Skip to main content
Top
Published in: Drugs & Aging 6/2019

01-06-2019 | Polyneuropathy | Review Article

Safety and Tolerability of Pharmacotherapies for Parkinson’s Disease in Geriatric Patients

Authors: Martin Klietz, Stephan Greten, Florian Wegner, Günter U. Höglinger

Published in: Drugs & Aging | Issue 6/2019

Login to get access

Abstract

Parkinson’s disease is a chronic neurodegenerative movement disorder affecting people mainly beyond their 50s. Geriatric patients with Parkinson’s disease experience a specific profile of comorbidities. Multimorbidity and resulting polypharmacotherapy are frequent at this age. Comorbid diseases, widely spread, involve arterial hypertension, ischemic heart disease, heart failure, atrial fibrillation, polyneuropathy, diabetes mellitus, cerebrovascular disease, sarcopenia, and frailty. Following years of drug development, levodopa is still the most effective drug for the treatment of motor symptoms. However, a wide range of other drugs are available with specific effects, contraindications, and complications. The treatment of geriatric patients with Parkinson’s disease is challenging and requires the cooperation of multidisciplinary teams. A careful assessment of a patient’s Parkinson’s disease symptoms, comorbidities, medication, vital signs, and resources is crucial for an effective and safe therapy. Laboratory tests can assist in the identification of contraindications for specific treatments. Identifying potentially inadequate drugs from prescription lists can lead to a better targeted treatment for geriatric patients with Parkinson’s disease. Future research should help develop a more evidence-based therapy of geriatric patients with Parkinson’s disease. For this purpose, randomized controlled trials of geriatric patients are urgently needed. An international register concerning issues of safer drug application and monitoring could help to implement a better treatment.
Literature
1.
2.
go back to reference Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s disease. N Engl J Med. 2003;348:1356–64.CrossRefPubMed Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s disease. N Engl J Med. 2003;348:1356–64.CrossRefPubMed
3.
go back to reference Politis M, Wu K, Molloy S, Bain PG, Chaudhuri KR, Piccini P. Parkinson’s disease symptoms: the patients perspective. Mov Disord. 2010;25:1646–51.CrossRefPubMed Politis M, Wu K, Molloy S, Bain PG, Chaudhuri KR, Piccini P. Parkinson’s disease symptoms: the patients perspective. Mov Disord. 2010;25:1646–51.CrossRefPubMed
4.
go back to reference Forsaa EB, Larsen JP, Wentzel-Larsen T, Herlofson K, Alves G. Predictors and course of health-related quality of life in Parkinson’s disease. Mov Disord. 2008;23:1420–7.CrossRefPubMed Forsaa EB, Larsen JP, Wentzel-Larsen T, Herlofson K, Alves G. Predictors and course of health-related quality of life in Parkinson’s disease. Mov Disord. 2008;23:1420–7.CrossRefPubMed
5.
go back to reference Soh S-E, Morris ME, McGinley JL. Determinants of health-related quality of life in Parkinson’s disease: a systematic review. Parkinsonism Relat Disord. 2011;17:1–9.CrossRefPubMed Soh S-E, Morris ME, McGinley JL. Determinants of health-related quality of life in Parkinson’s disease: a systematic review. Parkinsonism Relat Disord. 2011;17:1–9.CrossRefPubMed
6.
go back to reference Klietz M, Tulke A, Müschen LH, Paracka L, Schrader C, Dressler DW, et al. Impaired quality of life and need for palliative care in a German cohort of advanced Parkinson’s disease patients. Front Neurol. 2018;9:120.CrossRefPubMedPubMedCentral Klietz M, Tulke A, Müschen LH, Paracka L, Schrader C, Dressler DW, et al. Impaired quality of life and need for palliative care in a German cohort of advanced Parkinson’s disease patients. Front Neurol. 2018;9:120.CrossRefPubMedPubMedCentral
7.
go back to reference Kempster PA, O’Sullivan SS, Holton JL, Revesz T, Lees AJ. Relationships between age and late progression of Parkinson’s disease: a clinico-pathological study. Brain. 2010;133:1755–62.CrossRefPubMed Kempster PA, O’Sullivan SS, Holton JL, Revesz T, Lees AJ. Relationships between age and late progression of Parkinson’s disease: a clinico-pathological study. Brain. 2010;133:1755–62.CrossRefPubMed
8.
go back to reference Muller-Rebstein S, Trenkwalder C, Oertel WH, Culmsee C, Eckermann G, Höglinger GU. Pharmacotherapy of Parkinson’s disease: aspects of drug safety. Nervenarzt. 2017;88:888–94.CrossRefPubMed Muller-Rebstein S, Trenkwalder C, Oertel WH, Culmsee C, Eckermann G, Höglinger GU. Pharmacotherapy of Parkinson’s disease: aspects of drug safety. Nervenarzt. 2017;88:888–94.CrossRefPubMed
10.
go back to reference Lingor P, Csoti I, Koschel J, Schrader C, Winkler C, Wolz M, et al. The geriatric patient with Parkinson’s disease: a neurological challenge. Fortschr Neurol Psychiatry. 2016;84(Suppl. 1):S41–7. Lingor P, Csoti I, Koschel J, Schrader C, Winkler C, Wolz M, et al. The geriatric patient with Parkinson’s disease: a neurological challenge. Fortschr Neurol Psychiatry. 2016;84(Suppl. 1):S41–7.
11.
go back to reference Jakovljevic M, Ostojic L. Comorbidity and multimorbidity in medicine today: challenges and opportunities for bringing separated branches of medicine closer to each other. Psychiatr Danub. 2013;25(Suppl. 1):18–28.PubMed Jakovljevic M, Ostojic L. Comorbidity and multimorbidity in medicine today: challenges and opportunities for bringing separated branches of medicine closer to each other. Psychiatr Danub. 2013;25(Suppl. 1):18–28.PubMed
12.
13.
go back to reference Barnett K, Mercer SW, Norbury M, Watt G, Wyke S, Guthrie B. Epidemiology of multimorbidity and implications for health care, research, and medical education: a cross-sectional study. Lancet. 2012;380:37–43.CrossRefPubMed Barnett K, Mercer SW, Norbury M, Watt G, Wyke S, Guthrie B. Epidemiology of multimorbidity and implications for health care, research, and medical education: a cross-sectional study. Lancet. 2012;380:37–43.CrossRefPubMed
14.
go back to reference Hou JG, Wu LJ, Moore S, Ward C, York M, Atassi F, et al. Assessment of appropriate medication administration for hospitalized patients with Parkinson’s disease. Parkinsonism Relat Disord. 2012;18:377–81.CrossRefPubMed Hou JG, Wu LJ, Moore S, Ward C, York M, Atassi F, et al. Assessment of appropriate medication administration for hospitalized patients with Parkinson’s disease. Parkinsonism Relat Disord. 2012;18:377–81.CrossRefPubMed
15.
go back to reference McLean G, Hindle JV, Guthrie B, Mercer SW. Co-morbidity and polypharmacy in Parkinson’s disease: insights from a large Scottish primary care database. BMC Neurol. 2017;17:126.CrossRefPubMedPubMedCentral McLean G, Hindle JV, Guthrie B, Mercer SW. Co-morbidity and polypharmacy in Parkinson’s disease: insights from a large Scottish primary care database. BMC Neurol. 2017;17:126.CrossRefPubMedPubMedCentral
16.
go back to reference Choi J, Ahn A, Kim S, Won CW. Global prevalence of physical frailty by Fried’s criteria in community-dwelling elderly with national population-based surveys. J Am Med Dir Assoc. 2015;16:548–50.CrossRefPubMed Choi J, Ahn A, Kim S, Won CW. Global prevalence of physical frailty by Fried’s criteria in community-dwelling elderly with national population-based surveys. J Am Med Dir Assoc. 2015;16:548–50.CrossRefPubMed
17.
go back to reference Santos-Eggimann B, Cuenoud P, Spagnoli J, Junod J. Prevalence of frailty in middle-aged and older community-dwelling Europeans living in 10 countries. J Gerontol A Biol Sci Med Sci. 2009;64:675–81.CrossRefPubMed Santos-Eggimann B, Cuenoud P, Spagnoli J, Junod J. Prevalence of frailty in middle-aged and older community-dwelling Europeans living in 10 countries. J Gerontol A Biol Sci Med Sci. 2009;64:675–81.CrossRefPubMed
19.
go back to reference Riedel O, Dodel R, Deuschl G, Förstl H, Henn F, Heuser I, et al. Dementia and depression determine care dependency in Parkinson’s disease: analysis of 1,449 outpatients receiving nursing care in Germany. Nervenarzt. 2011;82:1012–9.CrossRefPubMed Riedel O, Dodel R, Deuschl G, Förstl H, Henn F, Heuser I, et al. Dementia and depression determine care dependency in Parkinson’s disease: analysis of 1,449 outpatients receiving nursing care in Germany. Nervenarzt. 2011;82:1012–9.CrossRefPubMed
20.
go back to reference Riedel O, Dodel R, Deuschl G, Klotsche J, Förstl H, Heuser I, et al. Depression and care-dependency in Parkinson’s disease: results from a nationwide study of 1449 outpatients. Parkinsonism Relat Disord. 2012;18:598–601.CrossRefPubMed Riedel O, Dodel R, Deuschl G, Klotsche J, Förstl H, Heuser I, et al. Depression and care-dependency in Parkinson’s disease: results from a nationwide study of 1449 outpatients. Parkinsonism Relat Disord. 2012;18:598–601.CrossRefPubMed
21.
go back to reference Leibson CL, Maraganore DM, Bower JH, Ransom JE, O’Brien PC, Rocca WA. Comorbid conditions associated with Parkinson’s disease: a population-based study. Mov Disord. 2005;21:446–55.CrossRef Leibson CL, Maraganore DM, Bower JH, Ransom JE, O’Brien PC, Rocca WA. Comorbid conditions associated with Parkinson’s disease: a population-based study. Mov Disord. 2005;21:446–55.CrossRef
22.
go back to reference Tönges L, Bartig D, Muhlack S, Jost W, Gold R, Krogias C. Characteristics and dynamics of inpatient treatment of patients with Parkinson’s disease in Germany: analysis of 1.5 million patient cases from 2010 to 2015. Nervenarzt. 2018;2018(16):552. Tönges L, Bartig D, Muhlack S, Jost W, Gold R, Krogias C. Characteristics and dynamics of inpatient treatment of patients with Parkinson’s disease in Germany: analysis of 1.5 million patient cases from 2010 to 2015. Nervenarzt. 2018;2018(16):552.
23.
go back to reference Müller-Rebstein S, Trenkwalder C, Ebentheuer J, Oertel WH, Culmsee C, Höglinger GU. Drug safety analysis in a real-life cohort of Parkinson’s disease patients with polypharmacy. CNS Drugs. 2018;31:1093–102.CrossRef Müller-Rebstein S, Trenkwalder C, Ebentheuer J, Oertel WH, Culmsee C, Höglinger GU. Drug safety analysis in a real-life cohort of Parkinson’s disease patients with polypharmacy. CNS Drugs. 2018;31:1093–102.CrossRef
24.
go back to reference Balzer-Geldsetzer M, Ferreira J, Odin P, Bloem BR, Meissner WG, Lorenzl S, et al. Study protocol: Care of Late-Stage Parkinsonism (CLaSP): a longitudinal cohort study. BMC Neurol. 2018;18:185.CrossRefPubMedPubMedCentral Balzer-Geldsetzer M, Ferreira J, Odin P, Bloem BR, Meissner WG, Lorenzl S, et al. Study protocol: Care of Late-Stage Parkinsonism (CLaSP): a longitudinal cohort study. BMC Neurol. 2018;18:185.CrossRefPubMedPubMedCentral
25.
go back to reference Zesiewicz TA, Strom JA, Borenstein AR, Hauser RA, Cimino CR, Fontanet HL, et al. Heart failure in Parkinson’s disease: analysis of the United States Medicare current beneficiary survey. Parkinsonism Relat Disord. 2004;10:417–20.CrossRefPubMed Zesiewicz TA, Strom JA, Borenstein AR, Hauser RA, Cimino CR, Fontanet HL, et al. Heart failure in Parkinson’s disease: analysis of the United States Medicare current beneficiary survey. Parkinsonism Relat Disord. 2004;10:417–20.CrossRefPubMed
26.
go back to reference Santiago JA, Bottero V, Potashkin JA. Biological and clinical implications of comorbidities in Parkinson’s disease. Front Aging Neurosci. 2017;9:394.CrossRefPubMedPubMedCentral Santiago JA, Bottero V, Potashkin JA. Biological and clinical implications of comorbidities in Parkinson’s disease. Front Aging Neurosci. 2017;9:394.CrossRefPubMedPubMedCentral
27.
go back to reference Lai S-W, Lin C-L, Liao K-F, Chang-Ou K-C. Increased risk of Parkinson’s disease in cataract patients: a population-based cohort study. Parkinsonism Relat Disord. 2015;21:68–71.CrossRefPubMed Lai S-W, Lin C-L, Liao K-F, Chang-Ou K-C. Increased risk of Parkinson’s disease in cataract patients: a population-based cohort study. Parkinsonism Relat Disord. 2015;21:68–71.CrossRefPubMed
28.
go back to reference Jellinger KA. Prevalence of cerebrovascular lesions in Parkinson’s disease: a postmortem study. Acta Neuropathol. 2003;105:415–9.PubMed Jellinger KA. Prevalence of cerebrovascular lesions in Parkinson’s disease: a postmortem study. Acta Neuropathol. 2003;105:415–9.PubMed
29.
go back to reference Hong CT, Hu H-H, Chan L, Bai C-H. Prevalent cerebrovascular and cardiovascular disease in people with Parkinson’s disease: a meta-analysis. Clin Epidemiol. 2018;10:1147–54.CrossRefPubMedPubMedCentral Hong CT, Hu H-H, Chan L, Bai C-H. Prevalent cerebrovascular and cardiovascular disease in people with Parkinson’s disease: a meta-analysis. Clin Epidemiol. 2018;10:1147–54.CrossRefPubMedPubMedCentral
30.
