Injections

in the wrist can cause hand weakness, and dysphagia, hoarseness, and breathiness

can occur with injections into the neck or vocal cords. The use of botulinum toxin

injections in the United States is also limited by cost. Treatment should occur with

the lowest dose, and the interval should be as long as possible between injections.

DBS of the ventral intermediate nucleus of the thalamus or unilateral thalamotomy is

possibly efficacious in reducing ET.

170 Greater improvement in self-reported

measures of function and fewer adverse events make DBS the preferred surgical

option of the two.

170

Because K.H. is otherwise healthy, she is a good candidate for propranolol

therapy. Propranolol can be initiated as needed or on a scheduled basis depending on

the degree of impairment and desire of the patient. If the decision is made with K.H.

to use propranolol on an as-needed basis, she should begin with one-half of a 20-mg

tablet administered 30 minutes to 1 hour before the desired effect. The dose can be

increased from one-half to two tablets. An example of a situation in which this may

occur is whether she wants to avoid embarrassment with attending a social activity

or before certain tasks requiring manual dexterity at work. Given the degree of her

impairment, she is probably a better candidate for chronic suppressive therapy with

propranolol. In this situation, she can be prescribed 10 mg twice daily, because it can

be safely and easily up-titrated every few days to a minimally effective dose,

generally not more than 120 to 360 mg/day in divided doses.

KEY REFERENCES AND WEBSITES

A full list of references for this chapter can be found at

http://thepoint.lww.com/AT11e. Below are the key references and websites for this

chapter, with the corresponding reference number in this chapter found in parentheses

after the reference.

Key References

Ferreira JJ et al. Summary of the recommendations of the EFNS/MDS-ES review on therapeutic management of

Parkinson’s disease. Eur J Neurol. 2013;20:5–15. (20)

Fox SH et al. Update on treatments for motor symptom of PD. Update for Website; 2015.

Garcia-Borreguero D et al; European Federation of Neurological Societies; European Neurological Society;

European Sleep Research Society. European guidelines on management of restless legs syndrome: report of a

joint task force by the European Federation of Neurological Societies, the European Neurological Society and

the European Sleep Research Society. Eur J Neurol. 2012;19(11):1385–1396. (157)

Garcia-Borreguero D et al. The long-term treatment of restless legs syndrome/Willis-Ekbom disease: evidencebased guidelines and clinical consensus best practice guidance: a report from the International Restless Legs

Syndrome Study Group. Sleep Med. 2013;14(7): 675–684. (156)

Miyasaki JM et al. Practice parameter: initiation of treatment for Parkinson’s disease: an evidence-based review:

report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2002;58:11.

(15)

Miyasaki JM et al. Practice parameter: evaluation and treatment of depression, psychosis, and dementia in

Parkinson’s disease (an evidence-based review): report of the Quality Standards Subcommittee of the American

Academy of Neurology. Neurology. 2006;66:996.

National Collaborating Centre for Chronic Conditions. Parkinson’s disease: national clinical guideline for diagnosis

and management in primary and secondary care. London: Royal College of Physicians; 2006. (17)

Pahwa R 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. Neurology. 2006;66:983. (16)

PD Med Collaborative Group, Gray R et al. Long-term effectiveness of dopamine agonists and monoamine

oxidase-B inhibitors compared with levodopa as initial treatment for Parkinson’s disease (PD MED): a large,

open-label, pragmatic randomised trial. Lancet. 2014;384(9949):1196–1205. (28)

Zesiewicz TA et al. Practice parameter: treatment of non-motor symptoms of Parkinson disease: report of the

Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2010;74:924. (130)

Zesiewicz TA et al. Evidence-based guideline update: treatment of essential tremor: report of the Quality

Standards subcommittee of the American Academy of Neurology. Neurology. 2011;77(19):1752–1755. (169)

COMPLETE REFERENCES CHAPTER 59 PARKINSON’s

DISEASE AND OTHER MOVEMENT DISORDERS

Rao G et al. Does this patient have Parkinson disease? JAMA. 2003;289:347.

