227
Views
36
CrossRef citations to date
0
Altmetric
Review

Treatment of advanced Parkinson’s disease

&
Pages 1181-1197 | Published online: 10 Jan 2014

References

  • Parkinson J. An essay on the shaking palsy. Medical Classics10, 964–967 (1817).
  • Cotzias GC, Van Woert MH, Schiffer LM. Aromatic amino acids and modification of parkinsonism. N. Engl. J Med.276(7), 374–379 (1967).
  • Jankovic J. Progression of Parkinson disease: are we making progress in charting the course? Arch. Neurol.62(3), 351–352 (2005).
  • Melamed E. Early-morning dystonia. A late side effect of long-term levodopa therapy in Parkinson’s disease. Arch. Neurol.36(5), 308–310 (1979).
  • Currie LJ, Harrison MB, Trugman JM, Bennett JP Jr, Wooten GF. Early morning dystonia in Parkinson’s disease. Neurology51(1), 283–285 (1998).
  • Hely MA, Morris JG, Reid WG et al. The Sydney Multicentre Study of Parkinson's disease: a randomised, prospective five-year study comparing low dose bromocriptine with low dose levodopa-carbidopa. J. Neurol. Neurosurg. Psychiatry57(8), 903–910 (1994).
  • Hely MA, Morris JG, Reid WG, Trafficante R. Sydney Multicenter Study of Parkinson’s disease: non-L-dopa-responsive problems dominate at 15 years. Mov. Disord.20(2), 190–199 (2005).
  • Marras C, Lang A, Krahn M, Tomlinson G, Naglie G. Quality of life in early Parkinson’s disease: impact of dyskinesias and motor fluctuations. Mov. Disord.19(1), 22–28 (2004).
  • Van Gerpen JA, Kumar N, Bower JH, Weigand S, Ahlskog JE. Levodopa-associated dyskinesia risk among Parkinson disease patients in Olmsted County, Minnesota, 1976–1990. Arch. Neurol.63(2), 205–209 (2006).
  • Diamond A, Jankovic J. The effect of deep brain stimulation on quality of life in movement disorders. J. Neurol. Neurosurg. Psychiatry76(9), 1188–1193 (2005).
  • Vidailhet M, Bonnet AM, Marconi R, Durif F, Agid Y. The phenomenology of L-dopa-induced dyskinesias in Parkinson’s disease. Mov. Disord.14(Suppl. 1), 13–18 (1999).
  • Jankovic J. Motor fluctuations and dyskinesias in Parkinson’s disease: clinical manifestations. Mov. Disord.20(Suppl. 11), S11–S16 (2005).
  • Fuente-Fernandez R, Sossi V, Huang Z et al. Levodopa-induced changes in synaptic dopamine levels increase with progression of Parkinson’s disease: implications for dyskinesias. Brain127(Pt 12), 2747–2754 (2004).
  • Lhermitte F, Agid Y, Feuerstein C et al. Abnormal movements caused by L-DOPA in patients with Parkinson’s disease: correlation with the plasma concentrations of DOPA and O-methyl-DOPA. Rev. Neurol.133(8–9), 445–454 (1977).
  • Lhermitte F, Agid Y, Signoret JL. Onset and end-of-dose levodopa-induced dyskinesias. Possible treatment by increasing the daily doses of levodopa Arch. Neurol.35(5), 261–263 (1978).
  • Olanow CW, Jankovic J. Neuroprotective therapy in Parkinson’s disease and motor complications: a search for a pathogenesis-targeted, disease-modifying strategy. Mov. Disord.20(Suppl. 11), S3–S10 (2005).
  • 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).
  • Fahn S, Oakes D, Shoulson I et al. Levodopa and the progression of Parkinson’s disease. N. Engl. J Med.351(24), 2498–2508 (2004).
  • Parkinson Study Group. Impact of deprenyl and tocopherol treatment on Parkinson’s disease in DATATOP patients requiring levodopa. Ann. Neurol.39(1), 37–45 (1996).
  • Braak H, Ghebremedhin E, Rub U, Bratzke H, Del Tredici K. Stages in the development of Parkinson’s disease-related pathology. Cell Tissue Res.318(1), 121–134 (2004).
  • Olanow CW, Agid Y, Mizuno Y et al. Levodopa in the treatment of Parkinson’s disease: current controversies. Mov. Disord.19(9), 997–1005 (2004).
  • Filion M, Tremblay L. Abnormal spontaneous activity of globus pallidus neurons in monkeys with MPTP-induced parkinsonism. Brain Res.547(1), 142–151 (1991).
  • Gerfen CR. D1 dopamine receptor supersensitivity in the dopamine-depleted striatum animal model of Parkinson’s disease. Neuroscientist9(6), 455–462 (2003).
  • Morissette M, Goulet M, Soghomonian JJ et al. Preproenkephalin mRNA expression in the caudate-putamen of MPTP monkeys after chronic treatment with the D2 agonist U91356A in continuous or intermittent mode of administration: comparison with L-DOPA therapy. Brain Res. Mol. Brain Res.49(1–2), 55–62 (1997).
  • Goulet M, Morissette M, Calon F et al. Continuous or pulsatile chronic D2 dopamine receptor agonist (U91356A) treatment of drug-naive 4-phenyl-1,2,3,6-tetrahydropyridine monkeys differentially regulates brain D1 and D2 receptor expression: in situ hybridization histochemical analysis. Neuroscience79(2), 497–507 (1997).
  • Nutt JG, Woodward WR, Hammerstad JP, Carter JH, Anderson JL. The “on-off” phenomenon in Parkinson’s disease. Relation to levodopa absorption and transport. N. Engl. J Med.310(8), 483–488 (1984).
