Skip to main content
Log in

Disease Modification in Parkinson’s Disease

  • Leading Article
  • Published:
Drugs & Aging Aims and scope Submit manuscript

Abstract

Parkinson’s disease (PD) is an age-related, progressive, multisystem neurodegenerative disorder resulting in significant morbidity and mortality, as well as a growing social and financial burden in an aging population. The hallmark of PD is loss of dopaminergic neurons of the substantia nigra pars compacta, leading to bradykinesia, rigidity and tremor. Current pharmacological treatment is therefore centred upon dopamine replacement to alleviate symptoms. However, two major problems complicate this approach: (i) motor symptoms continue to progress, requiring increasing doses of medication, which result in both short-term adverse effects and intermediate- to long-term motor complications; (ii) dopamine replacement does little to treat non-dopaminergic motor and non-motor symptoms, which are an important source of morbidity, including dementia, sleep disturbances, depression, orthostatic hypotension, and postural instability leading to falls. It is critical, therefore, to develop a broader and more fundamental therapeutic approach to PD, and major research efforts have focused upon developing neuroprotective interventions.

Despite many encouraging preclinical data suggesting the possibility of addressing the underlying pathophysiology by slowing cell loss, efforts to translate this into the clinical realm have largely proved disappointing in the past. Barriers to finding neuroprotective or disease-modifying drugs in PD include a lack of validated biomarkers of progression, which hampers clinical trial design and interpretation; difficulties separating symptomatic and neuroprotective effects of candidate neuroprotective therapies; and possibly fundamental flaws in some of the basic preclinical models and testing.

However, three recent clinical trials have used a novel delayed-start design in an attempt to overcome some of these roadblocks. While not examining markers of cell loss and function, which would determine neuroprotective effects, this trial design pragmatically tests whether earlier versus later intervention is beneficial. If positive (i.e. if an earlier intervention proves more effective), this demonstrates disease modification, which could result from neuroprotection or from other mechanisms. This strategy therefore provides a first step towards supporting neuroprotection in PD. Of the three delayed-start design clinical trials, two have investigated early versus later start of rasagiline, a specific irreversible monoamine oxidase B inhibitor. Each trial has supported, although not proven, disease-modifying effects. A third delayed-start-design clinical trial examining potential disease-modifying effects of pramipexole has unfortunately reportedly been negative according to preliminary presentations. The suggestion that rasagiline is disease modifying is made all the more compelling by in vitro and PD animal-model studies in which rasagiline was shown to have neuroprotective effects.

In this review, we examine efforts to demonstrate neuroprotection in PD to date, describe ongoing neuroprotection trials, and critically discuss the results of the most recent delayed-start clinical trials that test possible disease-modifying activities of rasagiline and pramipexole in PD.

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.

Institutional subscriptions

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Fahn S, Elton RL, UPDRS program members. Unified Parkinson’s Disease Rating Scale. Florham Park (NJ): Macmillan Healthcare Information, 1987

    Google Scholar 

  2. Fahn S, Oakes D, Shoulson I, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med 2004; 351: 2498–508

    Article  PubMed  CAS  Google Scholar 

  3. Goetz CG, Tilley BC, Shaftman SR, et al. Movement Disorder Society-sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord 2008; 23: 2129–70

    Article  PubMed  Google Scholar 

  4. Hart RG, Pearce LA, Ravina BM, et al. Neuroprotection trials in Parkinson’s disease: systematic review. Mov Disord 2009; 24: 647–54

    Article  PubMed  Google Scholar 

  5. Shults CW, Oakes D, Kieburtz K, et al. Effects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol 2002; 59: 1541–50

    Article  PubMed  Google Scholar 

  6. Castagnoli K, Palmer S, Castagnoli Jr N. Neuroprotection by (R)-deprenyl and 7-nitroindazole in the MPTP C57BL/6 mouse model of neurotoxicity. Neurobiology (Bp) 1999; 7: 135–49

    CAS  Google Scholar 

  7. Muralikrishnan D, Samantaray S, Mohanakumar KP. D-deprenyl protects nigrostriatal neurons against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurotoxicity. Synapse 2003; 50: 7–13