go back to reference Ebersbach G, Sojer M, Muller J, Ransmayr G, Wenning G, Poewe W. Dysequilibrium in idiopathic Parkinson disease: the effect of cerebrovascular comorbidity. Nervenarzt. 2002;73:162–5.CrossRefPubMed Ebersbach G, Sojer M, Muller J, Ransmayr G, Wenning G, Poewe W. Dysequilibrium in idiopathic Parkinson disease: the effect of cerebrovascular comorbidity. Nervenarzt. 2002;73:162–5.CrossRefPubMed
31.
go back to reference Kotagal V, Albin RL, Muller MLTM, Koeppe RA, Studenski S, Frey KA, et al. Advanced age, cardiovascular risk burden, and Timed Up and Go Test performance in Parkinson disease. J Gerontol A Biol Sci Med Sci. 2014;69:1569–75.CrossRefPubMedPubMedCentral Kotagal V, Albin RL, Muller MLTM, Koeppe RA, Studenski S, Frey KA, et al. Advanced age, cardiovascular risk burden, and Timed Up and Go Test performance in Parkinson disease. J Gerontol A Biol Sci Med Sci. 2014;69:1569–75.CrossRefPubMedPubMedCentral
32.
go back to reference Hu G, Jousilahti P, Bidel S, Antikainen R, Tuomilehto J. Type 2 diabetes and the risk of Parkinson’s disease. Diabetes Care. 2007;30:842–7.CrossRefPubMed Hu G, Jousilahti P, Bidel S, Antikainen R, Tuomilehto J. Type 2 diabetes and the risk of Parkinson’s disease. Diabetes Care. 2007;30:842–7.CrossRefPubMed
33.
go back to reference Cereda E, Barichella M, Cassani E, Caccialanza R, Pezzoli G. Clinical features of Parkinson disease when onset of diabetes came first: a case–control study. Neurology. 2012;78:1507–11.CrossRefPubMed Cereda E, Barichella M, Cassani E, Caccialanza R, Pezzoli G. Clinical features of Parkinson disease when onset of diabetes came first: a case–control study. Neurology. 2012;78:1507–11.CrossRefPubMed
34.
go back to reference Kotagal V, Albin RL, Müller MLTM, Koeppe RA, Frey KA, Bohnen NI. Diabetes is associated with postural instability and gait difficulty in Parkinson disease. Parkinsonism Relat Disord. 2013;19:522–6.CrossRefPubMedPubMedCentral Kotagal V, Albin RL, Müller MLTM, Koeppe RA, Frey KA, Bohnen NI. Diabetes is associated with postural instability and gait difficulty in Parkinson disease. Parkinsonism Relat Disord. 2013;19:522–6.CrossRefPubMedPubMedCentral
35.
go back to reference Bohnen NI, Kotagal V, Müller MLTM, Koeppe RA, Scott PJH, Albin RL, et al. Diabetes mellitus is independently associated with more severe cognitive impairment in Parkinson disease. Parkinsonism Relat Disord. 2014;20:1394–8.CrossRefPubMedPubMedCentral Bohnen NI, Kotagal V, Müller MLTM, Koeppe RA, Scott PJH, Albin RL, et al. Diabetes mellitus is independently associated with more severe cognitive impairment in Parkinson disease. Parkinsonism Relat Disord. 2014;20:1394–8.CrossRefPubMedPubMedCentral
36.
go back to reference Metta V, Sanchez TC, Padmakumar C. Osteoporosis: a hidden nonmotor face of Parkinson’s disease. Int Rev Neurobiol. 2017;134:877–90.CrossRefPubMed Metta V, Sanchez TC, Padmakumar C. Osteoporosis: a hidden nonmotor face of Parkinson’s disease. Int Rev Neurobiol. 2017;134:877–90.CrossRefPubMed
37.
go back to reference Deng Q, Zhou X, Chen J, Pan M, Gao H, Zhou J, et al. Lower hemoglobin levels in patients with Parkinson’s disease are associated with disease severity and iron metabolism. Brain Res. 2017;1655:145–51.CrossRefPubMed Deng Q, Zhou X, Chen J, Pan M, Gao H, Zhou J, et al. Lower hemoglobin levels in patients with Parkinson’s disease are associated with disease severity and iron metabolism. Brain Res. 2017;1655:145–51.CrossRefPubMed
38.
go back to reference Wielinski CL, Erickson-Davis C, Wichmann R, Walde-Douglas M, Parashos SA. Falls and injuries resulting from falls among patients with Parkinson’s disease and other parkinsonian syndromes. Mov Disord. 2005;20:410–5.CrossRefPubMed Wielinski CL, Erickson-Davis C, Wichmann R, Walde-Douglas M, Parashos SA. Falls and injuries resulting from falls among patients with Parkinson’s disease and other parkinsonian syndromes. Mov Disord. 2005;20:410–5.CrossRefPubMed
39.
go back to reference Huang Y-F, Cherng Y-G, Hsu SPC, Yeh C-C, Chou Y-C, Wu C-H, et al. Risk and adverse outcomes of fractures in patients with Parkinson’s disease: two nationwide studies. Osteoporos Int. 2015;26:1723–32.CrossRefPubMed Huang Y-F, Cherng Y-G, Hsu SPC, Yeh C-C, Chou Y-C, Wu C-H, et al. Risk and adverse outcomes of fractures in patients with Parkinson’s disease: two nationwide studies. Osteoporos Int. 2015;26:1723–32.CrossRefPubMed
41.
go back to reference Hornykiewicz O. Dopamine miracle: from brain homogenate to dopamine replacement. Mov Disord. 2002;17:501–8.CrossRefPubMed Hornykiewicz O. Dopamine miracle: from brain homogenate to dopamine replacement. Mov Disord. 2002;17:501–8.CrossRefPubMed
43.
44.
go back to reference Cedarbaum JM. Clinical pharmacokinetics of anti-parkinsonian drugs. Clin Pharmacokinet. 1987;13:141–78.CrossRefPubMed Cedarbaum JM. Clinical pharmacokinetics of anti-parkinsonian drugs. Clin Pharmacokinet. 1987;13:141–78.CrossRefPubMed
45.
go back to reference Wood LD. Clinical review and treatment of select adverse effects of dopamine receptor agonists in Parkinson’s disease. Drugs Aging. 2010;27:295–310.CrossRefPubMed Wood LD. Clinical review and treatment of select adverse effects of dopamine receptor agonists in Parkinson’s disease. Drugs Aging. 2010;27:295–310.CrossRefPubMed
46.
go back to reference Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov Disord. 2001;16:448–58.CrossRefPubMed Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov Disord. 2001;16:448–58.CrossRefPubMed
47.
go back to reference Schrag A, Ben-Shlomo Y, Quinn N. How common are complications of Parkinson’s disease? J Neurol. 2002;249:419–23.CrossRefPubMed Schrag A, Ben-Shlomo Y, Quinn N. How common are complications of Parkinson’s disease? J Neurol. 2002;249:419–23.CrossRefPubMed
48.
go back to reference Fahn S. Parkinson disease, the effect of levodopa, and the ELLDOPA trial earlier vs later l-DOPA. Arch Neurol. 1999;56:529–35.CrossRefPubMed Fahn S. Parkinson disease, the effect of levodopa, and the ELLDOPA trial earlier vs later l-DOPA. Arch Neurol. 1999;56:529–35.CrossRefPubMed
49.
go back to reference Olanow CW, Obeso JA, Stocchi F. Drug insight: continuous dopaminergic stimulation in the treatment of Parkinson’s disease. Nat Clin Pract Neurol. 2006;2:382–92.CrossRefPubMed Olanow CW, Obeso JA, Stocchi F. Drug insight: continuous dopaminergic stimulation in the treatment of Parkinson’s disease. Nat Clin Pract Neurol. 2006;2:382–92.CrossRefPubMed
50.
go back to reference Blanchet PJ, Grondin R, Bedard PJ, Shiosaki K, Britton DR. Dopamine D1 receptor desensitization profile in MPTP-lesioned primates. Eur J Pharmacol. 1996;309:13–20.CrossRefPubMed Blanchet PJ, Grondin R, Bedard PJ, Shiosaki K, Britton DR. Dopamine D1 receptor desensitization profile in MPTP-lesioned primates. Eur J Pharmacol. 1996;309:13–20.CrossRefPubMed
51.
go back to reference Olanow CW, Obeso JA, Stocchi F. Continuous dopamine-receptor treatment of Parkinson’s disease: scientific rationale and clinical implications. Lancet Neurol. 2006;5:677–87.CrossRefPubMed Olanow CW, Obeso JA, Stocchi F. Continuous dopamine-receptor treatment of Parkinson’s disease: scientific rationale and clinical implications. Lancet Neurol. 2006;5:677–87.CrossRefPubMed
52.
go back to reference Keber U, Klietz M, Carlsson T, Oertel WH, Weihe E, Schafer MKH, et al. Striatal tyrosine hydroxylase-positive neurons are associated with l-DOPA-induced dyskinesia in hemiparkinsonian mice. Neurosciene. 2015;298:302–17.CrossRef Keber U, Klietz M, Carlsson T, Oertel WH, Weihe E, Schafer MKH, et al. Striatal tyrosine hydroxylase-positive neurons are associated with l-DOPA-induced dyskinesia in hemiparkinsonian mice. Neurosciene. 2015;298:302–17.CrossRef
53.
go back to reference Klietz M, Keber U, Carlsson T, Chiu W-H, Höglinger GU, Weihe E, et al. l-DOPA-induced dyskinesia is associated with a deficient numerical downregulation of striatal tyrosine hydroxylase mRNA-expressing neurons. Neuroscience. 2016;331:120–33.CrossRefPubMed Klietz M, Keber U, Carlsson T, Chiu W-H, Höglinger GU, Weihe E, et al. l-DOPA-induced dyskinesia is associated with a deficient numerical downregulation of striatal tyrosine hydroxylase mRNA-expressing neurons. Neuroscience. 2016;331:120–33.CrossRefPubMed
54.
55.
go back to reference Othman AA, Dutta S. Population pharmacokinetics of levodopa in subjects with advanced Parkinson’s disease: levodopa-carbidopa intestinal gel infusion vs. oral tablets. Br J Clin Pharmacol. 2014;78:94–105.CrossRefPubMedPubMedCentral Othman AA, Dutta S. Population pharmacokinetics of levodopa in subjects with advanced Parkinson’s disease: levodopa-carbidopa intestinal gel infusion vs. oral tablets. Br J Clin Pharmacol. 2014;78:94–105.CrossRefPubMedPubMedCentral
56.
go back to reference Stocchi F, Vacca L, Stirpe P, Torti M. Pharmacokinetic drug evaluation of CVT-301 for the treatment of Parkinson’s disease. Expert Opin Drug Metab Toxicol. 2018;14:1189–95.CrossRefPubMed Stocchi F, Vacca L, Stirpe P, Torti M. Pharmacokinetic drug evaluation of CVT-301 for the treatment of Parkinson’s disease. Expert Opin Drug Metab Toxicol. 2018;14:1189–95.CrossRefPubMed
57.
58.
go back to reference Weintraub D, Chiang C, Kim HM, Wilkinson J, Marras C, Stanislawski B, et al. Association of antipsychotic use with mortality risk in patients with Parkinson disease. JAMA Neurol. 2016;73:535–41.CrossRefPubMedPubMedCentral Weintraub D, Chiang C, Kim HM, Wilkinson J, Marras C, Stanislawski B, et al. Association of antipsychotic use with mortality risk in patients with Parkinson disease. JAMA Neurol. 2016;73:535–41.CrossRefPubMedPubMedCentral
59.
go back to reference Weintraub D, Chiang C, Kim HM, Wilkinson J, Marras C, Stanislawski B, et al. Antipsychotic use and physical morbidity in Parkinson disease. Am J Geriatr Psychiatry. 2017;25:697–705.CrossRefPubMed Weintraub D, Chiang C, Kim HM, Wilkinson J, Marras C, Stanislawski B, et al. Antipsychotic use and physical morbidity in Parkinson disease. Am J Geriatr Psychiatry. 2017;25:697–705.CrossRefPubMed
60.
