Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-xtgtn Total loading time: 0 Render date: 2024-04-19T02:22:04.842Z Has data issue: false hasContentIssue false

Section I - The Pharmacological Basis for Parkinson's Disease Treatment

Published online by Cambridge University Press:  05 March 2016

Néstor Gálvez-Jiménez
Affiliation:
Cleveland Clinic, Florida
Hubert H. Fernandez
Affiliation:
Cleveland Clinic, Ohio
Alberto J. Espay
Affiliation:
University of Cincinnati
Susan H. Fox
Affiliation:
Toronto Western Hospital
Get access
Type
Chapter
Information
Parkinson's Disease
Current and Future Therapeutics and Clinical Trials
, pp. 1 - 92
Publisher: Cambridge University Press
Print publication year: 2016

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Standaert, DG, Roberson, ED. Treatment of central nervous system degenerative disorders. In: Hardman, JG, Molinoff, PB, Ruddon, RW and Gilman, AG, eds. Goodman and Gilman's The Pharmacological Basis of Therapeutics. New York: McGraw-Hill, 1995; 50319.Google Scholar
Schwab, RS, Tillmann, WR. Artane in the treatment of Parkinson's disease; a report of its effectiveness alone and in combination with benadryl and parpanit. N Engl J Med, 1949; 241: 4835.CrossRefGoogle ScholarPubMed
Ordenstein, L, Sur la Paralysie Agitante et la Sclérose en Plaques Generalisée., 1867; Paris: Martinet (two leaves of color plates).Google Scholar
Goetz, CG. The history of Parkinson's disease: early clinical descriptions and neurological therapies. Cold Spring Harb Perspect Med 2011; 1: a008862.CrossRefGoogle ScholarPubMed
Aosaki, T. Miura, M, Suzuki, T, Nishimura, K, Masuda, M. Acetylcholine-dopamine balance hypothesis in the striatum: an update. Geriatr Gerontol Int 2010; 10 (Suppl. 1): S14857.Google Scholar
Duvoisin, RC. Cholinergic-anticholinergic antagonism in parkinsonism. Arch Neurol 1967; 17: 12436.CrossRefGoogle ScholarPubMed
Zhou, FM, Liang, Y, Dani, JA. Endogenous nicotinic cholinergic activity regulates dopamine release in the striatum. Nat Neurosci 2001; 4: 12249.Google Scholar
Graybiel, AM. Neurochemically specified subsystems in the basal ganglia. Ciba Found Symp 1984; 107: 11449.Google ScholarPubMed
DeLong, M, Wichmann, T. Update on models of basal ganglia function and dysfunction. Parkinsonism Relat Disord 2009; 15 (Suppl. 3): S23740.CrossRefGoogle ScholarPubMed
Bolam, JP. Synapses of identified neurons in the neostriatum. Ciba Found Symp 1984; 107: 3047.Google Scholar
Kawaguchi, Y, Wilson, CJ, Augood, SJ, Emson, PC. Striatal interneurones: chemical, physiological and morphological characterization. Trends Neurosci 1995; 18: 52735.CrossRefGoogle ScholarPubMed
Pakhotin, P, Bracci, E. Cholinergic interneurons control the excitatory input to the striatum. J Neurosci 2007; 27: 391400.Google Scholar
Benarroch, EE. Effects of acetylcholine in the striatum. Recent insights and therapeutic implications. Neurology 2012; 79: 27481.Google Scholar
Rinne, JO, Ma, SY, Lee, MS, Collan, Y, Röyttä, M. Loss of cholinergic neurons in the pedunculopontine nucleus in Parkinson's disease is related to disability of the patients. Parkinsonism Relat Disord 2008; 14: 5537.CrossRefGoogle ScholarPubMed
Jellinger, K. The pedunculopontine nucleus in Parkinson's disease, progressive supranuclear palsy and Alzheimer's disease. J Neurol Neurosurg Psychiatry 1988; 51: 5403.Google Scholar
Benarroch, EE. Pedunculopontine nucleus: functional organization and clinical implications. Neurology 2013; 80: 114855.Google Scholar
Wang, HL, Morales, M. Pedunculopontine and laterodorsal tegmental nuclei contain distinct populations of cholinergic, glutamatergic and GABAergic neurons in the rat. Eur J Neurosci 2009; 29: 34058.Google Scholar
Griffiths, PD, Sambrook, MA, Perry, R, Crossman, AR. Changes in benzodiazepine and acetylcholine receptors in the globus pallidus in Parkinson's disease. J Neurol Sci 1990; 100: 1316.CrossRefGoogle ScholarPubMed
Sweeney, PJ. Parkinson's disease: managing symptoms and preserving function. Geriatrics 1995; 50: 2431.Google Scholar
Quik, M, O'Leary, K, Tanner, CM. Nicotine and Parkinson's disease: implications for therapy. Mov Disord 2008; 23: 164152.Google Scholar
Ebersbach, G, Stöck, M, Müller, J, et al. Worsening of motor performance in patients with Parkinson's disease following transdermal nicotine administration. Mov Disord 1999; 14: 101113.Google Scholar
Lemay, S, Chouinard, S, Blanchet, P, et al. Lack of efficacy of a nicotine transdermal treatment on motor and cognitive deficits in Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 2004; 28: 319.Google Scholar
Krueger, BK. Kinetics and block of dopamine uptake in synaptosomes from rat caudate nucleus. J Neurochem 1990; 55: 2607.Google Scholar
McDonough, JH Jr, Shih, TM. A study of the N-methyl-d-aspartate antagonistic properties of anticholinergic drugs. Pharmacol Biochem Behav 1995; 51: 24953.Google Scholar
Whiteman, PD, Fowle, AS, Hamilton, MJ, et al. Pharmacokinetics and pharmacodynamics of procyclidine in man. Eur J Clin Pharmacol 1985; 28: 738.Google Scholar
Grimaldi, R, Perucca, E, Ruberto, G, et al. Pharmacokinetic and pharmacodynamic studies following the intravenous and oral administration of the antiparkinsonian drug biperiden to normal subjects. Eur J Clin Pharmacol 1986; 29: 7357.Google Scholar
Yokogawa, K, Nakashima, E, Ishizaki, J, et al. Brain regional pharmacokinetics of biperiden in rats. Biopharm Drug Dispos 1992; 13: 13140.CrossRefGoogle ScholarPubMed
Ishizaki, J, Yokogawa, K, Nakashima, E, Ohkuma, S, Ichimura, F. Influence of ammonium chloride on the tissue distribution of anticholinergic drugs in rats. J Pharm Pharmacol 1998; 50: 7616.Google Scholar
Nakashima, E, Yokogawa, K, Ichimura, F, et al. Effects of fasting on biperiden pharmacokinetics in the rat. J Pharm Sci 1987; 76: 1013.Google Scholar
Paeme, G, Van Bossuyt, H, Vercruysse, A. Phenobarbital induction of procyclidine metabolism: in vitro study. Eur J Drug Metab Pharmacokinet 1982; 7: 22931.Google Scholar
He, H, McKay, G, Midha, KK. Phase I and II metabolites of benztropine in rat urine and bile. Xenobiotica 1995; 25: 85772.Google Scholar
Chew, ML, Mulsant, BH, Pollock, BG, et al. Anticholinergic activity of 107 medications commonly used by older adults. J Am Geriatr Soc 2008; 56: 133341.Google Scholar
Lang, AE. Treatment of Parkinson's disease with agents other than levodopa and dopamine agonists: controversies and new approaches. Can J Neurol Sci 1984; 11 (Suppl.): 21020.CrossRefGoogle ScholarPubMed
Hughes, AJ, Daniel, SE, Kilford, L, Lees, AJ. Accuracy of clinical diagnosis of idiopathic Parkinson's disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry 1992; 55: 1814.Google Scholar
Katzenschlager, R, Sampaio, C, Costa, J, Lees, A. Anticholinergics for symptomatic management of Parkinson's disease. Cochrane Database Syst Rev 2003: CD003735.Google Scholar
Thomsen, TR, Galpern, WR, Asante, A, Arenovich, T, Fox, SH. Ipratropium bromide spray as treatment for sialorrhea in Parkinson's disease. Mov Disord 2007; 22: 226873.Google Scholar
Arbouw, ME. Movig, KL, Koopmann, M, et al. Glycopyrrolate for sialorrhea in Parkinson disease: a randomized, double-blind, crossover trial. Neurology 2010; 74: 12037.Google Scholar
Lloret, SP, Nano, G, Carrosella, A, Gamzu, E, Merello, M. A double-blind, placebo-controlled, randomized, crossover pilot study of the safety and efficacy of multiple doses of intra-oral tropicamide films for the short-term relief of sialorrhea symptoms in Parkinson's disease patients. J Neurol Sci 2011; 310: 24850.Google Scholar
Bennett, N, O'Leary, M, Patel, AS, et al. Can higher doses of oxybutynin improve efficacy in neurogenic bladder? J Urol 2004; 171: 74951.Google Scholar
Corbin, KB. Trihexyphenidyl; evaluation of the new agent in the treatment of Parkinsonism. J Am Med Assoc 1949; 141: 37782.Google Scholar
Brumlik, J, Canter, G, Delatorre, R, et al. A critical analysis of the effects of trihexyphenidyl (artane) on the components of the Parkinsonian syndrome. J Nerv Ment Dis 1964; 138: 42431.Google ScholarPubMed
Iivanainen, M. KR 339 in the treatment of Parkinsonian tremor. Acta Neurol Scand 1974; 50: 46977.Google Scholar
Poewe, WH, Lees, AJ, Stern, GM. Dystonia in Parkinson's disease: clinical and pharmacological features. Ann Neurol 1988; 23: 738.Google Scholar
Jankovic, J. Treatment of dystonia. Lancet Neurol 2006; 5: 86472.Google Scholar
Machado, D, Jabbari, B. Treatment of Parkinson's disease with anticholinergic medication. In: Pfeiffer, RF., Wszolek, ZK, Ebadi, M, eds. Parkinson's Disease. Boca Raton: CRC Press, 2013; 81118.Google Scholar
Cantello, R, Riccio, A, Gilli, M, et al. Bornaprine vs placebo in Parkinson disease: double-blind controlled cross-over trial in 30 patients. Ital J Neurol Sci 1986; 7: 13943.Google Scholar
Tourtellotte, WW, Potvin, AR, Syndulko, K, et al. Parkinson's disease: Cogentin with Sinemet, a better response. Prog Neuropsychopharmacol Biol Psychiatry 1982; 6: 515.Google Scholar
Baba, Y, Higuchi, MA, Abe, H, et al. Anti-cholinergics for axial symptoms in Parkinson's disease after subthalamic stimulation. Clin Neurol Neurosurg 2012; 114: 130811.Google Scholar
Koller, WC. Pharmacologic treatment of parkinsonian tremor. Arch Neurol 1986; 43: 1267.Google Scholar
Parkes, JD, Baxter, RC, Marsden, CD, Rees, JE. Comparative trial of benzhexol, amantadine, and levodopa in the treatment of Parkinson's disease. J Neurol Neurosurg Psychiatry 1974; 37: 4226.Google Scholar
Schrag, A, Schelosky, L, Scholz, U, Poewe, W. Reduction of Parkinsonian signs in patients with Parkinson's disease by dopaminergic versus anticholinergic single-dose challenges. Mov Disord 1999; 14: 2525.Google Scholar
Roberts, J, Waller, DG, von Renwick, AG, et al. The effects of co-administration of benzhexol on the peripheral pharmacokinetics of oral levodopa in young volunteers. Br J Clin Pharmacol 1996; 41: 3317.CrossRefGoogle ScholarPubMed
Contin, M, Riva, R, Martinelli, P, et al. Combined levodopa-anticholinergic therapy in the treatment of Parkinson's disease. Effect on levodopa bioavailability. Clin Neuropharmacol 1991; 14: 14855.Google Scholar
Horrocks, M, Vicary, DJ, Rees, JE, Parkes, JD, Marsden, CD. Anticholinergic withdrawal and benzhexol treatment in Parkinson's disease. J Neurol Neurosurg Psychiatry 1973; 36: 93641.Google Scholar
Tune, LE, Anticholinergic effects of medication in elderly patients. J Clin Psychiatry 2001; 62 (Suppl. 21): 114.Google Scholar
Buter, TC, van den Hout, A, Matthews, FE, et al. Dementia and survival in Parkinson disease: a 12-year population study. Neurology 2008; 70: 101722.Google Scholar
Bohnen, NI, Kaufer, DI, Hendrickson, R, et al. Cortical cholinergic denervation is associated with depressive symptoms in Parkinson's disease and parkinsonian dementia. J Neurol Neurosurg Psychiatry 2007; 78: 6413.Google Scholar
Ehrt, U, Broich, K, Larsen, JP, Ballard, C, Aarsland, D. Use of drugs with anticholinergic effect and impact on cognition in Parkinson's disease: a cohort study. J Neurol Neurosurg Psychiatry 2010; 81: 1605.Google Scholar
Bedard, MA, Pillon, B, Dubois, B, et al. Acute and long-term administration of anticholinergics in Parkinson's disease: specific effects on the subcortico-frontal syndrome. Brain Cogn 1999; 40: 289313.Google Scholar
Cooper, JA, Sagar, HJ, Doherty, SM, et al. Different effects of dopaminergic and anticholinergic therapies on cognitive and motor function in Parkinson's disease. A follow-up study of untreated patients. Brain 1992; 115: 170125.Google Scholar
Quik, M, McIntosh, JM. Striatal α6* nicotinic acetylcholine receptors: potential targets for Parkinson's disease therapy. J Pharmacol Exp Ther 2006; 316: 4819.Google Scholar
Itti, E, Villafane, G, Malek, Z, et al. Dopamine transporter imaging under high-dose transdermal nicotine therapy in Parkinson's disease: an observational study. Nucl Med Commun 2009; 30: 51318.Google Scholar