Hoehn MM, Yahr HD. Parkinsonism: onset, progression, and mortality. Neurology. 1967;17:427.

Van Den Eeden SK et al. Incidence of Parkinson’s disease: variation by age, gender, and race/ethnicity. Am J

Epidemiol. 2003;157:1015.

Twelves D et al. Systematic review of incidence studies of Parkinson’s disease. Mov Disord. 2003;18:19.

Fall PA et al. Survival time, mortality, and cause of death in elderly patients with Parkinson’s disease: a 9-year

follow-up. Mov Disord. 2003;18:1312.

Suchowersky O et al. Practice parameter: diagnosis and prognosis of new onset Parkinson disease (an evidence

based review): report of the Quality Standards Subcommittee of the American Academy of Neurology.

Neurology. 2006;66:968.

de Lau et al. Epidemiology of Parkinson’s disease. Lancet Neurol. 2006;5(6):525–535.

SanyalJ et al. Environmental and familial risk factors of Parkinsons disease: a case-controlstudy. Can J Neurol

Sci. 2010;37:637.

Morley JF, Hurtig HI. Current understanding and management of Parkinson disease: five new things. Neurology.

2010;75(18, Suppl 1):S9.

Vance JM et al. Gene-environment interactions in Parkinson’s disease and other forms of parkinsonism.

Neurotoxicology. 2010;31:598.

Nutt JG, Wooten GF. Clinical practice. Diagnosis and initial management of Parkinson’s disease. N EnglJ Med.

2005;353:1021–1027.

Braak H et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197.

Savica R et al. When does Parkinson disease start? Arch Neurol. 2010;67:798.

Massano J, Bhatia KP. Clinical approach to Parkinson’s disease: features, diagnosis, and principles of

management. Cold Spring Harb Perspect Med. 2012;2(6):a008870.

MiyasakiJM et al. Practice parameter: initiation of treatment for Parkinson’s disease: an evidence-based review:

report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2002;58:11.

Pahwa R 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. Neurology. 2006;66:983.

National Collaborating Centre for Chronic Conditions. Parkinson’s disease: in adults. July 2017. Available at:

https://www.nice.org.uk/guidance/ng71. Accessed July 2017.

Connolly BS, Lang AE. Pharmacological treatment of Parkinson disease: a review. JAMA. 2014;311(16):1670–

1683.

Olanow CW et al. An algorithm (decision tree) for the management of Parkinson’s disease (2001): treatment

guidelines. Neurology. 2001;56(11) (S5):S1–S88.

Ferreira JJ et al. Summary of the recommendations of the EFNS/MDS-ES review on therapeutic management of

Parkinson’s disease. Eur J Neurol. 2013;20:5–15.

Shannon KM et al. Efficacy of pramipexole, a novel dopamine agonist, as monotherapy in mild to moderate

Parkinson’s disease. The Pramipexole Study Group [published correction appears in Neurology. 1998;50:838].

Neurology. 1997;49:724.

Rascol O 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 EnglJ Med. 2000;342:1484.

Holloway RG et al. Pramipexole vs levodopa as initial treatment for Parkinson’s disease: a 4-year randomized

controlled trial [published correction appears in Arch Neurol. 2005;62:430]. Arch Neurol. 2004;61:1044.

Parkinson Study Group. Pramipexole vs. levodopa as initial treatment for Parkinson disease: a randomized

controlled trial. Parkinson Study Group. JAMA. 2000;284:1931.

Parkinson Study Group CALM Cohort Investigators. Long term effect of initiating pramipexole vs levodopa in

early Parkinson disease. Arch Neurol. 2009;66:563.

Hauser RA et al. Ten-year follow-up of Parkinson’s disease patients randomized to initial therapy with ropinirole

or levodopa. Mov Disord. 2007;22:2409.

