Review: management of Parkinson's disease

David J Pedrosa, Lars Timmermann, David J Pedrosa, Lars Timmermann

Abstract

Parkinson's disease (PD) is one of the most frequent neurological diseases. Despite the modern imaging and nuclear techniques which help to diagnose it in a very early stage and lead to a better discrimination of similar diseases, PD has remained a clinical diagnosis. The increasing number of available treatment options makes the disease management often complicated even when the presence of PD seems undoubted. In addition, nonmotor symptoms and side effects of some therapies constitute some pitfalls already in the preclinical state or at the beginnings of the disease, especially with the progressive effect on patients. Therefore, this review aimed to summarize study results and depict recommended medical treatments for the most common motor and nonmotor symptoms in PD. Additionally, emerging new therapeutic options such as continuous pump therapies, eg, with apomorphine or parenteral levodopa, or the implantation of electrodes for deep brain stimulation were also considered.

Keywords: DBS; Parkinson’s disease; disease management; nonmotor symptoms; pump therapies; side effects.

Figures

Figure 1
Figure 1
German guidelines for treatment of PD 2012. Abbreviations: COMT-1, catechol-O-methyltransferase-Iinhibitor; DA, dopamine agonist; DBS, deep brain stimulation; PD, Parkinson’s disease.

References

    1. Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197–211.
    1. Kaufman MJ, Madras BK. Severe depletion of cocaine recognition sites associated with the dopamine transporter in Parkinson’s-diseased striatum. Synapse. 1991;9(1):43–49.
    1. Niznik HB, Fogel EF, Fassos FF, Seeman P. The dopamine transporter is absent in parkinsonian putamen and reduced in the caudate nucleus. J Neurochem. 1991;56(1):192–198.
    1. Eggers C, Pedrosa DJ, Kahraman D, et al. Parkinson subtypes progress differently in clinical course and imaging pattern. PLoS One. 2012;7(10):e46813.
    1. Rajput AH, Voll A, Rajput ML, Robinson CA, Rajput A. Course in Parkinson disease subtypes: a 39-year clinicopathologic study. Neurology. 2009;73(3):206–212.
    1. Ashburn A, Fazakarley L, Ballinger C, Pickering R, McLellan LD, Fitton C. A randomised controlled trial of a home based exercise programme to reduce the risk of falling among people with Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2007;78(7):678–684.
    1. Cakit BD, Saracoglu M, Genc H, Erdem HR, Inan L. The effects of incremental speed-dependent treadmill training on postural instability and fear of falling in Parkinson’s disease. Clin Rehabil. 2007;21(8):698–705.
    1. Ebersbach G, Ebersbach A, Edler D, et al. Comparing exercise in Parkinson’s disease – the Berlin LSVT®BIG study. Mov Disord. 2010;25(12):1902–1908.
    1. Ellis T, de Goede CJ, Feldman RG, Wolters EC, Kwakkel G, Wagenaar RC. Efficacy of a physical therapy program in patients with Parkinsons disease: a randomized controlled trial. Arch Phys Med Rehabil. 2005;86(4):626–632.
    1. Morris ME, Iansek R, Kirkwood B. A randomized controlled trial of movement strategies compared with exercise for people with Parkinson’s disease. Mov Disord. 2009;24(1):64–71.
    1. Barbe MT, Cepuran F, Amarell M, Schoenau E, Timmermann L. Long-term effect of robot-assisted treadmill walking reduces freezing of gait in Parkinson’s disease patients: a pilot study. J Neurol. 2013;260(1):296–298.
    1. Tomlinson CL, Patel S, Meek C, et al. Physiotherapy versus placebo or no intervention in Parkinson’s disease. Cochrane Database Syst Rev. 2012;7:CD002817.
    1. Chrisp P, Mammen GJ, Sorkin EM. Selegiline. A review of its pharmacology, symptomatic benefits and protective potential in Parkinson’s disease. Drugs Aging. 1991;1(3):228–248.
    1. Youdim MB, Bar Am O, Yogev-Falach M, et al. Rasagiline: neurodegeneration, neuroprotection, and mitochondrial permeability transition. J Neurosci Res. 2005;79(1–2):172–179.
    1. Zou L, Jankovic J, Rowe DB, Xie W, Appel SH, Le W. Neuroprotection by pramipexole against dopamine- and levodopa-induced cytotoxicity. Life Sci. 1999;64(15):1275–1285.
    1. Beal MF, Matthews RT, Tieleman A, Shults CW. Coenzyme Q10 attenuates the 1-methyl-4-phenyl-1,2,3,tetrahydropyridine (MPTP) induced loss of striatal dopamine and dopaminergic axons in aged mice. Brain Res. 1998;783(1):109–114.
