Skip to main content
Log in

Medical Management of Levodopa-Associated Motor Complications in Patients with Parkinson’s Disease

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

Abstract

Parkinson’s disease is a neurodegenerative disorder that affects approximately 1% of people over the age of 60 years. Levodopa is standard, and often initial, therapy for patients with this condition; however, with continued treatment and as the disease progresses, up to 80% of patients experience ‘wearing-off’ symptoms, dyskinesias and other motor complications. These levodopa-associated problems may become disabling and profoundly affect quality of life. Medications commonly used to manage these symptoms include monoamine oxidase type B (MAO-B) inhibitors, catechol-O-methyltransferase (COMT) inhibitors, the NMDA receptor antagonist amantadine and dopamine receptor agonists.

Agents that block MAO-B, such as rasagiline and selegiline, are used as both initial and adjunctive therapy in patients with Parkinson’s disease. These medications increase concentrations of dopamine in the brain by blocking its reuptake from the synaptic cleft, a mechanism that can slow motor decline, increase ‘on’ time and improve symptoms of Parkinson’s disease. Adverse events with these agents can include confusion, hallucination and orthostatic hypotension. MAO-B inhibition may elicit drug-drug interactions if administered with TCAs, SSRIs or serotonin-noradrenaline (norepinephrine) reuptake inhibitors. Conventional oral selegiline is associated with potentially harmful plasma concentrations of three major amfetamine metabolites, although metabolite concentrations are significantly lower with a new orally disintegrating tablet (ODT) selegeline formulation. Selegiline ODT is also absorbed more efficiently and shows less pharmacokinetic variability than conventional oral selegiline.

COMT mediates peripheral catabolism of levodopa. Therefore, agents that block COMT, such as tolcapone and entacapone, increase the elimination half-life of levodopa. Given adjunctively with levodopa, COMT inhibitors can decrease ‘off’ time and increase ‘on’ time, as well as lower the daily levodopa dose. Although more potent than entacapone, tolcapone requires monitoring for hepatotoxicity.

Amantadine is a noncompetitive NMDA receptor antagonist shown to lower dyskinesia scores and improve motor complications in patients with Parkinson’s disease when given adjunctively with levodopa.

Dopamine agonists, also used as initial and adjunctive therapy in Parkinson’s disease, improve motor response and decrease ‘off’ time purportedly through direct stimulation of dopamine receptors. Current dopamine agonists include bromocriptine, pergolide, cabergoline, lisuride, apomorphine, pramipexole, ropinirole and rotigotine. Although effective, this class of medications can be associated with cardiovascular and psychiatric adverse effects that can limit their utility.

All medications used to manage levodopa-associated motor complications in patients with Parkinson’s disease have had differing degrees of success. Although head-to-head comparisons of drugs within classes are rare, some differences have emerged related to effects on motor fluctuations, dyskinesias and on/off times, as well as to adverse effects. When choosing a drug to treat levodopa-induced complications, it is important to consider the risks and benefits of the different classes and of the specific agents within each class, given the different efficacy and safety profiles of each.

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
Table I

Similar content being viewed by others

Notes

  1. The use of trade names is for product identification purposes only and does not imply endorsement.

References

  1. Samii A, Nutt JG, Ransom BR. Parkinson’s disease. Lancet 2004; 363(9423): 1783–93

    Article  PubMed  CAS  Google Scholar 

  2. Hampton T. Parkinson disease registry launched. JAMA 2005; 293(2): 149

    Article  PubMed  CAS  Google Scholar 

  3. de Lau LML, Breteler MMB. Epidemiology of Parkinson’s disease. Lancet Neurol 2006; 5(6): 525–35

    Article  PubMed  Google Scholar 

  4. Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s disease. N Engl J Med 2003; 348(14): 1356–64

    Article  PubMed  CAS  Google Scholar 

  5. Jankovic J. Pathophysiology and assessment of parkinsonian symptoms and signs. In: Pahwa R, Lyons KE, Koller WC, editors. Handbook of Parkinson’s disease. 4th ed. New York: Taylor & Francis Group, LLC, 2007: 49–75

