Skip to main content
Log in

Monoamine Oxidase B Inhibitors for the Treatment of Parkinson’s Disease

A Review of Symptomatic and Potential Disease-Modifying Effects

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

Abstract

Parkinson’s disease is a disorder characterized pathologically by progressive neurodegeneration of the dopaminergic cells of the nigrostriatal pathway. Although the resulting dopamine deficiency is the cause of the typical motor features of Parkinson’s disease (bradykinesia, rigidity, tremor), additional non-motor symptoms appear at various timepoints and are the result of non-dopamine nerve degeneration. Monoamine oxidase B (MAO-B) inhibitors are used in the symptomatic treatment of Parkinson’s disease as they increase synaptic dopamine by blocking its degradation. Two MAO-B inhibitors, selegiline and rasagiline, are currently licensed in Europe and North America for the symptomatic improvement of early Parkinson’s disease and to reduce off-time in patients with more advanced Parkinson’s disease and motor fluctuations related to levodopa. A third MAO-B inhibitor (safinamide), which also combines additional non-dopaminergic properties of potential benefit to Parkinson’s disease, is currently under development in phase III clinical trials as adjuvant therapy to either a dopamine agonist or levodopa.

MAO-B inhibitors have also been studied extensively for possible neuroprotective or disease-modifying actions. There is considerable laboratory evidence that MAO-B inhibitors do exert some neuroprotective properties, at least in the Parkinson’s disease models currently available. However, these models have significant limitations and caution is required in assuming that such results may easily be extrapolated to clinical trials. Rasagiline 1 mg/day has been shown to provide improved motor control in terms of Unified Parkinson’s Disease Rating Scale (UPDRS) score at 18 months in those patients with early disease who began the drug 9 months before a second group. There are a number of possible explanations for this effect that may include a disease-modifying action; however, the US FDA recently declined an application for the licence of rasagiline to be extended to cover disease modification.

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. Schapira AH, Olanow CW. Drug selection and timing of initiation of treatment in early Parkinson’s disease. Ann Neurol 2008 Dec; 64 Suppl. 2: S47–55

    Article  PubMed  CAS  Google Scholar 

  2. Birkmayer W, Riederer P, Youdim MB, et al. The potentiation of the anti akinetic effect after L-dopa treatment by an inhibitor of MAO-B, Deprenil. J Neural Transm 1975; 36(3–4): 303–26

    Article  PubMed  CAS  Google Scholar 

  3. Birkmayer W, Riederer P, Ambrozi L, et al. Implications of combined treatment with ‘Madopar’ and L-deprenil in Parkinson’s disease: a long-term study. Lancet 1977 Feb 26; 1(8009): 439–43

    Article  PubMed  CAS  Google Scholar 

  4. Lees AJ, Shaw KM, Kohout LJ, et al. Deprenyl in Parkinson’s disease. Lancet 1977 Oct 15; 2(8042): 791–5

    Article  PubMed  CAS  Google Scholar 

  5. Schapira A, Bate G, Kirkpatrick P. Rasagiline. Nat Rev Drug Discov 2005 Aug; 4(8): 625–6

    Article  PubMed  CAS  Google Scholar 

  6. Schapira AH. Safinamide in the treatment of Parkinson’s disease. Expert Opin Pharmacother 2010 Sep; 11(13): 2261–8

    Article  PubMed  CAS  Google Scholar 

  7. The Parkinson Study Group. Effect of lazabemide on the progression of disability in early Parkinson’s disease. Ann Neurol 1996 Jul; 40(1): 99–107

    Article  Google Scholar 

  8. Kamada T, Chow T, Hiroi T, et al. Metabolism of selegiline hydrochloride, a selective monoamine b-type inhibitor, in human liver microsomes. Drug Metab Pharmacokinet 2002; 17(3): 199–206

    Article  PubMed  CAS  Google Scholar 

  9. Mandel S, Weinreb O, Amit T, et al. Mechanism of neuroprotective action of the anti-Parkinson drug rasagiline and its derivatives. Brain Res Brain Res Rev 2005 Apr; 48(2): 379–87

