Skip to main content
Top
Published in: Journal of Neural Transmission 11/2018

01-11-2018 | Neurology and Preclinical Neurological Studies - Review Article

Monoamine oxidase-B inhibitors in the treatment of Parkinson’s disease: clinical–pharmacological aspects

Authors: Peter Riederer, Thomas Müller

Published in: Journal of Neural Transmission | Issue 11/2018

Login to get access

Abstract

This invited narrative review emphasizes the role of MAO-B inhibition in the drug portfolio for dopamine substitution in patients with Parkinson’s disease. Neuronal and glial MAO-B inhibition contributes to more stable levels of dopamine and other biogenic amines in the synaptic cleft. Accordingly, symptomatic effects of MAO-B inhibition for a limited amelioration of impaired motor behaviour and wearing-off phenomena in patients with Parkinson’s disease are well proven, even when MAO-B inhibitors are only applied together with dopamine agonists. Delay of disease progression by MAO-B inhibition is under debate despite positive experimental findings. This discussion does not consider, that levodopa, respectively, dopamine agonists, are substrates, respectively, inhibitors of the ABCB1 (P-gp, MDR1, and CD243) transporter system. It supports toxin efflux over the blood–brain barrier. ABCB1 transporters have a limited capacity. MAO-B inhibitors do not weaken it. Treatment with MAO-B inhibitors is advantageous as it enables sparing of dopamine agonist and levodopa dosing.
Literature
go back to reference Apetauerova D, Scala SA, Hamill RW, Simon DK, Pathak S, Ruthazer R, Standaert DG, Yacoubian TA (2016) CoQ10 in progressive supranuclear palsy: a randomized, placebo-controlled, double-blind trial. Neurol Neuroimmunol Neuroinflamm 3:e266CrossRefPubMedPubMedCentral Apetauerova D, Scala SA, Hamill RW, Simon DK, Pathak S, Ruthazer R, Standaert DG, Yacoubian TA (2016) CoQ10 in progressive supranuclear palsy: a randomized, placebo-controlled, double-blind trial. Neurol Neuroimmunol Neuroinflamm 3:e266CrossRefPubMedPubMedCentral
go back to reference Barone P, Fernandez HH, Ferreira J, Müller T, Saint-Hilaire M, Stacy M, Tolosa E, Anand R (2013) Safinamide as an add-on therapy to a stable dose of a single dopamine agonist: results from a randomized, placebo-controlled, 24-week multicenter trial in early idiopathic Parkinson disease (PD) patients (MOTION Study). Neurology 80:P01–P061 Barone P, Fernandez HH, Ferreira J, Müller T, Saint-Hilaire M, Stacy M, Tolosa E, Anand R (2013) Safinamide as an add-on therapy to a stable dose of a single dopamine agonist: results from a randomized, placebo-controlled, 24-week multicenter trial in early idiopathic Parkinson disease (PD) patients (MOTION Study). Neurology 80:P01–P061
go back to reference Bartels AL, van Berckel BN, Lubberink M, Luurtsema G, Lammertsma AA, Leenders KL (2008a) Blood–brain barrier P-glycoprotein function is not impaired in early Parkinson’s disease. Parkinsonism Relat Disord 14:505–508CrossRefPubMed Bartels AL, van Berckel BN, Lubberink M, Luurtsema G, Lammertsma AA, Leenders KL (2008a) Blood–brain barrier P-glycoprotein function is not impaired in early Parkinson’s disease. Parkinsonism Relat Disord 14:505–508CrossRefPubMed
