511
Views
19
CrossRef citations to date
0
Altmetric
Drug Profile

Optimizing levodopa therapy to treat wearing-off symptoms in Parkinson’s disease: focus on levodopa/carbidopa/entacapone

&
Pages 119-131 | Published online: 09 Jan 2014

References

  • Karlsen KH, Tandberg E, Arsland D, Larsen JP. Health related quality of life in Parkinson’s disease: a prospective longitudinal study. J. Neurol. Neurosurg. Psychiatry69(5), 584–589 (2000).
  • De Rijk MC, Launer LJ, Berger K et al. Prevalence of Parkinson’s disease in Europe: a collaborative study of population-based cohorts. Neurologic Diseases in the Elderly Research Group. Neurology54(11 Suppl. 5), S21–S23 (2000).
  • Rajput AH. Frequency and cause of Parkinson’s disease. Can J. Neurol. Sci.19(1 Suppl.), 103–107 (1992).
  • Samii A, Nutt JG, Ransom BR. Parkinson’s disease. Lancet363(9423), 1783–1793 (2004).
  • Golbe LI. Young-onset Parkinson’s disease: a clinical review. Neurology41(2 Pt 1), 168–173 (1991).
  • Lang AE, Lozano AM. Parkinson’s disease. Second of two parts. N. Engl. J. Med.339(16), 1130–1143 (1998).
  • Obeso JA, Rodriguez-Oroz MC, Rodriguez M et al. Pathophysiology of the basal ganglia in Parkinson’s disease. Trends Neurosci.23(10 Suppl.), S8–S19 (2000).
  • Vingerhoets FJ, Snow BJ, Lee CS, Schulzer M, Mak E, Calne DB. Longitudinal fluorodopa positron emission tomographic studies of the evolution of idiopathic parkinsonism. Ann. Neurol.36(5), 759–764 (1994).
  • Fearnley JM, Lees AJ. Ageing and Parkinson’s disease: substantia nigra regional selectivity. Brain114(Pt 5), 2283–2301 (1991).
  • Olanow CW, Stern MB, Sethi K. The scientific and clinical basis for the treatment of Parkinson disease (2009). Neurology72(21 Suppl. 4), S1–S136 (2009).
  • Benamer TS, Patterson J, Grosset DG et al. Accurate differentiation of parkinsonism and essential tremor using visual assessment of [123I]-FP-CIT SPECT imaging: the [123I]-FP-CIT study group. Mov. Disord.15(3), 503–510 (2000).
  • Druschky A, Hilz MJ, Platsch G et al. Differentiation of Parkinson’s disease and multiple system atrophy in early disease stages by means of I-123-MIBG-SPECT. J. Neurol. Sci.175(1), 3–12 (2000).
  • Gerschlager W, Bencsits G, Pirker W et al. [123I]beta-CIT SPECT distinguishes vascular parkinsonism from Parkinson’s disease. Mov. Disord.17(3), 518–523 (2002).
  • Schapira AH, Obeso J. Timing of treatment initiation in Parkinson’s disease: a need for reappraisal? Ann. Neurol.59(3), 559–562 (2006).
  • Brooks DJ. Optimizing levodopa therapy for Parkinson’s disease with levodopa/carbidopa/entacapone: implications from a clinical and patient perspective. Neuropsychiatr. Dis. Treat.4(1), 39–47 (2008).
  • Schapira AH, Emre M, Jenner P, Poewe W. Levodopa in the treatment of Parkinson’s disease. Eur. J. Neurol.16(9), 982–989 (2009).
  • Poewe W, Antonini A, Zijlmans JCM, Burkhard PR, Vingerhoetz F. Levodopa in the treatment of Parkinson’s disease: an old drug still going strong. Clin. Interv. Aging5, 229–238 (2010).
  • Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov. Disord.16(3), 448–458 (2001).
  • Hattori N, Fujimoto K, Kondo T, Murata M, Stacy M. Patient perspectives towards levodopa therapy in Japan and the United States – results of two patient surveys. Mov. Disord.25(Suppl. 3), S569–S717 (2010).