    Article  PubMed  CAS  Google Scholar 

  10. Bar AO, Amit T, Youdim MB. Contrasting neuroprotective and neurotoxic actions of respective metabolites of anti-Parkinson drugs rasagiline and selegiline. Neurosci Lett 2004 Jan 30; 355(3): 169–72

    Article  Google Scholar 

  11. Youdim MB, Gross A, Finberg JP. Rasagiline [N-propargyl-1R(+)-aminoindan], a selective and potent inhibitor of mitochondrial monoamine oxidase B. Br J Pharmacol 2001 Jan; 132(2): 500–6

    Article  PubMed  CAS  Google Scholar 

  12. Chen JJ, Swope DM. Clinical pharmacology of rasagiline: a novel, second-generation propargylamine for the treatment of Parkinson disease. J Clin Pharmacol 2005 Aug; 45(8): 878–94

    Article  PubMed  CAS  Google Scholar 

  13. Youdim MB, Amit T, Bar-Am O, et al. Amyloid processing and signal transduction properties of antiparkinson-antialzheimer neuroprotective drugs rasagiline and TV3326. Ann N Y Acad Sci 2003 May; 993: 378–86

    Article  PubMed  CAS  Google Scholar 

  14. Binda C, Wang J, Pisani L, et al. Structures of human monoamine oxidase B complexes with selective noncovalent inhibitors: safinamide and coumarin analogs. J Med Chem 2007 Nov 15; 50(23): 5848–52

    Article  PubMed  CAS  Google Scholar 

  15. Marzo A, Dal BL, Monti NC, et al. Pharmacokinetics and pharmacodynamics of safinamide, a neuroprotectant with antiparkinsonian and anticonvulsant activity. Pharmacol Res 2004 Jul; 50(1): 77–85

    Article  PubMed  CAS  Google Scholar 

  16. Caccia C, Salvati P, Rossetti S, et al. Safinamide: modulation of dopaminergic and glutamatergic systems. Move Disord 2008; 23(1): S22–3

    Google Scholar 

  17. Ives NJ, Stowe RL, Marro J, et al. Monoamine oxidase type B inhibitors in early Parkinson’s disease: meta-analysis of 17 randomised trials involving 3525 patients. BMJ 2004 Sep 11; 329(7466): 593

    Article  PubMed  CAS  Google Scholar 

  18. The Parkinson Study Group. A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch Neurol 2002 Dec; 59(12): 1937–43

    Article  Google Scholar 

  19. The Parkinson Study Group. A controlled, randomized, delayed-start study of rasagiline in early Parkinson disease. Arch Neurol 2004 Apr; 61(4): 561–6

    Article  Google Scholar 

  20. 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 Mar 15; 24(4): 564–73

    Article  PubMed  Google Scholar 

  21. 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 Sep 24; 361(13): 1268–78

    Article  PubMed  CAS  Google Scholar 

  22. The Parkinson Study Group. A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations: the PRESTO study. Arch Neurol 2005 Feb; 62(2): 241–8

    Article  Google Scholar 

  23. Rascol O, Brooks DJ, Melamed E, et al. Rasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once daily, study): a randomised, double-blind, parallel-group trial. Lancet 2005 Mar 12; 365(9463): 947–54

    Article  PubMed  CAS  Google Scholar 

  24. Stocchi F, Arnold G, Onofrj M, et al. Improvement of motor function in early Parkinson disease by safinamide. Neurology 2004 Aug 24; 63(4): 746–8

    Article  PubMed  CAS  Google Scholar 

  25. Stocchi F, Vacca L, Grassini P, et al. Symptom relief in Parkinson disease by safinamide: biochemical and clinical evidence of efficacy beyond MAO-B inhibition. Neurology 2006 Oct 10; 67(7 Suppl. 2): S24–9

    Article  PubMed  CAS  Google Scholar 

  26. Sharma T, Stocchi F, Schapira AH, et al. Satinamide treatment improves cognition in Parkinson’s disease [abstract]. Move Disord 2007 Sep; 22(12): 29

