You are on page 1of 9

Review

A reassessment of risks and benefits of dopamine agonists


in Parkinson’s disease
Angelo Antonini, Eduardo Tolosa, Yoshikuni Mizuno, Mitsutoshi Yamamoto, Werner H Poewe

Neurologists have several choices of drugs that have been shown to be effective for the treatment of the symptoms of Lancet Neurol 2009; 8: 929–37
Parkinson’s disease. Among the first options are the dopamine agonists, which are commonly used both as an early Published Online
monotherapy and as an adjunct therapy to levodopa. However, before starting any treatment, the overall benefit-to- August 25, 2009
DOI:10.1016/S1474-
risk ratio to individual patients must be considered. For the dopamine agonists, the available evidence on their
4422(09)70225-X
symptomatic efficacy, effect on long-term levodopa-related motor complications, putative effect on progression of
Parkinson Institute, Istituti
disease, and adverse event profile must be taken into account. Recently, the ocurrence of adverse events such as leg Clinici di Perfezionamento,
oedema, daytime somnolence, impulse control disorders, and fibrosis have increasingly been recognised. The risks of Milan, Italy (A Antonini MD);
these potentially serious adverse events must therefore be taken into account and treatment decisions should be Neurology Service, Hospital
Clinic, University of Barcelona
based on considerations of risks versus benefits for individual patients.
and Centro de Investigación
Biomedica en Red sobre
Introduction Benefits of dopamine agonists in the medical Enfermedades
The use of oral dopamine agonists for the treatment of management of Parkinson’s disease Neurodegenerativas
(CIBERNED), Barcelona, Spain
Parkinson’s disease dates back to the 1970s when the Early monotherapy
(E Tolosa MD); Department of
ergolinic drug bromocriptine was first introduced. The On the basis of a consistent body of evidence from Neurology, Juntendo
first generation of dopamine agonists were all ergot randomised controlled trials, the dopamine agonists University School of Medicine,
derivatives and their pharmacological profile differed dihydroergocryptine, pergolide, pramipexole, and Tokyo, Japan (Y Mizuno MD);
Department of Neurology,
from that of levodopa in several ways. For example, ergot ropinirole11–18 have all been shown to be effective as
Kagawa Prefectural Central
derivatives had a longer half-life than levodopa and had a monotherapy in early Parkinson’s disease.8 The evidence Hospital, Kagawa, Japan
differential affinity primarily to D1-like and D2-like for efficacy was less strong for bromocriptine, cabergoline, (M Yamamoto MD); and
dopamine receptors. Although more than 50 years have and lisuride as there have not been randomised trials of Department of Neurology,
Medical University of
passed since the non-ergot agonist apomorphine was first these compounds in early Parkinson’s disease.19–22 In Innsbruck, Austria
reported to exert strong antiparkinsonian effects,1,2 most recent placebo-controlled trials, rotigotine23,24 and (W H Poewe MD)
of the currently used non-ergot dopamine agonists have piribedil25 have been shown to be efficacious in early Correspondence to:
entered the clinic more recently and include pramipexole, disease. Of note, cabergoline,7 pergolide,6 pramipexole,4,5 Werner H Poewe, Department of
ropinirole, rotigotine, and piribedil (table 1). and ropinirole3 have also been tested against levodopa in Neurology, Medical University of
Innsbruck, Anichstrasse 35,
The symptomatic efficacy of dopamine agonists to treat large randomised trials. The results from these trials
A-6020 Innsbruck, Austria
Parkinson’s disease is firmly established and several have all shown a significantly reduced risk of motor werner.poewe@i-med.ac.at
studies have also shown that early use of these drugs as complications compared with levodopa, in particular
initial monotherapy is associated with a reduced long-term dyskinesias, over double-blind follow-up periods of up to
incidence of motor complications (ie, motor fluctuations 5 years. However, the effect on symptoms as assessed
and dyskinesia) compared with levodopa.3–7 Although this with the unified Parkinson’s disease rating scale (UPDRS)
evidence has led to dopamine agonists being classified as was consistently greater for levodopa (table 2).
first-line options for initial monotherapy in early
Parkinson’s disease in many national and international Continuous dopaminergic stimulation and reduced risk
guidelines,8,9 there have also been recent concerns about of motor complications
the safety profile of these drugs in the longer term. These The exact reason why initial monotherapy with a
concerns are related to the risk of developing impulse dopamine agonist is associated with a reduced risk for
control disorders, peripheral oedema, daytime somnolence, motor complications, in particular dyskinesias, compared
and heart valve fibrosis. The recognition of the risk of with levodopa is not fully understood. The most popular
cardiac fibrotic valvulopathies with pergolide and hypothesis is that therapies with long half-lives provide
cabergoline10 has caused regulatory authorities in many more continuous stimulation of brain dopaminergic
countries to restrict the use of these drugs to second-line receptors and that such continuous stimulation is key to
options with specialised cardiac safety monitoring. the reduced risk of motor complications with longer-acting
In this Review, we first outline the benefits of using drugs compared with short-acting drugs, such as
dopamine agonists in the management of Parkinson’s levodopa. Short-acting drugs induce discontinuous or
disease. We then discuss recent evidence on each of the pulsatile stimulation, which is associated with altered
potential risks, outline the consequences for the gene expression and firing patterns in basal ganglia
management of Parkinson’s disease, and provide output neurons, particularly of the direct pathway, that
recommendations for clinical neurologists on how to cause development of levodopa-induced dyskinesias.26–28
individualise treatment decisions based on considerations This concept is supported by data from several preclinical
of their risks versus benefits. studies in primates treated with MPTP (1-methyl-4-

