107
Views
4
CrossRef citations to date
0
Altmetric
Review

Role and clinical utility of pramipexole extended release in the treatment of early Parkinson’s disease

, &
Pages 83-88 | Published online: 15 Mar 2012

Abstract

The aim of this article is to provide a short review of the most relevant pharmacological and clinical data on pramipexole extended release (ER) as well as to address the clinical utility and potential advantages of a once-daily formulation especially in the treatment of early Parkinson’s disease (PD). Pramipexole is widely established as a symptomatic treatment in early as well as advanced PD. The development of an ER formulation, with stable pramipexole plasma concentration over 24 hours, now offers a bioequivalent once-daily alternative. Double-blind randomized controlled trials in early and advanced PD, have established noninferiority of pramipexole ER compared with immediate release as well as superiority of both formulations over placebo. The overnight switch from the standard to the once-daily formulation was shown to be successful in >80% of patients without requiring any dose adjustments. Potential benefits of the prolonged-release design, which have not yet been formally demonstrated in the pivotal trial program, include improved compliance and a potential for better symptomatic control, particularly in patients with early disease that can be managed with monotherapy.

Role of dopamine agonists in the treatment of Parkinson’s disease

Although levodopa (LD) still remains the gold standard for symptomatic efficacy in reducing the motor symptoms of Parkinson’s disease (PD),Citation1 its long-term use is associated with the development of potentially disabling motor complications, including response oscillations as well as drug-induced dyskinesias (LD-induced dyskinesias), affecting approximately 30% of patients after only 2 years of LD exposure.Citation2 The precise underlying pathophysiology of LD-induced motor complications is still incompletely understood; however, it is believed that pulsatile dopamine-receptor stimulation, leading to neuroplastic changes in the basal ganglia circuitry, plays a pivotal role.Citation1,Citation3Citation5 Dopamine agonists (DAs) act directly on striatal dopamine receptors, with preferential effects on the D2 over D1 subfamily, and generally have considerably longer half-lives than LD ().Citation6 While the first agents of this class were ergolinic compounds with affinities to multiple non-DA-receptor types, the newer agents are nonergolinic and seem to lack the risk for cardiovalvular fibrosis, recently reported for ergolinic agonists,Citation7 which have since been withdrawn from many markets. In clinical trials involving patients with early PD, initial monotherapy with DAs was consistently associated with a significantly reduced risk of motor complications compared with LD, over double-blind follow-up periods of up to 5 years (),Citation8Citation10 such that DAs are currently recommended as first-line therapies particularly for patients with younger age at onset.Citation11,Citation12 In addition, DAs are established first-line therapies to reduce motor fluctuations in LD-treated patients.Citation11,Citation12 Both types of clinical benefit from DAs are likely to be related to their longer half-life, resulting in more continuous striatal DA-receptor stimulation. Continuous 24-hour delivery with once-daily dosing has become possible with a transdermal formulation of the short half-life agonist rotigotine and, recently, novel extended-release (ER) formulations have been developed for the nonergolinic oral compounds ropinirole and pramipexole. The latter principle is likely to increase convenience and possibly adherence, particularly for patients with early disease that can be managed with monotherapy.

Table 1 Pharmacological properties of dopamine agonists

Table 2 Results of the main trials of levodopa versus dopamine agonists in early Parkinson’s disease

