Skip to main content
Log in

Treatment of Parkinson’s Disease

What’s on the Horizon?

  • Leading Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

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.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Mouradian M. Recent advances in the genetics and pathogenesis of Parkinson’s disease. Neurology 2002; 58(2): 179–85

    Article  PubMed  Google Scholar 

  2. 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: 301–6

    Article  PubMed  CAS  Google Scholar 

  3. Parkinson Study Group. Entacapone improves motor fluctuations in levodopa-treated Parkinson’s disease patients. Ann Neurol 1997; 42(5): 747–55

    Article  Google Scholar 

  4. Rinne UK, Larsen JP, Siden A, et al. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations. Neurology 1998; 51: 1309–14

    Article  PubMed  CAS  Google Scholar 

  5. 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: 283–96

    Article  PubMed  CAS  Google Scholar 

  6. Jenner P. A2A antagonists as novel nondopaminergic therapy for motor dysfunction in PD. Neurology 2003; 61Suppl. 6: S33–8

    Google Scholar 

  7. Mori A, Shindou T. Modulation of GABAergic transmission in the striatopallidal system by adenosine A2A antagonists. Neurology 2003; 61Suppl. 6: S44–8

    Article  PubMed  CAS  Google Scholar 

  8. Penney Jr J, Young A. Striatal in homogeneities and basal ganglia origin. Mov Disord 1986; 1: 3–15

    Article  PubMed  Google Scholar 

  9. Bara-Jimenez W, Sherzai A, Dimitrova T, et al. Adenosine A2A receptor antagonist treatment of Parkinson’s disease. Neurology 2003; 61: 293–6

    Article  PubMed  CAS  Google Scholar 

  10. Chase T, Oh J. Striatal mechanisms and pathogenesis of parkinsonian signs and motor complications. Ann Neurol 2000; 27Suppl. 1: S122–9

    Google Scholar 

  11. Schiffmann S, Vanderhaeghen J. Adenosine A2A receptors regulate the gene expression of striatopallidal and striatonigral neurons. J Neurosci 1993; 13: 1080–7

    PubMed  CAS  Google Scholar 

  12. 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: 186–95

    Article  PubMed  CAS  Google Scholar 

  13. Franco R, Ferre S, Agnati L, et al. Evidence for adenosine/dopamine receptor interactions: indications for heteromerization. Neuropsychopharmacology 2000; 23: S50–9

    Article  PubMed  CAS  Google Scholar 

  14. Feigin A. Nondopaminergic symptomatic therapies for Parkinson’s disease. Neurology 2003; 61: 286–7

    Article  PubMed  Google Scholar 

  15. 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: S39–42

    Article  PubMed  CAS  Google Scholar 

  16. 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: 507–13

    Article  PubMed  CAS  Google Scholar 

  17. Hauser R, Hubble J, Truong D, et al. Randomized trial of the adenosine A2A receptor antagonist istradefylline in advanced PD. Neurology 2003; 61: 297–303

    Article  PubMed  CAS  Google Scholar 

  18. Ruottinen HM, Rinne UK. COMT inhibition in the treatment of Parkinson’s disease. J Neurol 1998; 245Suppl. 3: 25–34

    Article  Google Scholar 

  19. 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: 321–7

    Article  PubMed  CAS  Google Scholar 

  20. 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: 1673–7

    Article  PubMed  CAS  Google Scholar 

  21. Frucht S, Greene P, Fahn S. Sleep episodes in Parkinson’s disease: a wake-up call. Mov Disord 2000; 15: 601–3

    Article  PubMed  CAS  Google Scholar 

  22. 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: 1908–10

    Article  PubMed  CAS  Google Scholar 

  23. Paus S, Brecht H, Köster J, et al. Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s disease. Mov Disord 2003; 18: 659–67

    Article  PubMed  Google Scholar 

  24. Shaunak S, Wilkins A, Pillig J, et al. Pericardial, retroperitoneal, and pleural fibrosis induced by pergolide. J Neurol Neurosurg Psychiatry 1999; 66: 79–81

    Article  PubMed  CAS  Google Scholar 

  25. 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: S55–61

    Article  PubMed  CAS  Google Scholar 

  26. Pedata F, Pugliese A, Melani A, et al. A2A receptors in neuroprotection of dopaminergic neurons. Neurology 2003; 61Suppl. 6: S49–50

    Article  PubMed  Google Scholar 

  27. 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: 276–84

    Article  PubMed  Google Scholar 

  28. 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: 28–37

    Article  PubMed  CAS  Google Scholar 

  29. Ascherio A, Chen H. Caffeinated clues from epidemiology of Parkinson’s disease. Neurology 2001; 61Suppl. 6: S51–4

    Google Scholar 

  30. 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): 176–83

    Article  Google Scholar 

  31. Ward C. Does selegiline delay progression of Parkinson’s disease? A critical re-evaluation of the DATATOP study. J Neurol Neurosurg Psychiatry 1994; 57: 217–20