  • Grosset KA, Reid JL, Grosset DG. Medicine-taking behavior: implications of suboptimal compliance in Parkinson’s disease. Mov. Disord.20(11), 1397–1404 (2005).
  • Koller WC, Pahwa R. Treating motor fluctuations with controlled-release levodopa preparations. Neurology44(7 Suppl. 6), S23–S28 (1994).
  • Pahwa R, Factor SA, Lyons KE et al. Practice parameter: treatment of Parkinson disease with motor fluctuations and dyskinesia (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology66(7), 983–995 (2006).
  • Pahwa R, Busenbark K, Huber SJ et al. Clinical experience with controlled-release carbidopa/levodopa in Parkinson’s disease. Neurology43(4), 677–681 (1993).
  • Pahwa R, Lyons K, McGuire D et al. Early morning akinesia in Parkinson’s disease: effect of standard carbidopa/levodopa and sustained-release carbidopa/levodopa. Neurology46(4), 1059–1062 (1996).
  • Nilsson D, Hansson LE, Johansson K et al. Long-term intraduodenal infusion of a water based levodopa–carbidopa dispersion in very advanced Parkinson’s disease. Acta Neurol. Scand.97(3), 175–183 (1998).
  • Nilsson D, Nyholm D, Aquilonius SM. Duodenal levodopa infusion in Parkinson’s disease-long-term experience. Acta Neurol. Scand.104(6), 343–348 (2001).
  • Heikkinen H, Nutt JG, LeWitt PA, Koller WC, Gordin A. The effects of different repeated doses of entacapone on the pharmacokinetics of L-Dopa and on the clinical response to L-Dopa in Parkinson’s disease. Clin. Neuropharmacol.24(3), 150–157 (2001).
  • 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).
  • Kaakkola S. Clinical pharmacology, therapeutic use and potential of COMT inhibitors in Parkinson’s disease. Drugs59(6), 1233–1250 (2000).
  • Ahtila S, Kaakkola S, Gordin A et al. Effect of entacapone, a COMT inhibitor, on the pharmacokinetics and metabolism of levodopa after administration of controlled-release levodopa-carbidopa in volunteers. Clin. Neuropharmacol.18(1), 46–57 (1995).
  • Piccini P, Brooks DJ, Korpela K et al. The catechol-O-methyltransferase (COMT) inhibitor entacapone enhances the pharmacokinetic and clinical response to Sinemet CR in Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry68(5), 589–594 (2000).
  • Najib J. Entacapone: a catechol-O-methyltransferase inhibitor for the adjunctive treatment of Parkinson’s disease. Clin. Ther.23(6), 802–832 (2001).
  • Brooks DJ, Agid Y, Eggert K et al. Treatment of end-of-dose wearing-off in Parkinson’s disease: stalevo (levodopa/carbidopa/entacapone) and levodopa/DDCI given in combination with Comtess/Comtan (entacapone) provide equivalent improvements in symptom control superior to that of traditional levodopa/DDCI treatment. Eur. Neurol.53(4), 197–202 (2005).
  • 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).
  • 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. Lancet365(9463), 947–954 (2005).
  • 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).
  • Roberts JW, Cora-Locatelli G, Bravi D et al. Catechol-O-methyltransferase inhibitor tolcapone prolongs levodopa/carbidopa action in parkinsonian patients. Neurology43(12), 2685–2688 (1993).
  • Kurth MC, Adler CH, Hilaire MS et al. Tolcapone improves motor function and reduces levodopa requirement in patients with Parkinson’s disease experiencing motor fluctuations: a multicenter, double-blind, randomized, placebo-controlled trial. Tolcapone Fluctuator Study Group I. Neurology48(1), 81–87 (1997).
  • Adler CH, Singer C, O’Brien C et al. Randomized, placebo-controlled study of tolcapone in patients with fluctuating Parkinson disease treated with levodopa-carbidopa. Tolcapone Fluctuator Study Group III. Arch. Neurol.55(8), 1089–1095 (1998).
  • Dupont E, Burgunder JM, Findley LJ, Olsson JE, Dorflinger E. Tolcapone added to levodopa in stable parkinsonian patients: a double-blind placebo-controlled study. Tolcapone in Parkinson’s Disease Study Group II (TIPS II). Mov. Disord.12(6), 928–934 (1997).
  • Koller W, Lees A, Doder M, Hely M. Randomized trial of tolcapone versus pergolide as add-on to levodopa therapy in Parkinson’s disease patients with motor fluctuations. Mov. Disord.16(5), 858–866 (2001).
  • Assal F, Spahr L, Hadengue A, Rubbia-Brandt L, Burkhard PR. Tolcapone and fulminant hepatitis. Lancet352(9132), 958 (1998).
  • Watkins P. COMT inhibitors and liver toxicity. Neurology55(11 Suppl. 4), S51–S52 (2000).
  • Brooks DJ. Safety and tolerability of COMT inhibitors. Neurology62(1 Suppl. 1), S39–S46 (2004).
  • Golbe LI, Lieberman AN, Muenter MD et al. Deprenyl in the treatment of symptom fluctuations in advanced Parkinson’s disease. Clin. Neuropharmacol.11(1), 45–55 (1988).
  • Hubble JP. Novel drugs for Parkinson’s disease. Med. Clin. North Am.83(2), 525–536 (1999).
  • Seager H. Drug-delivery products and the Zydis fast-dissolving dosage form. J. Pharm. Pharmacol.50(4), 375–382 (1998).
  • Waters CH, Sethi KD, Hauser RA, Molho E, Bertoni JM. Zydis selegiline reduces off time in Parkinson’s disease patients with motor fluctuations: a 3-month, randomized, placebo-controlled study. Mov. Disord.19(4), 426–432 (2004).
