Scolaris Content Display Scolaris Content Display

Tratamiento con agonista dopaminérgico en la enfermedad de Parkinson precoz

Collapse all Expand all

References

References to studies included in this review

Argentina {published data only}

Herskovits E, Yorio A, Leston J. Long term bromocriptine treatment in de novo Parkinsonian patients. Medicina (B Aires) 1988;48:345‐50.

Barcelona {published data only}

Kulisevsky J, Garcia‐Sanchez C, Berthier M, Barbanoj M, Pascual‐Sedano B, Gironell A, et al. Chronic effects of dopaminergic replacement on cognitive function in Parkinson's disease: A two‐year follow‐up study of previously untreated patients. Movement Disorders 2000;15:613‐26.

Canada {published data only}

Riopelle RJ, Gawel MJ, Libman I, King DB, McLean DR, Paulseth R, et al. A double‐blind study of bromocriptine and L‐dopa in de novo Parkinson's disease. Short‐term results. European Neurology 1988;28(Suppl 1):11‐4.

Europe Bromocriptine {published data only}

Olsson JE, The European Multicentric Trial Group. Bromocriptine and levodopa in early combination in Parkinson's Disease: first results of the Collaborative European Multicentric Trial. In: Streifler MB, Korczyn AD, Melamed E, Youdim MBH editor(s). Advances in Neurology Vol 53: Parkinson's Disease: Anatomy, Pathology, and Therapy. New York: Raven Press, 1990.

Europe CQA 206‐291 {published data only}

Rascol O, Fabre N, Teravainen H, Poewe W, Lucking C, Rinne U, et al. CQA 206‐291: a novel dopamine agonist in the treatment of Parkinson's disease. Clinical Neuropharmacology 1990;13:303‐11.

Europe PELMOPET {published data only}

Hundemer HP, Lledo A, van Laar T, Quail D, Oertel W, Schwarz J, et al. The safety of pergolide monotherapy in early stage Parkinson's disease: One year interim analysis of a 3‐year double‐blind randomized study of pergolide versus levodopa. Movement Disorders 2000;15(Suppl 3):115 (P611).
Oertel WH, Wolters E, Sampaio C, Gimenez‐Roldan S, Bergamasco B, Dujardin M, et al. Pergolide versus levodopa monotherapy in early Parkinson's disease patients: The PELMOPET study. Movement Disorders 2006;21:343‐53.

France Lisuride {published data only}

Allain H, Destee A, Petit H, Patay M, Schuck S, Bentue‐Ferrer D, et al. Five‐year follow‐up of early lisuride and levodopa combination therapy versus levodopa monotherapy in de novo Parkinson's disease. The French Lisuride Study Group. European Neurology 2000;44:22‐30.

France Piribedil {published data only}

Ziegler M, Castro‐Caldas, Del Signore S, Rascol O. Efficacy of piribedil as early combination to levodopa in patients with stable Parkinson's disease: A 6‐month, randomized, placebo‐controlled study. Movement Disorders 2003;18:418‐25.

Germany/Aust PRADO {published data only}

Przuntek H, Welzel D, Gerlach M, Blumner E, Danielczyk W, Kaiser HJ, et al. Early institution of bromocriptine in Parkinson's disease inhibits the emergence of levodopa‐associated motor side effects. Long‐term results of the PRADO study. Journal of Neural Transmission 1996;103:699‐715.

Glasgow {published data only}

Bakheit AM, Henderson LM, Moore AP, Simpson JA, Thomas M. Long‐term double masked trial of early treatment with L‐dopa plus bromocriptine versus L‐dopa alone in Parkinson's disease. Interim results. European Neurology 1990;30:108‐11.

Int Pergolide Mono {published data only}

Barone P, Bravi D, Bermejo‐Pareja F, Marconi R, Kulisevsky J, Malagu S, et al. Pergolide monotherapy in the treatment of early PD: a randomized, controlled study. Pergolide Monotherapy Study Group. Neurology 1999;53:573‐9.

Int PKDS009 {published data only}

Bracco F, Battaglia A, Chouza C, Dupont E, Gershanik O, Marti Masso JF, et al. The long‐acting dopamine receptor agonist cabergoline in early Parkinson's disease: final results of a 5‐year, double‐blind, levodopa‐controlled study. The PKDS009 Study Group. CNS Drugs 2004;18:733‐46.
Rinne UK, Bracco F, Chouza C, Dupont E, Gershanik O, Marti Masso JF, et al. Cabergoline in the treatment of early Parkinson's disease: Results of the first year of treatment in a double‐blind comparison of cabergoline and levodopa. The PKDS009 Study Group. Neurology 1997;48:363‐8.
Rinne UK, Bracco F, Chouza C, Dupont E, Gershanik O, Marti Masso JF, et al. Early treatment of Parkinson's disease with cabergoline delays the onset of motor complications. Results of a double‐blind levodopa controlled trial. The PKDS009 Study Group. Drugs 1998;55(Suppl 1):23‐30.

Int PSG CALM‐PD {published data only}

Parkinson Study Group. Pramipexole vs. levodopa as initial treatment for Parkinson's disease: A 4‐year randomized controlled trial. Archives of Neurology 2004;61:1044‐53.
Parkinson Study Group. Pramipexole vs. levodopa as initial treatment for Parkinson's disease: A randomized controlled trial. The Journal of the American Medical Association 2000;284:1931‐8.

Int Ropinirole {published data only}

Brooks DJ, Abbott RJ, Lees AJ, Martignoni E, Philcox DV, Rascol O, et al. A placebo‐controlled evaluation of ropinirole, a novel D2 agonist, as sole dopaminergic therapy in Parkinson's disease. Clinical Neuropharmacology 1998;21:101‐7.

International 056 {published data only}

Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five‐year study of the incidence of dyskinesia in patients with early Parkinson's disease who were treated with ropinirole or levodopa. 056 Study Group. The New England Journal of Medicine 2000;342:1484‐91.

Italy A {published data only}

Bergamasco B, Frattola L, Muratorio A, Piccoli F, Mailland F, Parnetti L. Alpha‐dihydroergocryptine in the treatment of de novo Parkinsonian patients: results of a multicentre, randomized, double‐blind, placebo‐controlled study. Acta Neurologica Scandinavica 2000;101:372‐80.

Italy B {published data only}

Martignoni E, Pacchetti C, Sibilla L, Bruggi P, Pedevilla M, Nappi G. Dihydroergocryptine in the treatment of Parkinson's disease: a six months' double‐blind clinical trial. Clinical Neuropharmacology 1991;14:78‐83.

Milan {published data only}

Caraceni T, Musicco M. Levodopa or dopamine agonists, or deprenyl as initial treatment for Parkinson's disease. A randomized multicentre study. Parkinsonism and Related Disorders 2001;7:107‐14.

South America {published data only}

Alarcon F, Cevallos N, Lees AJ. Does combined levodopa and bromocriptine therapy in Parkinson's disease prevent late motor complications?. European Journal of Neurology 1998;5:255‐63.

Spain {published data only}

Gimenez‐Roldan S, Tolosa E, Burguera JA, Chacon J, Liano H, Forcadell F. Early combination of bromocriptine and levodopa in Parkinson's disease: a prospective randomized study of two parallel groups over a total follow‐up period of 44 months including an initial 8‐month double‐blind stage. Clinical Neuropharmacology 1997;20:67‐76.

Sydney {published data only}

Hely MA, Morris JG, Reid WG, O'Sullivan DJ, Williamson PM, Rail D, 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. Journal of Neurology, Neurosurgery, and Psychiatry 1994;57:903‐10.
Hely MA, Morris JG, Traficante R, Reid WG, O'Sullivan DJ, Williamson PM. The Sydney multicentre study of Parkinson's disease: progression and mortality at 10 years. Journal of Neurology, Neurosurgery, and Psychiatry 1999;67:300‐7.

UK‐PDRG {published data only}

Lees AJ, Katzenschlager R, Head J, Ben‐Shlomo Y, on behalf of the Parkinson's Disease Research Group of the United Kingdom. Ten‐year follow‐up of three different initial treatments in de‐novo PD: a randomized trial. Neurology 2001;57:1687‐94.
Parkinson's Disease Research Group in the United Kingdom. Comparisons of therapeutic effects of levodopa, levodopa and selegiline, and bromocriptine in patients with early, mild Parkinson's disease: three year interim report. BMJ 1993;307:469‐72.