  • Stocchi F. The levodopa wearing-off phenomenon in Parkinson’s disease: pharmacokinetic considerations. Expert Opin. Pharmacother.7(10), 1399–1407 (2006).
  • Olanow CW, Agid Y, Mizuno Y et al. Levodopa in the treatment of Parkinson’s disease: current controversies. Mov. Disord.19(9), 997 (2004).
  • Cotzias GC, Van Woert MH, Schiffer LM. Aromatic amino acids and modification of parkinsonism. N. Engl. J. Med.276(7), 374–379 (1967).
  • Birkmayer W, Hornykiewics O. The effect of l-3,4-dihydroxyphenlyalanine (=DOPA) on akinesia in parkinsonism. Parkinsonism Relat. Disord.4(2), 59–60 (1998).
  • Rajput AH. Levodopa prolongs life expectancy and is non-toxic to substantia nigra. Parkinsonism Relat. Disord.8(2), 95–100 (2001).
  • Rinne UK, Bracco F, Chouza C et al. Early treatment of Parkinson’s disease with cabergoline delays the onset of motor complications. Results of a double-blind levodopa controlled trial. The PKDS009 Study Group. Drugs55(Suppl. 1), 23–30 (1998).
  • Holloway RG, Shoulson I, Fahn S et al. Pramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. Arch. Neurol.61(7), 1044–1053 (2004).
  • Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: a randomised controlled trial. JAMA284(15), 1931–1938 (2000).
  • Hauser RA, Rascol O, Korczyn AD et al. Ten-year follow-up of Parkinson’s disease patients randomized to initial therapy with ropinirole or levodopa. Mov. Disord.22(16), 2409–2417 (2007).
  • Parkinson’s Disease Research Group in the United Kingdom. Comparisons of therapeutic effects of levodopa, levodopa and selegiline, and bromocriptine in patients with early, mild Parkinson’s disease: three year interim report. BMJ307, 469–472 (1993).
  • Oertel WH, Wolters E, Sampaio C et al. Pergolide versus levodopa monotherapy in early Parkinson’s disease patients: the PELMOPET study. Mov. Disord.21(3), 343–353 (2006).
  • Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. 056 Study Group. N. Engl. J. Med.342(20), 1484–1491 (2000).
  • Parkinson Study Group. Long-term effect of initiating pramipexole vs levodopa in early Parkinson disease. Arch. Neurol.66(5), 563–570 (2009).
  • Hauser RA, Holford NH. Quantitative description of loss of clinical benefit following withdrawal of levodopa–carbidopa and bromocriptine in early Parkinson’s disease. Mov. Disord.17(5), 961–968 (2002).
  • Hauser RA, Koller WC, Hubble JP, Malapira T, Busenbark K, Olanow CW. Time course of loss of clinical benefit following withdrawal of levodopa/carbidopa and bromocriptine in early Parkinson’ s disease. Mov. Disord.15(3), 485–489 (2000).
  • Obeso JA, Rodriguez-Oroz MC, Chana P, Lera G, Rodriguez M, Olanow CW. The evolution and origin of motor complications in Parkinson’s disease. Neurology55(11 Suppl. 4), S13–S20 (2000).
  • Chapuis S, Ouchchane L, Metz O, Gerbaud L, Durif F. Impact of the motor complications of Parkinson’s disease on the quality of life. Mov. Disord.20(2), 224–230 (2005).
  • Olanow CW, Watts RL, Koller WC. An algorithm (decision tree) for the management of Parkinson’s disease (2001): treatment guidelines. Neurology56(11 Suppl. 5), S1–S88 (2001).
  • Parkinson Study Group. Levodopa and the progression of Parkinson’s disease. N. Engl. J. Med.351(24), 2498–2508 (2004).
  • Katzenschlager R, Head J, Schrag A, Ben-Shlomo Y, Evans A, Lees AJ. Fourteen-year final report of the randomized PDRG-UK trial comparing three initial treatments in PD. Neurology71(7), 474–480 (2008).
