Trending Topic

3D illustration of human brain on black background
23 mins

Trending Topic

Developed by Touch
Mark CompleteCompleted
BookmarkBookmarked
Joseph Samaha, Jim Dagher, Shayan Abdollah Zadegan

Huntington’s disease (HD) is a neurodegenerative disease inherited in an autosomal dominant manner. It is caused by an expansion of cytosine, adenine, guanine (CAG) repeats within the huntingtin (HTT) gene, which is located on chromosome 4. This pathological expansion of CAG repeats results in the production of a mutant huntingtin protein with an abnormally long polyglutamine […]

Subcutaneous Apomorphine Infusion – An Update

Regina Katzenschlager
Share
Facebook
X (formerly Twitter)
LinkedIn
Via Email
Mark CompleteCompleted
BookmarkBookmarked
Copy LinkLink Copied
Download as PDF
Published Online: Jul 15th 2012 European Neurological Review, 2012;7(Suppl. 1):8–12 DOI: http://doi.org/10.17925/ENR.2012.07.S1.8
Select a Section…
1

Abstract

Overview

Continuous delivery of dopaminergic drugs is an important treatment strategy to delay or reverse motor complications in Parkinson’s disease (PD). Subcutaneous apomorphine (APO) infusion has been shown (in uncontrolled studies) to significantly reduce ‘off’ time and dyskinesia duration and severity, and long-term data show the beneficial effects persist for several years. There is some evidence that the maximum antidyskinetic effect of APO infusion may be attained when oral medications are reduced or discontinued, making monotherapy an important clinical goal. Recent studies demonstrate possible positive effects of APO infusion on the non-motor symptoms of PD. However, more trials are needed to assess the neuropsychiatric effects of this treatment. Moreover, randomised controlled trials are needed to compare APO infusion with best medical treatment and with other invasive treatments such as levodopa/carbidopa intestinal gel infusion and deep brain stimulation.

Keywords

Parkinson’s disease, levodopa, continuous dopaminergic stimulation, subcutaneous apomorphine, non-motor symptoms

2

Article

The management of the later stages of Parkinson’s disease (PD) is greatly impacted by non-dopaminergic problems, such as dementia, depression and falls, and by the emergence of motor complications including motor fluctuations and dyskinesias. Motor fluctuations, such as ‘wearing off’ and unpredictable ‘off’, affect 30–100 % of patients.1–4 Dyskinesias can be ‘on’ (mostly choreatic), biphasic (often dystonic) or ‘off’ (dystonic). In the later stages of the disease, there is a loss of nigrostriatal neurons and a concomitant loss of storage capacity. Positron emission tomography imaging of dyskinetic and non-dyskinetic patients showed no difference in dopamine receptor binding, which suggests that dyskinesias are unlikely to be the effect of alterations in striatal dopamine receptor binding.5 On the other hand, levodopa-induced changes in synaptic dopamine levels increase with the progression of PD.6 These changes in synaptic dopamine concentration may be a factor in the emergence of peak-dose dyskinesias.

The pharmacokinetics of levodopa in the periphery, such as plasma half-life clearance, volume of distribution and maximum plasma concentrations, remain unchanged.7 However, the absorption f oral levodopa, which takes place primarily in the duodenum, is affected as gastric emptying becomes more erratic.8,9 Pharmacodynamic postsynaptic striatal changes in gene expression,10 neuropeptide formation11 and discharge patterns of the basal ganglia12 result in complex feedback loops.13 Furthermore, non-dopaminergic factors such as glutamate, opioids and serotoninmay be involved in the development of dyskinesia.14 Sprouting of extrasynaptic dopaminergic terminals may also lead to dysregulateddopamine release.15

One of the most important factors associated with the risk of motor complications is the degree of neuronal loss. In rats whose nigrostriatal system had been lesioned unilaterally by 6-hydroxydopamine, the level of levodopa-induced motor complications was related to lesion size.16 In humans, if the first dose of levodopa is given at an advanced stage of the disease, motor complications may develop within a matter of weeks.17 Furthermore, patients with 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP)-induced chronic and severe parkinsonism developed dyskinesias or ‘on-off’ fluctuations within months of starting levodopa treatment.14 Additional factors that contribute to a greater risk of motor complications include younger age at disease onset,3,18 lower bodyweight19,20 and genetic factors.21

To view the full article in PDF or eBook formats, please click on the icons above.