    Google Scholar 

  6. Chaudhuri KR, Healy DG, Shapira AH, et al. Non-motor symp- toms of Parkinson’s disease: diagnosis and management. Lancet Neurol 2006; 5: 235–45

    Article  PubMed  Google Scholar 

  7. Olanow CW, Agid Y, Mizuno Y, et al. Levodopa in the treatment of Parkinson’s disease: current controversies. Mov Disord 2004; 19(9): 997–1005

    Article  PubMed  Google Scholar 

  8. Dewey Jr RB. Management of motor complications in Parkinson’s disease. Neurology 2004; 62Suppl. 4: S3–7

    Article  PubMed  Google Scholar 

  9. Jankovic J. Levodopa strengths and weaknesses. Neurology 2002; 58Suppl. 1: S19–32

    Article  PubMed  CAS  Google Scholar 

  10. Jankovic J. Motor fluctuations and dyskinesias in Parkinson’s disease: clinical manifestations. Mov Disord 2005; 20Suppl, 11: S11–6

    Article  PubMed  Google Scholar 

  11. Marjama-Lyons JM, Koller WC. Parkinson’s disease: update in diagnosis and symptom management. Geriatrics 2001; 56(8): 24–30, 33

    PubMed  CAS  Google Scholar 

  12. Obeso JA, Grandas F, Vaamonde J, et al. Motor complications associated with chronic levodopa therapy in Parkinson’s disease. Neurology 1989; 39Suppl. 2: 11–9

    PubMed  CAS  Google Scholar 

  13. Stacy M, Bowron A, Guttman M, et al. Identification of motor and nonmotor wearing-off in Parkinson’s disease: comparison of a patient questionnaire versus a clinician assessment. Mov Disord 2005; 20(6): 726–33

    Article  PubMed  Google Scholar 

  14. Stacy M, Hauser R, Oertel W, et al. End of dose wearing-off in Parkinson’s disease: a 9-question survey assessment. Clin Neuropharmacol 2006; 29(6): 312–32

    Article  PubMed  Google Scholar 

  15. Chapuis S, Ouchchane L, Metz O, et al. Impact of the motor complications of Parkinson’s disease on the quality of life. Mov Disord 2005; 20(2): 224–30

    Article  PubMed  Google Scholar 

  16. 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; 365(9463): 947–54

    Article  PubMed  CAS  Google Scholar 

  17. Jankovic J. An update on the treatment of Parkinson’s disease [Melvin D. Yahr memorial lecture]. Mt Sinai J Med 2006; 73(4): 682–9

    PubMed  Google Scholar 

  18. Olanow CW, Jankovic J. Neuroprotective therapy in Parkinson’s disease and motor complications: a search for a pathogenesis-targeted, disease-modifying strategy. Mov Disord 2005; 20Suppl. 11: S3–10

    Article  PubMed  Google Scholar 

  19. Kurth MC. Using liquid levodopa in the treatment of Parkinson’s disease: a practical guide. Drugs Aging 1997; 10(5): 332–40

    Article  PubMed  CAS  Google Scholar 

  20. Stocchi F, Vacca L, Ruggieri S, et al. Intermittent vs continuous levodopa administration in patients with advanced Parkinson disease: a clinical and pharmacokinetic study. Arch Neurol 2005; 62(6): 905–10

    Article  PubMed  Google Scholar 

  21. Nilsson D, Hansson LE, Johansson K, et al. Long-term intraduodenal infusion of a water based levodopa-carbidopa dispersion in very advanced Parkinson’s disease. Acta Neurol Scand 1998; 97(3): 175–83

    Article  PubMed  CAS  Google Scholar 

  22. Rao SS, Hofmann LA, Shakil A. Parkinson’s disease: diagnosis and treatment. Am Fam Physician 2006; 74(12): 2046–54

    PubMed  Google Scholar 

  23. Allain H, Destée A, Petit H, et al. Five-year follow-up of early lisuride and levodopa combination therapy versus levodopa monotherapy in de novo Parkinson’s disease. Eur Neurol 2000; 44(1): 22–30

