Skip to main content
Log in

Apomorphin bei der Behandlung des Morbus Parkinson

  • Übersichten
  • Published:
Der Nervenarzt Aims and scope Submit manuscript

Zusammenfassung

Apomorphin ist ein seit langem bekannter hochpotenter D2-, D3- und D4-Dopaminrezeptoragonist, der sich besonders durch eine zusätzliche D1-Dopaminrezeptorwirkung auszeichnet und in der Medizin vielfältige Anwendung gefunden hat. Wegen einer weitestgehenden Inaktivierung bei der Leberpassage wird Apomorphin in der Regel subkutan angewendet. Nach raschem Wirkungseintritt ist die Wirkungsdauer auf etwa eine Stunde begrenzt. Bei seiner Wirkung ist Apomorphin unabhängig von Mechanismen der enteralen Dopaminresorption und vom präsynaptischen System der Dopaminspeicherung und Dopaminfreisetzung. Die häufigste Nebenwirkung der subkutanen Apomorphingabe sind Gewebsverhärtungen an den Injektionsstellen. Periphere dopaminerge Nebenwirkungen, wie Übelkeit und orthostatische Hypotension, treten meist nur in der Anfangsphase der Behandlung auf und lassen sich mit Domperidon weitgehend unterdrücken. Halluzinationen treten seltener auf als beim Einsatz anderer Dopaminagonisten.

Beim Apomorphintest wird durch subkutane Bolusgaben die Levodopasensitivtät einer Parkinsonsymptomatik zur Abgrenzung eines M. Parkinson von atypischen Parkinsonsyndromen ermittelt. Dabei zeigt der Apomorphintest eine Levodopasensitivität in etwa 90% der Fälle an. Falsch-negative Ergebnisse können bei gering ausgeprägter Parkinsonsymptomatik vorkommen. Eine Metaanalyse zeigt, daß durch eine intermittierende subkutane Apomorphingabe (intermittierende Apomorphintherape) mittels eines Selbstapplikationsgeräts die tägliche Dauer unvorhersehbarer kurz anhaltender Off-Phasen um 48.8±8.7% reduziert werden kann. Dazu werden durchschnittlich 3,7±1,1 × 2,9±0,8 mg Apomorphin pro Tag appliziert. Durch eine kontinuierliche subkutane Apomorphingabe mittels einer Pumpe (kontinuierliche Apomorphintherapie) während der Wachphase läßt sich die tägliche Dauer der Off-Phasen um 64,8±13,5% und die zusätzlich notwendige Levodopadosis um 30,3±31,1% reduzieren. Bei Ganztagesgabe wird die tägliche Dauer der Off-Phasen um 63,5±19,1% und die zusätzlichen Levodopadosis um 65,5±21,9% reduziert. Levodopainduzierte Dyskinesien nehmen deutlich ab. Eine langsame Umstellung der Medikation auf eine Apomorphin-Monotherapie ist möglich. Auch bei langjähriger Anwendung zeigen sich keine wesentlichen Wirkungsverluste der Apomorphintherapie. Damit stellt die Apomorphintherapie eine sehr wirkungsvolle Therapie von Off-Phasen und levodopainduzierten Dyskinesien beim fortgeschrittenen Morbus Parkinson dar, die ausgeschöpft werden sollte, bevor invasivere Therapieansätze in Betracht gezogen werden.

Summary

Apomorphine has long been used in many medical specialties. It is a highly potent D2-, D3- and D4-dopamine receptor agonist with a particularly high D1-dopamine receptor affinity. Due to its almost complete inactivation during liver passage it is usually applied subcutaneously. After rapid onset its effect is waning after about one hour. Apomorphine’s action is not dependent on enteral dopamine resorption and on praesynaptic dopamine storage and dopamine secretion mechanisms. When applied subcutaneously its most common side effect is cutaneous nodules at the injection sites. Peripheral dopaminergic side effects, such as nausea and orthostatic hypotension, usually occur only during therapy initiation and respond well to domperidone. Hallucinations occur less frequently than with other dopamine receptor agonists.

