201
Views
26
CrossRef citations to date
0
Altmetric
Expert Opinion

Update on the use of pramipexole in the treatment of Parkinson’s disease

Pages 337-352 | Published online: 11 Apr 2008

Abstract

Pramipexole is a non-ergot dopamine agonist shown to be efficacious in the treatment of Parkinson’s disease (PD). This review addresses the literature concerning pramipexole’s efficacy in treating motor and non-motor symptoms in PD, its impact on the development of dyskinesias and response fluctuations, the issue of neuroprotection, and the risk for developing adverse events such as increased somnolence, attacks of sudden onset of sleep, cardiac valvulopathy and impulse control disturbances.

Parkinson’s disease

Parkinson’s disease (PD) is the most common neurodegenerative movement disorder, affecting almost 1% of the population above the age of 60 years. Its early manifestation is parkinsonism, a progressive impairment of movement control including rigidity, rest tremor, and bradykinesia, while postural instability is a later feature. Non-motor symptoms caused by autonomic disturbances, dysregulation of mood and sleep, changes in cognition and behavior, are present throughout the course of the disease and may become, in advanced stages, the most important cause of disability.

Introduction to the dopaminergic treatment of Parkinson’s disease

Levodopa

The most effective and widely used drug in PD is still levodopa, metabolized to dopamine in the brain. During the early phases, motor symptoms in PD respond generally well to levodopa treatment. With advancing disease, treatment related complications such as response fluctuations (“wearing off”, “on-off”), motor disturbances (dyskinesias, “off” dystonia), neuropsychiatric side effects (hallucinations, delusions, confusion), and diminished response to levodopa, emerge as major causes of disability.

Dopamine agonists

The levodopa related complications prompted the development of alternative antiparkinsonian drugs, such as cathecolamine-O-methyl-transferase inhibitors, monoamine-oxidase-B inhibitors, and the synthetic dopamine agonists (DA). The DA present several advantages over levodopa such as direct stimulation of striatal dopaminergic neurons, longer half-life providing a more continuous stimulation at the dopamine receptors, lack of oxidative metabolites, more reliable absorption and transport.

DA have been shown to be effective in PD, both as monotherapy and as adjuvants to levodopa (CitationCotzias et al 1975; CitationKartzinel et al 1976; CitationLibman et al 1987; CitationAhlskog et al 1994; CitationRinne et al 1997; CitationBrooks et al 1998; CitationParkinson Study Group 2000; CitationPoewe and Wenning 2000).

Pramipexole in Parkinson’s disease

This review will present the DA pramipexole in the treatment of PD, focusing on the latest developments.

Pharmacodynamics

Pramipexole is a non-ergotamine full agonist at the D2 subfamily of dopamine receptors, with higher selectivity for D3 than for D2 and D4 dopamine receptors (CitationMierau and Schingnitz 1992; CitationSvensson et al 1994; CitationMierau 1995; CitationMierau et al 1995; CitationBennett and Piercey 1999). Thereby, pramipexole is able to simultaneously excite the direct striatopallidal pathway (by D3 stimulation) and to inhibit the indirect striatopallidal pathway (by D2 stimulation), alleviating PD symptoms by mimicking dopamine’s effects in the striatum. Since the D3 receptors enjoy greatest predominance in the limbic system (CitationBouthenet et al 1991), pramipexole has the theoretical potential to also have an impact on psychiatric symptoms in PD (CitationBennett and Piercey 1999). In addition, pramipexole has a very low affinity for 5-HT2A, 5-HT2B, and D1 receptors, partly explaining its beneficial effects in the context of cardiac valvulopathy and dyskinesias, as discussed later.

Pharmacokinetics

Pramipexole has an absolute oral bioavailability greater than 90%, indicating good absorption and little first pass metabolism. It exhibits linear pharmacokinetics and less than 20% is protein bound; more than 90% of the absorbed dose is eliminated unchanged and almost exclusively by the kidneys, motivating a dose reduction with low creatinine clearance. Its elimination half-life is 8 hours in young, healthy volunteers (CitationHäselbarth 1994a, Citationb; CitationWright et al 1997; CitationBennett and Piercey 1999).

In men and postmenopausal women with PD, gender had no impact on the pharmacokinetics of pramipexole, and pramipexole did not alter levodopa’s bioavailability (CitationKompoliti et al 2002).

In contrast to other DA, pramipexole has been found to exert no strong cytochrome P 450 inhibition in vitro, minimizing the risk for drug-drug interactions (CitationWynalda and Wienkers 1997).

Clinical efficacy

Monotherapy in early Parkinson’s disease

Summary of previous results: The safety and efficacy of pramipexole as monotherapy in early PD were originally evaluated in 2 placebo-controlled clinical trials enrolling 599 PD subjects (CitationParkinson Study Group 1997; CitationShannon et al 1997). In addition, the Comparison of the Agonist Pramipexole with Levodopa on Motor Complications of PD (CALM-PD) study, conducted by the Parkinson Study Group (PSG), investigated initial treatment with pramipexole vs initial treatment with levodopa in 301 patients with early PD, requiring dopaminergic therapy. The subjects were randomly assigned to receive 1.5 mg pramipexole or 300 mg levodopa daily. The two treatment groups were of equal size. After the first 10 weeks of treatment, open-label levodopa (and during the last year of the study also other antiparkinsonian medication) could be added as needed, for better symptom control (CitationParkinson Study Group 2000). The CALM-PD study design and the subsequent long follow up (up to 58 months), generated a wealth of data regarding the effects of pramipexole on a variety of PD features.

The combined results of these three pivotal studies showed that pramipexole, started at low initial doses and up-titrated weekly, was effective and well tolerated in mild to moderate PD. No differences in effectiveness based on age or gender were detected, and no dose-response relationship could be demonstrated. Thus, doses of 1.5 mg, 3 mg, 4.5 mg, and 6 mg/day did provide similar significant benefit in reducing PD signs and symptoms compared with placebo. However, several side effects were dose related. On the Unified Parkinson’s Disease Rating Scale (UPDRS) (CitationLang 1989) significant improvement from baseline could be seen in both section II (around 2 points), and section III (around 5 points), compared with placebo (<0.6 points on both sections).

New studies: The 4-year results of the CALM-PD study showed that, after 4 years, initial treatment with pramipexole resulted in a significant reduction of the risk for developing treatment fluctuations and dyskinesias but in a higher risk for freezing, vs initial treatment with levodopa. On the other side, a greater improvement in the total UPDRS score was seen in the levodopa group vs the pramipexole group (2 ± 15.4 points vs −3.2 ± 17.3 points). More patients in the pramipexole group needed at 4 years open-label levodopa vs the levodopa group (72% vs 59%) but the mean daily levodopa dose was lower in the pramipexole group (434 ± 498 mg/day vs 702 ± 461 mg/day). More pramipexole patients withdrew prior to the final follow-up visit than levodopa patients (45% vs 33 %) due to side-effects (CitationHolloway et al 2004).

Adjuvant therapy in advanced Parkinson’s disease

Summary of previous results: Several clinical trials examined the medium and long term efficacy of pramipexole at a maximal dosage of 4.5 mg daily, as an adjuvant to levodopa therapy in moderate to advanced PD. The combined results showed that pramipexole was well tolerated, associated with a reduction in daily L-dopa usage, and in daily “off” time, together with improvement in several parkinsonism and disability scorings (CitationLieberman et al 1997; CitationBennett and Piercey 1999; CitationPinter et al 1999, Citation2000; CitationWeiner et al 2001).

New studies: Möller et al reported 2005 on 354 levodopa-treated PD patients with motor fluctuations. A first, double-blind, placebo controlled phase of 32 weeks was followed by an open-label phase with a maximum duration of 57 months. Pramipexole was administered t.i.d. at a total daily dosage varying between 0.375 and 4.5 mg. The effect emerged at a daily dose of 0.75 mg. The levodopa dosage could be decreased in 39% of the pramipexole treated patients vs in 12.8% of the placebo group. Pramipexole as an adjuvant therapy was found to be superior to placebo and effective at reducing UPDRS sum scores of parts II and III by 30%, and “off time” by 2.5 hours/day. Improvement was also seen on UPDRS part I through the items motivation/initiative and depression, but not intellectual impairment and thought disorder. Overall, pramipexole had a better anti tremor effect than placebo (change −2.5 and −0.5 respectively on the sum of UPDRS tremor-related items). Tremor was considered prominent when the baseline sum score of UPDRS III tremor-related items was at least 8 (6 if unilateral tremor) or the relative contribution of tremor items to the total UPDRS II and III score was >20%. Patients with prominent resting tremor benefited more from pramipexole than from placebo. Thus, the mean (SD) decrease in the total tremor score in the high tremor score group was 8 (4.2) for pramipexole compared with 3.7 (5.7) for placebo. The prevalence of somnolence was low (1.7% in the pramipexole group vs 2.2 in the placebo group), but this symptom was not specifically asked for, just recorded if spontaneously reported. There were more dyskinesias and visual hallucinations in the pramipexole group vs the placebo group (30% vs 8.7%, and 11.1% vs 4.4% respectively). During the double-blind phase, totally 27% of all patients (17.8% in the pramipexole group and 36.1% in the placebo group) had an adverse event that led to the discontinuation of the study medication, most frequently due to aggravated parkinsonism. During the open-label phase, 23.7% of all subjects had an adverse event that led to discontinuation of study medication, most often aggravated parkinsonism, visual hallucinations, and dyskinesias. Pramipexole’s long-term efficacy and tolerability were good (CitationMoller et al 2005).