    Article  PubMed  CAS  Google Scholar 

  32. Fahn S. Controversies in the therapy of Parkinson’s disease. Adv Neurol 1996; 69: 477–86

    PubMed  CAS  Google Scholar 

  33. Chase TN, Oh JD, Blanchet PJ. Neostriatal mechanisms in Parkinson’s disease. Neurology 1998; 51(2 Suppl. 2): S30–5

    Article  PubMed  CAS  Google Scholar 

  34. Chase TN, Engber TM, Mouradian MM. Contribution of dopaminergic and glutamatergic mechanisms to the pathogenesis of motor response complications in Parkinson’s disease. Adv Neurol 1996; 69: 497–501

    PubMed  CAS  Google Scholar 

  35. 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): 404–16

    Article  PubMed  CAS  Google Scholar 

  36. 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): 171–6

    Article  PubMed  CAS  Google Scholar 

  37. 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: 65–8

    PubMed  Google Scholar 

  38. Baas H. Dyskinesia in Parkinson’s disease: pathophysiology and clinical risk factors. J Neurol 2000; 247Suppl. 4: IV/12–6

    Google Scholar 

  39. 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): 150–9

    Article  PubMed  CAS  Google Scholar 

  40. 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): 433–42

    Article  PubMed  CAS  Google Scholar 

  41. Chase TN. Levodopa therapy: consequences of the nonphysiologic replacement of dopamine. Neurology 1998; 50(5 Suppl. 5): S17–25

    Article  PubMed  CAS  Google Scholar 

  42. Blanchet PJ, Konitsiotis S, Chase TN. Amantadine reduces levodopa-induced dyskinesias in parkinsonian monkeys. Mov Disord 1998; 13(5): 798–802

    Article  PubMed  CAS  Google Scholar 

  43. Papa SM, Chase TN. Levodopa-induced dyskinesias improved by a glutamate antagonist in Parkinsonian monkeys. Ann Neurol 1996; 39(5): 574–8

    Article  PubMed  CAS  Google Scholar 

  44. 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): 1323–6

    Article  PubMed  CAS  Google Scholar 

  45. Danysz W, Parsons CG, Kornhuber J, et al. Aminoadamantanes as NMDA receptor antagonists and antiparkinsonian agents: preclinical studies. Neurosci Biobehav Rev 1997; 21(4): 455–68

    Article  PubMed  CAS  Google Scholar 

  46. Konitsiotis S, Blanchet PJ, Verhagen L, et al. AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys. Neurology 2000; 54(8): 1589–95

    Article  PubMed  CAS  Google Scholar 

  47. 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): 381–91

    Article  PubMed  Google Scholar 

  48. 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): 27–39

    Article  PubMed  CAS  Google Scholar 

  49. 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): 203–13

    Article  PubMed  Google Scholar 

  50. 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): 179–82

    Article  PubMed  CAS  Google Scholar 

  51. 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): 267–74

    Article  PubMed  CAS  Google Scholar 

  52. 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): 40–7

    Article  PubMed  CAS  Google Scholar 

  53. Young AB, Fagg GE. Excitatory amino acid receptors in the brain: membrane binding and receptor autoradiographic approaches. Trends Pharmacol Sci 1990; 11(3): 126–33

    Article  PubMed  CAS  Google Scholar 

  54. 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): 35–48

    Article  PubMed  CAS  Google Scholar 

  55. 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): 819–33

    PubMed  CAS  Google Scholar 

  56. Ozawa S, Kamiya H, Tsuzuki K. Glutamate receptors in the mammalian central nervous system. Prog Neurobiol 1998; 54(5): 581–618

    Article  PubMed  CAS  Google Scholar 

  57. 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): 4748–54

    PubMed  CAS  Google Scholar 

  58. 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): 3269–74

    Article  PubMed  CAS  Google Scholar 

  59. 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): 571–4

    Article  PubMed  CAS  Google Scholar 

  60. Johnson SA, Luu NT, Herbst TA, et al. Synergistic interactions between ampakines and antipsychotic drugs. J Pharmacol Exp Ther 1999; 289(1): 392–7