    Google Scholar 

  27. Borgohain R, Szasz J, Bhatt M, et al. 2009 efficacy and safety of safinamide in patients with Parkinson’s disease experiencing motor fluctuations: results of a 6-month phase III, randomized, double-blind, placebo-controlled study [online]. Available from URL: http://www.movementdisorders.org/congress/congress09/late_breaking_abstracts. pdf [Accessed 2011 Sep 9]

  28. EMD Serono. MOTION, Safinamide in Early IPD, as Addon to Dopamine Agonist [ClinicalTrials.gov identifier NCT00605683]. US National Institutes of Health, ClinicalTrials.gov [online]. Available from URL: http://www.clinicaltrials.gov [Accessed 2010 Sep 16]

  29. EMD Serono. Safinamide in Idiopathic Parkinson’s Disease (IPD) With Motor Fluctuations, as add-on to Levodopa (SETTLE) [ClinicalTrials.govidentifierNCT00627640]. US National Institutes of Health, ClinicalTrials.gov [online]. Available from URL: http://www.clinicaltrials.gov [Accessed 2010 Sep 16]

  30. Heikkila RE, Manzino L, Cabbat FS, et al. Protection against the dopaminergic neurotoxicity of 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine by monoamine oxidase inhibitors. Nature 1984 Oct 4; 311(5985): 467–9

    Article  PubMed  CAS  Google Scholar 

  31. Schapira AH, Cooper JM, Dexter D, et al. Mitochondrial complex I deficiency in Parkinson’s disease [letter]. Lancet 1989 Jun 3; 1(8649): 1269

    Article  PubMed  CAS  Google Scholar 

  32. Schapira AH, Cooper JM, Dexter D, et al. Mitochondrial complex I deficiency in Parkinson’s disease. J Neurochem 1990 Mar; 54(3): 823–7

    Article  PubMed  CAS  Google Scholar 

  33. Owen AD, Schapira AH, Jenner P, et al. Oxidative stress and Parkinson’s disease. Ann N Y Acad Sci 1996 Jun 15; 786: 217–23

    Article  PubMed  CAS  Google Scholar 

  34. Schapira AH. Oxidative stress in Parkinson’s disease. Neuropathol Appl Neurobiol 1995 Feb; 21(1): 3–9

    Article  PubMed  CAS  Google Scholar 

  35. Morgan-Hughes JA, Sweeney MG, Cooper JM, et al. Mitochondrial DNA (mtDNA) diseases: correlation of genotype to phenotype. Biochim Biophys Acta 1995 May 24; 1271(1): 135–40

    Article  PubMed  Google Scholar 

  36. Schapira AH, Cooper JM, Morgan-Hughes JA, et al. Molecular basis of mitochondrial myopathies: polypeptide analysis in complex-I deficiency. Lancet 1988 Mar 5; 1(8584): 500–3

    Article  PubMed  CAS  Google Scholar 

  37. Tatton WG, Greenwood CE. Rescue of dying neurons: a new action for deprenyl in MPTP parkinsonism. J Neurosci Res 1991 Dec; 30(4): 666–72

    Article  PubMed  CAS  Google Scholar 

  38. Matsubara K, Senda T, Uezono T, et al. L-deprenyl prevents the cell hypoxia induced by dopaminergic neurotoxins, MPP(+) and beta-carbolinium: a microdialysis study in rats. Neurosci Lett 2001 Apr 20; 302(2–3): 65–8

    Article  PubMed  CAS  Google Scholar 

  39. Kupsch A, Sautter J, Gotz ME, et al. Monoamine oxidase-inhibition and MPTP-induced neurotoxicity in the non-human primate: comparison of rasagiline (TVP 1012) with selegiline. J Neural Transm 2001; 108(8–9): 985–1009

    Article  PubMed  CAS  Google Scholar 

  40. Gerlach M, Foley P, Riederer P. The relevance of preclinical studies for the treatment of Parkinson’s disease. J Neurol 2003 Feb; 250 Suppl. 1:I31–4

    Article  PubMed  Google Scholar 

  41. Koutsilieri E, O’Callaghan JF, Chen TS, et al. Selegiline enhances survival and neurite outgrowth of MPP(+)-treated dopaminergic neurons. Eur J Pharmacol 1994 Nov 15; 269(3): R3–4