www.thelancet.com/neurology Vol 8 October 2009 929


Review

and ropinirole3 both show that dyskinesia rates remain


D2/D3 receptor D1 receptor NE receptor 5-HT2B receptor Half-life
affinity affinity affinity affinity (h) lower for these two drugs than for levodopa monotherapy
even after most patients had received levodopa as an
Ergot agonists
adjunct therapy to maintain symptomatic control.
Bromocriptine D2 – + +/– 3–6
Recently, 6-year follow-up data for 222 of 301 patients
Cabergoline D3>D2 – + + 65
originally randomised in the CALM-PD (Long Term
Dihydroergocriptine D2 +/– + + 12–16
Impact of Initiating Pramipexole Versus Levodopa in
Lisuride D2 – + +* 2–3
Early Parkinson’s Disease) trial continued to show
Pergolide D3>D2 + + + 15–20
reduced overall dyskinesia rates for those patients initially
Non-ergot agonists
randomised to pramipexole.39 A similar trend was
Apomorphine D3>D2 + – – 0·5
observed at a 10-year follow-up time point for the
Piribedil D3>D2 – +/– – 20
ropinirole versus levodopa cohort, but this observation
Pramipexole D3>D2 – +/– – 10
was based on less than 20% of the original study cohort
Ropinirole D3>D2 – – – 6
remaining available for assessment.40 However, when
Rotigotine D3>D2 + – – 5–7†
analysing disabling dyskinesias separately, their rate was
–=no affinity. +=high affinity. +/–=moderate affinity. NE=norepinephrine. *Antagonist. †After transdermal application. similar at 4 years in the pramipexole versus levodopa trial
regardless of which drug was chosen for initial therapy.39
Table 1: Pharmacological properties of the dopamine agonists
Moreover, the 14-year results of the UK bromocriptine
trial suggest that the initially lower incidence of motor
Number Duration ΔUPDRS part III score Dyskinesia Wearing-off complications in patients who started on the agonist was
on (years) (% of patients) (% of patients) not maintained in the long term compared with those
levodopa
(agonist)
who started on levodopa, whereas initial levodopa therapy
resulted in better symptomatic control throughout the
Levodopa Agonist Levodopa Agonist Levodopa Agonist
observational period.41 Although the conclusions to be
Levodopa vs 89 (179) 5 –4·8±8·3 –0·8±10·1 45 20 34 23 drawn from these data are limited by the small number
ropinrole3
of patients who were evaluable after 14 years, they are
Levodopa vs 151 (150) 2 –7·3±8·6 –3·4±8·6 30·7 9·9 38·0 23·8
consistent with the understanding that there is an
pramipexole4
eventual need for levodopa in almost all patients with
Levodopa vs 146 (148) 3 –2·8±7·8 2·8±9·8 26·0 8·2 43·8 30·6*
pergolide6 Parkinson’s disease. The results from these studies
suggest that differences in initial risk of dyskinesia
ΔUPDRS=change in unified Parkinson’s disease rating scale. *Frequency of motor complications (fluctuations plus progressively diminish when patients are followed-up in
dyskinesia).
the long term.42
Table 2: Results of the main trials of levodopa versus dopamine agonists in early Parkinson’s disease
Neuroprotection
phenyl-1,2,3,6-tetrahydropyridine), which all showed that Halting or slowing disease progression remains a key
treatment with a long-acting dopamine agonist was unmet need in the management of Parkinson’s disease,
associated with reduced dyskinesias compared with and dopamine agonists have been extensively studied
levodopa.29–32 Additionally, results from clinical studies for their putative neuroprotective effects. Several
with continuous subcutaneous infusion of apomorphine33 mechanisms have been proposed and include indirect
or lisuride34 or with continuous enteral infusions of antioxidant effects via activation of presynaptic
levodopa35,36 have also shown fewer pre-existing levodopa- dopamine autoreceptors, leading to reduced dopamine
induced dyskinesias. There are, however, no studies that turnover, as well as antiapoptotic effects via activation of
indicate that continuous delivery of levodopa from the intracellular kinase systems (see elsewhere for review43).
start of treatment could prevent the development of On the basis of preclinical studies showing potential
dyskinesias. Studies that have used either slow-release neuroprotective activity of dopamine agonists, several
levodopa or combinations of levodopa with the catechol-O- clinical trials were undertaken that used surrogate
methyltransferase inhibitor entacapone have not shown biomarkers of nigrostriatal function as primary end-
decreased incidences of motor complications over points to try to avoid confounding symptomatic effects
periods of up to 5 years.37 On the contrary, results from that would hinder the interpretation of their putative
the recent STRIDE-PD (STalevo Reduction In Dyskinesia disease-modifying effects.
Evaluation-Parkinson’s Disease) trial, which compared The REAL-PET study (ReQuip as Early Therapy versus
initial levodopa plus carbidopa therapy with and without L-dopa using Positron Emission Tomography) compared
entacapone, have shown that early entacapone use is ropinirole with levodopa using striatal uptake of
associated with shorter latency to the development of ¹⁸F-fluorodopa on PET imaging as the primary
dyskinesias and, overall, increased dyskinesia rates.38 endpoint.44 For patients who had imaging in the
On the other hand, the 4-year and 5-year levo- CALM-PD trial (CALM-PD-CIT), pramipexole was
dopa-controlled monotherapy trials with pramipexole5 compared with levodopa using β-CIT uptake on SPECT

930 www.thelancet.com/neurology Vol 8 October 2009


Review

imaging (single photon emission computed associated with their “on-demand” delivery (ie,
tomography)45 as the primary outcome measure. Both apomorphine rescue injections) or continuous delivery
studies showed that treatment with the dopamine (ie, rotigotine patch, apomorphine infusions). When
agonist was associated with a significant decrease in the patients with advanced Parkinson’s disease who are
rate of decline of the imaging biomarker compared with treated with levodopa receive an agonist to reduce “off”
that seen for levodopa. However, as neither study episodes, dyskinesia can emerge or, if already present,
included a placebo group, they cannot conclusively can worsen. This risk can usually be controlled by
indicate whether the results reflect “protection” reducing the pre-existing total daily dose of levodopa.58,59
provided by the agonist or “toxicity” caused by levodopa.
The study results could also have been due to differences Risks associated with dopamine agonist use in
in the pharmacological effect of these drugs on the Parkinson’s disease
biomarker rather than on cell survival or function.46 The acute side-effects of dopamine agonists are similar
However, data from a recent study (INSPECT; to those observed with levodopa and include nausea,
Investigating Effects of Short-Term Treatment With vomiting, and postural hypotension. These adverse
Pramipexole or Levodopa on ¹²³I-β-CIT and SPECT events tend to occur with the initiation of treatment and
Imaging in Early Parkinson’s) indicated that short-term tend to abate as tolerance to the drug develops.60
pramipexole therapy did not modify dopamine Discontinuation rates associated with adverse events did
transporter binding measured with β-CIT SPECT.47 not differ between different dopamine agonists and
Nevertheless, this neuroimaging biomarker might be levodopa in randomised double-blind trials.19,61 However,
insufficiently sensitive to detect the effect of an compared with levodopa, dopamine agonists are
intervention on disease progression in the long associated with a higher rate of some dopaminergic
term.42,46,48,49 side-effects, including hallucinations, somnolence, and
More recently, delayed-start designs for trials assessing sudden-onset sleep, as well as impulse control disorders
disease-modifying versus symptomatic effects have been (figure).62 Additionally, certain adverse reactions,
used to investigate disease modification in Parkinson’s including peripheral oedema and fibrotic reactions,
disease.50,51 In a trial with a delayed-start design, patients seem to be specifically associated with the use of
are randomised to receive the study drug or placebo for a dopamine agonists and are not seen with other
fixed time interval (eg, 9 months), after which patients in dopaminergic therapies. Peripheral oedema can be
the placebo group are switched to the study drug and induced by all types of dopamine agonists, and possibly
followed for another fixed time interval. Whereas levodopa, whereas fibrosis seems mainly to be caused by
differences between the two groups at the end of the ergot derivatives.
placebo-controlled phase can be explained by both
symptomatic and/or disease-modifying effects, a Peripheral oedema
difference that is sustained until the end of the second Oedema is a poorly understood complication of agonist
phase, when patients in both groups are receiving the therapy in Parkinson’s disease. This adverse event has
same drug, is assumed to indicate a true disease-modifying probably been under-reported for a long time and can
effect. A recent trial of this design showed benefits of occur with both ergot and non-ergot therapy. A post-hoc
early versus delayed treatment with the monoamine analysis of the CALM-PD trial recently found the 4-year
oxidase B inhibitor rasagiline in a large cohort of patients incidence of leg oedema to be 45% in patients who were
with early Parkinson’s disease;51,52 furthermore, a trial initially randomised to pramipexole.63 Although this
combining a delayed-start design with dopamine percentage is higher than previous estimates,3,4 the
transporter binding visualised on SPECT imaging as a authors note that oedema seemed to emerge after at
surrogate marker for disease progression is underway to least 2 years of therapy. Thus, earlier studies, which
assess the disease-modifying effects of pramipexole (the tended to be of shorter duration, are likely to have
PROUD study; Assessment of Potential ImPact of missed this complication. Leg oedema also occurs as a
PRamipexole On Underlying Disease).53 complication of ergot-derived dopamine agonists,64 or
even levodopa, which suggests a class effect. This
Adjunct therapy in advanced Parkinson’s disease hypothesis would seem plausible as dopamine is an
Several dopamine agonists extend “on” time and reduce important regulator of the sympathetic nervous system,
“off” time disability when used as an adjunct therapy in aldosterone secretion, and ATP-mediated sodium and
patients treated with levodopa who have motor potassium channels.
fluctuations. Such efficacy probably reflects the longer
half-life of drugs such as pergolide, pramipexole, and Daytime sleepiness and sleep attacks
ropinirole, for which class I evidence from randomised Depending on the trial design and criteria used, excessive
placebo-controlled trials in Parkinson’s disease is daytime sleepiness was found in 16–51% of patients with
available. The efficacy of apomorphine injections or Parkinson’s disease (in studies that included more than
infusions54,55 or transdermal rotigotine56,57 is mainly 90 individuals; table 3). Sudden-onset sleep as a side-effect