UKPDRG Pergolide {published data only}

Grosset K, Grosset D, Lees A, on behalf of the Parkinson's Disease Research Group of the UK. Trial of Subtherapeutic Pergolide in de novo Parkinson's Disease. Movement Disorders 2005;20:363‐366.

USA {published data only}

Olanow CW, Hauser RA, Gauger L, Malapira T, Koller W, Hubble J, et al. The effect of deprenyl and levodopa on the progression of Parkinson's disease. Annals of Neurology 1995;38:771‐7.

USA / Canada PSG {published data only}

Parkinson Study Group. Safety and efficacy of pramipexole in early Parkinson's disease: A randomised dose‐ranging study. The Journal of the American Medical Association 1997;278:125‐30.

USA Bromocriptine {published data only}

Weiner WJ, Factor SA, Sanchez‐Ramos JR, Singer C, Sheldon C, Cornelius L, et al. Early combination therapy (bromocriptine and levodopa) does not prevent motor fluctuations in Parkinson's disease. Neurology 1993;43:21‐7.

USA Pramipexole A {published data only}

Hubble JP, Koller WC, Cutler NR, Sramek JJ, Friedman J, Goetz C, et al. Pramipexole in patients with early Parkinson's disease. Clinical Neuropharmacology 1995;18:338‐47.

USA Pramipexole B {published data only}

Shannon KM, Bennett JP, Friedman JH. Efficacy of pramipexole, a novel dopamine agonist, as monotherapy in mild to moderate Parkinson's disease. The Pramipexole Study Group. Neurology 1997;49:724‐8.

USA Ropinirole {published data only}

Adler CH, Sethi KD, Hauser RA, Davis TL, Hammerstad JP, Bertoni J, et al. Ropinirole for the treatment of early Parkinson's disease. The Ropinirole Study Group. Neurology 1997;49:393‐9.

References to studies excluded from this review

De Lucia {published data only}

De Lucia D, Fratta M, Terracciano M, Quarantiello M. Low incidence of dyskinesia in early Parkinson's disease treated with ropinirole. Parkinsonism and Related Disorders 2001;7:S1‐133 (P‐TU‐176).

Presthus {published data only}

Presthus J, Ankerhus J, Buren A, Bottcher J, Holmsen R, Mikkelsen B, et al. Dopaminergic agonist Ro 8‐4650 in Parkinson's disease. II. Patients not treated with dopa. Acta Neurologica Scandinavica 1978;58:77‐9.

Additional references

Anonymous 1980

Anonymous. Influence of adherence to treatment and response of cholesterol on mortality in the coronary drug project. The New England Journal of Medicine 1980;303:1038‐41.

Anonymous 1989

Anonymous. Effect of deprenyl on the progression of disability in early Parkinson's disease. The Parkinson Study Group. The New England Journal of Medicine 1989;321:1364‐71.

Clarke 2001

Clarke M, Halsey J. DICE 2: A further investigation of the effect of chance in life, death and subgroup analyses. International Journal of Clinical Practice 2001;55:240‐2.

Clarke 2002

Clarke CE. Medical management of Parkinson's disease. Journal of Neurology, Neurosurgery, and Psychiatry 2002;72(Suppl 1):122‐7.

Dickersin 1994

Dickersin K, Scherer R, Lefebvre C. Systematic Reviews: Identifying relevant studies for systematic reviews. BMJ 1994;309:1286‐91.

EBCTCG 1990

Early Breast Cancer Trialists' Collaborative Group. Treatment of Early Breast Cancer. Vol 1. Worldwide Evidence 1985‐1990. Oxford: Oxford University Press, 1990.

Fleiss 1993

Fleiss JL. The statistical basis of meta‐analysis. Statistical Methods in Medical Research 1993;2:121‐45.

GPDSSC 2002

Global Parkinson's Disease Survey Steering Committee. Factors impacting on quality of life in Parkinson's disease: results from an international survey. Movement Disorders 2002;17:60‐7.

Hills 2002

Hills R, Gray R, Stowe R, Bentham P. Drop‐out bias undermines findings if improved functionality with cholinesterase inhibitors. Neurobiology of Aging 2002;23(Suppl 1):S89 (Abstract 338).

Horwitz 1990

Horwitz RI, Viscoli CM, Berkman L, Donaldson RM, Horwitz SM, Murray CJ, et al. Treatment adherence and risk of death after a myocardial infarction. Lancet 1990;336:542‐5.

Jenkinson 1998

Jenkinson C, Fitzpatrick R, Peto V. The Parkinson's Disease Questionnaire. User manual for the PDQ‐39, PDQ‐8 and PDQ Summary Index. Health Services Research Unit. Oxford: University of Oxford, 1998.

Mantel 1959

Mantel N, Haenszel W. Statistical aspects of the analysis of data from retrospective studies of disease. Journal of the National Cancer Institute 1959;22:719‐48.

Parmar 1998

Parmar M, Torri V, Stewart L. Extracting summary statistics to perform meta‐analyses of the published literature for survival endpoints. Statistics in Medicine 1998;17:2815‐34.

Peto 1977

Peto R, Pike MC, Armitage P, Breslow NE, Cox DR, Howard SV, Mantel N, et al. Design and analysis of randomized clinical trials requiring prolonged observation of each patient. II. analysis and examples. British Journal of Cancer 1977;35:1‐39.

PSG 2000

Parkinson Study Group. Pramipexole vs. levodopa as initial treatment for Parkinson's disease: A randomized controlled trial. The Journal of the American Medical Association 2000;284:1931‐8.

PSG 2004

Parkinson Study Group. Levodopa and the Progress of Parkinson's Disease. The New England Journal of Medicine 2004;351:2498‐508.

Quinn 1997

Quinn NP. Parkinson's disease: clinical features. Baillieres Clinical Neurology 1997;6:1‐13.

Ramaker 2004(A)

Ramaker C, Hilten JJ van. Bromocriptine versus levodopa in early Parkinson's disease. Cochrane Database of Systematic Reviews 2004, Issue 2.

Ramaker 2004(B)

Ramaker C, Hilten JJ van. Bromocriptine/levodopa combined versus levodopa alone for early Parkinson's disease. Cochrane Database of Systematic Reviews 2004, Issue 2.

Rascol 2002

Rascol O, Goetz C, Koller W, Poewe W, Sampaio C. Treatment interventions for Parkinson's disease: an evidence based assessment. Lancet 2002;359:1589‐98.

Schapira 1999

Schapira AH. Science, medicine, and the future: Parkinson's disease. BMJ 1999;318:311‐4.

Wheatley 2002

Wheatley K, Stowe RL, Clarke CE, Hills RK, Williams AC, Gray R. Evaluating drug treatments for Parkinson's disease: how good are the trials?. BMJ 2002;324:1508‐11.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Jump to:

Argentina

Methods

Randomised: Yes (method used not given)

Blinding: Single (assessor)

Treatment period: Mean = 31 months

Participants

Eligibility:
Idiopathic PD patients who had not received any prior anti‐PD medication.

No. randomised
Total: 86
DA/Ctl: 26/31/29

Interventions

Comparison:
DA + LD vs. LD:
Bromocriptine
(<= 12.5 mg/d) + LD (<= 1000 mg/d) vs. LD (<= 1000 mg/d).

DA vs. LD
Bromocriptine (<= 20 mg/d) (± LD) vs. LD (<= 1000 mg/d).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose

Notes

Treatment Schedule:
Bromocriptine start 1.25 mg, increased by doses of 1.25 mg. LD start 125/12.5 mg, increased by 125/12.5 mg up to daily optimal dose.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Barcelona

Methods

Randomised: Yes (method used not given)

Blinding: Single (assessor)

Treatment period: 2 years

Participants

Eligibility:
Idiopathic PD patients who had never received anti‐PD medication.

No. randomised
Total: 20
DA/Ctl: 10/10

Interventions

Comparison:
Pergolide
(<= 3 mg/d) + LD
(<= 600 mg/d) vs.
LD (<= 600 mg/d)
(first 6 months ‐ DA vs. LD).