Pharmacology and pharmacokinetics of pramipexole ER

Pramipexole is a full intrinsic DA with a nonergot structure. The aminobenzothiazole compound is highly selective for the D2 receptor family and, intragroup, has preferential affinity for the D3 receptor subtype.Citation13 The agent lacks affinity for dopamine receptors D1 and D5 and displays only little affinity for D2 and D4 receptor subtypes.Citation14 Pramipexole ER was designed as a prolonged-release tablet with pramipexole dihydrochloride monohydrate dispersed homogeneously throughout the matrix. The active substance is released proportional to the square-root of timeCitation15 by two different mechanisms: diffusion and erosion,Citation15 reaching its maximum plasma concentration (Cmax) approximately 6 hours after oral administration (immediate release [IR]: Cmax 1–3 hours). In other respects the pharmacokinetic profile mostly coincides with the well-known IR formulation. Following oral administration, the agent shows a bioavailability of >90%.Citation15 In fact, pharmacokinetic studies observed an increase in Cmax with concomitant intake of a high-fat meal (24% after a single dose administration and about 20% after multiple dose administrations), but there was no significant change in overall area under the curve (AUC0–24 hours).Citation15,Citation16 Protein binding is <20% and the agent is metabolized only to a small extent and predominantly eliminated by renal excretion (~90%). With a renal clearance of approximately 400 mL/ min, the elimination half-life (t½) varies from 8 hours in the young to 12 hours in the elderly. Pramipexole ER is available in five dosage strengths (0.26 [0.375] mg, 0.52 [0.75] mg, 1.05 [1.5] mg, 2.1 [3] mg, and 3.15 [4.5] mg [all doses are expressed in terms of pramipexole base; corresponding dose strengths of pramipexole salt are given in brackets]) and is approved for use in early PD as well as adjunct therapy in advanced PD with motor complications.

Clinical efficacy in early PD

Two large randomized double-blind placebo-controlled Phase III trials were conducted to evaluate the clinical efficacy of pramipexole ER in early PD patients.Citation17,Citation18 These are summarized as follows.

Two hundred and fifty-nine patients with early PD at Hoehn and Yahr stage 1 to 3, diagnosed within the preceding 5 years, were randomized (2:2:1) to receive pramipexole ER (0.26–3.15 [0.375–4.5] mg qd), pramipexole IR (0.088–1.1 [0.125–1.5] mg tid), or placebo. Following a 7-week-flexible up-titration phase, drug doses were maintained for an additional 26 weeks, during which, to maximize patient retention in the trial, open-label LD rescue medication was permitted for subjects experiencing insufficient control of parkinsonism. Post-LD-rescue data were censored for the primary analysis. At 18 weeks an interim analysis on a subset of 253 patients was planned to evaluate superiority of efficacy of pramipexole ER to placebo on the Unified Parkinson’s Disease Rating Scale (UPDRS) II + III (primary endpoint).Citation18 At 33 weeks, the study aimed to demonstrate noninferiority of pramipexole ER to pramipexole IR in the combined score on UPDRS II + III (primary endpoint).Citation17 Noninferiority was predefined as a treatment-group difference for which the lower bound of the 95% confidence interval (CI) did not exceed −3 points. This margin had been chosen conservatively to be well outside the minimally clinically relevant difference on the UPDRS, which has been suggested to be −7 points for parts II and III combined.Citation19 At 18 weeks, the adjusted mean change in UPDRS II + III combined scores, censoring post-LD-rescue data, was −7.4 (1.1) in the pramipexole ER group, compared with −2.7 (1.3) in the placebo group (P = 0.0010 vs placebo) and −7.5 (1.1) in the pramipexole IR group (P = 0.0006 vs placebo).Citation18 Including data from subjects receiving LD rescue medication, the adjusted mean change was −5.1 (1.3) for placebo, −8.1 (1.1) for pramipexole ER (P = 0.0282 vs placebo), and −8.4 (1.1) for pramipexole IR (P = 0.0153 vs placebo).Citation18 Hence, using either approach, a statistically significant difference between pramipexole groups and placebo could be demonstrated from week 4 onward (ER P = 0.0111, IR P = 0.0042 vs placebo at week 4 by either approach).Citation18 At 33 weeks, UPDRS II + III change – censoring post-LD-rescue data – was −8.2 for ER and −8.7 for IR. The resultant treatment difference was −0.5 (95% CI: −2.3 to +1.3), thereby establishing noninferiority of the ER formulation. Including LD-rescue data, the adjusted mean decrease was −8.5 versus −9.4, revealing a difference of −0.9 (95% CI: −2.7, +0.9), which is still well within the predefined margin.Citation17 As already demonstrated at 18 weeks, the 33-week analysis confirmed the superiority of pramipexole ER over placebo (adjusted mean change in UPDRS II + III score: −8.2 ER vs −1.2 placebo [P < 0.0001]).Citation17 Consistent with the symptomatic efficacy of pramipexole, fewer patients on active treatment required LD-rescue medication (7.0% ER, 4.3% IR, 21.4% placebo).Citation17 Superiority of both pramipexole formulations could also be shown for secondary outcome measures, including Global Impression of Improvement responder rates (41.4% ER, 45.1% IR vs 20.6% placebo [P = 0.0003 and P < 0.0001]), Patient Global Impression of Improvement responder rates (34.4% ER, 32.4% IR vs 16.5% placebo [P = 0.0008 and P = 0.0020]), and UPDRS II + III responder rates (66.7% ER, 63.8% IR vs 35.0% placebo [P < 0.0001]).Citation17 A summary of the primary and key secondary endpoints at week 33 is given in .

