Aktuelle Neurologie 2013; 40(06): 338-342
DOI: 10.1055/s-0033-1343255
Übersicht
© Georg Thieme Verlag KG Stuttgart · New York

Stellenwert von L-Dopa in der Therapie der Parkinson-Krankheit

Value of Levodopa Therapy for Early and Advanced Parkinson’s Disease
H. Baas
1   Klinik für Neurologie, Klinikum Hanau GmbH
,
J. Hagenah
2   Klinik für Neurologie, Westküstenklinikum Heide
,
M. Hahne
3   Neurologische Klinik GmbH, Bad Neustadt a. d. Saale
,
C. Redecker
4   Klinik für Neurologie, Universitätsklinikum Jena
,
L. Wojtecki
5   Klinik für Neurologie & Institut für Klinische Neurowissenschaften und med. Psychologie, Heinrich-Heine-Universität, Düsseldorf
,
U. Wüllner
6   Klinik für Neurologie, Universitätsklinikum Bonn
› Author Affiliations
Further Information

Publication History

Publication Date:
14 August 2013 (online)

Zusammenfassung

Dopaminersatzstrategien sind die tragende Säule der pharmakologischen Behandlung der Parkin-son-Krankheit. Zur Verfügung stehen L-Dopa, Dopamin-Agonisten (DA) und Hemmstoffe des Do-pamin-Abbaus (Mono-aminooxydase-B- und Catechol-O-methyl-transferase-Hemmer). Amantadin und Budipin wirken vorwiegend als NMDA-Rezeptorantagonisten modulieren u. a. aber auch die Dopaminfreisetzung. Während der Einsatz von L-Dopa gegenüber den DA aufgrund des Auf-tretens motorischer Komplikationen und neurotoxischer Effekte kritisch diskutiert wurde, legen die ELLdopa-Studie und Langzeituntersuchungen nahe, den Stellenwert der L-Dopa-Therapie neu zu bewerten. Der Einsatz von L-Dopa und DA (wie der anderen Substanzen auch) muss sich an den individuellen Patientenbedürfnissen und dem Wirkungs- und Nebenwirkungsprofil orientieren. Ziel ist es, Symptome so zu beherrschen, dass berufliche Kompetenz und soziale Selbstständigkeit mit möglichst wenig Nebenwirkungen erhalten werden. Eine klare Empfehlung zur Therapieeinleitung kann nicht pauschal für alle Patienten gegeben werden. Studiendaten für die Wirksamkeit einer Kombination aus DA, MAOB-Hemmer und Amantadin oder Budipin fehlen. Das Risiko, Dyskinesien zu entwickeln, wird nicht nur durch die L-Dopa-Dosis, sondern auch durch Erkrankungsalter, Geschlecht und Körpergewicht sowie durch unbekannte hereditäre Komponenten beeinflusst. Patienten, die im Verlauf der Erkrankung wahrscheinlich motorische Komplikationen erleben könnten, sollten vorzugsweise mit DA behandelt werden, eine frühe Kombinationstherapie mit L-Dopa (< 400 mg Tagesdosis) erhöht das Dyskinesierisiko nur moderat. Der Stellenwert von L-Dopa steigt mit zunehmendem Alter und Schwere der motorischen Symptomatik sowie bei Vorliegen von Risikofaktoren für Impulskontrollstörungen.

Abstract

Dopamine substitution strategies are the main component of the pharmacotherapy of Parkinson’s disease. Available drugs include levodopa, dopamine agonists (DA) and inhibitors of dopamine degradation (monoaminooxidase B and catechol-O-methyl-transferase inhibitors). Amantadine and budipine primarily act as NMDA receptor Antagonists, but among other (also anticholinergic) effects modulate Dopamine levels. The use of levodopa has been the subject of critical discussions due to the increased occurrence of motor complications and putative neurotoxic effects. However, the ELLdopa trial and other long-term trials suggest that levodopa therapy should be reassesed. The use of levodopa and various DAs (as well as other substances) must be adapted to the requirements of the individual patient and the side effect profiles. The aim is to control the symptoms to such an extent that professional and social self-sufficiency can be maintained with as few side effects as possible. Recommendations how to start therapy cannot be given for all patients equally well. Trial data on the efficacy of combinations of DA, MAOB inhibitors and amantadine or budipine are lacking. The risk to develop dyskinesias is associated not only with levodopa dosage, but also with disease onset and probably gender and body weight as well as with still unknown hereditary components. Patients who are likely to experience motor complications in the disease course should be treated preferentially with DA, however a combination therapy with levodopa (< 400 mg daily dose) seems to increase daskinesia risk only modestly. The utility of levodopa increases with increasing age and severity of motor symptoms as well as in the presence of risk factors for impulse control disorders.