www.thelancet.com/neurology Vol 8 October 2009 931


Review

these disorders at some point during the course of the


Dyskinesia disease, including 11 of 272 (4·0%) patients with an active
Motor fluctuations impulse control disorder.82 Compulsive gambling and
Dopamine dysregulation syndrome
compulsive sexual behaviour were equally common.
There were no differences between the different
Oedema
dopamine agonists in their association with impulse
Somnolence
control disorders (p=0·21). In another study of
Impulse control disorders 297 patients, the lifetime prevalence of pathological
Hallucinations hypersexuality was 2·4% (7 patients) and compulsive
Nausea shopping was 0·7% (2 patients).83 When including data
Fibrosis*
on pathological gambling,85 the lifetime prevalence of
impulse dyscontrol was 6·1% in all patients with
Higher risk with Higher risk with Parkinson’s disease, which increased to 13·7% in patients
levodopa dopamine agonists
on dopamine agonists. A recent study of more than
Figure: Risk of motor complications and other adverse events with 3000 patients published in preliminary form has reported
dopamine agonists versus levodopa even higher figures: 17·1% of patients receiving dopamine
The length of the arrows indicates the relative extent of risk. *Ergot agonists vs agonists had at least one type of impulse control disorder
levodopa (see text).
and about a third of these patients had more than one of
these disorders.86 Recently, Cilia and co-workers87 used
of non-ergolinic dopamine agonists such as pramipexole functional imaging to show abnormal resting state
and ropinirole was first reported by Frucht and dysfunction of the mesocorticolimbic network in patients
co-workers79 and might occur in 4–6% of patients with with Parkinson’s disease with pathological gambling,
Parkinson’s disease who have had dopaminergic suggesting a drug-induced overstimulation of reward-
therapy.68,72 Both excessive daytime sleepiness and related neuronal systems.
sudden-onset sleep can be induced by all types of
currently used dopamine agonists and also, more rarely, Fibrotic reactions
by levodopa.80 In a questionnaire-based survey of In contrast to all of the above-mentioned adverse events
2952 patients with Parkinson’s disease, 22 of 769 (2·9%) associated with dopamine agonists, the development of
patients on levodopa monotherapy had had sudden-onset pleural, pericardial, peritoneal, or valvular fibrosis seems
sleep; occurrence was higher in patients on dopamine to be specifically associated with ergot compounds.
agonist monotherapy (7 of 131; 5·3%) and highest (137 of Retroperitoneal and pleuropulmonary fibrosis are often
1869; 7·3%) in patients on combined treatment with irreversible after drug withdrawal and can be fatal.88 Case
levodopa plus a dopamine agonist.72 reports of pleuropulmonary fibrosis with bromocriptine
were published soon after its introduction,89–95 and the
Impulse control disorders same complication was subsequently reported for
Dopaminergic therapy for Parkinson’s disease can also pergolide96 and cabergoline, for which there have also
be associated with behavioural abnormalities, which been rare reports of constrictive pericardial fibrosis.97
include dopamine dysregulation syndrome, abnormal Pleuropulmonary fibrosis can develop as late as 11 years
repetitive non-goal oriented behaviours (punding), and after initiation of pergolide therapy,98 highlighting the
reward or incentive-based compulsive actions. need for maintained vigilance during chronic therapy for
Collectively, these abnormalities are common and can Parkinson’s disease with ergolinic agonists.
have major and sometimes devastating psychosocial A possible link between pergolide and valvular heart
consequences. Whereas dopamine dysregulation disease was first reported by Pritchett and co-workers99
syndrome and punding have been primarily associated and was subsequently strongly suggested by echocardio-
with high-dose levodopa or continuous subcutaneous graphic studies.100–102 Further reports also implicated
apomorphine treatment, impulse control disorders seem cabergoline, and more recently bromocriptine, although
to be mainly associated with dopamine agonists. Impulse there is no firm evidence for cardiac valvular changes
control disorders are characterised by a failure to resist induced by lisuride at present.103 A meta-analysis104 of
an impulse, drive, or temptation to carry out an act that is heart valve abnormalities in patients with Parkinson’s
harmful to oneself or others.81 In Parkinson’s disease, disease treated with dopamine agonists indicated a high
typical impulse control disorders include hypersexuality, rate of moderate valvular changes in patients treated with
gambling, compulsive shopping, and compulsive eating. pergolide or cabergoline. This report included data from
In three systematic studies of the prevalence of impulse seven cross-sectional studies, including 477 patients
control disorders in Parkinson’s disease,82–84 6–7% of treated with these ergolinic dopamine agonists,
patients across all three studies met criteria for one of 127 patients with the non-ergot dopamine agonists
these disorders. In the first study, 18 of 272 (6·6%) ropinirole and pramipexole, and 364 control patients.
patients with Parkinson’s disease met criteria for one of Moderate to severe valvular changes were detected in

