Two-year follow-up of subthalamic deep brain stimulation in Parkinson’s disease. Mov Disord 2003; 18(11): 1332–7PubMedCrossRef Herzog J, Volkmann J, Krack P, et al. Two-year follow-up of subthalamic deep brain stimulation in Parkinson’s disease. Mov Disord 2003; 18(11): 1332–7PubMedCrossRef
92.
go back to reference Luo J, Kaplitt M, Fitzsimons H, et al. Subthalamic GAD gene therapy in a Parkinson’s disease rat model. Science 2002; 298(5592): 425–9PubMedCrossRef Luo J, Kaplitt M, Fitzsimons H, et al. Subthalamic GAD gene therapy in a Parkinson’s disease rat model. Science 2002; 298(5592): 425–9PubMedCrossRef
93.
go back to reference During M, Kaplitt M, Stern M, et al. Subthalamic GAD gene transfer in Parkinson disease patients who are candidates for deep brain stimulation. Hum Gene Ther 2001; 12(12): 1589–91PubMed During M, Kaplitt M, Stern M, et al. Subthalamic GAD gene transfer in Parkinson disease patients who are candidates for deep brain stimulation. Hum Gene Ther 2001; 12(12): 1589–91PubMed
94.
go back to reference Grady D, Kolata G. Gene therapy used to treat patients with Parkinson’s. New York Times 2003 Aug 19 Grady D, Kolata G. Gene therapy used to treat patients with Parkinson’s. New York Times 2003 Aug 19
95.
go back to reference Kordower J. In vivo gene delivery of glial cell line-derived neurotrophic factor for Parkinson’s disease. Ann Neurol 2003; 53Suppl. 3: S120–34PubMedCrossRef Kordower J. In vivo gene delivery of glial cell line-derived neurotrophic factor for Parkinson’s disease. Ann Neurol 2003; 53Suppl. 3: S120–34PubMedCrossRef
96.
go back to reference Björklund A, Kirik D, Rosenblad C, et al. Towards a neuroprotective gene therapy for Parkinson’s disease: use of adenovirus, AAV and lentivirus vectors for gene transfer of GDNF to the nigrostriatal system in the rat Parkinson model. Brain Res 2000; 886: 82–98PubMedCrossRef Björklund A, Kirik D, Rosenblad C, et al. Towards a neuroprotective gene therapy for Parkinson’s disease: use of adenovirus, AAV and lentivirus vectors for gene transfer of GDNF to the nigrostriatal system in the rat Parkinson model. Brain Res 2000; 886: 82–98PubMedCrossRef
97.
go back to reference Kordower JH, Emborg ME, Bloch J, et al. Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson’s disease. Science 2000; 290: 767–73PubMedCrossRef Kordower JH, Emborg ME, Bloch J, et al. Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson’s disease. Science 2000; 290: 767–73PubMedCrossRef
98.
go back to reference Business wire. Update of neurologix phase I clinical trial of gene therapy for Parkinson’s disease. Presented at AANS annual meeting; 2005 Apr 16–21; New Orleans (LA) Business wire. Update of neurologix phase I clinical trial of gene therapy for Parkinson’s disease. Presented at AANS annual meeting; 2005 Apr 16–21; New Orleans (LA)
99.
go back to reference Federoff H. GAD Zooks! Excitement to inhibition in one easy step? Gene Ther 2003; 10: 365–6CrossRef Federoff H. GAD Zooks! Excitement to inhibition in one easy step? Gene Ther 2003; 10: 365–6CrossRef
100.
go back to reference Zurn A, Widmer H, Aebischer P. Sustained delivery of GDNF: towards a treatment for Parkinson’s disease. Brain Res Rev 2001; 36: 222–9PubMedCrossRef Zurn A, Widmer H, Aebischer P. Sustained delivery of GDNF: towards a treatment for Parkinson’s disease. Brain Res Rev 2001; 36: 222–9PubMedCrossRef
101.
go back to reference Lapchak P, Gash D, Jiao S, et al. Glial cell line-derived neurotrophic factor: a novel therapeutic approach to treat motor dysfunction in Parkinson’s disease. Exp Neurol 1997; 144: 29–34PubMedCrossRef Lapchak P, Gash D, Jiao S, et al. Glial cell line-derived neurotrophic factor: a novel therapeutic approach to treat motor dysfunction in Parkinson’s disease. Exp Neurol 1997; 144: 29–34PubMedCrossRef
102.
go back to reference Grondin R, Gash D. Glial cell line-derived neurotrophic factor (GDNF): a drug candidate for the treatment of Parkinson’s disease. J Neurol 1998; 245Suppl. 3: P35–42PubMedCrossRef Grondin R, Gash D. Glial cell line-derived neurotrophic factor (GDNF): a drug candidate for the treatment of Parkinson’s disease. J Neurol 1998; 245Suppl. 3: P35–42PubMedCrossRef
103.