The apomorphine test determines the levodopasensitivity of a parkinsonian syndrome by application of an apomorphine bolus to distinguish between idiopathic Parkinson’s disease and atypical parkinsonian syndromes. It indicates a levodopasensitivity in about 90% of the patients tested. False-negative results can occur in mild parkinsonian syndromes. A meta analysis indicates that intermittend subutaneous apomorphine applications (intermittend apomorphine therapy) with a self application device can reduce the daily duration of unpredictable off-phases by 48.8±8.7%. For this an average of 3.7±1.1 × 2.9±0.8 mg apomorphine has to be applied per day. Continuous subcutaneous apomorphine application with an extracorporal pump (continuous apomorhine therapy) during the wake phase reduces the duration of daily off-phases by 64.8±13.5% and the additional levodopa dose by 30.3±31.1%. 24-hour application reduces the daily duration of the off-phases by 63.5±19.1% and the additional levodopa dose by 65.5±21.9%. Levodopa-induced dyskinesias are also substantially reduced. A gradual conversion to an apomorphine monotherapy is possible. Even after prolonged treatment times no significant loss of efficacy occurs. With this profile apomorphine is a highly successful treatment option for off-phases and levodopa-induced dyskinesias in advanced cases of idiopathic Parkinson’s disease which should be used before more invasive approaches are considered.

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.

Abb. 1
Abb. 2
Abb. 3

Literatur

  1. Acland KM, Churchyard A, Fletcher CL et al. (1998) Panniculitis in association with apomorphine infusion. Br J Dermatol 138:480–482

    Article  CAS  PubMed  Google Scholar 

  2. Bonuccelli U, Piccini P, Del Dotto P et al. (1993) Apomorphine test for dopaminergic responsiveness: a dose assessment study. Mov Disord 8:158–164

    CAS  PubMed  Google Scholar 

  3. Chaudhuri KR, Critchley P, Abbott RJ et al. (1988) Subcutaneous apomorphine for on-off oscillations in Parkinson’s disease. Lancet 2 (8622):1260

    Article  CAS  Google Scholar 

  4. Clarke CE, Davies P (2000) Systematic review of acute levodopa and apomorphine challenge tests in the diagnosis of idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry 69:590–594

    Article  CAS  PubMed  Google Scholar 

  5. Colzi A, Turner K, Lees AJ (1998) Continuous subcutaneous waking day apomorphine in the long term treatment of levodopa induced interdose dyskinesias in Parkinson’s disease. J Neurol Neurosurg Psychiatry 64:573–576

    CAS  PubMed  Google Scholar 

  6. Corsini GU, Pitzalis GF, Bernardi F et al. (1981) The use of dopamine agonists in the treatment of schizophrenia. Neuropharmacology 20:1309–1313

    CAS  PubMed  Google Scholar 

  7. D’Costa DF, Abbott RJ, Pye IF et al. (1991) The apomorphine test in parkinsonian syndromes. J Neurol Neurosurg Psychiatry 54:870–872

    PubMed  Google Scholar 

  8. Dewey RB Jr, Maraganore DM, Ahlskog JE et al. (1996) Intranasal apomorphine rescue therapy for parkinsonian “off” periods. Clin Neuropharmacol 19:193–201

    CAS  PubMed  Google Scholar 

  9. Dewey RB Jr, Hutton JT, LeWitt PA et al. (2001) A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch Neurol 58:1385–1392

    Article  PubMed  Google Scholar 

  10. Di Chiara G, Gessa GL (1978) Pharmacology and neurochemistry of apomorphine. Adv Pharmacol Chemother 15:87–160

    PubMed  Google Scholar 

  11. Eardley I, Wright P, MacDonagh R et al. (2004) An open-label, randomized, flexible-dose, crossover study to assess the comparative efficacy and safety of sildenafil citrate and apomorphine hydrochloride in men with erectile dysfunction. BJU Int 93:1271–1275

    Article  CAS  PubMed  Google Scholar 

  12. Ellis C, Lemmens G, Parkes JD et al. (1997) Use of apomorphine in parkinsonian patients with neuropsychiatric complications to oral treatment. Parkinsonism Relat Disord 3:103–107

    Article  Google Scholar 

  13. Esteban Munoz J, Marti MJ, Marin C et al. (1997) Long-term treatment with intermitent intranasal or subcutaneous apormorphine in patients with levodopa-related motor fluctuations. Clin Neuropharmacol 20:245–252

    PubMed  Google Scholar 

  14. Galvez-Jimenez N, Lang AE (1996) Perioperative problems in Parkinson’s disease and their management: apomorphine with rectal domperidone. Can J Neurol Sci 23:198–203

    CAS  PubMed  Google Scholar 

  15. Gancher ST, Woodward WR, Boucher B et al. (1989) Peripheral pharmacokinetics of apomorphine in humans. Ann Neurol 26:232–238