61.
go back to reference Fredericks D, Norton JC, Atchison C, Schoenhaus R, Pill MW. Parkinson’s disease and Parkinsons disease psychosis: a perspective on the challenges, treatments, and economic burden. Am J Manag Care. 2017;23:S83–92.PubMed Fredericks D, Norton JC, Atchison C, Schoenhaus R, Pill MW. Parkinson’s disease and Parkinsons disease psychosis: a perspective on the challenges, treatments, and economic burden. Am J Manag Care. 2017;23:S83–92.PubMed
62.
go back to reference Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA. 2005;294:1934–43.CrossRefPubMed Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA. 2005;294:1934–43.CrossRefPubMed
63.
go back to reference Meltzer HY, Mills R, Revell S, Williams H, Johnson A, Bahr D, et al. Pimavanserin, a serotonin(2A) receptor inverse agonist, for the treatment of Parkinson’s disease psychosis. Neuropsychopharmacology. 2010;35:881–92.CrossRefPubMed Meltzer HY, Mills R, Revell S, Williams H, Johnson A, Bahr D, et al. Pimavanserin, a serotonin(2A) receptor inverse agonist, for the treatment of Parkinson’s disease psychosis. Neuropsychopharmacology. 2010;35:881–92.CrossRefPubMed
65.
go back to reference Schapira AHV. Monoamine oxidase B inhibitors for the treatment of Parkinson’s disease: a review of symptomatic and potential disease-modifying effects. CNS Drugs. 2011;25:1061–71.CrossRefPubMed Schapira AHV. Monoamine oxidase B inhibitors for the treatment of Parkinson’s disease: a review of symptomatic and potential disease-modifying effects. CNS Drugs. 2011;25:1061–71.CrossRefPubMed
66.
go back to reference Cereda E, Cilia R, Canesi M, Tesei S, Mariani CB, Zecchinelli AL, et al. Efficacy of rasagiline and selegiline in Parkinson’s disease: a head-to-head 3-year retrospective case–control study. J Neurol. 2017;264:1254–63.CrossRefPubMedPubMedCentral Cereda E, Cilia R, Canesi M, Tesei S, Mariani CB, Zecchinelli AL, et al. Efficacy of rasagiline and selegiline in Parkinson’s disease: a head-to-head 3-year retrospective case–control study. J Neurol. 2017;264:1254–63.CrossRefPubMedPubMedCentral
67.
go back to reference Kumagai T, Nagayama H, Ota T, Nishiyama Y, Mishina M, Ueda M. Sex differences in the pharmacokinetics of levodopa in elderly patients with Parkinson disease. Clin Neuropharmacol. 2014;37:173–6.CrossRefPubMed Kumagai T, Nagayama H, Ota T, Nishiyama Y, Mishina M, Ueda M. Sex differences in the pharmacokinetics of levodopa in elderly patients with Parkinson disease. Clin Neuropharmacol. 2014;37:173–6.CrossRefPubMed
68.
go back to reference Nagayama H, Ueda M, Kumagai T, Tsukamoto K, Nishiyama Y, Nishimura S, et al. Influence of ageing on the pharmacokinetics of levodopa in elderly patients with Parkinson’s disease. Parkinsonism Relat Disord. 2011;17:150–2.CrossRefPubMed Nagayama H, Ueda M, Kumagai T, Tsukamoto K, Nishiyama Y, Nishimura S, et al. Influence of ageing on the pharmacokinetics of levodopa in elderly patients with Parkinson’s disease. Parkinsonism Relat Disord. 2011;17:150–2.CrossRefPubMed
69.
go back to reference Kanis JA, Cooper C, Rizzoli R, Reginster J-Y. European guidance for the diagnosis and management of osteoporosis in postmenopausal women. Osteoporos Int. 2019;30:3–44.CrossRefPubMed Kanis JA, Cooper C, Rizzoli R, Reginster J-Y. European guidance for the diagnosis and management of osteoporosis in postmenopausal women. Osteoporos Int. 2019;30:3–44.CrossRefPubMed
70.
go back to reference van den Bos F, Speelman AD, Samson M, Munneke M, Bloem BR, Verhaar HJJ. Parkinson’s disease and osteoporosis. Age Ageing. 2013;42:156–62.CrossRefPubMed van den Bos F, Speelman AD, Samson M, Munneke M, Bloem BR, Verhaar HJJ. Parkinson’s disease and osteoporosis. Age Ageing. 2013;42:156–62.CrossRefPubMed
71.
go back to reference Yasui K, Nakaso K, Kowa H, Takeshima T, Nakashima K. Levodopa-induced hyperhomocysteinaemia in Parkinson’s disease. Acta Neurol Scand. 2003;108:66–7.CrossRefPubMed Yasui K, Nakaso K, Kowa H, Takeshima T, Nakashima K. Levodopa-induced hyperhomocysteinaemia in Parkinson’s disease. Acta Neurol Scand. 2003;108:66–7.CrossRefPubMed
73.
go back to reference Szadejko K, Dziewiatowski K, Szabat K, Robowski P, Schinwelski M, Sitek E, et al. Polyneuropathy in levodopa-treated Parkinson’s patients. J Neurol Sci. 2016;371:36–41.CrossRefPubMed Szadejko K, Dziewiatowski K, Szabat K, Robowski P, Schinwelski M, Sitek E, et al. Polyneuropathy in levodopa-treated Parkinson’s patients. J Neurol Sci. 2016;371:36–41.CrossRefPubMed
74.
go back to reference Loens S, Chorbadzhieva E, Kleimann A, Dressler D, Schrader C. Effects of levodopa/carbidopa intestinal gel versus oral levodopa/carbidopa on B vitamin levels and neuropathy. Brain Behav. 2017;7:e00698.CrossRefPubMedPubMedCentral Loens S, Chorbadzhieva E, Kleimann A, Dressler D, Schrader C. Effects of levodopa/carbidopa intestinal gel versus oral levodopa/carbidopa on B vitamin levels and neuropathy. Brain Behav. 2017;7:e00698.CrossRefPubMedPubMedCentral
75.
go back to reference Velseboer DC, de Haan RJ, Wieling W, Goldstein DS, de Bie RMA. Prevalence of orthostatic hypotension in Parkinson’s disease: a systematic review and meta-analysis. Parkinsonism Relat Disord. 2011;17:724–9.CrossRefPubMedPubMedCentral Velseboer DC, de Haan RJ, Wieling W, Goldstein DS, de Bie RMA. Prevalence of orthostatic hypotension in Parkinson’s disease: a systematic review and meta-analysis. Parkinsonism Relat Disord. 2011;17:724–9.CrossRefPubMedPubMedCentral
76.
go back to reference Ferrer-Gila T, Rizea C. Orthostatic hypotension in the elderly. Rev Neurol. 2013;56:337–43.PubMed Ferrer-Gila T, Rizea C. Orthostatic hypotension in the elderly. Rev Neurol. 2013;56:337–43.PubMed
77.
go back to reference Jost WH, Augustis S. Severity of orthostatic hypotension in the course of Parkinson’s disease: no correlation with the duration of the disease. Parkinsonism Relat Disord. 2015;21:314–6.CrossRefPubMed Jost WH, Augustis S. Severity of orthostatic hypotension in the course of Parkinson’s disease: no correlation with the duration of the disease. Parkinsonism Relat Disord. 2015;21:314–6.CrossRefPubMed
78.
go back to reference Noack C, Schroeder C, Heusser K, Lipp A. Cardiovascular effects of levodopa in Parkinson’s disease. Parkinsonism Relat Disord. 2014;20:815–8.CrossRef Noack C, Schroeder C, Heusser K, Lipp A. Cardiovascular effects of levodopa in Parkinson’s disease. Parkinsonism Relat Disord. 2014;20:815–8.CrossRef
79.
go back to reference Kondo M, Ueda Y, Makino M, Nakajima K. Worsened orthostatic hypotension due to levodopa administration in a case of Parkinson’s disease. Nihon Ronen Igakkai Zasshi. 2000;37:255–8.CrossRefPubMed Kondo M, Ueda Y, Makino M, Nakajima K. Worsened orthostatic hypotension due to levodopa administration in a case of Parkinson’s disease. Nihon Ronen Igakkai Zasshi. 2000;37:255–8.CrossRefPubMed
80.
go back to reference Merola A, Sawyer RP, Artusi CA, Suri R, Berndt Z, Lopez-Castellanos JR, et al. Orthostatic hypotension in Parkinson disease: impact on health care utilization. Parkinsonism Relat Disord. 2018;47:45–9.CrossRefPubMed Merola A, Sawyer RP, Artusi CA, Suri R, Berndt Z, Lopez-Castellanos JR, et al. Orthostatic hypotension in Parkinson disease: impact on health care utilization. Parkinsonism Relat Disord. 2018;47:45–9.CrossRefPubMed
81.
go back to reference Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. Curr Treat Options Neurol. 2018;20:54.CrossRefPubMed Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. Curr Treat Options Neurol. 2018;20:54.CrossRefPubMed
82.
go back to reference Doi H, Sakakibara R, Sato M, Masaka T, Kishi M, Tateno A, et al. Plasma levodopa peak delay and impaired gastric emptying in Parkinson’s disease. J Neurol Sci. 2012;319:86–8.CrossRefPubMed Doi H, Sakakibara R, Sato M, Masaka T, Kishi M, Tateno A, et al. Plasma levodopa peak delay and impaired gastric emptying in Parkinson’s disease. J Neurol Sci. 2012;319:86–8.CrossRefPubMed
83.
go back to reference Muller T, Erdmann C, Bremen D, Schmidt WE, Muhlack S, Woitalla D, et al. Impact of gastric emptying on levodopa pharmacokinetics in Parkinson disease patients. Clin Neuropharmacol. 2006;29:61–7.CrossRefPubMed Muller T, Erdmann C, Bremen D, Schmidt WE, Muhlack S, Woitalla D, et al. Impact of gastric emptying on levodopa pharmacokinetics in Parkinson disease patients. Clin Neuropharmacol. 2006;29:61–7.CrossRefPubMed
84.
go back to reference Heetun ZS, Quigley EMM. Gastroparesis and Parkinson’s disease: a systematic review. Parkinsonism Relat Disord. 2012;18:433–40.CrossRefPubMed Heetun ZS, Quigley EMM. Gastroparesis and Parkinson’s disease: a systematic review. Parkinsonism Relat Disord. 2012;18:433–40.CrossRefPubMed
85.
go back to reference Greene RJ, Hall AD, Hider RC. The interaction of orally administered iron with levodopa and methyldopa therapy. J Pharm Pharmacol. 1990;42:502–4.CrossRefPubMed Greene RJ, Hall AD, Hider RC. The interaction of orally administered iron with levodopa and methyldopa therapy. J Pharm Pharmacol. 1990;42:502–4.CrossRefPubMed
86.
go back to reference Campbell NR, Hasinoff B. Ferrous sulfate reduces levodopa bioavailability: chelation as a possible mechanism. Clin Pharmacol Ther. 1989;45:220–5.CrossRefPubMed Campbell NR, Hasinoff B. Ferrous sulfate reduces levodopa bioavailability: chelation as a possible mechanism. Clin Pharmacol Ther. 1989;45:220–5.CrossRefPubMed
87.
go back to reference Deleu D, Jacob P, Chand P, Sarre S, Colwell A. Effects of caffeine on levodopa pharmacokinetics and pharmacodynamics in Parkinson disease. Neurology. 2006;67:897–9.CrossRefPubMed Deleu D, Jacob P, Chand P, Sarre S, Colwell A. Effects of caffeine on levodopa pharmacokinetics and pharmacodynamics in Parkinson disease. Neurology. 2006;67:897–9.CrossRefPubMed
88.
go back to reference Robertson DR, Higginson I, Macklin BS, Renwick AG, Waller DG, George CF. The influence of protein containing meals on the pharmacokinetics of levodopa in healthy volunteers. Br J Clin Pharmacol. 1991;31:413–7.CrossRefPubMedPubMedCentral Robertson DR, Higginson I, Macklin BS, Renwick AG, Waller DG, George CF. The influence of protein containing meals on the pharmacokinetics of levodopa in healthy volunteers. Br J Clin Pharmacol. 1991;31:413–7.CrossRefPubMedPubMedCentral
89.
go back to reference Simon N, Gantcheva R, Bruguerolle B, Viallet F. The effects of a normal protein diet on levodopa plasma kinetics in advanced Parkinson’s disease. Parkinsonism Relat Disord. 2004;10:137–42.CrossRefPubMed Simon N, Gantcheva R, Bruguerolle B, Viallet F. The effects of a normal protein diet on levodopa plasma kinetics in advanced Parkinson’s disease. Parkinsonism Relat Disord. 2004;10:137–42.CrossRefPubMed
91.
go back to reference Jenner P. Pharmacology of dopamine agonists in the treatment of Parkinson’s disease. Neurology. 2002;58(4 Suppl. 1):S1–8.CrossRefPubMed Jenner P. Pharmacology of dopamine agonists in the treatment of Parkinson’s disease. Neurology. 2002;58(4 Suppl. 1):S1–8.CrossRefPubMed
92.
go back to reference Piercey MF. Pharmacology of pramipexole, a dopamine D3-preferring agonist useful in treating Parkinson’s disease. Clin Neuropharmacol. 1998;21:141–51.PubMed Piercey MF. Pharmacology of pramipexole, a dopamine D3-preferring agonist useful in treating Parkinson’s disease. Clin Neuropharmacol. 1998;21:141–51.PubMed
93.
go back to reference 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;29:1452–67.CrossRefPubMed 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;29:1452–67.CrossRefPubMed
94.
go back to reference Tulloch IF. Pharmacologic profile of ropinirole: a nonergoline dopamine agonist. Neurology. 1997;49(1 Suppl. 1):S58–62.CrossRefPubMed Tulloch IF. Pharmacologic profile of ropinirole: a nonergoline dopamine agonist. Neurology. 1997;49(1 Suppl. 1):S58–62.CrossRefPubMed
95.
go back to reference Hagell P, Odin P. Apomorphine in the treatment of Parkinson’s disease. J Neurosci Nurs. 2001;33(21–34):37–8. Hagell P, Odin P. Apomorphine in the treatment of Parkinson’s disease. J Neurosci Nurs. 2001;33(21–34):37–8.