  • Nyholm D. Pharmacokinetic optimisation in the treatment of Parkinson’s disease: an update. Clin. Pharmacokinet.45(2), 109–136 (2006).
  • Pahwa R, Lyons KE. Levodopa-related wearing-off in Parkinson’s disease: identification and management. Curr. Med. Res. Opin.25(4), 841–849 (2009).
  • Oertel WH, Berardelli A, Bloem BR et al. Late (complicated) Parkinson’s disease. In: European Handbook of Neurological Management. Gilhus NE, Barnes MR, Brainin M (Eds). Wiley-Blackwell, Hoboken, NJ, USA, 584 (2010).
  • Kuoppamäki M, Korpela K, Marttila R et al. Comparison of pharmacokinetic profile of levodopa throughout the day between levodopa/carbidopa/entacapone and levodopa/carbidopa when administered four- or five-times daily. Eur. J. Clin. Pharmacol.65(5), 443–455 (2009).
  • Nutt JG, Carter JH, Lea ES, Woodward WR. Motor fluctuations during continuous levodopa infusions in patients with Parkinson’s disease. Mov. Disord.12(3), 285–292 (1997).
  • Khor SP, Hsu A. The pharmacokinetics and pharmacodynamics of levodopa in the treatment of Parkinson’s disease. Curr. Clin. Pharmacol.2(3), 234–243 (2007).
  • Nutt JG, Woodward WR, Beckner RM et al. Effect of peripheral catechol-O-methyltransferase inhibition on the pharmacokinetics and pharmacodynamics of levodopa in parkinsonian patients. Neurology44(5), 913–919 (1994).
  • Myllylä VV, Sotaniemi KA, Illi A, Suominen K, Keranen T. Effect of entacapone, a COMT inhibitor, on the pharmacokinetics of levodopa and on cardiovascular responses in patients with Parkinson’s disease. Eur. J. Clin. Pharmacol.45, 419–423 (1993).
  • Keränen T, Gordin A, Harjola VP et al. The effect of catechol-O-methyltransferase inhibition by entacapone on the pharmacokinetics and metabolism of levodopa in healthy volunteers. Clin. Neuropharmacol.16(2), 145–156 (1993).
  • Kaakkola S, Gordin A, Mannisto PT. General properties and clinical possibilities of new selective inhibitors of catechol O-methyltransferase. Gen. Pharmacol.25(5), 813–824 (1994).
  • Merello M, Lees AJ, Webster R, Bovingdon M, Gordin A. Effect of entacapone, a peripherally acting catechol-O-methyltransferase inhibitor, on the motor response to acute treatment with levodopa in patients with Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry57, 186–189 (1994).
  • 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.19(4), 283–296 (1996).
  • Sawle GV, Burn DJ, Morrish PK et al. The effect of entacapone (OR-611) on brain [18F]-6-L-fluorodopa metabolism: implications for levodopa therapy of Parkinson’s disease. Neurology44(7), 1292–1297 (1994).
  • Kuoppamäki M, Sauramo A, Korpela K et al. Night-time bioavailability of levodopa/carbidopa/entacapone is higher compared with controlled-release levodopa/carbidopa. Int. J. Clin. Pharmacol. Ther.48(11), 756–760 (2010 ).
  • Rinne UK, Larsen JP, Siden A, Worm-Petersen J. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations. NOMECOMT Study Group. Neurology51(5), 1309–1314 (1998).
  • Poewe WH, Deuschl G, Gordin A, Kultalahti ER, Leinonen M. Efficacy and safety of entacapone in Parkinson’s disease patients with suboptimal levodopa response: a 6-month randomized placebo-controlled double-blind study in Germany and Austria (Celomen study). Acta Neurol. Scand.105(4), 245–255 (2002).