go back to reference Hou J, Lin L, Mytilinenou C. Glial cell line-derived neurotrophic factor exerts neurotrophic effects on dopaminergic neurons in vitro and promotes their survival and regrowth after damage by 1-methyl-4-phenylpyridinium. J Neurochem 1996; 66: 74–82PubMedCrossRef Hou J, Lin L, Mytilinenou C. Glial cell line-derived neurotrophic factor exerts neurotrophic effects on dopaminergic neurons in vitro and promotes their survival and regrowth after damage by 1-methyl-4-phenylpyridinium. J Neurochem 1996; 66: 74–82PubMedCrossRef
104.
go back to reference Hudson J, Granholm A-C, Gerhardt G, et al. Glial cell line-derived neurotrophic factor augments midbrain dopaminergic circuits in vivo. Brain Res Bull 1995; 36: 425–32PubMedCrossRef Hudson J, Granholm A-C, Gerhardt G, et al. Glial cell line-derived neurotrophic factor augments midbrain dopaminergic circuits in vivo. Brain Res Bull 1995; 36: 425–32PubMedCrossRef
105.
go back to reference Kearns C, Gash DM. GDNF protects nigral dopamine neurons against 6-hydroxydopamine in vivo. Brain Res 1995; 672: 104–11PubMedCrossRef Kearns C, Gash DM. GDNF protects nigral dopamine neurons against 6-hydroxydopamine in vivo. Brain Res 1995; 672: 104–11PubMedCrossRef
106.
go back to reference Kearns C, Cass WA, Smoot K, et al. GDNF protection against 6-OHDA: time dependence and requirement for protein synthesis. J Neurosci 1997; 17: 7111–8PubMed Kearns C, Cass WA, Smoot K, et al. GDNF protection against 6-OHDA: time dependence and requirement for protein synthesis. J Neurosci 1997; 17: 7111–8PubMed
107.
go back to reference Tomac A, Lindqvist E, Lin L-FH, et al. Protection and repair of the nigrostriatal dopaminergic system by GDNF in vivo. Nature 1995; 373: 335–9PubMedCrossRef Tomac A, Lindqvist E, Lin L-FH, et al. Protection and repair of the nigrostriatal dopaminergic system by GDNF in vivo. Nature 1995; 373: 335–9PubMedCrossRef
108.
go back to reference Hoffer BJ, Hoffman A, Bowenkamp K, et al. Glial cell line-derived neurotrophic factor reverses toxin-induced injury to midbrain dopaminergic neurons in vivo. Neurosci Lett 1994; 182: 107–11PubMedCrossRef Hoffer BJ, Hoffman A, Bowenkamp K, et al. Glial cell line-derived neurotrophic factor reverses toxin-induced injury to midbrain dopaminergic neurons in vivo. Neurosci Lett 1994; 182: 107–11PubMedCrossRef
109.
go back to reference Lapchak P, Miller P, Collins F, et al. Glial cell line-derived neurotrophic factor attenuates behavioural deficits and regulates nigrostriatal dopaminergic and peptidergic markers in 6-hydroxydopamine-lesioned adult rats: comparison of intraventricular and intranigral delivery. Neuroscience 1997; 78(1): 61–72PubMedCrossRef Lapchak P, Miller P, Collins F, et al. Glial cell line-derived neurotrophic factor attenuates behavioural deficits and regulates nigrostriatal dopaminergic and peptidergic markers in 6-hydroxydopamine-lesioned adult rats: comparison of intraventricular and intranigral delivery. Neuroscience 1997; 78(1): 61–72PubMedCrossRef
110.
go back to reference Gash D, Zhang Z, Ovadia A, et al. Functional recovery in parkinsonian monkeys treated with GDNF. Nature 1996; 380: 252–5PubMedCrossRef Gash D, Zhang Z, Ovadia A, et al. Functional recovery in parkinsonian monkeys treated with GDNF. Nature 1996; 380: 252–5PubMedCrossRef
111.
go back to reference Zhang Z, Miyoshi Y, Lapchak P, et al. Dose response to intraventricular glial cell line-derived neurotrophic factor administration in parkinsonian monkeys. J Pharmacol Exp Ther 1997; 282: 1396–401PubMed Zhang Z, Miyoshi Y, Lapchak P, et al. Dose response to intraventricular glial cell line-derived neurotrophic factor administration in parkinsonian monkeys. J Pharmacol Exp Ther 1997; 282: 1396–401PubMed
112.