2

References

  1. Ahlskog JE, Muenter MD, Frequency of levodopa-related
    dyskinesias and motor fluctuations as estimated from the
    cumulative literature, Mov Disord, 2001;16:448–58.

  2. 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;20:726–33.

  3. Schrag A, Quinn N, Dyskinesias and motor fluctuations in
    Parkinson’s disease. A community-based study, Brain,
    2000;123:2297–305.

  4. Parkinson Study Group, Impact of deprenyl and tocopherol
    treatment on Parkinson’s disease in DATATOP patients
    requiring levodopa, Ann Neurol, 1996;39:37–45.

  5. Turjanski N, Lees AJ, Brooks DJ, In vivo studies on striatal
    dopamine D1 and D2 site binding in L-dopa-treated
    Parkinson’s disease patients with and without dyskinesias,
    Neurology, 1997;49:717–23.

  6. de la Fuente-Fernández R, Sossi V, Huang Z, et al.,
    Levodopa-induced changes in synaptic dopamine levels
    increase with progression of Parkinson’s disease:
    implications for dyskinesias, Brain, 2004;127:2747–54.

  7. Nutt JG, Woodward WR, Carter JH, Gancher ST, Effect of
    long-term therapy on the pharmacodynamics of levodopa.
    Relation to on-off phenomenon, Arch Neurol,
    1992;49:1123–30.

  8. Stocchi F, The hypothesis of the genesis of motor
    complications and continuous dopaminergic stimulation in
    the treatment of Parkinson’s disease, Parkinsonism Relat
    Disord, 2009;15(Suppl. 1):S9–S15.

  9. Nutt JG, Fellman JH, Pharmacokinetics of levodopa,
    Clin Neuropharmacol, 1984;7:35–49.

  10. Gerfen CR, Engber TM, Mahan LC, et al., D1 and D2
    dopamine receptor-regulated gene expression of striatonigral
    and striatopallidal neurons, Science, 1990;250:1429–32.

  11. Morissette M, Goulet M, Soghomonian JJ, et al.,
    Preproenkephalin mRNA expression in the caudateputamen
    of MPTP monkeys after chronic treatment with the
    D2 agonist U91356A in continuous or intermittent mode of
    administration: comparison with L-DOPA therapy, Brain Res
    Mol Brain Res, 1997;49:55–62.

  12. Obeso JA, Rodríguez-Oroz MC, Benitez-Temino B, et al.,
    Functional organization of the basal ganglia: therapeutic
    implications for Parkinson’s disease, Mov Disord,
    2008;23(Suppl. 3):S548–59.

  13. Jenner P, Molecular mechanisms of L-DOPA-induced
    dyskinesia, Nat Rev Neurosci, 2008;9:665–77.

  14. Carta M, Carlsson T, Kirik D, Björklund A, Dopamine released
    from 5-HT terminals is the cause of L-DOPA-induced
    dyskinesia in parkinsonian rats, Brain, 2007;130:1819–33.

  15. Lee J, Zhu WM, Stanic D, et al., Sprouting of dopamine
    terminals and altered dopamine release and uptake in
    Parkinsonian dyskinaesia, Brain, 2008;131:1574–87.

  16. Papa SM, Engber TM, Kask AM, Chase TN, Motor
    fluctuations in levodopa treated parkinsonian rats: relation
    to lesion extent and treatment duration, Brain Res,
    1994;662:69–74.

  17. Onofrj M, Paci C, Thomas A, Sudden appearance of
    invalidating dyskinesia-dystonia and off fluctuations after
    the introduction of levodopa in two dopaminomimetic drug
    naive patients with stage IV Parkinson’s disease, J Neurol
    Neurosurg Psychiatry, 1998;65:605–6.

  18. Kumar N, Van Gerpen JA, Bower JH, Ahlskog JE, Levodopadyskinesia
    incidence by age of Parkinson’s disease onset,
    Mov Disord, 2005;20:342–4.

  19. Arabia G, Zappia M, Bosco D, et al., Body weight, levodopa
    pharmacokinetics and dyskinesia in Parkinson’s disease,
    Neurol Sci, 2002;23(Suppl. 2):S53–4.

  20. Sharma JC, Macnamara L, Hasoon M, et al., Cascade of
    levodopa dose and weight-related dyskinesia in Parkinson’s
    disease (LD-WD-PD cascade), Parkinsonism Relat Disord,
    2006;12:499–505.

  21. Foltynie T, Cheeran B, Williams-Gray CH, et al., BDNF
    val66met influences time to onset of levodopa induced
    dyskinesia in Parkinson’s disease, J Neurol Neurosurg Psychiatry,
    2009;80:141–4.