go back to reference Bartels AL, Willemsen AT, Kortekaas R, de Jong BM, de Vries R, de Klerk O, van Oostrom JC, Portman A, Leenders KL (2008b) Decreased blood–brain barrier P-glycoprotein function in the progression of Parkinson’s disease, PSP and MSA. J Neural Transm 115:1001–1009CrossRefPubMed Bartels AL, Willemsen AT, Kortekaas R, de Jong BM, de Vries R, de Klerk O, van Oostrom JC, Portman A, Leenders KL (2008b) Decreased blood–brain barrier P-glycoprotein function in the progression of Parkinson’s disease, PSP and MSA. J Neural Transm 115:1001–1009CrossRefPubMed
go back to reference Beal MF, Oakes D, Shoulson I, Henchcliffe C, Galpern WR, Haas R, Juncos JL, Nutt JG, Voss TS, Ravina B, Shults CM, Helles K, Snively V, Lew MF, Griebner B, Watts A, Gao S, Pourcher E, Bond L, Kompoliti K, Agarwal P, Sia C, Jog M, Cole L, Sultana M, Kurlan R, Richard I, Deeley C, Waters CH, Figueroa A, Arkun A, Brodsky M, Ondo WG, Hunter CB, Jimenez-Shahed J, Palao A, Miyasaki JM, So J, Tetrud J, Reys L, Smith K, Singer C, Blenke A, Russell DS, Cotto C, Friedman JH, Lannon M, Zhang L, Drasby E, Kumar R, Subramanian T, Ford DS, Grimes DA, Cote D, Conway J, Siderowf AD, Evatt ML, Sommerfeld B, Lieberman AN, Okun MS, Rodriguez RL, Merritt S, Swartz CL, Martin WR, King P, Stover N, Guthrie S, Watts RL, Ahmed A, Fernandez HH, Winters A, Mari Z, Dawson TM, Dunlop B, Feigin AS, Shannon B, Nirenberg MJ, Ogg M, Ellias SA, Thomas CA, Frei K, Bodis-Wollner I, Glazman S, Mayer T, Hauser RA, Pahwa R, Langhammer A, Ranawaya R, Derwent L, Sethi KD, Farrow B, Prakash R, Litvan I, Robinson A, Sahay A, Gartner M, Hinson VK, Markind S, Pelikan M, Perlmutter JS, Hartlein J, Molho E, Evans S, Adler CH, Duffy A, Lind M, Elmer L, Davis K, Spears J, Wilson S, Leehey MA, Hermanowicz N, Niswonger S, Shill HA, Obradov S, Rajput A, Cowper M, Lessig S, Song D, Fontaine D, Zadikoff C, Williams K, Blindauer KA, Bergholte J, Propsom CS, Stacy MA, Field J, Mihaila D, Chilton M, Uc EY, Sieren J, Simon DK, Kraics L, Silver A, Boyd JT, Hamill RW, Ingvoldstad C, Young J, Thomas K, Kostyk SK, Wojcieszek J, Pfeiffer RF, Panisset M, Beland M, Reich SG, Cines M, Zappala N, Rivest J, Zweig R, Lumina LP, Hilliard CL, Grill S, Kellermann M, Tuite P, Rolandelli S, Kang UJ, Young J, Rao J, Cook MM, Severt L, Boyar K (2014) A randomized clinical trial of high-dosage coenzyme Q10 in early Parkinson disease: no evidence of benefit. JAMA Neurol 71:543–552CrossRefPubMed Beal MF, Oakes D, Shoulson I, Henchcliffe C, Galpern WR, Haas R, Juncos JL, Nutt JG, Voss TS, Ravina B, Shults CM, Helles K, Snively V, Lew MF, Griebner B, Watts A, Gao S, Pourcher E, Bond L, Kompoliti K, Agarwal P, Sia C, Jog M, Cole L, Sultana M, Kurlan R, Richard I, Deeley C, Waters CH, Figueroa A, Arkun A, Brodsky M, Ondo WG, Hunter CB, Jimenez-Shahed J, Palao A, Miyasaki JM, So J, Tetrud J, Reys L, Smith K, Singer C, Blenke A, Russell DS, Cotto C, Friedman JH, Lannon M, Zhang L, Drasby E, Kumar R, Subramanian T, Ford DS, Grimes DA, Cote D, Conway J, Siderowf AD, Evatt ML, Sommerfeld B, Lieberman AN, Okun MS, Rodriguez RL, Merritt S, Swartz CL, Martin WR, King P, Stover N, Guthrie S, Watts RL, Ahmed A, Fernandez HH, Winters A, Mari Z, Dawson TM, Dunlop B, Feigin AS, Shannon B, Nirenberg MJ, Ogg M, Ellias SA, Thomas CA, Frei K, Bodis-Wollner I, Glazman S, Mayer T, Hauser RA, Pahwa R, Langhammer A, Ranawaya R, Derwent L, Sethi KD, Farrow B, Prakash R, Litvan I, Robinson A, Sahay A, Gartner M, Hinson VK, Markind S, Pelikan M, Perlmutter JS, Hartlein J, Molho E, Evans S, Adler CH, Duffy A, Lind M, Elmer L, Davis K, Spears J, Wilson S, Leehey MA, Hermanowicz N, Niswonger S, Shill HA, Obradov S, Rajput A, Cowper M, Lessig