932 www.thelancet.com/neurology Vol 8 October 2009


Review

26% of patients treated with pergolide and cabergoline, an important role as initial monotherapy, particularly in
10% of patients treated with non-ergot agonists, and 10% younger patients. Over the past decade, the driving force
of control patients. Severe valvulopathy was observed in for the increased prominence of dopamine agonist
less than 1% of patients in both cross-sectional and monotherapy has been its documented benefits in
observational studies. Overall, the severity of reducing the risk of dyskinesias compared with
ergot-induced vavulopathies tends to correlate with levodopa.3,5 As motor complications are often difficult to
cumulative doses.101,105 On the basis of such findings, the manage, contribute substantially to overall disability
European Medicines Agency has added new warnings and cost of illness,107 and contribute to poor quality of
and contraindications to the product information of life in Parkinson’s disease,108 this benefit is considered
cabergoline and pergolide to reduce the risk of fibrosis. by many to outweigh both the inferior symptomatic
In addition to treatment of Parkinson’s disease, efficacy and the specific risks associated with dopamine
dopamine agonists are also first-line therapies for restless agonist therapy compared with levodopa. However,
legs syndrome. The smaller doses of cabergoline or excessive daytime sleepiness, sudden-onset sleep, and
pergolide used in patients with this syndrome, compared impulse control disorders can have serious consequences
with dose levels used in patients with Parkinson’s disease, both for the patient and their social relations. The need
might explain the absence of reports of cardiac for a balanced and individualised process by which to
valvulopathy in such patients. Neither cabergoline nor define the best initial monotherapy strategy for any
pergolide are approved for use in restless legs syndrome, given patient is also indicated by the results of the
and the non-ergot pramipexole and ropinirole are the pramipexole CALM-PD extension study.39 After 6 years,
only dopamine agonists currently licensed for this when about 90% of patients in both groups of the
indication, making it unlikely that cardiac valvulopathy original cohort were on levodopa, the outcome for
will become a serious drug-induced complication in overall impairment in activities of daily living was
restless legs syndrome. similar in the initial pramipexole and levodopa groups.
The mechanisms underlying cardiac valvulopathy The same was true for mean scores on quality of life
induced by cabergoline or pergolide are likely to involve scales and also for motor function as assessed by the
agonism of these two drugs at the 5-HT2B receptor UPDRS. However, motor complications, including
subtype, which is expressed on heart valve leaflets.106 wearing off, “on-off” effects, and dyskinesias, were more
This action might induce fibroblast mitogenesis, thereby common when patients were treated initially with
inducing thickening, retraction, and stiffening of valves levodopa than with pramipexole, although disabling
and causing incomplete leaflet coaptation and valvular dyskinesias were uncommon in both groups. Epworth
regurgitation. The extent to which fibrotic valvular sleepiness scale scores were higher with initial
changes are reversible after discontinuation, and pramipexole treatment, indicating more sleepiness in
whether switching to a non-ergot agonist prevents the inital pramipexole group than in the initial levodopa
further progression, is being studied. Preliminary group.
evidence from a prospective 2-year follow-up study of
21 patients who had developed moderate to severe Number of patients EDS screening EDS in patients EDS in controls
(controls) criteria or tool with PD (%) (%)
valvulopathy during ergot therapy and were switched to
non-ergot agonists showed regression of valve regurg- van Hilten et al65 90 (71) Questionnaire 44·4 31·0
itation in 13 patients. In 46 patients in the same study Tandberg et al66 245 (100) Questionnaire 15·5 1·0
who had continued therapy on cabergoline and pergolide, Ondo et al67 303 (··) ESS >10 50·2 ··
there was an 8% per year incidence of new cases of Hobson et al68 638 (··) ESS ≥7 51·0 ··
moderate to severe regurgitation in any heart valve Tan et al69 201 (214) ESS ≥10 19·9 9·8
(Antonini A, unpublished). Although the observation of Brodsky et al70 101 (100) ESS ≥10 40·6 19·0
regression of valve regurgitation in more than half of the Högl et al71 99 (44) ESS ≥10 33·0 11·0
patients who replaced ergot with non-ergot agonists is Paus et al72 177* (··) ESS ≥10 75·0 ··
encouraging, persistence of abnormalities in a large Marinus et al73 143 (104) ESS >10 27·0 3·0
proportion of the patients is worrisome. This persistence Kumar et al74 149 (115) ESS ≥8 21·0 3·0
could be due to flow turbulence induced by the valve Monaca et al75 222 (··) ESS >10 43·2 ··
dysfunction, which might contribute to worsening Ferreira et al76 176 (174) ESS ≥15 33·5 16·1
fibrosis even when the drug causing the complications Verbaan et al77 419 (150) SCOPA-DS ≥5 43·0 10·0
is withdrawn. Ghorayeb et al78 1625 (··) ESS ≥10 29·0 ··

Studies with more than 90 individuals were selected. ··=data not available. EDS=excessive daytime sleepiness.
Risks versus benefits of dopamine agonists in ESS=Epworth sleepiness score. PD=Parkinson’s disease. SCOPA-DS=scales for outcomes in PD-daytime sleepiness.
clinical practice *2952 patients were screened by telephone interview and 177 were identified to have EDS; after assessment, there
As a class, dopamine agonists have been successfully were 133 (75%) with an ESS of ≥10.
used for many years as an adjunct therapy to levodopa
Table 3: Frequency of EDS in individuals with Parkinson’s disease and controls
in patients who develop motor complications, and have