References

Joel, D. Open interconnected model of basal ganglia-thalamocortical circuitry and its relevance to the clinical syndrome of Huntington's disease. Mov Disord 2001; 16: 40723.Google Scholar
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: 274754.Google Scholar
Lee, JY, Lee, EK, Park, SS, et al. Association of DRD3 and GRIN2B with impulse control and related behaviors in disturbances in Parkinson disease. Parkinsonism Relat Disord 2009; 15: 180310.Google Scholar
Calabresi, P, Di Filippo, M, Ghiglieri, V, Tambasco, N, Picconi, B. Levodopa-induced dyskinesias in patients with Parkinson's disease: filling the bench-to-bedside gap. Lancet Neurol 2010; 9: 110617.Google Scholar
Voon, V, Fernagut, PO, Wickens, J, et al. Chronic dopaminergic stimulation in Parkinson's disease: from dyskinesias to impulse control disorders. Lancet Neurol 2009; 8: 11409.Google Scholar
Linazasoro, G. Dopamine dysregulation syndrome and levodopa-induced dyskinesias in Parkinson disease: common consequences of anomalous forms of neural plasticity. Clin Neuropharmacol 2009; 32: 227.Google Scholar
Kalivas, PW. The glutamate homeostasis hypothesis of addiction. Nat Rev Neurosci 2009; 10: 56172.Google Scholar
Cenci, MA, Lundblad, M. Post- versus presynaptic plasticity in l-DOPA-induced dyskinesia. J Neurochem 2006; 99: 38192.Google Scholar
van Eimeren, T, Ballanger, B, Pellecchia, G, et al. Dopamine agonists diminish value sensitivity of the orbitofrontal cortex: a trigger for pathological gambling in Parkinson's disease? Neuropsychopharmacology 2009; 34: 275866.Google Scholar
Allers, KA, Bergstrom, DA, Ghazi, LJ, Kreiss, DS, Walters, JR. MK801 and amantadine exert different effects on subthalamic neuronal activity in a rodent model of Parkinson's disease. Exp Neurol 2005; 191: 10418.Google Scholar
Verhagen-Metman, L, Blanchet, PJ, van den Munckhof, P, et al. A trial of dextromethorphan in parkinsonian patients with motor response complications. Mov Disord 1998; 13: 41417.Google Scholar
Spieker, S, Loschmann, PA, Klockgether, T. The NMDA antagonist budipine can alleviate levodopa-induced motor fluctuations. Mov Disord 1999; 14: 51719.Google Scholar
Rabey, JM, Nissipeanu, P, Korczyn, AD. Efficacy of memantine, an NMDA receptor antagonist, in the treatment of Parkinson's disease. Neural Transm Park Dis Dement Sect 1992; 4: 27782.CrossRefGoogle ScholarPubMed
Varanese, S, Howard, J, Di Rocco, A. NMDA antagonist memantine improves levodopa-induced dyskinesias and “on-off” phenomena in Parkinson's disease. Mov Disord 2010; 25: 50810.Google Scholar
Iderberg, H, Rylander, D, Bimpisidis, Z, Cenci, MA. Modulating mGluR5 and 5-HT1A/1B receptors to treat l-DOPA-induced dyskinesia: effects of combined treatment and possible mechanisms of action. Exp Neurol 2013; 250: 11624.Google Scholar
Vidal, EI, Fukushima, FB, Valle, AP, Villas Boas, PJ. Unexpected improvement in levodopa-induced dyskinesia and on-off phenomena after introduction of memantine for treatment of Parkinson's disease dementia. J Am Geriatr Soc 2013; 61: 1702.Google Scholar
Hanagasi, HA, Kaptanoglu, G, Sahin, HA, Emre, M. The use of NMDA antagonist memantine in drug-resistant dyskinesias resulting from l-dopa. Mov Disord 2000; 15: 101617.Google Scholar
Rajput, AH, Rajput, A, Lang, AE, et al. New use for an old drug: amantadine benefits levodopa-induced dyskinesia. Mov Disord 1998; 13: 851.Google Scholar
Movement Disorders Society. Amantadine and other antiglutamate agents: management of Parkinson's disease. Mov Disord 2002; 17 (Suppl. 4): S1322.Google Scholar
Crosby, NJ, Deane, KH, Clarke, CE. Amantadine for dyskinesia in Parkinson's disease. Cochrane Database Syst Rev 2003: CD003467.Google Scholar
Higgins, JPT, Green, S (eds). Cochrane Handbook for Systematic Reviews of Interventions. Chichester: Wiley-Blackwell, 2008.Google Scholar
Elahi, B, Phielipp, N, Chen, R. N-Methyl-d-Aspartate antagonists in levodopa induced dyskinesia: a meta-analysis. Can J Neurol Sci 2012; 39: 46572.Google Scholar
Riederer, P, Kornhuber, J, Gerlach, M, et al. Glutamatergic-dopaminergic imbalance in Parkinson's disease and paranoid hallucinatory psychosis. In: Rinne, UK, Nagatsu, T, Horowski, R (eds). International Workshop Berlin: Parkinson's Disease. The Netherlands: Medicom, , 1991; 1023.Google Scholar
Thomas, A, Iacono, D, Luciano, AL, et al. Duration of amantadine benefit on dyskinesia of severe Parkinson's disease. J Neurol Neurosurg Psychiatry 2004; 75: 1413.Google Scholar
Metman, LV, Del Dotto, P, LePoole, K, et al. Amantadine for levodopa-induced dyskinesias: a 1-year follow-up study. Arch Neurol 1999; 56: 13836.Google Scholar
Wolf, E, Seppi, K, Katzenschlager, R, et al. Long-term antidyskinetic efficacy of amantadine in Parkinson's disease. Mov Disord 2010; 25: 135763.CrossRefGoogle ScholarPubMed
Luginger, E, Wenning, GK, Bosch, S, Poewe, W. Beneficial effects of amantadine on l-dopa-induced dyskinesias in Parkinson's disease. Mov Disord 2000; 15: 8738.Google Scholar
Silva-Junior, FP, Braga-Neto, P, Sueli, MF, de Bruin, VM. Amantadine reduces the duration of levodopa-induced dyskinesia: a randomized, double-blind, placebo-controlled study. Parkinsonism Relat Disord 2005; 11: 44952.Google Scholar
Snow, BJ, Macdonald, L, McAuley, D, Wallis, W. The effect of amantadine on levodopa-induced dyskinesias in Parkinson's disease: a double-blind, placebo-controlled study. ClinNeuropharmacol 2000; 23: 825.Google Scholar
Verhagen, ML, Del Dotto, P, van den, Munckhof P, et al. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson's disease. Neurology 1998; 50: 13236.Google Scholar
Rajput, AH, Uitti, RJ, Lang, AE, et al. Amantidine ameliorates levodopa-induced dyskinesia. Neurology 1997; 48 (Suppl. 3): A328.Google Scholar
Sawada, H, Oeda, T, Kuno, S, et al. Amantadine for dyskinesias in Parkinson's disease: a randomized controlled trial. PLoS One 2010; 5: e15298.Google Scholar
Parkinson Study Group. Evaluation of dyskinesias in a pilot, randomized, placebo-controlled trial of remacemide in advanced Parkinson disease. Arch Neurol 2001; 58: 16608.Google Scholar
Merello, M, Nouzeilles, MI, Cammarota, A, Leiguarda, R. Effect of memantine (NMDA antagonist) on Parkinson's disease: a double-blind crossover randomized study. Clin Neuropharmacol 1999; 22: 2736.Google Scholar
Mukherjee, D, Patil, CG, Palestrant, D. Amantadine for severe traumatic brain injury. N Engl J Med 2012; 366: 2427.Google Scholar
Zafonte, RD, Watanabe, T, Mann, NR. Amantadine: a potential treatment for the minimally conscious state. Brain Inj 1998; 12: 61721.Google Scholar
Horiguchi, J, Inami, Y, Shoda, T. Effects of long-term amantadine treatment on clinical symptoms and EEG of a patient in a vegetative state. Clin Neuropharmacol 1990; 13: 848.Google Scholar
Inzelberg, R, Bonuccelli, U, Schechtman, E, et al. Association between amantadine and the onset of dementia in Parkinson's disease. Mov Disord 2006; 21: 13759.Google Scholar
Aarsland, D, Ballard, C, Walker, Z, et al. Memantine in patients with Parkinson's disease dementia or dementia with Lewy bodies: a double-blind, placebo-controlled, multicentre trial. Lancet Neurol 2009; 8: 61318.Google Scholar
Ballard, C, Kahn, Z, Corbett, A. Treatment of dementia with Lewy bodies and Parkinson's disease dementia. Drugs Aging 2011; 28: 76977.Google Scholar
Emre, M, Tsolaki, M, Bonuccelli, U, et al. Memantine for patients with Parkinson's disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol 2010; 9: 96977.Google Scholar
Onofrj, M, Thomas, A. Acute akinesia in Parkinson disease. Neurology 2005; 64: 11629.Google Scholar
Thomas, A, Onofrj, M. Akinetic crisis, acute akinesia, neuroleptic malignant-like syndrome, Parkinsonism-hyperpyrexia syndrome, and malignant syndrome are the same entity and are often independent of treatment withdrawal. Mov Disord 2005; 20: 1671; author reply 1671–2.Google Scholar
Margetić, B, Aukst-Margetić, B. Neuroleptic malignant syndrome and its controversies. Pharmacoepidemiol Drug Saf 2010; 19: 42935.Google Scholar
Eggert, KM, Oertel, WH, Reichmann, H (eds). Parkinson Syndrome – Diagnostik und Therapie. Leitlinien fuer Diagnostik und Therapie in der Neurologie. Berlin: Deutsche Gesellschaft fuer Neurologie, 2012.Google Scholar
Smith, EJ. Amantadine-induced psychosis in a young healthy patient. Am J Psychiatry 2008; 165: 1613.Google Scholar
Harper, RW, Knothe, UC. Colored Lilliputian hallucinations with amantadine. Med J Aust 1973; 1: 4445.Google Scholar
Snoey, ER, Bessen, HA. Acute psychosis after amantadine overdose. Ann Emerg Med 1990; 19: 66870.Google Scholar
Stroe, AE, Hall, J, Amin, F. Psychotic episode related to phenylpropanolamine and amantadine in a healthy female. Gen Hosp Psychiatry 1995; 17: 4578.Google Scholar
Flaherty, JA, Bellur, SN. Mental side effects of amantadine therapy: its spectrum and characteristics in a normal population. J Clin Psychiatry 1981; 42: 3445.Google Scholar
Walsh, RA, Lang, AE. Multiple impulse control disorders developing in Parkinson's disease after initiation of amantadine. Mov Disord 2012; 27: 326.Google Scholar
Riederer, P, Lange, KW, Kornhuber, J, Danielczyk, W. Pharmacotoxic psychosis after memantine in Parkinson's disease. Lancet 1991; 338: 10223.Google Scholar
Robles Bayón, A, Gude Sampedro, F. Inappropriate treatments for patients with cognitive decline. Neurologia 2012; 29: 52332.Google Scholar
Menendez-Gonzalez, M, Calatayud, MT, Blazquez-Menes, B. Exacerbation of Lewy bodies dementia due to memantine. J Alzheimers Dis 2005; 8: 28991.Google Scholar
Grant, JE, Chamberlain, SR, Odlaug, BL, Potenza, MN, Kim, SW. Memantine shows promise in reducing gambling severity and cognitive inflexibility in pathological gambling: a pilot study. Psychopharmacology (Berl) 2010; 212: 60312.Google Scholar
Gallini, A, Sommet, A, Salandini, AM, et al. Weight-loss associated with anti-dementia drugs in a patient with Parkinson's disease. Mov Disord 2007; 22: 19801.Google Scholar
Sani, G, Serra, G, Kotzalidis, GD, et al. The role of memantine in the treatment of psychiatric disorders other than the dementias: a review of current preclinical and clinical evidence. CNS Drugs 2012; 26: 66390.Google Scholar
Stewart, SE, Jenike, EA, Hezel, DM, et al. 2010; A single blinded case–control study of memantine insevere obsessive-compulsive disorder. J Clin Psychopharmacol 30: 349.Google Scholar
Pelc, I, Verbanck, P, Le Bon, O, et al. Efficacy and safety of acamprosate in the treatment of detoxified alcohol-dependent patients. A 90-day placebo-controlled dose-finding study. Br J Psychiatry 1997; 171: 737.Google Scholar
Thomas, A, Bonanni, L, Gambi, F, Di Iorio, A, Onofrj, M. Pathological gambling in Parkinson disease is reduced by amantadine. Ann Neurol 2010; 68: 4004.Google Scholar
Cera, N, Bifolchetti, S, Martinotti, G, et al. Amantadine and cognitive flexibility: decision making in Parkinson patients with severe pathological gambling and other impulse control disorders. Neuropsychiatr Dis Treat 2014; 10: 1093101.Google Scholar
Kashihara, K, Imamura, T. Amantadine may reverse punding in Parkinson's disease – observation in a patient. Mov Disord 2008; 23: 12930.Google Scholar
Pettorruso, M, Martinotti, G, Di Nicola, M, et al. Amantadine in the treatment of pathological gambling: a case report. Front Psychiatry 2012; 3: 102.Google Scholar
Fasano, A, Ricciardi, L, Pettorruso, M, Bentivoglio, AR. Management of punding in Parkinson's disease: an open-label prospective study. J Neurol 2011; 258: 65660.Google Scholar
Weintraub, D, Sohr, M, Siderowf, A, et al. Amantadine use associated with impulse control disorders in Parkinson disease. Ann Neurol 2010; 68: 9638.Google Scholar
Lee, JY, Kim, HJ, Jeon, BS. Is pathological gambling in Parkinson's disease reduced by amantadine? Ann Neurol 2011; 69: 21314.Google Scholar
Onofrj, M, Bonanni, L, Di Iorio, A, Thomas, A. Is pathological gambling in Parkinson's disease reduced by amantadine? Reply Ann Neurol 2011; 69: 21415.Google Scholar
Wang, O, Kilpatrick, RD, Critchlow, CW, et al. Relationship between epoetin alfa dose and mortality: findings from a marginal structural model. Clin J Am Soc Nephrol 2010; 5: 1828.Google Scholar
Fahn, S. The history of dopamine and levodopa in the treatment of Parkinson's disease. Mov Disord 2008; 23: S497508.Google Scholar
Bastiaens, J, Dorfman, BJ, Christos, PJ, Nirenberg, MJ. Prospective cohort study of impulse control disorders in Parkinson's disease. Mov Disord 2013; 28: 32733.Google Scholar
Yerkes, RM, Dodson, JD. The relation of strength of stimulus to rapidity of habit-formation. J Comp Neurol Psychol 1908; 18: 45982.Google Scholar
Cools, R. Dopaminergic modulation of cognitive function-implications for l-DOPA treatment in Parkinson's disease. Neurosci Biobehav Rev 2006; 30: 123.Google Scholar
Voon, V, Gao, J, Brezing, C, et al. Dopamine agonists and risk: impulse control disorders in Parkinson's disease. Brain 2011; 134: 143846.Google Scholar
Jenner, P. Molecular mechanisms of l-DOPA-induced dyskinesia. Nat Rev Neurosci 2008; 9: 66577.Google Scholar
Stocchi, F, Vacca, L, Ruggieri, S, Olanow, CW. Intermittent vs continuous levodopa administration in patients with advanced Parkinson disease: a clinical and pharmacokinetic study. Arch Neurol 2005; 62: 90510.Google Scholar
Chaudhuri, KR, Schapira, AH. Non-motor symptoms of Parkinson's disease: dopaminergic pathophysiology and treatment. Lancet Neurol 2009; 8: 46474.CrossRefGoogle ScholarPubMed
Agid, Y, Arnulf, I, Bejjani, P, et al. Parkinson's disease is a neuropsychiatric disorder. Adv Neurol 2003; 91: 36570.Google Scholar
Onofrj, M, Bonanni, L, Manzoli, L, Thomas, A. Cohort study on somatoform disorders in Parkinson disease and dementia with Lewy bodies. Neurology 2010; 74: 1598606.Google Scholar
Albin, RL, Dauer, WT. Parkinson syndrome. Heterogeneity of etiology; heterogeneity of pathogenesis? Neurology 2012; 79: 2023.Google Scholar
Rabinak, CA, Nirenberg, MJ. Dopamine agonist withdrawal syndrome in Parkinson disease. Arch Neurol 2010; 67: 5863.Google Scholar