96.
go back to reference Stacy M, Silver D. Apomorphine for the acute treatment of “off” episodes in Parkinson’s disease. Parkinsonism Relat Disord. 2008;14:85–92.CrossRefPubMed Stacy M, Silver D. Apomorphine for the acute treatment of “off” episodes in Parkinson’s disease. Parkinsonism Relat Disord. 2008;14:85–92.CrossRefPubMed
97.
go back to reference 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;33:147–61.CrossRefPubMed 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;33:147–61.CrossRefPubMed
98.
go back to reference Voon V, Hassan K, Zurowski M, Duff-Canning S, de Souza M, Fox S, et al. Prospective prevalence of pathologic gambling and medication association in Parkinson disease. Neurology. 2006;66:1750–2.CrossRefPubMed Voon V, Hassan K, Zurowski M, Duff-Canning S, de Souza M, Fox S, et al. Prospective prevalence of pathologic gambling and medication association in Parkinson disease. Neurology. 2006;66:1750–2.CrossRefPubMed
99.
go back to reference Voon V, Hassan K, Zurowski M, de Souza M, Thomsen T, Fox S, et al. Prevalence of repetitive and reward-seeking behaviors in Parkinson disease. Neurology. 2006;67:1254–7.CrossRefPubMed Voon V, Hassan K, Zurowski M, de Souza M, Thomsen T, Fox S, et al. Prevalence of repetitive and reward-seeking behaviors in Parkinson disease. Neurology. 2006;67:1254–7.CrossRefPubMed
100.
go back to reference Weintraub D, Siderowf AD, Potenza MN, Goveas J, Morales KH, Duda JE, et al. Association of dopamine agonist use with impulse control disorders in Parkinson disease. Arch Neurol. 2006;63:969–73.CrossRefPubMedPubMedCentral Weintraub D, Siderowf AD, Potenza MN, Goveas J, Morales KH, Duda JE, et al. Association of dopamine agonist use with impulse control disorders in Parkinson disease. Arch Neurol. 2006;63:969–73.CrossRefPubMedPubMedCentral
101.
go back to reference Bienfait KL, Menza M, Mark MH, Dobkin RD. Impulsive smoking in a patient with Parkinson’s disease treated with dopamine agonists. J Clin Neurosci. 2010;17:539–40.CrossRefPubMedPubMedCentral Bienfait KL, Menza M, Mark MH, Dobkin RD. Impulsive smoking in a patient with Parkinson’s disease treated with dopamine agonists. J Clin Neurosci. 2010;17:539–40.CrossRefPubMedPubMedCentral
102.
go back to reference Ceravolo R, Frosini D, Rossi C, Bonuccelli U. Impulse control disorders in Parkinson’s disease: definition, epidemiology, risk factors, neurobiology and management. Parkinsonism Relat Disord. 2009;15(Suppl. 4):S111–5.CrossRefPubMed Ceravolo R, Frosini D, Rossi C, Bonuccelli U. Impulse control disorders in Parkinson’s disease: definition, epidemiology, risk factors, neurobiology and management. Parkinsonism Relat Disord. 2009;15(Suppl. 4):S111–5.CrossRefPubMed
103.
go back to reference Miyasaki JM. Evidence-based initiation of dopaminergic therapy in Parkinson’s disease. J Neurol. 2010;257(Suppl. 2):S309–13.CrossRefPubMed Miyasaki JM. Evidence-based initiation of dopaminergic therapy in Parkinson’s disease. J Neurol. 2010;257(Suppl. 2):S309–13.CrossRefPubMed
104.
go back to reference Antonini A, Chaudhuri KR, Boroojerdi B, Asgharnejad M, Bauer L, Grieger F, et al. Impulse control disorder related behaviours during long-term rotigotine treatment: a post hoc analysis. Eur J Neurol. 2016;23:1556–65.CrossRefPubMedPubMedCentral Antonini A, Chaudhuri KR, Boroojerdi B, Asgharnejad M, Bauer L, Grieger F, et al. Impulse control disorder related behaviours during long-term rotigotine treatment: a post hoc analysis. Eur J Neurol. 2016;23:1556–65.CrossRefPubMedPubMedCentral
105.
go back to reference Moller JC, Eggert KM, Unger M, Odin P, Chaudhuri KR, Oertel WH. Clinical risk-benefit assessment of dopamine agonists. Eur J Neurol. 2008;15(Suppl. 2):15–23.CrossRefPubMed Moller JC, Eggert KM, Unger M, Odin P, Chaudhuri KR, Oertel WH. Clinical risk-benefit assessment of dopamine agonists. Eur J Neurol. 2008;15(Suppl. 2):15–23.CrossRefPubMed
106.
go back to reference Paus S, Brecht HM, Koster J, Seeger G, Klockgether T, Wullner U. Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s disease. Mov Disord. 2003;18:659–67.CrossRefPubMed Paus S, Brecht HM, Koster J, Seeger G, Klockgether T, Wullner U. Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s disease. Mov Disord. 2003;18:659–67.CrossRefPubMed
107.
go back to reference 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;26:439–44.CrossRefPubMed 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;26:439–44.CrossRefPubMed
108.
go back to reference Abbott RD, Petrovitch H, White LR, Masaki KH, Tanner CM, Curb JD, et al. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology. 2001;57:456–62.CrossRefPubMed Abbott RD, Petrovitch H, White LR, Masaki KH, Tanner CM, Curb JD, et al. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology. 2001;57:456–62.CrossRefPubMed
109.
go back to reference Gao X, Chen H, Schwarzschild MA, Ascherio A. A prospective study of bowel movement frequency and risk of Parkinson’s disease. Am J Epidemiol. 2011;174:546–51.CrossRefPubMedPubMedCentral Gao X, Chen H, Schwarzschild MA, Ascherio A. A prospective study of bowel movement frequency and risk of Parkinson’s disease. Am J Epidemiol. 2011;174:546–51.CrossRefPubMedPubMedCentral
110.
go back to reference Zhou C-Q, Zhang J-W, Wang M, Peng G-G. Meta-analysis of the efficacy and safety of long-acting non-ergot dopamine agonists in Parkinson’s disease. J Clin Neurosci. 2014;21:1094–101.CrossRefPubMed Zhou C-Q, Zhang J-W, Wang M, Peng G-G. Meta-analysis of the efficacy and safety of long-acting non-ergot dopamine agonists in Parkinson’s disease. J Clin Neurosci. 2014;21:1094–101.CrossRefPubMed
111.
go back to reference Tan EK, Jankovic J. Choosing dopamine agonists in Parkinson’s disease. Clin Neuropharmacol. 2001;24:247–53.CrossRefPubMed Tan EK, Jankovic J. Choosing dopamine agonists in Parkinson’s disease. Clin Neuropharmacol. 2001;24:247–53.CrossRefPubMed
112.
go back to reference Chaudhuri KR, Pal S, Brefel-Courbon C. ‘Sleep attacks’ or “unintended sleep episodes” occur with dopamine agonists: is this a class effect? Drug Saf. 2002;25:473–83.CrossRefPubMed Chaudhuri KR, Pal S, Brefel-Courbon C. ‘Sleep attacks’ or “unintended sleep episodes” occur with dopamine agonists: is this a class effect? Drug Saf. 2002;25:473–83.CrossRefPubMed
113.
go back to reference Razmy A, Lang AE, Shapiro CM. Predictors of impaired daytime sleep and wakefulness in patients with Parkinson disease treated with older (ergot) vs newer (nonergot) dopamine agonists. Arch. Neurol. 2004;61:97–102.CrossRefPubMed Razmy A, Lang AE, Shapiro CM. Predictors of impaired daytime sleep and wakefulness in patients with Parkinson disease treated with older (ergot) vs newer (nonergot) dopamine agonists. Arch. Neurol. 2004;61:97–102.CrossRefPubMed
114.
go back to reference Sprenger FS, Seppi K, Poewe W. Drug safety evaluation of rotigotine. Expert Opin Drug Saf. 2012;11:503–12.CrossRefPubMed Sprenger FS, Seppi K, Poewe W. Drug safety evaluation of rotigotine. Expert Opin Drug Saf. 2012;11:503–12.CrossRefPubMed
115.
go back to reference Ishiguro N, Saito A, Yokoyama K, Morikawa M, Igarashi T, Tamai I. Transport of the dopamine D2 agonist pramipexole by rat organic cation transporters OCT1 and OCT2 in kidney. Drug Metab Dispos. 2005;33:495–9.CrossRefPubMed Ishiguro N, Saito A, Yokoyama K, Morikawa M, Igarashi T, Tamai I. Transport of the dopamine D2 agonist pramipexole by rat organic cation transporters OCT1 and OCT2 in kidney. Drug Metab Dispos. 2005;33:495–9.CrossRefPubMed
116.
go back to reference Knop J, Hoier E, Ebner T, Fromm MF, Muller F. Renal tubular secretion of pramipexole. Eur J Pharm Sci. 2015;79:73–8.CrossRefPubMed Knop J, Hoier E, Ebner T, Fromm MF, Muller F. Renal tubular secretion of pramipexole. Eur J Pharm Sci. 2015;79:73–8.CrossRefPubMed
117.
go back to reference Wu MJ, Ing TS, Soung LS, Daugirdas JT, Hano JE, Gandhi VC. Amantadine hydrochloride pharmacokinetics in patients with impaired renal function. Clin Nephrol. 1982;17:19–23.PubMed Wu MJ, Ing TS, Soung LS, Daugirdas JT, Hano JE, Gandhi VC. Amantadine hydrochloride pharmacokinetics in patients with impaired renal function. Clin Nephrol. 1982;17:19–23.PubMed
118.
go back to reference Kaye CM, Nicholls B. Clinical pharmacokinetics of ropinirole. Clin Pharmacokinet. 2000;39:243–54.CrossRefPubMed Kaye CM, Nicholls B. Clinical pharmacokinetics of ropinirole. Clin Pharmacokinet. 2000;39:243–54.CrossRefPubMed
119.
go back to reference Bloomer JC, Clarke SE, Chenery RJ. In vitro identification of the P450 enzymes responsible for the metabolism of ropinirole. Drug Metab Dispos. 1997;25:840–4.PubMed Bloomer JC, Clarke SE, Chenery RJ. In vitro identification of the P450 enzymes responsible for the metabolism of ropinirole. Drug Metab Dispos. 1997;25:840–4.PubMed
120.
go back to reference Girndt M, Trocchi P, Scheidt-Nave C, Markau S, Stang A. The prevalence of renal failure: results from the German Health Interview and Examination Survey for Adults, 2008–2011 (DEGS1). Dtsch Arztebl Int. 2016;113:85–91.PubMedPubMedCentral Girndt M, Trocchi P, Scheidt-Nave C, Markau S, Stang A. The prevalence of renal failure: results from the German Health Interview and Examination Survey for Adults, 2008–2011 (DEGS1). Dtsch Arztebl Int. 2016;113:85–91.PubMedPubMedCentral
121.
go back to reference Schaeffner ES, Ebert N, Delanaye P, Frei U, Gaedeke J, Jakob O, et al. Two novel equations to estimate kidney function in persons aged 70 years or older. Ann Intern Med. 2012;157:471–81.CrossRefPubMed Schaeffner ES, Ebert N, Delanaye P, Frei U, Gaedeke J, Jakob O, et al. Two novel equations to estimate kidney function in persons aged 70 years or older. Ann Intern Med. 2012;157:471–81.CrossRefPubMed
122.
go back to reference Pellecchia MT, Vitale C, Sabatini M, Longo K, Amboni M, Bonavita V, et al. Ropinirole as a treatment of restless legs syndrome in patients on chronic hemodialysis: an open randomized crossover trial versus levodopa sustained release. Clin Neuropharmacol. 2004;27:178–81.CrossRefPubMed Pellecchia MT, Vitale C, Sabatini M, Longo K, Amboni M, Bonavita V, et al. Ropinirole as a treatment of restless legs syndrome in patients on chronic hemodialysis: an open randomized crossover trial versus levodopa sustained release. Clin Neuropharmacol. 2004;27:178–81.CrossRefPubMed
123.
go back to reference Cawello W, Ahrweiler S, Sulowicz W, Szymczakiewicz-Multanowska A, Braun M. Single dose pharmacokinetics of the transdermal rotigotine patch in patients with impaired renal function. Br J Clin Pharmacol. 2012;73:46–54.CrossRefPubMedPubMedCentral Cawello W, Ahrweiler S, Sulowicz W, Szymczakiewicz-Multanowska A, Braun M. Single dose pharmacokinetics of the transdermal rotigotine patch in patients with impaired renal function. Br J Clin Pharmacol. 2012;73:46–54.CrossRefPubMedPubMedCentral
124.
go back to reference Cieslak KP, Baur O, Verheij J, Bennink RJ, van Gulik TM. Liver function declines with increased age. HPB (Oxford). 2016;18:691–6.CrossRef Cieslak KP, Baur O, Verheij J, Bennink RJ, van Gulik TM. Liver function declines with increased age. HPB (Oxford). 2016;18:691–6.CrossRef
125.
go back to reference Cawello W, Fichtner A, Boekens H, Braun M. Influence of hepatic impairment on the pharmacokinetics of the dopamine agonist rotigotine. Eur J Drug Metab Pharmacokinet. 2014;39:155–63.CrossRefPubMed Cawello W, Fichtner A, Boekens H, Braun M. Influence of hepatic impairment on the pharmacokinetics of the dopamine agonist rotigotine. Eur J Drug Metab Pharmacokinet. 2014;39:155–63.CrossRefPubMed
126.
go back to reference Navacerrada F, Gonzalez-Alonso MR, Alonso-Navarro H, Pilo-de-la-Fuente B, Plaza-Nieto JF, Jimenez-Jimenez FJ. Liver toxicity possibly related with ropinirole use in the treatment of restless legs syndrome. Eur J Neurol. 2011;18:e65.CrossRefPubMed Navacerrada F, Gonzalez-Alonso MR, Alonso-Navarro H, Pilo-de-la-Fuente B, Plaza-Nieto JF, Jimenez-Jimenez FJ. Liver toxicity possibly related with ropinirole use in the treatment of restless legs syndrome. Eur J Neurol. 2011;18:e65.CrossRefPubMed
127.
go back to reference Dewey RBJ, Hutton JT, LeWitt PA, Factor SA. A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch Neurol. 2001;58:1385–92.CrossRefPubMed Dewey RBJ, Hutton JT, LeWitt PA, Factor SA. A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch Neurol. 2001;58:1385–92.CrossRefPubMed
128.
go back to reference Stibe CM, Lees AJ, Kempster PA, Stern GM. Subcutaneous apomorphine in parkinsonian on-off oscillations. Lancet. 1988;1:403–6.CrossRefPubMed Stibe CM, Lees AJ, Kempster PA, Stern GM. Subcutaneous apomorphine in parkinsonian on-off oscillations. Lancet. 1988;1:403–6.CrossRefPubMed
129.
go back to reference Humphrey SJ, Turman CN, Curry JT, Wheeler GJ. Cardiovascular and electrocardiographic effects of the dopamine receptor agonists ropinirole, apomorphine, and PNU-142774E in conscious beagle dogs. J Cardiovasc Pharmacol. 2006;47:337–47.PubMed Humphrey SJ, Turman CN, Curry JT, Wheeler GJ. Cardiovascular and electrocardiographic effects of the dopamine receptor agonists ropinirole, apomorphine, and PNU-142774E in conscious beagle dogs. J Cardiovasc Pharmacol. 2006;47:337–47.PubMed
130.
go back to reference Stocchi F, De Pandis MF, Delfino FA, Anselmo T, Frongillo D. Transient atrial fibrillation after subcutaneous apomorphine bolus. Mov Disord. 1996;11:584–5.CrossRefPubMed Stocchi F, De Pandis MF, Delfino FA, Anselmo T, Frongillo D. Transient atrial fibrillation after subcutaneous apomorphine bolus. Mov Disord. 1996;11:584–5.CrossRefPubMed
131.