  22. Grandas F, Galiano ML, Tabernero C, Risk factors for
    levodopa-induced dyskinesias in Parkinson’s disease,
    J Neurol, 1999;246:1127–33.

  23. Olanow CW, Obeso JA, Stocchi F, Continuous dopaminereceptor
    treatment of Parkinson’s disease: scientific rationale
    and clinical implications, Lancet Neurol, 2006;5:677–87.

  24. Rascol O, Brooks DJ, Korczyn AD, et al., 056 Study Group,
    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;342:1484–91.

  25. Lees AJ, Katzenschlager R, Head J, Ben-Shlomo Y, Ten-year
    follow-up of three different initial treatments in de-novo PD:
    a randomized trial, Neurology, 2001;57:1687–94.

  26. Bracco F, Battaglia A, Chouza C, et al., The long-acting
    dopamine receptor agonist cabergoline in early Parkinson’s
    disease: final results of a 5-year, double-blind, levodopacontrolled
    study, CNS Drugs, 2004;18:733–46.

  27. Katzenschlager R, Head J, Schrag A, et al., Fourteen-year
    final report of the randomized PDRG-UK trial comparing
    three initial treatments in PD, Neurology, 2008;71:474–80.

  28. Nilsson D, Nyholm D, Aquilonius SM, Duodenal levodopa
    infusion in Parkinson’s disease – long-term experience,
    Acta Neurol Scand, 2001;104:343–8.

  29. Nyholm D, Askmark H, Gomes-Trolin C, et al., Optimizing
    levodopa pharmacokinetics: intestinal infusion versus oral
    sustained-release tablets, Clin Neuropharmacol,
    2003;26:156–63.

  30. Syed N, Murphy J, Zimmerman T, Jr., et al., Ten years’
    experience with enteral levodopa infusions for motor
    fluctuations in Parkinson’s disease, Mov Disord,
    1998;13:336–8.

  31. Colzi A, Turner K, Lees AJ, Continuous subcutaneous
    waking day apomorphine in the long term treatment of
    levodopa induced interdose dyskinesias in Parkinson’s
    disease, J Neurol Neurosurg Psychiatry, 1998;64:573–6.

  32. Manson AJ, Turner K, Lees AJ, Apomorphine monotherapy
    in the treatment of refractory motor complications of
    Parkinson’s disease: long-term follow-up study of 64
    patients, Mov Disord, 2002;17:1235–41.

  33. Katzenschlager R, Hughes A, Evans A, et al., Continuous
    subcutaneous apomorphine therapy improves dyskinesias
    in Parkinson’s disease: a prospective study using
    single-dose challenges, Mov Disord, 2005;20:151–7.

  34. Dewey RB Jr, Maraganore DM, Ahlskog JE, Matsumoto JY,
    Intranasal apomorphine rescue therapy for parkinsonian
    ‘off’ periods, Clin Neuropharmacol, 1996;19:193–201.

  35. Dewey RB Jr, Hutton JT, LeWitt PA, Factor SA,
    A randomized, double-blind, placebo-controlled trial of
    subcutaneously injected apomorphine for parkinsonian
    off-state events, Arch Neurol, 2001;58:1385–92.

  36. Frankel JP, Lees AJ, Kempster PA, Stern GM, Subcutaneous
    apomorphine in the treatment of Parkinson’s disease,
    J Neurol Neurosurg Psychiatry, 1990;53:96–101.

  37. Hughes AJ, Bishop S, Kleedorfer B, et al., Subcutaneous
    apomorphine in Parkinson’s disease: response to chronic
    administration for up to five years, Mov Disord,
    1993;8:165–70.

  38. Wenning GK, Bösch S, Luginger E, et al., Effects of longterm,
    continuous subcutaneous apomorphine infusions on
    motor complications in advanced Parkinson’s disease, Adv Neurol, 1999;80:545–8.

  39. Poewe W, Kleedorfer B, Wagner M, et al., Continuous
    subcutaneous apomorphine infusions for fluctuating
    Parkinson’s disease. Long-term follow-up in 18 patients,
    Adv Neurol, 1993;60:656–9.

  40. Kanovsky P, Kubová D, Bares M, et al., Levodopa-induced
    dyskinesias and continuous subcutaneous infusions of
    apomorphine: results of a two-year, prospective follow-up,
    Mov Disord, 2002;17:188–91.

