Skip to main content
Erschienen in: Advances in Therapy 5/2022

Open Access 10.03.2022 | Commentary

P2B001 (Extended Release Pramipexole and Rasagiline): A New Treatment Option in Development for Parkinson’s Disease

verfasst von: Robert A. Hauser, Nir Giladi, Werner Poewe, Jonathan Brotchie, Hadas Friedman, Sheila Oren, Pninit Litman

Erschienen in: Advances in Therapy | Ausgabe 5/2022

Abstract

Despite levodopa’s superior efficacy in reducing the motor symptoms of Parkinson’s disease (PD), its risk to induce motor complications requires consideration of the pros and cons of initiating treatment with levodopa-sparing strategies. The current drive toward early levodopa monotherapy is primarily driven by safety and tolerability concerns with dopamine agonists and only mild efficacy of other available approaches. Recently, P2B001, a novel once-daily combination of low-dose, extended-release formulations of pramipexole and rasagiline (0.6 mg and 0.75 mg respectively), has entered clinical development. In this drug evaluation, we review the preclinical and current clinical data for P2B001 and its components. The P2B001 combination has the potential to provide greater efficacy than either pramipexole or rasagiline alone and a better tolerability profile compared to higher dosage dopamine agonist monotherapy, while maintaining the advantage of lower motor complication risk than levodopa.
Key Summary Points
P2B001 is in development as once-daily monotherapy for the treatment of the signs and symptoms early Parkinson’s disease.
P2B001 is a novel, fixed-dose, once-daily combination of extended-release formulations of pramipexole and rasagiline (0.6/0.75 mg), both components at low doses that are not individually available on the market.
The combination of pramipexole and rasagiline aims to improve striatal dopaminergic transmission via distinct and potentially synergistic mechanisms.
Phase 2 data demonstrated significant symptomatic efficacy of P2B001 versus placebo, with a benign safety profile that was similar to placebo.

Introduction

Parkinson’s disease (PD) is the fastest growing neurologic disorder across the globe [1]. It has been estimated that in 2016 there were approximately 6.1 million people living with the disease [2], with a life-time risk of PD as high as 1 in 15 [3]. A rapid increase in prevalence is not solely due to an aging population, but also due to increasing life expectancy [2, 3] and thus longer disease durations than in previous years. One French study estimated that by 2030 PD patients aged 65 will live an extra 3 years compared to patients in 2010 [4]. While the prevalence of PD increases steadily with age, almost a quarter of those affected had onset before the age of 65 years old [5, 6], with the implication that these patients can be expected to live for many years with the disease.
Once diagnosed, it is now generally agreed that there is no clinical rationale to postpone symptomatic treatment in people who develop PD-related disability [6]. Conventional pharmacotherapy for motor symptom control is primarily based on dopaminergic replacement strategies [6, 7]. Current European guidelines recommend levodopa, dopamine agonists and monoamine oxidase type B (MAO-B) inhibitors as having Level A evidence for the treatment of early untreated PD [8, 9]. The choice of first drug is left to clinical judgment depending on the need for symptomatic efficacy in improving motor disability (lowest with MAO-B inhibitors and highest with levodopa) compared with the risks of developing motor-complications (highest for levodopa) or risks for daytime somnolence, hallucinosis and problems with impulse control (lowest for MAO-B inhibitors and highest for dopamine agonists) [8, 9]. By contrast, the recent American Academy of Neurology (AAN) guidelines [10] recommend levodopa as the initial therapy for most patients with early PD seeking treatment for motor symptoms. While dopamine agonists should be avoided in subjects at high risk for dopaminergic adverse events (AEs), including patients aged > 70 years, those with a history of impulse control disorders (ICDs) or pre-existing excessive daytime sleepiness, cognitive impairment and hallucinosis, they may still be used in select patients aged ≤ 60 years who are at higher risk for the development of dyskinesia [10].
Despite levodopa’s superior efficacy in reducing the motor symptoms of PD [11, 12], its risk to induce motor complications requires consideration of the pros and cons of initiating treatment with levodopa-sparing strategies. Although their effect size in reducing motor symptoms is inferior to levodopa overall, dopamine agonists may provide satisfactory symptom control in early disease for several years and dopamine agonist monotherapy has a very low risk to induce motor complications [11, 12]. Likewise, in the early disease stages, when patients present with mild motor symptoms, MAO-B inhibitors may also provide sufficient benefit with low risk for adverse events [13]. Both of these levodopa-sparing approaches offer opportunity for once-daily drug regimens, which are more convenient for patients and have been shown to enhance adherence [14].
While early combination therapy approaches were initially explored for levodopa and dopamine agonists (with and without MAO-B inhibition) in the 1980s [1517], the use of initiating treatment with drug combinations has since been neglected in clinical research and the potential benefits of combining drugs with different mechanisms of action have remained largely unexplored. Recently, a novel drug formulation combining an MAO-B inhibitor with a low-dose dopamine agonist has entered clinical development [18]. This combination has the potential to provide greater efficacy than either agent alone and a better tolerability profile compared to higher dosage dopamine agonist monotherapy, while maintaining the advantage of lower motor complication risk than levodopa. In this drug evaluation, we review the data for P2B001, which combines lower than currently used doses of extended release (ER)-pramipexole and ER-rasagiline (0.6 mg and 0.75 mg, respectively) and is under development for patients with early PD.