go back to reference Kaminioti AN, Nikitas GT, Terlis AK, Manolis AG, Thomaides T, Panousopoulou AN. Ventricular bigeminy after subcutaneous administration of apomorphine in a patient with refractory Parkinson’s disease: a case report. J Mov Disord. 2013;6:9–12.CrossRefPubMedPubMedCentral Kaminioti AN, Nikitas GT, Terlis AK, Manolis AG, Thomaides T, Panousopoulou AN. Ventricular bigeminy after subcutaneous administration of apomorphine in a patient with refractory Parkinson’s disease: a case report. J Mov Disord. 2013;6:9–12.CrossRefPubMedPubMedCentral
132.
go back to reference Sartori M, Pratt CM, Young JB. Torsade de Pointe. Malignant cardiac arrhythmia induced by amantadine poisoning. Am J Med. 1984;77:388–91.CrossRefPubMed Sartori M, Pratt CM, Young JB. Torsade de Pointe. Malignant cardiac arrhythmia induced by amantadine poisoning. Am J Med. 1984;77:388–91.CrossRefPubMed
134.
go back to reference De Ponti F, Poluzzi E, Cavalli A, Recanatini M, Montanaro N. Safety of non-antiarrhythmic drugs that prolong the QT interval or induce torsade de pointes: an overview. Drug Saf. 2002;25:263–86.CrossRefPubMed De Ponti F, Poluzzi E, Cavalli A, Recanatini M, Montanaro N. Safety of non-antiarrhythmic drugs that prolong the QT interval or induce torsade de pointes: an overview. Drug Saf. 2002;25:263–86.CrossRefPubMed
135.
go back to reference Watanabe Y, Nakamura Y, Cao X, Ohara H, Yamazaki Y, Murayama N, et al. Intravenous administration of apomorphine does NOT induce long QT syndrome: experimental evidence from in vivo canine models. Basic Clin Pharmacol Toxicol. 2015;116:468–75.CrossRefPubMed Watanabe Y, Nakamura Y, Cao X, Ohara H, Yamazaki Y, Murayama N, et al. Intravenous administration of apomorphine does NOT induce long QT syndrome: experimental evidence from in vivo canine models. Basic Clin Pharmacol Toxicol. 2015;116:468–75.CrossRefPubMed
136.
go back to reference Halvorsen KA, Martensen-Larsen O. Apomorphine revived: fortified, prolonged, and improved therapeutical effect. Int J Addict. 1978;13:475–84.CrossRefPubMed Halvorsen KA, Martensen-Larsen O. Apomorphine revived: fortified, prolonged, and improved therapeutical effect. Int J Addict. 1978;13:475–84.CrossRefPubMed
137.
138.
go back to reference Rausten DS, Ochs MA. Apomorphine-naloxone controlled rapid emesis. J Am Coll Emerg Phys. 1973;2:44–5.CrossRef Rausten DS, Ochs MA. Apomorphine-naloxone controlled rapid emesis. J Am Coll Emerg Phys. 1973;2:44–5.CrossRef
139.
go back to reference Björklund A, Dunnett SB. Dopamine neuron systems in the brain: an update. Trends Neurosci. 2007;30:194–202.CrossRefPubMed Björklund A, Dunnett SB. Dopamine neuron systems in the brain: an update. Trends Neurosci. 2007;30:194–202.CrossRefPubMed
140.
go back to reference Muller T. Catechol-O-methyltransferase inhibitors in Parkinson’s disease. Drugs. 2015;75:157–74.CrossRefPubMed Muller T. Catechol-O-methyltransferase inhibitors in Parkinson’s disease. Drugs. 2015;75:157–74.CrossRefPubMed
141.
go back to reference Axelrod J, Tomchick R. Enzymatic O-methylation of epinephrine and other catechols. J Biol Chem. 1958;233:702–5.PubMed Axelrod J, Tomchick R. Enzymatic O-methylation of epinephrine and other catechols. J Biol Chem. 1958;233:702–5.PubMed
142.
go back to reference Guldberg HC, Marsden CA. Catechol-O-methyl transferase: pharmacological aspects and physiological role. Pharmacol Rev. 1975;27:135–206.PubMed Guldberg HC, Marsden CA. Catechol-O-methyl transferase: pharmacological aspects and physiological role. Pharmacol Rev. 1975;27:135–206.PubMed
143.
go back to reference Napolitano A, Del Dotto P, Petrozzi L, Dell’Agnello G, Bellini G, Gambaccini G, et al. Pharmacokinetics and pharmacodynamics of l-Dopa after acute and 6-week tolcapone administration in patients with Parkinson’s disease. Clin Neuropharmacol. 1999;22:24–9.CrossRefPubMed Napolitano A, Del Dotto P, Petrozzi L, Dell’Agnello G, Bellini G, Gambaccini G, et al. Pharmacokinetics and pharmacodynamics of l-Dopa after acute and 6-week tolcapone administration in patients with Parkinson’s disease. Clin Neuropharmacol. 1999;22:24–9.CrossRefPubMed
144.
go back to reference Ruottinen HM, Rinne UK. A double-blind pharmacokinetic and clinical dose-response study of entacapone as an adjuvant to levodopa therapy in advanced Parkinson’s disease. Clin Neuropharmacol. 1996;19:283–96.CrossRefPubMed Ruottinen HM, Rinne UK. A double-blind pharmacokinetic and clinical dose-response study of entacapone as an adjuvant to levodopa therapy in advanced Parkinson’s disease. Clin Neuropharmacol. 1996;19:283–96.CrossRefPubMed
145.
go back to reference Ferreira JJ, Lees A, Rocha J-F, Poewe W, Rascol O, Soares-da-Silva P. Opicapone as an adjunct to levodopa in patients with Parkinson’s disease and end-of-dose motor fluctuations: a randomised, double-blind, controlled trial. Lancet Neurol. 2016;15:154–65.CrossRef Ferreira JJ, Lees A, Rocha J-F, Poewe W, Rascol O, Soares-da-Silva P. Opicapone as an adjunct to levodopa in patients with Parkinson’s disease and end-of-dose motor fluctuations: a randomised, double-blind, controlled trial. Lancet Neurol. 2016;15:154–65.CrossRef
146.
go back to reference Factor SA, Molho ES, Feustel PJ, Brown DL, Evans SM. Long-term comparative experience with tolcapone and entacapone in advanced Parkinson’s disease. Clin Neuropharmacol. 2001;24:295–9.CrossRefPubMed Factor SA, Molho ES, Feustel PJ, Brown DL, Evans SM. Long-term comparative experience with tolcapone and entacapone in advanced Parkinson’s disease. Clin Neuropharmacol. 2001;24:295–9.CrossRefPubMed
147.
go back to reference Rocha J-F, Almeida L, Falcao A, Palma PN, Loureiro AI, Pinto R, et al. Opicapone: a short lived and very long acting novel catechol-O-methyltransferase inhibitor following multiple dose administration in healthy subjects. Br J Clin Pharmacol. 2013;76:763–75.CrossRefPubMedPubMedCentral Rocha J-F, Almeida L, Falcao A, Palma PN, Loureiro AI, Pinto R, et al. Opicapone: a short lived and very long acting novel catechol-O-methyltransferase inhibitor following multiple dose administration in healthy subjects. Br J Clin Pharmacol. 2013;76:763–75.CrossRefPubMedPubMedCentral
148.
go back to reference Davis TL, Roznoski M, Burns RS. Effects of tolcapone in Parkinson’s patients taking l-dihydroxyphenylalanine/carbidopa and selegiline. Mov Disord. 1995;10:349–51.CrossRefPubMed Davis TL, Roznoski M, Burns RS. Effects of tolcapone in Parkinson’s patients taking l-dihydroxyphenylalanine/carbidopa and selegiline. Mov Disord. 1995;10:349–51.CrossRefPubMed
149.
go back to reference Kuoppamaki M, Leinonen M, Poewe W. Efficacy and safety of entacapone in levodopa/carbidopa versus levodopa/benserazide treated Parkinson’s disease patients with wearing-off. J Neural Transm (Vienna). 2015;122:1709–14.CrossRef Kuoppamaki M, Leinonen M, Poewe W. Efficacy and safety of entacapone in levodopa/carbidopa versus levodopa/benserazide treated Parkinson’s disease patients with wearing-off. J Neural Transm (Vienna). 2015;122:1709–14.CrossRef
150.
go back to reference Rodrigues FB, Ferreira JJ. Opicapone for the treatment of Parkinson’s disease. Expert Opin Pharmacother. 2017;18:445–53.CrossRefPubMed Rodrigues FB, Ferreira JJ. Opicapone for the treatment of Parkinson’s disease. Expert Opin Pharmacother. 2017;18:445–53.CrossRefPubMed
151.
go back to reference Antonini A, Abbruzzese G, Barone P, Bonuccelli U, Lopiano L, Onofrj M, et al. COMT inhibition with tolcapone in the treatment algorithm of patients with Parkinson’s disease (PD): relevance for motor and non-motor features. Neuropsychiatr Dis Treat. 2008;4:1–9.CrossRefPubMedPubMedCentral Antonini A, Abbruzzese G, Barone P, Bonuccelli U, Lopiano L, Onofrj M, et al. COMT inhibition with tolcapone in the treatment algorithm of patients with Parkinson’s disease (PD): relevance for motor and non-motor features. Neuropsychiatr Dis Treat. 2008;4:1–9.CrossRefPubMedPubMedCentral
152.
go back to reference Suchowersky O, Bailey P, Pourcher E, Bulger L, Facciponte G. Comparison of two dosages of tolcapone added to levodopa in nonfluctuating patients with PD. Clin Neuropharmacol. 2001;24:214–20.CrossRefPubMed Suchowersky O, Bailey P, Pourcher E, Bulger L, Facciponte G. Comparison of two dosages of tolcapone added to levodopa in nonfluctuating patients with PD. Clin Neuropharmacol. 2001;24:214–20.CrossRefPubMed
153.
go back to reference Olanow CW, Watkins PB. Tolcapone: an efficacy and safety review (2007). Clin Neuropharmacol. 2007;30:287–94.CrossRefPubMed Olanow CW, Watkins PB. Tolcapone: an efficacy and safety review (2007). Clin Neuropharmacol. 2007;30:287–94.CrossRefPubMed
154.
go back to reference Lees AJ, Ratziu V, Tolosa E, Oertel WH. Safety and tolerability of adjunctive tolcapone treatment in patients with early Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2007;78:944–8.CrossRefPubMed Lees AJ, Ratziu V, Tolosa E, Oertel WH. Safety and tolerability of adjunctive tolcapone treatment in patients with early Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2007;78:944–8.CrossRefPubMed
155.
go back to reference Unger MM, Reese JP, Oertel WH, Eggert KM. Real-life evaluations of compliance with mandatory drug safety monitoring exemplified with tolcapone in Parkinson’s disease. Eur Neurol. 2008;60:122–6.CrossRefPubMed Unger MM, Reese JP, Oertel WH, Eggert KM. Real-life evaluations of compliance with mandatory drug safety monitoring exemplified with tolcapone in Parkinson’s disease. Eur Neurol. 2008;60:122–6.CrossRefPubMed
156.
go back to reference Heranval A, Lefaucheur R, Fetter D, Rouille A, Le Goff F, Maltete D. Drugs with potential cardiac adverse effects: retrospective study in a large cohort of parkinsonian patients. Rev Neurol (Paris). 2016;172:318–23.CrossRef Heranval A, Lefaucheur R, Fetter D, Rouille A, Le Goff F, Maltete D. Drugs with potential cardiac adverse effects: retrospective study in a large cohort of parkinsonian patients. Rev Neurol (Paris). 2016;172:318–23.CrossRef
157.
go back to reference Van Booven D, Marsh S, McLeod H, Carrillo MW, Sangkuhl K, Klein TE, et al. Cytochrome P450 2C9-CYP2C9. Pharmacogenet Genomics. 2010;20:277–81.PubMedPubMedCentral Van Booven D, Marsh S, McLeod H, Carrillo MW, Sangkuhl K, Klein TE, et al. Cytochrome P450 2C9-CYP2C9. Pharmacogenet Genomics. 2010;20:277–81.PubMedPubMedCentral
158.
go back to reference Dingemanse J, Meyerhoff C, Schadrack J. Effect of the catechol-O-methyltransferase inhibitor entacapone on the steady-state pharmacokinetics and pharmacodynamics of warfarin. Br J Clin Pharmacol. 2002;53:485–91.CrossRefPubMedPubMedCentral Dingemanse J, Meyerhoff C, Schadrack J. Effect of the catechol-O-methyltransferase inhibitor entacapone on the steady-state pharmacokinetics and pharmacodynamics of warfarin. Br J Clin Pharmacol. 2002;53:485–91.CrossRefPubMedPubMedCentral
159.
go back to reference European Medicines Agency. Ongentys: EMA assessment report. London, UK: European Medicines Agency; 2016. p. 1–140. European Medicines Agency. Ongentys: EMA assessment report. London, UK: European Medicines Agency; 2016. p. 1–140.
