295
Views
0
CrossRef citations to date
0
Altmetric
Review

Safety review of current pharmacotherapies for levodopa-treated patients with Parkinson’s disease

, &
Pages 563-579 | Received 24 Mar 2023, Accepted 15 Jun 2023, Published online: 04 Jul 2023

References

  • Birkmayer W, Hornykiewicz O. [The L-3,4-dioxyphenylalanine (DOPA)-effect in Parkinson-akinesia]. Wien Klin Wochenschr. 1961 Nov 10;73:787–788.
  • Fox SH, Katzenschlager R, Lim SY, et al. International Parkinson and movement disorder society evidence-based medicine review: update on treatments for the motor symptoms of Parkinson’s disease. Mov Disord. 2018 Aug;33(8):1248–1266.
  • Widnell K. Pathophysiology of motor fluctuations in Parkinson’s disease. Mov Disord. 2005;20(11):S17–22. doi:10.1002/mds.20459.
  • Nyholm D, Lennernas H, Gomes-Trolin C, et al. Levodopa pharmacokinetics and motor performance during activities of daily living in patients with Parkinson’s disease on individual drug combinations. Clin Neuropharmacol. 2002 Mar;25(2):89–96.
  • Lieberman A, Olanow CW, Sethi K, et al. A multicenter trial of ropinirole as adjunct treatment for Parkinson’s disease ropinirole study group. Neurology. 1998 Oct;51(4):1057–1062.
  • Nyholm D. The rationale for continuous dopaminergic stimulation in advanced Parkinson’s disease. Parkinsonism Relat Disord. 2007 Sep;13:S13–7. doi: 10.1016/j.parkreldis.2007.06.005
  • Adler CH. Relevance of motor complications in Parkinson’s disease. Neurology. 2002 Feb 26;58(4 Suppl 1):S51–6. doi: 10.1212/WNL.58.suppl_1.S51
  • Chou KL, Stacy M, Simuni T, et al. The spectrum of “off” in Parkinson’s disease: what have we learned over 40 years? Parkinsonism Relat Disord. 2018 Jun;51:9–16.
  • Aquino CC, Fox SH. Clinical spectrum of levodopa-induced complications. Mov Disord. 2015 Jan;30(1):80–89. doi: 10.1002/mds.26125
  • Contin M, Martinelli P. Pharmacokinetics of levodopa. J Neurol. 2010 Nov;257(Suppl 2):S253–61. doi: 10.1007/s00415-010-5728-8
  • Calabresi P, Di Filippo M, Ghiglieri V, et al. Levodopa-induced dyskinesias in patients with Parkinson’s disease: filling the bench-to-bedside gap. Lancet Neurol. 2010 Nov;9(11):1106–1117.
  • SINEMET® (Carbidopa-Levodopa) Tablets [prescribing information]. Princeton, NJ: Merck & Co., Inc. 2014. [cited 2023 Feb 10]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/017555s069lbl.pdf
  • DHIVY (carbidopa and levodopa) tablets [prescribing information]. Washington, DC: Riverside Pharmacy Corporation. 2021. [cited 2023 Mar 21]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/214869s000lbl.pdf
  • PARCOPA TM (carbidopa-levodopa orally disintegrating tablets) [prescribing information]. Eden Prairie, MN: Schwarz Pharma. 2007. [cited 2023 Feb 6]. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/076699_S000_PARCOPA_PRNTLBL.pdf
  • SINEMET® CR (carbidopa levodopa) Sustained-Release Tablets [prescribing information]. Whitehouse Station, NJ: Merck & Co., Inc. 2014. [cited 2023 Feb 10]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/019856s016s024s028lbl.pdf
  • DUOPA®(carbidopa and levodopa) enteral suspension [prescribing information]. North Chicago, IL: AbbVie Inc. 2022. [cited 2023 Feb 6]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/203952s004lbl.pdf
  • Rytary (carbidopa and levodopa) extended-release capsules [prescribing information]. Hayward, CA: Impax Pharmaceuticals. 2019. [cited 2023 Feb 10]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/203312s000lbl.pdf
  • INBRIJA™ (levodopa inhalation powder) [prescribing information]. Ardsley, NY: Acorda Therapeutics, Inc. 2022. [cited 2023 Feb 6]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/209184s000lbl.pdf
  • Markham CH, Treciokas LJ, Diamond SG. Parkinson’s disease and levodopa. A five-year follow-up and review. West J Med. 1974 Sep;121(3):188–206.
  • Block G, Liss C, Reines S, et al. Comparison of immediate-release and controlled release carbidopa/levodopa in Parkinson’s disease. A multicenter 5-year study. The CR First Study Group. Eur Neurol. 1997;37(1):23–27. DOI:10.1159/000117399
  • Ondo WG, Shinawi L, Moore S. Comparison of orally dissolving carbidopa/levodopa (Parcopa) to conventional oral carbidopa/levodopa: a single-dose, double-blind, double-dummy, placebo-controlled, crossover trial. Mov Disord. 2010 Dec 15;25(16):2724–2727. doi: 10.1002/mds.23158
  • Hauser RA, Hsu A, Kell S, et al. Extended-release carbidopa-levodopa (IPX066) compared with immediate-release carbidopa-levodopa in patients with Parkinson’s disease and motor fluctuations: a phase 3 randomised, double-blind trial. Lancet Neurol. 2013 Apr;12(4):346–356.