    1. Olanow CW, Rascol O, Hauser R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med. 2009;361(13):1268–1278.
    1. Parkinson Study Group. A controlled, randomized, delayed-start study of rasagiline in early Parkinson disease. Arch Neurol. 2004;61(4):561–566.
    1. Schapira AH, Albrecht S, Barone P, et al. Rationale for delayed-start study of pramipexole in Parkinson’s disease: the PROUD study. Mov Disord. 2010;25(11):1627–1632.
    1. Hart RG, Pearce LA, Ravina BM, Yaltho TC, Marler JR. Neuroprotection trials in Parkinson’s disease: systematic review. Mov Disord. 2009;24(5):647–654.
    1. Goetz CG, Poewe W, Rascol O, Sampaio C. Evidence-based medical review update: pharmacological and surgical treatments of Parkinson’s disease: 2001 to 2004. Mov Disord. 2005;20(5):523–539.
    1. 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. 1998;55(8):1089–1095.
    1. Deane KH, Spieker S, Clarke CE. Catechol-O-methyltransferase inhibitors for levodopa-induced complications in Parkinson’s disease. Cochrane Database Syst Rev. 2004;4:CD004554.
    1. 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. Neurology. 1997;48(1):81–87.
    1. Fahn S, Oakes D, Shoulson I, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med. 2004;351(24):2498–2508.
    1. Talati R, Baker WL, Patel AA, Reinhart K, Coleman CI. Adding a dopamine agonist to preexisting levodopa therapy vs levodopa therapy alone in advanced Parkinson’s disease: a meta analysis. Int J Clin Pract. 2009;63(4):613–623.
    1. Kvernmo T, Houben J, Sylte I. Receptor-binding and pharmacokinetic properties of dopaminergic agonists. Curr Top Med Chem. 2008;8(12):1049–1067.
    1. Fox SH, Katzenschlager R, Lim SY, et al. The Movement Disorder Society evidence-based medicine review update: treatments for the motor symptoms of Parkinson’s disease. Mov Disord. 2011;26(Suppl 3):S2–S41.
    1. 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. 2005;20(5):602–610.
    1. 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. 2004;61(7):1044–1053.
    1. 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. 2007;22(16):2409–2417.
    1. Olanow CW, Watkins PB. Tolcapone: an efficacy and safety review (2007) Clin Neuropharmacol. 2007;30(5):287–294.
    1. Reichmann H, Boas J, Macmahon D, Myllyla V, Hakala A, Reinikainen K. Efficacy of combining levodopa with entacapone on quality of life and activities of daily living in patients experiencing wearing-off type fluctuations. Acta Neurol Scand. 2005;111(1):21–28.
    1. Snow BJ, Macdonald L, Mcauley D, Wallis W. The effect of amantadine on levodopa-induced dyskinesias in Parkinson’s disease: a double-blind, placebo-controlled study. Clin Neuropharmacol. 2000;23(2):82–85.
    1. Luginger E, Wenning GK, Bosch S, Poewe W. Beneficial effects of amantadine on L-dopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2000;15(5):873–878.
    1. Hauser RA, Cantillon M, Pourcher E, et al. Preladenant in patients with Parkinson’s disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol. 2011;10(3):221–229.
    1. Hodgson RA, Bedard PJ, Varty GB, et al. Preladenant, a selective A(2A) receptor antagonist, is active in primate models of movement disorders. Exp Neurol. 2010;225(2):384–390.
    1. Postuma RB, Lang AE, Munhoz RP, et al. Caffeine for treatment of Parkinson disease: a randomized controlled trial. Neurology. 2012;79(7):651–658.
    1. Olanow CW, Obeso JA, Stocchi F. Drug insight: continuous dopaminergic stimulation in the treatment of Parkinson’s disease. Nat Clin Pract Neurol. 2006;2(7):382–392.
    1. Ondo WG, Sethi KD, Kricorian G. Selegiline orally disintegrating tablets in patients with Parkinson disease and “wearing off” symptoms. Clin Neuropharmacol. 2007;30(5):295–300.
    1. Crosby N, Deane KH, Clarke CE. Amantadine in Parkinson’s disease. Cochrane Database Syst Rev. 2003;1:CD003468.
    1. Pedrosa DJ, Reck C, Florin E, et al. Essential tremor and tremor in Parkinson’s disease are associated with distinct “tremor clusters” in the ventral thalamus. Exp Neurol. 2012;237(2):435–443.
    1. Helmich RC, Janssen MJ, Oyen WJ, Bloem BR, Toni I. Pallidal dysfunction drives a cerebellothalamic circuit into Parkinson tremor. Ann Neurol. 2011;69(2):269–281.
    1. Parkinson Study Group. Low-dose clozapine for the treatment of drug-induced psychosis in Parkinson’s disease. N Engl J Med. 1999;340(10):757–763.