References

Löhle, M, Reichmann, H. Clinical neuroprotection in Parkinson's disease – still waiting for the breakthrough. J Neurol Sci 2010; 289: 10414.Google Scholar
Löhle, M, Reichmann, H. Controversies in neurology: why monoamine oxidase B inhibitors could be a good choice for the initial treatment of Parkinson's disease. BMC Neurol 2011; 11: 112.Google Scholar
Fabbrini, G, Abbruzzese, G, Marconi, S, Zappia, M. Selegiline: a reappraisal of its role in Parkinson disease. Clinical Neuropharmacol 2012; 35: 13440.CrossRefGoogle ScholarPubMed
Goetz, CG, Koller, WC, Poewe, W. Management of Parkinson's disease: an evidence-based review. Mov Disord 2002; 17 (Suppl. 4): S1166.Google Scholar
Goetz, CG, Poewe, W, Rascol, O, Sampaio, C. Evidence-based medical review update: pharmacological and surgical treatments of Parkinson's disease: 2001 to 2004. Mov Disord 2005; 20: 52339.Google Scholar
Macleod, A, Counsell, C, Ives, N, Stowe, R. Monoamine oxidase B inhibitors for early Parkinson's disease. Cochrane Database Syst Rev 2005; 20: CD004898.Google Scholar
National Collaborating Centre for Chronic Conditions. Parkinson’s Disease. National Clinical Guideline for Diagnosis and Management in Primary and Secondary Care. Guidelines for the NHS by NICE. London: Royal College of Physicians, 2006.Google Scholar
Pahwa, R, Factor, SA, Lyons, KE, et al. Practice parameter: treatment of Parkinson disease with motor fluctuations and dyskinesia (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2006; 66: 98395.Google Scholar
Ryton, BA, Liddle, BJ. Implementing NICE clinical guidelines on Parkinson's disease Clin Med 2009; 9: 43640.Google Scholar
Caslake, R, Macleod, A, Ives, N, Stowe, R, Counsell, C. Monoamine oxidase B inhibitors versus other dopaminergic agents in early Parkinson's disease. Cochrane Database Syst Rev 2009; (4): CD006661.Google Scholar
Brainin, M, Barnes, M, Baron, JC, et al. Guidance for the preparation of neurological management guidelines by EFNS scientific task forces – revised recommendations 2004. Eur J Neurol 2004, 11: 57781.Google Scholar
Horstinka, ME, 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. Part I: early (uncomplicated) Parkinson's disease. Eur J Neurol 2006; 13: 117085.Google Scholar
Horstinka, ME, 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. Part II: late(complicated) Parkinson's disease. Eur J Neurol 2006; 13: 1186202.Google Scholar
Leone, MA, Brainin, M, Boon, P, et al. Guidance for the preparation of neurological management guidelines by EFNS scientific task forces – revised recommendations 2012. Eur J Neurol 2013; 20: 41019.Google Scholar
Fox, S, Katzenschlager, R, Lim, SY, et al. The Movement Disorder Society Evidence-Based Medicine Review Update: treatments for the motor symptoms of Parkinson's disease. Mov Dis 2011; 26 (Suppl. 3): S241.Google Scholar
Ferreira, JJ, Katzenschlager, R, Bloem, BR, et al. Summary of the recommendations of the EFNS/MDS-ES review on therapeutic management of Parkinson's disease. Eur J Neurol 2013; 20: 515.Google Scholar
Knoll, J, Vizi, ES, Somogyi, E. Phenylisopropyl-methylpropinylamine (E-250), a monoamine oxidase inhibitor antagonizing the effect of tyramine. Drug Res 1968; 18: 10912.Google Scholar
Szende, B, Bokonyi, G, Bocsi, J, et al. Anti-apoptotic and apoptotic action of (–)-deprenyl and its metabolites. J Neural Transm 2001; 108: 2533.Google Scholar
Matsubara, K, Senda, T, Uezono, T, et al. l-Deprenyl prevents the cell hypoxia induced by dopaminergic neurotoxins, MPP(+) and β-carbolinium: a microdialysis study in rats. Neurosci Lett 2001; 302: 658.Google Scholar
Tatton, W, Chalmers-Redman, R, Tatton, N. Neuroprotection by deprenyl and other propargylamines: glyceraldehyde-3-phosphate dehydrogenase rather than monoamine oxidase B. J Neural Transm 2003; 110: 50915.Google Scholar
Magyar, K, Szende, B. (–)-Deprenyl, a selective MAO-B inhibitor, with apoptotic and anti-apoptotic properties. Neurotoxicology 2004; 25: 23342.Google Scholar
Saravanan, KS, Sindhu, KM, Senthilkumar, KS, Mohanakumar, KP. l-Deprenyl protects against rotenone-induced, oxidative stress-mediated dopaminergic neurodegeneration in rats. Neurochem Int 2006; 49: 2840.Google Scholar
Birkmayer, W, Knoll, J, Riederer, P, et al. Increased life expectancy resulting from addition of L-deprenyl to Madopar treatment in Parkinson's disease: a longterm study. J Neural Transm 1985; 64: 11327.Google Scholar
Blume, E. Street drugs yield primate Parkinson's model. JAMA 1983; 250: 1314.Google Scholar
Langston, JW, Ballard, PA Jr. Parkinson's disease in a chemist working with 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. N Engl J Med 1983; 309: 310.Google Scholar
Tetrud, JW, Langston, JW. The effect of deprenyl (selegiline) on the natural history of Parkinson's disease. Science 1989; 245: 51922.Google Scholar
Myllylä, VV, Sotaniemi, KA, Aasly, J, et al. An open multicenter study of the efficacy of MDL 72,974A, a monoamine oxidase type B (MAO-B) inhibitor, in Parkinson's disease. Adv Neurol 1993; 60: 67680.Google Scholar
Parkinson Study Group. Effects of tocopherol and deprenyl on the progression and disability in early Parkinson's disease. N Engl J Med 1993; 328: 17683.Google Scholar
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: 60412.Google Scholar
Olanow, CW, Hauser, RA, Gauger, L, et al. The effect of deprenyl and levodopa on the progression of Parkinson's disease Ann Neurol 1995; 38: 77177.Google Scholar
Pålhagen, S, Heinonen, EH, Hägglund, J, et al. Selegiline delays the onset of disability in de novo parkinsonian patients. Swedish Parkinson Study Group. Neurology 1998; 51: 5205.Google Scholar
Pålhagen, S, Heinonen, EH, Hägglund, J, et al. Selegiline delays the onset of disability in de novo Parkinsonian patients. Swedish Parkinson Study Group. Neurology 1998; 51: 5205.Google Scholar
Pålhagen, SE, Heinonen, E. Use of selegiline as monotherapy and in combination with levodopa in the management of Parkinson's disease: perspectives from the MONOCOMB Study. Prog Neurother Neuropsychopharmacol 2008; 3: 4971.Google Scholar
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; 329: 593.Google Scholar
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: 2418.Google Scholar
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 randomized, double-blind, parallel-group trial. Lancet 2005; 365: 94754.Google Scholar
Parkinson Study Group. A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch Neurol 2002; 59: 193743.Google Scholar
Parkinson Study Group. A controlled, randomized, delayed-start study of rasagiline in early Parkinson disease. Arch Neurol 2004; 61: 5616.Google Scholar
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; 361: 126878.Google Scholar
Schapira, A, Monoamine oxidase B inhibitors for the treatment of Parkinson's disease: a review of symptomatic and potential disease-modifying effects. CNS Drugs 2011; 25: 106171.Google Scholar
Ondo, WG, Sethi, KD, Kricorian, G. Selegilime orally disintegrating tablets in patient with Parkinson's disease and “wearing off” symptoms. Clin Neuropharmacol 2007; 30: 295300.Google Scholar
Montastruc, JL, Chaumerliac, C, Desboeuf, K, et al. Adverse drug reactions to selegiline: a review of the French pharmaco vigilance database. Clin Neuropharmacol 2000; 23: 2715.Google Scholar
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 UK. BMJ 1995; 311: 16027.Google Scholar
Thorogood, M, Armstrong, B, Nichols, T, Hollowell, J. Mortality in people taking selegiline; observational study. BMJ 1998; 327: 2524.Google Scholar
Reichmann, H, Jost, WH. Efficacy and tolerability of rasagiline in daily clinical use – a post-marketing observational study in patients with Parkinson's disease. Eur J Neurol 2010; 17: 116471.Google Scholar
Jost, W, Friede, M, Schnitker, J. Indirect meta-analysis of randomised placebo-controlled clinical trials on rasagiline and selegiline in the symptomatic treatment of Parkinson's disease. Basal Ganglia 2012; 2: S1726.Google Scholar
Richard, IH, Kurland, R, Tanner, C, et al. Serotonin syndrome and the combined use of deprenyl and an antidepressant in Parkinson's disease. Parkinson Study Group. Neurology 1997; 48: 10707.Google Scholar
Connolly, BS, Fox, SH. Drug treatments for the neuropsychiatric complications of Parkinson's disease. Expert Rev Neurother 2012; 12: 143949.Google Scholar
Connolly, BS, Lang, AE. Pharmacological treatment of Parkinson disease. A review. JAMA 2014; 311: 167083.Google Scholar
Naoi, M, Maruyama, W, Inaba-Hasegawa, K. Revelation in the neuroprotective functions of rasagiline and selegiline: the induction of distinct genes by different mechanisms. Expert Rev Neurother 2013; 13: 67184.Google Scholar
Youdim, MB. Multi target neuroprotective and neurorestorative anti-Parkinson and anti-Alzheimer drugs ladostigil and m30 derived from rasagiline. Exp Neurobiol 2013; 22: 110.Google Scholar
Parkinson Study Group. Effect of deprenyl on the progression of disability in early Parkinson's disease. N Engl J Med 1989; 321: 136471.Google Scholar
Myllylä, VV, Sotaniemi, KA, Vuorinen, JA, Heinonen, EH. Selegiline as initial treatment in de novo parkinsonian patients. Neurology 1992; 42: 33943.Google Scholar
Przuntek, H, Kuhn, W. The effect of R-(–)-deprenyl in de novo Parkinson patients on combination therapy with levodopa and decarboxylase inhibitor. J Neural Transm Suppl 1987; 25: 97104.Google Scholar
Sivertsen, B, Dupont, E, Mikkelsen, B, et al. Selegiline and levodopa in early or moderately advanced Parkinson's disease: a double-blind controlled short- and long-term study. Acta Neurol Scand Suppl 1989; 126: 14752.Google Scholar
Nappi, G, Martignoni, E, Horowski, R, et al. Lisuride plus selegiline in the treatment of early Parkinson's disease. Acta Neurol Scand 1991; 83: 40710.Google Scholar
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; 311: 16027.Google Scholar
Larsen, JP, Boas, J. The effects of early selegiline therapy on long-term levodopa treatment and parkinsonian disability: an interim analysis of a Norwegian–Danish 5-year study. Norwegian–Danish Study Group. Mov Disord 1997; 12: 17582.Google Scholar
Larsen, JP, Boas, J, Erdal, JE. Does selegiline modify the progression of early Parkinson's disease? Results from a five-year study. The Norwegian–Danish Study Group. Eur J Neurol 1999; 6: 53947.Google Scholar
Parkinson's Disease Research Group in the United Kingdom. Comparisons of therapeutic effects of levodopa, levodopa and selegiline, and bromocriptine in patients with early, mild Parkinson's disease: three year interim report. BMJ 1993; 307: 46972.Google Scholar
Heinonen, EH, Myllylä, V. Safety of selegiline (deprenyl) in the treatment of Parkinson's disease. Drug Saf 1998; 19: 1122.Google Scholar
Lieberman, AN, Gopinathan, G, Neophytides, A, Foo, SH. Deprenyl versus placebo in Parkinson disease: a double-blind study. NY State J Med 1987; 87: 6469.Google Scholar
Golbe, LI, Lieberman, AN, Muenter, MD, et al. Deprenyl in the treatment of symptom fluctuations in advanced Parkinson's disease. Clin Neuropharmacol 1988; 11: 4555.Google Scholar