  • Brooks DJ, Sagar H. Entacapone is beneficial in both fluctuating and non-fluctuating patients with Parkinson’s disease: a randomised, placebo controlled, double blind, six month study. J. Neurol. Neurosurg. Psychiatry74(8), 1071–1079 (2003).
  • Parkinson Study Group. Entacapone improves motor fluctuations in levodopa-treated Parkinson’s disease patients. Ann. Neurol.42(5), 747–755 (1997).
  • Deane KH, Spieker S, Clarke CE. Catechol-O-methyltransferase inhibitors for levodopa-induced complications in Parkinson’s disease. Cochrane Database Syst. Rev. (4), CD004554 (2004).
  • Larsen JP, Worm-Petersen J, Siden A, Gordin A, Reinikainen K, Leinonen M. The tolerability and efficacy of entacapone over 3 years in patients with Parkinson’s disease. Eur. J. Neurol.10(2), 137–146 (2003).
  • Onofrj M, Thomas A, Vingerhoets F et al. Combining entacapone with levodopa/DDCI improves clinical status and quality of life in Parkinson’s Disease (PD) patients experiencing wearing-off, regardless of the dosing frequency: results of a large multicentre open-label study. J. Neural. Transm.111(8), 1053–1063 (2004).
  • Nissinen H, Kuoppamaki M, Leinonen M, Schapira AH. Early versus delayed initiation of entacapone in levodopa-treated patients with Parkinson’s disease: a long-term, retrospective analysis. Eur. J. Neurol.16(12), 1305–1311 (2009).
  • Eggert K, Skogar O, Amar K et al. Direct switch from levodopa/benserazide or levodopa/carbidopa to levodopa/carbidopa/entacapone in Parkinson’s disease patients with wearing-off: efficacy, safety and feasibility – an open-label, 6-week study. J. Neural. Transm.117(3), 333–342 (2010).
  • Koller W, Guarnieri M, Hubble J, Rabinowicz Al, Silver D. An open-label evaluation of the tolerability and safety of Stalevo® (carbidopa, levodopa and entacapone) in Parkinson’s disease patients experiencing wearing-off. J. Neural. Transm.112(2), 221–230 (2005).
  • Hauser RA, Panisset M, Abbruzzese G, Mancione L, Dronamraju N, Kakarieka A. Double-blind trial of levodopa/carbidopa/entacapone versus levodopa/carbidopa in early Parkinson’s disease. Mov. Disord.24(4), 541–550 (2009).
  • Stocchi F, Rascol O, Kieburtz K et al. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: the STRIDE-PD study. Ann. Neurol.68(1), 18–27 (2010).
  • Brooks DJ, Leinonen M, Kuoppamaki M, Nissinen H. Five-year efficacy and safety of levodopa/DDCI and entacapone in patients with Parkinson’s disease. J. Neural. Transm.115(6), 843–849 (2008).
  • Deuschl G, Vaitkus A, Fox GC, Roscher T, Schremmer D, Gordin A. Efficacy and tolerability of entacapone versus cabergoline in parkinsonian patients suffering from wearing-off. Mov. Disord.22(11), 1550–1555 (2007).
  • Brooks DJ. Safety and tolerability of COMT inhibitors. Neurology62(1 Suppl. 1), S39–S46 (2004).
  • Myllylä V, Haapaniemi T, Kaakkola S et al. Patient satisfaction with switching to Stalevo: an open-label evaluation in PD patients experiencing wearing-off (Simcom Study). Acta Neurol. Scand.114(3), 181–186 (2006).
  • Linazasoro G, Kulisevsky J, Hernandez B. Should levodopa dose be reduced when switched to Stalevo? Eur. J. Neurol.15(3), 257–261 (2008).
  • Grosset D, Antonini A, Canesi M et al. Adherence to antiparkinson medication in a multicenter European study. Mov. Disord.24(6), 826–832 (2009).
  • Ingman K, Naukkarinen T, Vahteristo M, Korpela I, Kuoppamäki M, Ellmén J. The effect of different dosing regimens of levodopa/carbidopa/entacapone on plasma levodopa concentrations. Eur. J. Clin. Pharmacol. doi:10.1007/s00228-011-1121-5 (2011) (Epub ahead of print).