    Article  PubMed  CAS  Google Scholar 

  42. Koutsilieri E, Chen TS, Rausch WD, et al. Selegiline is neuroprotective in primary brain cultures treated with 1-methyl-4-phenylpyridinium. Eur J Pharmacol 1996 Jun 13; 306(1–3): 181–6

    Article  PubMed  CAS  Google Scholar 

  43. Mytilineou C, Radcliffe P, Leonardi EK, et al. L-deprenyl protects mesencephalic dopamine neurons from glutamate receptor-mediated toxicity in vitro. J Neurochem 1997 Jan; 68(1): 33–9

    Article  PubMed  CAS  Google Scholar 

  44. Maruyama W, Naoi M. Neuroprotection by (−)-deprenyl and related compounds. Mech Ageing Dev 1999 Nov; 111(2–3): 189–200

    Article  PubMed  CAS  Google Scholar 

  45. Maruyama W, Nitta A, Shamoto-Nagai M, et al. N-propargyl-1 (R)-aminoindan, rasagiline, increases glial cell line-derived neurotrophic factor (GDNF) in neuroblastoma SH-SY5Y cells through activation of NF-kappaB transcription factor. Neurochem Int 2004 May; 44(6): 393–400

    Article  PubMed  CAS  Google Scholar 

  46. Youdim MB, Weinstock M. Molecular basis of neuroprotective activities of rasagiline and the anti-Alzheimer drug TV3326 [(N-propargyl-(3R)aminoindan-5-YL)-ethyl methyl carbamate]. Cell Mol Neurobiol 2001 Dec; 21(6): 555–73

    Article  PubMed  CAS  Google Scholar 

  47. 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 Jun; 939: 450–8

    Article  PubMed  CAS  Google Scholar 

  48. Maruyama W, Weinstock M, Youdim MB, et al. Antiapoptotic action of anti-Alzheimer drug, TV3326 [(N-propargyl)-(3R)-aminoindan-5-yl]-ethyl methyl carbamate, a novel cholinesterase-monoamine oxidase inhibitor. Neurosci Lett 2003 May 8; 341(3): 233–6

    Article  PubMed  CAS  Google Scholar 

  49. Bonneh-Barkay D, Ziv N, Finberg JP. Characterization of the neuroprotective activity of rasagiline in cerebellar granule cells. Neuropharmacology 2005 Mar; 48(3): 406–16

    Article  PubMed  CAS  Google Scholar 

  50. Finberg JP, Takeshima T, Johnston JM, et al. Increased survival of dopaminergic neurons by rasagiline, a monoamine oxidase B inhibitor. Neuroreport 1998 Mar 9; 9(4): 703–7

    Article  PubMed  CAS  Google Scholar 

  51. Sabi Sagi Y, Mandel S, Amit T, et al. Activation of tyrosine kinase receptor signaling pathway by rasagiline facilitates neurorescue and restoration of nigrostriatal dopamine neurons in post-MPTP-induced parkinsonism. Neurobiol Dis 2007 Jan; 25(1): 35–44

    Article  Google Scholar 

  52. Zhu W, Xie W, Pan T, et al. Comparison of neuroprotective and neurorestorative capabilities of rasagiline and selegiline against lactacystin-induced nigrostriatal dopaminergic degeneration. J Neurochem 2008 Jun; 105(5): 1970–8

    Article  PubMed  CAS  Google Scholar 

  53. Chau KY, Cooper JM, Schapira AH. Rasagiline protects against alpha-synuclein induced sensitivity to oxidative stress in dopaminergic cells. Neurochem Int 2010 Nov; 57(5): 525–9

    Article  PubMed  CAS  Google Scholar 

  54. Youdim MB, Bar AO, Yogev-Falach M, et al. Rasagiline: neurodegeneration, neuroprotection, and mitochondrial permeability transition. J Neurosci Res 2005 Jan 1; 79(1–2): 172–9