Two studies examined pramipexole’s efficacy in other ethnical groups than Caucasians. The first, a 15-week-long, randomized, double-blind, placebo-controlled, parallel-group study, was conducted in Hong Kong and Taiwan in 150 untreated and levodopa-treated Chinese patients with both early and advanced PD (CitationWong et al 2003). Pramipexole, both as monotherapy and as adjuvant to levodopa, was found to be more effective than placebo for symptom reduction measured with UPDRS Part I, Part III, and Parts I plus III. Pramipexole addition to levodopa led to a decrease in the number of “off” hours per day from 7.07 to 6.15 hours/day. In the placebo group the “off” time increased from 5.59 to 6.87 hours/day. Slightly more adverse events were reported overall for pramipexole treated patients (86%) compared with the placebo group (71%), with the largest differences for constipation, nausea, dry mouth, dyskinesias, somnolence and hallucinations. Interestingly, more tremor was reported in the placebo group (11.7%) compared with the pramipexole group (1.4%), possibly indicating a more specific antitremor effect for pramipexole.

The results of the second study were published recently. In a 10 weeks long, double-blind, randomized, placebo-controlled trial, pramipexole was administrated as an adjuvant therapy to 144 levodopa-treated PD patients of African American, Asian, and Hispanic heritage (CitationParkinson Study Group 2007). Overall, pramipexole was found to have a similar efficacy in all ethnical groups, and of the same magnitude as shown in former trials on primarily non-Hispanic whites. African Americans showed a statistically significant improvement in tremor measured by UPDRS part 3 tremor subscales. The overall tolerability and safety were also similar across the ethnical lines, but with different profiles in the different groups. More patients of Asian heritage reported asthenia compared with African Americans and Hispanics, while confusion was more frequent in the African heritage group. Hallucinations and insomnia were more common with pramipexole than with placebo. No sleep attacks were reported. Premature withdrawals occurred 3 times more frequently in persons of African heritage than in Asians, usually due to comorbid illnesses. These differences were not statistically significant but may indicate the need of more research in this field. A pharmacokinetic substudy on 40 patients did not show any differences in the pharmacokinetic profile among the ethnic groups.

Comparative studies

Rotigotine: In 506 subjects with advanced PD already treated with levodopa and other antiparkinsonian drugs, pramipexole was compared with rotigotine patch and placebo, as adjuvants. Primary efficacy variables were absolute change in total hours “off ” from baseline to end of study, as recorded by patients on home diaries, and responder rate, defined as the proportion of patients with a 30% or more reduction in absolute off time per day. Both active therapies showed similar efficacy in respect to both primary and secondary response parameters, and were superior to placebo. Responder rates and reduction in off-time were in absolute numbers somewhat higher for pramipexole, but not enough to show any statistically significance. Both pramipexole and rotigotine showed good tolerability, with more reports of hallucinations, dyskinesias and dizziness in the pramipexole group, and more reports of nausea and application site reactions in the rotigotine group (CitationPoewe et al 2007).

Bromocriptine: Pramipexole was compared with bromocriptine and placebo as adjuvant therapy in 325 patients with advanced PD. The primary end points were the change from baseline to study termination on UPDRS parts II and III. The magnitude of the observed changes on the primary endpoints was larger, the response came earlier, and the improvement from baseline to each subsequent visit was greater in the pramipexole group compared with bromocriptine, throughout the treatment. Statistically, the two treatment groups were similar, presumably due to an insufficient number of patients and low power. Both active treatments showed the same rate of responders and the rate of adverse events was similar for all 3 groups. Overall, pramipexole was shown to be clearly superior to placebo and not inferior to bromocriptine (CitationMizuno et al 2003).

Guttman et al compared adjuvant therapy with pramipexole and placebo in PD patients with a declining response to levodopa, including a bromocriptine group for comparison with placebo. The primary end points were UPDRS parts II and III. They found no differences in efficacy and adverse events between pramipexole and bromocriptine but the study was not powered for detecting any. Both DA were superior to placebo (CitationGuttman 1997).

Pergolide: Rektorová et al found no significant differences between pramipexole and pergolide as add-on therapies to levodopa in advanced PD patients with depression, as measured on UPDRS scales (CitationRektorova et al 2003).

In an open label study including 25 patients, Hanna et al found no significant difference in the efficacy of pramipexole vs pergolide as adjuvant therapies in mild to moderate PD, although there was a slight trend in favour or pramipexole (CitationHanna et al 2001).

Mixed: Goetz et al investigated slow (over 8 weeks) vs rapid (over 1 day) switch from bromocriptine/pergolide to pramipexole in 16 PD patients on stable levodopa therapy. The primary outcome was the time required to achieve a UPDRS motor score superior to baseline without increased side effects. The conversion rate to pramipexole was 1:1 for pergolide and 1:10 for bromocriptine. They found that a rapid titration based upon an equivalency dose calculation was preferable. Pramipexole was superior to both pergolide and bromocriptine in respect to motor UPDRS scores (CitationGoetz et al 1999).

Reichmann et al conducted a study in 1202 PD patients who were switched slowly or quickly to pramipexole from any other oral DA, as add-on therapy, because of insufficient effectiveness on motor performance, tremor, and mood (depression, anhedonia). The switch to pramipexole led to improvement of all symptoms. Resting tremor improved more than postural tremor. No difference was found between either switching procedure in respect to clinical efficacy, tolerability, and safety. Most patients tolerated pramipexole better or equally well, compared with their previous DA (CitationReichmann et al 2006).

An analysis of 7 trials on pramipexole, ropinirole, pergolide, entacapone and tolcapone as add-on therapies to levodopa in PD found that pramipexole and entacapone were the best choices for obtaining a greater reduction in levodopa dose and in “off ” time, and a better tolerability. All active drugs caused about the same amount of dyskinesias except higher dose tolcapone (600 mg) which caused more dyskinesias (CitationInzelberg et al 2000).

In conclusion, most data suggest that pramipexole and the other DA are similarly efficacious as adjuvant therapy to levodopa in advanced PD.

Specific symptoms

Drug-resistant tremor in Parkinson’s disease

Parkinsonian tremor can occasionally be unresponsive to dopaminergic therapy and notoriously difficult to treat.

Navan et al conducted a randomized, double-blind, 3-month parallel study in 30 PD patients, most of whom were already medicated. The aim was to investigate the effects of pramipexole, pergolide, and placebo on parkinsonian tremor. They found that both pergolide and pramipexole had significantly better overall antitremor effect than placebo, and their efficacy was similar. However, pergolide was superior to pramipexole in alleviating rest tremor and postural tremor scored clinically. Both drugs had similar beneficial effects on the UPDRS motor subscores and the tolerability was better for pramipexole (CitationNavan et al 2003b). The same group did also report a similar but modest reduction in rest tremor with a single 500 μg dose of either pramipexole or pergolide, compared with placebo (CitationNavan et al 2003a).

Pogarell et al investigated 84 early and advanced PD patients with marked drug-resistant tremor under a stable and optimized antiparkinsonian medication. Pramipexole or placebo were administered as add on medication. The primary end point was the absolute change in tremor score, defined as the sum of UPDRS tremor related items (16, 20, 21) in “on” periods. Pramipexole was found to be significantly superior to placebo with a difference between treatment groups in the mean absolute change in tremor score of −4.4 points ([95% CI] −6.2 to −2.5; p < 0.0001), corresponding to a difference in the mean percentage change of −34.7% in favor of pramipexole (CitationPogarell et al 2002).

In conclusion, existing data suggest that pramipexole may be effective for treating drug-resistant tremor in PD.

Non-motor symptoms in Parkinson’s disease

In addition to the noticeable motor symptoms, less obvious and sometimes more difficult to treat non-motor symptoms may greatly increase the distress and disability in PD. Increasing evidence supports pramipexole’s efficacy in ameliorating some of these symptoms.

Depression: In non-PD patients with both unipolar and bipolar major depression, pramipexole was shown to be effective in alleviating depressive symptoms, in monotherapy or as an adjuvant (CitationCorrigan et al 2000; CitationLattanzi et al 2002; CitationGoldberg et al 2004). Depression and other psychiatric disorders affect up to 50% of all PD patients (CitationDooneief et al 1992; CitationTandberg et al 1996) with negative impact on motor-related disability and quality of life (QOL) (CitationSchrag et al 2000).

In a randomized trial from 2005 including 67 PD patients with major depression and without motor complications, pramipexole was compared with sertraline in respect to recovery from depression, defined as a final HAM-D (CitationHamilton 1960) score ≤8 (CitationLemke et al 2006). Pramipexole led to recovery in 60.6% of patients, which was significantly higher compared with sertraline (27.3%). However, there was no difference between the two treatments when depression was assessed with Zung self-relating depression score (CitationZung 1965). There were more adverse effects in the sertraline recipients, responsible for a 15% withdrawal from the study (5 patients). None of the 33 pramipexole patients withdrew. In the pramipexole group there was a significant improvement in motor symptoms as measured by UPDRS part III. A noticeable limitation of the study was the lack of a placebo group, given that there is a high degree of placebo response in depressed PD patients (CitationWeintraub et al 2005).

A randomized, open-label, comparative study investigated the effect of adjuvant therapy with pramipexole and pergolide in 41 non-demented, levodopa-treated PD patients, all of which had motor complications, including fluctuations and dyskinesias. About a third of the patients in each DA group had mild to moderate depression at baseline. A significant decrease of Zung depression score (CitationZung 1965) was reported for both DA, but improvement on Montgomery-Åsberg Depression Rating Scale (CitationMontgomery and Asberg 1979) was seen only among the pramipexole recipients. At the same time, pramipexole improved motor symptoms including motor complications, and this may also have been a cause for improved depression, although pramipexole was superior even when controlling for the motor improvement. There was no significant difference between pramipexole and pergolide as adjuvant in respect to motor symptoms, activities of daily living, and treatment complications. The daily levodopa dose could be significantly decreased, more so in the pergolide group (CitationRektorova et al 2003).