  • Pahwa R, Lyons KE, Hauser RA, et al. Randomized trial of IPX066, carbidopa/levodopa extended release, in early Parkinson’s disease. Parkinsonism Relat Disord. 2014 Feb;20(2):142–148.
  • LeWitt PA, Hauser RA, Grosset DG, et al. A randomized trial of inhaled levodopa (CVT-301) for motor fluctuations in Parkinson’s disease. Mov Disord. 2016 Sep;31(9):1356–1365.
  • LeWitt PA, Pahwa R, Sedkov A, et al. Pulmonary Safety and Tolerability of Inhaled Levodopa (CVT-301) Administered to Patients with Parkinson’s Disease. J Aerosol Med Pulm Drug Deliv. 2018 Jun;31(3):155–161.
  • Grosset D, Dhall R, Gurevich T, et al. Long-term Efficacy of Inhaled Levodopa in Parkinson’s Disease Subjects with Motor Fluctuations: a Phase 3 Open-Label Randomized Study (S26.008). Neurology. 2018;90(15 Supplement):S26.008.
  • Grosset D, Dhall R, Gurevich T, et al. Long-Term Pulmonary Safety of Inhaled Levodopa in Parkinson’s Disease Subjects with Motor Fluctuations: a Phase 3 Open-Label Randomized Study (S26.006). Neurology. 2018;90(15 Supplement):S26.006.
  • LeWitt PA, Hauser RA, Pahwa R, et al. Safety and efficacy of CVT-301 (levodopa inhalation powder) on motor function during off periods in patients with Parkinson’s disease: a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Neurol. 2019 Feb;18(2):145–154.
  • Rispoli V, Simioni V, Capone JG, et al. Peripheral neuropathy in 30 duodopa patients with vitamins B supplementation. Acta Neurol Scand. 2017 Dec;136(6):660–667.
  • Fernandez HH, Boyd JT, Fung VSC, et al. Long-term safety and efficacy of levodopa-carbidopa intestinal gel in advanced Parkinson’s disease. Mov Disord. 2018 Jul;33(6):928–936.
  • Olanow CW, Kieburtz K, Odin P, et al. Continuous intrajejunal infusion of levodopa-carbidopa intestinal gel for patients with advanced Parkinson’s disease: a randomised, controlled, double-blind, double-dummy study. Lancet Neurol. 2014 Feb;13(2):141–149.
  • Isaacson SH, Pagan FL, Lew MF, et al. Should “on-demand” treatments for Parkinson’s disease off episodes be used earlier? Clin Park Relat Disord. 2022;7:100161. DOI:10.1016/j.prdoa.2022.100161
  • Fabbri M, Barbosa R, Rascol O. Off-time Treatment Options for Parkinson’s Disease. Neurol Ther. 2023 Jan 12;12(2):391–424. doi: 10.1007/s40120-022-00435-8
  • Lieberman A, Ranhosky A, Korts D. Clinical evaluation of pramipexole in advanced Parkinson’s disease: results of a double-blind, placebo-controlled, parallel-group study. Neurology. 1997 Jul;49(1):162–168. doi: 10.1212/WNL.49.1.162
  • Hubble JP, Koller WC, Cutler NR, et al. Pramipexole in patients with early Parkinson’s disease. Clin Neuropharmacol. 1995 Aug;18(4):338–347.
  • Molho ES, Factor SA, Weiner WJ, et al. The use of pramipexole, a novel dopamine (DA) agonist, in advanced Parkinson’s disease. J Neural Transm Suppl. 1995;45:225–230.
  • Guttman M. Double-blind comparison of pramipexole and bromocriptine treatment with placebo in advanced Parkinson’s disease. International Pramipexole-Bromocriptine Study Group. Neurology. 1997 Oct;49(4):1060–1065. doi: 10.1212/WNL.49.4.1060
  • Pinter MM, Pogarell O, Oertel WH. Efficacy, safety, and tolerance of the non-ergoline dopamine agonist pramipexole in the treatment of advanced Parkinson’s disease: a double blind, placebo controlled, randomised, multicentre study. J Neurol Neurosurg Psychiatry. 1999 Apr;66(4):436–441. doi: 10.1136/jnnp.66.4.436
  • Mizuno Y, Yanagisawa N, Kuno S, et al. Randomized, double-blind study of pramipexole with placebo and bromocriptine in advanced Parkinson’s disease. Mov Disord. 2003 Oct;18(10):1149–1156.
  • Moller JC, Oertel WH, Koster J, et al. Long-term efficacy and safety of pramipexole in advanced Parkinson’s disease: results from a European multicenter trial. Mov Disord. 2005 May;20(5):602–610.
  • Poewe WH, Rascol O, Quinn N, et al. Efficacy of pramipexole and transdermal rotigotine in advanced Parkinson’s disease: a double-blind, double-dummy, randomised controlled trial. Lancet Neurol. 2007 Jun;6(6):513–520.
  • Stowe R, Ives N, Clarke CE, et al. Evaluation of the efficacy and safety of adjuvant treatment to levodopa therapy in Parkinson s disease patients with motor complications. Cochrane Database Syst Rev. 2010 Jul;7(7):CD007166.