www.thelancet.com/neurology Vol 8 October 2009 933


Review

disability, whereas dopamine agonists and levodopa are


Search strategy and selection criteria needed for patients with greater impairment or whose
References for this Review were identified through searches professional work and performance on activities of daily
of PubMed (from 1966 to June, 2009) and the Cochrane living will be affected. Levodopa remains the gold
Library (1948 to June, 2009) with the search terms standard for symptomatic efficacy and should not be
“apomorphine”, “bromocriptine”, “cabergoline”, withheld from patients in whom sufficient symptomatic
“dihydroergocryptine”, “dopamine agonist”, “dyskinesia”, control cannot be otherwise obtained. Whether or not a
”fibrosis”, ”impulse control disorders”, ”motor complications”, dopamine agonist should be used as early monotherapy
”(o)edema”, ”pergolide”, ”piribedil”, ”pramipexole”, will largely depend on the perceived risk of dyskinesias—
”ropinirole”, ”rotigotine”, ”sleep”, ”somnolence”, and ”cardiac for which younger age is a major determinant. The use
valvulopathy”. Only peer-reviewed papers published in of ergolinic dopaminergic agonists such as pergolide or
English were considered. Additionally, reference lists in cabergoline in early Parkinson’s disease monotherapy is
published reviews were checked for relevant references, no longer recommended because of their poor overall
regulatory publications issued by the European Committee of benefit to risk ratio when compared with initial levodopa
Proprietary Medicinal Products and the US Food and Drugs monotherapy, based on their additional risk of potentially
Administration were reviewed, and recent releases of life-threatening pleuropulmonary or cardiac valvular
information (ie, congress abstracts and press releases) from fibrosis. However, dopamine agonists are also first-line
major Parkinson’s disease clinical trials were considered. drugs as an adjunct therapy in patients treated with
levodopa who have motor fluctuations, and ergolinic
agonists might still be needed for this indication in
As there have been no head-to-head studies of dopamine countries where newer drugs are not readily accessible.
agonists, there is little information available to help A clinician’s judgment on the different risk profiles of
choose which agonist to use, and physicians will need to monoamine oxidase B inhibitors, dopamine agonists,
take into account factors such as the patient’s lifestyle. and levodopa, as well as expectations about effect size,
However, the ergot-derived dopamine agonists are will substantially affect the final decision on which drug
associated with an additional risk of potentially to use. This decision should be based on careful and
life-threatening side-effects.103 Because of these effects, judicious counselling of patients and caregivers about
pergolide and cabergoline should be second-line the benefit to risk ratios of the different drug options and
therapies. Moreover, the need for these ergot-derived should remain an individualised decision for every new
dopamine agonists has largely been superseded by the patient.
newer agonists pramipexole and ropinirole, which seem Contributors
to have equal efficacy and do not seem to have fibrotic WHP and AA developed the concept of this Review, provided the first
side-effects.103 As one of the main reasons for prescribing draft, contributed to multiple revisions, and were responsible for the
final version of the manuscript. ET and YM reviewed the initial draft,
cabergoline was its long half-life and its associated and discussed and reviewed subsequent drafts. MY participated in the
benefits with compliance, recent developments in planning of the Review, and discussed, reviewed, and revised the
once-daily formulations for both ropinirole and manuscript.
pramipexole mean that this drug might no longer be Conflicts of interest
required. AA has received funding as an adviser and speaker in sponsored
symposia from Boehringer Ingelheim, GE Pharmaceuticals,
GlaxoSmithKline, Novartis, Solvay, UCB, and Valeant. ET has
Conclusions participated on advisory boards for UCB and Boehringer Ingelheim, and
The choice of drug when initiating therapy in early has received honoraria for lecturing in symposia from Boehringer
Parkinson’s disease depends on several factors, Ingelheim, GlaxoSmithKline, Lundbeck, Teva, and UCB. WHP has
including level of disability, age, comorbidities, and received lecture fees and consulting honoraria from Boehringer
Ingelheim, GlaxoSmithKline, Lundbeck, Merck Serono, Novartis, Solvay,
cognitive status, as well as the different risk profiles of Teva, and UCB. YM and MY have no conflicts of interest.
the available drugs. Most commonly, initial drug therapy
Acknowledgments
is with dopaminergic drugs, although amantadine or Editorial assistance was provided by Anita Chadha-Patel, who was
anticholinergic drugs might still be considered to be supported by an unrestricted grant from the Neureca Foundation for
appropriate in a few cases. None of the available drugs movement disorders, Milan, Italy.
has yet been proven to delay the progression of disability References
unequivocally and in a clinically meaningful way, 1 Struppler A, von Uexkull T. Studies of mechanism of action
of apomorphine on Parkinson’s tremor. Z Klein Med 1953;
although a recent study suggested evidence for 152: 46–57.
disease-modifying potential of rasagiline.109 Therefore, 2 Schwab RS, Amador LV, Lettvin JY. Apomorphine in Parkinson’s
selection among the available dopaminergic drug classes disease. Trans Am Neurol Assoc 1951; 56: 251–53.
3 Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE,
will largely depend on symptomatic efficacy and the Lang AE; for the 056 Study Group. A five-year study of the
degree of motor impairment. Monoamine oxidase B incidence of dyskinesia in patients with early Parkinson’s disease
inhibitors have a lesser effect on the main motor features who were treated with ropinirole or levodopa. N Engl J Med 2000;
342: 1484–91.
and are therefore an option for patients with mild