    Article  PubMed  CAS  Google Scholar 

  24. Churchyard A, Mathias CJ, Boonkongchuen P, et al. Autonomic effects of selegiline: possible cardiovascular toxicity in Parkinson’s disease. J Neurol Neurosurg Psychiatry 1997; 63(2): 228–34

    Article  PubMed  CAS  Google Scholar 

  25. Waters CH, Sethi KD, Hauser RA, et al. Zydis selegiline reduces off time in Parkinson’s disease patients with motor fluctuations: a 3-month, randomized, placebo-controlled study. Mov Disord 2004; 19(4): 426–32

    Article  PubMed  Google Scholar 

  26. Guttman M, Kish SJ, Furukawa Y. Current concepts in the diagnosis and management of Parkinson’s disease. CMAJ 2003; 168(3): 293–301

    PubMed  Google Scholar 

  27. Churchyard A, Mathias CJ, Lees AJ. Selegiline-induced postural hypotension in Parkinson’s disease: a longitudinal study on the effects of drug withdrawal. Mov Disord 1999; 14(2): 246–51

    Article  PubMed  CAS  Google Scholar 

  28. 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; 62(2): 241–8

    Article  Google Scholar 

  29. Radad K, Gille G, Rausch W-D. Short review on dopamine agonists: insight into clinical and research studies relevant to Parkinson’s disease. Pharmacol Rep 2005; 57(6): 701–12

    PubMed  CAS  Google Scholar 

  30. Goetz CG, Poewe W, Rascol O, et al. Evidence-based medical review update: pharmacological and surgical treatments of Parkinson’s disease, 2001 to 2004. Mov Disord 2005; 20(5): 523–39

    Article  PubMed  Google Scholar 

  31. Henchcliffe C, Schumacher HC, Burgut FT. Recent advances in Parkinson’s disease therapy: use of monoamine oxidase inhibitors. Expert Rev Neurother 2005; 5(6): 811–21

    Article  PubMed  CAS  Google Scholar 

  32. Dostinex [package insert]. New York; Pfizer Inc, 2004

  33. Deuschl G, Schade-Brittinger C, Krack P, et al. A randomized trial of deep-brain stimulation for Parkinson’s disease. N Engl J Med 2006; 355(9): 896–908

    Article  PubMed  CAS  Google Scholar 

  34. Horstink M, Tolosa E, Bonuccelli U, et al. Review of the therapeutic management of Parkinson’s disease. Report of a joint task force of the European Federation of Neurological Societies and the Movement Disorder Society-European Section: Pt I. Early (uncomplicated) Parkinson’s disease. Eur J Neurol 2006; 13(11): 1170–85

    CAS  Google Scholar 

  35. Watts RL, Jankovic J, Waters C, et al. Randomized, blind, controlled trial of transdermal rotigotine in early Parkinson disease. Neurology 2007; 68(4): 272–6

    Article  PubMed  CAS  Google Scholar 

  36. Mahmood I. Clinical pharmacokinetics and pharmacodynamics of selegiline: an update. Clin Pharmacokinet 1997; 33(2): 91–102

    Article  PubMed  CAS  Google Scholar 

  37. Mercuri NB, Scarponi M, Bonci A, et al. Monoamine oxidase inhibition causes a long-term prolongation of the dopamine-induced responses in rat midbrain dopaminergic cells. J Neurosci 1997; 17(7): 2267–72

    PubMed  CAS  Google Scholar 

  38. Goetz CG, Schwid SR, Eberly SW, et al. Safety of rasagiline in elderly patients with Parkinson disease. Neurology 2006; 66(9): 1427–9