    CAS  PubMed  Google Scholar 

  16. Gancher ST, Bennett W, English J (1989) Studies of renal function in animals chronically treated with apomorphine. Res Commun Chem Pathol Pharmacol 66:163–166

    CAS  PubMed  Google Scholar 

  17. Gancher S (1995) Pharmacokinetics of apomorphine in Parkinson’s disease. J Neural Transm Suppl 45:137–141

    CAS  PubMed  Google Scholar 

  18. Gancher ST, Nutt JG, Woodward WR (1995) Apomorphine infusional therapy in Parkinson’s disease: clinical utility and lack of tolerance. Mov Disord 10:37–43

    CAS  PubMed  Google Scholar 

  19. Gassen M, Gross A, Youdim MB (1999) Apomorphine, a dopamine receptor agonist with remarkable antioxidant and cytoprotective properties. Adv Neurol 80:297–302

    CAS  PubMed  Google Scholar 

  20. Grunblatt E, Mandel S, Berkuzki T et al. (1999) Apomorphine protects against MPTP-induced neurotoxicity in mice. Mov Disord 14:612–618

    Article  CAS  PubMed  Google Scholar 

  21. Hagell P, Odin P (2001) Apomorphine in the treatment of Parkinson’s disease. J Neurosci Nurs 33:21–38

    CAS  PubMed  Google Scholar 

  22. Helmers JH (1977) Preliminary report of domperidone (R 33182), a new antiemetic compound. A pilot study. Acta Anaesthesiol Belg 28:245–250

    CAS  PubMed  Google Scholar 

  23. Hughes AJ, Lees AJ, Stern GM (1990) Apomorphine test to predict dopaminergic responsiveness in parkinsonian syndromes. Lancet 336 (8706):32–34

    Article  CAS  PubMed  Google Scholar 

  24. Hughes AJ, Bishop S, Kleedorfer B et al. (1993) Subcutaneous apomorphine in Parkinson’s disease: response to chronic administration for up to five years. Mov Disord 8:165–170

    CAS  PubMed  Google Scholar 

  25. Jenner P (2003) Dopamine agonists, receptor selectivity and dyskinesia induction in Parkinson’s disease. Curr Opin Neurol 16 [Suppl 1]:S3–7

    Google Scholar 

  26. Kanovsky P, Kubova D, Bares M et al. (2002) Levodopa-induced dyskinesias and continuous subcutaneous infusions of apomorphine: results of a two-year, prospective follow-up. Mov Disord 17:188–191

    Article  PubMed  Google Scholar 

  27. Kempster PA, Iansek R, Larmour I (1991) Intermittent subcutaneous apomorphine injection treatment for parkinsonian motor oscillations. Aust N Z J Med 21:314–318

    CAS  PubMed  Google Scholar 

  28. Kreczy-Kleedorfer B, Wagner M, Bosch S et al. (1993) Langzeitergebnisse kontinuierlicher subkutaner Apomorphinpumpentherapie bei Patienten mit fortgeschrittener Parkinson-Krankheit. Nervenarzt 64:221–225

    CAS  PubMed  Google Scholar 

  29. Lees AJ, Montastruc JL, Turjanski N et al. (1989) Sublingual apomorphine and Parkinson’s disease. J Neurol Neurosurg Psychiatry 52:1440

    CAS  PubMed  Google Scholar 

  30. Lees AJ, Richardson C, Turner K (1998) Treatment of Parkinson’s disease with apomorphine, Shared Guidelines, 3rd edn. UCL Hospitals NHS Trust

  31. Manson AJ, Turner K, Lees AJ (2002) Apomorphine monotherapy in the treatment of refractory motor complications of Parkinson’s disease: long-term follow-up study of 64 patients. Mov Disord 17:1235–1241

    Article  PubMed  Google Scholar 

  32. Matthiessen A, Wright CRA (1869) Researches into the chemical constitution of the opium bases Part I: on the action of hydrochloric acid on morhia. Proc R Soc Lond B Biol Sci 17:455–460

    Google Scholar 

  33. Merello M, Leiguarda R (1995) Tratamiento de las fluctuationes motoras de la enfermedad de parkinson con inyecciones subcutaneas de apomorfina. Medicina (B Aires) 55:5–10

  34. Merello M, Pikielny R, Cammarota A et al. (1997) Comparison of subcutaneous apomorphine versus dispersible madopar latency and effect duration in Parkinson’s disease patients: a double-blind single-dose study. Clin Neuropharmacol 20:165–167