Katzenschlager R et al; Parkinson’s Disease Research Group of the United Kingdom. Fourteen-year final report

of the randomized PDRG-UK trial comparing three initial treatments in PD. Neurology. 2008;71(7):474–480.

PD Med Collaborative Group, Gray R et al. Long-term effectiveness of dopamine agonists and monoamine

oxidase B inhibitors compared with levodopa as initial treatment for Parkinson’s disease (PD MED): a large,

open-label, pragmatic randomised trial. Lancet. 2014;384(9949):1196–1205.

Turnbull K et al. Monoamine oxidase B inhibitors for early Parkinson’s disease. Cochrane Database of

Systematic Reviews 2005, Issue 3.

Ives NJ et al. Monoamine oxidase type B inhibitors in early Parkinsons’s disease: meta analysis of 17 randomised

trials involving 3525 patients. Br Med J. 2004;329(7466):593–596.

Schade R et al. Dopamine agonists and the risk of cardiac valve regurgitation. N EnglJ Med. 2007;356:29.

Zanettini R et al. Valvular heart disease and the use of dopamine agonists for Parkinson’s disease. N EnglJ Med.

2007;356:39.

Beaulieu JM, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol

Rev. 2011;63(1):182–217.

Mirapex [package insert]. Ridgefield, CT: Boehringer Ingelheim Pharmaceuticals; 2015.

Hubble JP et al. Pramipexole in patients with early Parkinson’s disease. Clin Neuropharmacol. 1995;18:338.

Parkinson Study Group. Safety and efficacy of pramipexole in early Parkinson’s disease. A randomized doseranging study. Parkinson Study Group. JAMA. 1997;278:125.

Requip [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2014.

Rascol O et al. Ropinirole in the treatment of early Parkinson’s disease: a 6-month interim report of a 5-year

levodopa controlled study. 056 Study Group. Mov Disord. 1998;13:39.

Adler CH et al. Ropinirole for the treatment of early Parkinson’s disease. The Ropinirole Study Group.

Neurology. 1997;49:393.

Korczyn AD et al. Ropinirole versus bromocriptine in the treatment of early Parkinson’s disease: a 6-month

interim report of a 3-year study. 053 Study Group. Mov Disord. 1998;13:46.

The Parkinson Study Group. A controlled trial of rotigotine monotherapy in early Parkinson’s disease. Arch

Neurol. 2003;60:1721.

Güldenpfennig WM et al. Safety, tolerability, and efficacy of continuous transdermal dopaminergic stimulation

with rotigotine patch in early-stage idiopathic Parkinson disease. Clin Neuropharmacol. 2005;28:106.

Watts RL et al. Randomized, blind, controlled trial of transdermal rotigotine in early Parkinson disease. [published

corrections appear in Neurology. 2007;69:2187; Neurology. 2007;69:617]. Neurology. 2007;68:272.

Giladi N et al. Rotigotine transdermal patch in early Parkinson’s disease: a randomized, double-blind, controlled

study versus placebo and ropinirole. Mov Disord. 2007;22:2398–2404.

LeWitt PA et al. Advanced Parkinson disease treated with rotigotine transdermal system: PREFER Study.

Neurology. 2007;68:1262.

Poewe WH et al. Efficacy of pramipexole and transdermal rotigotine in advanced Parkinson’s disease: a doubleblind, double-dummy, randomised controlled trial. Lancet Neurol. 2007;6:513–520.

Nomoto M et al; Rotigotine Trial Group. Transdermal rotigotine in early stage Parkinson’s disease: a randomized,

double-blind, placebo-controlled trial. Mov Disord. 2013;28(10):1447–1450.

Fox SH et al. Update on treatments for motor symptom of PD. (http://www.movementdisorders.org/MDSFiles1/PDFs/EBM-Papers/update-on-treatments-for-motor-symptoms-of-PD.pdf).

Zhou CQ et al. Meta-analysis of the efficacy and safety of long-acting non- ergot dopamine agonists in

Parkinson’s disease. J Clin Neurosci. 2014;21(7): 1094–1101.