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals
Levodopa dose

Notes

Treatment Schedule:
Pergolide dose increased every 3 days (start 0.05 mg, then 0.1, 0.2, 0.25 and 0.5 mg tid), reaching 1.5 mg/d by day 15. LD dose increased every 3 days (start 25 mg, then 50, 100 mg tid), reaching 300 mg/d in 9 days. Thereafter, doses titrated individually every month. Patients on pergolide started LD after 6 months.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Canada

Methods

Randomised: Yes (method used not given)

Blinding: Double blind

Treatment period: 23 weeks

Participants

Eligibility:
Idiopathic PD patients, no previous anti‐PD therapy (other than anticholinergics).

No. randomised
Total: 81
DA/Ctl: 42/39

Interventions

Comparison:
Bromocriptine
(<= 30 mg/d) vs. LD (<= 300 mg/d) in identical capsules.

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose

Notes

Treatment Schedule:
Phase (1): 2‐week baseline. Phase (2): 15‐week titration period‐medication increased every 3 weeks until stable improvement or maximum 30 mg/d bromocriptine or 300/75 mg LD achieved.
Phase (3): 6‐week maintenance period of constant dosage.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Europe Bromocriptine

Methods

Randomised: Yes (method used not given)

Blinding: Open

Treatment period: 5 years
(12 month data reported)

Participants

Eligibility:
Idiopathic PD patients aged < 65 years at onset of disease in Hoehn & Yahr stages I‐IV. Taken LD therapy for < 2 years.

No. randomised
Total: 277
DA/Ctl: 137/140

Interventions

Comparison:
Bromocriptine
(<= 30 mg/d) + LD (stable dose) vs. LD (stable dose).

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals
Levodopa dose

Notes

Treatment Schedule:
Bromocriptine 1.25 mg/d week 1, 2.5 mg/d week 2; thereafter, increased weekly by 1.25 mg, up to maximum 30 mg/d. LD dose stable as possible.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Europe CQA 206‐291

Methods

Randomised: Yes (method used not given)

Blinding: Double blind

Treatment period: 8 weeks

Participants

Eligibility:
Idiopathic PD patients treated with LD with basal motor status rating of at least 2 in two of the cardinal extrapyramidal symptoms using 0 to 4 UPDRS. LD dosage stable for > 1 month before start of trial.

No. randomised
Total: 36
DA/Ctl: 24/12

Interventions

Comparison:
CQA 206‐291 (<= 30 mg/d) + LD (stable dose) vs. Placebo + LD (stable dose).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals

Notes

Treatment Schedule:
CQA 206‐291 titrated within 4 weeks from 0.2 mg/d up to maximum 30 mg/d. Maintenance dose given for additional 4 weeks.
No change in LD regimen or dose allowed during study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Europe PELMOPET

Methods

Randomised: Yes (computer generated list)

Blinding: Double blind

Treatment period: 3 years

Participants

Eligibility:
Aged 30 to 75 years, diagnosed < 2 years, modified Hoehn & Yahr stage 1 to 2.5, UPDRS part III score 5 to 25 and positive response to apomorphine challenge. Patients had to be LD or dopamine agonist drug naive.

No. randomised
Total: 294
DA/Ctl: 148/146

Interventions

Comparison:
Pergolide (<= 5 mg/d) vs. LD (<= 1200 mg/d).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose
Mortality

Notes

Treatment Schedule:
Study period I: 2‐week non‐randomised, non‐treatment observational period.
Study period II: 8 week fixed‐dose titration schedule, pergolide 0.05 to 0.75 mg/d and LD 50 to 150 mg/d.
Study period III: 34‐month treatment phase with flexible‐titration schedule. Maximum doses allowed 5 mg/d pergolide and 1200 mg/d LD. After 3‐years, 1‐week washout period, then 2‐weeks off medication.

Additional Treatment:
No other anti‐PD medications allowed. Rescue therapy with LD not permitted during trial.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

France Lisuride

Methods

Randomised: Yes (method used not given)

Blinding: Double blind for 1 year then open label.

Treatment period: 5 years

Participants

Eligibility:
Idiopathic PD with disease duration <= 3 years and Hoehn & Yahr stage <= 3. Patients on LD prior to study could be included if treatment duration < 1 year and daily dose < 300mg.

No. randomised
Total: 82
DA/Ctl: 41/41

Interventions

Comparison:
Lisuride
(<= 1.2 mg/d) + LD (stable dose for first year) vs. Placebo + LD (stable dose for first year)

(Placebo‐controlled for 1‐year then open dose variable study).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose

Notes

Treatment Schedule:
Adapted lisuride/placebo dose (up to maximum 1.2 mg/d) during first year without modifying initial LD dose (300 mg).
During 4 years of open trial, daily dose of LD or lisuride (in priority over LD) could be modified to obtain optimal efficacy/tolerance ratio.

Additional Treatment:
At 12 months, selegiline (10 mg/d) added to both groups. Other anti‐PD drugs prohibited during study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

France Piribedil

Methods

Randomised: Yes (blocks of 4, stratified by country)

Blinding: Double blind

Treatment period: 6 months

Participants

Eligibility:
Idiopathic PD patients aged 35 to 75 years, PD for < 10 years (Hoehn & Yahr stage I to III). Had to be receiving LD for > 6 months but < 8 years. Prior treatment with dopamine agonist, anticholinergics and amantadine discontinued for > 1 month before screening. Patients treated with selegiline could be included as long as on stable dosage for > 1 month before entry.

No. randomised
Total: 115
DA/Ctl: 61/54

Interventions

Comparison:
Piribedil (<= 450 mg/d) + LD (4 month stable dose) vs. Placebo + LD (4 month stable dose).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose

Notes

Treatment Schedule:
Placebo run‐in period of 15 days then piribedil/matching placebo for 6 months. Titration starting dose 50 mg od, increased every 2 weeks by 50 mg up to 150 mg tid. Daily dose could be adjusted by 50 mg as necessary. Week 5 ‐ up to 6 months, patients received dosage level achieved in titration. LD kept stable until 4th month but could be adjusted afterwards.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Germany/Aust PRADO

Methods

Randomised: Yes (method used not given)

Blinding: Open

Treatment period: 4.5 years (Stopped after 4 years, due to increased mortality in LD arm)

Participants

Eligibility:
Idiopathic PD patients in Hoehn & Yahr stages I to IV, never treated with anticholinergics, amantadine or dopamine agonist and previous LD therapy < 6 months duration.

No. randomised
Total: 674 (587 analysed)
DA/Ctl: 285/302

Interventions

Comparison:
Bromocriptine (mean = 14 mg/d) + LD (60% stable dose) vs. LD (stable dose).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose
Mortality

Notes

Treatment Schedule:
Both groups: 3 month LD optimal monotherapy phase. In bromocriptine + LD group between 3 and 6 months gradual substitution of LD by up to 40 +/‐ 10% of bromocriptine. Followed by long‐term treatment phase from months 7 to 54, with dose adjusted as required.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Glasgow

Methods

Randomised: Yes (method used not given)

Blinding: Double blind

Treatment period: 5 years
(1 year interim analysis reported)

Participants

Eligibility:
Idiopathic PD patients established on LD (< 2 years). Anticholinergic drugs or previous treatment with amantadine were allowed, but not previous treatment with bromocriptine or selegiline.

No. randomised
Total: 31
DA/Ctl: 16/15

Interventions

Comparison:
Bromocriptine
(<= 30 mg/d) + LD (40%, <= 750 mg/d) vs. Placebo + LD (<= 750 mg/d).

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals
Levodopa dose

Notes

Treatment Schedule:
Bromocriptine/matching placebo (start 1.25 mg bid, 2.5 bid week 3, 2.5 mg tid week 5, 5 mg tid week 7, 7.5 mg tid week 9 and 10 mg tid week 10). Maximum LD dose 750 mg/d; reduced each week (week 10 LD dosage 40% of pre‐study LD).

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Int Pergolide Mono

Methods

Randomised: Yes (computer generated blocks of 4)

Blinding: Double blind

Treatment period: 14 weeks

Participants

Eligibility:
PD for <= 3 years, modified Hoehn & Yahr stage I to III, UPDRS Motor Score > 14 points. Previous LD allowed if < 12 weeks duration. Any previous selegiline therapy discontinued > 8 weeks before entry; LD, dopamine agonists, amantadine and anticholinergics discontinued > 4 weeks before entry.

No. randomised
Total: 105
DA/Ctl: 53/52

Interventions

Comparison:
Pergolide (<= 3 mg/d) vs. Placebo.