  41. Stocchi F, Vacca L, De Pandis MF, et al., Subcutaneous
    continuous apomorphine infusion in fluctuating patients
    with Parkinson’s disease: long-term results, Neurol Sci,
    2001;22:93–4.

  42. Antonini A, Isaias IU, Rodolfi G, et al., A 5-year prospective
    assessment of advanced Parkinson disease patients treated
    with subcutaneous apomorphine infusion or deep brain
    stimulation, J Neurol, 2011;258:579–85.

  43. García Ruiz PJ, Sesar Ignacio A, Ares Pensado B, et al.,
    Efficacy of long-term continuous subcutaneous
    apomorphine infusion in advanced Parkinson’s disease with
    motor fluctuations: a multicenter study, Mov Disord,
    2008;23:1130–6.

  44. Sixel-Döring F, Klinke H, Hahn K, et al., [Subcutaneous
    apomorphine infusion therapy in advanced Parkinson’s
    disease: Long-term follow-up of effectivity, tolerability and
    patient satisfaction], Akt Neurol, 2011;38:S27–S33.

  45. Manson AJ, Hanagasi H, Turner K, et al., Intravenous
    apomorphine therapy in Parkinson’s disease: clinical and
    pharmacokinetic observations, Brain, 2001;124:331–40.

  46. Martinez-Martin P, Reddy P, Antonini A, et al., Chronic
    subcutaneous infusion therapy with apomorphine in
    advanced Parkinson’s disease compared to conventional
    therapy: a real life study of non motor effect, J Parkinsons Dis,
    2011;1:197–203.

  47. Alegret M, Valldeoriola F, Martí M, et al., Comparative
    cognitive effects of bilateral subthalamic stimulation and
    subcutaneous continuous infusion of apomorphine in
    Parkinson’s disease, Mov Disord, 2004;19:1463–9.

  48. De Gaspari D, Siri C, Landi A, et al., Clinical and
    neuropsychological follow up at 12 months in patients with
    complicated Parkinson’s disease treated with subcutaneous
    apomorphine infusion or deep brain stimulation of the
    subthalamic nucleus, J Neurol Neurosurg Psychiatry,
    2006;77:450–3.

  49. Morgante L, Basile G, Epifanio A, et al., Continuous
    apomorphine infusion (CAI) and neuropsychiatric
    disorders in patients with advanced Parkinson’s disease:
    a follow-up of two years, Arch Gerontol Geriatr Suppl,
    2004:291–6.

  50. Di Rosa AE, Epifanio A, Antonini A, et al., Continuous
    apomorphine infusion and neuropsychiatric disorders: a
    controlled study in patients with advanced Parkinson’s
    disease, Neurol Sci, 2003;24:174–5.

  51. Deleu D, Hanssens Y, Northway MG, Subcutaneous
    apomorphine : an evidence-based review of its use in
    Parkinson’s disease, Drugs Aging, 2004;21:687–709.

  52. Kehr J, Hu XJ, Goiny M, Scheller DK, Continuous delivery of
    rotigotine decreases extracellular dopamine suggesting
    continuous receptor stimulation, J Neural Transm,
    2007;114:1027–31.

  53. Hely MA, Reid WG, Adena MA, et al., The Sydney
    multicenter study of Parkinson’s disease: the inevitability of
    dementia at 20 years, Mov Disord, 2008;23:837–44.

3

Article Information

Disclosure

Regina Katzenschlager has received consulting and lecturing fees from Abbott, Cephalon, Lundbeck, Teva, Boehringer, GlaxoSmithKline, Novartis and Genus.

Correspondence

Regina Katzenschlager, Department of Neurology, Danube Hospital, Langobardenstr. 122, 1220 Vienna, Austria. E: regina.katzenschlager@chello.at

Support

The V International Forum on Parkinson’s Disease (Helsinki, Finland, 6–7 May 2011) was funded by an unrestricted educational grant from Abbott. Abbott funded the development of this supplement by ESP Bioscience (Crowthorne, UK). Emily Chu and Nicole Meinel of ESP Bioscience provided medical writing and editorial support to the author in the development of this publication. Abbott had the opportunity to review and comment on the publication’s content; however, all decisions regarding content were made by the author.

Received

2013-06-22T00:00:00

4

Further Resources

Share
Facebook
X (formerly Twitter)
LinkedIn
Via Email
Mark CompleteCompleted
BookmarkBookmarked
Copy LinkLink Copied
Download as PDF
Close Popup