In conclusion, existing data suggest that pramipexole may play a role in reducing symptoms of depression in PD.

Anhedonia: An observational study on 657 PD patients in early and advanced disease stages (Hoehn and Yahr 1–5 [CitationHoehn and Yahr 1967]) found that pramipexole as add-on to levodopa therapy improved anhedonia, the reduced capacity to experience pleasure, as evaluated by the Snaith-Hamilton Pleasure Scale (CitationSnaith et al 1995). Notably, this was seen in both depressed and in non depressed PD patients, and was therefore not only due to an antidepressant effect. Meanwhile, in line with the results of other studies, it was seen that severe, moderate, and mild depression also decreased (CitationReichmann et al 2003; CitationLemke et al 2005; CitationLemke et al 2006).

Cognition: Cognitive impairment is common in PD, and up to a third of PD patients develop overt dementia (CitationAarsland et al 2005). As results from studies in healthy controls suggest that pramipexole may worsen cognitive abilities (Pizzagalli et al 2007), it is important to investigate its impact on cognition in PD.

Rektorová et al examined in a randomized study the effects of pramipexole and pergolide as adjuvant therapy to levodopa in 41 non-demented patients with advanced PD and a current depressive episode. While confirming that pramipexole had an antidepressant effect and that both DA improved motor performance, the results showed no significant effect on the neuropsychological tests applied. The conclusion was that both pramipexole and pergolide as adjuvant therapy to levodopa are safe in terms of effects on cognitive performance (CitationRektorova et al 2005).

Relja et al evaluated over 6 months the effects of pramipexole as add-on therapy in PD patients treated with levodopa. An exclusion criterion was evidence of possible dementia and only patients scoring above 25 on the mini-mental state examination (MMSE) were included. They found no statistically significant difference in performance on the cognitive tests applied, between those treated with levodopa in monotherapy and those adding pramipexole, but there was a trend for a greater decline on Letter Fluency (CitationBenton 1989) and Trail Making test (CitationReitan 1992) in the latter group (CitationRelja and Klepac 2006).

Brusa et al investigated in a randomized study 20 patients with early/mild PD treated with levodopa or with pramipexole. Patients with dementia were excluded. They found that pramipexole but not levodopa produced a significant impairment of short term verbal memory, attentional-executive functions, and verbal fluency (CitationBrusa et al 2003).

Given the fact that cognitive impairment is common in advanced PD, it would be of great interest to know more about pramipexole’s effects in a cognitively impaired PD population. In the previously mentioned studies, no demented PD subjects were included and no studies on pramipexole in a cognitively disabled PD population could be identified. A case report regarding an 85 years old demented lady with parkinsonism indicated that pramipexole might have contributed to a worsening of the patient’s cognition and general condition (CitationScoyni et al 2007). As cognitive decline is more common with advancing age and disease stage in PD, the results of one study on the use of DA in very elderly patients with PD are especially interesting in this context (CitationShulman et al 2000). In total, 43 non-demented PD patients (24 between 80 and 85, 14 between 86 and 90, and 5 between 91 and 95 years of age), with a mean disease duration of 11 years, were treated with pramipexole (mean daily dose 2.7 mg). The overall success rate over a mean treatment time of 14 months was 40% (17 of 43 patients). The adverse effect event rate was of the same magnitude as in younger patients (around 60 years old), and the adverse events profile was similar with that reported in younger patients, with the exception of hallucinations, which were more common in this very old age group. Thus 21% of the patients treated with pramipexole reported hallucinations. Unfortunately, the design of the study did not provide any specific data on cognition and the effects of pramipexole therapy.

Biglan et al found that better cognitive function defined as MMSE >28 was associated with a decreased risk for developing hallucinations after pramipexole therapy in PD (CitationBiglan et al 2007).

In the light of the aforementioned results and especially considering the higher risk for hallucinations with pramipexole therapy, there is reason to suspect that pramipexole may have deleterious effects on cognition in an advanced PD population showing cognitive deficits.

In conclusion, there are mixed results in respect to pramipexole’s impact on cognition in a non-demented PD population, with studies showing no deleterious effects and studies suggesting cognitive worsening. No studies suggesting that pramipexole treatment may lead to an improvement in cognition were identified. Although no studies examining the effects of pramipexole in a cognitively impaired PD population could be found, caution is recommended in this respect.

Apathy: Apathy is a frequent finding in PD, significantly associated with cognitive impairment, executive dysfunction, anxiety and depression (CitationStarkstein et al 1992; CitationAarsland et al 1999; CitationPluck and Brown 2002). Apathy levels in PD are higher than in equally disabled patients with other diagnoses (CitationPluck and Brown 2002). While apathy does not seem to respond to levodopa, there are some indications from clinical trials that pramipexole’s D3 agonist properties may be beneficial in this respect (CitationGuttman and Jaskolka 2001; CitationKumru et al 2006).

Miscellaneous symptoms

Restless legs syndrome: There may be an overrepresentation of restless legs syndrome (RLS) in PD and the relationship between the two diseases is not completely elucidated (CitationPoewe and Hogl 2004). Pramipexole has been shown to be effective in treating pure RLS (CitationMontplaisir et al 1999), as well as other DA, but no studies investigating pramipexole’s efficacy in the treatment of RLS in PD patients have been identified.

Weight loss: Weight loss is seen in PD patients already before diagnosis and continues throughout the course of the disease. Possible causes include: (1) increased energy expenditure due to increased muscular activity (rigidity, dyskinesias); (2) decreased energy intake, due to general movement impairment (difficulties in feeding, accessing food, and swallowing), and decreased appetite caused by depression; and (3) an inherent metabolic abnormality in PD (CitationChen et al 2003; CitationLorefalt et al 2006; CitationToth et al 1997). Increased body weight, as well as motor and mood improvement, were seen in 28 non-depressed, advanced PD patients on stable medication, 3 months after starting treatment with pramipexole (CitationKumru et al 2006). In addition, many patients treated with pramipexole reported a decrease in anxiety and apathy together with enhanced attention span and interest in daily activities. No patient reported dysphagia, vomiting or changes in eating behavior but 12 reported increased hunger. There was no relationship between changes in BMI and in UPDRS part III and the authors speculate that the weight increase was due to a direct or indirect action of pramipexole on the feeding centers in the hypothalamus.

Rapid eye movement (REM) sleep behavior disorder (RBD): Rapid eye movement (REM) sleep behaviour disorder (RBD) may involve a dopaminergic deficiency, as it is associated with both PD and RLS. The traditional first-line therapy for RBD is clonazepam. Nevertheless, 2 studies were found that reported on pramipexole in RBD, although none was directed at PD patients.

Fantini et al reported about 8 patients with idiopathic RBD, treated with pramipexole, 7 of which experienced a reduction in the frequency and intensity of RBD. In 5 patients this reduction was sustained (CitationFantini et al 2003).

Schmidt et al reported about 10 patients with RBD, 3 of which had parkinsonism. Pramipexole reduced markedly the frequency and severity of RBD in 89% of the patients (CitationSchmidt et al 2006).

The results of these studies are interesting but they have to be replicated in a PD population before gaining any acceptance in the treatment of PD related RBD.

Fatigue: Fatigue is an early symptom in some PD patients (CitationSchifitto 2006). While fatigue improved in patients with fibromyalgia with pramipexole treatment (CitationHolman and Myers 2005), it was reported as an adverse event in PD patients treated with pramipexole (CitationPinter et al 1999; CitationPogarell et al 2002).

Dopaminergic complications in Parkinson’s disease

The risk of experiencing response fluctuations (wearing off, on-off) or dyskinesias in PD has been evaluated at 40% after about 5 years of levodopa treatment (CitationAhlskog and Muenter 2001).

The CALM-PD study examined the risk for developing dopaminergic complications in early PD individuals initially treated with pramipexole monotherapy versus levodopa monotherapy. Eventually all patients could add levodopa for emerging disability. After 2 years, 28 % of subjects assigned to pramipexole developed dopaminergic complications vs 51% in the levodopa group (CitationParkinson Study Group 2000).

The 4-years results of the CALM-PD study showed that initial treatment with pramipexole vs initial treatment with levodopa resulted in a significant reduction of the risk for developing dyskinesias (24.5% vs 54%), and wearing off (47% vs 62.7%), but in a higher risk for freezing (37.1% vs 25.3%). The occurrence of disabling dyskinesias was uncommon and did not differ between the two treatment groups. The risk for on-off fluctuations was the same in both groups. Ten patients in the pramipexole group developed dyskinesias prior to the open-label levodopa treatment, 7 of which had never been exposed to levodopa (CitationParkinson Study Group 2000; CitationHolloway et al 2004). Initial pramipexole treatment was associated with the appearance of motor fluctuations before dyskinesias (CitationHauser et al 2006).

A comparison of several published studies on the treatment of early PD with levodopa, pramipexole, cabergoline, or ropinirole, showed a reduction in the risk for developing dyskinesias for all three DA compared with levodopa. This reduction was slightly higher for pramipexole and ropinirole compared with cabergoline (absolute risk reductions 20%, 25%, and 8% respectively). These findings are not unexpected, as dyskinesias seem to be mediated by the D1 receptor, for which pramipexole has a very low affinity compared with other compounds, such as cabergoline (CitationInzelberg et al 2003).