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals

Notes

Treatment Schedule:
3‐week fixed‐dose titration period 0.05 mg/d to 0.75 mg/d. Only patients tolerating fixed‐dose titration were eligible to continue; 9‐week flexible dose period (0.75 to 3 mg/d), then fixed dose for last 2‐weeks of this period. Finally, 1 to 2‐week de‐titration period.

Additional Treatment:
No concomitant treatment with any anti‐PD agent allowed.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Int PKDS009

Methods

Randomised: Yes (computer generated list)

Blinding: Double blind

Treatment period: Until development of motor complications or maximum of 5 years treatment

Participants

Eligibility:
Newly and recently diagnosed PD patients (Hoehn & Yahr stage I to III) requiring treatment. Prior use of LD, selegiline or dopamine agonists ‐ exclusion criteria. Previous treatment with amantadine/anticholinergics allowed but had to be discontinued > 4 weeks prior to study.

No. randomised
Total: 420
DA/Ctl: 211/209

Interventions

Comparison:
Cabergoline (<= 4 mg/d) + Placebo (±LD) vs. LD (<= 600 mg/d) + Placebo (±LD).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose
Mortality

Notes

Treatment Schedule:
Cabergoline/LD titrated over 24 weeks, from 0.25 mg/d cabergoline or 100 mg/d LD to 4 mg/d cabergoline or 600 mg/d LD. Titration continued until achievement of optimal/maximum dose. Thereafter, optimal/maximum dose continued until the onset of motor complications or up to 3 years (minimum) and 5 years (maximum).

Additional Treatment:
If diminishing control open‐label LD could be added to both arms.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

Int PSG CALM‐PD

Methods

Randomised: Yes (blocks, stratified by investigator)

Blinding: Double blind

Treatment period: 48 months

Participants

Eligibility:
Idiopathic PD patients aged >= 30 years, diagnosed for < 7 years with Hoehn & Yahr stage I to III, requiring dopaminergic anti‐PD therapy. Patients who had taken LD or dopaminergic agonist in 2 months prior to enrolment excluded.

No. randomised
Total: 301
DA/Ctl: 151/150

Interventions

Comparison:
Pramipexole
(<= 4.5 mg/d) + Placebo (±LD) vs. LD (<=600 mg/d) + Placebo (±LD).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose
Mortality

Notes

Treatment Schedule:
10‐week dose escalation period to 1.5 mg/d pramipexole (or matching placebo) or 300/75 mg/d LD (or matching placebo). Participants requiring additional therapy could escalate to 3 mg pramipexole or 450/112.5 mg LD, or 4.5 mg pramipexole or 600/150 mg LD.

Additional Treatment:
During maintenance open‐label carbidopa/levodopa prescribed as needed. Sustained release carbidopa/levodopa preparations not permitted.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

Int Ropinirole

Methods

Randomised: Yes (method used not given)

Blinding: Double blind

Treatment period: 12 weeks

Participants

Eligibility:
Idiopathic PD patients aged 30 to 80 years (Hoehn & Yahr stages I to IV). Patients untreated or had received LD or dopamine agonists for < 6 months (discontinued > 2 weeks before screening visit).

No. randomised
Total: 63
DA/Ctl: 41/22

Interventions

Comparison:
Ropinirole (<= 10 mg/d) vs. Placebo.

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals

Notes

Treatment Schedule:
Start 0.5 mg bid, increased weekly by 0.5 or 1 mg bid to maximum dose of 5 mg bid.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

International 056

Methods

Randomised: Yes (stratified by selegiline use)

Blinding: Double blind

Treatment period: 5 years

Participants

Eligibility:
PD patients in Hoehn & Yahr stage I to III who required dopaminergic therapy. Patients could have received prior LD or dopamine agonist therapy for < 6 weeks (but discontinued > 2 weeks before entry).

No. randomised
Total: 268
DA/Ctl: 179/89

Interventions

Comparison:
Ropinirole (<= 24 mg/d) + Placebo (±LD) vs. LD (<= 1200 mg/d) + Placebo (±LD).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose
Mortality

Notes

Treatment Schedule:
Ropinirole start 0.75 mg/d and LD 50 mg od plus placebo bid. Dose titration weekly intervals; maximum doses 24 mg/d ropinirole and 1200 mg/d LD.

Additional Treatment:
No other anti‐PD therapies (except rescue LD) allowed.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Italy A

Methods

Randomised: Yes (within centre randomisation)

Blinding: Double blind

Treatment period: 18 months
(Stopped after 3 month interim analysis)

Participants

Eligibility:
Aged < 80 years, with untreated idiopathic PD of <= 5 years duration, Hoehn & Yahr stages I & II.

No. randomised
Total: 123
DA/Ctl: 62/61

Interventions

Comparison:
Alpha‐DHEC (<= 120 mg/d) vs. Placebo.

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals

Notes

Treatment Schedule:
Increased from 5 mg bid week 1/2, 10 mg bid week 3/4, to 20 mg bid week 5/6; then dose adjusted as necessary, up to a maximum dose of 120 mg/d.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Italy B

Methods

Randomised: Yes (method used not given)

Blinding: Double blind

Treatment period: 6 months

Participants

Eligibility:
Idiopathic PD patients with stable response to LD without on‐off phenomena or dystonia/dyskinesia.

No. randomised
Total: 20
DA/Ctl: 10/10

Interventions

Comparison:
Alpha‐DHEC
(<= 60 mg/d) + LD (stable dose) vs. Placebo + LD (stable dose).

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals

Notes

Treatment Schedule:
2‐week placebo run‐in phase in addition to usual LD treatment. Then 6‐month alpha‐DHEC/matching placebo (start 3 mg/d, increased at every control without exceeding 60 mg/d).

Additional Treatment:
No dopamine agonists, anticholinergics, amantadine, selegiline or other drugs acting on CNS allowed, in addition to LD.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Milan

Methods

Randomised: Yes (blocks of 6, stratified by centre)

Blinding: Open

Treatment period:
Median = 34 months

Participants

Eligibility:
PD diagnosed < 2 years who had not taken any of the study drugs for > 4 months.

No. randomised
Total: 335
DA/Ctl: 169/166

Interventions

Comparison:
Bromocriptine
(<= 60 mg/d) or Lisuride (<= 6 mg/d) (±LD) vs. LD
(<= 750 mg/d).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Withdrawals
Mortality

Notes

Treatment Schedule:
Drug doses increased over 2‐4 weeks until optimum dose acheived. Maximum doses; 60 mg bromocriptine, 6 mg lisuride, 750 mg LD.

Additional Treatment:
If dopamine agonists were/became ineffective, LD added.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

South America

Methods

Randomised: Yes (method used not given)

Blinding: Single (assessor)

Treatment period: Until development of motor complication or 3 years treatment

Participants

Eligibility:
Idiopathic PD patients aged 20 to 80 years in Hoehn & Yahr stages I to IV. No LD or dopaminergic agonists before study and any treatment with minor anti‐PD drugs (i.e. anticholinergic drugs) suspended > 4 weeks before entry.

No. randomised
Total: 87 (78 analysed)
DA/Ctl: 40/38

Interventions

Comparison:
Bromocriptine
(<= 15 mg/d) + LD (<= 1000 mg/d) vs. LD (<= 750 mg/d).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose

Notes

Treatment Schedule:
Bromocriptine start 1.25 mg/d at month 3, then increased every 2 weeks by same amount (maximum dose 15 mg/d). LD dose start 125 mg/d, increased each week by same amount then adjusted to achieve maximum "on" time/as little "off" time as possible.

Additional Treatment:
If diminishing clinical control, LD or bromocriptine added on an open basis.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Spain

Methods

Randomised: Yes (method used not given)

Blinding: Double blind for 1st 8 months then open label.

Treatment period: 44 months

Participants

Eligibility:
Aged 40 to 70 years in Hoehn & Yahr stage <= 3 and responsive to LD therapy introduced 2 to 6 months before study entry. No previous exposure to any dopaminergic agonist allowed.

No. randomised
Total: 57 (50 analysed)
DA/Ctl: 27/23

Interventions

Comparison:
Bromocriptine
(<= 30 mg/d) + LD (75% stable dose) vs. Placebo + LD (75% stable dose).