  • Sethi KD, Hauser RA, Isaacson SH, Mcclain T. Levodopa/carbidopa/entacapone 200/50/200 mg (Stalevo 200) in the treatment of Parkinson’s disease: a case series. Cases J.2, 7134 (2009).
  • Findley L, Aujla M, Bain PG et al. Direct economic impact of Parkinson’s disease: a research survey in the United Kingdom. Mov. Disord.18(10), 1139–1145 (2003).
  • Dengler I, Leukel N, Meuser T, Jost WH. [Prospective study of the direct and indirect costs of idiopathic Parkinson’s disease]. Der. Nervenarzt.77(10), 1204–1209 (2006).
  • Keranen T, Kaakkola S, Sotaniemi K et al. Economic burden and quality of life impairment increase with severity of PD. Parkinsonism Relat. Disord.9(3), 163–168 (2003).
  • Dodel RC, Berger K, Oertel WH. Health-related quality of life and healthcare utilisation in patients with Parkinson’s disease: impact of motor fluctuations and dyskinesias. Pharmacoeconomics19(10), 1013–1038 (2001).
  • Findley LJ, Lees A, Apajasalo M, Pitkanen A, Turunen H. Cost-effectiveness of levodopa/carbidopa/entacapone (Stalevo) compared with standard care in UK Parkinson’s disease patients with wearing-off. Curr. Med. Res. Opin.21(7), 1005–1014 (2005).
  • Palmer CS, Nuijten MJ, Schmier JK, Subedi P, Snyder EH. Cost effectiveness of treatment of Parkinson’s disease with entacapone in the United States. Pharmacoeconomics20(9), 617–628 (2002).
  • Delea TE, Thomas SK, Hagiwara M. The association between adherence to levodopa/carbidopa/entacapone therapy and healthcare utilization and costs among patients with Parkinson’s disease: a retrospective claims-based analysis. CNS Drugs25(1), 53–66 (2010).
  • Hauser RA, Ellenbogen AL, Metman LV et al. Crossover comparison of IPX066 and a standard levodopa formulation in advanced Parkinson’s disease. Mov. Disord.26(12), 2246–2252 (2011).
  • Kiss LE, Ferreira HS, Torrao L et al. Discovery of a long-acting, peripherally selective inhibitor of catechol-O-methyltransferase. J. Med. Chem.53(8), 3396–3411 (2010).
  • Kaakkola S. Problems with the present inhibitors and a relevance of new and improved COMT inhibitors in Parkinson’s disease. Int. Rev. Neurobiol.95, 207–225 (2010).
  • Morelli M, Carta AR, Jenner P. Adenosine A2A receptors and Parkinson’s disease. Handb. Exp. Pharmacol. (193), 589–615 (2009).
  • Sommer DB, Stacy MA. What’s in the pipeline for the treatment of Parkinson’s disease? Expert Rev. Neurother.8(12), 1829–1839 (2008).
  • Quik M, Bordia T, Huang L, Perez X. Targeting nicotinic receptors for parkinson’s disease therapy. CNS Neurol. Disord. Drug Targets.10(6), 651–658 (2011).
  • Rangasamy SB, Soderstrom K, Bakay RA, Kordower JH. Neurotrophic factor therapy for Parkinson’s disease. Prog. Brain Res.184, 237–264 (2010).
  • Witt J, Marks WJ Jr. An update on gene therapy in Parkinson’s disease. Curr. Neurol. Neurosci. Rep.11(4), 362–370 (2011).
  • Hilker R, Brotchie JM, Chapman J. Pros and cons of a prion-like pathogenesis in Parkinson’s disease. BMC Neurology11, 74 (2011).
  • Olanow CW, Mcnaught K. Parkinson’s disease, proteins, and prions: milestones. Mov. Disord.26(6), 1056–1071 (2011).

Website

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.