  • Am OB, Amit T, Youdim MB. Contrasting neuroprotective and neurotoxic actions of respective metabolites of anti-Parkinson drugs rasagiline and selegiline. Neurosci. Lett.355(3), 169–172 (2004).
  • Speiser Z, Levy R, Cohen S. Effects of N-propargyl-1-(R)aminoindan (rasagiline) in models of motor and cognition disorders. J. Neural. Transm.52(Suppl. 5) 2287–3000 (1998).
  • Hauser RA, Lew MF, Hurtig H, Ondo W, Wojcieszek J. Early treatment with rasagiline is more beneficial than delayed treatment start in the long-term management of parkinson’s disease: Analyses of the TEMPO ITT cohort Parkinsonism. Relat. Disord.11(Suppl. 2), 129 (2005).
  • Parkinson Study Group. A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch. Neurol.59(12), 1937–1943 (2002).
  • Parkinson Study Group. A controlled, randomized, delayed-start study of rasagiline in early Parkinson disease. Arch. Neurol.61(4), 561–566 (2004).
  • Siderowf A, Eberly S, Oakes D. Reexamination of the TEMPO study. Arch. Neurol.621320–1321 (2005).
  • Stern MB, Marek KL, Friedman J et al. Double-blind, randomized, controlled trial of rasagiline as monotherapy in early Parkinson’s disease patients. Mov. Disord.19(8), 916–923 (2004).
  • Lew MF, Hauser RA, Hurtig H, Ondo W, Wojcieszek J. Long-term efficacy of rasagiline in Parkinson’s disease patients Parkinsonism. Relat. Disord.11(Suppl. 2), 129 (2005).
  • Rascol O, Brooks DJ, Melamed E et al. Rasagiline is effective and well tolerated in the treatment of moderate to advanced Parkinson’s disease (PD) patients receiving concomitant dopamine agonist therapy Parkinsonism. Relat. Disord.11(Suppl. 2), 129 (2005).
  • Parkinson Study Group. A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations: the PRESTO study. Arch. Neurol.62(2), 241–248 (2005).
  • Biglan KM, Schwid S, Eberly S et al. Rasagiline improves quality of life in patients with early Parkinson’s disease. Mov. Disord.21(5), 616–623 (2006).
  • Hobson DE, Pourcher E, Martin WR. Ropinirole and pramipexole, the new agonists Can. J. Neurol. Sci.26(Suppl. 2) S27–S33 (1999).
  • Maj J, Rogoi Z, Margas W, Kata M, Dziedzicka-Wasylewska M. The effect of repeated treatment with pramipexole on the central dopamine D3 system. J. Neural. Transm.107(12), 1369–1379 (2000).
  • Piercey MF, Walker EL, Feldpausch DL, Camacho-Ochoa M. High-affinity binding for pramipexole, a dopamine D3 receptor ligand, in rat striatum. Neurosci. Lett.219(2), 138–140 (1996).
  • Wright CE, Sisson TL, Ichhpurani AK, Peters GR. Steady state pharmacokinetic properties of pramipexole in healthy volunteers. J. Clin. Pharmacol.37(6), 520–525 (1997).
  • Moller JC, Oertel WH, Koster J, Pezzoli G, Provinciali L. Long-term efficacy and safety of pramipexole in advanced Parkinson’s disease: results from a European multicenter trial. Mov. Disord.20(5), 602–610 (2005).
  • Guttman M. Double-blind comparison of pramipexole and bromocriptine treatment with placebo in advanced Parkinson’s disease. International Pramipexole-Bromocriptine Study Group. Neurology49(4), 1060–1065 (1997).
  • Coldwell MC, Boyfield I, Brown T, Hagan JJ, Middlemiss DN. Comparison of the functional potencies of ropinirole and other dopamine receptor agonists at human D2(long), D3 and D4. 4 receptors expressed in Chinese hamster ovary cells. Br. J. Pharmacol.127(7), 1696–1702 (1999).
  • Lieberman A, Olanow CW, Sethi K et al. A multicenter trial of ropinirole as adjunct treatment for Parkinson’s disease. Ropinirole Study Group. Neurology51(4), 1057–1062 (1998).
  • 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.19(3), 234–245 (1996).
  • Blin O. The pharmacokinetics of pergolide in Parkinson’s disease. Curr. Opin. Neurol.16(Suppl. 1), S9–12 (2003).
  • Jenner P. Dopamine agonists, receptor selectivity and dyskinesia induction in Parkinson’s disease. Curr. Opin. Neurol.16(Suppl. 1), S3–S7 (2003).
  • Thalamas C, Rajman I, Kulisevsky J et al. Pergolide: multiple-dose pharmacokinetics in patients with mild-to-moderate Parkinson disease. Clin. Neuropharmacol.28(3), 120–125 (2005).
  • Jankovic J. Controlled trial of pergolide mesylate in Parkinson’s disease and progressive supranuclear palsy. Neurology33(4), 505–507 (1983).
  • Jankovic J. Long-term study of pergolide in Parkinson’s disease. Neurology35(3), 296–299 (1985).
  • Jankovic J. Long-term use of dopamine agonists in Parkinson’s disease. Clin. Neuropharmacol.8(2), 131–140 (1985).
  • Jankovic J, Orman J. Parallel double-blind study of pergolide in Parkinson’s disease. Adv. Neurol.45, 551–554 (1987).
  • Olanow CW, Fahn S, Muenter M et al. A multicenter double-blind placebo-controlled trial of pergolide as an adjunct to Sinemet in Parkinson’s disease. Mov. Disord.9(1), 40–47 (1994).
  • Van Camp G, Flamez A, Cosyns B et al. Heart valvular disease in patients with Parkinson’s disease treated with high-dose pergolide. Neurology61(6), 859–861 (2003).