Lieberman A et al. Clinical evaluation of pramipexole in advanced Parkinsons disease: results of a double-blind,

placebo-controlled, parallel-group study. Neurology. 1997;49:162.

Elmer LW et al. Long term safety and tolerability of rotigotine transdermal system in idiopathic Parkinson’s

disease: a prospective, open label extension study. Parkinsonism Relat Disord. 2012;18(5):488–493.

Chung SJ et al. Switch from oral pramipexole or ropinirole to rotigotine transdermal system in advanced

Parkinson’s disease: an open-labelstudy. Expert Opin Pharmacother. 2105;16(7):961–970.

Lieberman A et al. A multicenter trial of ropinirole as adjunct treatment for Parkinson’s disease. Ropinirole Study

Group [published correction appears in Neurology. 1999;52:435]. Neurology. 1998;51:1057.

Homann CN et al. Sleep attacks in patients taking dopamine agonists: review. BMJ. 2002;324(7352):1483–1487.

Weintraub D et al. Impulse control disorders in Parkinson disease: a cross-sectionalstudy of 3090 patients. Arch

Neurol. 2010;67:589.

Avanzi M et al. Prevalence of pathological gambling in patients with Parkinson’s disease. Mov Disord.

2006;21:2068.

O’Sullivan SS et al. Dopamine dysregulation syndrome: an overview of its epidemiology, mechanisms and

management. CNS Drugs. 2009;23(2):157–170.

Evans AH et al. Factors influencing susceptibility to compulsive dopaminergic drug use in Parkinson disease.

Neurology. 2005;65:1570.

Ceravolo R et al. Spectrum of addictions in Parkinson’s disease: from dopamine dysregulation syndrome to

impulse control disorders. J Neurol. 2010;257(Suppl 2):S276.

Zelapar [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals North America LLC; 2014.

Parkinson Study Group. Effect of deprenyl on the progression of disability in early Parkinson’s disease. N EnglJ

Med. 1989;321:1364.

Parkinson Study Group. Effects of Tocopherol and Deprenyl on the Progression of Disability in Early Parkinson’s

Disease. N EnglJ Med. 1993;328:176–183.

Shoulson I et al. Impact of sustained deprenyl (selegiline) in levodopa-treated Parkinson’s disease: a randomized

placebo-controlled extension of the deprenyl and tocopherol antioxidative therapy of parkinsonism trial. Ann

Neurol. 2002;51:604–612.

Palhagen S et al. Selegiline delays the onset of disability in de novo parkinsonian patients. Swedish Parkinson

Study Group. Neurology. 1998;51:520–525.

Knudsen GD. Selegiline and Rasagiline: twins or distant cousins? Guidelines. Consult Pharm. 2011;1(4):48–51.

Parkinson Study Group. A controlled trial of rasagiline in early Parkinson disease: the TEMPO study. Arch

Neurol. 2002;59:1937.

Parkinson Study Group. A controlled, randomized, delayed start study of rasagiline in early Parkinson disease.

Arch Neurol. 2004;61:561.

Olanow CW et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med.

2009;361:1268.

Jost WH et al. Indirect meta-analysis of randomised placebo-controlled clinical trials on rasagiline and selegiline in

the symptomatic treatment of Parkinson’s disease. Basal Ganglia. 2012;2:S17–S26.

Azilect [package insert]. Overland Park, KS; Teva Neuroscience; 2014.

deMarcaida JA et al. Effects of tyramine administration in Parkinson’s disease patients treated with selective

MAO-B inhibitor rasagiline. Mov Disord. 2006;21:1716.

Eldepryl [Package insert]. St Leonards, NSW: Aspen Pharmacare Australia Pty Ltd; 2010.

Sinemet [package insert]. Morgantown, WV: Merck & Co; 2014.

Fahn S et al. Levodopa and the progression of Parkinson’s disease. N EnglJ Med. 2004;351:2498.