    CAS  PubMed  Google Scholar 

  35. Merello M, Piran Arce G, Nouzeilles M et al. (2000) Antidyskinetic effect of long-term waking day continuous infusion of apomorphine compared to posteroventral pallidotomy. Mov Disord 15 [Suppl 3]:54

  36. Muller T, Benz S, Bornke C et al. (2003) Repeated rating improves value of diagnostic dopaminergic challenge tests in Parkinson’s disease. J Neural Transm 110:603–609

    Article  PubMed  Google Scholar 

  37. Ostergaard L, Werdelin L, Odin P et al. (1995) Pen injected apomorphine against off phenomena in late Parkinson’s disease: a double blind, placebo controlled study. J Neurol Neurosurg Psychiatry 58:681–687

    CAS  PubMed  Google Scholar 

  38. Pietz K, Hagell P, Odin P (1998) Subcutaneous apomorphine in late stage Parkinson’s disease: a long term follow up. J Neurol Neurosurg Psychiatry 65:709–716

    CAS  PubMed  Google Scholar 

  39. Podolec Z, Vetulani J, Bednarczyk B et al. (1979) Central dopamine receptors regulate blood eosinophilia in the rat. Allergy 34:103–110

    CAS  PubMed  Google Scholar 

  40. Poewe W, Kleedorfer B, Gerstenbrand F et al. (1989) Die Behandlung von Parkinsonpatienten mit L-Dopa-Wirkungsfluktuation mittels subkutanen Apomorphingaben. Acta Neurol 16:73–77

    Google Scholar 

  41. Pollak P, Champay AS, Gaio JM et al. (1990) Administration sous-cutanee d’apomorphine dans les fuctuations motrices de la maladie de Parkinson. Rev Neurol (Paris) 146:116–122

    CAS  Google Scholar 

  42. Priano L, Albani G, Calderoni S et al. (2002). Controlled-release transdermal apomorphine treatment for motor fluctuations in Parkinson’s disease. Neurol Sci 23 [Suppl 2]:S99–100

    Google Scholar 

  43. Przedborski S, Levivier M, Raftopoulos C et al. (1995) Peripheral and central pharmacokinetics of apomorphine and its effect on dopamine metabolism in humans. Mov Disord 10:28–36

    CAS  PubMed  Google Scholar 

  44. Rodriguez M, Lera G, Vaamonde J et al. (1994) Motor response to apomorphine and levodopa in asymmetric Parkinson’s disease. J Neurol Neurosurg Psychiatry 57:562–566

    CAS  PubMed  Google Scholar 

  45. Schwab RS, Amador LV, Lettvin JY (1951) Apomorphine in Parkinson’s disease. Trans Am Neurol Assoc 76:251–253

    Google Scholar 

  46. Stocchi F, Bramante L, Monge A et al. (1993) Apomorphine and lisuride infusion. A comparative chronic study. Adv Neurol 60:653–655

    CAS  PubMed  Google Scholar 

  47. Stocchi F, Farina C, Nordera G et al. (1999) Implantable venous access system for apomorphine infusion in complicated Parkinson’s disease. Mov Disord 14:358

    Article  CAS  Google Scholar 

  48. van Laar T, Jansen EN, Essink AW et al. (1993) A double-blind study of the efficacy of apomorphine and its assessment in ‘off’-periods in Parkinson’s disease. Clin Neurol Neurosurg 95:231–235

    Article  PubMed  Google Scholar 

  49. van Laar T, Jansen EN, Neef C et al. (1995) Pharmacokinetics and clinical efficacy of rectal apomorphine in patients with Parkinson’s disease: a study of five different suppositories. Mov Disord 10:433–439

    PubMed  Google Scholar 

  50. Wang HC, Hsieh Y (2001) Treatment of neuroleptic malignant syndrome with subcutaneous apomorphine monotherapy. Mov Disord 16:765–767

    Article  CAS  PubMed  Google Scholar 

  51. Weil E (1884) De L’apomorphine dans certain troubles nerveaux. Lyon Med 48:411–419

    Google Scholar 

Download references

Interessenkonflikt:

Der korrespondierende Autor versichert, dass keine Verbindungen mit einer Firma, deren Produkt in dem Artikel genannt ist, oder einer Firma, die ein Konkurrenzprodukt vertreibt, bestehen.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to D. Dressler.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Dressler, D. Apomorphin bei der Behandlung des Morbus Parkinson. Nervenarzt 76, 681–689 (2005). https://doi.org/10.1007/s00115-004-1830-4

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00115-004-1830-4

Schlüsselwörter

Keywords

Navigation