934 www.thelancet.com/neurology Vol 8 October 2009


Review

4 Parkinson Study Group. Pramipexole vs levodopa as initial 26 Olanow CW, Obeso JA, Stocchi F. Continuous dopamine-receptor
treatment for Parkinson disease: a randomized controlled trial. treatment of Parkinson’s disease: scientific rationale and clinical
JAMA 2000; 284: 1931–38. implications. Lancet Neurol 2006; 5: 677–87.
5 Holloway RG, Shoulson I, Fahn S, et al. Pramipexole vs levodopa as 27 Olanow CW, Obeso JA, Stocchi F. Drug insight: continuous
initial treatment for Parkinson disease: a 4-year randomized dopaminergic stimulation in the treatment of Parkinson’s disease.
controlled trial. Arch Neurol 2004; 61: 1044–53. Nat Clin Pract Neurol 2006; 2: 382–92.
6 Oertel WH, Wolters E, Sampaio C, et al. Pergolide versus levodopa 28 Cenci MA. Dopamine dysregulation of movement control in
monotherapy in early Parkinson’s disease patients: the PELMOPET L-DOPA-induced dyskinesia. Trends Neurosci 2007; 30: 236–43.
study. Mov Disord 2005; 21: 343–53. 29 Bedard PJ, Di Paolo T, Falardeau P, Boucher R. Chronic treatment
7 Bracco F, Battaglia A, Chouza C, et al. The long-acting dopamine with L-DOPA, but not bromocriptine induces dyskinesia in
receptor agonist cabergoline in early Parkinson’s disease: final MPTP-parkinsonian monkeys. Correlation with [3H]spiperone
results of a 5-year, double-blind, levodopa-controlled study. binding. Brain Res 1986; 379: 294–99.
CNS Drugs 2004; 18: 733–46. 30 Maratos EC, Jackson MJ, Pearce RK, Jenner P. Antiparkinsonian
8 Horstink M, Tolosa E, Bonuccelli U, et al. Review of the activity and dyskinesia risk of ropinirole and L-DOPA combination
therapeutic management of Parkinson’s disease. Report of a joint therapy in drug naive MPTP-lesioned common marmosets
task force of the European Federation of Neurological Societies (Callithrix jacchus). Mov Disord 2001; 16: 631–41.
and the Movement Disorder Society-European Section. Part I: 31 Smith LA, Tel BC, Jackson MJ, et al. Repeated administration of
early (uncomplicated) Parkinson’s disease. Eur J Neurol 2006; piribedil induces less dyskinesia than L-dopa in MPTP-treated
13: 1170–85. common marmosets: a behavioural and biochemical investigation.
9 Miyasaki JM, Martin W, Suchowersky O, Weiner WJ, Lang AE. Mov Disord 2002; 17: 887–901.
Practice parameter: initiation of treatment for Parkinson’s disease: 32 Maratos EC, Jackson MJ, Pearce RK, Cannizzaro C, Jenner P. Both
an evidence-based review. Report of the Quality Standards short- and long-acting D-1/D-2 dopamine agonists induce less
Subcommittee of the American Academy of Neurology. Neurology dyskinesia than L-DOPA in the MPTP-lesioned common marmoset
2002; 58: 11–17. (Callithrix jacchus). Exp Neurol 2003; 179: 90–102.
10 Horvath J, Fross RD, Kleiner-Fisman G, et al. Severe multivalvular 33 Katzenschlager R, Hughes A, Evans A, et al. Continuous
heart disease: a new complication of the ergot derivative dopamine subcutaneous apomorphine therapy improves dyskinesias in
agonists. Mov Disord 2004; 19: 656–62. Parkinson‘s disease: a prospective study using single-dose
11 Bergamasco B, Frattola L, Muratorio A, Piccoli F, Mailland F, challenges. Mov Disord 2005; 20: 151–57.
Parnetti L. Alpha-dihydroergocryptine in the treatment of de novo 34 Stocchi F, Ruggieri S, Vacca L, Olanow CW. Prospective
parkinsonian patients: results of a multicentre, randomized, randomized trial of lisuride infusion versus oral levodopa in
double-blind, placebo-controlled study. Acta Neurol Scand 2000; patients with Parkinson’s disease. Brain 2002; 125: 2058–66.
101: 372–80. 35 Stocchi F, Vacca L, Ruggieri S, Olanow CW. Intermittent vs
12 Barone P, Bravi D, Bermejo-Pareja F, et al; for the Pergolide continuous levodopa administration in patients with advanced
Monotherapy Study Group. Pergolide monotherapy in the Parkinson disease: a clinical and pharmacokinetic study.
treatment of early PD: a randomized, controlled study. Neurology Arch Neurol 2005; 62: 905–10.
1999; 53: 573–79. 36 Antonini A, Isaias IU, Canesi M, et al. Duodenal levodopa infusion
13 Shannon KM, Bennett JP Jr, Friedman JH; for the Pramipexole for advanced Parkinson’s disease: 12-month treatment outcome.
Study Group. Efficacy of pramipexole, a novel dopamine agonist, as Mov Disord 2007; 22: 1145–49.
monotherapy in mild to moderate Parkinson‘s disease. Neurology 37 Koller WC, Hutton JT, Tolosa E, Capilldeo R; for the Carbidopa/
1997; 49: 724–28. Levodopa Study Group. Immediate-release and controlled-release
14 Adler CH, Sethi DK, Hauser RA, et al. Ropinirole for the treatment carbidopa/levodopa in PD: a 5-year randomized multicenter study.
of early Parkinson’s disease. Neurology 1997; 49: 393–99. Neurology 1999; 53: 1012–19.
15 DA agonists—non-ergot derivatives. Ropinirole: management of 38 PDtrials. Primary objective of STRIDE-PD study was not achieved.
Parkinson’s disease. Mov Disord 2002; 17 (suppl 4): S98–102. http://www.pdtrials.org/en/clinical_research_news/release/
16 DA agonists—non-ergot derivatives. Pramipexole: management of pr_1236180097 (accessed Aug 17, 2009).
Parkinson’s disease. Mov Disord 2002; 17 (suppl 4): S93–97. 39 Parkinson Study Group CALM Cohort Investigators. Long-term
17 DA agonists—non-ergot derivatives. Piribedil: management of effect of initiating pramipexole vs levodopa in early Parkinson
Parkinson’s disease. Mov Disord 2002; 17 (suppl 4): S90–92. disease. Arch Neurol 2009; 66: 563–70.
18 DA agonists—ergot derivatives. Pergolide: management of 40 Hauser RA, Rascol O, Korczyn AD, et al. Ten-year follow-up of
Parkinson’s disease. Mov Disord 2002; 17 (suppl 4): S79–82. Parkinson’s disease patients randomized to initial therapy with
19 Goetz CG, Poewe W, Rascol O, Sampaio C. Evidence-based medical ropinirole or levodopa. Mov Disord 2007; 22: 2409–17.
review update: pharmacological and surgical treatments of 41 Katzenschlager R, Head J, Schrag A, Ben-Shlomo Y, Evans A,
Parkinson’s disease: 2001 to 2004. Mov Disord 2005; 20: 523–39. Lees AJ. Fourteen-year final report of the randomized PDRG-UK
20 Montastruc JL, Rascol O, Senard JM, Rascol O. A randomised trial comparing three initial treatments in PD. Neurology 2008;
controlled study comparing bromocriptine to which levodopa 71: 474–80.
was later added, with levodopa alone in previously untreated 42 European Medicines Agency. Guideline on clinical investigation of
patients with Parkinson’s disease: a five year follow up. medicinal products in the treatment of Parkinson’s disease
J Neurol Neurosurg Psychiatry 1994; 57: 1034–38. (CHMP/563/95 Rev.1). July, 2008. http://www.emea.europa.eu/
21 Rinne UK, Bracco F, Chouza C, et al; for the PKDS009 Study pdfs/human/ewp/056395en.pdf (accessed Aug 7, 2009).
Group. Early treatment of Parkinson’s disease with cabergoline 43 Olanow CW. Can we achieve neuroprotection with currently
delays the onset of motor complications. Results of a double-blind available anti-parkinsonian interventions? Neurology 2009;
levodopa controlled trial. Drugs 1998; 55 (suppl 1): 23–30. 72: S59–64.
22 Rinne UK. Lisuride, a dopamine agonist in the treatment of early 44 Whone AL, Remy P, Davis MR, et al. The REAL-PET study: Slower
Parkinson’s disease. Neurology 1989; 39: 336–39. progression in early Parkinson’s disease treated with ropinirole
23 Jankovic J, Watts RL, Martin W, Boroojerdi B. Transdermal compared with levodopa. Neurology 2002; 58 (suppl 3): A82.
rotigotine: double-blind, placebo-controlled trial in Parkinson 45 Dopamine transporter brain imaging to assess the effects of
disease. Arch Neurol 2007; 64: 676–82. pramipexole vs levodopa on Parkinson disease progression.
24 Watts RL, Jankovic J, Waters C, Rajput A, Boroojerdi B, Rao J. JAMA 2002; 287: 1653–61.
Randomized, blind, controlled trial of transdermal rotigotine in 46 Ahlskog JE. Slowing Parkinson’s disease progression: recent
early Parkinson disease. Neurology 2007; 68: 272–76. dopamine agonist trials. Neurology 2003; 60: 381–89.
25 Rascol O, Dubois B, Caldas AC, Senn S, Del Signore S, Lees A. 47 Jennings D, Tabamo R, Seibyl J, Marek K. InSPECT: investigating
Early piribedil monotherapy of Parkinson’s disease: a planned the effect of short-term treatment with pramipexole or levodopa on
seven-month report of the REGAIN study. Mov Disord 2006; 123b-CIT and SPECT imaging. Mov Disord 2007; 22 (suppl 16):
21: 2110–15. S143 (abstract).