  • Schapira AH, Barone P, Hauser RA, et al. Extended-release pramipexole in advanced Parkinson disease: a randomized controlled trial. Neurology. 2011 Aug 23;77(8):767–774. doi:10.1212/WNL.0b013e31822affdb.
  • Hauser RA, Schapira AH, Barone P, et al. Long-term safety and sustained efficacy of extended-release pramipexole in early and advanced Parkinson’s disease. Eur J Neurol. 2014 May;21(5):736–743.
  • Schapira AH, Barone P, Hauser RA, et al. Success rate, efficacy, and safety/tolerability of overnight switching from immediate- to extended-release pramipexole in advanced Parkinson’s disease. Eur J Neurol. 2013 Jan;20(1):180–187.
  • Pahwa R, Stacy MA, Factor SA, et al. Ropinirole 24-hour prolonged release: randomized, controlled study in advanced Parkinson disease. Neurology. 2007 Apr 3;68(14):1108–1115. doi:10.1212/01.wnl.0000258660.74391.c1.
  • LeWitt PA, Lyons KE, Pahwa R, et al. Advanced Parkinson disease treated with rotigotine transdermal system: pREFER Study. Neurology. 2007 Apr 17;68(16):1262–1267. DOI:10.1212/01.wnl.0000259516.61938.bb
  • Dewey, RB Jr., Hutton JT, LeWitt PA, et al. A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch Neurol. 2001 Sep;58(9):1385–1392. doi:10.1001/archneur.58.9.1385.
  • Olanow CW, Factor SA, Espay AJ, et al. Apomorphine sublingual film for off episodes in Parkinson’s disease: a randomised, double-blind, placebo-controlled phase 3 study. Lancet Neurol. 2020 Feb;19(2):135–144.
  • Waters CH, Sethi KD, Hauser RA, et al. Zydis selegiline reduces off time in Parkinson’s disease patients with motor fluctuations: a 3-month, randomized, placebo-controlled study. Mov Disord. 2004 Apr;19(4):426–432.
  • Schapira AH, Fox SH, Hauser RA, et al. Assessment of Safety and Efficacy of Safinamide as a Levodopa Adjunct in Patients with Parkinson Disease and Motor Fluctuations: a Randomized Clinical Trial. JAMA Neurol. 2017 Feb 1;74(2):216–224. doi:10.1001/jamaneurol.2016.4467.
  • Group PS. A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations: the PRESTO study. Arch Neurol. 2005 Feb;62(2):241–248.
  • Rascol O, Brooks DJ, Melamed E, et al. Rasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once daily, study): a randomised, double-blind, parallel-group trial. Lancet. 2005 Mar 12-18;365(9463):947–954. doi:10.1016/S0140-6736(05)71083-7.
  • Borgohain R, Szasz J, Stanzione P, et al. Randomized trial of safinamide add-on to levodopa in Parkinson’s disease with motor fluctuations. Mov Disord. 2014 Feb;29(2):229–237.
  • Rajput AH, Martin W, Saint-Hilaire MH, et al. Tolcapone improves motor function in parkinsonian patients with the “wearing-off” phenomenon: a double-blind, placebo-controlled, multicenter trial. Neurology. 1997 Oct;49(4):1066–1071.
  • Group PS. Entacapone improves motor fluctuations in levodopa-treated Parkinson’s disease patients. Ann Neurol. 1997 Nov;42(5):747–755. doi:10.1002/ana.410420511
  • Baas H, Beiske AG, Ghika J, et al. Catechol-O-methyltransferase inhibition with tolcapone reduces the “wearing off” phenomenon and levodopa requirements in fluctuating parkinsonian patients. J Neurol Neurosurg Psychiatry. 1997 Oct;63(4):421–428.
  • Rinne UK, Larsen JP, Siden A, et al. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations nomecomt study group. Neurology. 1998 Nov;51(5):1309–1314.
  • Mizuno Y, Kondo T, Japanese Istradefylline Study G. Adenosine A2A receptor antagonist istradefylline reduces daily off time in Parkinson’s disease. Mov Disord. 2013 Jul;28(8):1138–1141. doi: 10.1002/mds.25418
  • Lees AJ, Ferreira J, Rascol O, et al. Opicapone as Adjunct to Levodopa Therapy in Patients with Parkinson Disease and Motor Fluctuations: a Randomized Clinical Trial. JAMA Neurol. 2017 Feb 1;74(2):197–206. doi:10.1001/jamaneurol.2016.4703.
  • Ferreira JJ, Lees A, Rocha JF, et al. 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 Feb;15(2):154–165.
  • Pahwa R, Tanner CM, Hauser RA, 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 Aug 1;74(8):941–949. doi:10.1001/jamaneurol.2017.0943
  • Oertel W, Eggert K, Pahwa R, 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 Dec;32(12):1701–1709. doi:10.1002/mds.27131.
  • LeWitt PA, Guttman M, Tetrud JW, et al. Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces “off” time in Parkinson’s disease: a double-blind, randomized, multicenter clinical trial (6002-US-005). Ann Neurol. 2008 Mar;63(3):295–302.