    1. Hariz MI, Krack P, Alesch F, et al. Multicentre European study of thalamic stimulation for parkinsonian tremor: a 6 year follow-up. J Neurol Neurosurg Psychiatry. 2008;79(6):694–699.
    1. Pillon B, Dubois B, Bonnet AM, et al. Cognitive slowing in Parkinson’s disease fails to respond to levodopa treatment: the 15-objects test. Neurology. 1989;39(6):762–768.
    1. Levy G, Louis ED, Cote L, et al. Contribution of aging to the severity of different motor signs in Parkinson disease. Arch Neurol. 2005;62(3):467–472.
    1. Rahman S, Griffin HJ, Quinn NP, Jahanshahi M. The factors that induce or overcome freezing of gait in Parkinson’s disease. Behav Neurol. 2008;19(3):127–136.
    1. Nieuwboer A, Kwakkel G, Rochester L, et al. Cueing training in the home improves gait-related mobility in Parkinson’s disease: the RESCUE trial. J Neurol Neurosurg Psychiatry. 2007;78(2):134–140.
    1. Arias P, Cudeiro J. Effect of rhythmic auditory stimulation on gait in parkinsonian patients with and without freezing of gait. PLoS One. 2010;5(3):e9675.
    1. Nieuwboer A, Baker K, Willems AM, et al. The short-term effects of different cueing modalities on turn speed in people with Parkinson’s disease. Neurorehabil Neural Repair. 2009;23(8):831–836.
    1. Nieuwboer A. Cueing for freezing of gait in patients with Parkinson’s disease: a rehabilitation perspective. Mov Disord. 2008;23(Suppl 2):S475–S481.
    1. Allen NE, Canning CG, Sherrington C, et al. The effects of an exercise program on fall risk factors in people with Parkinson’s disease: a randomized controlled trial. Mov Disord. 2010;25(9):1217–1225.
    1. Giladi N, McDermott MP, Fahn S, et al. Freezing of gait in PD: prospective assessment in the DATATOP cohort. Neurology. 2001;56(12):1712–1721.
    1. Jankovic J. Long-term study of pergolide in Parkinson’s disease. Neurology. 1985;35(3):296–299.
    1. Giladi N. Medical treatment of freezing of gait. Mov Disord. 2008;23(Suppl 2):S482–S488.
    1. Ferraye MU, Debu B, Fraix V, et al. Effects of subthalamic nucleus stimulation and levodopa on freezing of gait in Parkinson disease. Neurology. 2008;70(16 Pt 2):1431–1437.
    1. Davis JT, Lyons KE, Pahwa R. Freezing of gait after bilateral subthalamic nucleus stimulation for Parkinson’s disease. Clin Neurol Neurosurg. 2006;108(5):461–464.
    1. Moreau C, Defebvre L, Destee A, et al. STN-DBS frequency effects on freezing of gait in advanced Parkinson disease. Neurology. 2008;71(2):80–84.
    1. Ferraye MU, Debu B, Fraix V, et al. Effects of pedunculopontine nucleus area stimulation on gait disorders in Parkinson’s disease. Brain. 2010;133(Pt 1):205–214.
    1. Finsterer J, Strobl W. Presentation, etiology, diagnosis, and management of camptocormia. Eur Neurol. 2010;64(1):1–8.
    1. Jankovic J. Camptocormia, head drop and other bent spine syndromes: heterogeneous etiology and pathogenesis of parkinsonian deformities. Mov Disord. 2010;25(5):527–528.
    1. Claassen DO, Josephs KA, Ahlskog JE, Silber MH, Tippmann-Peikert M, Boeve BF. REM sleep behavior disorder preceding other aspects of synucleinopathies by up to half a century. Neurology. 2010;75(6):494–499.
    1. Olson EJ, Boeve BF, Silber MH. Rapid eye movement sleep behaviour disorder: demographic, clinical and laboratory findings in 93 cases. Brain. 2000;123(Pt 2):331–339.
    1. Seppi K, Weintraub D, Coelho M, et al. The Movement Disorder Society evidence-based medicine review update: treatments for the non-motor symptoms of Parkinson’s disease. Mov Disord. 2011;26(Suppl 3):S42–S80.
    1. Aurora RN, Kristo DA, Bista SR, et al. The treatment of restless legs syndrome and periodic limb movement disorder in adults – an update for 2012: practice parameters with an evidence-based systematic review and meta-analyses: an American Academy of Sleep Medicine Clinical Practice Guideline. Sleep. 2012;35(8):1039–1062.
    1. Oertel W, Trenkwalder C, Benes H, et al. Long-term safety and efficacy of rotigotine transdermal patch for moderate-to-severe idiopathic restless legs syndrome: a 5-year open-label extension study. Lancet Neurol. 2011;10(8):710–720.