Neuroprotection

In vitro and in vivo studies

Pramipexole has been shown to have several neuroprotective actions, presumably through its antiapoptotic, antioxidant, and antitoxic effects, and through induction of neurotrophic factors. In cell cultures pramipexole protected against dopaminergic neuronal death induced by glutamate (CitationIzumi et al 2007), 1-methyl-4-phenylpyridinium (MPP+) (CitationPresgraves et al 2004), 6-hydroxydopamine (6-OHDA) (CitationLe et al 2000), rotenone (CitationGu et al 2004), L-dihydroxyphenylalanine (L-DOPA), and methamphetamine (CitationHall et al 1996); in a recent study, pramipexole was shown to prevent neurotoxicity induced by oligomers of beta-amyloid, suggesting that pramipexole may have a role to play also in the treatment of other neurodegenerative diseases related to beta-amyloid proteins (CitationUberti et al 2007); in a dopaminergic cell line, pramipexole attenuated neurotoxicity induced by dopamine and levodopa, possibly through a mechanism related to antioxidant activity, scavenging of free radicals such as hydrogen peroxide, and inhibition of apoptosis (CitationZou et al 1999). Pramipexole stimulated the production of a dopaminergic neurotrophic factor in tissue cultures (CitationCarvey et al 2001), and increased the Nurr1 gene expression and protein levels (CitationPan et al 2005). It has been hypothesized that by suppressing STN overactivity, pramipexole may ameliorate the subthalamicus nucleus mediated excitotoxicity. Finally, pramipexole reduced mitochondrial swelling and could thereby have an antiapoptotic effect (CitationCassarino et al 1998; CitationSayeed et al 2006). In mice, pramipexole treatment completely antagonized the neurotoxic effects of MPTP (CitationJoyce et al 2004).

In humans

The CALM-PD study attempted to assess pramipexole’s neuroprotective effect in humans, by quantifying the rate of decline from baseline in striatal ([123]I)beta-CIT-SPECT uptake, a measure of the density of striatal presynaptic dopaminergic innervation, reflecting the degeneration of substantia nigra. Forty-two PD subjects were randomized to initiate therapy with pramipexole and 40 with levodopa, both as monotherapy. Eventually, both groups could add levodopa when needed. After both 2 and 4 years of treatment, the group initially receiving pramipexole showed a slower rate of decline in striatal ([123]I)beta -CIT SPECT uptake compared with the group initially randomized to levodopa (7.1% vs 13.5% at 22 months (p = 0.004); and 16.0% vs 25.5% at 46 months (p = 0.01). In addition, the percentage loss from baseline in striatal ([123]I)beta -CIT uptake was correlated with the change from baseline in UPDRS at the 46-month evaluation (r = −0.40; p = 0.001) (CitationParkinson Study Group 2002). The interpretation of these results is equivocal due to the lack of a placebo group, and because it can not be ruled out that the differences in the rate of decline from baseline may be due to direct pharmacological effects of pramipexole on the ligand receptors, and not due to a slowing of the disease process with pramipexole (CitationRavina et al 2005). Both animal and human studies show for example that short term therapy with pramipexole can down-regulate striatal dopamine transporters (CitationGuttman et al 2001).

The results of a subanalysis of CALM-PD data argue against a neuroprotective action of pramipexole, which, as initial therapy in PD, did not lower the risk for dyskinesias once levodopa was added to the treatment, after controlling for levodopa dose. Nonetheless, pramipexole delayed the point in time when levodopa treatment was needed and thereby delayed also the appearance of dyskinesias (CitationConstantinescu et al 2007).

In conclusion, despite promising results in preclinical studies, there are still no convincing clinical data confirming a neuroprotective effect of pramipexole in PD.

Patient-focused perspective in Parkinson’s disease

Cost-effectiveness

Using data from the CALM-PD study, Noyes et al made a cost-effectiveness assessment of initial treatment with pramipexole vs levodopa, in early PD. The results reflecting the first 2 years of treatment suggested that pramipexole was not welfare enhancing due to higher costs compared with levodopa (CitationNoyes et al 2004). However, after 4 years, the cost-effectiveness of pramipexole increased, with the largest incremental benefit being noticed during the fourth year of the trial. The increased effectiveness was not due to a reduction in costs compared with levodopa, but to a greater gain in quality adjusted life years (QUALY). Subjects with lower QOL and more depressive symptoms at baseline showed the highest improvement in QUALY gains over time (CitationNoyes et al 2005).

Quality of life (QOL)

Health-related QOL (HRQOL) evaluates a person’s wellbeing. Several studies have shown that QOL decreases in PD patients as the disease progresses (CitationKarlsen et al 2000). Data from the CALM-PD study was used to compare the effect of pramipexole vs levodopa on disease-specific QOL. Although at 24 months, the levodopa group showed better QOL scores than the pramipexole group (CitationParkinson Study Group, 2000), there was no difference in the mean changes in the HRQOL between baseline and 48 months follow up (CitationHolloway et al 2004). However, this approach ignored the dynamics of HRQOL changes. Consequently, when taking into account the HRQOL values for all visits and accumulating gains and losses over time, by each treatment arm, a different pattern emerged. During the first year, HRQOL increased, and during years 2–4, it decreased in both arms. The deterioration was greater in the levodopa arm, although, at all times, there was a 5-point UPDRS benefit favoring levodopa, suggesting that pramipexole induced its improvement thorough non-motor and levodopa through motor mechanisms. The difference in HRQOL between the two treatment arms widened in years 3 and 4 in favor of pramipexole suggesting that a longer follow up may show even larger differences. The clinical relevance of these findings is not clear at this time (CitationNoyes et al 2006).

Tolerability and adverse events

As already discussed in section 3.3, pramipexole is generally well tolerated up to a total daily dose of 4.5 mg, both as monotherapy and as an adjuvant to levodopa in PD patients at different disease stages. The highest dosage of 6 mg daily led to a higher incidence of neuropsychiatric side effects, particularly somnolence, and there is generally a direct relationship between pramipexole dose and adverse events. Adverse events that appear to occur more often with pramipexole compared with placebo are sleep disturbances, gastrointestinal symptoms (nausea, constipation), postural hypotension, peripheral edema, impulse control disorders, and hallucinosis (CitationLieberman et al 1997; CitationMoller et al 2005; CitationParkinson Study Group, 1997; CitationParkinson Study Group, 2000; CitationPinter et al 1999; CitationShannon et al 1997; CitationWeiner et al 2001). The most alarming adverse events will be discussed in the following sections.

Increased somnolence and attacks of sudden onset of sleep

Sleep disturbances such as excessive daytime sleepiness (somnolence) and disorders of night time sleep patterns are common in PD and contribute to a general deterioration of the QOL (CitationTandberg et al 1998). Intuitively, increased somnolence should also be related to potentially fatal attacks of sudden onset of sleep while driving. Indeed, 75% of PD patients reporting such attacks have high scores on the Epworth Sleepiness Scale (ESS) (CitationJohns 1991). However, 25% of the patients have low scores on ESS, suggesting that sleep attacks may be a sleep phenomenon not necessarily related to increased somnolence (CitationPaus et al 2003). There are contradictory reports on the role of pramipexole in the development of somnolence and attacks of sudden onset of sleep, and, being a cause of great concern, this issue will be discussed at large in this review.

The initial report by Frucht et al from 1999, on 8 PD patients treated with pramipexole who experienced sudden, irresistible attacks of sleep, and sustained motor vehicle accidents, led to a driving ban for pramipexole treated patients in some countries (CitationFrucht et al 1999).

The following year Hauser et al reported data collected prior to Frucht’s report. Six of 22 (27%) PD patients randomized to pramipexole reported somnolence, compared with only 2 of 18 (11%) placebo patients. In addition, during the open-label extension period, 21 of 37 (57%) patients receiving pramipexole reported somnolence as an adverse event, 7 (19%) reported falling asleep while driving, and 2 (5%) reported minor motor vehicle accidents. Sleep latency normalized after withdrawal of pramipexole (CitationHauser et al 2000).

The 4-year results of the CALM-PD study showed that more somnolence was reported in the pramipexole group at 4 years, and while only 1 subject (0.7%) in the levodopa group withdrew because of somnolence, 12 subjects (8%) in the pramipexole group did that. Of these, 8 subjects (66%) described their somnolence as “sudden” or “unexpected” and 5 (41%) reported that these episodes occurred while driving. Two serious adverse events related to driving were reported in the levodopa group and 5 in the pramipexole group. Somnolence seemed to occur mostly during the escalation phase of pramipexole (CitationHolloway et al 2004). A secondary analysis of this data found that initial treatment with pramipexole in patients with early PD almost doubled the risk for developing somnolence (CitationBiglan et al 2007).

Etminan et al analyzed in two separate analyses the risk for somnolence in PD patients taking pramipexole and ropinirole vs placebo (4 trials) and patients taking these two DA as adjuvant therapy to levodopa (7 trials). They concluded that the risk for somnolence was higher with pramipexole or ropinirole both as monotherapy an as adjuvant to levodopa, compared with placebo or levodopa alone (CitationEtminan et al 2001). Pramipexole gave a lower risk for somnolence and hypotension compared to ropinirole, but a greater risk for hallucinations (CitationEtminan et al 2003).

Happe et al found that sedation in PD may be a class effect of DA, with no difference between ergot and non-ergot DA (CitationHappe and Berger, 2001).

O’Suilleabhain et al reported 2002 on 368 PD patients treated with levodopa and DA, in monotherapy or in combination therapy (CitationO’Suilleabhain and Dewey 2002). One hundred and seven patients took pramipexole. ESS was higher in PD as a group, compared with controls, and most of the increase could be accounted for by levodopa use, DA use, and PD severity. There was no significant difference between the DA, when controlling for levodopa dose, but there was a trend toward higher total ESS for those using pramipexole (12.2) and pergolide (12.8) compared with ropinirole (10.6) or bromocriptine (8).