  • Hauser RA, Hattori N, Fernandez H, et al. Efficacy of Istradefylline, an Adenosine A2A Receptor Antagonist, as Adjunctive Therapy to Levodopa in Parkinson’s Disease: a Pooled Analysis of 8 Phase 2b/3 Trials. J Parkinsons Dis. 2021;11(4):1663–1675. doi: 10.3233/JPD-212672
  • Hauser RA, Shulman LM, Trugman JM, et al. Study of istradefylline in patients with Parkinson’s disease on levodopa with motor fluctuations. Mov Disord. 2008 Nov 15;23(15):2177–2185. DOI:10.1002/mds.22095
  • Golbe LI, Lieberman AN, Muenter MD, et al. Deprenyl in the treatment of symptom fluctuations in advanced Parkinson’s disease. Clin Neuropharmacol. 1988 Feb;11(1):45–55.
  • Mizuno Y, Hasegawa K, Kondo T, et al. Clinical efficacy of istradefylline (KW-6002) in Parkinson’s disease: a randomized, controlled study. Mov Disord. 2010 Jul 30;25(10):1437–1443. doi:10.1002/mds.23107
  • MIRAPEX® (pramipexole dihydrochloride) tablets: Boehringer Ingelheim Pharmaceuticals, Inc; 2018 [February 26, 2023]; Prescribing Information]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/020667s036lbl.pdf
  • Foley P, Gerlach M, Double KL, et al. Dopamine receptor agonists in the therapy of Parkinson’s disease. J Neural Transm (Vienna). 2004 Oct;111(10–11):1375–1446.
  • Piercey MF. Pharmacology of pramipexole, a dopamine D3-preferring agonist useful in treating Parkinson’s disease. Clin Neuropharmacol. 1998 May;21(3):141–151.
  • Pfeiffer RF. A promising new technology for Parkinson’s disease. Neurology. 2005 Jul 26;65(2 Suppl 1):S6–10. doi: 10.1212/WNL.65.2_suppl_1.S6
  • MIRAPEX ER® (pramipexole dihydrochloride) extended-release tablets: Boehringer Ingelheim Pharmaceuticals, Inc; 2018 [February 24, 2023]; Prescribing Information]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/022421s017lbl.pdf
  • Jenner P, Konen-Bergmann M, Schepers C, et al. Pharmacokinetics of a once-daily extended-release formulation of pramipexole in healthy male volunteers: three studies. Clin Ther. 2009 Nov;31(11):2698–2711.
  • Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. N Engl J Med. 2000 May 18;342(20):1484–1491. DOI:10.1056/NEJM200005183422004
  • Ceravolo R, Rossi C, Del Prete E, et al. A review of adverse events linked to dopamine agonists in the treatment of Parkinson’s disease. Expert Opin Drug Saf. 2016;15(2):181–198. DOI:10.1517/14740338.2016.1130128
  • Antonini A, Tolosa E, Mizuno Y, et al. A reassessment of risks and benefits of dopamine agonists in Parkinson’s disease. Lancet Neurol. 2009 Oct;8(10):929–937.
  • Voon V, Napier TC, Frank MJ, et al. Impulse control disorders and levodopa-induced dyskinesias in Parkinson’s disease: an update. Lancet Neurol. 2017 Mar;16(3):238–250.
  • Zhou CQ, Lou JH, Zhang YP, et al. Long-acting versus standard non-ergot dopamine agonists in Parkinson’s disease: a meta-analysis of randomized controlled trials. CNS Neurosci Ther. 2014 Apr;20(4):368–376.
  • Tulloch IF. Pharmacologic profile of ropinirole: a nonergoline dopamine agonist. Neurology. 1997 Jul;49(1 Suppl 1):S58–62. doi: 10.1212/WNL.49.1_Suppl_1.S58
  • Adler CH, Sethi KD, Hauser RA, et al. Ropinirole for the treatment of early Parkinson’s disease. The Ropinirole Study Group. Neurology. 1997 Aug;49(2):393–399.
  • Rascol O, Lees AJ, Senard JM, et al. Ropinirole in the treatment of levodopa-induced motor fluctuations in patients with Parkinson’s disease. Clin Neuropharmacol. 1996 Jun;19(3):234–245.
  • Brunt ER, Brooks DJ, Korczyn AD, et al. A six-month multicentre, double-blind, bromocriptine-controlled study of the safety and efficacy of ropinirole in the treatment of patients with Parkinson’s disease not optimally controlled by L-dopa. J Neural Transm (Vienna). 2002 Apr;109(4):489–502.
  • Im JH, Ha JH, Cho IS, et al. Ropinirole as an adjunct to levodopa in the treatment of Parkinson’s disease: a 16-week bromocriptine controlled study. J Neurol. 2003 Jan;250(1):90–96.
  • Mizuno Y, Abe T, Hasegawa K, et al. Ropinirole is effective on motor function when used as an adjunct to levodopa in Parkinson’s disease: sTRONG study. Mov Disord. 2007 Oct 15;22(13):1860–1865. DOI:10.1002/mds.21313
  • Zesiewicz TA, Chriscoe S, Jimenez T, et al. A randomized, fixed-dose, dose-response study of ropinirole prolonged release in advanced Parkinson’s disease. Neurodegener Dis Manag. 2017 Feb;7(1):61–72.