Introduction to P2B001 and its Components

P2B001 is a combination capsule developed to slowly release ER-pramipexole and ER-rasagiline simultaneously throughout the day. As will be discussed below, both components have a large evidence base supporting their efficacy. Whereas pramipexole provides the stronger symptomatic effect of the two, its use is limited by the development of dose-related adverse events [19]. Therefore, the P2B001 ER-pramipexole component dose is limited to 0.6 mg/day. Whereas rasagiline has a safety profile similar to placebo, its use as monotherapy is often short-lived because of mild efficacy in the face of a progressive disease. The combination of pramipexole and rasagiline was chosen for clinical development because they are thought to act in potentially complementary mechanisms [20, 21].

Chemistry and Pharmacodynamics of P2B001 Components

Pramipexole

Pramipexole is a full, nonergot dopamine agonist with high relative in vitro specificity and full intrinsic activity at the dopamine D2-like subfamily of dopamine receptors, binding with higher affinity for D3 than to D2 or D4 receptor subtypes (Ki's of 0.5 nM vs. 3.3–3.9 nM, respectively) [2224]. The ratio of selectivity for binding to D3 over D2 is about 6.5–8, with some evidence of selectivity in functional assays [24]. Affinity for D1 receptors is insignificant, being around 200,000 times lower than that to D3 receptors [23]. Pramipexole may have multiple mechanisms of action that are not fully elucidated. However, it is known that pramipexole acts on presynaptic as well as postsynaptic dopamine receptors [23]. Whereas in the intact striatum stimulation of presynaptic autoreceptors of the D3 and D2 type is thought to reduce the synthesis and synaptic release of dopamine [25], in the parkinsonian state, the postsynaptic D3 and D2 receptors on the striatofugal neurons of the direct and indirect pathways are additionally stimulated. It is hypothesized that, in the parkinsonian state, pramipexole corrects the increased inhibitory impact of the direct pathway on motor activity by enhancing the direct transmission (through D3 receptors) and reducing the indirect transmission (through D2 receptors), thereby mimicking dopamine’s effects in the striatum [26]. Several potential mechanisms for neuroprotection have also been proposed [2729]; however, the PRIDE delayed-start study did not find evidence of a disease-modifying effect for pramipexole monotherapy [30].
The symptomatic efficacy of pramipexole in early PD is well established [11, 31, 32]. While its effect size is inferior to that of levodopa, pramipexole has a reduced propensity to cause motor complications [11]. This has led to its use as an option for initial monotherapy, especially in younger patients who have greater risk to develop dyskinesias in response to levodopa. However, there are safety concerns, including the induction of daytime sleepiness and sudden-onset sleep as well as ICDs. These side effects are often dose-related, with higher doses resulting in a higher risk compared with small to medium doses [3336].