References

Hornykiewicz, OD. Physiologic, biochemical, and pathological backgrounds of levodopa and possibilities for the future. Neurology 1970; 20 (Suppl.): 15.Google Scholar
Cotzias, GC, Van Woert, MH, Schiffer, LM. Aromatic amino acids and modification of parkinsonism. N Engl J Med 1967; 276: 3749.Google Scholar
Schrag, A, Quinn, N. Dyskinesias and motor fluctuations in Parkinson's disease. A community-based study. Brain 2000; 123: 2297305.Google Scholar
Fabbrini, G, Brotchie, JM, Grandas, F, Nomoto, M, Goetz, CG. Levodopa-induced dyskinesias. Mov Disord 2007; 22: 137989; quiz 1523.Google Scholar
Linazasoro, G. Pathophysiology of motor complications in Parkinson disease: postsynaptic mechanisms are crucial. Arch Neurol 2007; 64: 13740.Google Scholar
Nandhagopal, R, Kuramoto, L, Schulzer, M, et al. Longitudinal evolution of compensatory changes in striatal dopamine processing in Parkinson's disease. Brain 2011; 134: 32908.Google Scholar
de la Fuente-fernández, R, Schulzer, M, Mak, E, Calne, DB, Stoessl, AJ. Presynaptic mechanisms of motor fluctuations in Parkinson's disease: a probabilistic model. Brain 2004; 127: 88899.Google Scholar
Hall, TR, Figueroa, HR, Yurgens, PB. Effects of inhibitors on monoamine oxidase activity in mouse brain. Pharmacol Res Commun 1982; 14: 44353.Google Scholar
Dolphin, AC, Jenner, P, Sawaya, MC, Marsden, CD, Testa, B. The effect of bromocriptine on locomotor activity and cerebral catecholamines in rodents. J Pharm Pharmacol 1977; 29: 72734.Google Scholar
Birkmayer, W. Long term treatment with l-deprenyl. J Neural Transm 1978; 43: 23944.Google Scholar
Lees, AJ, Stern, GM. Sustained bromocriptine therapy in previously untreated patients with Parkinson's disease. J Neurol Neurosurg Psychiatry 1981; 44: 10203.Google Scholar
Parkes, JD, Debono, AG, Marsden, CD. Bromocriptine in Parkinsonism: long-term treatment, dose response, and comparison with levodopa. J Neurol Neurosurg Psychiatry 1976; 39: 11018.Google Scholar
Montastruc, JL, Rascol, O, Senard, JM, Rascol, A. A randomised controlled study comparing bromocriptine to which levodopa was later added, with levodopa alone in previously untreated patients with Parkinson's disease: a five year follow up. J Neurol Neurosurg Psychiatry 1994; 57: 10348.Google Scholar
Rinne, UK. Early combination of bromocriptine and levodopa in the treatment of Parkinson's disease: a 5-year follow-up. Neurology 1987; 37: 8268.Google Scholar
Schachter, M, Sheehy, MP, Parkes, JD, Marsden, CD. Lisuride in the treatment of Parkinsonism. Acta Neurol Scand 1980; 62: 3825.Google Scholar
Massel, D, Suskin, N. Pergolide and cabergoline increased risk for valvular heart disease in Parkinson disease. ACP J Club 2007; 146: 756.Google Scholar
Van Camp, G, Flamez, A, Cosyns, B, et al. Treatment of Parkinson's disease with pergolide and relation to restrictive valvular heart disease. Lancet 2004; 363: 117983.Google Scholar
Zhou, CQ, Li, SS, Chen, ZM, Li, FQ, Lei, P, Peng, GG. Rotigotine transdermal patch in Parkinson's disease: a systematic review and meta-analysis. PLoS One. 2013; 8: e69738.Google Scholar
Voon, V, Sohr, M, Lang, AE, et al. Impulse control disorders in Parkinson disease: a multicenter case – control study. Ann Neurol 2011; 69: 98696.Google Scholar
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: 58995.Google Scholar
NICE. Parkinson's disease: Diagnosis and Management in Primary and Secondary Care. NICE guidelines [CG35]. National Institute for Health and Care Excellence, 2011. Available at: https://www.nice.org.uk/guidance/cg35.Google Scholar
Miyasaki, JM, Martin, W, Suchowersky, O, Weiner, WJ, Lang, AE. Practice parameter: initiation of treatment for Parkinson's disease: an evidence-based review: report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2002; 58: 117.Google Scholar
Pahwa, R, Factor, SA, Lyons, KE, et al. Practice Parameter: treatment of Parkinson disease with motor fluctuations and dyskinesia (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2006; 66: 98395.Google Scholar
Olanow, CW, Obeso, JA, Stocchi, F. Continuous dopamine-receptor treatment of Parkinson's disease: scientific rationale and clinical implications. Lancet Neurol 2006; 5: 67787.Google Scholar
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 Exp Ther 1995; 272: 8549.Google Scholar
Bibbiani, F, Costantini, LC, Patel, R, Chase, TN. Continuous dopaminergic stimulation reduces risk of motor complications in parkinsonian primates. Exp Neurol 2005; 192: 738.Google Scholar
Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: A randomized controlled trial. JAMA 2000; 284: 19318.Google Scholar
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: 148491.Google Scholar
Whone, AL, Watts, RL, Stoessl, AJ, et al. Slower progression of Parkinson's disease with ropinirole versus levodopa: the REAL-PET study. Ann Neurol 2003; 54: 93101.Google Scholar
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: 47480.Google Scholar
Tan, EK, Jankovic, J. Choosing dopamine agonists in Parkinson's disease. Clin Neuropharmacol 2001; 24: 24753.Google Scholar
Tonini, M, Cipollina, L, Poluzzi, E, et al. Review article: clinical implications of enteric and central D2 receptor blockade by antidopaminergic gastrointestinal prokinetics. Aliment Pharmacol Ther 2004; 19: 37990.Google Scholar
Fox, S. Domperidone as a treatment for dopamine agonist-induced peripheral edema in patients with Parkinson's disease. ClinicalTrials.gov 2012; NCT00305331.Google Scholar
Gizer, IR, Ficks, C, Waldman, ID. Candidate gene studies of ADHD: a meta-analytic review. Hum Genet 2009; 126: 5190.Google Scholar
Rashid, AJ, So, CH, Kong, MM, et al. D1-D2 dopamine receptor heterooligomers with unique pharmacology are coupled to rapid activation of Gq/11 in the striatum. Proc Natl Acad Sci U S A 2007; 104: 6549.Google Scholar
van Wieringen, JP, Booij, J, Shalgunov, V, Elsinga, P, Michel, MC. Agonist high- and low-affinity states of dopamine D2 receptors: methods of detection and clinical implications. Naunyn Schmiedebergs Arch Pharmacol 2013; 386: 13554.Google Scholar
Zhang, J, Xiong, B, Zhen, X, Zhang, A. Dopamine D1 receptor ligands: where are we now and where are we going. Med Res Rev 2009; 29: 27294.Google Scholar
Yun, JY, Kim, HJ, Lee, JY, et al. Comparison of once-daily versus twice-daily combination of ropinirole prolonged release in Parkinson's disease. BMC Neurol 2013; 13: 113.Google Scholar
Kvernmo, T, Härtter, S, Burger, E. A review of the receptor-binding and pharmacokinetic properties of dopamine agonists. Clin Ther 2006; 28: 106578.Google Scholar
Tippmann-Peikert, M, Park, JG, Boeve, BF, Shepard, JW, Silber, MH. Pathologic gambling in patients with restless legs syndrome treated with dopaminergic agonists. Neurology 2007; 68: 3013.Google Scholar
Thobois, S. Proposed dose equivalence for rapid switch between dopamine receptor agonists in Parkinson's disease: a review of the literature. Clin Ther 2006; 28: 112.Google Scholar
Tomlinson, CL, Stowe, R, Patel, S, et al. Systematic review of levodopa dose equivalency reporting in Parkinson's disease. Mov Disord 2010; 25: 264953.Google Scholar
Wright, CE, Sisson, TL, Ichhpurani, AK, Peters, GR. Steady-state pharmacokinetic properties of pramipexole in healthy volunteers. J Clin Pharmacol. 1997; 37: 5205.Google Scholar
Clarke, CE, Speller, JM, Clarke, JA. Pramipexole for levodopa-induced complications in Parkinson's disease. Cochrane Database Syst Rev 2000; 3: CD002261.Google Scholar
Moller, JC, Oertel, WH, Koster, J, Pezzoli, G, Provinciali, L. Long-term efficacy and safety of pramipexole in advanced Parkinson's disease: results from a European multicenter trial. Mov Disord 2005; 20: 60210.Google Scholar
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: 1628.CrossRefGoogle ScholarPubMed
Parkinson Study Group CALM Cohort Investigators. Long-term effect of initiating pramipexole vs levodopa in early Parkinson disease. Arch Neurol 2009; 66: 56370.Google Scholar
Inzelberg, R, Carasso, RL, Schechtman, E, Nisipeanu, P. A comparison of dopamine agonists and catechol-O-methyltransferase inhibitors in Parkinson's disease. Clin Neuropharmacol 2000; 23: 2626.Google Scholar
Herrero, MT, Pagonabarraga, J, Linazasoro, G. Neuroprotective role of dopamine agonists: evidence from animal models and clinical studies. Neurologist 2011; 17 (Suppl. 1): S5466.Google Scholar
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; 361: 126878.Google Scholar
Schapira, AH, McDermott, MP, Barone, P, et al. Pramipexole in patients with early Parkinson's disease (PROUD): a randomised delayed-start trial. Lancet Neurol 2013; 12: 74755.Google Scholar
Ostow, M. Pramipexole for depression. Am J Psychiatry 2002; 159: 3201.Google Scholar
Leentjens, AF, Koester, J, Fruh, B, et al. The effect of pramipexole on mood and motivational symptoms in Parkinson's disease: a meta-analysis of placebo-controlled studies. Clin Ther 2009; 31: 8998.Google Scholar
Rektorova, I, Rektor, I, Bares, M, et al. Pramipexole and pergolide in the treatment of depression in Parkinson's disease: a national multicentre prospective randomized study. Eur J Neurol 2003; 10: 399406.Google Scholar
Barone, P, Poewe, W, Albrecht, S, et al. Pramipexole for the treatment of depressive symptoms in patients with Parkinson's disease: a randomised, double-blind, placebo-controlled trial. Lancet Neurol 2010; 9: 57380.Google Scholar
Barone, P, Scarzella, L, Marconi, R, et al. Pramipexole versus sertraline in the treatment of depression in Parkinson's disease: a national multicenter parallel-group randomized study. J Neurol 2006; 253: 6017.Google Scholar