  • Tintner R, Manian P, Gauthier P, Jankovic J. Pleuropulmonary fibrosis after long-term treatment with the dopamine agonist pergolide for Parkinson Disease. Arch. Neurol.62(8), 1290–1295 (2005).
  • Van Camp G, Flamez A, Cosyns B et al. Treatment of Parkinson’s disease with pergolide and relation to restrictive valvular heart disease. Lancet363(9416), 1179–1183 (2004).
  • Horvath J, Fross RD, Kleiner-Fisman G et al. Severe multivalvular heart disease: a new complication of the ergot derivative dopamine agonists. Mov. Disord.19(6), 656–662 (2004).
  • Del Dotto P, Bonuccelli U. Clinical pharmacokinetics of cabergoline. Clin. Pharmacokinet.42(7), 633–645 (2003).
  • Marsden CD. Clinical experience with cabergoline in patients with advanced Parkinson’s disease treated with levodopa. Drugs55(Suppl. 1), 17–22 (1998).
  • Rabey JM, Oberman Z, Scharf M et al. The influence of levodopa in the pharmacokinetics of bromocriptine in Parkinson’s disease. Clin. Neuropharmacol.12(5), 440–447 (1989).
  • 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.18(10), 1149–1156 (2003).
  • Gomez-Mancilla B, Bedard PJ. Effect of D1 and D2 agonists and antagonists on dyskinesia produced by L-dopa in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated monkeys. J. Pharmacol. Exp. Ther.259(1), 409–413 (1991).
  • Frucht SJ, Greene PE, Fahn S. Sleep episodes in Parkinson’s disease: a wake-up call. Mov. Disord.15(4), 601–603 (2000).
  • Frucht S, Rogers JD, Greene PE, Gordon MF, Fahn S. Falling asleep at the wheel: motor vehicle mishaps in persons taking pramipexole and ropinirole. Neurology52(9), 1908–1910 (1999).
  • Hobson DE, Lang AE, Martin WR et al. Excessive daytime sleepiness and sudden-onset sleep in Parkinson disease: a survey by the Canadian Movement Disorders Group. JAMA287(4), 455–463 (2002).
  • Driver-Dunckley E, Samanta J, Stacy M. Pathological gambling associated with dopamine agonist therapy in Parkinson’s disease. Neurology61(3), 422–423 (2003).
  • Kurlan R. Disabling repetitive behaviors in Parkinson’s disease. Mov. Disord.19(4), 433–437 (2004).
  • Dodd ML, Klos KJ, Bower JH et al. Pathological gambling caused by drugs used to treat Parkinson disease. Arch. Neurol.62(9), 1377–1381 (2005).
  • Morgan JC, Iyer SS, Sethi KD. Impulse control disorders and dopaminergic drugs. Arch. Neurol.63(2), 298–299 (2006).
  • Tan EK, Ondo W. Clinical characteristics of pramipexole-induced peripheral edema. Arch. Neurol.57(5), 729–732 (2000).
  • Shannon KM, Goetz CG, Carroll VS, Tanner CM, Klawans HL. Amantadine and motor fluctuations in chronic Parkinson’s disease. Clin. Neuropharmacol.10(6), 522–526 (1987).
  • Luginger E, Wenning GK, Bosch S, Poewe W. Beneficial effects of amantadine on L-dopa-induced dyskinesias in Parkinson’s disease. Mov. Disord.15(5), 873–878 (2000).
  • Parkes JD, Zilkha KJ, Marsden P, Baxter RC, Knill-Jones RP. Amantadine dosage in treatment of Parkinson’s disease. Lancet1(7657), 1130–1133 (1970).
  • Singer C, Papapetropoulos S, Gonzalez MA, Roberts EL, Lieberman A. Rimantadine in Parkinson’s disease patients experiencing peripheral adverse effects from amantadine: report of a case series. Mov. Disord.20(7), 873–877 (2005).
  • Nutt JG, Burchiel KJ, Comella CL et al. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology60(1), 69–73 (2003).
  • Kordower JH, Palfi S, Chen EY et al. Clinicopathological findings following intraventricular glial-derived neurotrophic factor treatment in a patient with Parkinson’s disease. Ann. Neurol.46(3), 419–424 (1999).
  • Gill SS, Patel NK, Hotton GR et al. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat. Med.9(5), 589–595 (2003).
  • Lang AE, Gill S, Patel NK et al. Randomized-controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann. Neurol.59(3), 459–466 (2006).
  • Colosimo C, Merello M, Albanese A. clinical usefulness of apomorphine in movement disorders. Clin. Neuropharmacol.17(3), 243–259 (1994).
  • Frankel JP, Lees AJ, Kempster PA, Stern GM. Subcutaneous apomorphine in the treatment of Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry53(2), 96–101 (1990).
  • Stacy M. Apomorphine: North American clinical experience. Neurology62(6 Suppl. 4), S18–S21 (2004).
  • Dewey RB Jr, Hutton JT, LeWitt PA, Factor SA. A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch. Neurol.58(9), 1385–1392 (2001).
  • Dewey RB Jr, Maraganore DM, Ahlskog JE, Matsumoto JY. Intranasal apomorphine rescue therapy for parkinsonian “off” periods. Clin. Neuropharmacol.19(3), 193–201 (1996).
  • Dewey RB Jr, Maraganore DM, Ahlskog JE, Matsumoto JY. A double-blind, placebo-controlled study of intranasal apomorphine spray as a rescue agent for off-states in Parkinson’s disease. Mov. Disord.13(5), 782–787 (1998).
  • Ondo W, Hunter C, Almaguer M, Jankovic J. A novel sublingual apomorphine treatment for patients with fluctuating Parkinson’s disease. Mov. Disord.14(4), 664–668 (1999).