Table 3 Efficacy results at week 33 (FAS/LOCF with levodopa rescue censored)

Switch study

Rascol et al conducted a randomized, double-blind, double-dummy, parallel-group study to assess efficacy, safety, and tolerability of an overnight switch from pramipexole IR to ER in early PD patients.Citation20 After a 2–4-week open-label run-in on pramipexole IR tid, 156 patients were switched overnight either to ER or to IR at an unchanged daily dosage (randomized 2:1). Subjects were allowed a one-step dose adjustment as required for efficacy and/or tolerability 4 and 5 weeks after switching. Dosage levels then had to remain stable for another 4 weeks. The primary efficacy endpoint was defined as the proportion of patients successfully switched (with or without any dosage adjustment) at the end of week 9, determined as no worsening from baseline UPDRS II + III score >15% and no withdrawal due to drug-related adverse events (AEs). Noninferiority was predefined as a 95% CI with a lower bound not exceeding −15%. At week 9, 84.5% of patients in the ER versus 94.2% in the IR group were considered successfully switched (). The absolute difference between groups was −9.76% (95% CI: −18.81%, +1.66%), hence noninferiority of pramipexole ER was not formally demonstrated. However, after 9 weeks, 80.6% of ER and 84.6% of IR recipients were successfully switched without requiring any dose adjustments. In the ER group, 16.5% had increased and 2.9% had decreased their dosage, while corresponding percentages in the IR group were 13.5% versus 1.9% (). The between-group difference for increased versus unchanged/decreased dosage was not significant (P = 0.6190). The mean pramipexole dosage at 9 weeks was 2.75 (±0.95) mg/day (+0.12 mg/day from baseline) in the ER, compared with 2.83 (±0.86) mg/day (+0.09 mg/day from baseline) in the IR group.

Table 4 Rates of successful switching after 9 weeks

Table 5 Dosage change from baseline to 9 weeks

Safety and tolerability

Pooled safety data of the clinical trials in early and advanced PD, comprising information from 803 PD patients exposed to clinically effective doses of pramipexole ER, show a slightly higher rate of AEs for both pramipexole IR and ER compared with placebo. However, no significant difference in AE profiles has been found between the two pramipexole formulations. A summary of the most common side effects is given in .Citation15 Safety evaluation of the 33-week trial in early PDCitation17 also identified somnolence, gastrointestinal complaints, and dizziness as the most frequent AEs. In this study, a small numerical increase in Epworth Sleepiness Scale score was observed in both pramipexole groups but mean values remained below the cut-off for excessive daytime sleepiness (≤10). Impulse control disorders were also slightly more common in both active treatment groups (four patients in the ER, three in the IR, and one in the placebo group). Taken as a whole, pramipexole ER showed the same safety and tolerability profile as pramipexole IR.

Table 6 Frequency of adverse events (%)