go back to reference Miyoshi Y, Zhang Z, Ovadia A, et al. Glial cell line-derived neurotrophic factor-levodopa interactions and reduction of side effects in parkinsonian monkeys. Ann Neurol 1997; 42: 208–14PubMedCrossRef Miyoshi Y, Zhang Z, Ovadia A, et al. Glial cell line-derived neurotrophic factor-levodopa interactions and reduction of side effects in parkinsonian monkeys. Ann Neurol 1997; 42: 208–14PubMedCrossRef
113.
go back to reference Grondin R, Zhang Z, Yi A, et al. Chronic, controlled GDNF infusion promotes structural and functional recovery in advanced parkinsonian monkeys. Brain 2002; 125: 2191–201PubMedCrossRef Grondin R, Zhang Z, Yi A, et al. Chronic, controlled GDNF infusion promotes structural and functional recovery in advanced parkinsonian monkeys. Brain 2002; 125: 2191–201PubMedCrossRef
114.
go back to reference Nutt J, Burchiel K, Comella C, et al. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in Parkinson’s disease. Neurology 2003; 60: 69–73PubMedCrossRef Nutt J, Burchiel K, Comella C, et al. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in Parkinson’s disease. Neurology 2003; 60: 69–73PubMedCrossRef
115.
go back to reference Kordower J, Palfi S, Chen E, et al. Clinicopathological findings following intraventricular glial-derived neurotrophic factor treatment in a patient with Parkinson’s disease. Ann Neurol 1999; 47(3): 419–24CrossRef Kordower J, Palfi S, Chen E, et al. Clinicopathological findings following intraventricular glial-derived neurotrophic factor treatment in a patient with Parkinson’s disease. Ann Neurol 1999; 47(3): 419–24CrossRef
116.
go back to reference Gill S, Patel N, Hotton G, et al. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson’s disease. Nat Med 2003; 9(5): 589–95PubMedCrossRef Gill S, Patel N, Hotton G, et al. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson’s disease. Nat Med 2003; 9(5): 589–95PubMedCrossRef
117.
go back to reference Wood MJ, Charlton HM, Wood KJ, et al. Immune responses to adenovius vectors in the nervous system. Trends Neurosci 1996; 19: 497–501PubMedCrossRef Wood MJ, Charlton HM, Wood KJ, et al. Immune responses to adenovius vectors in the nervous system. Trends Neurosci 1996; 19: 497–501PubMedCrossRef
118.
go back to reference Kajiwara K, Byrnes AP, Charlton HM, et al. Immune responses to adenoviral vectors during gene transfer in the brain. Hum Gene Ther 1997; 8: 253–65PubMedCrossRef Kajiwara K, Byrnes AP, Charlton HM, et al. Immune responses to adenoviral vectors during gene transfer in the brain. Hum Gene Ther 1997; 8: 253–65PubMedCrossRef
119.
go back to reference Rosenbald C, Gronborg M, Hansen C, et al. In vivo protection of nigral dopmaine neurons by lentiviral gene transfer of the novel GDNF-family member Neublastin/Artemin. Mol Cell Neurosci 2000; 15: 199–214CrossRef Rosenbald C, Gronborg M, Hansen C, et al. In vivo protection of nigral dopmaine neurons by lentiviral gene transfer of the novel GDNF-family member Neublastin/Artemin. Mol Cell Neurosci 2000; 15: 199–214CrossRef
120.
go back to reference Mandel RJ, Snyder RO, Leff SE. Recombinant adeno-associated viral vector-mediated glial cell line-derived neurotrophic factor gene transfer protects nigral dopamine neurons after onset of progressive degeneration in a rat model of Parkinson’s disease. Exp Neurol 1999; 160: 205–14PubMedCrossRef Mandel RJ, Snyder RO, Leff SE. Recombinant adeno-associated viral vector-mediated glial cell line-derived neurotrophic factor gene transfer protects nigral dopamine neurons after onset of progressive degeneration in a rat model of Parkinson’s disease. Exp Neurol 1999; 160: 205–14PubMedCrossRef
121.
go back to reference Mandel RJ, Spratt SK, Snyder RO, et al. Midbrain injection of recombinant adeno-associated virus encoding rat glial cell line-derived neurotrophic factor protects nigral neurons in a progressive 6-hydroxydopamine-induced degeneration model of Parkinson’s disease in rats. Proc Natl Acad Sci U S A 1997; 94: 14083–8PubMedCrossRef Mandel RJ, Spratt SK, Snyder RO, et al. Midbrain injection of recombinant adeno-associated virus encoding rat glial cell line-derived neurotrophic factor protects nigral neurons in a progressive 6-hydroxydopamine-induced degeneration model of Parkinson’s disease in rats. Proc Natl Acad Sci U S A 1997; 94: 14083–8PubMedCrossRef
122.