Lemke, MR, Brecht, HM, Koester, J, Kraus, PH, Reichmann, H. Anhedonia, depression, and motor functioning in Parkinson's disease during treatment with pramipexole. J Neuropsychiatry Clin Neurosci 2005; 17: 21420.Google Scholar
Kirsch-Darrow, L, Marsiske, M, Okun, MS, Bauer, R, Bowers, D. Apathy and depression: separate factors in Parkinson's disease. J Int Neuropsychol Soc 2011; 17: 105866.Google Scholar
Mayberg, HS. Limbic-cortical dysregulation: a proposed model of depression. J Neuropsychiatry Clin Neurosci 1997; 9: 47181.Google Scholar
Kaye, CM, Nicholls, B. Clinical pharmacokinetics of ropinirole. Clin Pharmacokinet 2000; 39: 24354.Google Scholar
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: 105762.Google Scholar
Rektorova, I, Balaz, M, Svatova, J, et al. Effects of ropinirole on nonmotor symptoms of Parkinson disease: a prospective multicenter study. Clin Neuropharmacol 2008; 31: 2616.Google Scholar
Zagmutt, FJ, Tarrants, ML. Indirect comparisons of adverse events and dropout rates in early Parkinson's disease trials of pramipexole, ropinirole, and rasagiline. Int J Neurosci 2012; 122: 34553.Google Scholar
Kulisevsky, J, Pagonabarraga, J. Tolerability and safety of ropinirole versus other dopamine agonists and levodopa in the treatment of Parkinson's disease: meta-analysis of randomized controlled trials. Drug Saf 2010; 33: 14761.Google Scholar
Scheller, D, Ullmer, C, Berkels, R, Gwarek, M, Lubbert, H. The in vitro receptor profile of rotigotine: a new agent for the treatment of Parkinson's disease. Naunyn Schmiedebergs Arch Pharmacol 2009; 379: 7386.Google Scholar
Reynolds, NA, Wellington, K, Easthope, SE. Rotigotine: in Parkinson's disease. CNS Drugs 2005; 19: 97381.Google Scholar
Guldenpfennig, WM, Poole, KH, Sommerville, KW, Boroojerdi, B. Safety, tolerability, and efficacy of continuous transdermal dopaminergic stimulation with rotigotine patch in early-stage idiopathic Parkinson disease. Clin Neuropharmacol 2005; 28: 10610.Google Scholar
Giladi, N, Boroojerdi, B, Korczyn, AD, et al. Rotigotine transdermal patch in early Parkinson's disease: a randomized, double-blind, controlled study versus placebo and ropinirole. Mov Disord 2007; 22: 2398404.Google Scholar
LeWitt, PA, Lyons, KE, Pahwa, R. Advanced Parkinson disease treated with rotigotine transdermal system: PREFER Study. Neurology 2007; 68: 12627.Google Scholar
Poewe, WH, Rascol, O, Quinn, N, et al. Efficacy of pramipexole and transdermal rotigotine in advanced Parkinson's disease: a double-blind, double-dummy, randomised controlled trial. Lancet Neurol 2007; 6: 51320.Google Scholar
Trenkwalder, C, Kies, B, Rudzinska, M, et al. Rotigotine effects on early morning motor function and sleep in Parkinson's disease: a double-blind, randomized, placebo-controlled study (RECOVER). Mov Disord 2011; 26: 909.Google Scholar
Ray Chaudhuri, K, Martinez-Martin, P, Antonini, A, et al. Rotigotine and specific non-motor symptoms of Parkinson's disease: post hoc analysis of RECOVER. Parkinsonism Relat Disord 2013; 19: 6605.Google Scholar
Ghys, L, Surmann, E, Whitesides, J, Boroojerdi, B. Effect of rotigotine on sleep and quality of life in Parkinson's disease patients: post hoc analysis of RECOVER patients who were symptomatic at baseline. Expert Opin Pharmacother 2011; 12: 198598.Google Scholar
Sarati, S, Guiso, G, Garattini, S, Caccia, S. Kinetics of piribedil and effects on dopamine metabolism: hepatic biotransformation is not a determinant of its dopaminergic action in rats. Psychopharmacology (Berl) 1991; 105: 5415.Google Scholar
Emile, J, Chanelet, J, Truelle, JL, Bastard, J. Action of piribedil in Parkinson's disease: I.V. test and oral treatment. Adv Neurol 1975; 9: 40913.Google Scholar
Lebrun-Frenay, C, Borg, M. Choosing the right dopamine agonist for patients with Parkinson's disease. Curr Med Res Opin. 2002; 18: 20914.Google Scholar
Ziegler, M, Rondot, P. [Action of piribedil in Parkinson disease. Multicenter study]. Presse Med 1999; 28: 141418 (in French).Google Scholar
Post, RM, Gerner, RH, Carman, JS, et al. Effects of a dopamine agonist piribedil in depressed patients: relationship of pretreatment homovanillic acid to antidepressant response. Arch Gen Psychiatry 1978; 35: 60915.Google Scholar
Thobois, S, Lhommée, E, Klinger, H, et al. Parkinsonian apathy responds to dopaminergic stimulation of D2/D3 receptors with piribedil. Brain 2013; 136: 156877.Google Scholar
Nash, JR, Wilson, SJ, Potokar, JP, Nutt, DJ. Mirtazapine induces REM sleep behavior disorder (RBD) in parkinsonism. Neurology 2003; 61: 1161; author reply 1161.Google Scholar
Passouant, P, Besset, A, Billiard, M, Negre, C. [Effect of piribedil on nocturnal sleep (author's transl)]. Rev Electroencephalogr Neurophysiol Clin 1978; 8: 32634.Google Scholar
Peralta, C, Wolf, E, Alber, H, et al. Valvular heart disease in Parkinson's disease vs. controls: An echocardiographic study. Mov Disord 2006; 21: 110913.Google Scholar
Frucht, S, Rogers, JD, Greene, PE, Gordon, MF, Fahn, S. Falling asleep at the wheel: motor vehicle mishaps in persons taking pramipexole and ropinirole. Neurology 1999; 52: 190810.Google Scholar
Hauser, RA, Gauger, L, Anderson, WM, Zesiewicz, TA. Pramipexole-induced somnolence and episodes of daytime sleep. Mov Disord 2000; 15: 65863.Google Scholar
Etminan, M, Samii, A, Takkouche, B, Rochon, PA. Increased risk of somnolence with the new dopamine agonists in patients with Parkinson's disease: a meta-analysis of randomised controlled trials. Drug Saf 2001; 24: 8638.Google Scholar
Paus, S, Brecht, HM, Koster, J, et al. Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson's disease. Mov Disord 2003; 18: 65967.Google Scholar
Avorn, J, Schneeweiss, S, Sudarsky, LR, et al. Sudden uncontrollable somnolence and medication use in Parkinson disease. Arch Neurol 2005; 62: 12428.Google Scholar
Happe, S, Berger, K. The association of dopamine agonists with daytime sleepiness, sleep problems and quality of life in patients with Parkinson's disease – a prospective study. J Neurol 2001; 248: 10627.Google Scholar
Kleiner-Fisman, G, Fisman, DN. Risk factors for the development of pedal edema in patients using pramipexole. Arch Neurol 2007; 64: 8204.Google Scholar
Tan, EK. Peripheral edema and dopamine agonists in Parkinson disease. Arch Neurol 2007; 64: 15467; author reply 1547.Google Scholar
Senard, JM, Rai, S, Lapeyre-Mestre, M, et al. Prevalence of orthostatic hypotension in Parkinson's disease. J Neurol Neurosurg Psychiatry 1997; 63: 5849.Google Scholar
Perez-Lloret, S, Rey, MV, Fabre, N, et al. Factors related to orthostatic hypotension in Parkinson's disease. Parkinsonism Relat Disord 2012; 18: 5015.Google Scholar
Kujawa, K, Leurgans, S, Raman, R, Blasucci, L, Goetz, CG. Acute orthostatic hypotension when starting dopamine agonists in Parkinson's disease. Arch Neurol 2000; 57: 14613.Google Scholar
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: 43944.Google Scholar
Onofrj, M, Thomas, A, D'Andreamatteo, G, et al. Incidence of RBD and hallucination in patients affected by Parkinson's disease: 8-year follow-up. Neurol Sci. 2002; 23 (Suppl. 2): S914.Google Scholar
Kamakura, K, Mochizuki, H, Kaida, K, et al. Therapeutic factors causing hallucination in Parkinson's disease patients, especially those given selegiline. Parkinsonism Relat Disord 2004; 10: 23542.Google Scholar
Williams, DR, Lees, AJ. Visual hallucinations in the diagnosis of idiopathic Parkinson's disease: a retrospective autopsy study. Lancet Neurol 2005; 4: 60510.Google Scholar
Harding, AJ, Stimson, E, Henderson, JM, Halliday, GM. Clinical correlates of selective pathology in the amygdala of patients with Parkinson's disease. Brain 2002; 125: 243145.Google Scholar
Holman, AJ. Impulse control disorder behaviors associated with pramipexole used to treat fibromyalgia. J Gambl Stud 2009; 25: 42531.Google Scholar
Almanzar, S, Zapata-Vega, MI, Raya, JA. Dopamine agonist-induced impulse control disorders in a patient with prolactinoma. Psychosomatics 2013; 54: 38791.Google Scholar
Schreglmann, SR, Gantenbein, AR, Eisele, G, Baumann, CR. Transdermal rotigotine causes impulse control disorders in patients with restless legs syndrome. Parkinsonism Relat Disord 2012; 18: 2079.Google Scholar
Avila, A, Cardona, X, Martin-Baranera, M, Bello, J, Sastre, F. Impulsive and compulsive behaviors in Parkinson's disease: a one-year follow-up study. J Neurol Sci 2011; 310: 197201.Google Scholar
Mamikonyan, E, Siderowf, AD, Duda, JE, et al. Long-term follow-up of impulse control disorders in Parkinson's disease. Mov Disord 2008; 23: 7580.Google Scholar
Dang, D, Cunnington, D, Swieca, J. The emergence of devastating impulse control disorders during dopamine agonist therapy of the restless legs syndrome. Clin Neuropharmacol 2011; 34: 6670.Google Scholar
Ye, Z, Hammer, A, Camara, E, Munte, TF. Pramipexole modulates the neural network of reward anticipation. Hum Brain Mapp 2011; 32: 80011.Google Scholar
Pondal, M, Marras, C, Miyasaki, J, et al. Clinical features of dopamine agonist withdrawal syndrome in a movement disorders clinic. J Neurol Neurosurg Psychiatry 2013; 84: 1305.Google Scholar
Rabinak, CA, Nirenberg, MJ. Dopamine agonist withdrawal syndrome in Parkinson disease. Arch Neurol 2010; 67: 5863.Google Scholar
Nirenberg, MJ. Dopamine agonist withdrawal syndrome: implications for patient care. Drugs Aging 2013; 30: 58792.Google Scholar
Edwards, MJ. Dopamine agonist withdrawal syndrome (DAWS): perils of flicking the dopamine ‘switch’. J Neurol Neurosurg Psychiatry 2013; 84: 120.Google Scholar