S, Song D, Fontaine D, Zadikoff C, Williams K, Blindauer KA, Bergholte J, Propsom CS, Stacy MA, Field J, Mihaila D, Chilton M, Uc EY, Sieren J, Simon DK, Kraics L, Silver A, Boyd JT, Hamill RW, Ingvoldstad C, Young J, Thomas K, Kostyk SK, Wojcieszek J, Pfeiffer RF, Panisset M, Beland M, Reich SG, Cines M, Zappala N, Rivest J, Zweig R, Lumina LP, Hilliard CL, Grill S, Kellermann M, Tuite P, Rolandelli S, Kang UJ, Young J, Rao J, Cook MM, Severt L, Boyar K (2014) A randomized clinical trial of high-dosage coenzyme Q10 in early Parkinson disease: no evidence of benefit. JAMA Neurol 71:543–552CrossRefPubMed
go back to reference Calzetti S, Negrotti A, Cassio A (1995) l-deprenyl as an adjunct to low-dose bromocriptine in early Parkinson’s disease: a short-term, double-blind, and prospective follow-up study. Clin Neuropharmacol 18:250–257CrossRefPubMed Calzetti S, Negrotti A, Cassio A (1995) l-deprenyl as an adjunct to low-dose bromocriptine in early Parkinson’s disease: a short-term, double-blind, and prospective follow-up study. Clin Neuropharmacol 18:250–257CrossRefPubMed
go back to reference de Rijk MC, Launer LJ, Berger K, Breteler MM, Dartigues JF, Baldereschi M, Fratiglioni L, Lobo A, Martinez-Lage J, Trenkwalder C, Hofman A (2000) Prevalence of Parkinson’s disease in Europe: a collaborative study of population-based cohorts. Neurologic Diseases in the Elderly Research Group. Neurology 54:S21–S23PubMed de Rijk MC, Launer LJ, Berger K, Breteler MM, Dartigues JF, Baldereschi M, Fratiglioni L, Lobo A, Martinez-Lage J, Trenkwalder C, Hofman A (2000) Prevalence of Parkinson’s disease in Europe: a collaborative study of population-based cohorts. Neurologic Diseases in the Elderly Research Group. Neurology 54:S21–S23PubMed
go back to reference Dutheil F, Beaune P, Tzourio C, Loriot MA, Elbaz A (2010) Interaction between ABCB1 and professional exposure to organochlorine insecticides in Parkinson disease. Arch Neurol 67:739–745CrossRefPubMed Dutheil F, Beaune P, Tzourio C, Loriot MA, Elbaz A (2010) Interaction between ABCB1 and professional exposure to organochlorine insecticides in Parkinson disease. Arch Neurol 67:739–745CrossRefPubMed
go back to reference Ekesbo A, Rydin E, Torstenson R, Sydow O, Laengstrom B, Tedroff J (1999) Dopamine autoreceptor function is lost in advanced Parkinson’s disease. Neurology 52:120–125CrossRefPubMed Ekesbo A, Rydin E, Torstenson R, Sydow O, Laengstrom B, Tedroff J (1999) Dopamine autoreceptor function is lost in advanced Parkinson’s disease. Neurology 52:120–125CrossRefPubMed
go back to reference Fuller RW, Clemens JA, Hynes MD III (1982) Degree of selectivity of pergolide as an agonist at presynaptic versus postsynaptic dopamine receptors: implications for prevention or treatment of tardive dyskinesia. J Clin Psychopharmacol 2:371–375CrossRefPubMed Fuller RW, Clemens JA, Hynes MD III (1982) Degree of selectivity of pergolide as an agonist at presynaptic versus postsynaptic dopamine receptors: implications for prevention or treatment of tardive dyskinesia. J Clin Psychopharmacol 2:371–375CrossRefPubMed
go back to reference Garcia-Ruiz PJ, Martinez Castrillo JC, Alonso-Canovas A, Herranz BA, Vela L, Sanchez AP, Mata M, Olmedilla GN, Mahillo Fernandez I (2014) Impulse control disorder in patients with Parkinson’s disease under dopamine agonist therapy: a multicentre study. J Neurol Neurosurg Psychiatry 85:840–844CrossRefPubMed Garcia-Ruiz PJ, Martinez Castrillo JC, Alonso-Canovas A, Herranz BA, Vela L, Sanchez AP, Mata M, Olmedilla GN, Mahillo Fernandez I (2014) Impulse control disorder in patients with Parkinson’s disease under dopamine agonist therapy: a multicentre study. J Neurol Neurosurg Psychiatry 85:840–844CrossRefPubMed