www.thelancet.com/neurology Vol 8 October 2009 935


Review

48 Ravina B, Eidelberg D, Ahlskog JE, et al. The role of radiotracer 71 Högl B, Seppi K, Brandauer E, et al. Increased daytime sleepiness
imaging in Parkinson disease. Neurology 2005; 64: 208–15. in Parkinson‘s disease: a questionnaire survey. Mov Disord 2003;
49 Marek K, Jennings D, Tamagnan G, Seibyl J. Biomarkers for 18: 319–23.
Parkinson’s disease: tools to assess Parkinson’s disease onset and 72 Paus S, Brecht HM, Koster J, Seeger G, Klockgether T, Wullner U.
progression. Ann Neurol 2008; 64 (suppl 2): S111–21. Sleep attacks, daytime sleepiness, and dopamine agonists in
50 Parkinson Study Group. A controlled, randomized, delayed-start Parkinson’s disease. Mov Disord 2003; 18: 659–67.
study of rasagiline in early Parkinson disease. Arch Neurol 2004; 73 Marinus J, Visser M, van Hilten JJ, Lammers GJ, Stiggelbout AM.
61: 561–66. Assessment of sleep and sleepiness in Parkinson disease. Sleep
51 Olanow CW, Hauser RA, Jankovic J, et al. A randomized, 2003; 26: 1049–54.
double-blind, placebo-controlled, delayed start study to assess 74 Kumar S, Bhatia M, Behari M. Excessive daytime sleepiness in
rasagiline as a disease modifying therapy in Parkinson’s disease Parkinson’s disease as assessed by Epworth Sleepiness Scale (ESS).
(the ADAGIO study): rationale, design, and baseline characteristics. Sleep Med 2003; 4: 339–42.
Mov Disord 2008; 23: 2194–201. 75 Monaca C, Duhamel A, Jacquesson JM, et al. Vigilance troubles in
52 Olanow CW, Rascol O. Early rasagiline treatment slows UPDRS Parkinson’s disease: a subjective and objective polysomnographic
decline in the ADAGIO delayed start study. Ann Neurol 2008; study. Sleep Med 2006; 7: 448–53.
64 (suppl 12): S68 (abstract). 76 Ferreira JJ, Desboeuf K, Galitzky M, et al. Sleep disruption, daytime
53 Schapira A, Hsu HH, Scrine MF, Gordon KL, Marek K. PROUD: somnolence and ’sleep attacks’ in Parkinson’s disease: a clinical
The impact of early vs delayed treatment with pramipexole on survey in PD patients and age-matched healthy volunteers.
new onset Parkinson’s disease. Mov Disord 2008; 23 (suppl 1): Eur J Neurol 2006; 13: 209–14.
S194 (abstract). 77 Verbaan D, Marinus J, Visser M, van Rooden SM, Stiggelbout AM,
54 Poewe W, Kleedorfer B, Wagner M, Bosch S, Schelosky L. van Hilten JJ. Patient-reported autonomic symptoms in Parkinson
Continuous subcutaneous apomorphine infusions for fluctuating disease. Neurology 2007; 69: 333–41.
Parkinson’s disease. Long-term follow-up in 18 patients. 78 Ghorayeb I, Loundou A, Auquier P, Dauvilliers Y, Bioulac B,
Adv Neurol 1993; 60: 656–59. Tison F. A nationwide survey of excessive daytime sleepiness in
55 Dewey RB Jr, Hutton JT, LeWitt PA, Factor SA. A randomized, Parkinson’s disease in France. Mov Disord 2007; 22: 1567–72.
double-blind, placebo-controlled trial of subcutaneously injected 79 Frucht S, Rogers JD, Greene PE, Gordon MF, Fahn S. Falling asleep
apomorphine for parkinsonian off-state events. Arch Neurol 2001; at the wheel: motor vehicle mishaps in persons taking pramipexole
58: 1385–92. and ropinirole. Neurology 1999; 52: 1908–10.
56 LeWitt PA, Lyons KE, Pahwa R. Advanced Parkinson disease treated 80 Seppi K, Hogl B, Diem A, Peralta C, Wenning GK, Poewe W.
with rotigotine transdermal system: PREFER study. Neurology 2007; Levodopa-induced sleepiness in the Parkinson variant of multiple
68: 1262–67. system atrophy. Mov Disord 2006; 21: 1281–83.
57 Poewe WH, Rascol O, Quinn N, et al. Efficacy of pramipexole and 81 Voon V, Fox SH. Medication-related impulse control and repetitive
transdermal rotigotine in advanced Parkinson’s disease: a behaviors in Parkinson disease. Arch Neurol 2007; 64: 1089–96.
double-blind, double-dummy, randomised controlled trial. 82 Weintraub D, Siderowf AD, Potenza MN, et al. Association of
Lancet Neurol 2007; 6: 513–20. dopamine agonist use with impulse control disorders in Parkinson
58 Facca A, Sanchez-Ramos J. High-dose pergolide monotherapy in disease. Arch Neurol 2006; 63: 969–73.
the treatment of severe levodopa-induced dyskinesias. Mov Disord 83 Voon V, Hassan K, Zurowski M, et al. Prevalence of repetitive and
1996; 11: 327–29. reward-seeking behaviors in Parkinson disease. Neurology 2006;
59 Cristina S, Zangaglia R, Mancini F, Martignoni E, Nappi G, 67: 1254–57.
Pacchetti C. High-dose ropinirole in advanced Parkinson’s disease 84 Isaias IU, Siri C, Cilia R, De Gaspari D, Pezzoli G, Antonini A. The
with severe dyskinesias. Clin Neuropharmacol 2003; 26: 146–50. relationship between impulsivity and impulse control disorders in
60 Yamamoto M, Schapira AH. Dopamine agonists in Parkinson’s Parkinson’s disease. Mov Disord 2008; 23: 411–15.
disease. Expert Rev Neurother 2008; 8: 671–77. 85 Voon V, Hassan K, Zurowski M, et al. Prospective prevalence of
61 Rascol O, Goetz C, Koller W, Poewe W, Sampaio C. Treatment pathologic gambling and medication association in Parkinson
interventions for Parkinson’s disease: an evidence based disease. Neurology 2006; 66: 1750–52.
assessment. Lancet 2002; 359: 1589–98. 86 Weintraub D. Dopamine and impulse control disorders in
62 Stowe RL, Ives NJ, Clarke C, et al. Dopamine agonist therapy in Parkinson’s disease. Ann Neurol 2008; 64 (suppl 2): S93–100.
early Parkinson’s disease. Cochrane Database Syst Rev 2008; 87 Cilia R, Siri C, Marotta G, et al. Functional abnormalities
2: CD006564. underlying pathological gambling in Parkinson disease.
63 Biglan KM, Holloway RG Jr, McDermott MP, Richard IH. Risk Arch Neurol 2008; 65: 1604–11.
factors for somnolence, edema, and hallucinations in early 88 Shaunak S, Wilkins A, Pilling JB, Dick DJ. Pericardial,
Parkinson disease. Neurology 2007; 69: 187–95. retroperitoneal, and pleural fibrosis induced by pergolide.
64 Blackard WG. Edema--an infrequently recognized complication of J Neurol Neurosurg Psychiatry 1999; 66: 79–81.
bromocriptine and other ergot dopaminergic drugs. Am J Med 1993; 89 Bowler JV, Ormerod IE, Legg NJ. Retroperitoneal fibrosis and
94: 445. bromocriptine. Lancet 1986; 2: 466.
65 van Hilten JJ, Weggeman M, van der Velde EA, Kerkhof GA, 90 Demonet JF, Rostin M, Dueymes JM, Ioualalen A, Montastruc JL,
van Dijk JG, Roos RA. Sleep, excessive daytime sleepiness and Rascol A. Retroperitoneal fibrosis and treatment of Parkinson’s
fatigue in Parkinson’s disease. J Neural Transm Park Dis Dement Sect disease with high doses of bromocriptine. Clin Neuropharmacol
1993; 5: 235–44. 1986; 9: 200–01.
66 Tandberg E, Larsen JP, Karlsen K. Excessive daytime sleepiness and 91 Wiggins J, Skinner C. Bromocriptine induced pleuropulmonary
sleep benefit in Parkinson’s disease: a community-based study. fibrosis. Thorax 1986; 41: 328–30.
Mov Disord 1999; 14: 922–27.
92 Ward CD, Thompson J, Humby MD. Pleuropulmonary and
67 Ondo WG, Dat Vuong K, Khan H, Atassi F, Kwak C, Jankovic J. retroperitoneal fibrosis associated with bromocriptine treatment.
Daytime sleepiness and other sleep disorders in Parkinson’s J Neurol Neurosurg Psychiatry 1987; 50: 1706–07.
disease. Neurology 2001; 57: 1392–96.
93 Kains JP, Hardy JC, Chevalier C, Collier A. Retroperitoneal fibrosis
68 Hobson DE, Lang AE, Martin WR, Razmy A, Rivest J, Fleming J. in two patients with Parkinson’s disease treated with bromocriptine.
Excessive daytime sleepiness and sudden-onset sleep in Parkinson Acta Clin Belg 1990; 45: 306–10.
disease: a survey by the Canadian Movement Disorders Group.
94 Todman DH, Oliver WA, Edwards RL. Pleuropulmonary fibrosis
JAMA 2002; 287: 455–63.
due to bromocriptine treatment for Parkinson’s disease.
69 Tan EK, Lum SY, Fook-Chong SM, et al. Evaluation of somnolence Clin Exp Neurol 1990; 27: 79–82.
in Parkinson’s disease: comparison with age- and sex-matched
95 Hely MA, Morris JG, Lawrence S, Jeremy R, Genge S. Retroperitoneal
controls. Neurology 2002; 58: 465–68.
fibrosis, skin and pleuropulmonary changes associated with
70 Brodsky MA, Godbold J, Roth T, Olanow CW. Sleepiness in bromocriptine therapy. Aust N Z J Med 1991; 21: 82–84.
Parkinson’s disease: a controlled study. Mov Disord 2003; 18: 668–72.