    1. Arnulf I, Bejjani BP, Garma L, et al. Improvement of sleep architecture in PD with subthalamic nucleus stimulation. Neurology. 2000;55(11):1732–1734.
    1. Iranzo A, Valldeoriola F, Santamaria J, Tolosa E, Rumia J. Sleep symptoms and polysomnographic architecture in advanced Parkinson’s disease after chronic bilateral subthalamic stimulation. J Neurol Neurosurg Psychiatry. 2002;72(5):661–664.
    1. Monaca C, Ozsancak C, Jacquesson JM, et al. Effects of bilateral subthalamic stimulation on sleep in Parkinson’s disease. J Neurol. 2004;251(2):214–218.
    1. Kedia S, Moro E, Tagliati M, Lang AE, Kumar R. Emergence of restless legs syndrome during subthalamic stimulation for Parkinson disease. Neurology. 2004;63(12):2410–2412.
    1. Arnulf I, Bejjani BP, Garma L, et al. Effect of low and high frequency thalamic stimulation on sleep in patients with Parkinson’s disease and essential tremor. J Sleep Res. 2000;9(1):55–62.
    1. Larsen JP, Tandberg E. Sleep disorders in patients with Parkinson’s disease: epidemiology and management. CNS Drugs. 2001;15(4):267–275.
    1. Wakabayashi K, Takahashi H, Ohama E, Takeda S, Ikuta F. Lewy bodies in the visceral autonomic nervous system in Parkinson’s disease. Adv Neurol. 1993;60:609–612.
    1. Lahrmann H, Cortelli P, Hilz M, Mathias CJ, Struhal W, Tassinari M. EFNS guidelines on the diagnosis and management of orthostatic hypotension. Eur J Neurol. 2006;13(9):930–936.
    1. Aranda B, Cramer P. Effects of apomorphine and L-dopa on the parkinsonian bladder. Neurourol Urodyn. 1993;12(3):203–209.
    1. Christmas TJ, Kempster PA, Chapple CR, et al. Role of subcutaneous apomorphine in parkinsonian voiding dysfunction. Lancet. 1988;2(8626–8627):1451–1453.
    1. Soykan I, Sarosiek I, Shifflett J, Wooten GF, McCallum RW. Effect of chronic oral domperidone therapy on gastrointestinal symptoms and gastric emptying in patients with Parkinson’s disease. Mov Disord. 1997;12(6):952–957.
    1. Restivo DA, Palmeri A, Marchese-Ragona R. Botulinum toxin for cricopharyngeal dysfunction in Parkinson’s disease. N Engl J Med. 2002;346(15):1174–1175.
    1. El Sharkawi A, Ramig L, Logemann JA, et al. Swallowing and voice effects of Lee Silverman Voice Treatment (LSVT): a pilot study. J Neurol Neurosurg Psychiatry. 2002;72(1):31–36.
    1. Briganti A, Salonia A, Gallina A, et al. Drug insight: oral phosphodiesterase type 5 inhibitors for erectile dysfunction. Nat Clin Pract Urol. 2005;2(5):239–247.
    1. Lawrence AD, Evans AH, Lees AJ. Compulsive use of dopamine replacement therapy in Parkinson’s disease: reward systems gone awry? Lancet Neurol. 2003;2(10):595–604.
    1. Evans AH, Lees AJ. Dopamine dysregulation syndrome in Parkinson’s disease. Curr Opin Neurol. 2004;17(4):393–398.
    1. Thomas A, Bonanni L, Gambi F, Di Iorio A, Onofrj M. Pathological gambling in Parkinson disease is reduced by amantadine. Ann Neurol. 2010;68(3):400–404.
    1. Cummings JL. Behavioral complications of drug treatment of Parkinson’s disease. J Am Geriatr Soc. 1991;39(7):708–716.
    1. Henderson MJ, Mellers JDC. Psychosis in Parkinson’s disease: “between a rock and a hard place.”. Int Rev Psychiatry. 2000;12(4):319–334.
    1. Sanchez-Ramos JR, Ortoll R, Paulson GW. Visual hallucinations associated with Parkinson disease. Arch Neurol. 1996;53(12):1265–1268.
    1. Pacchetti C, Manni R, Zangaglia R, et al. Relationship between hallucinations, delusions, and rapid eye movement sleep behavior disorder in Parkinson’s disease. Mov Disord. 2005;20(11):1439–1448.
    1. Aarsland D, Larsen JP, Cummins JL, Laake K. Prevalence and clinical correlates of psychotic symptoms in Parkinson disease: a community-based study. Arch Neurol. 1999;56(5):595–601.
    1. Fenelon G, Mahieux F, Huon R, Ziegler M. Hallucinations in Parkinson’s disease: prevalence, phenomenology and risk factors. Brain. 2000;123(Pt 4):733–745.