Camargo SM et al. The molecular mechanism of intestinal levodopa absorption and its possible implications for

the treatment of Parkinson’s disease. J Pharmacol Exp Ther. 2014;351(1):114–123.

Riley DE, Lang AE. The spectrum of levodopa-related fluctuations in Parkinson’s disease. Neurology.

1993;43:1459.

Goodwin FK. Psychiatric side effects of levodopa in man. JAMA. 1971;218:1915.

Thanvi B, Lo T. Long term motor complications of levodopa: clinical features, mechanisms, and management

strategies. Postgrad Med J. 2004;80(946):452–458.

Marques de Sousa S, Massano J. Motor complications in Parkinson’s disease: a comprehensive review of

emergent management strategies. CNS Neurol Disord Drug Targets. 2013;12:1017–1049.

Duodopa [package insert]. St-Laurent, QC: AbbVie Corporation; 2015.

100.

101.

102.

103.

104.

105.

106.

107.

108.

109.

110.

111.

Sinemet CR [package insert]. Morgantown, WV: Merck & Co; July 2014.

Hauser RA et al. Crossover comparison of IPX066 and a standard levodopa formulation in advanced Parkinson’s

disease. Mov Disord. 2011;26:2246–2252.

Rytary [package insert]. Hayward, CA: Impax Laboratories; 2015.

Hauser RA et al. Extended-release carbidopa-levodopa (IPX066) compared with immediate-release carbidopalevodopa in patients with Parkinson’s disease and motor fl uctuations: a phase 3 randomised, double-blind trial.

Lancet Neurol. 2013;12:346–356.

Lieberman A et al. Clinical evaluation of pramipexole in advanced Parkinson’s disease: results of a double-blind,

placebo-controlled, parallel-group study. Neurology. 1997;49:162.

Watts RL et al. Onset of dyskinesia with adjunct ropinirole prolonged- release or additional levodopa in early

Parkinson’s disease. Mov Disord. 2010;25:858.

Hersh BP et al. Early treatment benefits of ropinirole prolonged release in Parkinson’s disease patients with

motor fluctuations. Mov Disord. 2010;25:927.

Stocchi F et al. PREPARED: comparison of prolonged and immediate release ropinirole in advanced Parkinson’s

disease. Mov Disord. 2011;26(7): 1259–1265.

Apokyn [package insert]. Louisville, KY: US World Meds, LLC’; 2014.

Schwab RS et al. Amantadine in the treatment of Parkinson’s disease. JAMA. 1969;208:1168.

Fahn S, Isgreen WP. Long-term evaluation of amantadine and levodopa combination in parkinsonism by doubleblind crossover analyses. Neurology. 1975;25:695.

Bailey EV, Stone TW. The mechanism of action of amantadine in Parkinsonism: a review. Arch Int

Pharmacodyn Ther. 1975;216:246.

Greenamayre JT, O’Brien CF. N-methyl-D-aspartate antagonists in the treatment of Parkinson’s disease. Arch

Neurol. 1991;48:977.

Verhagen Metman L et al. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s

disease. Neurology. 1998;50:1323.

Metman LV et al. Amantadine for levodopa-induced dyskinesias: a 1-year follow-up study. Arch Neurol.

1999;56:1383.

Snow BJ et al. The effect of amantadine on levodopa-induced dyskinesias in Parkinson’s disease: a double-blind,

placebo-controlled study. Clin Neuropharmacol. 2000;23:82.

Thomas A et al. Duration of amantadine benefit on dyskinesia of severe Parkinson’s disease. J Neurol Neurosurg

Psychiatry. 2004;75:141.

Ory-Magne F et al; NS-Park CIC Network. Withdrawing amantadine in dyskinetic patients with Parkinson

disease: the AMANDYSK trial. Neurology. 2014;82(4):300–307.

Crosby N et al. Amantadine in Parkinson’s disease. Cochrane Database Syst Rev. 2003;(1):CD003468.