  • Ondo W, Hunter C, Almaguer M, Gancher S, Jankovic J. Efficacy and tolerability of a novel sublingual apomorphine preparation in patients with fluctuating Parkinson’s disease. Clin. Neuropharmacol.22(1), 1–4 (1999).
  • Stocchi F, Vacca L, De Pandis MF et al. Subcutaneous continuous apomorphine infusion in fluctuating patients with Parkinson’s disease: long-term results. Neurol. Sci.22(1), 93–94 (2001).
  • Priano L, Albani G, Calderoni S et al. Controlled-release transdermal apomorphine treatment for motor fluctuations in Parkinson’s disease. Neurol. Sci.23(Suppl. 2), S99–S100 (2002).
  • Djaldetti R, Baron J, Ziv I, Melamed E. Gastric emptying in Parkinson’s disease: patients with and without response fluctuations. Neurology46(4), 1051–1054 (1996).
  • Contin M, Riva R, Martinelli P, Albani F, Baruzzi A. Effect of meal timing on the kinetic-dynamic profile of levodopa/carbidopa controlled release [corrected] in parkinsonian patients. Eur. J. Clin. Pharmacol.54(4), 303–308 (1998).
  • Kurth MC, Tetrud JW, Irwin I, Lyness WH, Langston JW. Oral levodopa/carbidopa solution versus tablets in Parkinson’s patients with severe fluctuations: a pilot study. Neurology43(5), 1036–1039 (1993).
  • Metman LV, Hoff J, Mouradian MM, Chase TN. Fluctuations in plasma levodopa and motor responses with liquid and tablet levodopa/carbidopa. Mov. Disord.9(4), 463–465 (1994).
  • Pappert EJ, Goetz CG, Niederman F et al. Liquid levodopa/carbidopa produces significant improvement in motor function without dyskinesia exacerbation. Neurology47(6), 1493–1495 (1996).
  • Claxton AJ, Cramer J, Pierce C. A systematic review of the associations between dose regimens and medication compliance. Clin. Ther.23(8), 1296–1310 (2001).
  • Djaldetti R, Melamed E. Levodopa ethylester: a novel rescue therapy for response fluctuations in Parkinson’s disease. Ann. Neurol.39(3), 400–404 (1996).
  • Blindauer K, Shoulson I, Oakes D et al. A randomized controlled trial of etilevodopa in patients with Parkinson disease who have motor fluctuations. Arch. Neurol.63(2), 210–216 (2006).
  • Rascol O, Brooks DJ, Korczyn AD et al. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. 056 Study Group. N. Engl. J. Med.342(20), 1484–1491 (2000).
  • Clarke CE, Speller JM, Clarke JA. Pramipexole versus bromocriptine for levodopa-induced complications in Parkinson’s disease. Cochrane Database Syst. Rev. (3), CD002259 (2000).
  • Metman LV, Del Dotto P, LePoole K et al. Amantadine for levodopa-induced dyskinesias: a 1-year follow-up study. Arch. Neurol.56(11), 1383–1386 (1999).
  • Rajput AH, Rajput A, Lang AE et al. New use for an old drug: amantadine benefits levodopa-induced dyskinesia. Mov. Disord.13(5), 851 (1998).
  • Silva-Junior FP, Braga-Neto P, Sueli MF, de Bruin VM. Amantadine reduces the duration of levodopa-induced dyskinesia: a randomized, double-blind, placebo-controlled study. Parkinsonism Relat. Disord.11(7), 449–452 (2005).
  • Thomas A, Iacono D, Luciano AL et al. Duration of amantadine benefit on dyskinesia of severe Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry75(1), 141–143 (2004).
  • Merims D, Ziv I, Djaldetti R, Melamed E. Riluzole for levodopa-induced dyskinesias in advanced Parkinson’s disease. Lancet353(9166), 1764–1765 (1999).
  • Lokk J. Memantine can relieve certain symptoms in Parkinson disease. Improvement achieved in two out of three described cases with dyskinesia and cognitive failure. Lakartidningen.101(23), 2003–2006 (2004).
  • Verhagen ML, Del Dotto P, Natte R, van den MP, Chase TN. Dextromethorphan improves levodopa-induced dyskinesias in Parkinson’s disease. Neurology51(1), 203–206 (1998).
  • Durif F, Debilly B, Galitzky M et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology62(3), 381–388 (2004).
  • Morgante L, Epifanio A, Spina E et al. Quetiapine and clozapine in parkinsonian patients with dopaminergic psychosis. Clin. Neuropharmacol.27(4), 153–156 (2004).
  • Sa DS, Lang AE. Olanzapine and clozapine: comparative effects on motor function in hallucinating PD patients. Neurology57(4), 747 (2001).
  • Carpentier AF, Bonnet AM, Vidailhet M, Agid Y. Improvement of levodopa-induced dyskinesia by propranolol in Parkinson’s disease. Neurology46(6), 1548–1551 (1996).
  • Tousi B, Subramanian T. The effect of levetiracetam on levodopa induced dyskinesia in patients with Parkinson’s disease. Parkinsonism Relat. Disord.11(5), 333–334 (2005).
  • Zesiewicz TA, Sullivan KL, Maldonado JL, Tatum WO, Hauser RA. Open-label pilot study of levetiracetam (Keppra) for the treatment of levodopa-induced dyskinesias in Parkinson’s disease. Mov. Disord.20(9), 1205–1209 (2005).
  • Silverdale MA, Nicholson SL, Crossman AR, Brotchie JM. Topiramate reduces levodopa-induced dyskinesia in the MPTP-lesioned marmoset model of Parkinson’s disease. Mov. Disord.20(4), 403–409 (2005).