    1. Aarsland D, Ballard C, Larsen JP, McKeith I. A comparative study of psychiatric symptoms in dementia with Lewy bodies and Parkinson’s disease with and without dementia. Int J Geriatr Psychiatry. 2001;16(5):528–536.
    1. Marsh L, Williams JR, Rocco M, Grill S, Munro C, Dawson TM. Psychiatric comorbidities in patients with Parkinson disease and psychosis. Neurology. 2004;63(2):293–300.
    1. Factor SA, Friedman JH, Lannon MC, Oakes D, Bourgeois K. Clozapine for the treatment of drug-induced psychosis in Parkinson’s disease: results of the 12 week open label extension in the PSYCLOPS trial. Mov Disord. 2001;16(1):135–139.
    1. Morgante L, Epifanio A, Spina E, et al. Quetiapine and clozapine in parkinsonian patients with dopaminergic psychosis. Clin Neuropharmacol. 2004;27(4):153–156.
    1. Ondo WG, Tintner R, Voung KD, Lai D, Ringholz G. Double-blind, placebo-controlled, unforced titration parallel trial of quetiapine for dopaminergic-induced hallucinations in Parkinson’s disease. Mov Disord. 2005;20(8):958–963.
    1. Merims D, Balas M, Peretz C, Shabtai H, Giladi N. Rater-blinded, prospective comparison: quetiapine versus clozapine for Parkinson’s disease psychosis. Clin Neuropharmacol. 2006;29(6):331–337.
    1. Bergman J, Lerner V. Successful use of donepezil for the treatment of psychotic symptoms in patients with Parkinson’s disease. Clin Neuropharmacol. 2002;25(2):107–110.
    1. Aarsland D, Hutchinson M, Larsen JP. Cognitive, psychiatric and motor response to galantamine in Parkinson’s disease with dementia. Int J Geriatr Psychiatry. 2003;18(10):937–941.
    1. Bohnen NI, Kaufer DI, Ivanco LS, et al. Cortical cholinergic function is more severely affected in parkinsonian dementia than in Alzheimer disease: an in vivo positron emission tomographic study. Arch Neurol. 2003;60(12):1745–1748.
    1. Emre M, Aarsland D, Albanese A, et al. Rivastigmine for dementia associated with Parkinson’s disease. N Engl J Med. 2004;351(24):2509–2518.
    1. Aarsland D, Ballard C, Walker Z, et al. Memantine in patients with Parkinson’s disease dementia or dementia with Lewy bodies: a double-blind, placebo-controlled, multicentre trial. Lancet Neurol. 2009;8(7):613–618.
    1. Leroi I, Overshott R, Byrne EJ, Daniel E, Burns A. Randomized controlled trial of memantine in dementia associated with Parkinson’s disease. Mov Disord. 2009;24(8):1217–1221.
    1. Lemke MR, Fuchs G, Gemende I, et al. Depression and Parkinson’s disease. J Neurol. 2004;251(Suppl 6):VI/24–VI/27.
    1. Global Parkinson’s Disease Survey Steering Committee. Factors impacting on quality of life in Parkinson’s disease: results from an international survey. Mov Disord. 2002;17(1):60–67.
    1. Kuopio AM, Marttila RJ, Helenius H, Toivonen M, Rinne UK. The quality of life in Parkinson’s disease. Mov Disord. 2000;15(2):216–223.
    1. Schrag A, Jahanshahi M, Quinn N. What contributes to quality of life in patients with Parkinson’s disease? J Neurol Neurosurg Psychiatry. 2000;69(3):308–312.
    1. Remy P, Doder M, Lees A, Turjanski N, Brooks D. Depression in Parkinson’s disease: loss of dopamine and noradrenaline innervation in the limbic system. Brain. 2005;128(Pt 6):1314–1322.
    1. Maj J, Rogoz Z, Skuza G, Kolodziejczyk K. The behavioural effects of pramipexole, a novel dopamine receptor agonist. Eur J Pharmacol. 1997;324(1):31–37.
    1. Willner P, Lappas S, Cheeta S, Muscat R. Reversal of stress-induced anhedonia by the dopamine receptor agonist, pramipexole. Psychopharmacology (Berl) 1994;115(4):454–462.
    1. Lemke MR, Brecht HM, Koester J, Kraus PH, Reichmann H. Anhedonia, depression, and motor functioning in Parkinson’s disease during treatment with pramipexole. J Neuropsychiatry Clin Neurosci. 2005;17(2):214–220.
    1. Antonini A, Tolosa E. Apomorphine and levodopa infusion therapies for advanced Parkinson’s disease: selection criteria and patient management. Expert Rev Neurother. 2009;9(6):859–867.