160.
go back to reference Shitara Y, Maeda K, Ikejiri K, Yoshida K, Horie T, Sugiyama Y. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in hepatic clearance and intestinal absorption. Biopharm Drug Dispos. 2013;34:45–78.CrossRefPubMed Shitara Y, Maeda K, Ikejiri K, Yoshida K, Horie T, Sugiyama Y. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in hepatic clearance and intestinal absorption. Biopharm Drug Dispos. 2013;34:45–78.CrossRefPubMed
161.
go back to reference Yu J, Zhou Z, Tay-Sontheimer J, Levy RH, Ragueneau-Majlessi I. Intestinal drug interactions mediated by OATPs: a systematic review of preclinical and clinical findings. J Pharm Sci. 2017;106:2312–25.CrossRefPubMed Yu J, Zhou Z, Tay-Sontheimer J, Levy RH, Ragueneau-Majlessi I. Intestinal drug interactions mediated by OATPs: a systematic review of preclinical and clinical findings. J Pharm Sci. 2017;106:2312–25.CrossRefPubMed
162.
go back to reference Kalgutkar AS, Dalvie DK, Castagnoli NJ, Taylor TJ. Interactions of nitrogen-containing xenobiotics with monoamine oxidase (MAO) isozymes A and B: SAR studies on MAO substrates and inhibitors. Chem Res Toxicol. 2001;14:1139–62.CrossRefPubMed Kalgutkar AS, Dalvie DK, Castagnoli NJ, Taylor TJ. Interactions of nitrogen-containing xenobiotics with monoamine oxidase (MAO) isozymes A and B: SAR studies on MAO substrates and inhibitors. Chem Res Toxicol. 2001;14:1139–62.CrossRefPubMed
163.
go back to reference Fernandez HH, Chen JJ. Monoamine oxidase-B inhibition in the treatment of Parkinson’s disease. Pharmacotherapy. 2007;27:174S–85S.CrossRefPubMed Fernandez HH, Chen JJ. Monoamine oxidase-B inhibition in the treatment of Parkinson’s disease. Pharmacotherapy. 2007;27:174S–85S.CrossRefPubMed
164.
go back to reference Chang Y, Wang L-B, Li D, Lei K, Liu S-Y. Efficacy of rasagiline for the treatment of Parkinson’s disease: an updated meta-analysis. Ann Med. 2017;49:421–34.CrossRefPubMed Chang Y, Wang L-B, Li D, Lei K, Liu S-Y. Efficacy of rasagiline for the treatment of Parkinson’s disease: an updated meta-analysis. Ann Med. 2017;49:421–34.CrossRefPubMed
165.
go back to reference Dashtipour K, Chen JJ, Kani C, Bahjri K, Ghamsary M. Clinical outcomes in patients with Parkinson’s disease treated with a monoamine oxidase type-B inhibitor: a cross-sectional, cohort study. Pharmacotherapy. 2015;35:681–6.CrossRefPubMedPubMedCentral Dashtipour K, Chen JJ, Kani C, Bahjri K, Ghamsary M. Clinical outcomes in patients with Parkinson’s disease treated with a monoamine oxidase type-B inhibitor: a cross-sectional, cohort study. Pharmacotherapy. 2015;35:681–6.CrossRefPubMedPubMedCentral
166.
go back to reference Dezsi L, Vecsei L. Monoamine oxidase B inhibitors in Parkinson’s disease. CNS Neurol Disord Drug Targets. 2017;16:425–39.CrossRefPubMed Dezsi L, Vecsei L. Monoamine oxidase B inhibitors in Parkinson’s disease. CNS Neurol Disord Drug Targets. 2017;16:425–39.CrossRefPubMed
167.
go back to reference Riederer P, Lachenmayer L. Selegiline’s neuroprotective capacity revisited. J Neural Transm (Vienna). 2003;110:1273–8.CrossRef Riederer P, Lachenmayer L. Selegiline’s neuroprotective capacity revisited. J Neural Transm (Vienna). 2003;110:1273–8.CrossRef
168.
go back to reference Olanow CW, Rascol O, Hauser R, Feigin PD, Jankovic J, Lang A, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med. 2009;361:1268–78.CrossRefPubMed Olanow CW, Rascol O, Hauser R, Feigin PD, Jankovic J, Lang A, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med. 2009;361:1268–78.CrossRefPubMed
169.
go back to reference Sadeghian M, Mullali G, Pocock JM, Piers T, Roach A, Smith KJ. Neuroprotection by safinamide in the 6-hydroxydopamine model of Parkinson’s disease. Neuropathol Appl Neurobiol. 2016;42:423–35.CrossRefPubMed Sadeghian M, Mullali G, Pocock JM, Piers T, Roach A, Smith KJ. Neuroprotection by safinamide in the 6-hydroxydopamine model of Parkinson’s disease. Neuropathol Appl Neurobiol. 2016;42:423–35.CrossRefPubMed
170.
go back to reference Hauser RA, Abler V, Eyal E, Eliaz RE. Efficacy of rasagiline in early Parkinson’s disease: a meta-analysis of data from the TEMPO and ADAGIO studies. Int J Neurosci. 2016;126:942–6.CrossRefPubMed Hauser RA, Abler V, Eyal E, Eliaz RE. Efficacy of rasagiline in early Parkinson’s disease: a meta-analysis of data from the TEMPO and ADAGIO studies. Int J Neurosci. 2016;126:942–6.CrossRefPubMed
171.
go back to reference Jenner P, Langston JW. Explaining ADAGIO: a critical review of the biological basis for the clinical effects of rasagiline. Mov Disord. 2011;26:2316–23.CrossRefPubMed Jenner P, Langston JW. Explaining ADAGIO: a critical review of the biological basis for the clinical effects of rasagiline. Mov Disord. 2011;26:2316–23.CrossRefPubMed
172.
go back to reference Romberg RW, Needleman SB, Snyder JJ, Greedan A. Methamphetamine and amphetamine derived from the metabolism of selegiline. J Forensic Sci. 1995;40:1100–2.CrossRefPubMed Romberg RW, Needleman SB, Snyder JJ, Greedan A. Methamphetamine and amphetamine derived from the metabolism of selegiline. J Forensic Sci. 1995;40:1100–2.CrossRefPubMed
173.
go back to reference Stocchi F, Torti M. Adjuvant therapies for Parkinson’s disease: critical evaluation of safinamide. Drug Des Dev Ther. 2016;10:609–18.CrossRef Stocchi F, Torti M. Adjuvant therapies for Parkinson’s disease: critical evaluation of safinamide. Drug Des Dev Ther. 2016;10:609–18.CrossRef
174.
go back to reference Parkinson Study Group. A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch Neurol. 2002;59:1937–43.CrossRef Parkinson Study Group. A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch Neurol. 2002;59:1937–43.CrossRef
175.
go back to reference Elmer L, Schwid S, Eberly S, Goetz C, Fahn S, Kieburtz K, et al. Rasagiline-associated motor improvement in PD occurs without worsening of cognitive and behavioral symptoms. J Neurol Sci. 2006;248:78–83.CrossRefPubMed Elmer L, Schwid S, Eberly S, Goetz C, Fahn S, Kieburtz K, et al. Rasagiline-associated motor improvement in PD occurs without worsening of cognitive and behavioral symptoms. J Neurol Sci. 2006;248:78–83.CrossRefPubMed
176.
go back to reference Parkinson Study Group. Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease. N Engl J Med. 1993;328:176–83.CrossRef Parkinson Study Group. Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease. N Engl J Med. 1993;328:176–83.CrossRef
177.
go back to reference Hauser RA, Silver D, Choudhry A, Eyal E, Isaacson S. Randomized, controlled trial of rasagiline as an add-on to dopamine agonists in Parkinson’s disease. Mov Disord. 2014;29:1028–34.CrossRefPubMed Hauser RA, Silver D, Choudhry A, Eyal E, Isaacson S. Randomized, controlled trial of rasagiline as an add-on to dopamine agonists in Parkinson’s disease. Mov Disord. 2014;29:1028–34.CrossRefPubMed
178.
go back to reference Richard IH, Kurlan R, Tanner C, Factor S, Hubble J, Suchowersky O, et al. Serotonin syndrome and the combined use of deprenyl and an antidepressant in Parkinson’s disease in Parkinson Study Group. Neurology. 1997;48:1070–7.CrossRefPubMed Richard IH, Kurlan R, Tanner C, Factor S, Hubble J, Suchowersky O, et al. Serotonin syndrome and the combined use of deprenyl and an antidepressant in Parkinson’s disease in Parkinson Study Group. Neurology. 1997;48:1070–7.CrossRefPubMed
179.
go back to reference Panisset M, Chen JJ, Rhyee SH, Conner J, Mathena J. Serotonin toxicity association with concomitant antidepressants and rasagiline treatment: retrospective study (STACCATO). Pharmacotherapy. 2014;34:1250–8.CrossRefPubMed Panisset M, Chen JJ, Rhyee SH, Conner J, Mathena J. Serotonin toxicity association with concomitant antidepressants and rasagiline treatment: retrospective study (STACCATO). Pharmacotherapy. 2014;34:1250–8.CrossRefPubMed
180.
go back to reference Aboukarr A, Giudice M. Interaction between monoamine oxidase B inhibitors and selective serotonin reuptake inhibitors. Can J Hosp Pharm. 2018;71:196–207.PubMedPubMedCentral Aboukarr A, Giudice M. Interaction between monoamine oxidase B inhibitors and selective serotonin reuptake inhibitors. Can J Hosp Pharm. 2018;71:196–207.PubMedPubMedCentral
181.
go back to reference Taylor JJ, Wilson JW, Estes LL. Linezolid and serotonergic drug interactions: a retrospective survey. Clin Infect Dis. 2006;43:180–7.CrossRefPubMed Taylor JJ, Wilson JW, Estes LL. Linezolid and serotonergic drug interactions: a retrospective survey. Clin Infect Dis. 2006;43:180–7.CrossRefPubMed
182.
go back to reference Chen JJ, Swope DM. Clinical pharmacology of rasagiline: a novel, second-generation propargylamine for the treatment of Parkinson disease. J Clin Pharmacol. 2005;45:878–94.CrossRefPubMed Chen JJ, Swope DM. Clinical pharmacology of rasagiline: a novel, second-generation propargylamine for the treatment of Parkinson disease. J Clin Pharmacol. 2005;45:878–94.CrossRefPubMed
183.
go back to reference Churchyard A, Mathias CJ, Boonkongchuen P, Lees AJ. Autonomic effects of selegiline: possible cardiovascular toxicity in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1997;63:228–34.CrossRefPubMedPubMedCentral Churchyard A, Mathias CJ, Boonkongchuen P, Lees AJ. Autonomic effects of selegiline: possible cardiovascular toxicity in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1997;63:228–34.CrossRefPubMedPubMedCentral
184.
go back to reference Laine K, Anttila M, Helminen A, Karnani H, Huupponen R. Dose linearity study of selegiline pharmacokinetics after oral administration: evidence for strong drug interaction with female sex steroids. Br J Clin Pharmacol. 1999;47:249–54.CrossRefPubMedPubMedCentral Laine K, Anttila M, Helminen A, Karnani H, Huupponen R. Dose linearity study of selegiline pharmacokinetics after oral administration: evidence for strong drug interaction with female sex steroids. Br J Clin Pharmacol. 1999;47:249–54.CrossRefPubMedPubMedCentral
185.
go back to reference Palovaara S, Anttila M, Nyman L, Laine K. Effect of concomitant hormone replacement therapy containing estradiol and levonorgestrel on the pharmacokinetics of selegiline. Eur J Clin Pharmacol. 2002;58:259–63.CrossRefPubMed Palovaara S, Anttila M, Nyman L, Laine K. Effect of concomitant hormone replacement therapy containing estradiol and levonorgestrel on the pharmacokinetics of selegiline. Eur J Clin Pharmacol. 2002;58:259–63.CrossRefPubMed
186.
go back to reference Cruz MP. Xadago (Safinamide): a monoamine oxidase B inhibitor for the adjunct treatment of motor symptoms in Parkinson’s disease. P T. 2017;42:622–37.PubMedPubMedCentral Cruz MP. Xadago (Safinamide): a monoamine oxidase B inhibitor for the adjunct treatment of motor symptoms in Parkinson’s disease. P T. 2017;42:622–37.PubMedPubMedCentral
187.
go back to reference Finberg JPM. Pharmacology of rasagiline, a new MAO-B inhibitor drug for the treatment of Parkinson’s disease with neuroprotective potential. Rambam Maimonides Med J. 2010;1:e0003.CrossRefPubMedPubMedCentral Finberg JPM. Pharmacology of rasagiline, a new MAO-B inhibitor drug for the treatment of Parkinson’s disease with neuroprotective potential. Rambam Maimonides Med J. 2010;1:e0003.CrossRefPubMedPubMedCentral
188.
go back to reference Chen JJ, Wilkinson JR. The monoamine oxidase type B inhibitor rasagiline in the treatment of Parkinson disease: is tyramine a challenge? J Clin Pharmacol. 2012;52:620–8.CrossRefPubMed Chen JJ, Wilkinson JR. The monoamine oxidase type B inhibitor rasagiline in the treatment of Parkinson disease: is tyramine a challenge? J Clin Pharmacol. 2012;52:620–8.CrossRefPubMed
189.
go back to reference Marquet A, Kupas K, Johne A, Astruc B, Patat A, Krosser S, et al. The effect of safinamide, a novel drug for Parkinson’s disease, on pressor response to oral tyramine: a randomized, double-blind, clinical trial. Clin Pharmacol Ther. 2012;92:450–7.PubMed Marquet A, Kupas K, Johne A, Astruc B, Patat A, Krosser S, et al. The effect of safinamide, a novel drug for Parkinson’s disease, on pressor response to oral tyramine: a randomized, double-blind, clinical trial. Clin Pharmacol Ther. 2012;92:450–7.PubMed
190.
go back to reference Anttila M, Sotaniemi EA, Pelkonen O, Rautio A. Marked effect of liver and kidney function on the pharmacokinetics of selegiline. Clin Pharmacol Ther. 2005;77:54–62.CrossRefPubMed Anttila M, Sotaniemi EA, Pelkonen O, Rautio A. Marked effect of liver and kidney function on the pharmacokinetics of selegiline. Clin Pharmacol Ther. 2005;77:54–62.CrossRefPubMed
191.
go back to reference Rudolph JL, Salow MJ, Angelini MC, McGlinchey RE. The anticholinergic risk scale and anticholinergic adverse effects in older persons. Arch Intern Med. 2008;168:508–13.CrossRefPubMed Rudolph JL, Salow MJ, Angelini MC, McGlinchey RE. The anticholinergic risk scale and anticholinergic adverse effects in older persons. Arch Intern Med. 2008;168:508–13.CrossRefPubMed
192.