go back to reference Kirik D, Rosenbald C, Björklund A, et al. Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson’s model: intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system. J Neurosci 2000; 20: 4686–700PubMed Kirik D, Rosenbald C, Björklund A, et al. Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson’s model: intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system. J Neurosci 2000; 20: 4686–700PubMed
123.
go back to reference Emborg M, Ma S, Mufson E, et al. Age-related declines in nigral neuronal function correlate with motor impairments in Rhesus monkeys. J Comp Neurol 1998; 401: 253–63PubMedCrossRef Emborg M, Ma S, Mufson E, et al. Age-related declines in nigral neuronal function correlate with motor impairments in Rhesus monkeys. J Comp Neurol 1998; 401: 253–63PubMedCrossRef
124.
go back to reference Bowenkamp K, David D, Lapchak P, et al. 6-hydroxydopamine induces the loss of the dopaminergic phenotype in substantia nigra neurons of the rat: a possible mechanism for restoration of the nigrostriatal circuit mediated by glial cell line-derived neurotrophic factor. Exp Brain Res 1996; 111: 1–7PubMedCrossRef Bowenkamp K, David D, Lapchak P, et al. 6-hydroxydopamine induces the loss of the dopaminergic phenotype in substantia nigra neurons of the rat: a possible mechanism for restoration of the nigrostriatal circuit mediated by glial cell line-derived neurotrophic factor. Exp Brain Res 1996; 111: 1–7PubMedCrossRef
125.
go back to reference Georgievska B, Kirik D, Björklund A. Aberrant sprouting and downregulation of tyrosine hydroxylase in lesioned nigrostriatal dopamine neurons induced by long-lasting overexpression of glial cell line derived neurotrophic factor in the striatum by lentiviral gene transfer. Exp Neurol 2002; 177: 461–74PubMedCrossRef Georgievska B, Kirik D, Björklund A. Aberrant sprouting and downregulation of tyrosine hydroxylase in lesioned nigrostriatal dopamine neurons induced by long-lasting overexpression of glial cell line derived neurotrophic factor in the striatum by lentiviral gene transfer. Exp Neurol 2002; 177: 461–74PubMedCrossRef
127.
go back to reference Lindner MD, Winn SR, Baetge EE, et al. Implantation of encapsulated catecholamine and GDNF-producing cells in rats with unilateral dopamine depletions and parkinsonian symptoms. Exp Neurol 1995; 132: 62–76PubMedCrossRef Lindner MD, Winn SR, Baetge EE, et al. Implantation of encapsulated catecholamine and GDNF-producing cells in rats with unilateral dopamine depletions and parkinsonian symptoms. Exp Neurol 1995; 132: 62–76PubMedCrossRef
128.
go back to reference Tseng JL, Baetge EE, Zurn AD, et al. GDNF reduces drug-induced rotational behavior after medial forebrain bundle transection by a mechanism not involving striatal dopamine. J Neurosci 1997; 1: 325–33 Tseng JL, Baetge EE, Zurn AD, et al. GDNF reduces drug-induced rotational behavior after medial forebrain bundle transection by a mechanism not involving striatal dopamine. J Neurosci 1997; 1: 325–33
129.
go back to reference Akerud P, Canals JM, Snyder EY, et al. Neuroprotection through delivery of glial cell-lined derived neurotrophic factor by neural stem cells in a mouse model of Parkinson’s disease. J Neurosci 2001; 21: 8108–18PubMed Akerud P, Canals JM, Snyder EY, et al. Neuroprotection through delivery of glial cell-lined derived neurotrophic factor by neural stem cells in a mouse model of Parkinson’s disease. J Neurosci 2001; 21: 8108–18PubMed
130.
go back to reference Freed C, Greene P, Breeze R, et al. Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. N Eng J Med 2001; 344(10): 710-9CrossRef Freed C, Greene P, Breeze R, et al. Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. N Eng J Med 2001; 344(10): 710-9CrossRef
Metadata
Title
Treatment of Parkinson’s Disease
What’s on the Horizon?
Authors
Dr Stacy S. Wu
Steven J. Frucht
Publication date
01-09-2005
Publisher
Springer International Publishing
Published in
CNS Drugs / Issue 9/2005
Print ISSN: 1172-7047
Electronic ISSN: 1179-1934
DOI
https://doi.org/10.2165/00023210-200519090-00001

Other articles of this Issue 9/2005

CNS Drugs 9/2005 Go to the issue

Adis Drug Evaluation

Modafinil