  • REQUIP XL (ropinirole) extended-release tablets [prescribing information]. Research Triangle Park, NC: GlaxoSmithKline; 2021. [cited 2023 Feb 24]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/022008s003s004s007s008lbledt.pdf
  • Tompson D, Oliver-Willwong R. Pharmacokinetic and pharmacodynamic comparison of ropinirole 24-hour prolonged release and ropinirole immediate release in patients with Parkinson’s disease. Clin Neuropharmacol. 2009 May;32(3):140–148. doi: 10.1097/WNF.0b013e318176c505
  • REQUIP® (ropinirole hydrochloride) Tablets [prescribing information]. Research Triangle Park, NC: GlaxoSmithKline; 2007. [cited 2023 Feb 23]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/020658s018s020s021lbl.pdf
  • Weintraub D, Koester J, Potenza MN, et al. Impulse control disorders in Parkinson disease: a cross-sectional study of 3090 patients. Arch Neurol. 2010 May;67(5):589–595.
  • Wood M, Dubois V, Scheller D, et al. Rotigotine is a potent agonist at dopamine D1 receptors as well as at dopamine D2 and D3 receptors. Br J Pharmacol. 2015 Feb;172(4):1124–1135.
  • Scheller D, Ullmer C, Berkels R, et al. The in vitro receptor profile of rotigotine: a new agent for the treatment of Parkinson’s disease. Naunyn Schmiedebergs Arch Pharmacol. 2009 Jan;379(1):73–86.
  • LeWitt PA, Boroojerdi B, Surmann E, et al. Rotigotine transdermal system for long-term treatment of patients with advanced Parkinson’s disease: results of two open-label extension studies, CLEOPATRA-PD and PREFER. J Neural Transm (Vienna). 2013 Jul;120(7):1069–1081.
  • NEUPRO (rotigotine transdermal system) [prescribing information]. Smyrna, GA: UCB, Inc. 2021. [cited 2023 Feb 10]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/021829s001lbl.pdf
  • Gupta HV, Lyons KE, Pahwa R. Old Drugs, New Delivery Systems in Parkinson’s Disease. Drugs Aging. 2019 Sep;36(9):807–821. doi: 10.1007/s40266-019-00682-9
  • Reynolds NA, Wellington K, Easthope SE. Rotigotine: in Parkinson’s disease. CNS Drugs. 2005;19(11):973–981. doi:10.2165/00023210-200519110-00006.
  • Chen JJ, Swope DM, Dashtipour K, et al. Transdermal rotigotine: a clinically innovative dopamine-receptor agonist for the management of Parkinson’s disease. Pharmacotherapy. 2009 Dec;29(12):1452–1467.
  • Deleu D, Hanssens Y, Northway MG. Subcutaneous apomorphine: an evidence-based review of its use in Parkinson’s disease. Drugs Aging. 2004;21(11):687–709. doi:10.2165/00002512-200421110-00001.
  • Clinical Review Report: Apomorphine (Movapo):Paladin Labs, Inc. Indication: The acute, intermittent treatment of hypomobility “off” episodes (“end-of-dose wearing off” and unpredictable “on/off” episodes) in patients with advanced Parkinson’s disease. Ottawa, Ontario: Canadian Agency for Drugs and Technologies in Health; 2018.
  • Pfeiffer RF, Gutmann L, Hull KL Jr., et al. Continued efficacy and safety of subcutaneous apomorphine in patients with advanced Parkinson’s disease. Parkinsonism Relat Disord. 2007 Mar;13(2):93–100.
  • Pahwa R, Koller WC, Trosch RM, et al. Subcutaneous apomorphine in patients with advanced Parkinson’s disease: a dose-escalation study with randomized, double-blind, placebo-controlled crossover evaluation of a single dose. J Neurol Sci. 2007 Jul 15;258(1–2):137–143. DOI:10.1016/j.jns.2007.03.013
  • APOKYN® (apomorphine hydrochloride injection) [prescribing information]. Louisville, KY: US WorldMeds, LLC; 2016. Accessed 27 February 2023. : https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021264s014lbl.pdf
  • Isaacson SH, Dewey RB Jr., Pahwa R, et al. How to manage the initiation of apomorphine therapy without antiemetic pretreatment: a review of the literature. Clin Park Relat Disord. 2023;8:100174. DOI:10.1016/j.prdoa.2022.100174
  • Ondo WG, Hunter C, Ferrara JM, et al. Apomorphine injections: predictors of initial common adverse events and long term tolerability. Parkinsonism Relat Disord. 2012 Jun;18(5):619–622.
  • Hauser RA, Isaacson S, Clinch T, et al. Randomized, placebo-controlled trial of trimethobenzamide to control nausea and vomiting during initiation and continued treatment with subcutaneous apomorphine injection. Parkinsonism Relat Disord. 2014 Nov;20(11):1171–1176.