    PubMed  CAS  Google Scholar 

  61. 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): 1681–8

    Article  PubMed  CAS  Google Scholar 

  62. 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): 1689–702

    Article  PubMed  CAS  Google Scholar 

  63. 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): 1183–6

    Article  Google Scholar 

  64. 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

    Google Scholar 

  65. Thatte U. NS-2330 (Neurosearch). Curr Opin Investig Drugs 2001; 2(11): 1592–4

    PubMed  CAS  Google Scholar 

  66. A fourteen-week placebo-controlled dose-response efficacy and safety study of NS 2330 in early Parkinson’s disease patients (study for proof of concept in early Parkinson’s disease of a triple reuptake inhibitor, NS 2330 / SCEPTRE) [online]. Available from URL: http://health.uchc.edu/clinical-trials/trials.asp?Therap=Neurology&SubTherap=Parkinson@s+Disease. [Accessed 2004 Aug 14]

  67. Tran M, Mitchell T, Kennedy D, et al. Role of coenzyme Q10 in chronic heart failure, angina, and hypertension. Pharmacotherapy 2001; 7: 797–806

    Article  Google Scholar 

  68. Huntington Study Group. A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington’s disease. Neurology 2001; 57(3): 397–404

    Google Scholar 

  69. Przedborski S, Jackson-Lewis V. Mechanisms of MPTP toxicity. Mov Disord 1998; 13Suppl. 1: 35–8

    PubMed  Google Scholar 

  70. Krige D, Carroll M, Cooper J, et al. Platelet mitochondrial dysfunction in Parkinson’s disease. Ann Neurol 1992; 32: 782–8

    Article  PubMed  CAS  Google Scholar 

  71. Parker Jr W, Boyson S, Parks J. Abnormalities of the electron transport chain in idiopathic Parkinson’s disease. Ann Neurol 1989; 26: 719–23

    Article  PubMed  Google Scholar 

  72. Turner C, Schapira A. Mitochondrial dysfunction in neurodegenerative disorders and ageing. Adv Exp Med Biol 2001; 487: 229–51

    Article  PubMed  CAS  Google Scholar 

  73. 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: 201–4

    Article  PubMed  CAS  Google Scholar 

  74. 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: 109–14

    Article  PubMed  CAS  Google Scholar 

  75. 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: 8892–7

    Article  PubMed  CAS  Google Scholar 

  76. 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: 793–5

    Article  PubMed  CAS  Google Scholar 

  77. Shults C, Oakes D, Kieburtz K, et al. Effects of coenzyme Q10 in early Parkinson disease. Arch Neurol 2002; 59: 1541–50

    Article  PubMed  Google Scholar 

  78. Hirsch EC, Hunot S, Faucheux B, et al. Dopaminergic neurons degenerate by apoptosis in Parkinson’s disease. Mov Disord 1999; 14(2): 383–5

    Article  PubMed  CAS  Google Scholar 

  79. 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): 2875–80

    Article  PubMed  CAS  Google Scholar 

  80. 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): 1901–12

    PubMed  CAS  Google Scholar 

  81. 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): 67–74

    Article  PubMed  CAS  Google Scholar 

  82. Gallo KA, Johnson GL. Mixed-lineage kinase control of JNK and p38 MAPK pathways. Nat Rev Mol Cell Biol 2002; 3(9): 663–72

    Article  PubMed  CAS  Google Scholar 

  83. 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): 25302–8

    Article  PubMed  CAS  Google Scholar 

  84. Murakata C, Kaneko M, Gessner G, et al. Mixed lineage kinase activity of indolocarbazole analogues. Bioorg Med Chem Lett 2002; 12(2): 147–50

    Article  PubMed  CAS  Google Scholar 

  85. 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): 421–7

    PubMed  CAS  Google Scholar 

  86. 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): 1200–8

    Article  PubMed  CAS  Google Scholar 

  87. Parkinson Study Group. The safety and tolerability of a mixed lineage kinase inhibitor (CEP-1347) in PD. Neurology 2004; 62(2): 330–2

    Article  CAS  Google Scholar 

  88. PRECEPT: Parkinson Research Examination of CEP-1347 Trial [online]. Available from URL: http://www.parkinson-study-group.org. [Accessed 2004 Aug 14]

  89. PRECEPT [online]. Available from URL: http://bidmc.harvard.edu/display.asp?.node_id=1214. [Accessed 2004 Aug 14]

  90. 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): 682–4

    Article  PubMed  CAS  Google Scholar 

  91. 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): 1332–7

    Article  PubMed  Google Scholar 

  92. Luo J, Kaplitt M, Fitzsimons H, et al. Subthalamic GAD gene therapy in a Parkinson’s disease rat model. Science 2002; 298(5592): 425–9

    Article  PubMed  CAS  Google Scholar 

  93. 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): 1589–91

    PubMed  CAS  Google Scholar 

  94. Grady D, Kolata G. Gene therapy used to treat patients with Parkinson’s. New York Times 2003 Aug 19

  95. Kordower J. In vivo gene delivery of glial cell line-derived neurotrophic factor for Parkinson’s disease. Ann Neurol 2003; 53Suppl. 3: S120–34