    1. Lundqvist C. Continuous levodopa for advanced Parkinson’s disease. Neuropsychiatr Dis Treat. 2007;3(3):335–348.
    1. Devos D. Patient profile, indications, efficacy and safety of duodenal levodopa infusion in advanced Parkinson’s disease. Mov Disord. 2009;24(7):993–1000.
    1. Poewe W, Kleedorfer B, Wagner M, Bosch S, Schelosky L. Continuous subcutaneous apomorphine infusions for fluctuating Parkinson’s disease. Long-term follow-up in 18 patients. Adv Neurol. 1993;60:656–659.
    1. Stibe CM, Lees AJ, Kempster PA, Stern GM. Subcutaneous apomorphine in parkinsonian on–off oscillations. Lancet. 1988;1(8582):403–406.
    1. Frankel JP, Lees AJ, Kempster PA, Stern GM. Subcutaneous apomorphine in the treatment of Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1990;53(2):96–101.
    1. McIntyre CC, Savasta M, Kerkerian-Le Goff L, Vitek JL. Uncovering the mechanism(s) of action of deep brain stimulation: activation, inhibition, or both. Clin Neurophysiol. 2004;115(6):1239–1248.
    1. Allert N, Lehrke R, Sturm V, Volkmann J. Secondary failure after ten years of pallidal neurostimulation in a patient with advanced Parkinson’s disease. J Neural Transm. 2010;117(3):349–351.
    1. Volkmann J. Deep brain stimulation for the treatment of Parkinson’s disease. J Clin Neurophysiol. 2004;21(1):6–17.
    1. Volkmann J, Daniels C, Witt K. Neuropsychiatric effects of subthalamic neurostimulation in Parkinson disease. Nat Rev Neurol. 2010;6(9):487–498.
    1. Voon V, Krack P, Lang AE, et al. A multicentre study on suicide outcomes following subthalamic stimulation for Parkinson’s disease. Brain. 2008;131(Pt 10):2720–2728.
    1. Daniels C, Krack P, Volkmann J, et al. Risk factors for executive dysfunction after subthalamic nucleus stimulation in Parkinson’s disease. Mov Disord. 2010;25(11):1583–1589.
    1. York MK, Wilde EA, Simpson R, Jankovic J. Relationship between neuropsychological outcome and DBS surgical trajectory and electrode location. J Neurol Sci. 2009;287(1–2):159–171.
    1. Weaver FM, Follett KA, Stern M, et al. Randomized trial of deep brain stimulation for Parkinson disease: thirty-six-month outcomes. Neurology. 2012;79(1):55–65.
    1. Krack P, Dostrovsky J, Ilinsky I, et al. Surgery of the motor thalamus: problems with the present nomenclatures. Mov Disord. 2002;17(Suppl 3):S2–S8.
    1. Wojtecki L, Timmermann L, Jorgens S, et al. Frequency-dependent reciprocal modulation of verbal fluency and motor functions in subthalamic deep brain stimulation. Arch Neurol. 2006;63(9):1273–1276.
    1. Mason A, Ilinsky IA, Maldonado S, Kultas-Ilinsky K. Thalamic terminal fields of individual axons from the ventral part of the dentate nucleus of the cerebellum in Macaca mulatta. J Comp Neurol. 2000;421(3):412–428.
    1. Hamani C, Moro E, Lozano AM. The pedunculopontine nucleus as a target for deep brain stimulation. J Neural Transm. 2011;118(10):1461–1468.
    1. Stefani A, Lozano AM, Peppe A, et al. Bilateral deep brain stimulation of the pedunculopontine and subthalamic nuclei in severe Parkinson’s disease. Brain. 2007;130(Pt 6):1596–1607.
    1. Costa A, Carlesimo GA, Caltagirone C, et al. Effects of deep brain stimulation of the peduncolopontine area on working memory tasks in patients with Parkinson’s disease. Parkinsonism Relat Disord. 2010;16(1):64–67.
    1. Stefani A, Pierantozzi M, Ceravolo R, Brusa L, Galati S, Stanzione P. Deep brain stimulation of pedunculopontine tegmental nucleus (PPTg) promotes cognitive and metabolic changes: a target-specific effect or response to a low-frequency pattern of stimulation? Clin EEG Neurosci. 2010;41(2):82–86.
    1. Zahodne LB, Okun MS, Foote KD, et al. Greater improvement in quality of life following unilateral deep brain stimulation surgery in the globus pallidus as compared to the subthalamic nucleus. J Neurol. 2009;256(8):1321–1329.
    1. Follett KA, Weaver FM, Stern M, et al. Pallidal versus subthalamic deep-brain stimulation for Parkinson’s disease. N Engl J Med. 2010;362(22):2077–2091.
    1. Okun MS, Fernandez HH, Wu SS, et al. Cognition and mood in Parkinson’s disease in subthalamic nucleus versus globus pallidus interna deep brain stimulation: the COMPARE trial. Ann Neurol. 2009;65(5):586–595.