  • Durif F, Deffond D, Dordain G, Tournilhac M. Apomorphine and diphasic dyskinesia. Clin. Neuropharmacol.17(1), 99–102 (1994).
  • Pacchetti C, Albani G, Martignoni E et al. “Off” painful dystonia in Parkinson’s disease treated with botulinum toxin. Mov. Disord.10(3), 333–336 (1995).
  • Poewe WH, Lees AJ, Stern GM. Dystonia in Parkinson’s disease: clinical and pharmacological features. Ann. Neurol.23(1), 73–78 (1988).
  • Pogarell O, Gasser T, van Hilten JJ et al. Pramipexole in patients with Parkinson’s disease and marked drug resistant tremor: a randomised, double blind, placebo- controlled multicentre study. J. Neurol. Neurosurg. Psychiatry72(6), 713–720 (2002).
  • Navan P, Findley LJ, Jeffs JA, Pearce RK, Bain PG. Randomized, double-blind, 3-month parallel study of the effects of pramipexole, pergolide, and placebo on parkinsonian tremor. Mov. Disord.18(11), 1324–1331 (2003).
  • Navan P, Findley LJ, Undy MB, Pearce RK, Bain PG. A randomly assigned double-blind cross-over study examining the relative anti-parkinsonian tremor effects of pramipexole and pergolide. Eur. J. Neurol.12(1), 1–8 (2005).
  • Marjama-Lyons J, Koller W. Tremor-predominant Parkinson’s disease. Approaches to treatment. Drugs Aging16(4), 273–278 (2000).
  • Koller WC, Herbster G. Adjuvant therapy of parkinsonian tremor. Arch. Neurol.44(9), 921–923 (1987).
  • Crosby NJ, Deane KH, Clarke CE. β-blocker therapy for tremor in Parkinson’s disease. Cochrane Database Syst. Rev. (1), CD003361 (2003).
  • Nakanishi I, Kohmoto J, Miwa H, Kondo T. Effect of zonisamide on resting tremor resistant to antiparkinsonian medication. No To Shinkei55(8), 685–689 (2003).
  • Jansen EN. Clozapine in the treatment of tremor in Parkinson’s disease. Acta Neurol. Scand.89(4), 262–265 (1994).
  • Katzenschlager R, Sampaio C, Costa J, Lees A. Anticholinergics for symptomatic management of Parkinson’s disease. Cochrane Database Syst. Rev. (2), CD003735 (2003).
  • Giladi N, McDermott MP, Fahn S et al. Freezing of gait in PD: prospective assessment in the DATATOP cohort. Neurology56(12), 1712–1721 (2001).
  • Giladi N. Freezing of gait. Clinical overview. Adv. Neurol.87, 191–197 (2001).
  • Giladi N, Treves TA, Simon ES et al. Freezing of gait in patients with advanced Parkinson’s disease. J. Neural. Transm.108(1), 53–61 (2001).
  • Tohgi H, Abe T, Takahashi S et al. Monoamine metabolism in the cerebrospinal fluid in Parkinson’s disease: relationship to clinical symptoms and subsequent therapeutic outcomes. J. Neural. Transm. Park Dis. Dement. Sect.5(1), 17–26 (1993).
  • Tohgi H, Abe T, Takahashi S. The effects of L-threo-3,4-dihydroxyphenylserine on the total norepinephrine and dopamine concentrations in the cerebrospinal fluid and freezing gait in parkinsonian patients. J. Neural. Transm. Park Dis. Dement. Sect.5(1), 27–34 (1993).
  • Tohgi H, Abe T, Takahashi S et al. Effect of a synthetic norepinephrine precursor, L-threo-3,4- dihydroxyphenylserine on the total norepinephrine concentration in the cerebrospinal fluid of parkinsonian patients. Neurosci. Lett.116(1–2), 194–197 (1990).
  • Giladi N, Gurevich T, Shabtai H, Paleacu D, Simon ES. The effect of botulinum toxin injections to the calf muscles on freezing of gait in parkinsonism: a pilot study. J. Neurol.248(7), 572–576 (2001).
  • Fernandez HH, Lannon MC, Trieschmann ME, Friedman JH. Botulinum toxin type B for gait freezing in Parkinson’s disease. Med. Sci. Monit.10(7), CR282–CR284 (2004).
  • Wieler M, Camicioli R, Jones CA, Martin WR. Botulinum toxin injections do not improve freezing of gait in Parkinson disease. Neurology65(4), 626–628 (2005).
  • Witjas T, Kaphan E, Azulay JP et al. Nonmotor fluctuations in Parkinson’s disease: frequent and disabling. Neurology59(3), 408–413 (2002).
  • Aarsland D, Andersen K, Larsen JP, Lolk A, Kragh-Sorensen P. Prevalence and characteristics of dementia in Parkinson disease: an 8-year prospective study. Arch. Neurol.60(3), 387–392 (2003).
  • Aarsland D, Zaccai J, Brayne C. A systematic review of prevalence studies of dementia in Parkinson’s disease. Mov. Disord.20(10), 1255–1263 (2005).
  • Miyasaki JM, Shannon KM, Voon V et al. Practice parameter: evaluation and treatment of depression, psychosis, and dementia in Parkinson disease (an evidence-based review). Neurology661–1 (2006).
  • Suchowersky O, Reich S, Perlmutter J et al. Practice parameter: diagnosis and prognosis of new onset Parkinson disease (an evidence-based review). Neurology66(7), 978–975 (2006).
  • Aarsland D, Litvan I, Salmon D et al. Performance on the dementia rating scale in Parkinson’s disease with dementia and dementia with Lewy bodies: comparison with progressive supranuclear palsy and Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry74(9), 1215–1220 (2003).