Hobson et al surveyed 638 PD patients, including 420 drivers. Patients taking different DA and levodopa, in monotherapy or in combination, were compared with respect to attacks of sudden onset of sleep. Forty-nine drivers (11.6%) reported such attacks. All medications and all combinations thereof could induce sudden onset of sleep, with the same risk in pramipexole users (13/143 or 9%) as in levodopa users (43/407 or 10.5%). No statistically significant difference could be shown between the different DA, although the frequency of sudden sleep attacks was nominally highest in the ropinirole group (24/90 or 26%) (CitationHobson et al 2002).

In addition, in a review article from the same year, Homann and colleagues identified 20 publications reporting sleep events in 124 PD patients, including 32 on pramipexole, 84 on other DA, and 8 on levodopa monotherapy. Ninety six sleep attacks were identified. Overall, 6.6% of PD patients taking DA reported sleep events (range 0%–30%), without any difference among the different medications, making sleep events a class effect of all dopaminergic drugs. The frequency of road accidents in PD patients was not higher than in age matched controls, whether or not they were taking dopamine medication (CitationHomann et al 2002).

Paus et al investigated 2592 PD patients, 284 of which were on a combination of pramipexole and levodopa therapy. Sudden sleep attacks were reported by 177 patients (6%) on phone interviews. The frequency of sleep attacks was lowest for untreated PD patients (1.4%), higher for levodopa monotherapy (2.9%), DA monotherapy (5.3%), combination therapy levodopa plus one DA (7.3%), and it was highest for levodopa plus multiple DA therapy (9.2%). For pramipexole plus levodopa therapy, the frequency of sleep attacks without warning signs was 5.6% (95% CI 4.3–7.0). There was no significant difference in this regard between pramipexole and the other DA (CitationPaus et al 2003).

Similar results came from a study from 2005 on 929 patients, 39% of which used pramipexole, 18.5% ropinirole, 20% levodopa alone, and the rest a combination of different antiparkinsonian drugs. After controlling for all available patient characteristics, there was a significant association between sudden uncontrollable somnolence and pramipexole therapy vs levodopa monotherapy (OR 2.22. 95% CI 1.43–3.43). There was no significant difference in this regard between the other DA tested (ropinirole, pergolide, bromocriptine) and pramipexole. There was also a dose-response relationship for pramipexole, with high doses resulting in a higher risk for somnolence, compared with small to medium doses (OR 2.79 vs 2.08 respectively) (CitationAvorn et al 2005).

Romigi et al investigated one single PD patient with polysomnography. They found an increase in both the diurnal and the nocturnal sleep under pramipexole plus levodopa vs cabergoline plus levodopa therapy. The ESS scorings showed mild sleepiness on pramipexole and levodopa therapy, but were normal on cabergoline and levodopa and on levodopa alone. The patient did experience sleep episodes and irresistible onset of daytime sleep, but no sleep attacks, and no sudden onset of daytime sleep, while on pramipexole and L-dopa therapy but not on other drug combinations. The authors conclude that there may exist an individual susceptibility to specific antiparkinsonian drugs with respect to sleepiness (CitationRomigi et al 2005).

Razmy et al conducted a polysomnography study in PD patients treated with pramipexole, ropinirole, bromocriptine or pergolide, finding that the total dopaminergic drug dose given, rather than the specific DA used, was the best predictor of daytime sleepiness (CitationRazmy et al 2004).

In conclusion, the available evidence shows that somnolence, while being part of PD itself, can be induced by all dopaminergic drugs, DA as well as levodopa itself. In addition, many other factors other than medication play an important role in this regard. Increased somnolence may be a risk factor for attacks with sudden onset of sleep, but sleep attacks can also occur without a prior increase in somnolence. The risk for increased somnolence is higher with DA, including pramipexole, compared with levodopa, but it seems to be similar in all DA, ergot, and non-ergot. Combination therapies increase the risk which is directly proportional with the total dose of dopaminergic equivalents. In the light of these findings, disabling somnolence can be reduced with monotherapy, preferably with levodopa only, by reducing the total dopaminergic doses, by improving the night sleep, and by finding alternative therapies. In the particular patient, switch to another DA may be attempted, as the susceptibility to particular DA may differ between patients. Modafinil treatment may be tried in difficult cases although the evidence supporting the use of modafinil in PD is conflicting (CitationNieves and Lang 2002; CitationAdler et al 2003; CitationOndo et al 2005). Counseling is important for all patients treated with dopaminergic drugs, including pramipexole, and should include advice on night sleep hygiene, avoidance of sedatives and alcohol, normalizing diurnal rhythms, pulling over to stop if a wave of sleepiness is perceived. PD patients should abstain altogether from driving and other activities which require intense alertness, when suffering from generalized drowsiness, and if falling asleep during inactivity, regardless of their antiparkinsonian medication (CitationHolloway et al 2004).

Cardiac valvulopathy

The recent finding that pergolide and cabergoline can cause cardiac valvulopathies led to a wave or research pertaining to valvular abnormalities in patients treated with non-ergot DA as well.

Peralta et al investigated 75 PD patients and controls with echocardiography. Twenty-five of the patients were treated with pramipexole. While the exposure to pergolide and cabergoline was associated with higher frequencies of valvular regurgitation compared to controls, that was not the case for the pramipexole treated patients, which showed about the same risk as controls, around 10% (CitationPeralta et al 2006).

One study from Japan comprising 16 pramipexole treated PD patients found a higher frequency of insignificant forms of valvulopathy in the pramipexole group (25%) compared with the control group (17.6%), although the difference was not statistically significant (OR 1.62 [0.45–5.87]). The authors point out that a similar frequency of valvulopathy was found in a normal-population study (CitationYamamoto et al 2006).

Zanettini et al included in their study 36 PD patients treated with pramipexole, none of which showed any clinical important valvular regurgitation (CitationZanettini et al 2007).

Junghanns et al evaluated 85 PD patients treated with ergot and non-ergot DA (23 on pramipexole) together with 38 age-matched controls. The frequencies of valvular heart disease were similar in the control group (37%) and the pramipexole group (26%) (CitationJunghanns et al 2007).

In a large British case-study involving 11417 people who were prescribed drugs for PD, there were 31 patients with cardiac valve regurgitation, but none of them had been treated with pramipexole (CitationSchade et al 2007).

Dewey et al included 21 pramipexole patients in their study comparing ergot (pergolide) and non-ergot DA (ropinirole and pramipexole) with respect to regurgitation. The conclusion was that the risk of cardiac valve regurgitation appeared to be low when using non – ergot-derived DA, much lower than for pergolide (CitationDewey et al 2007).

Thus, pramipexole does not seem to cause valvular abnormalities. These results are not unexpected as the cause of valvular fibrosis appears to be fibroblast activation mediated through the 5-HT2B serotonergic receptors (CitationRothman et al 2000). While cabergoline and pergolide are strong agonists at this receptor, pramipexole’s affinity for the 5-HT2B receptor is low, which explains its low risk for cardiac valvulopathy, comparable with that seen in controls.

Still, Chaudhuri et al reported in 2004 in a letter to the Editors of the journal Movement Disorders that they were “aware of pramipexole being implicated as well, at least in one case” in respect with “fibrotic reaction with non-ergot agonists” (CitationChaudhuri et al 2004). As no further information is provided, this remark adds nothing to this discussion, and may even be confusing. However, it is included here for the sake of comprehensiveness.

In conclusion, although the evidence concerning pramipexole and valvulopathy is reassuring so far, continued pharmacovigilance is advisable.

Impulse control disturbances

Recently, pramipexole and other DA have been reported to cause impulse control disorders (ICD) such as gambling, compulsive buying, and pathological hypersexuality.

Dodd et al investigated 11 PD patients who had developed pathological gambling and other behavioral disorders. Eighty-two percent of these were treated with pramipexole, discontinuation of which led to improvement. In a literature survey, the authors found 17 similar case reports, 10 (59%) of which were also treated with pramipexole, while the rest were taking other DA (CitationDodd et al 2005).

Klos et al reported on pathological hypersexuality in 13 PD patients, 6 of which were treated with pramipexole, and in 2 multiple-system atrophy (MSA) patients, 1 of whom was treated with pramipexole (CitationKlos et al 2005). Several of these patients had other obsessive-compulsive behaviors in addition to hypersexuality. The conditions resolved after stopping pramipexole and/or adding antipsychotics. The authors identified 14 additional cases in the literature, all treated with levodopa plus DA, although the agonists were others than pramipexole. They suggest that excessive stimulation of the D2 receptor class and specifically the D3 receptor subclass may have caused this behavior.

Weintraub et al investigated the prevalence of ICD in 272 PD patients, half of which were treated with a DA (pramipexole, ropinirole, pergolide) (CitationWeintraub et al 2006). Eighteen patients (6.6%) met criteria for ICD at some point during their disease course and all of these were taking a DA while symptomatic. Pramipexole was the most frequently prescribed DA, representing more than half of all DA use. There was no difference between the three agonists in respect to their association with ICD and the authors concluded that the risk associated with ICD was a class effect of the DA and not specific to a certain agonist. The main risk for developing ICD in the context of DA treatment was a history of ICD symptoms prior to PD. A long term follow-up of 15 of these patients (mean time period 29 months) showed that 80% of them had discontinued or significantly reduced the DA treatment, and all these patients went into full or partial remission. However, most of these patients had increased the levodopa dosage, and the total levodopa equivalent daily dosage, as well as the UPDRS motor score, were similar at follow up. Of the three patients who had the same DA treatment at follow up, one was taking pramipexole. His compulsive sexuality had gone into full remission after DBS surgery and decreased amantadine dose (CitationMamikonyan et al 2008).