    Article  PubMed  CAS  Google Scholar 

  39. Azilect [package insert]. Kfar Saba; Teva Pharmaceutical Industries Ltd, 2006

  40. Clarke CE. Rasagiline for motor complications in Parkinson’s disease. Lancet 2005; 365(9463): 914–6

    Article  PubMed  Google Scholar 

  41. Demarcaida JA, Schwid SR, White WB, et al. Effects of tyramine administration in Parkinson’s disease patients treated with selective MAO-B inhibitor rasagiline. Mov Disord 2006; 21(10): 1716–21

    Article  PubMed  Google Scholar 

  42. Shin H-S. Metabolism of selegiline in humans: identification, excretion, and stereochemistry of urine metabolites. Drug Metab Dispos 1997; 25(6): 657–62

    PubMed  CAS  Google Scholar 

  43. Cedarbaum JM, Toy LH, Green-Parsons A. L-deprenyl (selegiline) added to Sinemet CR in the management of Parkinson’s disease patients with motor response fluctuations. Clin Neuropharmacol 1991; 14(3): 228–34

    Article  PubMed  CAS  Google Scholar 

  44. 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; 51(5): 604–12

    Article  PubMed  CAS  Google Scholar 

  45. Pålhagen S, Heinonen E, Hägglund J, et al. Selegiline slows the progression of the symptoms of Parkinson disease. Neurology 2006; 66: 1200–6

    Article  PubMed  Google Scholar 

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

    Article  Google Scholar 

  47. Clarke A, Jankovic J. Selegiline orally disintegrating tablet in the treatment of Parkinson’s disease. Therapy 2006; 3(3): 349–56

    CAS  Google Scholar 

  48. Lew MF. Selegiline orally disintegrating tablets for the treatment of Parkinson’s disease. Expert Rev Neurother 2005; 5(6): 705–12

    Article  PubMed  CAS  Google Scholar 

  49. Zhang H, Zhang J, Streisand JB. Oral mucosal drug delivery: clinical pharmacokinetics and therapeutic applications. Clin Pharmacokinet 2002; 41(9): 661–80

    Article  PubMed  CAS  Google Scholar 

  50. Fuh J-L, Lee R-C, Wang S-J, et al. Swallowing difficulty in Parkinson’s disease. Clin Neurol Neurosurg 1997; 99(2): 106–12

    PubMed  CAS  Google Scholar 

  51. Zelapar (selegiline hydrochloride) orally disintegrating tablets [package insert]. Costa Mesa (CA); Valeant Pharmaceuticals International, 2006

  52. Clarke A, Brewer F, Johnson ES, et al. A new formulation of selegiline: improved bioavailability and selectivity for MAO-B inhibition. J Neural Transm 2003; 110(11): 1241–55

    Article  PubMed  CAS  Google Scholar 

  53. Heikkinen H, Nutt JG, LeWitt PA, et al. The effects of different repeated doses of entacapone on the pharmacokinetics of L-dopa and on the clinical response to L-dopa in Parkinson’s disease. Clin Neuropharmacol 2001; 24(3): 150–7

    Article  PubMed  CAS  Google Scholar 

  54. Nyholm D. Pharmacokinetic optimisation in the treatment of Parkinson’s disease: an update. Clin Pharmacokinet 2006; 45(2): 109–36

    Article  PubMed  CAS  Google Scholar 

  55. Widnell KL, Cornelia C. Role of COMT inhibitors and dopamine agonists in the treatment of motor fluctuations. Mov Disord 2005; 20Suppl. 11: S30–7

    Article  PubMed  Google Scholar 

  56. Olanow CW. Tolcapone and hepatotoxic effects. Arch Neurol 2000; 57(2): 263–7

    Article  PubMed  CAS  Google Scholar 

  57. Lew M, Kricorian G. Results from a 2-year centralized tolcapone liver enzyme monitoring program [abstract no. 1035]. J Neurol Sci 2005; 238Suppl. 1: S363

    Google Scholar 

  58. Factor SA, Molho ES, Feustel PJ, et al. Long-term comparative experience with tolcapone and entacapone in advanced Parkinson’s disease. Clin Neuropharmacol 2001; 24(5): 295–9