  • Litvan I, Bhatia KP, Burn DJ et al. Movement Disorders Society Scientific Issues Committee report: SIC task force appraisal of clinical diagnostic criteria for Parkinsonian disorders. Mov. Disord.18(5), 467–486 (2003).
  • Emre M, Aarsland D, Albanese A et al. Rivastigmine for dementia associated with Parkinson’s disease. N. Engl. J. Med.351(24), 2509–2518 (2004).
  • Wesnes KA, McKeith I, Edgar C, Emre M, Lane R. Benefits of rivastigmine on attention in dementia associated with Parkinson disease. Neurology65(10), 1654–1656 (2005).
  • Ravina B, Putt M, Siderowf A et al. Donepezil for dementia in Parkinson’s disease: a randomised, double blind, placebo-controlled, crossover study. J. Neurol. Neurosurg. Psychiatry76(7), 934–939 (2005).
  • Thomas AJ, Burn DJ, Rowan EN et al. A comparison of the efficacy of donepezil in Parkinson’s disease with dementia and dementia with Lewy bodies. Int. J. Geriatr. Psychiatry20(10), 938–944 (2005).
  • Harding AJ, Stimson E, Henderson JM, Halliday GM. Clinical correlates of selective pathology in the amygdala of patients with Parkinson’s disease. Brain125(Pt 11), 2431–2445 (2002).
  • Harding AJ, Broe GA, Halliday GM. Visual hallucinations in Lewy body disease relate to Lewy bodies in the temporal lobe. Brain125(Pt 2), 391–403 (2002).
  • Factor SA, Feustel PJ, Friedman JH et al. Longitudinal outcome of Parkinson’s disease patients with psychosis. Neurology60(11), 1756–1761 (2003).
  • Goetz CG, Stebbins GT. Risk factors for nursing home placement in advanced Parkinson’s disease. Neurology43(11), 2227–2229 (1993).
  • Goetz CG, Stebbins GT. Mortality and hallucinations in nursing home patients with advanced Parkinson’s disease. Neurology45(4), 669–671 (1995).
  • Goetz CG, Vogel C, Tanner CM, Stebbins GT. Early dopaminergic drug-induced hallucinations in parkinsonian patients. Neurology51(3), 811–814 (1998).
  • 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).
  • Friedman JH, Factor SA. Atypical antipsychotics in the treatment of drug-induced psychosis in Parkinson’s disease. Mov. Disord.15(2), 201–211 (2000).
  • Burn DJ. Beyond the iron mask: towards better recognition and treatment of depression associated with Parkinson’s disease. Mov. Disord.17(3), 445–454 (2002).
  • Burn DJ. Depression in Parkinson’s disease. Eur. J. Neurol.9(Suppl. 3), 44–54 (2002).
  • Rektorova I, Rektor I, Bares M et al. Pramipexole and pergolide in the treatment of depression in Parkinson’s disease: a national multicentre prospective randomized study. Eur. J. Neurol.10(4), 399–406 (2003).
  • Chou KL, Hurtig HI, Jaggi JL et al. Electroconvulsive therapy for depression in a Parkinson’s disease patient with bilateral subthalamic nucleus deep brain stimulators. Parkinsonism Relat. Disord.11(6), 403–406 (2005).
  • Fall PA, Granerus AK. Maintenance ECT in Parkinson’s disease. J. Neural. Transm.106(7–8), 737–741 (1999).
  • Shoulson I, Glaubiger GA, Chase TN. On-off response. Clinical and biochemical correlations during oral and intravenous levodopa administration in parkinsonian patients. Neurology25(12), 1144–1148 (1975).
  • Zesiewicz TA, Baker MJ, Wahba M, Hauser RA. Autonomic nervous system dysfunction in Parkinson’s disease. Curr. Treat. Options Neurol.5(2), 149–160 (2003).
  • Edwards LL, Quigley EM, Harned RK, Hofman R, Pfeiffer RF. Characterization of swallowing and defecation in Parkinson’s disease. Am. J. Gastroenterol.89(1), 15–25 (1994).
  • Sakakibara R, Odaka T, Uchiyama T et al. Colonic transit time and rectoanal videomanometry in Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry74(2), 268–272 (2003).
  • Abbott RD, Petrovitch H, White LR et al. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology57(3), 456–462 (2001).
  • Edwards LL, Quigley EM, Harned RK, Hofman R, Pfeiffer RF. Defecatory function in Parkinson’s disease: response to apomorphine. Ann. Neurol.33(5), 490–493 (1993).
  • Albanese A, Maria G, Bentivoglio AR et al. Severe constipation in Parkinson’s disease relieved by botulinum toxin. Mov. Disord.12(5), 764–766 (1997).
  • Sullivan KL, Staffetti JF, Hauser RA, Dunne PB, Zesiewicz TA. Tegaserod (Zelnorm) for the treatment of constipation in Parkinson’s disease. Mov. Disord.21(1), 115–116 (2006).
  • Restivo DA, Palmeri A, Marchese-Ragona R. Botulinum toxin for cricopharyngeal dysfunction in Parkinson’s disease. N. Engl. J. Med.346(15), 1174–1175 (2002).
  • Pal PK, Calne DB, Calne S, Tsui JK. Botulinum toxin A as treatment for drooling saliva in PD. Neurology54(1), 244–247 (2000).
  • Ondo WG, Hunter C, Moore W. A double-blind placebo-controlled trial of botulinum toxin B for sialorrhea in Parkinson’s disease. Neurology62(1), 37–40 (2004).
  • Dogu O, Apaydin D, Sevim S, Talas DU, Aral M. Ultrasound-guided versus ‘blind’ intraparotid injections of botulinum toxin-A for the treatment of sialorrhoea in patients with Parkinson’s disease. Clin. Neurol. Neurosurg.106(2), 93–96 (2004).