    Article  PubMed  CAS  Google Scholar 

  55. Seaton TA, Cooper JM, Schapira AH. Cyclosporin inhibition of apoptosis induced by mitochondrial complex I toxins. Brain Res 1998 Oct 26; 809(1): 12–7

    Article  PubMed  CAS  Google Scholar 

  56. Weinreb O, Bar-Am O, Amit T, et al. Neuroprotection via pro-survival protein kinase C isoforms associated with Bcl-2 family members. FASEB J 2004 Sep; 18(12): 1471–3

    PubMed  CAS  Google Scholar 

  57. Maruyama W, Akao Y, Youdim MB, et al. Transfection-enforced Bcl-2 overexpression and an anti-Parkinson drug, rasagiline, prevent nuclear accumulation of glyceraldehyde3-phosphate dehydrogenase induced by an endogenous dopaminergic neurotoxin, N-methyl(R)salsolinol. J Neurochem 2001 Aug; 78(4): 727–35

    Article  PubMed  CAS  Google Scholar 

  58. Blandini F, Armentero MT, Fancellu R, et al. Neuroprotective effect of rasagiline in a rodent model of Parkinson’s disease. Exp Neurol 2004 Jun; 187(2): 455–9

    Article  PubMed  CAS  Google Scholar 

  59. Weinreb O, Bar-Am O, Prosolovich K, et al. Does 1-(R)-aminoindan possess neuroprotective properties against experimental Parkinson’s disease? Antioxid Redox Signal 2011 Mar 1; 14(5): 767–75

    Article  PubMed  CAS  Google Scholar 

  60. Bar-Am O, Weinreb O, Amit T, et al. The neuroprotective mechanism of 1-(R)-aminoindan, the major metabolite of the anti-parkinsonian drug rasagiline. J Neurochem 2010 Mar; 112(5): 1131–7

    Article  PubMed  CAS  Google Scholar 

  61. Schapira AH. Challenges to the development of disease-modifying therapies in Parkinson’s disease. Eur J Neurol 2011 Mar; 18 Suppl. 1: 16–21

    Article  Google Scholar 

  62. Schapira AH, Tolosa E. Molecular and clinical prodrome of Parkinson disease: implications for treatment. Nat Rev Neurol 2010 Jun; 6(6): 309–17

    Article  PubMed  CAS  Google Scholar 

  63. Olanow CW, Kieburtz K, Schapira AH. Why have we failed to achieve neuroprotection in Parkinson’s disease? Ann Neurol 2008 Dec; 64 Suppl. 2: S101–10

    Article  PubMed  CAS  Google Scholar 

  64. Olanow CW, Schapira AH, LeWitt PA, et al. TCH346 as a neuroprotective drug in Parkinson’s disease: a double-blind, randomised, controlled trial. Lancet Neurol 2006 Dec; 5(12): 1013–20

    Article  PubMed  CAS  Google Scholar 

  65. Parkinson Study Group. Effect of deprenyl on the progression of disability in early Parkinson’s disease. N Engl J Med 1989 Nov 16; 321(20): 1364–71

    Article  Google Scholar 

  66. LeWitt PA. Clinical trials of neuroprotection for Parkinson’s disease. Neurology 2004 Oct 12; 63(7 Suppl. 2): S23–31

    Article  PubMed  Google Scholar 

  67. 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 parkinsonism trial. Ann Neurol 2002 May; 51(5): 604–12

    Article  PubMed  CAS  Google Scholar 

  68. Jenner P. Preclinical evidence for neuroprotection with monoamine oxidase-B inhibitors in Parkinson’s disease. Neurology 2004 Oct 12; 63(7 Suppl. 2): S13–22

    Article  PubMed  Google Scholar 

  69. Palhagen S, Heinonen E, Hagglund J, et al. Selegiline slows the progression of the symptoms of Parkinson disease. Neurology 2006 Apr 25; 66(8): 1200–6

    Article  PubMed  CAS  Google Scholar 

  70. Parkinson Study Group. Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease: the Parkinson Study Group. N Engl J Med 1993 Jan 21; 328(3): 176–83