    Article  PubMed  CAS  Google Scholar 

  8. Miyake Y, Fukushima W, Tanaka K, et al. Dietary intake of antioxidant vitamins and risk of Parkinson’s disease: a case-control study in Japan. Eur J Neurol 2011; 18: 106–13

    Article  PubMed  CAS  Google Scholar 

  9. Etminan M, Gill SS, Samii A. Intake of vitamin E, vitamin C, and carotenoids and the risk of Parkinson’s disease: a meta-analysis. Lancet Neurol 2005; 4: 362–5

    Article  PubMed  CAS  Google Scholar 

  10. Birkmayer W, Knoll J, Riederer P, et al. Increased life expectancy resulting from addition of L-deprenyl to Mado-par treatment in Parkinson’s disease: a long term study. J Neural Transm 1985; 64: 113–27

    Article  PubMed  CAS  Google Scholar 

  11. Tetrud JW, Langston JW. The effect of deprenyl (selegiline) on the natural history of Parkinson’s disease. Science 1989; 245: 519–22

    Article  PubMed  CAS  Google Scholar 

  12. 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: 176–83

  13. Shoulson I, Oakes D, Fahn S, et al. Impact of sustained deprenyl (selegiline) in levodopa-treated Parkinson’s disease: a randomized placebo-controlled extension of the deprenyl and tocopherol antioxidative therapy of parkin-sonism trial. Ann Neurol 2002; 51: 604–12

    Article  PubMed  CAS  Google Scholar 

  14. Palhagen S, Heinonen EH, Hagglund J, et al. Selegiline delays the onset of disability in de novo parkinsonian patients. Swedish Parkinson Study Group. Neurology 1998; 51: 520–5

    Article  PubMed  CAS  Google Scholar 

  15. Pramipexole vs levodopa as initial treatment for Parkinson disease: a randomized controlled trial. Parkinson Study Group. JAMA 2000; 284: 1931–8

  16. Long-term effect of initiating pramipexole vs levodopa in early Parkinson disease. Parkinson Study Group CALM Cohort Investigators. Arch Neurol 2009; 66: 563–70

  17. Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. Parkinson Study Group. JAMA 2002; 287: 1653–61

  18. Albrecht S, Buerger E. Potential neuroprotection mechanisms in PD: focus on dopamine agonist pramipexole. Curr Med Res Opin 2009; 25: 2977–87

    Article  PubMed  CAS  Google Scholar 

  19. Li C, Guo Y, Xie W, et al. Neuroprotection of pramipexole in UPS impairment induced animal model of Parkinson’s disease. Neurochem Res 2010; 35: 1546–56

    Article  PubMed  CAS  Google Scholar 

  20. Du F, Li R, Huang Y, et al. Dopamine D3 receptor-preferring agonists induce neurotrophic effects on mesencephalic dop-amine neurons. Eur J Neurosci 2005; 22: 2422–30

    Article  PubMed  Google Scholar 

  21. Ling ZD, Robie HC, Tong CW, et al. Both the antioxidant and D3 agonist actions of pramipexole mediate its neuroprotective actions in mesencephalic cultures. J Pharmacol Exp Ther 1999; 289: 202–10

    PubMed  CAS  Google Scholar 

  22. Iida M, Miyazaki I, Tanaka K, et al. Dopamine D2 receptor-mediated antioxidant and neuroprotective effects of ropinirole, a dopamine agonist. Brain Res 1999; 838: 51–9

    Article  PubMed  CAS  Google Scholar 

  23. Whone AL, Watts RL, Stoessl AJ, et al. Slower progression of Parkinson’s disease with ropinirole versus levodopa: The REAL-PET study. Ann Neurol 2003; 54: 93–101

    Article  PubMed  CAS  Google Scholar 

  24. Pavese N, Kiferle L, Piccini P. Neuroprotection and imaging studies in Parkinson’s disease. Parkinsonism Relat Disord 2009; 15Suppl. 4: S33–7

    Article  PubMed  Google Scholar 

  25. Guttman M, Stewart D, Hussey D, et al. Influence of L-dopa and pramipexole on striatal dopamine transporter in early PD. Neurology 2001; 56: 1559–64