    Article  PubMed  CAS  Google Scholar 

  96. 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: 82–98

    Article  PubMed  Google Scholar 

  97. 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: 767–73

    Article  PubMed  CAS  Google Scholar 

  98. 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. Federoff H. GAD Zooks! Excitement to inhibition in one easy step? Gene Ther 2003; 10: 365–6

    Article  CAS  Google Scholar 

  100. Zurn A, Widmer H, Aebischer P. Sustained delivery of GDNF: towards a treatment for Parkinson’s disease. Brain Res Rev 2001; 36: 222–9

    Article  PubMed  CAS  Google Scholar 

  101. 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: 29–34

    Article  PubMed  CAS  Google Scholar 

  102. 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: P35–42

    Article  PubMed  CAS  Google Scholar 

  103. 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: 74–82

    Article  PubMed  CAS  Google Scholar 

  104. 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: 425–32

    Article  PubMed  CAS  Google Scholar 

  105. Kearns C, Gash DM. GDNF protects nigral dopamine neurons against 6-hydroxydopamine in vivo. Brain Res 1995; 672: 104–11

    Article  PubMed  CAS  Google Scholar 

  106. Kearns C, Cass WA, Smoot K, et al. GDNF protection against 6-OHDA: time dependence and requirement for protein synthesis. J Neurosci 1997; 17: 7111–8

    PubMed  CAS  Google Scholar 

  107. Tomac A, Lindqvist E, Lin L-FH, et al. Protection and repair of the nigrostriatal dopaminergic system by GDNF in vivo. Nature 1995; 373: 335–9

    Article  PubMed  CAS  Google Scholar 

  108. 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: 107–11

    Article  PubMed  CAS  Google Scholar 

  109. 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): 61–72

    Article  PubMed  CAS  Google Scholar 

  110. Gash D, Zhang Z, Ovadia A, et al. Functional recovery in parkinsonian monkeys treated with GDNF. Nature 1996; 380: 252–5

    Article  PubMed  CAS  Google Scholar 

  111. 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: 1396–401

    PubMed  CAS  Google Scholar 

  112. 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: 208–14

    Article  PubMed  CAS  Google Scholar 

  113. Grondin R, Zhang Z, Yi A, et al. Chronic, controlled GDNF infusion promotes structural and functional recovery in advanced parkinsonian monkeys. Brain 2002; 125: 2191–201

    Article  PubMed  Google Scholar 

  114. 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: 69–73

    Article  PubMed  CAS  Google Scholar 

  115. 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): 419–24

    Article  Google Scholar 

  116. 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): 589–95

    Article  PubMed  CAS  Google Scholar 

  117. Wood MJ, Charlton HM, Wood KJ, et al. Immune responses to adenovius vectors in the nervous system. Trends Neurosci 1996; 19: 497–501

    Article  PubMed  CAS  Google Scholar 

  118. Kajiwara K, Byrnes AP, Charlton HM, et al. Immune responses to adenoviral vectors during gene transfer in the brain. Hum Gene Ther 1997; 8: 253–65

    Article  PubMed  CAS  Google Scholar 

  119. 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: 199–214

    Article  CAS  Google Scholar 

  120. 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: 205–14

    Article  PubMed  CAS  Google Scholar 

  121. 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: 14083–8

    Article  PubMed  CAS  Google Scholar 

  122. 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: 4686–700

    PubMed  CAS  Google Scholar 

  123. 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: 253–63

    Article  PubMed  CAS  Google Scholar 

  124. 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: 1–7

    Article  PubMed  CAS  Google Scholar 

  125. 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: 461–74

    Article  PubMed  CAS  Google Scholar 

  126. Lysaght MJ, Aebischer P. Encapsulated cells as therapy. Sci Am 1999; 280: 76–82

    Article  PubMed  CAS  Google Scholar 

  127. 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: 62–76

    Article  PubMed  CAS  Google Scholar 

  128. 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

    Google Scholar 

  129. 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: 8108–18

    PubMed  CAS  Google Scholar 

  130. 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): 710-9

    Article  CAS  Google Scholar 

Download references

Acknowledgements

No sources of funding were used to assist in the preparation of this review. The author has no conflicts of interest that are directly relevant to the content of this review.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Stacy S. Wu.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wu, S.S., Frucht, S.J. Treatment of Parkinson’s Disease. CNS Drugs 19, 723–743 (2005). https://doi.org/10.2165/00023210-200519090-00001

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00023210-200519090-00001

Keywords

Navigation