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Withdrawals
Levodopa dose
Mortality

Notes

Treatment Schedule:
Bromocriptine 1.25 mg for 7 days, then increased by 1.25 mg weekly; after 6 weeks, increased by 2.5 mg/week, up to a maximum 15 mg by day 63. After a month, clinician attempted to reduce the LD dose by 20 to 30% (bromocriptine could be increased to maximum of 30 mg/d, with LD dose unchanged).

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Sydney

Methods

Randomised: Yes (method used not given)

Blinding: Double blind

Treatment period: 5 years (Mortality at 10 years)

Participants

Eligibility:
Idiopathic de novo PD patients (including patients who had received LD for < 3 months but discontinued 1 month prior to study entry).

No. randomised
Total: 133
DA/Ctl: 66/67

Interventions

Comparison:
Bromocriptine
(<= 30 mg/d) (±LD) vs.
LD (<= 600 mg/d) in identical capsules.

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose
Mortality

Notes

Treatment Schedule:
1 month placebo baseline phase. Then 1 mg/d of bromocriptine or 20/5 mg/d LD. Dose gradually increased at weekly, then monthly intervals. Maximum doses 30 mg/d bromocriptine and 600/150 mg/d LD.

Additional Treatment:
Rescue LD could replace or be added to bromocriptine arm.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

UK‐PDRG

Methods

Randomised: Yes (blocks of 4)

Blinding: Open

Treatment period: 10 years (Drop‐outs at 3 years)

Participants

Eligibility:
Untreated PD patients of any age requiring dopaminergic treatment. Patients known to have failed to respond to dopaminergic drugs excluded.

No. randomised
Total: 511
DA/Ctl: 262/249

Interventions

Comparison:
Bromocriptine
(<= 120 mg/d) vs. LD.

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Withdrawals
Mortality

Notes

Treatment Schedule:
Bromocriptine start 2.5 mg, increasing by 2.5 mg every 3 days to dose of 30 mg/d. Maximum dose allowed 40 mg tid. LD start 62.5 mg tid, increased to 125 mg tid and maintained for > 3 months before further increments considered.

Additional Treatment:
Additional anti‐PD drugs allowed.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

UKPDRG Pergolide

Methods

Randomised: Yes (blocks of 4)

Blinding: Double blind

Treatment period: Until LD needed (Mean = 16 months)

Participants

Eligibility:
Aged 30 to 79 years with disease duration < 5 years, not taking nor requiring anti‐PD therapy.

No. randomised
Total: 106
DA/Ctl: 53/53

Interventions

Comparison:
Pergolide (50 µg/d) vs. Placebo.

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals

Notes

Treatment Schedule:
25 µg twice daily from week 0.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

USA

Methods

Randomised: Yes (method used not given)

Blinding: Double blind

Treatment period: 14 months

Participants

Eligibility:
PD patients with Hoehn & Yahr stage I to III, who had >= 2 of resting tremor, bradykinesia or rigidity. Patients on no medication at baseline/received no selegiline during preceding 60 days or other anti‐PD therapy for >= 30 days prior entry.

No. randomised
Total: 101
DA/Ctl: 52/49

Interventions

Comparison:
Bromocriptine
(<= 20 mg/d) (±LD) vs. LD
(+/‐ selegiline in each arm).

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals
Levodopa dose
Mortality

Notes

Treatment Schedule:
Initiated on 10 mg/d of selegiline or matching placebo. On 3rd day started either bromocriptine (5 mg) or LD (25/100). Dose of bromocriptine titrated up or down as appropriate.

Additional Treatment:
Bromocriptine patients could receive supplementary LD once 20 mg/d bromocriptine achieved.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

USA / Canada PSG

Methods

Randomised: Yes (computer generated blocks of 10, stratified by investigator)

Blinding: Double blind

Treatment period: 11 weeks

Participants

Eligibility:
Aged > 30 years with idiopathic PD for < 7 years; Hoehn & Yahr stages I to III. Did not require anti‐PD treatment and had not taken such medication within 3 months prior to enrolment.

No. randomised
Total: 264
DA/Ctl: 213/51

Interventions

Comparison:
Pramipexole (1.5, 3, 4.5 or 6 mg/d) vs. Placebo.

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals
Mortality

Notes

Treatment Schedule:
Titration schedule used (including up to 4 weeks of matching placebo); target/achieved dose (1.5, 3, 4.5 or 6 mg/d) by week 6 for 4 weeks, then 1‐week discontinuation period.

Additional Treatment:
LD or other dopamine agonists not permitted during study. Selegiline, anticholinergics and amantadine permitted if stable dosages for 30 days prior to and during study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

USA Bromocriptine

Methods

Randomised: Yes (method used not given)

Blinding: Double blind

Treatment period: 4 years (Mean = 40 months)

Participants

Eligibility:
PD patients previously untreated with bromocriptine or LD.

No. randomised
Total: 25
DA/Ctl: 7/8/10

Interventions

Comparison:
DA + LD vs. Placebo + LD:
Bromocriptine (<= 18 mg/d) + LD
(<= 1200 mg/d) vs. Placebo + LD (<= 1200 mg/d).

DA + LD:
Bromocriptine (<= 30 mg/d) + Placebo vs. LD (<= 1200 mg/d) + Placebo.

Outcomes

Data Reported:
Clinician rated disability
Motor complications
Side‐effects
Withdrawals
Levodopa dose

Notes

Treatment Schedule:
Bromocriptine start 2.5 mg/d, then after 2 weeks LD (100/25 mg) added. Doses could be gradually increased to maximum doses of 18 mg bromocriptine and 1200/300 mg LD.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

USA Pramipexole A

Methods

Randomised: Yes (method used not given)

Blinding: Double blind

Treatment period: 9 weeks

Participants

Eligibility:
Aged > 20 years with early idiopathic PD (Hoehn & Yahr stages I to III).

No. randomised
Total: 56
DA/Ctl: 28/28

Interventions

Comparison:
Pramipexole (<= 4.5 mg/d) vs. Placebo.

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals

Notes

Treatment Schedule:
Start 0.3 mg/d, increased weekly up to 4.5 mg/d by week 6, then 3‐week fixed maintenance dose phase, then dose reduction phase.

Additional Treatment:
Selegiline (10 mg/d) received throughout. No other anti‐PD medications allowed.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

USA Pramipexole B

Methods

Randomised: Yes (stratified by selegiline and centre)

Blinding: Double blind

Treatment period: 8 months

Participants

Eligibility:
Aged > 25 years with idiopathic PD in Hoehn & Yahr stage I to III, not taking LD. Up to 180 days of prior LD exposure allowed, but LD‐free for 60 days prior to entry.

No. randomised
Total: 335
DA/Ctl: 165/171

Interventions

Comparison:
Pramipexole (<= 4.5 mg/d) vs. Placebo.

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals

Notes

Treatment Schedule:
Dose titration over 7‐week period, 0.375 to 4.5 mg/d. Then 6 month maintenance phase (on maximum dose), then 1‐week dose‐reduction phase.

Additional Treatment:
Selegiline allowed if dose stable for 30 days and did not exceed 10 mg/d.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

USA Ropinirole

Methods

Randomised: Yes (stratified by selegiline use)

Blinding: Double blind

Treatment period: 6 months

Participants

Eligibility:
Early PD (Hoehn & Yahr stages I to III) requiring dopaminergic therapy, but no LD or any dopaminergic agonist for > 6 weeks prior study entry. All anti‐PD therapies, except selegiline, discontinued > 4 weeks prior to entry.

No. randomised
Total: 241
DA/Ctl: 116/125

Interventions

Comparison:
Ropinirole (<= 24 mg/d) (±LD) vs. Placebo (±LD).

Outcomes

Data Reported:
Clinician rated disability
Side‐effects
Withdrawals

Notes

Treatment Schedule:
Start 0.25 mg tid, then titration up to optimal response (minimum 1.5 mg tid, maximum 8 mg tid) for remainder of study.

Additional Treatment:
Patients on selegiline remained on stable dose for 4 weeks prior to entry and during study. Additional therapy (LD) permitted if patient first been titrated to highest tolerated.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Unclear risk

B ‐ Unclear

Note: In all cases the length of follow up equalled the planned treatment period except for Europe Bromocriptine, Germany/Aust PRADO, Glasgow, Italy A and USA Bromocriptine.
d: day
DA/Ctl: Dopamine agonist/Control
LD: Levodopa
od: Once a day
PD: Parkinson's disease
tid: Three times a day
vs: versus

Characteristics of excluded studies [ordered by study ID]

Jump to:

Study

Reason for exclusion

De Lucia

Abstract with no outcome data reported.