  • Hattori N, Nomoto M, Study G. Sustained efficacy of apomorphine in Japanese patients with advanced Parkinson’s disease. Parkinsonism Relat Disord. 2014 Aug;20(8):819–823. doi: 10.1016/j.parkreldis.2014.04.008
  • Deffond D, Durif F, Tournilhac M. Apomorphine in treatment of Parkinson’s disease: comparison between subcutaneous and sublingual routes. J Neurol Neurosurg Psychiatry. 1993 Jan;56(1):101–103. doi: 10.1136/jnnp.56.1.101
  • Lees AJ, Montastruc JL, Turjanski N, et al. Sublingual apomorphine and Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1989 Dec;52(12):1440.
  • Hauser RA, Olanow CW, Dzyngel B, et al. Sublingual apomorphine (APL-130277) for the acute conversion of off to on in Parkinson’s disease. Mov Disord. 2016 Sep;31(9):1366–1372.
  • KYNMOBI TM (apomorphine hydrochloride) sublingual film [prescribing information]. Marlborough, MA: Sunovion Pharmaceuticals Inc.; 2022. Accessed 27 February 2023.: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2020/210875Orig1s000lbl.pdf
  • Robakis D, Fahn S. Defining the Role of the Monoamine Oxidase-B Inhibitors for Parkinson’s Disease. CNS Drugs. 2015 Jun;29(6):433–441. doi: 10.1007/s40263-015-0249-8
  • Jost WH. A critical appraisal of MAO-B inhibitors in the treatment of Parkinson’s disease. J Neural Transm (Vienna). 2022 Jun;129(5–6):723–736. doi: 10.1007/s00702-022-02465-w
  • Tabi T, Vecsei L, Youdim MB, et al. Selegiline: a molecule with innovative potential. J Neural Transm (Vienna). 2020 May;127(5):831–842.
  • Shoulson I, Oakes D, Fahn S, et al. Impact of sustained deprenyl (selegiline) in levodopa-treated Parkinson’s disease: a randomized placebo-controlled extension of the deprenyl and tocopherol antioxidative therapy of parkinsonism trial. Ann Neurol. 2002 May;51(5):604–612.
  • Eldepryl (Selegiline Hydrochloride) [prescribing information]. Tampa, FL: Somerset Pharmaceuticals, Inc; 2008. Accessed 30 January 2023. : https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/020647s006s007lbl.pdf
  • Cedarbaum JM. Clinical pharmacokinetics of anti-parkinsonian drugs. Clin Pharmacokinet. 1987 Sep;13(3):141–178. doi: 10.2165/00003088-198713030-00002
  • Lew MF. Selegiline orally disintegrating tablets for the treatment of Parkinson’s disease. Expert Rev Neurother. 2005 Nov;5(6):705–712. doi: 10.1586/14737175.5.6.705
  • ZELAPAR® (Selegiline hydrochloride) orally disintegrating tablets [prescribing information]. Bridgewater, NJ: Bausch Health US, LLC; 2021. Accessed 30 January 2023. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/021479s010lbl.pdf
  • AZILECT® (rasagiline mesylate) Tablets [prescribing information]. Kansas City, MO: Teva Neuroscience, Inc.; 2003. Accessed 10 February 2023. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/021641s002s003s004s005s007s008s010lbl.pdf
  • Thebault JJ, Guillaume M, Levy R. Tolerability, safety, pharmacodynamics, and pharmacokinetics of rasagiline: a potent, selective, and irreversible monoamine oxidase type B inhibitor. Pharmacotherapy. 2004 Oct;24(10):1295–1305. doi: 10.1592/phco.24.14.1295.43156
  • Blair HA, Dhillon S. Safinamide: a Review in Parkinson’s Disease. CNS Drugs. 2017 Feb;31(2):169–176. doi: 10.1007/s40263-017-0408-1
  • Chase TN, Bibbiani F, Oh JD. Striatal glutamatergic mechanisms and extrapyramidal movement disorders. Neurotox Res. 2003;5(1–2):139–146. doi:10.1007/BF03033378.
  • Blandini F, Porter RH, Greenamyre JT. Glutamate and Parkinson’s disease. Mol Neurobiol. 1996 Feb;12(1):73–94. doi: 10.1007/BF02740748
  • Cattaneo C, Sardina M, Bonizzoni E. Safinamide as Add-On Therapy to Levodopa in Mid- to Late-Stage Parkinson’s Disease Fluctuating Patients: post hoc Analyses of Studies 016 and SETTLE. J Parkinsons Dis. 2016;6(1):165–173. doi:10.3233/JPD-150700.
  • XADAGO (safinamide) tablets [prescribing information]. Louisville, KY: US WorldMeds, LLC; 2021. Accessed 10 February 2023. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/207145lbl.pdf
  • Borgohain R, Szasz J, Stanzione P, et al. Two-year, randomized, controlled study of safinamide as add-on to levodopa in mid to late Parkinson’s disease. Mov Disord. 2014 Sep;29(10):1273–1280.
  • Aboukarr A, Giudice M. Interaction between Monoamine Oxidase B Inhibitors and Selective Serotonin Reuptake Inhibitors. Can J Hosp Pharm. 2018 May;71(3):196–207. doi: 10.4212/cjhp.v71i3.2586
  • Edinoff AN, Swinford CR, Odisho AS, et al. Clinically Relevant Drug Interactions with Monoamine Oxidase Inhibitors. Health Psychol Res. 2022;10(4):39576. DOI:10.52965/001c.39576
  • Kurth MC, Adler CH. COMT inhibition: a new treatment strategy for Parkinson’s disease. Neurology. 1998 May;50(5 Suppl 5):S3–14. doi: 10.1212/WNL.50.5_Suppl_5.S3
  • Artusi CA, Sarro L, Imbalzano G, et al. Safety and efficacy of tolcapone in Parkinson’s disease: systematic review. Eur J Clin Pharmacol. 2021 Jun;77(6):817–829.