There is one case report of pathological gambling associated with pramipexole in a patient with restless legs syndrome (CitationQuickfall and Suchowersky 2007).

In conclusion, according to several reports, there is a significant risk for developing ICD in patients treated with pramipexole and other DA. This risk seems to be a class effect and is not specific to pramipexole alone. In North America and also in many European countries, pramipexole is the most used DA, implying that more patients are exposed to it compared with the other agonists. This may lead to more reports on gambling and other behavioral disorders during pramipexole treatment, although the risk may be the same with other DA (CitationLu et al 2006).

Hallucinations

In studies on early PD, hallucinations occurred in 9% of pramipexole-treated patients, compared with 2.6% of patients on placebo. The same pattern was seen in advanced PD, with almost 17% of pramipexole treated patients developing hallucinations, compared with 3.8% in the placebo group. The development of hallucinations was one of the most common causes of study termination in patients with advanced PD (2.7% on pramipexole vs 0.4% on placebo) (CitationPinter et al 1999; CitationShannon et al 1997; CitationParkinson Study Group 2000; CitationMizuno et al 2003; CitationMoller et al 2005). A meta-analysis found that, compared with placebo, pramipexole was associated with a higher risk for hallucinations than ropinirole (RR 5.2 [1.97–13.72] vs 2.75 [0.55–13.73]) (CitationEtminan et al 2003).

The 4-year results of the CALM-PD study showed that while hallucinations were reported to be more common in the pramipexole group vs the levodopa group after 2 years, there was no significant difference at 4 years (CitationParkinson Study Group 2000; CitationHolloway et al 2004;). The risk for developing hallucinations was higher in younger patients initially treated with pramipexole, compared with younger patients initially treated with levodopa, but this was not seen in the older cohort. Cognitive disturbances, older age and greater comorbid illness were associated with more hallucinations (CitationBiglan et al 2007).

In conclusion, there is an increased risk for hallucinations in PD patients treated with pramipexole.

Edema

Edema is a known adverse event associated with pramipexole treatment (CitationShannon et al 1997).

Tan and Ondo reported on 15 PD patients and 2 RLS patients who developed pedal edema, out of 300 patients treated with pramipexole. Edema abated after pramipexole discontinuation but it returned with reintroduction of pramipexole. Its intensity was dose dependent. No predisposing factors could be identified (CitationTan and Ondo 2000).

The 4-year results of the CALM-PD study showed that edema and cellulitis were later phenomena, reported more often in the pramipexole group. Five patients (3.3%) in the pramipexole group withdrew because of edema, but none in the levodopa group (CitationHolloway et al 2004).

In a retrospective medical record review, Kleiner-Fisman et al found a 7.7% (95% CI, 4.5%–12.9%) risk for development of pedal edema in the first year after initiation of pramipexole therapy in 237 PD patients, with more rapid development of edema among those with a history of coronary artery disease. In total, 16% of all patients developed pedal edema, about the same prevalence as reported from the CALM-PD study (14.6%) (CitationParkinson Study Group 2000). There was no relationship between dose of pramipexole and incidence and severity of pedal edema, and the edema resolved with withdrawal of pramipexole in 71% of cases (CitationKleiner-Fisman and Fisman 2007).

In a secondary analysis of the CALM-PD trial, Biglan et al showed that initial pramipexole treatment in patients with early PD therapy was associated with development of edema (HR 3.18, 95% CI 1.95–5.18, p < 0.0001), together with female gender and comorbid cardiac disease. Edema occurred mostly after 2 years of treatment, and not initially, which may explain why previous studies did not report edema as a common complication (CitationBiglan et al 2007).

In conclusion, pedal edema is a relatively common outcome in patients with PD receiving pramipexole, with a higher risk in patients with a history of cardiac disease.

Conclusion: Pramipexole’s place in the therapy of Parkinson’s disease

Pramipexole is generally well tolerated and efficacious in treating motor symptoms in both early and advanced PD. Its efficacy and tolerability are similar in African Americans, Asians, Caucasians and Hispanics. As initial treatment it may delay the onset of dyskinesias by delaying the start of levodopa therapy. It appears to have beneficial effects on non-motor symptoms such as depression, and it may improve treatment-resistant tremor, anhedonia and weight loss. Pramipexole seems to have neuroprotective effects in vitro and in animal models but these findings have not been replicated in humans. Compared with levodopa treatment, pramipexole therapy is associated with a higher risk for developing hallucinations, somnolence (and indirectly attacks of sudden onset of sleep), impulse control disturbances, peripheral edema, but not cardiac valvular disease. The risk for adverse events increases with patient age, pramipexole dose, severity of PD, concomitant combination therapy, and comorbidity, especially cardio-vascular. Continued pharmacovigilance in respect to adverse events, especially increased somnolence, cardiac valvulopathy, and psychiatric effects is necessary. Although pramipexole therapy is more costly than levodopa, the cost effectiveness increases over time due to a higher gain in QoL. More research is needed regarding pramipexole’s neuroprotective potential and its impact on cognition, apathy, fatigue, QoL, and treatment costs in PD. Pramipexole seems especially well suited for younger PD patients (<65 years), without cognitive disturbances, with low comorbidity, at high risk for developing dyskinesias, and with particular PD features shown to be responsive to pramipexole.

Limitations

The literature search was limited to Medline. Although a lot of effort was made for identifying all publications related to every topic, there is always the risk that some publications have not been found. No publication judged to be relevant for this topic has been deliberately omitted. offers a summary of the most important studies used for this review. No government agencies monitoring adverse events or pharmaceutical companies were contacted. This may bias the selection of publications.

Table 1 Summary of the most important studies used for this review

Disclosures

Radu Constantinescu is a member of Boehringer-Ingelheim’s Advisory Board in Sweden. This review was written without any financial support from Boehringer-Ingelheim or elsewhere. Between the end of 2005 and June 2006, Radu Constantinescu participated in the preparation of the “Efficacy of Pramipexole Taken Twice a Day in People with Early Parkinson’s Disease” study.