    Article  PubMed  CAS  Google Scholar 

  26. Jennings DL, Tabama R, Seibyl JP, et al. Investigating the effect of short term treatment with pramipexole or levodopa on [123I]-beta-CIT-SPECT imaging. Mov Dis 2007; 22 Suppl.: S143

    Google Scholar 

  27. Fahn S. Parkinson disease, the effect of levodopa, and the ELLDOPA trial: earlier vs later L-DOPA. Arch Neurol 1999; 56: 529–35

    Article  PubMed  CAS  Google Scholar 

  28. Fernagut PO, Li Q, Dovero S, et al. Dopamine transporter binding is unaffected by L-DOPA administration in normal and MPTP-treated monkeys. PLoS One 2010 Nov 22; 5(11): e14053

    Article  PubMed  Google Scholar 

  29. Olanow CW, Rascol O, Hauser R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 2009; 361: 1268–78

    Article  PubMed  CAS  Google Scholar 

  30. D’Agostino Sr RB. The delayed-start study design. N Engl J Med 2009; 361: 1304–6

    Article  PubMed  Google Scholar 

  31. Freedman NM, Mishani E, Krausz Y, et al. In vivo measurement of brain monoamine oxidase B occupancy by rasagiline, using(11)C-l-deprenyl and PET. JNucl Med 2005; 46:1618–24

    CAS  Google Scholar 

  32. Parkinson Study Group. A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch Neurol 2002; 59: 1937–43

    Article  Google Scholar 

  33. Parkinson Study Group. A controlled, randomized, delayed-start study of rasagiline in early Parkinson disease. Arch Neurol 2004; 61: 561–6

    Article  Google Scholar 

  34. Hauser RA, Lew MF, Hurtig HI, et al. Long-term outcome of early versus delayed rasagiline treatment in early Parkinson’s disease. Mov Disord 2009; 24: 564–73

    Article  PubMed  Google Scholar 

  35. Finberg JP, Takeshima T, Johnston JM, et al. Increased survival of dopaminergic neurons by rasagiline, a mono-amine oxidase B inhibitor. Neuroreport 1998; 9: 703–7

    Article  PubMed  CAS  Google Scholar 

  36. Heikkila RE, Duvoisin RC, Finberg JP, et al. Prevention of MPTP-induced neurotoxicity by AGN-1133 and AGN-1135, selective inhibitors of monoamine oxidase-B. Eur J Pharmacol 1985; 116: 313–7

    Article  PubMed  CAS  Google Scholar 

  37. Akao Y, Maruyama W, Yi H, et al. An anti-Parkinson’s disease drug, N-propargyl-1(R)-aminoindan (rasagiline), enhances expression of anti-apoptotic bc1-2 in human dopaminergic SH-SY5Y cells. Neurosci Lett 2002; 326: 105–8

    Article  PubMed  CAS  Google Scholar 

  38. Maruyama W, Akao Y, Carrillo MC, et al. Neuroprotection by propargylamines in Parkinson’s disease: suppression of apoptosis and induction of prosurvival genes. Neurotox-icol Teratol 2002; 24: 675–82

    Article  CAS  Google Scholar 

  39. Chau KY, Cooper JM, Schapira AH. Rasagiline protects against alpha-synuclein induced sensitivity to oxidative stress in dopaminergic cells. Neurochem Int 2010; 57: 525–9

    Article  PubMed  CAS  Google Scholar 

  40. Weinreb O, Amit T, Bar-Am O, et al. Rasagiline: a novel anti-Parkinsonian monoamine oxidase-B inhibitor with neuroprotective activity. Prog Neurobiol 2010; 92: 330–44

    Article  PubMed  CAS  Google Scholar 

  41. Weinreb O, Amit T, Bar-Am O, et al. Induction of neuro-trophic factors GDNF and BDNF associated with the mechanism of neurorescue action of rasagiline and lados-tigil: new insights and implications for therapy. Ann N Y Acad Sci 2007; 1122: 155–68