 
  • Literatur

  • 1 Fahn S. and the Parkinson Study Group. Does levodopa slow or hasten the rate of progression of Parkinson’s disease?. J Neurol 2005; 252 (Suppl. 04) IV37-IV42
  • 2 Parkkinen L, O’Sullivan SS, Kuoppamäki M et al. Does levodopa accelerate the pathologic process in Parkinson disease brain?. Neurology 2011; 77: 1420-1426
  • 3 Rajput AH. Levodopa prolongs life expectancy and is non-toxic to substantia nigra. Parkinsonism Relat Disord 2001; 8: 95-100
  • 4 Müller T, van Laar T, Cornblath DR et al. Peripheral neuropathy in Parkinson’s disease: levodopa exposure and implications for duodenal delivery. Parkinsonism Relat Disord 2013; 19: 501-507
  • 5 Stowe RL, Ives NJ, Clarke C et al. Dopamine agonist therapy in early Parkinson’s disease. Cochrane Database Syst Rev 2008; (2) CD006564
  • 6 Biglan KM, Holloway Jr RG, McDermott MP et al. Parkinson Study Group CALM-PD Investigators. Risk factors for somnolence, edema, and hallucinations in early Parkinson disease. Neurology 2007; 69: 187-195
  • 7 Hauser RA, Rascol O, Korczyn AD et al. Ten-year follow-up of Parkinson’s disease patients randomized to initial therapy with ropinirole or levodopa. Mov Disord 2007; 22: 2409-2417
  • 8 Paus S, Brecht HM, Köster J et al. Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s disease. Mov Disord 2003; 18: 659-667
  • 9 Barone P. Treatment of depressive symptoms in Parkinson’s disease. Eur J Neurol 2011; 18 (Suppl. 01) 11-15
  • 10 Schade R, Andersohn F, Suissa S et al. Dopamine agonists and the risk of cardiac-valve regurgitation. N Engl J Med 2007; 356: 29-38
  • 11 Goetz CG, Leurgans S, Pappert EJ et al. Prospective longitudinal assessment of hallucinations in Parkinson’s disease. Neurology 2001; 57: 2078-2082
  • 12 Weintraub D, Koester J, Potenza MN et al. Impulse control disorders in Parkinson disease: a cross-sectional study of 3090 patients. Arch Neurol 2010; 67: 589-595
  • 13 Bostwick JM, Hecksel KA, Stevens SR et al. Frequency of new-onset pathologic compulsive gambling or hypersexuality after drug treatment of idiopathic Parkinson disease. Mayo Clin Proc 2009; 84: 310-316
  • 14 Karlsen KH, Tandberg E, Arsland D et al. Health related quality of life in Parkinson’s disease: a prospective longitudinal study. J Neurol Neurosurg Psychiatry 2000; 69: 584-589
  • 15 Katzenschlager R, Head J, Schrag A et al. Parkinson’s Disease Research Group of the United Kingdom. Fourteen-year final report of the randomized PDRG-UK trial comparing three initial treatments in PD. Neurology 2008; 71: 474-480
  • 16 Barbeau A, Roy M. Six-year results of treatment with levodopa plus benzerazide in Parkinson’s disease. Neurology 1976; 26: 399-404
  • 17 Rajput AH, Stern W, Laverty WH. Chronic low-dose levodopa therapy in Parkinson’s disease: An argument for delaying levodopa therapy. Neurology 1984; 34: 991-996
  • 18 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. 056 Study Group. N Engl J Med 2000; 342: 1484-1491
  • 19 Rinne UK, Bracco F, Chouza C et al. Early treatment of Parkinson’s disease with cabergoline delays the onset of motor complications. Results of a double-blind levodopa controlled trial. The PKDS009 Study Group. Drugs 1998; 55 (Suppl. 01) 23-30
  • 20 Przuntek H, Conrad B, Dichgans J et al. SELEDO: a 5-year long-term trial on the effect of selegiline in early Parkinsonian patients treated with levodopa. Eur J Neurol 1999; 6: 141-150
  • 21 Stocchi F, Rascol O, Kieburtz K et al. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: the STRIDE-PD study. Ann Neurol 2010; 68: 18-27
  • 22 Schapira AH, Poewe W, Kieburtz K, Rascol O, Stocchi F, Nissinen H, Leinonen M, Olanow W. for the STRIDE-PD investigators. Poster presented at the 16th International Congress of Parkinson’s Disease and Movement Disorders 17–21 June. Dublin, Ireland: 2012
  • 23 S2-Leitlinien der Deutschen Gesellschaft für Neurologie. http://www.awmf.org/leitlinien/detail/ll/030-010.html
  • 24 Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson’s disease: a 4-year randomized controlled trial. Arch Neurol 2004; 61: 1044-1053
  • 25 Parkinson Study Group. Long-term effect of initiating pramipexole vs levodopa in early Parkinson disease. Arch Neurol 2009; 66: 563-570
  • 26 Chase TN, Oh JD. Striatal dopamine- and glutamate-mediated dysregulation in experimental parkinsonism. Trends Neurosci 2000; 23 (Suppl. 01) 86-91
  • 27 Marin C, Papa S, Engber TM et al. MK-801 prevents levodopa-induced motor response alterations in parkinsonian rats. Brain Res 1996; 736: 202-205
  • 28 Blanchet PJ, Calon F, Martel JC et al. Continuous administration decreases and pulsatile administration increases behavioral sensitivity to a novel dopamine D2 agonist (U-91356A) in MPTP exposed monkeys. J Pharmacol Expt Ther 1995; 272: 854-859
  • 29 LeWitt PA, Guttman M, Tetrud JW et al. 6002-US-005 Study Group. Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces “off” time in Parkinson’s disease: a double-blind, randomized, multicenter clinical trial (6002-US-005). Ann Neurol 2008; 63: 295-302
  • 30 Wills AM, Eberly S, Tennis M et al. on behalf of the Parkinson Study Group. Caffeine consumption and risk of dyskinesia in CALM-PD. Mov Disord 2013; epub
  • 31 Shaw KM, Lees AJ, Stern GM. The impact of treatment with levodopa on Parkinson’s disease. Q J Med 1980; 49: 283-293