go back to reference Götz ME, Gerstner A, Harth R, Dirr A, Janetzky B, Kuhn W, Riederer P, Gerlach M (2000) Altered redox state of platelet coenzyme Q10 in Parkinson’s disease. J Neural Transm 107:41–48CrossRefPubMed Götz ME, Gerstner A, Harth R, Dirr A, Janetzky B, Kuhn W, Riederer P, Gerlach M (2000) Altered redox state of platelet coenzyme Q10 in Parkinson’s disease. J Neural Transm 107:41–48CrossRefPubMed
go back to reference Gregory AM, Buysse DJ, Willis TA, Rijsdijk FV, Maughan B, Rowe R, Cartwright S, Barclay NL, Eley TC (2011) Associations between sleep quality and anxiety and depression symptoms in a sample of young adult twins and siblings. J Psychosom Res 71:250–255CrossRefPubMed Gregory AM, Buysse DJ, Willis TA, Rijsdijk FV, Maughan B, Rowe R, Cartwright S, Barclay NL, Eley TC (2011) Associations between sleep quality and anxiety and depression symptoms in a sample of young adult twins and siblings. J Psychosom Res 71:250–255CrossRefPubMed
go back to reference Hauser RA, Silver D, Choudhry A, Eyal E, Isaacson S (2014) Randomized, controlled trial of rasagiline as an add-on to dopamine agonists in Parkinson’s disease. Mov Disord 29:1028–1034CrossRefPubMed Hauser RA, Silver D, Choudhry A, Eyal E, Isaacson S (2014) Randomized, controlled trial of rasagiline as an add-on to dopamine agonists in Parkinson’s disease. Mov Disord 29:1028–1034CrossRefPubMed
go back to reference Häussermann P, Kuhn W, Przuntek H, Müller T (2001) Integrity of the blood-cerebrospinal fluid barrier in early Parkinson’s disease. Neurosci Lett 300:182–184CrossRefPubMed Häussermann P, Kuhn W, Przuntek H, Müller T (2001) Integrity of the blood-cerebrospinal fluid barrier in early Parkinson’s disease. Neurosci Lett 300:182–184CrossRefPubMed
go back to reference Korchounov A, Winter Y, Rossy W (2012) Combined beneficial effect of rasagiline on motor function and depression in de novo PD. Clin Neuropharmacol 35:121–124CrossRefPubMed Korchounov A, Winter Y, Rossy W (2012) Combined beneficial effect of rasagiline on motor function and depression in de novo PD. Clin Neuropharmacol 35:121–124CrossRefPubMed
go back to reference Kovacs N, Janszky J, Nagy F (2011) Cost effectiveness of rasagiline and pramipexole as treatment strategies in early Parkinson’s disease in the UK setting: an economic Markov model evaluation. Drugs Aging 28:161–162CrossRefPubMed Kovacs N, Janszky J, Nagy F (2011) Cost effectiveness of rasagiline and pramipexole as treatment strategies in early Parkinson’s disease in the UK setting: an economic Markov model evaluation. Drugs Aging 28:161–162CrossRefPubMed
go back to reference Kronstrand R, Ahlner J, Dizdar N, Larson G (2003) Quantitative analysis of desmethylselegiline, methamphetamine, and amphetamine in hair and plasma from Parkinson patients on long-term selegiline medication. J Anal Toxicol 27:135–141CrossRefPubMed Kronstrand R, Ahlner J, Dizdar N, Larson G (2003) Quantitative analysis of desmethylselegiline, methamphetamine, and amphetamine in hair and plasma from Parkinson patients on long-term selegiline medication. J Anal Toxicol 27:135–141CrossRefPubMed