Presthus

Cross‐over study with data split by treatment period not reported.

Data and analyses

Open in table viewer
Comparison 1. Symptom control in trials of dopamine agonists versus levodopa

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Freezing Show forest plot

5

712

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.58 [1.14, 2.18]

Analysis 1.1

Comparison 1 Symptom control in trials of dopamine agonists versus levodopa, Outcome 1 Freezing.

Comparison 1 Symptom control in trials of dopamine agonists versus levodopa, Outcome 1 Freezing.

1.1 DA + LD versus LD

3

130

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.26 [0.62, 2.56]

1.2 DA versus LD

3

582

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.67 [1.17, 2.40]

Open in table viewer
Comparison 2. Motor complications in trials of dopamine agonists versus levodopa

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Dyskinesia Show forest plot

16

3404

Peto Odds Ratio (95% CI)

0.51 [0.43, 0.59]

Analysis 2.1

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 1 Dyskinesia.

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 1 Dyskinesia.

1.1 DA + LD versus LD

8

1019

Peto Odds Ratio (95% CI)

0.74 [0.53, 1.04]

1.2 DA versus LD

10

2385

Peto Odds Ratio (95% CI)

0.45 [0.37, 0.54]

2 Dystonia Show forest plot

9

1896

Peto Odds Ratio (95% CI)

0.64 [0.51, 0.81]

Analysis 2.2

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 2 Dystonia.

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 2 Dystonia.

2.1 DA + LD versus LD

4

203

Peto Odds Ratio (95% CI)

0.67 [0.25, 1.81]

2.2 DA versus LD

7

1693

Peto Odds Ratio (95% CI)

0.64 [0.50, 0.81]

3 Motor Fluctuations Show forest plot

11

2580

Peto Odds Ratio (95% CI)

0.75 [0.63, 0.90]

Analysis 2.3

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 3 Motor Fluctuations.

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 3 Motor Fluctuations.

3.1 DA + LD versus LD

5

813

Peto Odds Ratio (95% CI)

0.89 [0.61, 1.28]

3.2 DA versus LD

7

1767

Peto Odds Ratio (95% CI)

0.71 [0.58, 0.87]

Open in table viewer
Comparison 3. Non‐motor side‐effects in trials of dopamine agonists

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oedema Show forest plot

6

1650

Peto Odds Ratio (Peto, Fixed, 95% CI)

3.48 [2.53, 4.79]

Analysis 3.1

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 1 Oedema.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 1 Oedema.

1.1 DA versus Placebo

1

333

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.28 [0.90, 5.75]

1.2 DA (+/‐LD) versus LD

5

1317

Peto Odds Ratio (Peto, Fixed, 95% CI)

3.68 [2.62, 5.18]

2 Somnolence Show forest plot

11

2423

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.18 [1.75, 2.72]

Analysis 3.2

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 2 Somnolence.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 2 Somnolence.

2.1 DA versus Placebo

5

1006

Peto Odds Ratio (Peto, Fixed, 95% CI)

3.68 [2.62, 5.18]

2.2 DA (+/‐LD) versus LD

6

1417

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.49 [1.12, 2.00]

3 Constipation Show forest plot

8

1961

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.80 [1.36, 2.39]

Analysis 3.3

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 3 Constipation.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 3 Constipation.

3.1 DA versus Placebo

4

944

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.19 [1.39, 3.43]

3.2 DA (+/‐ LD) versus LD

4

1017

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.59 [1.11, 2.28]

4 Dizziness Show forest plot

13

2294

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.60 [1.28, 2.01]

Analysis 3.4

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 4 Dizziness.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 4 Dizziness.

4.1 DA versus Placebo

7

957

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.90 [1.32, 2.74]

4.2 DA (+/‐LD) versus LD

6

1337

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.45 [1.09, 1.92]

5 Hallucinations Show forest plot

14

2479

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.22 [1.58, 3.12]

Analysis 3.5

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 5 Hallucinations.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 5 Hallucinations.

5.1 DA versus Placebo

5

956

Peto Odds Ratio (Peto, Fixed, 95% CI)

4.55 [2.38, 8.71]

5.2 DA (+/‐LD) versus LD

9

1523

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.69 [1.13, 2.52]

6 Nausea Show forest plot

15

2631

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.86 [1.56, 2.23]

Analysis 3.6

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 6 Nausea.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 6 Nausea.

6.1 DA versus Placebo

7

1184

Peto Odds Ratio (Peto, Fixed, 95% CI)

3.11 [2.35, 4.11]

6.2 DA (+/‐ LD) versus LD

8

1447

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.32 [1.05, 1.66]

7 Insomnia Show forest plot

10

2315

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.29 [1.02, 1.64]

Analysis 3.7

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 7 Insomnia.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 7 Insomnia.

7.1 DA versus Placebo

5

998

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.78 [1.20, 2.63]

7.2 DA (+/‐LD) versus LD

5

1317

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.07 [0.79, 1.44]

8 Headache Show forest plot

11

2168

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.26 [0.96, 1.67]

Analysis 3.8

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 8 Headache.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 8 Headache.

8.1 DA versus Placebo

5

789

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.11 [0.68, 1.79]

8.2 DA (+/‐LD) versus LD

6

1379

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.35 [0.96, 1.90]

9 Vomiting Show forest plot

7

998

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.30 [0.78, 2.18]

Analysis 3.9

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 9 Vomiting.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 9 Vomiting.

9.1 DA versus Placebo

4

624

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.37 [0.61, 3.05]

9.2 DA (+/‐LD) versus LD

3

374

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.26 [0.64, 2.46]

10 Hypotension Show forest plot

10

1866

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.25 [0.88, 1.78]

Analysis 3.10

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 10 Hypotension.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 10 Hypotension.

10.1 DA versus Placebo

3

451

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.73 [0.90, 3.35]

10.2 DA (+/‐LD) versus LD

7

1415

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.11 [0.73, 1.67]

11 Anxiety Show forest plot

5

1113

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.14 [0.75, 1.73]

Analysis 3.11

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 11 Anxiety.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 11 Anxiety.

11.1 DA versus Placebo

1

105

Peto Odds Ratio (Peto, Fixed, 95% CI)

7.39 [0.46, 119.79]

11.2 DA (+/‐LD) versus LD

4

1008

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.09 [0.72, 1.66]

12 Depression Show forest plot

6

1628

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.91 [0.68, 1.22]

Analysis 3.12

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 12 Depression.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 12 Depression.

12.1 DA versus Placebo

2

347

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.48 [0.20, 1.14]

12.2 DA (+/‐LD) versus LD

4

1281

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.99 [0.72, 1.34]

Open in table viewer
Comparison 4. Levodopa dose in trials of DA + LD versus LD

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Levodopa Dose (mg/day) Show forest plot

7

455

Mean Difference (IV, Fixed, 95% CI)

‐82.21 [‐110.13, ‐54.29]

Analysis 4.1

Comparison 4 Levodopa dose in trials of DA + LD versus LD, Outcome 1 Levodopa Dose (mg/day).

Comparison 4 Levodopa dose in trials of DA + LD versus LD, Outcome 1 Levodopa Dose (mg/day).

Open in table viewer
Comparison 5. Mortality in trials of dopamine agonists (by randomised treatment comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality Show forest plot

11

3228

Peto Odds Ratio (95% CI)

1.05 [0.86, 1.28]

Analysis 5.1

Comparison 5 Mortality in trials of dopamine agonists (by randomised treatment comparison), Outcome 1 Mortality.

Comparison 5 Mortality in trials of dopamine agonists (by randomised treatment comparison), Outcome 1 Mortality.

1.1 DA versus Placebo

1

264

Peto Odds Ratio (95% CI)

0.0 [0.0, 0.0]

1.2 DA + LD versus LD

2

637

Peto Odds Ratio (95% CI)

0.45 [0.21, 0.97]

1.3 DA versus LD

8

2327

Peto Odds Ratio (95% CI)

1.11 [0.91, 1.37]

Open in table viewer
Comparison 6. Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall patient withdrawal Show forest plot

27

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

Analysis 6.1

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 1 Overall patient withdrawal.

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 1 Overall patient withdrawal.