  • Araki I, Kitahara M, Oida T, Kuno S. Voiding dysfunction and Parkinson’s disease: urodynamic abnormalities and urinary symptoms. J. Urol.164(5), 1640–1643 (2000).
  • Zesiewicz TA, Helal M, Hauser RA. Sildenafil citrate (Viagra) for the treatment of erectile dysfunction in men with Parkinson’s disease. Mov. Disord.15(2), 305–308 (2000).
  • Hussain IF, Brady CM, Swinn MJ, Mathias CJ, Fowler CJ. Treatment of erectile dysfunction with sildenafil citrate (Viagra) in parkinsonism due to Parkinson’s disease or multiple system atrophy with observations on orthostatic hypotension. J. Neurol. Neurosurg. Psychiatry71(3), 371–374 (2001).
  • Pavone C, Curto F, Anello G et al. Prospective, randomized, crossover comparison of sublingual apomorphine (3 mg) with oral sildenafil (50 mg) for male erectile dysfunction. J. Urol.172(6 Pt 1), 2347–2349 (2004).
  • Evans AH, Lees AJ. Dopamine dysregulation syndrome in Parkinson’s disease. Curr. Opin. Neurol.17(4), 393–398 (2004).
  • Wright RA, Kaufmann HC, Perera R et al. A double-blind, dose-response study of midodrine in neurogenic orthostatic hypotension. Neurology51(1), 120–124 (1998).
  • Low PA, Gilden JL, Freeman R, Sheng KN, McElligott MA. Efficacy of midodrine vs placebo in neurogenic orthostatic hypotension. A randomized, double-blind multicenter study. Midodrine Study Group. JAMA277(13), 1046–1051 (1997).
  • Hoehn MM. Levodopa-induced postural hypotension. Treatment with fludrocortisone. Arch. Neurol.32(1), 50–51 (1975).
  • Abate G, Polimeni RM, Cuccurullo F, Puddu P, Lenzi S. Effects of indomethacin on postural hypotension in parkinsonism. Br. Med J.2(6203), 1466–1468 (1979).
  • Swinn L, Schrag A, Viswanathan R et al. Sweating dysfunction in Parkinson’s disease. Mov. Disord.18(12), 1459–1463 (2003).
  • Partinen M. Sleep disorder related to Parkinson’s disease. J. Neurol.244(4 Suppl. 1), S3–S6 (1997).
  • Lees AJ, Blackburn NA, Campbell VL. The nighttime problems of Parkinson’s disease. Clin. Neuropharmacol.11(6), 512–519 (1988).
  • Askenasy JJ, Yahr MD. Parkinsonian tremor loses its alternating aspect during non-REM sleep and is inhibited by REM sleep. J. Neurol. Neurosurg. Psychiatry53(9), 749–753 (1990).
  • Boeve BF, Silber MH, Ferman TJ, Lucas JA, Parisi JE. Association of REM sleep behavior disorder and neurodegenerative disease may reflect an underlying synucleinopathy. Mov. Disord.16(4), 622–630 (2001).
  • Boeve BF, Silber MH, Ferman TJ. Melatonin for treatment of REM sleep behavior disorder in neurologic disorders: results in 14 patients. Sleep Med.4(4), 281–284 (2003).
  • Fantini ML, Gagnon JF, Filipini D, Montplaisir J. The effects of pramipexole in REM sleep behavior disorder. Neurology61(10), 1418–1420 (2003).
  • Tandberg E, Larsen JP, Karlsen K. Excessive daytime sleepiness and sleep benefit in Parkinson’s disease: a community-based study. Mov. Disord.14(6), 922–927 (1999).
  • Ondo WG, Dat VK, Khan H et al. Daytime sleepiness and other sleep disorders in Parkinson’s disease. Neurology57(8), 1392–1396 (2001).
  • Happe S, Pirker W, Sauter C, Klosch G, Zeitlhofer J. Successful treatment of excessive daytime sleepiness in Parkinson’s disease with modafinil. J. Neurol.248(7), 632–634 (2001).
  • Ondo WG, Fayle R, Atassi F, Jankovic J. Modafinil for daytime somnolence in Parkinson’s disease: double blind, placebo- controlled parallel trial. J. Neurol. Neurosurg. Psychiatry76(12), 1636–1639 (2005).
  • Paus S, Brecht HM, Koster J et al. Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s disease. Mov. Disord.18(6), 659–667 (2003).
  • Hauser RA, Wahba MN, Zesiewicz TA, McDowell AW. Modafinil treatment of pramipexole-associated somnolence. Mov. Disord.15(6), 1269–1271 (2000).
  • Allen RP, Kushida CA, Atkinson MJ. Factor analysis of the International Restless Legs Syndrome Study Group’s scale for restless legs severity. Sleep Med.4(2), 133–135 (2003).
  • Ondo WG, Vuong KD, Jankovic J. Exploring the relationship between Parkinson disease and restless legs syndrome. Arch. Neurol.59(3), 421–424 (2002).
  • Lang AE, Johnson K. Akathisia in idiopathic Parkinson’s disease. Neurology37(3), 477–481 (1987).
  • Comella CL, Goetz CG. Akathisia in Parkinson’s disease. Mov. Disord.9(5), 545–549 (1994).
  • Lyons KE, Pahwa R. Efficacy and tolerability of levetiracetam in Parkinson disease patients with levodopa-induced dyskinesia. Clin. Neuropharmacol.29, 148–153 (2006).
  • Evans AH, Pavese N, Lawrence AD et al. Compulsive drug use linked to sensitized ventral striatal dopamine transmission. Ann. Neurol.59, 852–858 (2006).

Website

  • Food and Drug Administration www.fda.gov/medwatch/SAFETY/1998/ tasmar.htm

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.