go back to reference Laine K, Anttila M, Heinonen E, Helminen A, Huupponen R, Maki-Ikola O, Reinikainen K, Scheinin M (1997) Lack of adverse interactions between concomitantly administered selegiline and citalopram. Clin Neuropharmacol 20:419–433CrossRefPubMed Laine K, Anttila M, Heinonen E, Helminen A, Huupponen R, Maki-Ikola O, Reinikainen K, Scheinin M (1997) Lack of adverse interactions between concomitantly administered selegiline and citalopram. Clin Neuropharmacol 20:419–433CrossRefPubMed
go back to reference Lecht S, Haroutiunian S, Hoffman A, Lazarovici P (2007) Rasagiline—a novel MAO B inhibitor in Parkinson’s disease therapy. Ther Clin Risk Manag 3:467–474PubMedPubMedCentral Lecht S, Haroutiunian S, Hoffman A, Lazarovici P (2007) Rasagiline—a novel MAO B inhibitor in Parkinson’s disease therapy. Ther Clin Risk Manag 3:467–474PubMedPubMedCentral
go back to reference Mao A, Freeman KA, Tallarida RJ (1996) Transient loss of dopamine autoreceptor control in the presence of highly potent dopamine agonists. Life Sci 59:L317–L324CrossRef Mao A, Freeman KA, Tallarida RJ (1996) Transient loss of dopamine autoreceptor control in the presence of highly potent dopamine agonists. Life Sci 59:L317–L324CrossRef
go back to reference Mizuno Y, Hattori N, Kondo T, Nomoto M, Origasa H, Takahashi R, Yamamoto M, Yanagisawa N (2017) A Randomized double-blind placebo-controlled phase III trial of selegiline monotherapy for early Parkinson disease. Clin Neuropharmacol 40:201–207CrossRefPubMedPubMedCentral Mizuno Y, Hattori N, Kondo T, Nomoto M, Origasa H, Takahashi R, Yamamoto M, Yanagisawa N (2017) A Randomized double-blind placebo-controlled phase III trial of selegiline monotherapy for early Parkinson disease. Clin Neuropharmacol 40:201–207CrossRefPubMedPubMedCentral
go back to reference Muhlack S, Herrmann L, Salmen S, Müller T (2014) Fewer fluctuations, higher maximum concentration and better motor response of levodopa with catechol-O-methyltransferase inhibition. J Neural Transm 121:1357–1366CrossRefPubMed Muhlack S, Herrmann L, Salmen S, Müller T (2014) Fewer fluctuations, higher maximum concentration and better motor response of levodopa with catechol-O-methyltransferase inhibition. J Neural Transm 121:1357–1366CrossRefPubMed
go back to reference Müller T (2012) Drug therapy in patients with Parkinson’s disease. Transl Neurodegener 1:1–10CrossRef Müller T (2012) Drug therapy in patients with Parkinson’s disease. Transl Neurodegener 1:1–10CrossRef
go back to reference Müller T (2014) Pharmacokinetic/pharmacodynamic evaluation of rasagiline mesylate for Parkinson’s disease. Expert Opin Drug Metab Toxicol 10:1423–1432CrossRefPubMed Müller T (2014) Pharmacokinetic/pharmacodynamic evaluation of rasagiline mesylate for Parkinson’s disease. Expert Opin Drug Metab Toxicol 10:1423–1432CrossRefPubMed
go back to reference Müller T, Foley P (2017) Clinical pharmacokinetics and pharmacodynamics of safinamide. Clin Pharmacokinet 56:251–261CrossRefPubMed Müller T, Foley P (2017) Clinical pharmacokinetics and pharmacodynamics of safinamide. Clin Pharmacokinet 56:251–261CrossRefPubMed
go back to reference Müller T, Eising EG, Reiners C, Przuntek H, Jacob M, Kuhn W (1997) 2-[123I]-iodolisuride SPET visualizes dopaminergic loss in de-novo parkinsonian patients: is it a marker of striatal pre-synaptic degeneration? Nucl Med Commun 18:1115–1121CrossRefPubMed Müller T, Eising EG, Reiners C, Przuntek H, Jacob M, Kuhn W (1997) 2-[123I]-iodolisuride SPET visualizes dopaminergic loss in de-novo parkinsonian patients: is it a marker of striatal pre-synaptic degeneration? Nucl Med Commun 18:1115–1121CrossRefPubMed