go back to reference Aizenberg D, Sigler M, Weizman A, Barak Y. Anticholinergic burden and the risk of falls among elderly psychiatric inpatients: a 4-year case–control study. Int Psychogeriatr. 2002;14:307–10.CrossRefPubMed Aizenberg D, Sigler M, Weizman A, Barak Y. Anticholinergic burden and the risk of falls among elderly psychiatric inpatients: a 4-year case–control study. Int Psychogeriatr. 2002;14:307–10.CrossRefPubMed
193.
go back to reference 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;81:160–5.CrossRefPubMed 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;81:160–5.CrossRefPubMed
194.
go back to reference Crispo JAG, Willis AW, Thibault DP, Fortin Y, Hays HD, McNair DS, et al. Associations between anticholinergic burden and adverse health outcomes in Parkinson disease. PLoS One. 2016;11:e0150621.CrossRefPubMedPubMedCentral Crispo JAG, Willis AW, Thibault DP, Fortin Y, Hays HD, McNair DS, et al. Associations between anticholinergic burden and adverse health outcomes in Parkinson disease. PLoS One. 2016;11:e0150621.CrossRefPubMedPubMedCentral
195.
go back to reference Vezina P, Mohr E, Grimes D. Deprenyl in Parkinson’s disease: mechanisms, neuroprotective effect, indications and adverse effects. Can J Neurol Sci. 1992;19:142–6.CrossRefPubMed Vezina P, Mohr E, Grimes D. Deprenyl in Parkinson’s disease: mechanisms, neuroprotective effect, indications and adverse effects. Can J Neurol Sci. 1992;19:142–6.CrossRefPubMed
196.
go back to reference Glavin GB, Dugani AM, Pinsky C. L-deprenyl attenuates stress ulcer formation in rats. Neurosci Lett. 1986;70:379–81.CrossRefPubMed Glavin GB, Dugani AM, Pinsky C. L-deprenyl attenuates stress ulcer formation in rats. Neurosci Lett. 1986;70:379–81.CrossRefPubMed
197.
go back to reference Geyer M, Stamenic I, Buhler H, Bertschinger P. Epidemiology of gastrointestinal bleeding in the elderly. Praxis (Bern 1994). 2006;95:757–65.CrossRef Geyer M, Stamenic I, Buhler H, Bertschinger P. Epidemiology of gastrointestinal bleeding in the elderly. Praxis (Bern 1994). 2006;95:757–65.CrossRef
198.
go back to reference Kyaw MH, Chan FKL. Pharmacologic options in the management of upper gastrointestinal bleeding: focus on the elderly. Drugs Aging. 2014;31:349–61.CrossRefPubMed Kyaw MH, Chan FKL. Pharmacologic options in the management of upper gastrointestinal bleeding: focus on the elderly. Drugs Aging. 2014;31:349–61.CrossRefPubMed
199.
go back to reference Moriyoshi K, Masu M, Ishii T, Shigemoto R, Mizuno N, Nakanishi S. Molecular cloning and characterization of the rat NMDA receptor. Nature. 1991;354:31–7.CrossRefPubMed Moriyoshi K, Masu M, Ishii T, Shigemoto R, Mizuno N, Nakanishi S. Molecular cloning and characterization of the rat NMDA receptor. Nature. 1991;354:31–7.CrossRefPubMed
201.
go back to reference Hubsher G, Haider M, Okun MS. Amantadine: the journey from fighting flu to treating Parkinson disease. Neurology. 2012;78:1096–9.CrossRefPubMed Hubsher G, Haider M, Okun MS. Amantadine: the journey from fighting flu to treating Parkinson disease. Neurology. 2012;78:1096–9.CrossRefPubMed
202.
go back to reference Schwab RS, England ACJ, Poskanzer DC, Young RR. Amantadine in the treatment of Parkinson’s disease. JAMA. 1969;208:1168–70.CrossRefPubMed Schwab RS, England ACJ, Poskanzer DC, Young RR. Amantadine in the treatment of Parkinson’s disease. JAMA. 1969;208:1168–70.CrossRefPubMed
203.
go back to reference Mizoguchi K, Yokoo H, Yoshida M, Tanaka T, Tanaka M. Amantadine increases the extracellular dopamine levels in the striatum by re-uptake inhibition and by N-methyl-d-aspartate antagonism. Brain Res. 1994;662:255–8.CrossRefPubMed Mizoguchi K, Yokoo H, Yoshida M, Tanaka T, Tanaka M. Amantadine increases the extracellular dopamine levels in the striatum by re-uptake inhibition and by N-methyl-d-aspartate antagonism. Brain Res. 1994;662:255–8.CrossRefPubMed
204.
go back to reference Pahwa R, Tanner CM, Hauser RA, Isaacson SH, Nausieda PA, Truong DD, et al. ADS-5102 (amantadine) extended-release capsules for levodopa-induced dyskinesia in Parkinson disease (EASE LID Study): a randomized clinical trial. JAMA Neurol. 2017;74:941–9.CrossRefPubMedPubMedCentral Pahwa R, Tanner CM, Hauser RA, Isaacson SH, Nausieda PA, Truong DD, et al. ADS-5102 (amantadine) extended-release capsules for levodopa-induced dyskinesia in Parkinson disease (EASE LID Study): a randomized clinical trial. JAMA Neurol. 2017;74:941–9.CrossRefPubMedPubMedCentral
205.
go back to reference Oertel W, Eggert K, Pahwa R, Tanner CM, Hauser RA, Trenkwalder C, et al. Randomized, placebo-controlled trial of ADS-5102 (amantadine) extended-release capsules for levodopa-induced dyskinesia in Parkinson’s disease (EASE LID 3). Mov Disord. 2017;32:1701–9.CrossRefPubMedPubMedCentral Oertel W, Eggert K, Pahwa R, Tanner CM, Hauser RA, Trenkwalder C, et al. Randomized, placebo-controlled trial of ADS-5102 (amantadine) extended-release capsules for levodopa-induced dyskinesia in Parkinson’s disease (EASE LID 3). Mov Disord. 2017;32:1701–9.CrossRefPubMedPubMedCentral
206.
go back to reference Hiraoka M, Hirano Y, Kawano S, Fan Z, Sawanobori T. Amantadine-induced afterpotentials and automaticity in guinea pig ventricular myocytes. Circ Res. 1989;65:880–93.CrossRefPubMed Hiraoka M, Hirano Y, Kawano S, Fan Z, Sawanobori T. Amantadine-induced afterpotentials and automaticity in guinea pig ventricular myocytes. Circ Res. 1989;65:880–93.CrossRefPubMed
207.
go back to reference Manini AF, Raspberry D, Hoffman RS, Nelson LS. QT prolongation and torsades de pointes following overdose of ziprasidone and amantadine. J Med Toxicol. 2007;3:178–81.CrossRefPubMedPubMedCentral Manini AF, Raspberry D, Hoffman RS, Nelson LS. QT prolongation and torsades de pointes following overdose of ziprasidone and amantadine. J Med Toxicol. 2007;3:178–81.CrossRefPubMedPubMedCentral
209.
go back to reference Akturk IF, Erol MK. Bradyarrhythmias and pacemaker indications in elderly patients. Turk Kardiyol Dern Ars. 2017;45:71–4.PubMed Akturk IF, Erol MK. Bradyarrhythmias and pacemaker indications in elderly patients. Turk Kardiyol Dern Ars. 2017;45:71–4.PubMed
210.
go back to reference Dandamudi S, Slusser J, Mahoney DW, Redfield MM, Rodeheffer RJ, Chen HH. The prevalence of diabetic cardiomyopathy: a population-based study in Olmsted County, Minnesota. J Card Fail. 2014;20:304–9.CrossRefPubMedPubMedCentral Dandamudi S, Slusser J, Mahoney DW, Redfield MM, Rodeheffer RJ, Chen HH. The prevalence of diabetic cardiomyopathy: a population-based study in Olmsted County, Minnesota. J Card Fail. 2014;20:304–9.CrossRefPubMedPubMedCentral
211.
go back to reference Nakata M, Ito S, Shirai W, Hattori T. Severe reversible neurological complications following amantadine treatment in three elderly patients with renal insufficiency. Eur Neurol. 2006;56:59–61.CrossRefPubMed Nakata M, Ito S, Shirai W, Hattori T. Severe reversible neurological complications following amantadine treatment in three elderly patients with renal insufficiency. Eur Neurol. 2006;56:59–61.CrossRefPubMed
212.
go back to reference Bleidner WE, Harmon JB, Hewes WE, Lynes TE, Hermann EC. Absorption, distribution and excretion of amantadine hydrochloride. J Pharmacol Exp Ther. 1965;150:484–90.PubMed Bleidner WE, Harmon JB, Hewes WE, Lynes TE, Hermann EC. Absorption, distribution and excretion of amantadine hydrochloride. J Pharmacol Exp Ther. 1965;150:484–90.PubMed
213.
go back to reference Ing TS, Rahn AC, Armbruster KF, Oyama JH, Klawans HL. Letter: accumulation of amantadine hydrochloride in renal insufficiency. N Engl J Med. 1974;291:1257.PubMed Ing TS, Rahn AC, Armbruster KF, Oyama JH, Klawans HL. Letter: accumulation of amantadine hydrochloride in renal insufficiency. N Engl J Med. 1974;291:1257.PubMed
214.
go back to reference Stoof JC, Booij J, Drukarch B, Wolters EC. The anti-parkinsonian drug amantadine inhibits the N-methyl-d-aspartic acid-evoked release of acetylcholine from rat neostriatum in a non-competitive way. Eur J Pharmacol. 1992;213:439–43.CrossRefPubMed Stoof JC, Booij J, Drukarch B, Wolters EC. The anti-parkinsonian drug amantadine inhibits the N-methyl-d-aspartic acid-evoked release of acetylcholine from rat neostriatum in a non-competitive way. Eur J Pharmacol. 1992;213:439–43.CrossRefPubMed
215.
go back to reference Lupp A, Lucking CH, Koch R, Jackisch R, Feuerstein TJ. Inhibitory effects of the antiparkinsonian drugs memantine and amantadine on N-methyl-d-aspartate-evoked acetylcholine release in the rabbit caudate nucleus in vitro. J Pharmacol Exp Ther. 1992;263:717–24.PubMed Lupp A, Lucking CH, Koch R, Jackisch R, Feuerstein TJ. Inhibitory effects of the antiparkinsonian drugs memantine and amantadine on N-methyl-d-aspartate-evoked acetylcholine release in the rabbit caudate nucleus in vitro. J Pharmacol Exp Ther. 1992;263:717–24.PubMed
217.
go back to reference Neagoe AD. Delirium with manic and psychotic features associated with amantadine. Gen Hosp Psychiatry. 2013;35(680):e7–8. Neagoe AD. Delirium with manic and psychotic features associated with amantadine. Gen Hosp Psychiatry. 2013;35(680):e7–8.
218.
go back to reference Postma JU, Van Tilburg W. Visual hallucinations and delirium during treatment with amantadine (Symmetrel). J Am Geriatr Soc. 1975;23:212–5.CrossRefPubMed Postma JU, Van Tilburg W. Visual hallucinations and delirium during treatment with amantadine (Symmetrel). J Am Geriatr Soc. 1975;23:212–5.CrossRefPubMed
219.
go back to reference Flaherty JA, Bellur SN. Mental side effects of amantadine therapy: its spectrum and characteristics in a normal population. J Clin Psychiatry. 1981;42:344–5.PubMed Flaherty JA, Bellur SN. Mental side effects of amantadine therapy: its spectrum and characteristics in a normal population. J Clin Psychiatry. 1981;42:344–5.PubMed
221.
go back to reference Moreno O, Garcia PT, Sanchez D, Sancho T, Lecumberri B. Cognitive impairment and severe hypocalcemia in a patient with hypoparathyroidism and systemic sclerosis: report of a case. Endocrinol Nutr. 2015;62:356–8.CrossRefPubMed Moreno O, Garcia PT, Sanchez D, Sancho T, Lecumberri B. Cognitive impairment and severe hypocalcemia in a patient with hypoparathyroidism and systemic sclerosis: report of a case. Endocrinol Nutr. 2015;62:356–8.CrossRefPubMed
222.
go back to reference Fujioka S, Fukae J, Ogura H, Mishima T, Yanamoto S, Higuchi M-A, et al. Hospital-based study on emergency admission of patients with Parkinson’s disease. eNeurologicalSci. 2016;4:19–21.CrossRefPubMedPubMedCentral Fujioka S, Fukae J, Ogura H, Mishima T, Yanamoto S, Higuchi M-A, et al. Hospital-based study on emergency admission of patients with Parkinson’s disease. eNeurologicalSci. 2016;4:19–21.CrossRefPubMedPubMedCentral
223.
224.
go back to reference Braga M, Pederzoli M, Antonini A, Beretta F, Crespi V. Reasons for hospitalization in Parkinson’s disease: a case–control study. Parkinsonism Relat Disord. 2014;20:488–92.CrossRefPubMed Braga M, Pederzoli M, Antonini A, Beretta F, Crespi V. Reasons for hospitalization in Parkinson’s disease: a case–control study. Parkinsonism Relat Disord. 2014;20:488–92.CrossRefPubMed
225.
go back to reference Begg DP. Disturbances of thirst and fluid balance associated with aging. Physiol Behav. 2017;178:28–34.CrossRefPubMed Begg DP. Disturbances of thirst and fluid balance associated with aging. Physiol Behav. 2017;178:28–34.CrossRefPubMed
226.
go back to reference Hindle JV. The practical management of cognitive impairment and psychosis in the older Parkinson’s disease patient. J Neural Transm (Vienna). 2013;120:649–53.CrossRef Hindle JV. The practical management of cognitive impairment and psychosis in the older Parkinson’s disease patient. J Neural Transm (Vienna). 2013;120:649–53.CrossRef
227.