    Article  PubMed  CAS  Google Scholar 

  59. Entacapone to tolcapone switch: multicenter double-blind, randomized, active-controlled trial in advanced Parkinson’s disease. Mov Disord 2007; 22 (1): 14-9

    Google Scholar 

  60. Luginger E, Wenning GK, Bosch S, et al. Beneficial effects of amantadine on L-dopa-induced dyskinesias in Parkinson’s disease. Mov Disord 2000; 15(5): 873–8

    Article  PubMed  CAS  Google Scholar 

  61. Snow BJ, Macdonald L, Mcauley D, et al. The effect of amantadine on levodopa-induced dyskinesias in Parkinson’s disease: a double-blind, placebo-controlled study. Clin Neuropharmacol 2000; 23(2): 82–5

    Article  PubMed  CAS  Google Scholar 

  62. Verhagen Metman L, Del Dotto P, Blanchet PJ, et al. Blockade of glutamatergic transmission as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Amino Acids 1998; 14(1–3): 75–82

    Google Scholar 

  63. 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 

  64. Bonuccelli U. Comparing dopamine agonists in Parkinson’s disease. Curr Opin Neurol 2003; 16Suppl. 1: S13–9

    Article  PubMed  CAS  Google Scholar 

  65. Bonnet AM, Serre I, Marconi R, et al. A “combined” levodopa test as a useful method for evaluating the efficacy of dopamine agonists: application to pergolide and bromocriptine. Mov Disord 1995; 10(5): 668–71

    Article  PubMed  CAS  Google Scholar 

  66. Tintner R, Manian P, Gauthier P, et al. Pleuropulmonary fibrosis after long-term treatment with the dopamine agonist pergolide for Parkinson disease. Arch Neurol 2005; 62(8): 1290–5

    Article  PubMed  Google Scholar 

  67. Hayashi R, Tako K, Mikashita H, et al. Efficacy of low-dose lisuride infusion in Parkinson’s disease. Intern Med 1998; 37(5): 444–8

    Article  PubMed  CAS  Google Scholar 

  68. Woitalla D, Müller T, Benz S, et al. Transdermal lisuride delivery in the treatment of Parkinson’s disease. J Neural Transm Suppl 2004; 68: 89–95

    Article  PubMed  CAS  Google Scholar 

  69. Guttman M, the International Pramipexole-Bromocriptine Study Group. Double-blind comparison of pramipexole and bromocriptine treatment with placebo in advanced Parkinson’s disease. Neurology 1997; 49(4): 1060–5

    Article  PubMed  CAS  Google Scholar 

  70. Lieberman A, Ranhosky A, Korts D. Clinical evaluation of pramipexole in advanced Parkinson’s disease: results of a double-blind, placebo-controlled, parallel-group study. Neurology 1997; 49(1): 162–8

    Article  PubMed  CAS  Google Scholar 

  71. Lieberman A, Olanow CW, Sethi K, et al. A multicenter trial of ropinirole as adjunct treatment for Parkinson’s disease. Ropinirole Study Group. Neurology 1998; 51(4): 1057–62

    CAS  Google Scholar 

  72. Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. N Engl J Med 2000; 342(20): 1484–91

    Article  PubMed  CAS  Google Scholar 

  73. Whone AL, Watts RL, Stoessl AJ, et al. Slower progression in early Parkinson’s disease treated with ropinirole compared with 1-dopa: the REAL-PET study. Ann Neurol 2003; 54(1): 93–101

    Article  PubMed  CAS  Google Scholar 

  74. Jankovic J, Watts RL, Martin W, et al. Transdermal rotigotine: double-blind, placebo-controlled trial in Parkinson’s disease. Arch Neurol 2007; 64: 676–82

    Article  PubMed  Google Scholar 

  75. Kenney C, Jankovic J. Rotigotine transdermal patch in the treatment of Parkinson’s disease and restless legs syndrome. Expert Opin Pharmacother 2007; 8(9): 1329–35