  • TASMAR® (tolcapone) tablets [prescribing information]. Cost Mesa, CA: Valeant Pharmaceuticals International; 2006. Accessed 28 January 2023. https://www.accessdata.fda.gov/drugsatfda_docs/label/2007/020697s012lbl.pdf
  • Sedek G, Jorga K, Schmitt M, et al. Effect of tolcapone on plasma levodopa concentrations after coadministration with levodopa/carbidopa to healthy volunteers. Clin Neuropharmacol. 1997 Dec;20(6):531–541.
  • Fabbri M, Ferreira JJ, Rascol O. COMT Inhibitors in the Management of Parkinson’s Disease. CNS Drugs. 2022 Mar;36(3):261–282. doi: 10.1007/s40263-021-00888-9
  • Olanow CW. Tolcapone and hepatotoxic effects. Tasmar Advisory Panel. Arch Neurol. 2000 Feb;57(2):263–267. doi: 10.1001/archneur.57.2.263
  • Olanow CW, Watkins PB. Tolcapone: an efficacy and safety review (2007). Clin Neuropharmacol. 2007 Sep;30(5):287–294. doi: 10.1097/wnf.0b013e318038d2b6
  • Li J, Lou Z, Liu X, et al. Efficacy and Safety of Adjuvant Treatment with Entacapone in Advanced Parkinson’s Disease with Motor Fluctuation: a Systematic Meta-Analysis. Eur Neurol. 2017;78(3–4):143–153. DOI:10.1159/000479555
  • COMTAN® (entacapone) Tablets [prescribing information]. East Hanover, NJ: Novartis Pharmaceuticals; 2010. Accessed 28 January 2023. https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/020796s15lbl.pdf
  • Stalevo® (carbidopa, levodopa and entacapone) Tablets [prescribing information]. East Hanover, NJ: Novartis Pharmacy Corporation. 2010. [cited 2023 Mar 10]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/021485s20lbl.pdf
  • Ongentys (opicapone) capusles [prescribing information]. San Diego, CA: Neurocrine Biosciences, Inc; 2020. [cited 2023 Jan 30]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/212489s000lbl.pdf
  • Rocha JF, Santos A, Falcao A, et al. Effect of moderate liver impairment on the pharmacokinetics of opicapone. Eur J Clin Pharmacol. 2014 Mar;70(3):279–286.
  • Castro Caldas A, Teodoro T, Ferreira JJ. The launch of opicapone for Parkinson’s disease: negatives versus positives. Expert Opin Drug Saf. 2018 Mar;17(3):331–337. doi: 10.1080/14740338.2018.1433659
  • Jackson GG, Muldoon RL, Akers LW. Serological Evidence for Prevention of Influenzal Infection in Volunteers by an Anti-Influenzal Drug Adamantanamine Hydrochloride. Antimicrob Agents Chemother (Bethesda). 1963;161:703–707.
  • Wendel HA, Snyder MT, Pell S. Trial of amantadine in epidemic influenza. Clin Pharmacol Ther. 1966 Jan;7(1):38–43. doi: 10.1002/cpt19667138
  • Togo Y, Hornick RB, Dawkins AT Jr. Studies on induced influenza in man. I. Double-blind studies designed to assess prophylactic efficacy of amantadine hydrochloride against a2/Rockville/1/65 strain. JAMA. 1968 Mar 25;203(13):1089–1094. doi: 10.1001/jama.1968.03140130001001
  • Schwab RS, England AC Jr., Poskanzer DC, et al. Amantadine in the treatment of Parkinson’s disease. JAMA. 1969 May 19;208(7):1168–1170. DOI:10.1001/jama.1969.03160070046011
  • Rascol O, Goetz C, Koller W, et al. Treatment interventions for Parkinson’s disease: an evidence based assessment. Lancet. 2002 May 4;359(9317):1589–1598. DOI:10.1016/S0140-6736(02)08520-3
  • Rascol O, Fabbri M, Poewe W. Amantadine in the treatment of Parkinson’s disease and other movement disorders. Lancet Neurol. 2021 Dec;20(12):1048–1056. doi: 10.1016/S1474-4422(21)00249-0
  • Danysz W, Dekundy A, Scheschonka A, et al. Amantadine: reappraisal of the timeless diamond-target updates and novel therapeutic potentials. J Neural Transm (Vienna). 2021 Feb;128(2):127–169.
  • Butzer JF, Silver DE, Sahs AL. Amantadine in Parkinson’s disease. A double-blind, placebo-controlled, crossover study with long-term follow-up. Neurology. 1975 Jul;25(7):603–606. doi: 10.1212/WNL.25.7.603
  • Crosby N, Deane KH, Clarke CE. Amantadine in Parkinson’s disease. Cochrane Database Syst Rev. 2003;2003(1):CD003468. doi:10.1002/14651858.CD003468.