go back to reference Müller T, Hoffmann JA, Dimpfel W, Oehlwein C (2013) Switch from selegiline to rasagiline is beneficial in patients with Parkinson’s disease. J Neural Transm 120:761–765CrossRefPubMed Müller T, Hoffmann JA, Dimpfel W, Oehlwein C (2013) Switch from selegiline to rasagiline is beneficial in patients with Parkinson’s disease. J Neural Transm 120:761–765CrossRefPubMed
go back to reference Müller T, Öhm G, Eilert K, Möhr K, Rotter S, Haas T, Küchler M, Lütge S, Marg M, Rothe H (2017) Benefit on motor and non-motor behavior in a specialized unit for Parkinson’s disease. J Neural Transm 124:715–720CrossRefPubMed Müller T, Öhm G, Eilert K, Möhr K, Rotter S, Haas T, Küchler M, Lütge S, Marg M, Rothe H (2017) Benefit on motor and non-motor behavior in a specialized unit for Parkinson’s disease. J Neural Transm 124:715–720CrossRefPubMed
go back to reference Nappi G, Martignoni E, Horowski R, Pacchetti C, Rainer E, Bruggi P, Runge I (1991) Lisuride plus selegiline in the treatment of early Parkinson’s disease. Acta Neurol Scand 83:407–410CrossRefPubMed Nappi G, Martignoni E, Horowski R, Pacchetti C, Rainer E, Bruggi P, Runge I (1991) Lisuride plus selegiline in the treatment of early Parkinson’s disease. Acta Neurol Scand 83:407–410CrossRefPubMed
go back to reference Olanow CW, Rascol O, Hauser R, Feigin PD, Jankovic J, Lang A, Langston W, Melamed E, Poewe W, Stocchi F, Tolosa E (2009) A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 361:1268–1278CrossRefPubMed Olanow CW, Rascol O, Hauser R, Feigin PD, Jankovic J, Lang A, Langston W, Melamed E, Poewe W, Stocchi F, Tolosa E (2009) A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 361:1268–1278CrossRefPubMed
go back to reference Parkinson Study Group (1996) Impact of deprenyl and tocopherol treatment on Parkinson’s disease in DATATOP patients requiring levodopa. Ann Neurol 39:37–45CrossRef Parkinson Study Group (1996) Impact of deprenyl and tocopherol treatment on Parkinson’s disease in DATATOP patients requiring levodopa. Ann Neurol 39:37–45CrossRef
go back to reference Pilleri M, Antonini A (2014) Novel levodopa formulations in the treatment of Parkinson’s disease. Expert Rev Neurother 14:143–149CrossRefPubMed Pilleri M, Antonini A (2014) Novel levodopa formulations in the treatment of Parkinson’s disease. Expert Rev Neurother 14:143–149CrossRefPubMed
go back to reference Pingili R, Vemulapalli S, Mullapudi SS, Nuthakki S, Pendyala S, Kilaru N (2016) Pharmacokinetic interaction study between flavanones (hesperetin, naringenin) and rasagiline mesylate in wistar rats. Drug Dev Ind Pharm 42:1110–1117CrossRefPubMed Pingili R, Vemulapalli S, Mullapudi SS, Nuthakki S, Pendyala S, Kilaru N (2016) Pharmacokinetic interaction study between flavanones (hesperetin, naringenin) and rasagiline mesylate in wistar rats. Drug Dev Ind Pharm 42:1110–1117CrossRefPubMed
go back to reference Przuntek H, Conrad B, Dichgans J, Kraus PH, Krauseneck P, Pergande G, Rinne U, Schimrigk K, Schnitker J, Vogel HP (1999) SELEDO: a 5-year long-term trial on the effect of selegiline in early Parkinsonian patients treated with levodopa. Eur J Neurol 6:141–150CrossRefPubMed Przuntek H, Conrad B, Dichgans J, Kraus PH, Krauseneck P, Pergande G, Rinne U, Schimrigk K, Schnitker J, Vogel HP (1999) SELEDO: a 5-year long-term trial on the effect of selegiline in early Parkinsonian patients treated with levodopa. Eur J Neurol 6:141–150CrossRefPubMed