    Article  Google Scholar 

  71. Mally J, Kovacs AB, Stone TW. Delayed development of symptomatic improvement by (−)-deprenyl in Parkinson’s disease. J Neurol Sci 1995 Dec; 134(1–2): 143–5

    Article  PubMed  CAS  Google Scholar 

  72. Lees AJ. Comparison of therapeutic effects and mortality data of levodopa and levodopa combined with selegiline in patients with early, mild Parkinson’s disease: Parkinson’s Disease Research Group of the United Kingdom. BMJ 1995 Dec 16; 311(7020): 1602–7

    Article  PubMed  CAS  Google Scholar 

  73. Schapira AH. Molecular and clinical pathways to neuroprotection of dopaminergic drugs in Parkinson disease. Neurology 2009 Feb 17; 72(7 Suppl. ): S44–50

    Article  PubMed  CAS  Google Scholar 

  74. Schapira AH. Science, medicine, and the future: Parkinson’s disease. BMJ 1999 Jan 30; 318(7179): 311–4

    Article  PubMed  CAS  Google Scholar 

  75. Fahn S, Oakes D, Shoulson I, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med 2004 Dec 9; 351(24): 2498–508

    Article  PubMed  CAS  Google Scholar 

  76. Nissinen H, Kuoppamaki M, Leinonen M, et al. Early versus delayed initiation of entacapone in levodopa-treated patients with Parkinson’s disease: a long-term, retrospective analysis. Eur J Neurol 2009 Dec; 16(12): 1305–11

    Article  PubMed  CAS  Google Scholar 

  77. Schapira AH, Bezard E, Brotchie J, et al. Novel pharmacological targets for the treatment of Parkinson’s disease. Nat Rev Drug Discov 2006 Oct; 5(10): 845–54

    Article  PubMed  CAS  Google Scholar 

  78. Schapira AH, Emre M, Jenner P, et al. Levodopa in the treatment of Parkinson’s disease. Eur J Neurol 2009 Sep; 16(9): 982–9

    Article  PubMed  CAS  Google Scholar 

  79. Schapira AH. Neurobiology and treatment of Parkinson’s disease. Trends Pharmacol Sci 2009 Jan; 30(1): 41–7

    Article  PubMed  CAS  Google Scholar 

  80. Schapira AH, Obeso J. Timing of treatment initiation in Parkinson’s disease: a need for reappraisal? Ann Neurol 2006 Mar; 59(3): 559–62

    Article  PubMed  Google Scholar 

  81. Schapira AH. Movement disorders: advances in cause and treatment. Lancet Neurol 2010 Jan; 9(1): 6–7

    Article  PubMed  Google Scholar 

  82. Rascol O, Fitzer-Attas CJ, Hauser R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease (the ADAGIO study): prespecified and post-hoc analyses of the need for additional therapies, changes in UPDRS scores, and non-motor outcomes. Lancet Neurol 2011 May; 10(5): 415–23

    Article  PubMed  CAS  Google Scholar 

  83. Schapira AH, Schrag A. Parkinson disease: Parkinson disease clinical subtypes and their implications. Nat Rev Neurol 2011 May; 7(5): 247–8

    Article  PubMed  Google Scholar 

  84. Schapira AH. Treatment options in the modern management of Parkinson disease. Arch Neurol 2007 Aug; 64(8): 1083–8

    Article  PubMed  Google Scholar 

  85. Schapira AH, Agid Y, Barone P, et al. Perspectives on recent advances in the understanding and treatment of Parkinson’s disease. Eur J Neurol 2009 Oct; 16(10): 1090–9

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

This article was written solely by the author. No funds were provided to prepare this review. The author has received honoraria for educational symposia from GSK, BI, EMD Serono, TEVA Lundbeck and Orion Novartis, as well as consultancy advice to the same companies, and grants from the MRC, Wellcome, BI and Parkinson UK.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anthony H. V. Schapira.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Schapira, A.H.V. Monoamine Oxidase B Inhibitors for the Treatment of Parkinson’s Disease. CNS Drugs 25, 1061–1071 (2011). https://doi.org/10.2165/11596310-000000000-00000

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/11596310-000000000-00000

Keywords

Navigation