1.1 DA versus Placebo

8

1293

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.43 [1.06, 1.93]

1.2 DA + LD versus LD

11

1348

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.85 [0.66, 1.10]

1.3 DA versus LD

10

2388

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.02 [1.70, 2.40]

2 Patient withdrawal due to adverse events Show forest plot

23

4719

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.49 [2.08, 2.98]

Analysis 6.2

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 2 Patient withdrawal due to adverse events.

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 2 Patient withdrawal due to adverse events.

2.1 DA versus Placebo

8

1293

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.43 [1.60, 3.68]

2.2 DA + LD versus LD

9

1220

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.62 [1.74, 3.93]

2.3 DA versus LD

8

2206

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.47 [1.96, 3.11]

3 Patient withdrawal due to lack of efficacy Show forest plot

18

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

Analysis 6.3

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 3 Patient withdrawal due to lack of efficacy.

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 3 Patient withdrawal due to lack of efficacy.

3.1 DA versus Placebo

6

946

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.28 [0.11, 0.70]

3.2 DA + LD versus LD

9

1021

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.26 [0.14, 0.47]

3.3 DA versus LD

5

1151

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.93 [1.94, 4.42]

original image
Figures and Tables -
Figure 1

original image
Figures and Tables -
Figure 2

original image
Figures and Tables -
Figure 3

original image
Figures and Tables -
Figure 4

original image
Figures and Tables -
Figure 5

original image
Figures and Tables -
Figure 6

original image
Figures and Tables -
Figure 7

original image
Figures and Tables -
Figure 8

original image
Figures and Tables -
Figure 9

original image
Figures and Tables -
Figure 10

original image
Figures and Tables -
Figure 11

original image
Figures and Tables -
Figure 12

original image
Figures and Tables -
Figure 13

original image
Figures and Tables -
Figure 14

original image
Figures and Tables -
Figure 15

original image
Figures and Tables -
Figure 16

original image
Figures and Tables -
Figure 17

original image
Figures and Tables -
Figure 18

original image
Figures and Tables -
Figure 19

original image
Figures and Tables -
Figure 20

original image
Figures and Tables -
Figure 21

original image
Figures and Tables -
Figure 22

Comparison 1 Symptom control in trials of dopamine agonists versus levodopa, Outcome 1 Freezing.
Figures and Tables -
Analysis 1.1

Comparison 1 Symptom control in trials of dopamine agonists versus levodopa, Outcome 1 Freezing.

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 1 Dyskinesia.
Figures and Tables -
Analysis 2.1

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 1 Dyskinesia.

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 2 Dystonia.
Figures and Tables -
Analysis 2.2

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 2 Dystonia.

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 3 Motor Fluctuations.
Figures and Tables -
Analysis 2.3

Comparison 2 Motor complications in trials of dopamine agonists versus levodopa, Outcome 3 Motor Fluctuations.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 1 Oedema.
Figures and Tables -
Analysis 3.1

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 1 Oedema.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 2 Somnolence.
Figures and Tables -
Analysis 3.2

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 2 Somnolence.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 3 Constipation.
Figures and Tables -
Analysis 3.3

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 3 Constipation.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 4 Dizziness.
Figures and Tables -
Analysis 3.4

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 4 Dizziness.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 5 Hallucinations.
Figures and Tables -
Analysis 3.5

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 5 Hallucinations.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 6 Nausea.
Figures and Tables -
Analysis 3.6

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 6 Nausea.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 7 Insomnia.
Figures and Tables -
Analysis 3.7

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 7 Insomnia.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 8 Headache.
Figures and Tables -
Analysis 3.8

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 8 Headache.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 9 Vomiting.
Figures and Tables -
Analysis 3.9

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 9 Vomiting.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 10 Hypotension.
Figures and Tables -
Analysis 3.10

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 10 Hypotension.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 11 Anxiety.
Figures and Tables -
Analysis 3.11

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 11 Anxiety.

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 12 Depression.
Figures and Tables -
Analysis 3.12

Comparison 3 Non‐motor side‐effects in trials of dopamine agonists, Outcome 12 Depression.

Comparison 4 Levodopa dose in trials of DA + LD versus LD, Outcome 1 Levodopa Dose (mg/day).
Figures and Tables -
Analysis 4.1

Comparison 4 Levodopa dose in trials of DA + LD versus LD, Outcome 1 Levodopa Dose (mg/day).

Comparison 5 Mortality in trials of dopamine agonists (by randomised treatment comparison), Outcome 1 Mortality.
Figures and Tables -
Analysis 5.1

Comparison 5 Mortality in trials of dopamine agonists (by randomised treatment comparison), Outcome 1 Mortality.

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 1 Overall patient withdrawal.
Figures and Tables -
Analysis 6.1

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 1 Overall patient withdrawal.

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 2 Patient withdrawal due to adverse events.
Figures and Tables -
Analysis 6.2

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 2 Patient withdrawal due to adverse events.

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 3 Patient withdrawal due to lack of efficacy.
Figures and Tables -
Analysis 6.3

Comparison 6 Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison), Outcome 3 Patient withdrawal due to lack of efficacy.

Table 1. Clinician‐rated disability scale data reported in trials

Favours Ctl

Favours Ctl

Favours DA

Favours DA

Study ID

Assessment Time Point

Method of analysis

P < 0.01

P: 0.05 to 0.01

No significant difference

P: 0.05 to 0.01

P < 0.01

DA versus Placebo

USA Pramipexole A

9 weeks

Observed Case

UPDRS motor (P = 0.1)

UPDRS ADL (P = 0.002)

USA / Canada PSG

10 weeks

Last Observation Carried Forward (LOCF)

UPDRS mental

UPDRS motor & ADL

UPDRS total (P < 0.005)

International Ropinirole

12 weeks

LOCF
(patients with >= 1 post dose assessment)

Finger‐tap test, UPDRS ADL & mental and Hoehn & Yahr

UPDRS motor (P = 0.018)

Clinician's Global Evaluation (P = 0.008)

International Pergolide Monotherapy

12 weeks

Intention to Treat (ITT)
(patients with baseline and >= 1 follow up assessment)

UPDRS total, ADL & motor, Schwab & England ADL, and Clinical Global Impression (CGI) (P < 0.001)

USA Ropinirole

6 months

ITT

UPDRS motor and CGI
(P < 0.001)

USA Pramipexole B

8 months

LOCF
(patients with >= 1 post dose assessment)

UPDRS ADL & motor (P < 0.001)

UKPDRG Pergolide

16 months

LOCF
(patients with >= 1 efficacy measurement)

Hoehn & Yahr, ADL and UPDRS ADL & motor

Italy A

3 months

LOCF
(patients with >= 1 post dose assessment)

UPDRS motor (P = 0.014)

UPDRS total, ADL & mental (P <= 0.006)

DA + LD versus Placebo + LD

European CQA 206‐291

2 months

ITT

Schwab & England ADL

UPDRS total (P < 0.01)

France Piribedil

6 months

LOCF
(patients with >= 1 post dose assessment)

UPDRS ADL and Schwab & England

UPDRS motor (P = 0.04)

Italy B

6 months

ITT

Columbia University Rating Scale (CURS) and North Western University Disability Scale (NWUDS) (P <= 0.002)

Spain

44 months

ITT

UPDRS ADL and Hoehn & Yahr

UPDRS total & motor

USA Bromocriptine

48 months

ITT

CURS and ADL score

France Lisuride

60 months

LOCF

UPDRS mental and Hoehn & Yahr

UPDRS total, ADL & motor and Schwab & England (P < 0.01)

Glasgow

12 months

ITT

NWUDS (P = 0.04)

DA + LD versus LD

Europe Bromocriptine

12 months

ITT

Webster and NWUDS

Barcelona

24 months

ITT

UPDRS mental, ADL & motor

Argentina

31 months

ITT

Hoehn & Yahr and Webster

South America

36 months

ITT

Hoehn & Yahr, Webster, CURS, NWUDS and UPDRS motor

Germany / Austria PRADO

48 months

ITT

Webster and Hoehn & Yahr

DA versus LD

Canada

6 months

ITT

Hoehn & Yahr, CURS and NWUDS

USA

14 months

ITT

UPDRS total, ADL & motor

International PSG CALM‐PD

48 months

Multiple Imputation
(Little & Yau)

UPDRS total & motor (P < 0.003)

UPDRS ADL (P = 0.02)

UPDRS mental (P = 0.1)