  • Bauer RB, McHenry JT. Comparison of amantadine, placebo, and levodopa in Parkinson’s disease. Neurology. 1974 Aug;24(8):715–720. doi: 10.1212/WNL.24.8.715
  • Kornhuber J, Bormann J, Hubers M, et al. Effects of the 1-amino-adamantanes at the MK-801-binding site of the NMDA-receptor-gated ion channel: a human postmortem brain study. Eur J Pharmacol. 1991 Apr 25;206(4):297–300. DOI:10.1016/0922-4106(91)90113-V
  • Kornhuber J, Bormann J, Retz W, et al. Memantine displaces [3H]MK-801 at therapeutic concentrations in postmortem human frontal cortex. Eur J Pharmacol. 1989 Aug 3;166(3):589–590. DOI:10.1016/0014-2999(89)90384-1
  • Metman LV, Del Dotto P, LePoole K, et al. Amantadine for levodopa-induced dyskinesias: a 1-year follow-up study. Arch Neurol. 1999 Nov;56(11):1383–1386.
  • Snow BJ, Macdonald L, McAuley D, et al. The effect of amantadine on levodopa-induced dyskinesias in Parkinson’s disease: a double-blind, placebo-controlled study. Clin Neuropharmacol. 2000 Mar;23(2):82–85.
  • Luginger E, Wenning GK, Bosch S, et al. Beneficial effects of amantadine on L-dopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2000 Sep;15(5):873–878.
  • SYMMETREL® (Amantadine Hydrochloride, USP) Tablets and Syrup [prescribing information]. Chadds Ford, PA: Endo Pharmaceuticals, Inc., 2002. [cited 2023 Jan 23]. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2003/016023_S037_SYMMETREL%20SYRUP%20AND%20TABLETS_PRNTLBL.pdf
  • Aoki FY, Sitar DS. Clinical pharmacokinetics of amantadine hydrochloride. Clin Pharmacokinet. 1988 Jan;14(1):35–51. doi: 10.2165/00003088-198814010-00003
  • Horadam VW, Sharp JG, Smilack JD, et al. Pharmacokinetics of amantadine hydrochloride in subjects with normal and impaired renal function. Ann Intern Med. 1981 Apr;94(4 pt 1):454–458.
  • GOCOVRI TM (amantadine) extended release capsules [prescribing information]. Emeryville, CA: Adamas Pharma LLC; 2021. [cited 2023 Jan 23]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/208944lbl.pdf
  • Hauser RA, Pahwa R, Wargin WA, et al. Pharmacokinetics of ADS-5102 (Amantadine) Extended Release Capsules Administered Once Daily at Bedtime for the Treatment of Dyskinesia. Clin Pharmacokinet. 2019 Jan;58(1):77–88.
  • Devries T, Dentiste A, Handiwala L, et al. Bioavailability and Pharmacokinetics of Once-Daily Amantadine Extended-Release Tablets in Healthy Volunteers: results from Three Randomized, Crossover, Open-Label, Phase 1 Studies. Neurol Ther. 2019;8(2):449–460. DOI:10.1007/s40120-019-0144-1
  • Walker JE, Albers JW, Tourtellotte WW, et al. A qualitative and quantitative evaluation of amantadine in the treatment of Parkinson’s disease. J Chronic Dis. 1972 Mar;25(3):149–182.
  • Walker JE, Potvin A, Tourtellotte W, et al. Amantadine and levodopa in the treatment of Parkinson’s disease. Clin Pharmacol Ther. 1972 Jan;13(1):28–36.
  • Savery F. Amantadine and a fixed combination of levodopa and carbidopa in the treatment of Parkinson’s disease. Dis Nerv Syst. 1977 Aug;38(8):605–608.
  • Kong M, Ba M, Ren C, et al. An updated meta-analysis of amantadine for treating dyskinesia in Parkinson’s disease. Oncotarget. 2017 Aug 22;8(34):57316–57326. DOI:10.18632/oncotarget.17622
  • Collins LE, Galtieri DJ, Collins P, et al. Interactions between adenosine and dopamine receptor antagonists with different selectivity profiles: effects on locomotor activity. Behav Brain Res. 2010 Aug 25;211(2):148–155. DOI:10.1016/j.bbr.2010.03.003
  • Bennett KA, Tehan B, Lebon G, et al. Pharmacology and structure of isolated conformations of the adenosine A(2)A receptor define ligand efficacy. Mol Pharmacol. 2013 May;83(5):949–958.
  • Perez V, Sosti V, Rubio A, et al. Modulation of the motor response to dopaminergic drugs in a parkinsonian model of combined dopaminergic and noradrenergic degeneration. Eur J Pharmacol. 2007 Dec 8;576(1–3):83–90. DOI:10.1016/j.ejphar.2007.08.024
  • NOURIANZ™ (istradefylline) tablets [prescribing information]. Bedminster, NJ: Kyowa Kirin, Inc.; 2020. [cited 2023 Jan 20]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/022075s000lbl.pdf
  • Mukai M, Uchimura T, Zhang X, et al. Effects of Rifampin on the Pharmacokinetics of a Single Dose of Istradefylline in Healthy Subjects. J Clin Pharmacol. 2018 Feb;58(2):193–201.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.