Symmetrel [package insert]. Chadds Ford, PA: Endo Pharmaceuticals; 2009.

Tasmar [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals North America, LLC; 2013.

Comtan [package insert]. East Hanover, NJ; Novartis Pharmaceuticals Corporation; 2014.

De Santi C et al. Catechol-O-methyltransferase: variation in enzyme activity and inhibition by entacapone and

tolcapone. Eur J Clin Pharmacol. 1998;54:215.

Stocchi F et al. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: the

STRIDE-PD study [published correction appears in Ann Neurol. 2010;68:412]. Ann Neurol. 2010;68:18.

Rinne UK et al. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations.

Nomecomt Study Group. Neurology. 1998;51:1309.

Parkinson Study Group. Entacapone improves motor fluctuations in levodopa-treated Parkinson’s disease

patients. [published correction appears in Ann Neurol. 1998;44:292]. Ann Neurol. 1997;42:747.

Gottwald MD. Entacapone, a catechol-O-methyltransferase inhibitor for treating Parkinson’s disease: review

and current status. Expert Opin Investig Drugs. 1999;8(4):453–462.

FDA Drug Safety Communication: FDA Review Found No Increased Cardiovascular Risks with Parkinson’s

Disease Drug Entacapone; 2015.

Golbe LI. Deprenyl as symptomatic therapy in Parkinson’s disease. Clin Neuropharmacol. 1988;11:387.

Elizam TS et al. Selegiline as an adjunct to conventional levodopa therapy in Parkinson’s disease. Experience

with this type B monoamine oxidase inhibitor in 200 patients [published correction appears in Arch Neurol.

1990;47:160]. Arch Neurol. 1989;46:1280.

Waters CH et al. Zydis selegiline reduces off-time in Parkinson’s disease patients with motor fluctuations: a 3-

month, randomized, placebo-controlled study. Mov Disord. 2004;19:426.

112.

113.

114.

115.

116.

117.

118.

119.

120.

121.

122.

123.

124.

125.

126.

127.

128.

129.

130.

131.

132.

133.

134.

135.

136.

137.

138.

Ongo WG et al. Selegiline orally disintegrating tablets in patients with Parkinson disease and ‘‘Wearing Off’’

symptoms. Clin Neuropharmacol. 2007;30(5):295–300.

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. 2005;62:241.

Rascol O 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, doubleblind, parallel-group trial. Lancet. 2005;365:947.

Lang AE et al. Posteroventral medial pallidotomy in advanced Parkinson’s disease. N Engl J Med.

1997;337:1036.

Jankovic J et al. Outcome after stereotactic thalamotomy for parkinsonian, essential and other types of tremor.

Neurosurgery. 1995;37:680.

Weaver FM et al. Bilateral deep brain stimulation vs best medical therapy for patients with advanced Parkinson

disease: a randomized controlled trial. JAMA. 2009;301:63.

Follett KA et al. Pallidal versus subthalamic deep-brain stimulation for Parkinson’s disease. N Engl J Med.

2010;362:2077.

Deuschl G et al. A randomized trial of deep-brain stimulation for Parkinson’s disease [published correction

appears in N EnglJ Med. 2006;355:1289]. N EnglJ Med. 2006;355:896.

Krack P et al. Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson’s

disease. N EnglJ Med. 2003;349:1925.

Rodriguez-Oroz MC et al. Bilateral deep brain stimulation in Parkinson’s disease: a multicentre study with 4

years follow-up. Brain. 2005;128 (Pt 10):2240.

Parkinson Study Group. DATATOP: a multicenter controlled clinical trial in early Parkinson’s disease. Arch

Neurol. 1989;46:1052.

Parkinson Study Group. Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s

disease. N EnglJ Med. 1993;328:176.

Suchowersky O et al. Practice parameter: neuroprotective strategies and alternative therapies for Parkinson

disease (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy

of Neurology [published correction appears in Neurology. 2006;67:299]. Neurology. 2006;66:976.