Argentina

31 months

ITT

Hoehn & Yahr and Webster

Milan

72 months

ITT

Motor score/signs

Europe PELMOPET

36 months

LOCF
(patients with both a baseline and post baseline score)

UPDRS total, ADL, motor, CGI and Patient Global Impressions (P < 0.001). Schwab & England (P = 0.008)

USA Bromocriptine

48 months

ITT

UPDRS ADL & motor

International 056

60 months

LOCF
(patients with >= 1 post dose assessment)

UPDRS ADL & motor

International PKDS009

60 months

ITT

UPDRS motor
(P < 0.01)

UPDRS ADL

CGI

Sydney

60 months

Per Protocol

CURS and NWUDS

UK‐PDRG

108 months

ITT

At 1 year: Webster (P = 0.006)

At 9 years: Webster

Figures and Tables -
Table 1. Clinician‐rated disability scale data reported in trials
Comparison 1. Symptom control in trials of dopamine agonists versus levodopa

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Freezing Show forest plot

5

712

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.58 [1.14, 2.18]

1.1 DA + LD versus LD

3

130

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.26 [0.62, 2.56]

1.2 DA versus LD

3

582

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.67 [1.17, 2.40]

Figures and Tables -
Comparison 1. Symptom control in trials of dopamine agonists versus levodopa
Comparison 2. Motor complications in trials of dopamine agonists versus levodopa

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Dyskinesia Show forest plot

16

3404

Peto Odds Ratio (95% CI)

0.51 [0.43, 0.59]

1.1 DA + LD versus LD

8

1019

Peto Odds Ratio (95% CI)

0.74 [0.53, 1.04]

1.2 DA versus LD

10

2385

Peto Odds Ratio (95% CI)

0.45 [0.37, 0.54]

2 Dystonia Show forest plot

9

1896

Peto Odds Ratio (95% CI)

0.64 [0.51, 0.81]

2.1 DA + LD versus LD

4

203

Peto Odds Ratio (95% CI)

0.67 [0.25, 1.81]

2.2 DA versus LD

7

1693

Peto Odds Ratio (95% CI)

0.64 [0.50, 0.81]

3 Motor Fluctuations Show forest plot

11

2580

Peto Odds Ratio (95% CI)

0.75 [0.63, 0.90]

3.1 DA + LD versus LD

5

813

Peto Odds Ratio (95% CI)

0.89 [0.61, 1.28]

3.2 DA versus LD

7

1767

Peto Odds Ratio (95% CI)

0.71 [0.58, 0.87]

Figures and Tables -
Comparison 2. Motor complications in trials of dopamine agonists versus levodopa
Comparison 3. Non‐motor side‐effects in trials of dopamine agonists

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Oedema Show forest plot

6

1650

Peto Odds Ratio (Peto, Fixed, 95% CI)

3.48 [2.53, 4.79]

1.1 DA versus Placebo

1

333

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.28 [0.90, 5.75]

1.2 DA (+/‐LD) versus LD

5

1317

Peto Odds Ratio (Peto, Fixed, 95% CI)

3.68 [2.62, 5.18]

2 Somnolence Show forest plot

11

2423

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.18 [1.75, 2.72]

2.1 DA versus Placebo

5

1006

Peto Odds Ratio (Peto, Fixed, 95% CI)

3.68 [2.62, 5.18]

2.2 DA (+/‐LD) versus LD

6

1417

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.49 [1.12, 2.00]

3 Constipation Show forest plot

8

1961

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.80 [1.36, 2.39]

3.1 DA versus Placebo

4

944

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.19 [1.39, 3.43]

3.2 DA (+/‐ LD) versus LD

4

1017

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.59 [1.11, 2.28]

4 Dizziness Show forest plot

13

2294

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.60 [1.28, 2.01]

4.1 DA versus Placebo

7

957

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.90 [1.32, 2.74]

4.2 DA (+/‐LD) versus LD

6

1337

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.45 [1.09, 1.92]

5 Hallucinations Show forest plot

14

2479

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.22 [1.58, 3.12]

5.1 DA versus Placebo

5

956

Peto Odds Ratio (Peto, Fixed, 95% CI)

4.55 [2.38, 8.71]

5.2 DA (+/‐LD) versus LD

9

1523

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.69 [1.13, 2.52]

6 Nausea Show forest plot

15

2631

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.86 [1.56, 2.23]

6.1 DA versus Placebo

7

1184

Peto Odds Ratio (Peto, Fixed, 95% CI)

3.11 [2.35, 4.11]

6.2 DA (+/‐ LD) versus LD

8

1447

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.32 [1.05, 1.66]

7 Insomnia Show forest plot

10

2315

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.29 [1.02, 1.64]

7.1 DA versus Placebo

5

998

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.78 [1.20, 2.63]

7.2 DA (+/‐LD) versus LD

5

1317

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.07 [0.79, 1.44]

8 Headache Show forest plot

11

2168

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.26 [0.96, 1.67]

8.1 DA versus Placebo

5

789

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.11 [0.68, 1.79]

8.2 DA (+/‐LD) versus LD

6

1379

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.35 [0.96, 1.90]

9 Vomiting Show forest plot

7

998

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.30 [0.78, 2.18]

9.1 DA versus Placebo

4

624

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.37 [0.61, 3.05]

9.2 DA (+/‐LD) versus LD

3

374

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.26 [0.64, 2.46]

10 Hypotension Show forest plot

10

1866

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.25 [0.88, 1.78]

10.1 DA versus Placebo

3

451

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.73 [0.90, 3.35]

10.2 DA (+/‐LD) versus LD

7

1415

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.11 [0.73, 1.67]

11 Anxiety Show forest plot

5

1113

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.14 [0.75, 1.73]

11.1 DA versus Placebo

1

105

Peto Odds Ratio (Peto, Fixed, 95% CI)

7.39 [0.46, 119.79]

11.2 DA (+/‐LD) versus LD

4

1008

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.09 [0.72, 1.66]

12 Depression Show forest plot

6

1628

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.91 [0.68, 1.22]

12.1 DA versus Placebo

2

347

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.48 [0.20, 1.14]

12.2 DA (+/‐LD) versus LD

4

1281

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.99 [0.72, 1.34]

Figures and Tables -
Comparison 3. Non‐motor side‐effects in trials of dopamine agonists
Comparison 4. Levodopa dose in trials of DA + LD versus LD

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Levodopa Dose (mg/day) Show forest plot

7

455

Mean Difference (IV, Fixed, 95% CI)

‐82.21 [‐110.13, ‐54.29]

Figures and Tables -
Comparison 4. Levodopa dose in trials of DA + LD versus LD
Comparison 5. Mortality in trials of dopamine agonists (by randomised treatment comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality Show forest plot

11

3228

Peto Odds Ratio (95% CI)

1.05 [0.86, 1.28]

1.1 DA versus Placebo

1

264

Peto Odds Ratio (95% CI)

0.0 [0.0, 0.0]

1.2 DA + LD versus LD

2

637

Peto Odds Ratio (95% CI)

0.45 [0.21, 0.97]

1.3 DA versus LD

8

2327

Peto Odds Ratio (95% CI)

1.11 [0.91, 1.37]

Figures and Tables -
Comparison 5. Mortality in trials of dopamine agonists (by randomised treatment comparison)
Comparison 6. Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Overall patient withdrawal Show forest plot

27

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

1.1 DA versus Placebo

8

1293

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.43 [1.06, 1.93]

1.2 DA + LD versus LD

11

1348

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.85 [0.66, 1.10]

1.3 DA versus LD

10

2388

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.02 [1.70, 2.40]

2 Patient withdrawal due to adverse events Show forest plot

23

4719

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.49 [2.08, 2.98]

2.1 DA versus Placebo

8

1293

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.43 [1.60, 3.68]

2.2 DA + LD versus LD

9

1220

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.62 [1.74, 3.93]

2.3 DA versus LD

8

2206

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.47 [1.96, 3.11]

3 Patient withdrawal due to lack of efficacy Show forest plot

18

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

3.1 DA versus Placebo

6

946

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.28 [0.11, 0.70]

3.2 DA + LD versus LD

9

1021

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.26 [0.14, 0.47]

3.3 DA versus LD

5

1151

Peto Odds Ratio (Peto, Fixed, 95% CI)

2.93 [1.94, 4.42]

Figures and Tables -
Comparison 6. Patient withdrawal in trials of dopamine agonists (by randomised treatment comparison)