References

Kvernmo, T, Härtter, S, Burger, E. A review of the receptor-binding and pharmacokinetic properties of dopamine agonists. Clin Ther 2006; 28: 106578.Google Scholar
Jenner, P. Preventing and controlling dyskinesia in Parkinson's disease – a view of current knowledge and future opportunities. Mov Disord 2008; 23 (Suppl. 3): S5858.Google Scholar
Chase, TN, Baronti, F, Fabbrini, G, et al. Rationale for continuous dopaminomimetic therapy of Parkinson's disease. Neurology 1989; 39 (Suppl. 2): 710.Google Scholar
Mouradian, MM, Heuser, IJ, Baronti, F, Chase, TN. Modification of central dopaminergic mechanisms by continuous levodopa therapy for advanced Parkinson's disease. Ann Neurol 1990; 27: 1823.Google Scholar
Brotchie, J, Jenner, P. New approaches to therapy. In: Brotchie, J, Bezard, E, Jenner, P, eds. Pathophysiology, Pharmacology, and Biochemistry of Dyskinesia. San Diego: Elsevier Academic Press, 2011; 12350.Google Scholar
Chaudhuri, KR, Schapira, AH. Non-motor symptoms of Parkinson's disease: dopaminergic pathophysiology and treatment. Lancet Neurol 2009; 8: 46474.Google Scholar
Storch, A, Schneider, CB, Wolz, M, et al. Nonmotor fluctuations in Parkinson disease: severity and correlation with motor complications. Neurology 2013; 80 : 8009.Google Scholar
Ray Chaudhuri, K, Rizos, A, Sethi, K. Motor and nonmotor complications in Parkinson's disease: an argument for continuous drug delivery? J Neural Transm 2013; 120: 130520.Google Scholar
Mouradian, MM. Scientific rationale for continuous dopaminergic stimulation in Parkinson's disease. Eur Neurol Rev 2006; 626.Google Scholar
Marinnan, S, Emmanuel, A, Burn, D. Delayed gastric emptying in Parkinson's disease. Mov Disord 2014; 29: 2332.Google Scholar
Naidu, Y, Ray Chaudhuri, K. Transdermal rotigotine: a new non-ergot dopamine agonist for the treatment of Parkinson's disease. Expert Opin Drug Deliv 2007; 4: 11118.Google Scholar
Jenner, P. A novel dopamine agonist for the transdermal treatment of Parkinson's disease. Neurology 2005; 65 (Suppl. 1): S35.Google Scholar
Paul, ML, Graybiel, AM, David, JC, Robertson, HA. D1-like and D2-like dopamine receptors synergistically activate rotation and c-fos expression in the dopamine-depleted striatum in a rat model of Parkinson's disease. J Neurosci 1992; 12: 372942.Google Scholar
Albanese, A, Jenner, P, Marsden, CD, Stephenson, JD. Bladder hyperreflexia induced in marmosets by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Neurosci Lett. 1988; 87: 4650.Google Scholar
Tomiyama, M, Kimura, T, Maeda, T, et al. A serotonin 5-HT1A receptor agonist prevents behavioral sensitization to L-DOPA in a rodent model of PD. Neurosci Res 2005; 52: 18594.Google Scholar
Srinivasan, J, Schmidt, WJ. Treatment with α2 – adrenoceptor antagonists, 2 methoxy idazaoxan, protects 6- hydroxydopamine induced Parkinsonian symptoms in rats: neurochemical and behavioral evidence. Behav Brain Res 2004; 154: 35363.Google Scholar
Cawello, W, Braun, M, Boekens, H. Absorption, disposition, metabolic fate, and elimination of the dopamine agonist rotigotine in man: administration by intravenous infusion or transdermal delivery drug metabolism and disposition. Drug Metab Dispos 2009; 37: 205560.Google Scholar
LeWitt, PA, Lyons, KE, Pahwa, R, SP 650 Study Group. Advanced Parkinson disease treated with rotigotine transdermal system: PREFER study. Neurology 2007; 68: 12627.Google Scholar
Poewe, WH, Rascol, O, Quinn, N, et al. Efficacy of pramipexole and transdermal rotigotine in advanced Parkinson's disease: a double-blind, double-dummy, randomised controlled trial. Lancet Neurol 2007; 6: 51320.Google Scholar
Trenkwalder, C, Kies, B, Rudzinska, M, et al. Rotigotine effects on early morning motor function and sleep in Parkinson's disease: a double-blind, randomized, placebo-controlled study (RECOVER). Mov Disord 2011; 26: 909.Google Scholar
Ray Chaudhuri, K, Martinez-Martin, P, Antonini, A, et al. Rotigotine and specific non-motor symptoms of Parkinson's disease: post hoc analysis of RECOVER. Parkinsonism Relat Disord 2013; 19: 6605.Google Scholar
Kassubek, J, Ray Chaudhuri, K, Zesiewicz, T, et al. Rotigotine transdermal system and evaluation of pain in patients with Parkinson's disease: a post hoc analysis of the RECOVER study. BMC Neurology 2014; 14: 42.Google Scholar
Trenkwalder, C, Kies, B, Dioszeghy, P, et al. Rotigotine transdermal system for the management of motor function and sleep disturbances in Parkinson's disease: results from a 1-year, open-label extension of the RECOVER study. Basal Ganglia 2012; 2: 7985.Google Scholar
Chen, JJ, Swope, DM, Dashtipour, K, Lyons, KE. Transdermal rotigotine: a clinically innovative dopamine-receptor agonist for the management of Parkinson's disease. Pharmacotherapy 2009; 29: 145267.Google Scholar
Metta, V, Muzerengi, S, Ray Chadhuri, K. Rotigotine: first dopamine agonist transdermal patch. Prescriber 2007; 18: 1930.Google Scholar
Weintraub, D, Nirenberg, MJ. Impulse control and related disorders in Parkinson's disease. Neurodegener Dis 2013; 11: 6371.Google Scholar
Rizos, A, Martinez-Martin, P, Martin, A, et al. European multicentre survey of tolerability rates and impulse control behaviour trends of prolonged release dopamine agonists in young and old PD. Mov Disord 2012; 27 (Suppl. 1): 161.Google Scholar
Priano, L, Albani, G, Brioschi, A, et al. Transdermal Apomorphine permeation from microemulsions: a new treatment in Parkinson's disease. Mov Disord 2004; 19: 93742.Google Scholar
Woitalla, D, Müller, T, Benz, S, Horowski, R, Przuntek, H. Transdermal lisuride delivery in the treatment of Parkinson's disease. J Neural Transm Suppl 2004; 68: 8995.Google Scholar
Dimitrova, T, Bara-Jiminez, W, Thomas, M, et al. Continuous dopaminergic stimulation with lisuride TTS (patch) in moderately advanced parkinsonian patients. Neurology 2006; 66 (Suppl. 2): A185.Google Scholar
Corrodi, H, Farnebo, LO, Fuxe, K, Hamberger, B, Ungerstedt, U. ET 495 (piribedil) and brain catecholamine mechanisms: evidence for stimulation of dopamine receptors. Eur J Pharmacol 1972; 20: 195204.Google Scholar
Montastruc, JL, Ziegler, M, Rascol, O, Malbezin, M. A randomized, double-blind study of a skin patch of a dopaminergic agonist, piribedil, in Parkinson's disease. Mov Disord 1999; 14: 33641.Google Scholar
Kapoor, R, Turjanski, N, Frankel, J, et al. Intranasal apomorphine: a new treatment in Parkinson's disease. J Neurol Neurosurg Psychiatry 1990; 53: 1015.Google Scholar
Kleedorfer, B, Turjanski, N, Ryan, R, et al. Intranasal apomorphine in Parkinson's disease. Neurology 1991; 41: 7612.Google Scholar
van Laar, T, Jansen, EN, Essink, AW, Neef, C. Intranasal apomorphine in parkinsonian on–off fluctuations. Arch Neurol 1992; 49: 4824.Google Scholar
Sam, E, Jeanjean, AP, Maloteaux, JM, Verbeke, N. Apomorphine pharmacokinetics in parkinsonism after intranasal and subcutaneous application. Eur J Drug Metab Pharmacokinet. 1995; 20: 2733.Google Scholar
Dewey, RB Jr, Maraganore, DM, Ahlskog, JE, Matsumoto, JY. Intranasal apomorphine rescue therapy for parkinsonian “off” periods. Clin Neuropharmacol 1996; 19: 193201.Google Scholar
Esteban Muñoz, J, Martí, MJ, Marín, C, Tolosa, E. Long-term treatment with intermitent intranasal or subcutaneous apormorphine in patients with levodopa-related motor fluctuations. Clin Neuropharmacol 1997; 20: 24552.Google Scholar
Dewey, RB Jr, Maraganore, DM, Ahlskog, JE, Matsumoto, JY. A double-blind, placebo-controlled study of intranasal apomorphine spray as a rescue agent for off-states in Parkinson's disease. Mov Disord 1998; 13: 7827.Google Scholar
Wickremaratchi, M, Hadjikoutis, S, Weiser, R, et al. Efficacy and tolerability of intranasal apomorphine powder (INAP) 5mg as rescue therapy in subjects with Parkinson's disease (PD) complicated by motor fluctuations. J Neurol Neurosurg Psychiatry 2003; 74: 144860.Google Scholar
Deleu, D, Hanssens, Y, Northway, MG. Subcutaneous apomorphine: an evidence-based review of its use in Parkinson's disease. Drugs Aging 2004; 21: 687709.Google Scholar
Schwab, RS, Amador, LV, Lettvin, JY. Apomorphine in Parkinson's disease. Trans Am Neurol Assoc 1951; 56: 2513.Google Scholar
Cotzias, GC, Papavasiliou, PS, Tolosa, ES, Mendez, JS, Bell-Midura, M. Treatment of Parkinson's disease with apomorphines. Possible role of growth hormone. The N Engl J Med 1976; 294: 56772.Google Scholar
Stibe, CM, Lees, AJ, Kempster, PA, Stern, GM. Subcutaneous apomorphine in parkinsonian on–off oscillations. Lancet 1988; 331: 4036.Google Scholar
Chaudhuri, KR, Critchley, P, Abbott, RJ, Pye, IF, Millac, PAH. Subcutaneous apomorphine for on–off oscillations in Parkinson's disease. Lancet 1988; 332: 1260.Google Scholar
LeWitt, PA. Subcutaneously administered apomorphine: pharmacokinetics and metabolism. Neurology 2004; 62 (Suppl. 4): S811.Google Scholar
Nicolle, E, Pollak, P, Serre-Debeauvais, F, et al. Pharmacokinetics of apomorphine in parkinsonian patients. Fundam Clin Pharmacol 1993; 7: 24552.Google Scholar
Manson, AJ, Hanagasi, H, Turner, K, et al. Intravenous apomorphine therapy in Parkinson's disease: clinical and pharmacokinetic observations. Brain 2001; 124: 33140.Google Scholar
Hofstee, DJ, Neef, C, van Laar, T, Jansen, EN. Pharmacokinetics of apomorphine in Parkinson's disease: plasma and cerebrospinal fluid levels in relation to motor responses. Clin Neuropharmacol 1994; 17: 4552.Google Scholar
Frankel, JP, Lees, AJ, Kempster, PA, Stern, GM. Subcutaneous apomorphine in the treatment of Parkinson's disease. J Neurol Neurosurg Psychiatry 1990; 53: 96101.Google Scholar
Pollak, P, Champay, AS, Gaio, JM, et al. [Subcutaneous administration of apomorphine in motor fluctuations in Parkinson's disease]. Rev Neurol (Paris) 1990; 146 : 11622 (in French).Google Scholar
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: 16570.Google Scholar
Stocchi, F, Bramante, L, Monge, A, et al. Apomorphine and lisuride infusion. A comparative long-term study. Adv Neurol 1993; 60: 6535.Google Scholar
Poewe, W, Kleedorfer, B, Wagner, M, Bosch, S, Schelosky, L. Continuous subcutaneous apomorphine infusions for fluctuating Parkinson's disease. Long-term follow-up in 18 patients. Adv Neurol 1993; 60: 6569.Google Scholar
Kreczy-Kleedorfer, B, Wagner, M, Bösch, S, Poewe, W. [Long-term results of continuous subcutaneous apomorphine pump therapy in patients with advanced Parkinson disease]. Nervenarzt 1993; 64: 2215 (in German).Google Scholar
Gancher, ST, Nutt, JG, Woodward, WR. Apomorphine infusional therapy in Parkinson's disease: clinical utility and lack of tolerance. Mov Disord 1995; 10: 3743.Google Scholar
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: 5736.Google Scholar
Pietz, K, Hagell, P, Odin, P. Subcutaneous apomorphine in late stage Parkinson's disease: a long-term follow-up. J Neurol Neurosurg Psychiatry 1998; 65: 70916.Google Scholar
Wenning, GK, Bösch, S, Luginger, E, Wagner, M, Poewe, W. Effects of long-term, continuous subcutaneous apomorphine infusions on motor complications in advanced Parkinson's disease. Adv Neurol 1999; 80: 5458.Google Scholar
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: 934.Google Scholar
Kanovsky, P, Kubova, 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: 18891.Google Scholar
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: 123541.Google Scholar
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: 1745.Google Scholar
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; 9: 2916.Google Scholar
Tyne, HL, Parsons, J, Sinnott, A, et al. A 10 year retrospective audit of long-term apomorphine use in Parkinson's disease. J Neurol 2004; 251: 13704.Google Scholar
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: 1517.Google Scholar
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: 4503.Google Scholar
Garcia 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: 11306.Google Scholar
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 Parkinson's Dis 2011; 1: 197203.Google Scholar
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: 57985.Google Scholar
Drapier, S, Gillioz, AS, Leray, E, et al. Apomorphine infusion in advanced Parkinson's patients with subthalamic stimulation contraindications. Parkinsonism Relat Disord 2012; 18: 404.Google Scholar
Alegret, M, Valldeoriola, F, Marti, M, et al. Comparative cognitive effects of bilateral subthalamic stimulation and subcutaneous continuous infusion of apomorphine in Parkinson's disease. Mov Disord 2004; 19: 14639.Google Scholar
Todorova, A Ray Chaudhuri, K. Subcutaneous apomorphine and non motor symptoms in Parkinson's disease. Parkinsonism Relat Disord 2013; 19: 10738.Google Scholar
Ray Chaudhuri, K, Abbott, RJ, Millac, PAH. Subcutaneous apomorphine for parkinsonian patients with psychiatric side effects on oral treatment. J Neurol Neurosurg Psychiatry 1991; 54: 3723.Google Scholar
Ellis, C, Lemmens, G, Parkes, JD, et al. Use of apomorphine in parkinsonian patients with neurospsychiatric complications of oral treatment. Parkinsonism Relat Disord 1997; 3: 1037.Google Scholar
van Laar, T, Postuma, AG, Drent, M. Continuous subcutaneous infusion of apomorphine can be used safely in patients with Parkinson's disease and pre-existing visual hallucinations. Parkinsonism Relat Disord 2010; 16: 712.Google Scholar
Issacson, S, Ray Chaudhuri, K. Morning akinesia and the potential role of gastroparesis – managing delayed onset of first daily dose of oral levodopa in patients with Parkinson's disease. European Neurological Review 2013; 8: 824.Google Scholar
Mathers, SE, Kempster, PA, Law, PJ, et al. Anal sphincter dysfunction in Parkinson's disease. Arch Neurol 1989; 46: 10614.Google Scholar
Edwards, LL, Quigley, EM, Harned, RK, Hofman, R, Pfeiffer, RF. Defecatory function n Parkinson's disease: response to apomorphine. Ann Neurol 1993; 33: 4903.Google Scholar
Tison, F, Wiart, L, Guatterie, M, et al. Effects of central dopaminergic stimulation by apomorphine on swallowing disorders in Parkinson's disease. Mov Disord 1996; 11: 72932.Google Scholar
Reuter, I, Ellis, CM, Ray Chaudhuri, K. Nocturnal subcutaneous apomorphine infusion in Parkinson's disease and restless legs syndrome. Acta Neurol Scand 1999; 100: 1637.Google Scholar
Tribl, GG, Sycha, T, Kotzailias, N, Zeitlhoger, J, Auff, E. Apomorphine in idiopathic restless legs syndrome: an exploratory study. J Neurol Neurosurg Psychiatry 2005; 76: 1815.Google Scholar
Magennis, B, Cashell, A, O'Brien, D, Lynch, T. An audit of apomorphine in the management of complex idiopathic Parkinson's disease in Ireland. Mov Disord 2012; 27 (Suppl. 1): S44.Google Scholar
Todorova, A, Martin, A, Okai, D, et al. Assessment of impulse control disorders in Parkinson's patients with infusion therapies: a single centre experience. Mov Disord 2013; 28 (Suppl. 1): S133.Google Scholar
Corsini, GU, Del Zompo, M, Gessa, GL, Mangoni, A. Therapeutic efficacy of apomorphine combined with an extracerebral inhibitor of dopamine receptors in Parkinson's disease. Lancet 1979; 313: 9546.Google Scholar
Stocchi, F, Ruggieri, S, Antonini, A, et al. Subcutaneous lisuride infusion in Parkinson's disease: clinical results using different modes of administration. J Neural Transm 1988; 27: 2733.Google Scholar
Krause, W, Nieuweboer, B, Ruggieri, S, Stocchi, F, Suchy, I. Pharmacokinetics of lisuride after subcutaneous infusion. J Neural Transm Suppl 1988; 27: 714.Google Scholar
Stocchi, F, Ruggieri, S, Vacca, L, Olanow, CW. Prospective randomized trial of lisuride infusion versus oral levodopa in patients with Parkinson's disease. Brain 2002; 125: 205866.Google Scholar
Heinz, A, Suchy, I, Klewin, I, et al. Long-term observation of chronic subcutaneous administration of lisuride in the treatment of motor fluctuations in Parkinson's disease. J Neural Transm Park Dis Dement Sect 1992; 4: 291301.Google Scholar
Obeso, JA, Luquin, MR, Vaamonde, J, Martînez Lage, JM. Subcutaneous administration of lisuride in the treatment of complex motor fluctuations in Parkinson's disease. J Neural Transm Suppl 1988; 27: 1725.Google Scholar
Critchley, PH, Grandas Perez, F, Quinn, NP, Parkes, JD, Marsden, CD. Continuous subcutaneous lisuride infusions in Parkinson's disease. J Neural Transm Suppl. 1988; 27: 5560.Google Scholar
Vaamonde, J, Luquin, MR, Obeso, JA. Subcutaneous lisuride infusion in Parkinson's disease. Response to chronic administration in 34 patients. Brain 1991; 114: 60117.Google Scholar
Hayashi, R, Tako, K, Makishita, H, Koyama, J, Yanagisawa, N. Efficacy of a low-dose subcutaneous lisuride infusion in Parkinson's disease. Intern Med 1998; 37: 4448.Google Scholar
Hofmann, C, Penner, U, Dorow, R, et al. Lisuride, a dopamine receptor agonist with 5-HT2B receptor antagonist properties: absence of cardiac valvulopathy adverse drug reaction reports supports the concept of a crucial role for 5-HT2B receptor agonism in cardiac valvular fibrosis. Clin Neuropharmacol 2006; 29: 806.Google Scholar
Canesi, M, Mariani, C, Isaias, IU, Pezzoli, G. Night-time use of Rotigotine in advanced Parkinson's disease. Funct Neurol 2010; 25: 2013.Google Scholar
Todorova, A, Martinez-Martin, P, Martin, A, et al. Daytime apomorphine infusion combined with transdermal Rotigotine patch therapy is tolerated at 2 years: a 24-h treatment option in Parkinson's disease. Basal Ganglia 2013; 3: 12730.Google Scholar