    Article  PubMed  CAS  Google Scholar 

  76. Kolls B, Stacy M. Apomorphine: a rapid rescue agent for the management of motor fluctuations in advanced Parkinson disease. Clin Neuropharmacol 2006; 29(5): 292–301

    Article  PubMed  CAS  Google Scholar 

  77. Dewey Jr RB, Hutton JT, LeWitt PA, et al. A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch Neurol 2001; 58(9): 1385–92

    Article  PubMed  Google Scholar 

  78. Factor SA. Literature review: intermittent subcutaneous apomorphine therapy in Parkinson’s disease. Neurology 2004; 62(6 Suppl. 4): S12–7

    Article  PubMed  CAS  Google Scholar 

  79. Schade R, Andersohn F, Suissa S, et al. Dopamine agonists and the risk of cardiac-valve regurgitation. N Engl J Med 2007; 356(1): 29–38

    Article  PubMed  CAS  Google Scholar 

  80. Zanettini R, Antonini A, Gatto G, et al. Valvular heart disease and the use of dopamine agonists for Parkinson’s disease. N Engl J Med 2007; 356(1): 39–46

    Article  PubMed  CAS  Google Scholar 

  81. Lu C, Bharmal A, Suchowersky O. Gambling and Parkinson disease [letter]. Arch Neurol 2006; 63(2): 298

    Article  PubMed  Google Scholar 

  82. Nirenberg MJ, Waters C. Compulsive eating and weight gain related to dopamine agonist use. Mov Disord 2006; 21(4): 524–9

    Article  PubMed  Google Scholar 

  83. Stocchi F. Pathological gambling in Parkinson’s disease. Lancet Neurol 2005; 4(10): 590–2

    Article  PubMed  Google Scholar 

  84. Prescrire editorial staff. Hypersexuality due to dopaminergic drugs. Prescrire Int 2005; 14(80): 224

    Google Scholar 

  85. Driver-Dunckley E, Samanta J, Stacy M. Pathological gambling associated with dopamine agonist therapy in Parkinson’s disease. Neurology 2003; 61(3): 422–3

    Article  PubMed  CAS  Google Scholar 

  86. Homann CN, Wenzel K, Suppan K, et al. Sleep attacks in patients taking dopamine agonists: review. BMJ 2002; 324(7352): 1483–7

    Article  PubMed  CAS  Google Scholar 

  87. Etminan M, Gill S, Samii A. Comparison of the risk of adverse events with pramipexole and ropinirole in patients with Parkinson’s disease: a meta-analysis. Drug Saf 2003; 26(6): 439–44

    Article  PubMed  CAS  Google Scholar 

  88. Low-dose clozapine for the treatment of drug-induced psychosis in Parkinson’s disease. The Parkinson Study Group. N Engl J Med 1999; 340 (10): 757-63

  89. Durif F, Debilly B, Galitzky M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology 2004; 62(3): 381–8

    Article  PubMed  CAS  Google Scholar 

  90. Diamond A, Jankovic J. Treatment of advanced Parkinson’s disease. Expert Rev Neurother 2006; 6(8): 1181–97

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

No sources of funding were used to assist in the preparation of this review. Dr Jankovic has acted as a consultant for Allergan, Eisai, Merz and Teva, and has received grants from Allergan, Amersham, Boehringer-Ingelheim, Cergene, Medtronic, Merz, Prestwick, Teva and Schwarz Pharma. Dr Stacy has acted as a consultant for and/or received honoraria and grants from GlaxoSmithKline, Novartis, Boehringer-Ingelheim, Vernalis, EMD Biosciences, Schantz and Schwarz Pharma.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Joseph Jankovic.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jankovic, J., Stacy, M. Medical Management of Levodopa-Associated Motor Complications in Patients with Parkinson’s Disease. CNS Drugs 21, 677–692 (2007). https://doi.org/10.2165/00023210-200721080-00005

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00023210-200721080-00005

Keywords

Navigation