Patient-focused perspectives

Poor compliance has been identified as a major issue in several disease areasCitation21Citation24 including PD.Citation25,Citation26 The irregular intake of prescribed medication affects health care on many levels, including poor symptom control and reduced quality of life,Citation27 distortion of treatment effectivenessCitation28 as well as increased health care expenditure.Citation29,Citation30 The reasons for noncompliance are manifold and include fear of side effects, complex drug regimens incompatible with everyday life, and dementia. In one study, poorer compliance was identified to be more likely among younger patients and those with complex drug regimens, depression, and lower quality of life.Citation26 Grosset et al demonstrated an inverse correlation of compliance and drug doses per day in PD patientsCitation31 and showed that low therapy adherence was significantly associated with poor motor scores (UPDRS), more days absent from work, and worse mobility (PDQ39).Citation31 In this study, all measures of therapy adherence (total number of days adherent, timing adherence, and total therapy adherence) were significantly higher for once-daily medications, including DAs taken once versus three times daily.Citation31 Therefore, the availability of once-daily formulations of DAs with established efficacy in PD represents an advantage even if there is no added benefit of enhanced clinical efficacy or safety. In addition, from the patient perspective there is the obvious advantage in terms of convenience and ease of use, particularly in early disease where monotherapy with a single dose per day is possible. Due to blinding purposes, the possible beneficial effects of a once-daily formulation on convenience and adherence have not been possible to assess in the Phase III trials.Citation17,Citation18,Citation20,Citation32 ER formulations of a long-acting DA such as pramipexole may also contribute to improved symptom control during the night, although this has not been formally studied to date and is more relevant to advanced rather than early PD. Furthermore, there is a possibility that the pharmacokinetic profile of a slow-release formulation may reduce the risk of peripheral dopaminergic side effects such as nausea and vomiting as well as central adverse reactions, including somnolence and daytime sleepiness, by avoiding rapid plasma level increases and high peak concentrations as compared with their IR counterparts. This has, however, not been demonstrated in the pivotal clinical trials with pramipexole ER.

Conclusion

Pramipexole is widely established as a symptomatic treatment in early as well as advanced PD. The development of an ER formulation, with stable pramipexole plasma concentration over 24 hours, now offers a bioequivalent once-daily alternative. Double-blind randomized controlled trials in early as well as advanced PD have established the noninferiority of pramipexole ER compared with IR as well as the superiority of both formulations over placebo. The overnight switch from the standard to the once-daily formulation was shown to be successful in >80% of patients without requiring any dose adjustments. Potential benefits of prolonged-release once-daily DA formulations include improved compliance and a potential for better symptomatic control over the day as well as during the night. However, the latter, along with reduced risk of dopaminergic side effects, has not been formally demonstrated in the pivotal trial program. From a patient perspective there is little doubt that once-daily drugs offer major advantages in terms of convenience, especially for initial monotherapy in early PD.

Disclosure

Klaus Seppi has received honoraria for speaking and/or consulting from: Novartis, AstraZeneca, Boehringer Ingelheim, Lundbeck, Schwarz Pharma, UCB Pharma, Teva, and GlaxoSmithKline, and grants/research funding from: The Movement Disorders Society, Michael J Fox Foundation for Parkinson’s Research, Österreichische Nationalbank, Austrian Science Fund, and Medical University Innsbruck. Werner Poewe has received consultancy and lecture fees from: AstraZeneca, Teva, Novartis, GlaxoSmithKline, Boehringer-Ingelheim, UCB, Orion Pharma, and Merck Serono in relation to clinical drug development programs for PD, and grants/research funding from: The Movement Disorders Society, Michael J Fox Foundation for Parkinson’s Research, Österreichische Nationalbank, Austrian Science Fund, and Medical University Innsbruck. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in, or financial conflict with, the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.