References

Fearnley, JM, Lees, AJ. Ageing and Parkinson's disease: substantia nigra regional selectivity. Brain 1991; 114 : 2283301.Google Scholar
Brooks, DJ, Frey, KA, Marek, KL, et al. Assessment of neuroimaging techniques as biomarkers of the progression of Parkinson's disease. Exp Neurol 2003; 184 (Suppl. 1): S6879.Google Scholar
Chaudhuri, KR, Odin, P, Antonini, A, Martinez-Martin, P. Parkinson's disease: the non-motor issues. Parkinsonism Relat Disord 2011; 17: 71723.Google Scholar
Cenci, MA, Ohlin, KE, Odin, P. Current options and future possibilities for the treatment of dyskinesia and motor fluctuations in Parkinson's disease. CNS Neurol Disord Drug Targets 2011; 10: 67084.Google Scholar
Salat, D, Tolosa, E. Levodopa in the treatment of Parkinson's disease: current status and new developments. J Parkinsons Dis 2013; 3: 25569.Google Scholar
Heremans, E, Nieuwboer, A, Vercruysse, S. Freezing of gait in Parkinson's disease: where are we now? Curr Neurol Neurosci Rep 2013; 13: 350.Google Scholar
Carlsson, A, Lindqvist, M, Magnusson, T. 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists. Nature 1957; 180: 1200.Google Scholar
Hornykiewicz, O. Die Topische Lokalisation und das Verhalten von Noradrenalin und Dopamin in der Substantia Nigra des normalen und Parkinsonkranken Menschen. Wien Klin Wochenschr 1963; 75: 30912.Google Scholar
Birkmayer, W, Hornykiewicz, O. Der L-Dioxyphenylalanin (=DOPA – Effekt bei der Parkinson-akinase. Wien Klin Wschr 1961; 73: 7878.Google Scholar
Barbeau, A, Sourkes, TL, Murphy, GF. Les catécholamines dans la maladie de Parkinson. In: de Ajuriaguerra, J, ed. Monoamines et Systéme Nerveaux Central. Geneva: Georg & Cie SA; 1962; 24762.Google Scholar
Cotzias, G, Van Woert, M, Schiffer, L. Aromatic amino acids and modification of parkinsonism. N Engl J Med 1967; 276: 3749.Google Scholar
Calne, DB, Reid, JL, Vakil, SD et al. Idiopathic Parkinsonism treated with an extracerebral decarboxylase inhibitor in combination with levodopa. BMJ 1971; 3: 72932.Google Scholar
Cenci, MA, Lundblad, M. Post- versus presynaptic plasticity in l-DOPA-induced dyskinesia. J Neurochem 2006; 99: 38192.Google Scholar
Nutt, JG, Holford, NH. The response to levodopa in Parkinson's disease: imposing pharmacological law and order. Ann Neurol 1996; 39: 56173.Google Scholar
Nyholm, D, Lennernas, H, Gomes-Trolin, C, Aquilonius, SM. Levodopa pharmacokinetics and motor performance during activities of daily living in patients with Parkinson's disease on individual drug combinations. Clin Neuropharmacol 2002; 25: 8996.Google Scholar
Contin, M, Martinelli, P. Pharmacokinetics of levodopa. J Neurol 2010; 257 (Suppl. 2): S25361.Google Scholar
Nyholm, D, Stepien, V. Levodopa fractionation in Parkinson's disease. J Parkinsons Dis 2014; 4: 8996.Google Scholar
Fasano, A, Bove, F, Gabrielli, M, et al. The role of small intestinal bacterial overgrowth in Parkinson's disease. Mov Disord 2013; 28: 12419.Google Scholar
Chapuis, S, Ouchchane, L, Metz, O, Gerbaud, L, Durif, F. Impact of the motor complications of Parkinson's disease on the quality of life. Mov Disord 2005; 20: 22430.Google Scholar
Fabbrini, G, Brotchie, JM, Grandas, F, Nomoto, M, Goetz, CG. Levodopa-induced dyskinesias. Mov Disord 2007; 22: 137989; quiz 523.Google Scholar
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: 72633.Google Scholar
Fahn, S. Parkinson disease, the effect of levodopa, and the ELLDOPA trial. Earlier vs later l-DOPA. Arch Neurol 1999; 56: 52935.Google Scholar
Fahn, S. A new look at levodopa based on the ELLDOPA study. J Neural Transm Suppl 2006: 41926.Google Scholar
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: 1827.Google Scholar
Castro-Garcia, A. [Psychiatric complications of L-dopa: physiopathology and treatment]. Rev Neurol. 1997; 25 (Suppl. 2): S15762.Google Scholar
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: 58995.Google Scholar
O'Sullivan, SS, Evans, AH, Lees, AJ. Dopamine dysregulation syndrome: an overview of its epidemiology, mechanisms and management. CNS Drugs 2009; 23: 15770.Google Scholar
Spencer, AH, Rickards, H, Fasano, A, Cavanna, AE. The prevalence and clinical characteristics of punding in Parkinson's disease. Mov Disord 2011; 26: 57886.Google Scholar
Storch, A, Schneider, CB, Wolz, M, et al. Nonmotor fluctuations in Parkinson disease: severity and correlation with motor complications. Neurology 2013; 80: 8009.Google Scholar
Rossi, P, Colosimo, C, Moro, E, Tonali, P, Albanese, A. Acute challenge with apomorphine and levodopa in Parkinsonism. Eur Neurol. 2000; 43: 95101.Google Scholar
Cedarbaum, JM, Kutt, H, McDowell, FH. A pharmacokinetic and pharmacodynamic comparison of Sinemet CR (50/200) and standard Sinemet (25/100). Neurology 1989; 39 (Suppl. 2): 3844; discussion 59.Google Scholar
MacMahon, DG, Sachdev, D, Boddie, HG, et al. A comparison of the effects of controlled-release levodopa (Madopar CR) with conventional levodopa in late Parkinson's disease. J Neurol Neurosurg Psychiatry 1990; 53: 2203.Google Scholar
Gauthier, S, Amyot, D. Sustained release antiparkinson agents: controlled release levodopa. Can J Neurol Sci 1992; 19 (Suppl.): 1535.Google Scholar
Sage, JI, Mark, MH. Pharmacokinetics of continuous-release carbidopa/levodopa. Clin Neuropharmacol 1994; 17 (Suppl. 2): S16.Google Scholar
Block, G, Liss, C, Reines, S, Irr, J, Nibbelink, D. Comparison of immediate-release and controlled release carbidopa/levodopa in Parkinson's disease. A multicenter 5-year study. The CR First Study Group. Eur Neurol 1997; 37: 237.Google Scholar
Wolters, EC, Tesselaar, HJ. International (NL-UK) double-blind study of Sinemet CR and standard Sinemet (25/100) in 170 patients with fluctuating Parkinson's disease. J Neurol 1996; 243: 23540.Google Scholar
Jankovic, J, Schwartz, K, Vander Linden, C. Comparison of Sinemet CR4 and standard Sinemet: double blind and long-term open trial in parkinsonian patients with fluctuations. Mov Disord 1989; 4: 3039.Google Scholar
Koller, WC, Hutton, JT, Tolosa, E, Capilldeo, R. Immediate-release and controlled-release carbidopa/levodopa in PD: a 5-year randomized multicenter study. Carbidopa/Levodopa Study Group. Neurology 1999; 53: 10129.Google Scholar
Baas, H, Zehrden, F, Selzer, R, et al. Pharmacokinetic-pharmacodynamic relationship of levodopa with and without tolcapone in patients with Parkinson's disease. Clin Pharmacokinet 2001; 40: 38393.Google Scholar
Kuoppamaki, M, Korpela, K, Marttila, R, et al. Comparison of pharmacokinetic profile of levodopa throughout the day between levodopa/carbidopa/entacapone and levodopa/carbidopa when administered four or five times daily. Eur J Clin Pharmacol 2009; 65: 44355.Google Scholar
Adler, CH, Singer, C, O'Brien, C, et al. Randomized, placebo-controlled study of tolcapone in patients with fluctuating Parkinson disease treated with levodopa-carbidopa. Tolcapone Fluctuator Study Group III. Arch Neurol 1998; 55: 108995.Google Scholar
Deane, KH, Spieker, S, Clarke, CE. Catechol-O-methyltransferase inhibitors for levodopa-induced complications in Parkinson's disease. Cochrane Database Syst Rev 2004; CD004554.Google Scholar
Kurth, MC, Adler, CH, Hilaire, MS, et al. Tolcapone improves motor function and reduces levodopa requirement in patients with Parkinson's disease experiencing motor fluctuations: a multicenter, double-blind, randomized, placebo-controlled trial. Tolcapone Fluctuator Study Group I. Neurology 1997; 48: 817.Google Scholar
Rocha, JF, Santos, A, Falcao, A, et al. Effect of moderate liver impairment on the pharmacokinetics of opicapone. Eur J Clin Pharmacol 2014; 70: 27986.Google Scholar
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: 94754.Google Scholar
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; 361: 126878.Google Scholar
Adler, CH, Sethi, KD, Hauser, RA, et al. Ropinirole for the treatment of early Parkinson's disease. The Ropinirole Study Group. Neurology 1997; 49: 3939.Google Scholar
Shannon, KM, Bennett, JP Jr, Friedman, JH. Efficacy of pramipexole, a novel dopamine agonist, as monotherapy in mild to moderate Parkinson's disease. The Pramipexole Study Group. Neurology 1997; 49: 7248.Google Scholar
Fahn, S, Oakes, D, Shoulson, I, et al. Levodopa and the progression of Parkinson's disease. N Engl J Med 2004; 351: 2498508.Google Scholar
Rinne, UK, Bracco, F, Chouza, C, et al. Cabergoline in the treatment of early Parkinson's disease: results of the first year of treatment in a double-blind comparison of cabergoline and levodopa. The PKDS009 Collaborative Study Group. Neurology 1997; 48: 3638.Google Scholar
Oertel, WH, Wolters, E, Sampaio, C, et al. Pergolide versus levodopa monotherapy in early Parkinson's disease patients: The PELMOPET study. Mov Disord 2006; 21: 34353.Google Scholar
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: 148491.Google Scholar
Butzer, JF, Silver, DE, Sahs, AL. Amantadine in Parkinson's disease. A double-blind, placebo-controlled, crossover study with long-term follow-up. Neurology 1975; 25: 6036.Google Scholar
Fox, SH, Katzenschlager, R, Lim, SY, et al. The Movement Disorder Society Evidence-Based Medicine Review Update: Treatments for the motor symptoms of Parkinson's disease. Mov Disord 2011; 26 (Suppl. 3): S241.Google Scholar
Hauser, RA, Ellenbogen, AL, Metman, LV, et al. Crossover comparison of IPX066 and a standard levodopa formulation in advanced Parkinson's disease. Mov Disord 2011; 26: 224652.Google Scholar
Hauser, RA, Hsu, A, Kell, S, et al. Extended-release carbidopa-levodopa (IPX066) compared with immediate-release carbidopa-levodopa in patients with Parkinson's disease and motor fluctuations: a phase 3 randomised, double-blind trial. Lancet Neurol 2013; 12: 34656.Google Scholar
Hauser, RA. Future treatments for Parkinson's disease: surfing the PD pipeline. Int J Neurosci. 2011; 121 (Suppl. 2): 5362.Google Scholar
LeWitt, PA, Huff, FJ, Hauser, RA, et al. Double-blind study of the actively transported levodopa prodrug XP21279 in Parkinson's disease. Mov Disord 2014; 29: 7582.Google Scholar
Stocchi, F, Zappia, M, Dall'Armi, V, et al. Melevodopa/carbidopa effervescent formulation in the treatment of motor fluctuations in advanced Parkinson's disease. Mov Disord 2010; 25: 18817.Google Scholar
Nyholm, D, Ehrnebo, M, Lewander, T, et al. Frequent administration of levodopa/carbidopa microtablets vs levodopa/carbidopa/entacapone in healthy volunteers. Acta Neurol Scand 2013; 127: 12432.Google Scholar
Shoulson, I, Glaubiger, GA, Chase, TN. On–off response. Clinical and biochemical correlations during oral and intravenous levodopa administration in parkinsonian patients. Neurology 1975; 25: 11448.Google Scholar
Quinn, N, Marsden, CD, Parkes, JD. Complicated response fluctuations in Parkinson's disease: response to intravenous infusion of levodopa. Lancet 1982; 320: 4125.Google Scholar
Kurlan, R, Rubin, AJ, Miller, C, et al. Duodenal delivery of levodopa for on–off fluctuations in parkinsonism: preliminary observations. Ann Neurol 1986; 20: 2625.Google Scholar
Nyholm, D. Enteral levodopa/carbidopa gel infusion for the treatment of motor fluctuations and dyskinesias in advanced Parkinson's disease. Expert Rev Neurother 2006; 6: 140311.Google Scholar
Fernandez, HH, Odin, P. Levodopa-carbidopa intestinal gel for treatment of advanced Parkinson's disease. Curr Med Res Opin. 2011; 27: 90719.Google Scholar
Olanow, CW, Kieburtz, K, Odin, P, et al. Continuous intrajejunal infusion of levodopa-carbidopa intestinal gel for patients with advanced Parkinson's disease: a randomised, controlled, double-blind, double-dummy study. Lancet Neurol 2014; 13: 1419.Google Scholar
Fernandez, HH, Vanagunas, A, Odin, P, et al. Levodopa-carbidopa intestinal gel in advanced Parkinson's disease open-label study: interim results. Parkinsonism Relat Disord 2013; 19: 33945.Google Scholar
Honig, H, Antonini, A, Martinez-Martin, P, et al. Intrajejunal levodopa infusion in Parkinson's disease: a pilot multicenter study of effects on nonmotor symptoms and quality of life. Mov Disord 2009; 24: 146874.Google Scholar
Busk, K, Nyholm, D. Long-term 24-h levodopa/carbidopa gel infusion in Parkinson's disease. Parkinsonism Relat Disord 2012; 18: 10001.Google Scholar
Seppi, K, Weintraub, D, Coelho, M, et al. The Movement Disorder Society Evidence-Based Medicine Review Update: treatments for the non-motor symptoms of Parkinson's disease. Mov Disord 2011; 26 (Suppl. 3): S4280.Google Scholar
Ferreira, JJ, Katzenschlager, R, Bloem, BR, et al. Summary of the recommendations of the EFNS/MDS-ES review on therapeutic management of Parkinson's disease. Eur J Neurol 2013; 20: 515.Google Scholar