References

  • AarslandDLarsenJPLimNGRange of neuropsychiatric disturbances in patients with Parkinson’s diseaseJ Neurol Neurosurg Psychiatry199967492610486397
  • AarslandDZaccaiJBrayneCA systematic review of prevalence studies of dementia in Parkinson’s diseaseMov Disord20052012556316041803
  • AdlerCHCavinessJNHentzJGRandomized trial of modafinil for treating subjective daytime sleepiness in patients with Parkinson’s diseaseMov Disord2003182879312621632
  • AhlskogJEMuenterMDFrequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literatureMov Disord2001164485811391738
  • AhlskogJEMuenterMDMaraganoreDMFluctuating Parkinson’s disease. Treatment with the long-acting dopamine agonist cabergolineArch Neurol1994511236417986180
  • AvornJSchneeweissSSudarskyLRSudden uncontrollable somnolence and medication use in Parkinson diseaseArch Neurol2005621242816087765
  • BennettJPJrPierceyMFPramipexole – a new dopamine agonist for the treatment of Parkinson’s diseaseJ Neurol Sci1999163253110223406
  • BentonAHHamsherKMultilingual aphasia examination manual1989Iowa City, IAUniversity of Iowa
  • BiglanKMHollowayRGJrMcDermottMPRisk factors for somnolence, edema, and hallucinations in early Parkinson diseaseNeurology2007691879517620552
  • BouthenetMLSouilEMartresMPLocalization of dopamine D3 receptor mRNA in the rat brain using in situ hybridization histochemistry: comparison with dopamine D2 receptor mRNABrain Res1991564203191839781
  • BrooksDJAbbottRJLeesAJA placebo-controlled evaluation of ropinirole, a novel D2 agonist, as sole dopaminergic therapy in Parkinson’s diseaseClin Neuropharmacol19982110179579296
  • BrusaLBassiAStefaniAPramipexole in comparison to l-dopa: a neuropsychological studyJ Neural Transm20031103738012658365
  • CarveyPMMcGuireSOLingZDNeuroprotective effects of D3 dopamine receptor agonistsParkinsonism Relat Disord2001721322311331189
  • CassarinoDSFallCPSmithTSPramipexole reduces reactive oxygen species production in vivo and in vitro and inhibits the mitochondrial permeability transition produced by the parkinsonian neurotoxin methylpyridinium ionJ Neurochem1998712953019648878
  • ChaudhuriKRDhawanVBasuSValvular heart disease and fibrotic reactions may be related to ergot dopamine agonists, but non-ergot agonists may also not be sparedMov Disord2004191522315515009
  • ChenHZhangSMHernanMAWeight loss in Parkinson’s diseaseAnn Neurol200353676912731005
  • ConstantinescuRRomerMMcDermottMPImpact of pramipexole on the onset of levodopa-related dyskinesiasMov Disord2007221317917534955
  • CorriganMHDenahanAQWrightCEComparison of pramipexole, fluoxetine, and placebo in patients with major depressionDepress Anxiety200011586510812530
  • CotziasGCPapavasiliouPSTolosaEAporphines in Parkinson’s diseaseTrans Am Neurol Assoc1975100178811226602
  • DeweyRB2ndReimoldSCO’SuilleabhainPECardiac valve regurgitation with pergolide compared with nonergot agonists in Parkinson diseaseArch Neurol2007643778017353380
  • DoddMLKlosKJBowerJHPathological gambling caused by drugs used to treat Parkinson diseaseArch Neurol20056213778116009751
  • DooneiefGMirabelloEBellKAn estimate of the incidence of depression in idiopathic Parkinson’s diseaseArch Neurol19924930571536634
  • EtminanMGillSSamiiAComparison of the risk of adverse events with pramipexole and ropinirole in patients with Parkinson’s disease: a meta-analysisDrug Saf2003264394412688834
  • EtminanMSamiiATakkoucheBIncreased risk of somnolence with the new dopamine agonists in patients with Parkinson’s disease: a meta-analysis of randomised controlled trialsDrug Saf200124863811665873
  • FantiniMLGagnonJFFilipiniDThe effects of pramipexole in REM sleep behavior disorderNeurology20036114182014638967
  • FruchtSRogersJDGreenePEFalling asleep at the wheel: motor vehicle mishaps in persons taking pramipexole and ropiniroleNeurology19995219081010371546
  • GoetzCGBlasucciLStebbinsGTSwitching dopamine agonists in advanced Parkinson’s disease: is rapid titration preferable to slow?Neurology1999521227910214748
  • GoldbergJFBurdickKEEndickCJPreliminary randomized, double-blind, placebo-controlled trial of pramipexole added to mood stabilizers for treatment-resistant bipolar depressionAm J Psychiatry2004161564614992985
  • GuMIravaniMMCooperJMPramipexole protects against apoptotic cell death by non-dopaminergic mechanismsJ Neurochem20049110758115569251
  • GuttmanMDouble-blind comparison of pramipexole and bromocriptine treatment with placebo in advanced Parkinson’s disease. International Pramipexole-Bromocriptine Study GroupNeurology199749106059339690
  • GuttmanMJaskolkaJThe use of pramipexole in Parkinson’s disease: are its actions D(3) mediated?Parkinsonism Relat Disord20017231411331191
  • GuttmanMStewartDHusseyDInfluence of L-dopa and pramipexole on striatal dopamine transporter in early PDNeurology20015615596411402115
  • HallEDAndrusPKOostveenJANeuroprotective effects of the dopamine D2/D3 agonist pramipexole against postischemic or methamphetamine-induced degeneration of nigrostriatal neuronsBrain Res19967428089117424
  • HamiltonMA rating scale for depressionJ Neurol Neurosurg Psychiatry196023566214399272
  • HannaPARatkosLOndoWGSwitching from pergolide to pramipexole in patients with Parkinson’s diseaseJ Neural Transm2001108637011261747
  • HappeSBergerKThe association of dopamine agonists with daytime sleepiness, sleep problems and quality of life in patients with Parkinson’s disease – a prospective studyJ Neurol20012481062712013583
  • HauserRAGaugerLAndersonWMPramipexole-induced somnolence and episodes of daytime sleepMov Disord2000156586310928575
  • HauserRAMcDermottMPMessingSFactors associated with the development of motor fluctuations and dyskinesias in Parkinson diseaseArch Neurol20066317566017172616
  • HobsonDELangAEMartinWRExcessive daytime sleepiness and sudden-onset sleep in Parkinson disease: a survey by the Canadian Movement Disorders GroupJAMA20022874556311798367
  • HoehnMMYahrMDParkinsonism: onset, progression and mortalityNeurology196717427426067254
  • HollowayRGShoulsonIFahnSPramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trialArch Neurol20046110445315262734
  • HolmanAJMyersRRA randomized, double-blind, placebo-controlled trial of pramipexole, a dopamine agonist, in patients with fibromyalgia receiving concomitant medicationsArthritis Rheum200552249550516052595
  • HomannCNWenzelKSuppanKSleep attacks in patients taking dopamine agonists: reviewBMJ20023241483712077032
  • HäselbarthVFJustus-ObenauerHPeilHPharmacokinectics and bioavailability of pramipexole: Comparison of plasma levels after intravenous and oral administration in healthy volunteers (M/2730/0029)Upjohn Technical Report1994a 7215–94–016
  • HäselbarthVKLohmannJJustus-ObenauerHPharmacokinetics and metabolism of [14c]pramipexole after single intravenous and oral doses in healthy volunteers (Protocol M/2730/0030)Upjohn Technical Report1994b 7215–94–014
  • InzelbergRCarassoRLSchechtmanEA comparison of dopamine agonists and catechol-O-methyltransferase inhibitors in Parkinson’s diseaseClin Neuropharmacol200023262611154093
  • InzelbergRSchechtmanENisipeanuPCabergoline, pramipexole and ropinirole used as monotherapy in early Parkinson’s disease: an evidence-based comparisonDrugs Aging2003208475512964891
  • IzumiYSawadaHYamamotoNNovel neuroprotective mechanisms of pramipexole, an anti-Parkinson drug, against endogenous dopamine-mediated excitotoxicityEur J Pharmacol20075571324017161393
  • JohnsMWA new method for measuring daytime sleepiness: the Epworth sleepiness scaleSleep19911454051798888
  • JoyceJNWoolseyCRyooHLow dose pramipexole is neuroprotective in the MPTP mouse model of Parkinson’s disease, and downregulates the dopamine transporter via the D3 receptorBMC Biol200422215473914
  • JunghannsSFuhrmannJTSimonisGValvular heart disease in Parkinson’s disease patients treated with dopamine agonists: a reader-blinded monocenter echocardiography studyMov Disord200722234817094087
  • KarlsenKHTandbergEArslandDHealth related quality of life in Parkinson’s disease: a prospective longitudinal studyJ Neurol Neurosurg Psychiatry200069584911032608
  • KartzinelRTeychennePGillespieMMBromocriptine and levodopa (with or without carbidopa) in parkinsonismLancet19762272559850
  • Kleiner-FismanGFismanDNRisk factors for the development of pedal edema in patients using pramipexoleArch Neurol200764820417420306
  • KlosKJBowerJHJosephsKAPathological hypersexuality predominantly linked to adjuvant dopamine agonist therapy in Parkinson’s disease and multiple system atrophyParkinsonism Relat Disord200511381616109498
  • KompolitiKAdlerCHRamanRGender and pramipexole effects on levodopa pharmacokinetics and pharmacodynamicsNeurology20025814182212011296
  • KumruHSantamariaJValldeoriolaFIncrease in body weight after pramipexole treatment in Parkinson’s diseaseMov Disord2006211972416972276
  • LangAFMunsatTLAssessment of Parkinson’s diseaseQuantification of Neurologic Deficit1989Boston, MassButterworth-Heinemann285309
  • LattanziLDell’OssoLCassanoPPramipexole in treatment-resistant depression: a 16-week naturalistic studyBipolar Disord200243071412479663
  • LeWDJankovicJXieWAntioxidant property of pramipexole independent of dopamine receptor activation in neuroprotectionJ Neural Transm200010711657311129106
  • LemkeMRBrechtHMKoesterJAnhedonia, depression, and motor functioning in Parkinson’s disease during treatment with pramipexoleJ Neuropsychiatry Clin Neurosci2005172142015939976
  • LemkeMRBrechtHMKoesterJEffects of the dopamine agonist pramipexole on depression, anhedonia and motor functioning in Parkinson’s diseaseJ Neurol Sci20062482667016814808
  • LibmanIGawelMJRiopelleRJA comparison of bromocriptine (Parlodel) and levodopa-carbidopa (Sinemet) for treatment of “de novo” Parkinson’s disease patientsCan J Neurol Sci198714576803319120
  • LiebermanARanhoskyAKortsDClinical evaluation of pramipexole in advanced Parkinson’s disease: results of a double-blind, placebo-controlled, parallel-group studyNeurology19974916289222185
  • LorefaltBGranerusAKUnossonMAvoidance of solid food in weight losing older patients with Parkinson’s diseaseJ Clin Nurs20061514041217038101
  • LuCBharmalASuchowerskyOGambling and Parkinson diseaseArch Neurol20066329816476827
  • MamikonyanESiderowfADDudaJELong-term follow-up of impulse control disorders in Parkinson’s diseaseMov Disord200823758017960796
  • MierauJPramipexole: A dopamine-receptor agonist for treatment of Parkinson’s DiseaseClin Neuropharmacol199518Suppl 1S195S206
  • MierauJSchingnitzGBiochemical and pharmacological studies on pramipexole, a potent and selective dopamine D2 receptor agonistEur J Pharmacol1992215161701356788