    1. Charles PD, Gill CE, Davis TL, Konrad PE, Benabid AL. Is deep brain stimulation neuroprotective if applied early in the course of PD? Nat Clin Pract Neurol. 2008;4(8):424–426.
    1. Deuschl G, Schade-Brittinger C, Krack P, et al. A randomized trial of deep-brain stimulation for Parkinson’s disease. N Engl J Med. 2006;355(9):896–908.
    1. Weaver F, Follett K, Hur K, Ippolito D, Stern M. Deep brain stimulation in Parkinson disease: a metaanalysis of patient outcomes. J Neurosurg. 2005;103(6):956–967.
    1. Williams A, Gill S, Varma T, et al. Deep brain stimulation plus best medical therapy versus best medical therapy alone for advanced Parkinson’s disease (PD SURG trial): a randomised, open-label trial. Lancet Neurol. 2010;9(6):581–591.
    1. Rascol O, Dubois B, Caldas AC, Senn S, Del Signore S, Lees A. Early piribedil monotherapy of Parkinson’s disease: a planned seven-month report of the REGAIN study. Mov Disord. 2006;21(12):2110–2115.
    1. Parkinson Study Group CALM Cohort Investigators. Long-term effect of initiating pramipexole vs levodopa in early Parkinson disease. Arch Neurol. 2009;66(5):563–570.
    1. Barone P, Scarzella L, Marconi R, et al. Pramipexole versus sertraline in the treatment of depression in Parkinson’s disease: a national multicenter parallel-group randomized study. J Neurol. 2006;253(5):601–607.
    1. Pahwa R, Lyons KE. Options in the treatment of motor fluctuations and dyskinesias in Parkinson’s disease: a brief review. Neurol Clin. 2004;22(Suppl 3):S35–S52.
    1. Wolf E, Seppi K, Katzenschlager R, et al. Long-term antidyskinetic efficacy of amantadine in Parkinson’s disease. Mov Disord. 2010;25(10):1357–1363.
    1. Durif F, Debilly B, Galitzky M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology. 2004;62(3):381–388.
    1. Spieker S, Eisebitt R, Breit S, et al. Tremorlytic activity of budipine in Parkinson’s disease. Clin Neuropharmacol. 1999;22(2):115–119.
    1. Friedman JH, Koller WC, Lannon MC, Busenbark K, Swanson-Hyland E, Smith D. Benztropine versus clozapine for the treatment of tremor in Parkinson’s disease. Neurology. 1997;48(4):1077–1081.
    1. Anderson KN, Shneerson JM. Drug treatment of REM sleep behavior disorder: the use of drug therapies other than clonazepam. J Clin Sleep Med. 2009;5(3):235–239.
    1. Stocchi F, Barbato L, Nordera G, Berardelli A, Ruggieri S. Sleep disorders in Parkinson’s disease. J Neurol. 1998;245(Suppl 1):S15–S18.
    1. Ferini-Strambi L, Aarskog D, Partinen M, et al. Effect of pramipexole on RLS symptoms and sleep: a randomized, double-blind, placebo-controlled trial. Sleep Med. 2008;9(8):874–881.
    1. Montagna P, Hornyak M, Ulfberg J, et al. Randomized trial of pramipexole for patients with restless legs syndrome (RLS) and RLS-related impairment of mood. Sleep Med. 2011;12(1):34–40.
    1. Oertel WH, Stiasny-Kolster K, Bergtholdt B, et al. Efficacy of pramipexole in restless legs syndrome: a six-week, multicenter, randomized, double-blind study (effect-RLS study) Mov Disord. 2007;22(2):213–219.
    1. Bogan RK, Fry JM, Schmidt MH, Carson SW, Ritchie SY. Ropinirole in the treatment of patients with restless legs syndrome: a US-based randomized, double-blind, placebo-controlled clinical trial. Mayo Clin Proc. 2006;81(1):17–27.
    1. Trenkwalder C, Garcia-Borreguero D, Montagna P, et al. Ropinirole in the treatment of restless legs syndrome: results from the TREAT RLS 1 study, a 12 week, randomised, placebo controlled study in 10 European countries. J Neurol Neurosurg Psychiatry. 2004;75(1):92–97.
    1. Hening WA, Allen RP, Ondo WG, et al. Rotigotine improves restless legs syndrome: a 6-month randomized, double-blind, placebo-controlled trial in the United States. Mov Disord. 2010;25(11):1675–1683.
    1. Oertel WH, Benes H, Garcia-Borreguero D, et al. Rotigotine transdermal patch in moderate to severe idiopathic restless legs syndrome: a randomized, placebo-controlled polysomnographic study. Sleep Med. 2010;11(9):848–856.