936 www.thelancet.com/neurology Vol 8 October 2009


Review

96 Agarwal P, Fahn S, Frucht SJ. Diagnosis and management of 103 Antonini A, Poewe W. Fibrotic heart-valve reactions to
pergolide-induced fibrosis. Mov Disord 2004; 19: 699–704. dopamine-agonist treatment in Parkinson’s disease.
97 Townsend M, MacIver DH. Constrictive pericarditis and Lancet Neurol 2007; 6: 826–29.
pleuropulmonary fibrosis secondary to cabergoline treatment for 104 Simonis G, Fuhrmann JT, Strasser RH. Meta-analysis of heart valve
Parkinson’s disease. Heart 2004; 90: e47. abnormalities in Parkinson’s disease patients treated with
98 Tintner R, Manian P, Gauthier P, Jankovic J. Pleuropulmonary dopamine agonists. Mov Disord 2007; 22: 1936–42.
fibrosis after long-term treatment with the dopamine agonist 105 Zanettini R, Antonini A, Gatto G, Gentile R, Tesei S, Pezzoli G.
pergolide for Parkinson Disease. Arch Neurol 2005; 62: 1290–95. Valvular heart disease and the use of dopamine agonists for
99 Pritchett AM, Morrison JF, Edwards WD, Schaff HV, Connolly HM, Parkinson’s disease. N Engl J Med 2007; 356: 39–46.
Espinosa RE. Valvular heart disease in patients taking pergolide. 106 Roth BL. Drugs and valvular heart disease. N Engl J Med 2007;
Mayo Clin Proc 2002; 77: 1280–86. 356: 6–9.
100 Van Camp G, Flamez A, Cosyns B, Goldstein J, Perdaens C, 107 Spottke AE, Reuter M, Machat O, et al. Cost of illness and its
Schoors D. Heart valvular disease in patients with Parkinson’s predictors for Parkinson‘s disease in Germany.
disease treated with high-dose pergolide. Neurology 2003; Pharmacoeconomics 2005; 23: 817–36.
61: 859–61. 108 Chapuis S, Ouchchane L, Metz O, Gerbaud L, Durif F. Impact of
101 Van Camp G, Flamez A, Cosyns B, et al. Treatment of Parkinson’s the motor complications of Parkinson’s disease on the quality of
disease with pergolide and relation to restrictive valvular heart life. Mov Disord 2005; 20: 224–30.
disease. Lancet 2004; 363: 1179–83. 109 University of South Florida. Patients starting Parkinson’s drug
102 Bleumink GS, van der Molen-Eijgenraam M, Strijbos JH, rasagaline earlier do better. http://hscweb3.hsc.usf.edu/health/
Sanwikarja S, van Puijenbroek EP, Stricker BH. now/?p=3564 (accessed Aug 18, 2009).
Pergolide-induced pleuropulmonary fibrosis.
Clin Neuropharmacol 2002; 25: 290–93.

www.thelancet.com/neurology Vol 8 October 2009 937

You might also like