  • MierauJSchneiderFJEnsingerHAPramipexole binding and activation of cloned and expressed dopamine D2, D3 and D4 receptorsEur J Pharmacol199529029367664822
  • MizunoYYanagisawaNKunoSRandomized, double-blind study of pramipexole with placebo and bromocriptine in advanced Parkinson’s diseaseMov Disord20031811495614534919
  • MollerJCOertelWHKosterJLong-term efficacy and safety of pramipexole in advanced Parkinson’s disease: results from a European multicenter trialMov Disord2005206021015726540
  • MontgomerySAAsbergMA new depression scale designed to be sensitive to changeBr J Psychiatry19791343829444788
  • MontplaisirJNicolasADenesleRRestless legs syndrome improved by pramipexole: a double-blind randomized trialNeurology1999529384310102409
  • NavanPFindleyLJJeffsJADouble-blind, single-dose, cross-over study of the effects of pramipexole, pergolide, and placebo on rest tremor and UPDRS part III in Parkinson’s diseaseMov Disord2003a181768012539211
  • NavanPFindleyLJJeffsJARandomized, double-blind, 3-month parallel study of the effects of pramipexole, pergolide, and placebo on Parkinsonian tremorMov Disord2003b1813243114639675
  • NievesAVLangAETreatment of excessive daytime sleepiness in patients with Parkinson’s disease with modafinilClin Neuropharmacol200225111411981239
  • NoyesKDickAWHollowayRGPramipexole v. levodopa as initial treatment for Parkinson’s disease: a randomized clinical-economic trialMed Decis Making2004244728515358996
  • NoyesKDickAWHollowayRGPramipexole and levodopa in early Parkinson’s disease: dynamic changes in cost effectivenessPharmacoeconomics20052312577016336019
  • NoyesKDickAWHollowayRGPramipexole versus levodopa in patients with early Parkinson’s disease: effect on generic and disease-specific quality of lifeValue Health20069283816441522
  • O’SuilleabhainPEDeweyRBJrContributions of dopaminergic drugs and disease severity to daytime sleepiness in Parkinson diseaseArch Neurol200259986912056935
  • OndoWGFayleRAtassiFModafinil for daytime somnolence in Parkinson’s disease: double blind, placebo controlled parallel trialJ Neurol Neurosurg Psychiatry2005761636916291885
  • PanTXieWJankovicJBiological effects of pramipexole on dopaminergic neuron-associated genes: relevance to neuroprotectionNeurosci Lett2005377106915740846
  • Parkinson Study GroupSafety and efficacy of pramipexole in early Parkinson disease. A randomized dose-ranging study. Parkinson Study GroupJAMA1997278125309214527
  • Parkinson Study GroupPramipexole vs levodopa as initial treatment for Parkinson disease: A randomized controlled trial. Parkinson Study GroupJAMA20002841931811035889
  • Parkinson Study GroupDopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progressionJAMA200228716536111926889
  • Parkinson Study GroupPramipexole in levodopa-treated Parkinson disease patients of African, Asian, and Hispanic heritageClin Neuropharmacol200730728517414939
  • PausSBrechtHMKosterJSleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s diseaseMov Disord2003186596712784269
  • PeraltaCWolfEAlberHValvular heart disease in Parkinson’s disease vs. controls: An echocardiographic studyMov Disord20062111091316622856
  • PinterMMPogarellOOertelWHEfficacy, safety, and tolerance of the non-ergoline dopamine agonist pramipexole in the treatment of advanced Parkinson’s disease: a double blind, placebo controlled, randomised, multicentre studyJ Neurol Neurosurg Psychiatry1999664364110201413
  • PinterMMRutgersAWHebenstreitEAn open-label, multicentre clinical trial to determine the levodopa dose-sparing capacity of pramipexole in patients with idiopathic Parkinson’s diseaseJ Neural Transm200010713072311145006
  • PizzagalliDAEvinsAESchetterECSingle dose of a dopamine agonist impairs reinforcement learning in humans: Behavioral evidence from a laboratory-based measure of reward responsivenessPsychopharmacology, (Berl)20081962213217909750
  • PluckGCBrownRGApathy in Parkinson’s diseaseJ Neurol Neurosurg Psychiatry2002736364212438462
  • PoeweWHoglBAkathisia, restless legs and periodic limb movements in sleep in Parkinson’s diseaseNeurology200463S12615505135
  • PoeweWWenningGKApomorphine: an underutilized therapy for Parkinson’s diseaseMov Disord2000157899411009181
  • PoeweWHRascolOQuinnNEfficacy of pramipexole and transdermal rotigotine in advanced Parkinson’s disease: a double-blind, double-dummy, randomised controlled trialLancet Neurol200765132017509486
  • PogarellOGasserTvan HiltenJJPramipexole in patients with Parkinson’s disease and marked drug resistant tremor: a randomised, double blind, placebo controlled multicentre studyJ Neurol Neurosurg Psychiatry2002727132012023411
  • PresgravesSPBorwegeSMillanMJInvolvement of dopamine D(2)/D(3) receptors and BDNF in the neuroprotective effects of S32504 and pramipexole against 1-methyl-4-phenylpyridinium in terminally differentiated SH-SY5Y cellsExp Neurol20041901577015473989
  • QuickfallJSuchowerskyOPathological gambling associated with dopamine agonist use in restless legs syndromeParkinsonism Relat Disord200713535617270485
  • RavinaBEidelbergDAhlskogJEAlbinRLBrooksDJCarbonMThe role of radiotracer imaging in Parkinson diseaseNeurology2005642081515668415
  • RazmyALangAEShapiroCMPredictors of impaired daytime sleep and wakefulness in patients with Parkinson disease treated with older (ergot) vs newer (nonergot) dopamine agonistsArch Neurol2004619710214732626
  • ReichmannHBrechtMHKosterJPramipexole in routine clinical practice: a prospective observational trial in Parkinson’s diseaseCNS Drugs2003179657314533946
  • ReichmannHOdinPBrechtHMChanging dopamine agonist treatment in Parkinson’s disease: experiences with switching to pramipexoleJ Neural Transm Suppl2006172517447411
  • ReitanRTrail making testManual for administration and scoring1992South TusconReitan Neuropsychology Laboratory
  • RektorovaIRektorIBaresMPramipexole and pergolide in the treatment of depression in Parkinson’s disease: a national multicentre prospective randomized studyEur J Neurol20031039940612823492
  • RektorovaIRektorIBaresMCognitive performance in people with Parkinson’s disease and mild or moderate depression: effects of dopamine agonists in an add-on to L-dopa therapyEur J Neurol20051291515613141
  • ReljaMKlepacNA dopamine agonist, pramipexole, and cognitive functions in Parkinson’s diseaseJ Neurol Sci2006248251416797593
  • RinneUKBraccoFChouzaCCabergoline 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 Collaborative Study GroupNeurology19974836389040722
  • RomigiABrusaLMarcianiMGSleep episodes and daytime somnolence as result of individual susceptibility to different dopaminergic drugs in a PD patient: a polysomnographic studyJ Neurol Sci200522871015607203
  • RothmanRBBaumannMHSavageJEEvidence for possible involvement of 5-HT(2B) receptors in the cardiac valvulopathy associated with fenfluramine and other serotonergic medicationsCirculation200010228364111104741
  • SayeedIParvezSWinkler-StuckKPatch clamp reveals powerful blockade of the mitochondrial permeability transition pore by the D2-receptor agonist pramipexoleFASEB J200620556816407457
  • SchadeRAndersohnFSuissaSDopamine agonists and the risk of cardiac-valve regurgitationN Engl J Med2007356293817202453
  • SchifittoGFOakesDShulmanLMR Fatigue in levodopa-naïve subjects with early Parkinson’s disease, abstract at the First World Parkinson Congress2006Washington DC, USA
  • SchmidtMHKoshalVBSchmidtHSUse of pramipexole in REM sleep behavior disorder: results from a case seriesSleep Med200674182316815751
  • SchragAJahanshahiMQuinnNWhat contributes to quality of life in patients with Parkinson’s disease?J Neurol Neurosurg Psychiatry2000693081210945804
  • ScoyniRMAielloLTraniIDrug adverse events and drop-out risk: a clinical caseArch Gerontol Geriatr200744Suppl 13596417317475
  • ShannonKMBennettJPJrFriedmanJHEfficacy of pramipexole, a novel dopamine agonist, as monotherapy in mild to moderate Parkinson’s disease. The Pramipexole Study GroupNeurology19974972489305331
  • ShulmanLMMinagarARabinsteinAThe use of dopamine agonists in very elderly patients with Parkinson’s diseaseMov Disord200015664810928576
  • SnaithRPHamiltonMMorleySA scale for the assessment of hedonic tone the Snaith-Hamilton Pleasure ScaleBr J Psychiatry1995167991037551619
  • StarksteinSEMaybergHSPreziosiTJReliability, validity, and clinical correlates of apathy in Parkinson’s diseaseJ Neuropsychiatry Clin Neurosci1992413491627973
  • SvenssonKCarlssonAHuffRMBehavioral and neurochemical data suggest functional differences between dopamine D2 and D3 receptorsEur J Pharmacol1994263235437843260
  • TanEKOndoWClinical characteristics of pramipexole-induced peripheral edemaArch Neurol2000577293210815140
  • TandbergELarsenJPAarslandDThe occurrence of depression in Parkinson’s disease. A community-based studyArch Neurol19965317598639068
  • TandbergELarsenJPKarlsenKA community-based study of sleep disorders in patients with Parkinson’s diseaseMov Disord19981389599827612
  • TothMJFishmanPSPoehlmanETFree-living daily energy expenditure in patients with Parkinson’s diseaseNeurology19974888919008499
  • UbertiDBianchiIOlivariLPramipexole prevents neurotoxicity induced by oligomers of beta-amyloidEur J Pharmacol2007569194617572405
  • WeinerWJFactorSAJankovicJThe long-term safety and efficacy of pramipexole in advanced Parkinson’s diseaseParkinsonism Relat Disord200171152011248592
  • WeintraubDMoralesKHMobergPJAntidepressant studies in Parkinson’s disease: a review and meta-analysisMov Disord2005201161915954137
  • WeintraubDSiderowfADPotenzaMNAssociation of dopamine agonist use with impulse control disorders in Parkinson diseaseArch Neurol2006639697316831966
  • WongKSLuCSShanDEEfficacy, safety, and tolerability of pramipexole in untreated and levodopa-treated patients with Parkinson’s diseaseJ Neurol Sci200321681714607306
  • WrightCESissonTLIchhpuraniAKSteady-state pharmacokinetic properties of pramipexole in healthy volunteersJ Clin Pharmacol19973752059208359
  • WynaldaMAWienkersLCAssessment of potential interactions between dopamine receptor agonists and various human cytochrome P450 enzymes using a simple in vitro inhibition screenDrug Metab Dispos199725121149321526
  • YamamotoMUesugiTNakayamaTDopamine agonists and cardiac valvulopathy in Parkinson disease: a case-control studyNeurology2006671225917030757
  • ZanettiniRAntoniniAGattoGValvular heart disease and the use of dopamine agonists for Parkinson’s diseaseN Engl J Med2007356394617202454
  • ZouLJankovicJRoweDBNeuroprotection by pramipexole against dopamine- and levodopa-induced cytotoxicityLife Sci19996412758510227583
  • ZungWWA Self-Rating Depression ScaleArch Gen Psychiatry196512637014221692

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.