228.
go back to reference Mokhles MM, Trifiro G, Dieleman JP, Haag MD, van Soest EM, Verhamme KMC, et al. The risk of new onset heart failure associated with dopamine agonist use in Parkinson’s disease. Pharmacol Res. 2012;65:358–64.CrossRefPubMed Mokhles MM, Trifiro G, Dieleman JP, Haag MD, van Soest EM, Verhamme KMC, et al. The risk of new onset heart failure associated with dopamine agonist use in Parkinson’s disease. Pharmacol Res. 2012;65:358–64.CrossRefPubMed
229.
go back to reference Hasenfuss G, Kasper W, Meinertz T, Busch W, Lehmann M, Krause T, et al. Evaluation of long-term oral levodopa therapy in chronic congestive heart failure. Klin Wochenschr. 1987;65:1087–94.CrossRefPubMed Hasenfuss G, Kasper W, Meinertz T, Busch W, Lehmann M, Krause T, et al. Evaluation of long-term oral levodopa therapy in chronic congestive heart failure. Klin Wochenschr. 1987;65:1087–94.CrossRefPubMed
230.
go back to reference Montastruc JL, Rascol O, Montastruc P. Naloxone or haloperidol but not yohimbine reverse apomorphine-induced respiratory depression. Clin Neuropharmacol. 1992;15:404–7.CrossRefPubMed Montastruc JL, Rascol O, Montastruc P. Naloxone or haloperidol but not yohimbine reverse apomorphine-induced respiratory depression. Clin Neuropharmacol. 1992;15:404–7.CrossRefPubMed
231.
go back to reference Srinivasan M, Lagercrantz H, Yamamoto Y. A possible dopaminergic pathway mediating hypoxic depression in neonatal rabbits. J Appl Physiol. 1985;1989(67):1271–6. Srinivasan M, Lagercrantz H, Yamamoto Y. A possible dopaminergic pathway mediating hypoxic depression in neonatal rabbits. J Appl Physiol. 1985;1989(67):1271–6.
232.
go back to reference Gibbons CH, Simon DK, Huang M, Tilley B, Aminoff MJ, Bainbridge JL, et al. Autonomic and electrocardiographic findings in Parkinson’s disease. Auton Neurosci. 2017;205:93–8.CrossRefPubMedPubMedCentral Gibbons CH, Simon DK, Huang M, Tilley B, Aminoff MJ, Bainbridge JL, et al. Autonomic and electrocardiographic findings in Parkinson’s disease. Auton Neurosci. 2017;205:93–8.CrossRefPubMedPubMedCentral
233.
go back to reference Kannankeril PJ, Roden DM. Drug-induced long QT and torsade de pointes: recent advances. Curr Opin Cardiol. 2007;22:39–43.CrossRefPubMed Kannankeril PJ, Roden DM. Drug-induced long QT and torsade de pointes: recent advances. Curr Opin Cardiol. 2007;22:39–43.CrossRefPubMed
235.
go back to reference Jahn K, Kressig RW, Bridenbaugh SA, Brandt T, Schniepp R. Dizziness and unstable gait in old age: etiology, diagnosis and treatment. Dtsch Arztebl Int. 2015;112:387–93.PubMedPubMedCentral Jahn K, Kressig RW, Bridenbaugh SA, Brandt T, Schniepp R. Dizziness and unstable gait in old age: etiology, diagnosis and treatment. Dtsch Arztebl Int. 2015;112:387–93.PubMedPubMedCentral
236.
go back to reference Hanewinckel R, van Oijen M, Ikram MA, van Doorn PA. The epidemiology and risk factors of chronic polyneuropathy. Eur J Epidemiol. 2016;31:5–20.CrossRefPubMed Hanewinckel R, van Oijen M, Ikram MA, van Doorn PA. The epidemiology and risk factors of chronic polyneuropathy. Eur J Epidemiol. 2016;31:5–20.CrossRefPubMed
237.
go back to reference Nyholm D, Nilsson Remahl AIM, Dizdar N, Constantinescu R, Holmberg B, Jansson R, et al. Duodenal levodopa infusion monotherapy vs oral polypharmacy in advanced Parkinson disease. Neurology. 2005;64:216–23.CrossRefPubMed Nyholm D, Nilsson Remahl AIM, Dizdar N, Constantinescu R, Holmberg B, Jansson R, et al. Duodenal levodopa infusion monotherapy vs oral polypharmacy in advanced Parkinson disease. Neurology. 2005;64:216–23.CrossRefPubMed
239.
go back to reference Krause O, Wiese B, Doyle I-M, Kirsch C, Thürmann P, Wilm S, et al. Multidisciplinary intervention to improve medication safety in nursing home residents: protocol of a cluster randomised controlled trial (HIOPP-3-iTBX study). BMC Geriatr. 2019;19:24.CrossRefPubMedPubMedCentral Krause O, Wiese B, Doyle I-M, Kirsch C, Thürmann P, Wilm S, et al. Multidisciplinary intervention to improve medication safety in nursing home residents: protocol of a cluster randomised controlled trial (HIOPP-3-iTBX study). BMC Geriatr. 2019;19:24.CrossRefPubMedPubMedCentral
240.
241.
go back to reference Yildirim AB, Kilinc AY. Polypharmacy and drug interactions in elderly patients. Turk Kardiyol Dern Ars. 2017;45:17–21.PubMed Yildirim AB, Kilinc AY. Polypharmacy and drug interactions in elderly patients. Turk Kardiyol Dern Ars. 2017;45:17–21.PubMed
242.
go back to reference Moriarty F, Hardy C, Bennett K, Smith SM, Fahey T. Trends and interaction of polypharmacy and potentially inappropriate prescribing in primary care over 15 years in Ireland: a repeated cross-sectional study. BMJ Open. 2015;5:e008656.CrossRefPubMedPubMedCentral Moriarty F, Hardy C, Bennett K, Smith SM, Fahey T. Trends and interaction of polypharmacy and potentially inappropriate prescribing in primary care over 15 years in Ireland: a repeated cross-sectional study. BMJ Open. 2015;5:e008656.CrossRefPubMedPubMedCentral
243.
go back to reference Muhic N, Mrhar A, Brvar M. Comparative analysis of three drug–drug interaction screening systems against probable clinically relevant drug–drug interactions: a prospective cohort study. Eur J Clin Pharmacol. 2017;73:875–82.CrossRefPubMed Muhic N, Mrhar A, Brvar M. Comparative analysis of three drug–drug interaction screening systems against probable clinically relevant drug–drug interactions: a prospective cohort study. Eur J Clin Pharmacol. 2017;73:875–82.CrossRefPubMed
244.
go back to reference Somogyi-Vegh A, Nyaka B, Vida RG, Lovasz A, Botz L. Comprehensive evaluation of drug interaction screening programs: discrepancies and concordances. Orv Hetil. 2015;156:720–30.CrossRefPubMed Somogyi-Vegh A, Nyaka B, Vida RG, Lovasz A, Botz L. Comprehensive evaluation of drug interaction screening programs: discrepancies and concordances. Orv Hetil. 2015;156:720–30.CrossRefPubMed
245.
go back to reference Kheshti R, Aalipour M, Namazi S. A comparison of five common drug–drug interaction software programs regarding accuracy and comprehensiveness. J Res Pharm Pract. 2016;5:257–63.CrossRefPubMedPubMedCentral Kheshti R, Aalipour M, Namazi S. A comparison of five common drug–drug interaction software programs regarding accuracy and comprehensiveness. J Res Pharm Pract. 2016;5:257–63.CrossRefPubMedPubMedCentral
246.
go back to reference Moura CS, Prado NM, Belo NO, Acurcio FA. Evaluation of drug–drug interaction screening software combined with pharmacist intervention. Int J Clin Pharm. 2012;34:547–52.CrossRefPubMed Moura CS, Prado NM, Belo NO, Acurcio FA. Evaluation of drug–drug interaction screening software combined with pharmacist intervention. Int J Clin Pharm. 2012;34:547–52.CrossRefPubMed
247.
go back to reference Pazan F, Weiss C, Wehling M. The FORTA (Fit fOR The Aged) List 2015: update of a validated clinical tool for improved pharmacotherapy in the elderly. Drugs Aging. 2016;33:447–9.CrossRefPubMed Pazan F, Weiss C, Wehling M. The FORTA (Fit fOR The Aged) List 2015: update of a validated clinical tool for improved pharmacotherapy in the elderly. Drugs Aging. 2016;33:447–9.CrossRefPubMed
248.
go back to reference Kuhn-Thiel AM, Weiss C, Wehling M. Consensus validation of the FORTA (Fit fOR The Aged) List: a clinical tool for increasing the appropriateness of pharmacotherapy in the elderly. Drugs Aging. 2014;31:131–40.CrossRefPubMed Kuhn-Thiel AM, Weiss C, Wehling M. Consensus validation of the FORTA (Fit fOR The Aged) List: a clinical tool for increasing the appropriateness of pharmacotherapy in the elderly. Drugs Aging. 2014;31:131–40.CrossRefPubMed
249.
go back to reference Brown JD, Hutchison LC, Li C, Painter JT, Martin BC. Predictive validity of the Beers and Screening Tool of Older Persons’ Potentially Inappropriate Prescriptions (STOPP) criteria to detect adverse drug events, hospitalizations, and emergency department visits in the United States. J Am Geriatr Soc. 2016;64:22–30.CrossRefPubMedPubMedCentral Brown JD, Hutchison LC, Li C, Painter JT, Martin BC. Predictive validity of the Beers and Screening Tool of Older Persons’ Potentially Inappropriate Prescriptions (STOPP) criteria to detect adverse drug events, hospitalizations, and emergency department visits in the United States. J Am Geriatr Soc. 2016;64:22–30.CrossRefPubMedPubMedCentral
250.
go back to reference Hamilton H, Gallagher P, Ryan C, Byrne S, O’Mahony D. Potentially inappropriate medications defined by STOPP criteria and the risk of adverse drug events in older hospitalized patients. Arch Intern Med. 2011;171:1013–9.CrossRefPubMed Hamilton H, Gallagher P, Ryan C, Byrne S, O’Mahony D. Potentially inappropriate medications defined by STOPP criteria and the risk of adverse drug events in older hospitalized patients. Arch Intern Med. 2011;171:1013–9.CrossRefPubMed
251.
go back to reference Siebert S, Elkeles B, Hempel G, Kruse J, Smollich M. The PRISCUS list in clinical routine: practicability and comparison to international PIM lists. Z Gerontol Geriatr. 2013;46:35–47.CrossRefPubMed Siebert S, Elkeles B, Hempel G, Kruse J, Smollich M. The PRISCUS list in clinical routine: practicability and comparison to international PIM lists. Z Gerontol Geriatr. 2013;46:35–47.CrossRefPubMed
252.
go back to reference Cooper JA, Cadogan CA, Patterson SM, Kerse N, Bradley MC, Ryan C, et al. Interventions to improve the appropriate use of polypharmacy in older people: a Cochrane systematic review. BMJ Open. 2015;5:e009235.CrossRefPubMedPubMedCentral Cooper JA, Cadogan CA, Patterson SM, Kerse N, Bradley MC, Ryan C, et al. Interventions to improve the appropriate use of polypharmacy in older people: a Cochrane systematic review. BMJ Open. 2015;5:e009235.CrossRefPubMedPubMedCentral
253.
go back to reference Kim J, Parish AL. Polypharmacy and medication management in older adults. Nurs Clin North Am. 2017;52:457–68.CrossRefPubMed Kim J, Parish AL. Polypharmacy and medication management in older adults. Nurs Clin North Am. 2017;52:457–68.CrossRefPubMed
254.
go back to reference O’Mahony D, O’Ullivan D, Byrne S, O’Connor MN, Ryan C, Gallagher P. STOPP/START criteria for potentially inappropriate prescribing in older people: version 2. Age Ageing. 2015;44:213–8.CrossRefPubMed O’Mahony D, O’Ullivan D, Byrne S, O’Connor MN, Ryan C, Gallagher P. STOPP/START criteria for potentially inappropriate prescribing in older people: version 2. Age Ageing. 2015;44:213–8.CrossRefPubMed
255.
256.
go back to reference Antimisiaris D, Cutler T. Managing polypharmacy in the 15-minute office visit. Prim Care. 2017;44:413–28.CrossRefPubMed Antimisiaris D, Cutler T. Managing polypharmacy in the 15-minute office visit. Prim Care. 2017;44:413–28.CrossRefPubMed
257.
go back to reference Jansen J, Naganathan V, Carter SM, McLachlan AJ, Nickel B, Irwig L, et al. Too much medicine in older people? Deprescribing through shared decision making. BMJ. 2016;353:i2893.CrossRefPubMed Jansen J, Naganathan V, Carter SM, McLachlan AJ, Nickel B, Irwig L, et al. Too much medicine in older people? Deprescribing through shared decision making. BMJ. 2016;353:i2893.CrossRefPubMed
258.
go back to reference Scott IA, Hilmer SN, Reeve E, Potter K, Le Couteur D, Rigby D, et al. Reducing inappropriate polypharmacy: the process of deprescribing. JAMA Intern Med. 2015;175:827–34.CrossRefPubMed Scott IA, Hilmer SN, Reeve E, Potter K, Le Couteur D, Rigby D, et al. Reducing inappropriate polypharmacy: the process of deprescribing. JAMA Intern Med. 2015;175:827–34.CrossRefPubMed
259.
go back to reference Loffler C, Drewelow E, Paschka SD, Frankenstein M, Eger J, Jatsch L, et al. Optimizing polypharmacy among elderly hospital patients with chronic diseases: study protocol of the cluster randomized controlled POLITE-RCT trial. Implement Sci. 2014;9:151.CrossRefPubMedPubMedCentral Loffler C, Drewelow E, Paschka SD, Frankenstein M, Eger J, Jatsch L, et al. Optimizing polypharmacy among elderly hospital patients with chronic diseases: study protocol of the cluster randomized controlled POLITE-RCT trial. Implement Sci. 2014;9:151.CrossRefPubMedPubMedCentral
Metadata
Title
Safety and Tolerability of Pharmacotherapies for Parkinson’s Disease in Geriatric Patients
Authors
Martin Klietz
Stephan Greten
Florian Wegner
Günter U. Höglinger
Publication date
01-06-2019
Publisher
Springer International Publishing
Published in
Drugs & Aging / Issue 6/2019
Print ISSN: 1170-229X
Electronic ISSN: 1179-1969
DOI
https://doi.org/10.1007/s40266-019-00654-z

Other articles of this Issue 6/2019

Drugs & Aging 6/2019 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.