go back to reference Przuntek H, Bittkau S, Bliesath H, Buttner U, Fuchs G, Glass J, Haller H, Klockgether T, Kraus P, Lachenmayer L, Müller D, Müller T, Rathay B, Sgonina J, Steinijans V, Teshmar E, Ulm G, Volc D (2002) Budipine provides additional benefit in patients with Parkinson disease receiving a stable optimum dopaminergic drug regimen. Arch Neurol 59:803–806CrossRefPubMed Przuntek H, Bittkau S, Bliesath H, Buttner U, Fuchs G, Glass J, Haller H, Klockgether T, Kraus P, Lachenmayer L, Müller D, Müller T, Rathay B, Sgonina J, Steinijans V, Teshmar E, Ulm G, Volc D (2002) Budipine provides additional benefit in patients with Parkinson disease receiving a stable optimum dopaminergic drug regimen. Arch Neurol 59:803–806CrossRefPubMed
go back to reference Przuntek H, Müller T, Riederer P (2004) Diagnostic staging of Parkinson’s disease: conceptual aspects. J Neural Transm 111:201–216CrossRefPubMed Przuntek H, Müller T, Riederer P (2004) Diagnostic staging of Parkinson’s disease: conceptual aspects. J Neural Transm 111:201–216CrossRefPubMed
go back to reference Rajput AH, Birdi S (1997) Epidemiology of Parkinson’s disease. Parkinsonism Relat Disord 3:175–186CrossRefPubMed Rajput AH, Birdi S (1997) Epidemiology of Parkinson’s disease. Parkinsonism Relat Disord 3:175–186CrossRefPubMed
go back to reference Rascol O, Hauser RA, Stocchi F, Fitzer-Attas CJ, Sidi Y, Abler V, Olanow CW, Investigators AFU (2016) Long-term effects of rasagiline and the natural history of treated Parkinson’s disease. Mov Disord 31:1489–1496CrossRefPubMed Rascol O, Hauser RA, Stocchi F, Fitzer-Attas CJ, Sidi Y, Abler V, Olanow CW, Investigators AFU (2016) Long-term effects of rasagiline and the natural history of treated Parkinson’s disease. Mov Disord 31:1489–1496CrossRefPubMed
go back to reference Reynolds GP, Riederer P, Sandler M, Jellinger K, Seeman D (1978) Amphetamine and 2-phenylethylamine in post-mortem Parkinsonian brain after (-)deprenyl administration. J Neural Transm 43:271–277CrossRefPubMed Reynolds GP, Riederer P, Sandler M, Jellinger K, Seeman D (1978) Amphetamine and 2-phenylethylamine in post-mortem Parkinsonian brain after (-)deprenyl administration. J Neural Transm 43:271–277CrossRefPubMed
go back to reference Riederer P, Jellinger K, Danielczyk W, Seemann D, Ulm G, Reynolds GP, Birkmayer W, Koppel H (1983) Combination treatment with selective monoamine oxidase inhibitors and dopaminergic agonists in Parkinson’s disease: biochemical and clinical observations. Adv Neural 37:159–176 Riederer P, Jellinger K, Danielczyk W, Seemann D, Ulm G, Reynolds GP, Birkmayer W, Koppel H (1983) Combination treatment with selective monoamine oxidase inhibitors and dopaminergic agonists in Parkinson’s disease: biochemical and clinical observations. Adv Neural 37:159–176
go back to reference Schapira AH, McDermott MP, Barone P, Comella CL, Albrecht S, Hsu HH, Massey DH, Mizuno Y, Poewe W, Rascol O, Marek K (2013) Pramipexole in patients with early Parkinson’s disease (PROUD): a randomised delayed-start trial. Lancet Neurol 12:747–755CrossRefPubMedPubMedCentral Schapira AH, McDermott MP, Barone P, Comella CL, Albrecht S, Hsu HH, Massey DH, Mizuno Y, Poewe W, Rascol O, Marek K (2013) Pramipexole in patients with early Parkinson’s disease (PROUD): a randomised delayed-start trial. Lancet Neurol 12:747–755CrossRefPubMedPubMedCentral