    1. Kushida CA, Becker PM, Ellenbogen AL, Canafax DM, Barrett RW. Randomized, double-blind, placebo-controlled study of XP13512/GSK1838262 in patients with RLS. Neurology. 2009;72(5):439–446.
    1. Kushida CA, Walters AS, Becker P, et al. A randomized, double-blind, placebo-controlled, crossover study of XP13512/GSK1838262 in the treatment of patients with primary restless legs syndrome. Sleep. 2009;32(2):159–168.
    1. Lee DO, Ziman RB, Perkins AT, Poceta JS, Walters AS, Barrett RW. A randomized, double-blind, placebo-controlled study to assess the efficacy and tolerability of gabapentin enacarbil in subjects with restless legs syndrome. J Clin Sleep Med. 2011;7(3):282–292.
    1. Walters AS, Ondo WG, Kushida CA, et al. Gabapentin enacarbil in restless legs syndrome: a phase 2b, 2-week, randomized, double-blind, placebo-controlled trial. Clin Neuropharmacol. 2009;32(6):311–320.
    1. Winkelman JW, Bogan RK, Schmidt MH, Hudson JD, DeRossett SE, Hill-Zabala CE. Randomized polysomnography study of gabapentin enacarbil in subjects with restless legs syndrome. Mov Disord. 2011;26(11):2065–2072.
    1. Hogl B, Paulus W, Clarenbach P, Trenkwalder C. Restless legs syndrome: diagnostic assessment and the advantages and risks of dopaminergic treatment. J Neurol. 2006;253(Suppl 4):IV22–IV28.
    1. Garcia-Borreguero D, Larrosa O, de la Llave Y, Verger K, Masramon X, Hernandez G. Treatment of restless legs syndrome with gabapentin: a double-blind, cross-over study. Neurology. 2002;59(10):1573–1579.
    1. Happe S, Sauter C, Klosch G, Saletu B, Zeitlhofer J. Gabapentin versus ropinirole in the treatment of idiopathic restless legs syndrome. Neuropsychobiology. 2003;48(2):82–86.
    1. Schoffer KL, Henderson RD, O’Maley K, O’Sullivan JD. Nonpharmacological treatment, fludrocortisone, and domperidone for orthostatic hypotension in Parkinson’s disease. Mov Disord. 2007;22(11):1543–1549.
    1. Zangaglia R, Martignoni E, Glorioso M, et al. Macrogol for the treatment of constipation in Parkinson’s disease. A randomized placebo-controlled study. Mov Disord. 2007;22(9):1239–1244.
    1. 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 Psychiatry. 2001;71(3):371–374.
    1. Fernandez HH, Okun MS, Rodriguez RL, et al. Quetiapine improves visual hallucinations in Parkinson disease but not through normalization of sleep architecture: results from a double-blind clinical-polysomnography study. Int J Neurosci. 2009;119(12):2196–2205.
    1. Rabey JM, Prokhorov T, Miniovitz A, Dobronevsky E, Klein C. Effect of quetiapine in psychotic Parkinson’s disease patients: a double-blind labeled study of 3 months’ duration. Mov Disord. 2007;22(3):313–318.
    1. Shotbolt P, Samuel M, David A. Quetiapine in the treatment of psychosis in Parkinson’s disease. Ther Adv Neurol Disord. 2010;3(6):339–350.
    1. Emre M, Tsolaki M, Bonuccelli U, et al. Memantine for patients with Parkinson’s disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010;9(10):969–977.
    1. Barone P, Poewe W, Albrecht S, et al. Pramipexole for the treatment of depressive symptoms in patients with Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010;9(6):573–580.
    1. 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. 2003;10(4):399–406.
    1. Devos D, Dujardin K, Poirot I, et al. Comparison of desipramine and citalopram treatments for depression in Parkinson’s disease: a double-blind, randomized, placebo-controlled study. Mov Disord. 2008;23(6):850–857.
    1. Menza M, Dobkin RD, Marin H, et al. A controlled trial of antidepressants in patients with Parkinson disease and depression. Neurology. 2009;72(10):886–892.
    1. Eggert K, Oertel WH, Reichmann H, et al. Parkinson-Syndrome: Diagnostik und Therapie. In: Diener HC, Weimar C, editors. Leitlinien für Diagnostik und Therapie in der Neurologie. Vol. 82. Stuttgart: Thieme; 2012. p. 112. Leitlinien für Diagnostik und Therapie in der Neurologie 5 überarb Auflage. [German]
    1. Schuepbach WM, Rau J, Knudsen K, et al. EARLYSTIM Study Group. Neurostimulation for Parkinson’s disease with early motor complications. N Engl J Med. 2013 Feb 14;368(7):610–622. doi: 10.1056/NEJMoa1205158.

Source: PubMed

3
Sottoscrivi