    Article  PubMed  CAS  Google Scholar 

  42. Carrillo MC, Minami C, Kitani K, et al. Enhancing effect of rasagiline on superoxide dismutase and catalase activities in the dopaminergic system in the rat. Life Sci 2000; 67: 577–85

    Article  PubMed  CAS  Google Scholar 

  43. Youdim MB, Wadia A, Tatton W, et al. The anti-Parkinson drug rasagiline and its cholinesterase inhibitor derivatives exert neuroprotection unrelated to MAO inhibition in cell culture and in vivo. Ann N Y Acad Sci 2001; 939: 450–8

    Article  PubMed  CAS  Google Scholar 

  44. Schapira AH, Barone P, Comella C, et al. Immediate vs. delayed-start pramipexole in early Parkinson’s disease: the PROUD study [abstract]. Parkinsonism Relat Disord 2009; 15: S81

    Article  Google Scholar 

  45. Schapira AH, Albrecht S, Barone P, et al. Rationale for delayed-start study of pramipexole in Parkinson’s disease: The PROUD study. Mov Disord 2010; 25: 1627–32

    Article  PubMed  Google Scholar 

  46. A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. NINDS NET-PD Investigators. Neurology 2006; 66: 664–71

  47. Surmeier DJ. Calcium, ageing, and neuronal vulnerability in Parkinson’s disease. Lancet Neurol 2007; 6: 933–8

    Article  PubMed  CAS  Google Scholar 

  48. Weisskopf MG, O’Reilly E, Chen H, et al. Plasma urate and risk of Parkinson’s disease. Am J Epidemiol 2007; 166: 561–7

    Article  PubMed  CAS  Google Scholar 

  49. Ascherio A, LeWitt PA, Xu K, et al. Urate as a predictor of the rate of clinical decline in Parkinson disease. Arch Neurol 2009; 66: 1460–8

    Article  PubMed  Google Scholar 

  50. Peterson AL, Nutt JG. Treatment of Parkinson’s disease with trophic factors. Neurotherapeutics 2008; 5: 270–80

    Article  PubMed  CAS  Google Scholar 

  51. Kaplitt MG. Parkinson disease: another player in gene therapy for Parkinson disease. Nat Rev Neurol 2010; 6: 7–8

    Article  PubMed  CAS  Google Scholar 

  52. Ahlskog JE, Uitti RJ. Rasagiline, Parkinson neuroprotection, and delayed-start trials: still no satisfaction? Neurology 2010; 74: 1143–8

    Article  PubMed  CAS  Google Scholar 

  53. Olanow CW, Rascol O. The delayed-start study in Parkinson disease: can’t satisfy everyone. Neurology 2010; 74: 1149–50

    Article  PubMed  Google Scholar 

  54. Clarke CE. Are delayed-start design trials to show neuroprotection in Parkinson’s disease fundamentally flawed? Mov Disord 2008; 23: 784–9

    Article  PubMed  Google Scholar 

  55. Schrag A, Sampaio C, Counsell N, et al. Minimal clinically important change on the Unified Parkinson’s Disease Rating Scale. Mov Disord 2006; 21: 1200–7

    Article  PubMed  Google Scholar 

  56. Schapira AH, Obeso J. Timing of treatment initiation in Parkinson’s disease: a need for reappraisal? Ann Neurol 2006; 59: 559–62

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

No sources of funding were used to assist in the preparation of this article. Claire Henchcliffe has acted as a continuing medical education lecturer for Vindico, TCL and PRIME; has served on advisory boards for UCB, Teva and GE; and has received speakers’ bureau honoraria from Boehringer-Ingelheim, GlaxoSmithKline, Novartis and Teva. W. Lawrence Severt has served on advisory boards for and received speakers’ bureau honoraria from Teva.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Claire Henchcliffe M.D., D.Phil..

Rights and permissions

Reprints and permissions

About this article

Cite this article

Henchcliffe, C., Severt, W.L. Disease Modification in Parkinson’s Disease. Drugs Aging 28, 605–615 (2011). https://doi.org/10.2165/11591320-000000000-00000

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/11591320-000000000-00000

Keywords

Navigation