go back to reference Sian-Hülsmann J, Mandel S, Youdim MB, Riederer P (2011) The relevance of iron in the pathogenesis of Parkinson’s disease. J Neurochem 118:939–957CrossRefPubMed Sian-Hülsmann J, Mandel S, Youdim MB, Riederer P (2011) The relevance of iron in the pathogenesis of Parkinson’s disease. J Neurochem 118:939–957CrossRefPubMed
go back to reference Sian-Hülsmann J, Monoranu C, Strobel S, Riederer P (2015) Lewy bodies: a spectator or salient killer? CNS Neurol Disord Drug Targets 14:947–955CrossRefPubMed Sian-Hülsmann J, Monoranu C, Strobel S, Riederer P (2015) Lewy bodies: a spectator or salient killer? CNS Neurol Disord Drug Targets 14:947–955CrossRefPubMed
go back to reference Tanner CM, Ross GW, Jewell SA, Hauser RA, Jankovic J, Factor SA, Bressman S, Deligtisch A, Marras C, Lyons KE, Bhudhikanok GS, Roucoux DF, Meng C, Abbott RD, Langston JW (2009) Occupation and risk of parkinsonism: a multicenter case-control study. Arch Neurol 66:1106–1113CrossRefPubMed Tanner CM, Ross GW, Jewell SA, Hauser RA, Jankovic J, Factor SA, Bressman S, Deligtisch A, Marras C, Lyons KE, Bhudhikanok GS, Roucoux DF, Meng C, Abbott RD, Langston JW (2009) Occupation and risk of parkinsonism: a multicenter case-control study. Arch Neurol 66:1106–1113CrossRefPubMed
go back to reference Vautier S, Fernandez C (2009) ABCB1: the role in Parkinson’s disease and pharmacokinetics of antiparkinsonian drugs. Expert Opin Drug Metab Toxicol 5:1349–1358CrossRefPubMed Vautier S, Fernandez C (2009) ABCB1: the role in Parkinson’s disease and pharmacokinetics of antiparkinsonian drugs. Expert Opin Drug Metab Toxicol 5:1349–1358CrossRefPubMed
go back to reference Vautier S, Milane A, Fernandez C, Buyse M, Chacun H, Farinotti R (2008) Interactions between antiparkinsonian drugs and ABCB1/P-glycoprotein at the blood–brain barrier in a rat brain endothelial cell model. Neurosci Lett 442:19–23CrossRefPubMed Vautier S, Milane A, Fernandez C, Buyse M, Chacun H, Farinotti R (2008) Interactions between antiparkinsonian drugs and ABCB1/P-glycoprotein at the blood–brain barrier in a rat brain endothelial cell model. Neurosci Lett 442:19–23CrossRefPubMed
go back to reference Yoritaka A, Kawajiri S, Yamamoto Y, Nakahara T, Ando M, Hashimoto K, Nagase M, Saito Y, Hattori N (2015) Randomized, double-blind, placebo-controlled pilot trial of reduced coenzyme Q10 for Parkinson’s disease. Parkinsonism Relat Disord 21:911–916CrossRefPubMed Yoritaka A, Kawajiri S, Yamamoto Y, Nakahara T, Ando M, Hashimoto K, Nagase M, Saito Y, Hattori N (2015) Randomized, double-blind, placebo-controlled pilot trial of reduced coenzyme Q10 for Parkinson’s disease. Parkinsonism Relat Disord 21:911–916CrossRefPubMed
Metadata
Title
Monoamine oxidase-B inhibitors in the treatment of Parkinson’s disease: clinical–pharmacological aspects
Authors
Peter Riederer
Thomas Müller
Publication date
01-11-2018
Publisher
Springer Vienna
Published in
Journal of Neural Transmission / Issue 11/2018
Print ISSN: 0300-9564
Electronic ISSN: 1435-1463
DOI
https://doi.org/10.1007/s00702-018-1876-2

Other articles of this Issue 11/2018

Journal of Neural Transmission 11/2018 Go to the issue

Neurology and Preclinical Neurological Studies - Review Article

The structure of monoamine oxidases: past, present, and future

Psychiatry and Preclinical Psychiatric Studies - Review Article

Epigenetic signature of MAOA and MAOB genes in mental disorders