References

Korczyn, AD, Keren, O. The effect of dopamine on the papillary diameter in mice. Life Sci 1980; 26: 75763.Google Scholar
Korczyn, AD. Pathophysiology of drug-induced dyskinesias. Neuropharmacology 1973; 11: 6017.Google Scholar
Routtinen, HM, Rinne, UK. Entacapone prolongs levodopa response in one-month double-blind study in Parkinsonian patients with levodopa-related fluctuations. J Neurol Neurosurg Psychiatry 1996; 60: 3640.Google Scholar
Ceravalo, R, Piccini, P, Bailey, DL, et al. 18F-dopa PET evidence that tolcapone acts as a central COMT inhibitor in Parkinson's disease. Synapse 2002; 43: 2017.Google Scholar
Jorga, KM. COMT inhibitors: pharmacokinetics and pharmacodynamic comparisons. Clin Neuropharmacol 1998; 21 (Suppl.): S916.Google Scholar
Corvol, JC, Bonnet, C, Charbonnier-Beaupel, F, et al. The COMT Val158Met polymorphism affects the response to entacapone in Parkinson's disease: a randomized crossover clinical trial. Ann Neurol 2011; 69: 1118.Google Scholar
Maltête, D, Cottard, AM, Mihout, B, Costentin, J. Erythrocytes catechol-O-methyl transferase activity is up-regulated after a 3-month treatment by entacapone in parkinsonian patients. Clin Neuropharmacol 2011; 34: 213.Google Scholar
Agid, Y, Destée, A, Durif, F, et al. Tolcapone, bromocriptine, and Parkinson's disease. French Tolcapone Study Group. Lancet 1997; 350: 71213.Google Scholar
Baas, H, Beiske, AG, Ghika, J. COMT inhibitors with tolcapone reduces ‘wearing-off’ phenomenon and levodopa requirements in fluctuating Parkinsonian patients. J Neurol Neurosurg Psychiatry 1997; 63: 4218.Google Scholar
Adler, CH, Singer, C, O'Brien, C., et al. Randomized placebo-controlled study of tolcapone in patients with fluctuating Parkinson's disease treated with levodopa carbidopa. Arch Neurol 1998; 55: 108995.Google Scholar
Brooks, DJ, Sagar, H, UK–Irish Entacapone Study Group. Entacapone is beneficial in both fluctuating and non-fluctuating patients in Parkinson's disease: a randomized, placebo controlled, double-blind, six months study. J Neurol Neurosurg Psychiatry 2003; 74: 10719.Google Scholar
Fenelon, G, Gimenez-Roldan, S, Monstrut, JL, et al. Efficacy and tolerability of entacapone in patients with Parkinson's disease treated with levodopa plus a dopamine agonist and experiencing wearing-off motor fluctuations. A randomized, double-blind, multicentre study. J Neural Transm 2008; 110: 23951.Google Scholar
Parkinson Study Group. Entacapone improves motor fluctuations in levodopa-treated Parkinson's disease patients. Ann Neurol 1997; 42: 74755.Google Scholar
Myllylä, VV, Kultalahti, ER, Haapaniemi, H, Leinonen, M, FILOMEN Study Group. Twelve-month safety of entacapone in patients with Parkinson's disease. Eur J Neurol 2001; 8: 5360.Google Scholar
Larsen, JP, Worm-Petersen, J, Sideo, A, et al. The tolerability and efficacy of entacapone over 3 years in patients with Parkinson's disease. Eur J Neurol 2008; 10: 13746.Google Scholar
Rinne, UK, Larsen, JP, Siden, A, et al. Entacapone enhances the response to levodopa in Parkinsonian patients with motor fluctuations. Neurology 1998; 51: 130914.Google Scholar
Stocchi, F, Rascol, O, Kieburtz, K, et al. Initiating levodopa/carbidopa therapy with and without entacapone in early PD: the STRIDE-PD study. Ann Neurol 2010; 68: 1827.Google Scholar
Fox, SH, Katzenschlager, R, Lim, SY, et al. The Movement Disorder Society Evidence-Based Medicine Review Update: treatments for the motor symptoms of Parkinson's disease. Mov Disord 2011; 26 (Suppl. 3): S241.Google Scholar
US FDA. FDA Drug Safety Communication: ongoing safety review of Stalevo (entacapone/carbidopa/levodopa) and possible development of prostate cancer. Available at: http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/ucm206363.htm.Google Scholar
Zesiewicz, TA, Wecker, L, Sullivan, KL, Merlin, LR, Hauser, RA. The controversy concerning plasma homocysteine in Parkinson disease patients treated with levodopa alone or with entacapone: effects of vitamin status. Clin Neuropharmacol 2006; 29: 10611.Google Scholar
Stowe, R, Ives, N, Clarke, CE, et al. Meta-analysis of the comparative efficacy and safety of adjuvant treatment to levodopa in later Parkinson's disease. Mov Disord 2011; 26: 58798.Google Scholar
Entacapone to Tolcapone Switch Study Investigators. Entacapone to tolcapone switch: multicenter double-blind, randomized, active-controlled trial in advanced Parkinson's disease. Mov Disord 2007; 22: 1419.Google Scholar
Valeant Pharmaceuticals. TASMAR® (tolcapone). Available at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/020697s004lbl.pdf.Google Scholar
Ferreira, JJ, Rascol, O, Poewe, W, et al. A double-blind, randomized, placebo and active-controlled study of nebicapone for the treatment of motor fluctuations in Parkinson's disease. CNS Neurosci Ther 2010; 16: 33747.Google Scholar
Korczyn, AD, Nisipeanu, P, Newer therapies for Parkinson's disease. Neurol Neurochir Pol 1996; 30 (Suppl. 2): 10511.Google Scholar
Inzelberg, R, Nisipeanu, P, Rabey, JM, et al. Double-blind comparison of caberigolineand bromocriptine in Parkinson's disease patients with motor fluctuations. Neurology 1996; 47: 7858.Google Scholar
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 levidopa. N Engl J Med 2000; 342: 148491.Google Scholar
Parkinson Study Group. A controlled trial of rotigotine monotherapy in early Parkinson's disease. Arch Neurol 2003; 60: 17218.Google Scholar
Korczyn, AD. Transdermal therapy in Parkinson's disease. Lancet Neurol 2007; 6: 4756.Google Scholar
Kurth, MC, Adler, CH, Saint Hilaire, MH. Tolcapone improves motor function and reduces levodopa requirement in patients with Parkinson's disease experiencing motor fluctuations: a multicenter, double-blind, randomized, placebo-controlled trial. Neurology 1997; 48: 817.Google Scholar
Rajput, AH, Martin, W, Saint Hilaire, MH, Dorflinger, E, Pedder, S. Tolcapone improves motor function in Parkinsonian patients with the ‘wearing-off’ phenomenon: a double-blind, placebo-controlled, multicenter trial. Neurology 1997; 49: 106671.Google Scholar
Rabey, M, Sagi, I, Huberman, M, et al. Rasagiline mesylate, a new MAO-B inhibitor for the treatment of Parkinson's disease. Clin Neuropharmacol 2000; 23: 32430.Google Scholar

References

Devos, D. Patient profile, indications, efficacy and safety of duodenal levodopa infusion in advanced Parkinson's disease. Mov Disord 2009; 24: 9931000.Google Scholar
Nyholm, D, Klangemo, K, Johansson, A. Levodopa/carbidopa intestinal gel infusion long-term therapy in advanced Parkinson's disease. Eur J Neurol 2012; 19: 107985.Google Scholar
Zibetti, M, Merola, A, Artusi, CA, et al. Levodopa/carbidopa intestinal gel infusion in advanced Parkinson's disease: a 7-year experience. Eur J Neurol 2014; 21: 3128.Google Scholar
Samanta, J, Hauser, RA. Duodenal levodopa infusion for the treatment of Parkinson's disease. Expert Opin Pharmacother 2007; 8: 65764.Google Scholar
Fernandez, HH, Odin, P. Levodopa-carbidopa intestinal gel for treatment of advanced Parkinson's disease. Curr Med Res Opin. 2011; 27: 90719.Google Scholar
Nyholm, D, Odin, P, Johansson, A, et al. Pharmacokinetics of levodopa, carbidopa, and 3-O-methyldopa following 16-hour jejunal infusion of levodopa-carbidopa intestinal gel in advanced Parkinson's disease patients. AAPS J 2013; 15: 31623.Google Scholar
Nyholm, D, Askmark, H, Gomes-Trolin, C, et al. Optimizing levodopa pharmacokinetics: intestinal infusion versus oral sustained-release tablets. Clin Neuropharmacol 2003; 26: 15663.Google Scholar
Olanow, CW, Kieburtz, K, Odin, P, et al. Continuous intrajejunal infusion of levodopa-carbidopa intestinal gel for patients with advanced Parkinson's disease: a randomised, controlled, double-blind, double-dummy study. Lancet Neurol 2014; 13: 1419.Google Scholar
Fernandez, HH, Vanagunas, A, Odin, P, et al. Levodopa-carbidopa intestinal gel in advanced Parkinson's disease open-label study: interim results. Parkinsonism Relat Disord 2013; 19: 33945.Google Scholar
Espay, AJ. Management of motor complications in Parkinson disease: current and emerging therapies. Neurol Clin. 2010; 28: 91325.Google Scholar
Hauser, RA, Ellenbogen, AL, Metman, LV, et al. Crossover comparison of IPX066 and a standard levodopa formulation in advanced Parkinson's disease. Mov Disord 2011; 26: 224652.Google Scholar
Hauser, RA, Hsu, A, Kell, S, et al. Extended-release carbidopa-levodopa (IPX066) compared with immediate-release carbidopa-levodopa in patients with Parkinson's disease and motor fluctuations: a phase 3 randomised, double-blind trial. Lancet Neurol 2013; 12: 34656.Google Scholar
Pahwa, R, Lyons, KE, Hauser, RA, et al. Randomized trial of IPX066, carbidopa/levodopa extended release, in early Parkinson's disease. Parkinsonism Relat Disord 2014; 20: 1428.Google Scholar
LeWitt, PA, Huff, FJ, Hauser, RA, et al. Double-blind study of the actively transported levodopa prodrug XP21279 in Parkinson's disease. Mov Disord 2014; 29: 7582.Google Scholar
LeWitt, PA, Friedman, H, Giladi, N, et al. Accordion pill carbidopa/levodopa for improved treatment of advanced Parkinson's disease symptoms [abstract]. 2013; 27 (Suppl. 1): S1301.Google Scholar
Metman, LV, Stover, N, Cuiping, C, Cowles, VE, Sweeney, M. Improved motor performance associated with smoother levodopa time-concentration profiles with DM-1992, a gastroretentive, extended-release carbidopa/levodopa tablet, in patients with advanced Parkinson's disease. Neurology 2014; 82: poster 7.082.Google Scholar
Depomed Announces Positive Results from Phase 2 Study of DM-1992 for Parkinson's Disease [press release]. PHARMABIZ.COM, November 9, 2012.Google Scholar
Freed, MF, Grosset, D, Worth, P, et al. Rapid levodopa augmentation following inhaled CVT-301 results in rapid improvement in motor response when administered to PD patients in the OFF state. Neurology 2014; 82: S7.007.Google Scholar
Hauser, RA, Schwarzschild, MA. Adenosine A2A receptor antagonists for Parkinson's disease: rationale, therapeutic potential and clinical experience. Drugs Aging 2005; 22: 47182.Google Scholar
Kanda, T, Jackson, MJ, Smith, LA, et al. Combined use of the adenosine A2A antagonist KW-6002 with l-DOPA or with selective D1 or D2 dopamine agonists increases antiparkinsonian activity but not dyskinesia in MPTP-treated monkeys. Exp Neurol 2000; 162: 3217.Google Scholar
Kase, H, Aoyama, S, Ichimura, M, et al. Progress in pursuit of therapeutic A2A antagonists: the adenosine A2A receptor selective antagonist KW6002: research and development toward a novel nondopaminergic therapy for Parkinson's disease. Neurology 2003; 61 (Suppl. 6): S97–100.Google Scholar
Kalda, A, Yu, L, Oztas, E, Chen, JF. Novel neuroprotection by caffeine and adenosine A2A receptor antagonists in animal models of Parkinson's disease. J Neurol Sci 2006; 248: 915.Google Scholar
Dungo, R, Deeks, ED. Istradefylline: first global approval. Drugs 2013; 73: 87582.Google Scholar
Hauser, RA, Hubble, JP, Truong, DD, Istradefylline US-001 Study Group. Randomized trial of the adenosine A2A receptor antagonist istradefylline in advanced PD. Neurology 2003; 61: 297303.Google Scholar
LeWitt, PA, Guttman, M, Tetrud, JW, et al. 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.Google Scholar
Stacy, M, Silver, D, Mendis, T, et al. A 12-week, placebo-controlled study (6002-US-006) of istradefylline in Parkinson disease. Neurology 2008; 70: 223340.Google Scholar
Hauser, RA, Shulman, LM, Trugman, JM, et al. Study of istradefylline in patients with Parkinson's disease on levodopa with motor fluctuations. Mov Disord 2008; 23: 217785.Google Scholar
Pourcher, E, Fernandez, HH, Stacy, M, et al. Istradefylline for Parkinson's disease patients experiencing motor fluctuations: results of the KW-6002-US-018 study. Parkinsonism Relat Disord 2012; 18: 17884.Google Scholar
Hauser, RA, Cantillon, M, Pourcher, E, et al. Preladenant in patients with Parkinson's disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol 2011; 10: 2219.Google Scholar
Hauser, R, Sticchi, F, Rascol, O, et al. Phase-3 Clinical Trials of Adjunctive Therapy with Preladenant, an Adenosine 2a Antagonist, in Patients with Parkinson's Disease. American Academy of Neurology Annual Meeting; 2014;Google Scholar
Black, KJ CampBell, CM, Dickerson, W, et al., eds. A randomized, double-blind, placebo-controlled cross-over trial of the adenosine 2a antagonist SYN115 in Parkinson disease. Neurology; 2010; 82: poster P7.087.Google Scholar
Hauser, RA Olanow, CW, Kieburtz, KD, et al. A phase 2, placebo-controlled, randomized, double-blind trial of tozadenant (SYN-115) in patients with Parkinson's disease with wearing-off fluctuations on levodopa. Mov Disord 2013; 333 (Suppl. 1): e119.Google Scholar
Dall'Igna, OP, Porciuncula, LO, Souza, DO, Cunha, RA, Lara, DR. Neuroprotection by caffeine and adenosine A2A receptor blockade of β-amyloid neurotoxicity. Br J Pharmacol 2003; 138: 12079.Google Scholar
Stocchi, F, Borgohain, R, Onofrj, M, et al. A randomized, double-blind, placebo-controlled trial of safinamide as add-on therapy in early Parkinson's disease patients. Mov Disord 2012; 27: 10612.Google Scholar
Stocchi, F, Borgohain, R, Onofrj, M, et al. A randomized, double-blind, placebo-controlled trial of safinamide as add-on therapy in early Parkinson's disease patients. Mov Disord 2012; 27: 10612.Google Scholar
Gottwald, MD, Aminoff, MJ. Therapies for dopaminergic-induced dyskinesias in Parkinson disease. Ann Neurol 2011; 69: 91927.Google Scholar
Pahwa, R, Tanner, CM, Hauser, RA, et al. Randomized trial of extended release amantadine in Parkinson's disease patients with levodopa-induced dyskinesia (EASED Study). Mov Disord 2013;28 (Suppl. 1): 443.Google Scholar
Lewitt, PA, Hauser, RA, Lu, M, et al. Randomized clinical trial of fipamezole for dyskinesia in Parkinson disease (FJORD study). Neurology 2012; 79: 1639.Google Scholar
Dimitrova, T, Bara-Jimenez, W, Savola, JM, et al. Alpha-2 adrenergic antagonist effects in Parkinson's disease. Mov Disord 2009; 24 (Suppl. 1): S261.Google Scholar
Johnston, TH, Fox, SH, McIldowie, MJ, Piggott, MJ, Brotchie, JM. Reduction of l-DOPA-induced dyskinesia by the selective metabotropic glutamate receptor 5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson's disease. J Pharmacol Exp Ther 2010; 333: 86573.Google Scholar
Stocchi, F, Rascol, O, Destée, A, et al. AFQ056 in Parkinson patients with levodopa-induced dyskinesia: 13-week, randomized, dose-finding study. Mov Disord 2013; 28: 183846.Google Scholar
Parkinson Study Group. Phase II safety, tolerability, and dose selection study of isradipine as a potential disease-modifying intervention in early Parkinson's disease (STEADY-PD). Mov Disord 2013; 28: 182331.Google Scholar
Parkinson Study Group SURE-P Inbestigators, Schwarzschild, MA, Ascherio, A, et al. Inosine to increase serum and cerebrospinal fluid urate in Parkinson disease: a randomized clinical trial. JAMA Neurol 2014; 71: 14150.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×