References

  • OlanowCAgidYMizunoYLevodopa in the treatment of Parkinson’s disease: current controversiesMov Disord2004199997100515372588
  • HollowayRShoulsonIFahnSPramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trialArch Neurol20046171044105315262734
  • GerfenCEngberTMahanLD1 and D2 dopamine receptor-regulated gene expression of striatonigral and striatopallidal neuronsScience19902504986142914322147780
  • HerreroMAugoodSHirschEEffects of L-DOPA on preproen-kephalin and preprotachykinin gene expression in the MPTP-treated monkey striatumNeuroscience1995684118911988544992
  • JolkkonenJJennerPMarsdenCL-DOPA reverses altered gene expression of substance P but not enkephalin in the caudate-putamen of common marmosets treated with MPTPBrain Res Mol Brain Res19953222973077500841
  • AntoniniATolosaEMizunoYYamamotoMPoeweWHA reassessment of risks and benefits of dopamine agonists in Parkinson’s diseaseLancet Neurol200981092993719709931
  • AntoniniAPoeweWFibrotic heart-valve reactions to dopamine-agonist treatment in Parkinson’s diseaseLancet Neurol20076982682917706566
  • RascolOBrooksDKorczynADe DeynPClarkeCLangAA five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. 056 Study GroupN Engl J Med2000342201484149110816186
  • GroupPSPramipexole vs levodopa as initial treatment for Parkinson disease: A randomized controlled trial. Parkinson Study GroupJAMA2000284151931193811035889
  • OertelWWoltersESampaioCPergolide versus levodopa monotherapy in early Parkinson’s disease patients: The PELMOPET studyMov Disord200621334335316211594
  • HorstinkMTolosaEBonuccelliUReview of the therapeutic management of Parkinson’s disease. Report of a joint task force of the European Federation of Neurological Societies (EFNS) and the Movement Disorder Society-European Section (MDS-ES). Part II: late (complicated) Parkinson’s diseaseEur J Neurol200613111186120217038032
  • MiyasakiJMMartinWSuchowerskyOWeinerWJLangAEPractice parameter: initiation of treatment for Parkinson’s disease: an evidence-based review: report of the Quality Standards Subcommittee of the American Academy of NeurologyNeurology2002581111711781398
  • GerlachMDoubleKArzbergerTLeblhuberFTatschnerTRiedererPDopamine receptor agonists in current clinical use: comparative dopamine receptor binding profiles defined in the human striatumJ Neural Transm20031101119112714523624
  • KohnoYTakeuchiSPharmacological profiles and clinical effects of antiparkinsonian agent, pramipexoleNihon Yakurigaku Zasshi2004123642944015170083
  • European Medicines AgencyAssessment Report for Sifrol EMEA/703892/2009LondonEuropean Medicines Agency2009 Available from: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Assessment_Report_-_Variation/human/000133/WC500049957.pdfAccessed August 9, 2010
  • JennerPKönen-BergmannMSchepersCHaertterSPharmacokinetics of a once-daily extended-release formulation of pramipexole in healthy male volunteers: three studiesClin Ther200931112698271120110012
  • PoeweWRascolOBaronePExtended-release pramipexole in early Parkinson disease: A 33-week randomized controlled trialNeurology20117775976621832218
  • HauserRASchapiraAHRascolORandomized, double-blind, multicenter evaluation of pramipexole extended release once daily in early Parkinson’s diseaseMov Disord201025152542254920669317
  • SchragASpottkeAQuinnNDodelRComparative responsiveness of Parkinson’s disease scales to change over timeMov Disord200924681381819199355
  • RascolOBaronePHauserRAEfficacy, safety, and tolerability of overnight switching from immediate- to once daily extended-release pramipexole in early Parkinson’s diseaseMov Disord201025142326233220669265
  • VillerFGuilleminFBriançonSMoumTSuurmeijerTvan den HeuvelWCompliance to drug treatment of patients with rheumatoid arthritis: a 3 year longitudinal studyJ Rheumatol199926102114212210529126
  • CramerJVachonLDesforgesCSussmanNMDose frequency and dose interval compliance with multiple antiepileptic medications during a controlled clinical trialEpilepsia19953611111111177588455
  • CramerJAA systematic review of adherence with medications for diabetesDiabetes Care20042751218122415111553
  • SackettDLHaynesRBGibsonESTaylorDWRobertsRSJohnsonALPatient compliance with antihypertensive regimensPatient Couns Health Educ197811182110238880
  • LeopoldNAPolanskyMHurkaMRDrug adherence in Parkinson’s diseaseMov Disord200419551351715133814
  • GrossetKABoneIGrossetDGSuboptimal medication adherence in Parkinson’s diseaseMov Disord200520111502150716037924
  • GrossetKAReidJLGrossetDGMedicine-taking behavior: implications of suboptimal compliance in Parkinson’s diseaseMov Disord200520111397140416092116
  • HaynesRBDantesRPatient compliance and the conduct and interpretation of therapeutic trialsControl Clin Trials19878112193568692
  • RichterAAntonSEKochPDennettSLThe impact of reducing dose frequency on health outcomesClin Ther200325823072335 discussion 230614512137
  • DavisKLEdinHMAllenJKPrevalence and cost of medication nonadherence in Parkinson’s disease: evidence from administrative claims dataMov Disord201025447448020131374
  • GrossetDAntoniniACanesiMAdherence to antiparkinson medication in a multicenter European studyMov Disord200924682683219191340
  • SchapiraAHBaronePHauserRAExtended-release pramipexole in advanced Parkinson disease: a randomized controlled trialNeurology201177876777421832216