Storch A et al. Randomized, double-blind, placebo-controlled trial on symptomatic effects of coenzyme Q(10) in

Parkinson disease. Arch Neurol. 2007;64(7):938–944.

The NINDS NET-PD Investigators. A randomized clinical trial of coenzyme Q10 and GPI-1485 in early

Parkinson disease. Neurology. 2007;68: 20–28.

The Parkinson Study Group QE3 Investigators. A Randomized Clinical Trial of High-Dosage Coenzyme Q10 in

Early Parkinson DiseaseNo Evidence of Benefit. JAMA Neurol. 2014;71(5):543–552.

The NINDS NET-PD Investigators. A randomized, double-blind, futility clinical trial of creatine and minocycline

in early Parkinson disease. Neurology. 2006;66:664.

The NINDS NET-PD Investigators. Effect of creatine monohydrate on clinical progression in patients with

Parkinson disease: a randomized clinical trial. JAMA. 2015;313(6):584–593.

Zesiewicz TA et al. Practice parameter: treatment of non-motor symptoms of Parkinson disease: report of the

Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2010;74:924.

Barone P et al. The PRIAMO study: a multicenter assessment of nonmotor symptoms and their impact on

quality of life in Parkinson’s disease. Mov Disord. 2009;24:1641.

Chaudhuri KR et al. Non-motor symptoms of Parkinson’s disease: diagnosis and management. Lancet Neurol.

2006;5(3):235–245.

Hely MA et al. The Sydney multicenter study of Parkinson’s disease: the inevitability of dementia at 20 years.

Mov Disord. 2008;23:837.

Maidment I et al. Cholinesterase inhibitors for Parkinson’s disease dementia. Cochrane Database Syst Rev.

2006;(1):CD004747.

van Laar T et al. Effects of cholinesterase inhibitors in Parkinson’s disease dementia: a review of clinical trial

data. CNS Neurosci Ther. 2011;17(5): 428–441.

Emre M et al. Rivastigmine for dementia associated with Parkinson’s disease. N EnglJ Med. 2004;351:2509.

Miyasaki JM et al. Practice parameter: evaluation and treatment of depression, psychosis, and dementia in

Parkinson’s disease (an evidence-based review): report of the Quality Standards Subcommittee of the

American Academy of Neurology. Neurology. 2006;66:996.

Litvinenko IV et al. Efficacy and safety of galantamine (reminyl) for dementia in patients with Parkinson’s

139.

140.

141.

142.

143.

144.

145.

146.

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

157.

158.

159.

160.

161.

162.

163.

164.

165.

166.

disease (an open controlled trial). Neurosci Behav Physiol. 2008;38(9):937–945.

Poewe W et al. Long-term benefits of rivastigmine in dementia associated with Parkinson’s disease: an active

treatment extension study. Mov Disord 2006. 21(4):456–461.

Schmitt FA et al. Evaluating rivastigmine in mild-to-moderate Parkinson’s disease dementia using ADAS-cog

items. Am J Alzheimers Dis Other Demen. 2010;25(5):407–413.

Madhusoodanan S et al. Extrapyramidal symptoms associated with antidepressants—a review of the literature

and an analysis of spontaneous reports. Ann Clin Psychiatry. 2010;22(3):148–156.

Chen JJ, Marsh L. Anxiety in Parkinson’s disease: identification and management. Ther Adv Neurol Disord.

2014;7(1):52–59.

Fénelon G, Alves G. Epidemiology of psychosis in Parkinson’s disease. J Neurol Sci. 2010;289(1–2):12–17.

Forsaa EB et al. A 12-year population-based study of psychosis in Parkinson disease. Arch Neurol. 2010;67:996.

Rich SS et al. Risperidone versus clozapine in the treatment of psychosis in six patients with Parkinson’s disease

and other akinetic-rigid syndromes. J Clin Psychiatry. 1995;56:556.

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

TRIPASS XR تري باس

Kana Brax Laberax

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

CELEPHI 200 MG, Gélule

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more