Rasagiline

Rasagiline (N-propargyl-1-(R)-aminoindan) is a potent and irreversible inhibitor of MAO-B, which is attributed to irreversible covalent binding of its propargyl moiety to the flavin adenine dinucleotide (FAD) moiety of the enzyme. By binding to the FAD moiety, rasagiline prevents the access of dopamine to MAO-B, thereby inhibiting oxidative deamination to 3,4-dihydroxphenlyacetic acid (DOPAC) and raising the levels of dopamine [37]. MAO-B inhibition with rasagiline also increases the availability of phenylethylamine, which can enhance striatal dopamine release [38]. Striatal dopamine release may also be enhanced by the rasagiline active metabolite, 1-aminoindan, an effect which is thought to be separate from MAO-B inhibition (although 1-aminoindan is also a weak MAO-B inhibitor) [39, 40]. Both rasagiline and 1-aminoindan contain a propargylamine moiety which have been shown to inhibit apoptosis in both in vitro and in vivo models of PD [41]. Such effects are thought to be driven at the mitochondrial level where propargylamine inhibits apoptotic pathways, induces glial cell-derived neurotrophic factor (GDNF) and brain cell-derived neurotrophic factor (BDNF), and stimulates protein kinase C phosphorylation [42].
The symptomatic efficacy and safety of rasagiline monotherapy in early disease has also been firmly established by randomized controlled trials (RCTs) [13, 43]. The effect size is mild, but in early disease, rasagiline monotherapy has been sustained for years without the need for adjunct medications in a sizable proportion of subjects [44]. Rasagiline monotherapy is generally very well tolerated with little difference in the rate of adverse events between rasagiline and placebo [13, 43]. In addition, the neuroprotective properties of rasagiline seen in preclinical studies [42] did not readily translate to the clinic where one large randomized, delayed-start design trial of rasagiline monotherapy found that rasagiline 1.0 mg, but not 2.0 mg, had statistically significant effects on clinical progression [13].

Pharmacokinetics and Metabolism of P2B001

P2B001 contains both pramipexole and rasagiline formulated using a proprietary ER coating system. While ER pramipexole formulations have been available for over a decade, P2B001 contains the first ER formulation of rasagiline and is, additionally, the first combination of both.
Pramipexole has an absolute oral bioavailability > 90%, with steady absorption across the intestine and little first pass metabolism [26]. It exhibits linear pharmacokinetics, and < 20% is protein bound; > 90% of the absorbed dose is eliminated unchanged and almost exclusively by the kidneys. Rasagiline is rapidly absorbed, reaching peak plasma concentration (Cmax) in approximately 1 hour. The absolute bioavailability of rasagiline is about 36%, and plasma protein binding ranges from 88 to 94% with mean extent of binding of 61–63% to human albumin over the concentration range of 1–100 ng/ml [45]. At steady state, the mean AUC and Cmax for rasagiline and 1-aminoindan are linearly proportional to the rasagiline dose [46]. Rasagiline undergoes almost complete biotransformation in the liver prior to excretion. The metabolism of rasagiline proceeds through two main CYP-450-dependent pathways: N-dealkylation and/or hydroxylation to yield 1-aminoindan (major active metabolite), 3-hydroxy-N-propargyl-1-aminoindan and 3-hydroxy-1-aminoindan. Glucuronide conjugation of rasagiline and its metabolites, with subsequent urinary excretion, is the major elimination pathway. There is no correlation of pharmacokinetics with the MAO-B inhibition of rasagiline because of its irreversible inhibition of MAO-B. However, it is hypothesized that other mechanisms of efficacy (e.g., via 1-amino-indan [47, 48]) may be in effect, which may be better leveraged by using an extended-release formulation.
A comparative bioavailability study was performed in 22 healthy volunteers (aged 18–55 years) to assess the relative rasagiline and pramipexole systemic exposure of P2B001 under fasting conditions. The study was conducted in accordance with the Declaration of Helsinki and International Conference on Harmonization Good Clinical Practice Guidelines, and all participants provided written informed consent. The randomized, single-dose, crossover study compared a single dose of P2B001 (0.6 mg ER-pramipexole/0.75 mg ER-rasagiline) with the combination of branded pramipexole-ER (Mirapex ER®, Boehringer Ingelheim, Germany) plus branded rasagiline (Azilect®, Teva Pharmaceuticals, Israel) at currently used doses (0.75 mg and 1 mg, respectively). The overall pharmacokinetic profile of the P2B001 pramipexole component was similar to that seen with the branded pramipexole-ER plus rasagiline combination (albeit with lower Cmax and AUC due to lower dosing), while the Cmax of the P2B001 rasagiline component was significantly lower (from 5774 to 537 pg/ml) and the t1/2 significantly longer (from 3.9 to 12.5 h) than that obtained with the branded rasagiline due to the extended-release formulation (Fig. 1A, Table 1).
Table 1
Comparative pharmacokinetics of P2B001 components versus branded products
 
Cmax (pg/ml)
Tmax (h)
AUC0−inf (h*pg/ml)
t½ (h)
Pramipexole-ER (0.75 mg) + Rasagiline (1 mg) (N = 19)
PPX: 804 ± 222
RAS: 5774 ± 2447
PPX: 9.1 ± 3.4
RAS: 0.6 ± 0.3
PPX: 21,451 ± 5388
RAS: 4680 ± 1573
PPX: 12.2 ± 2.5
RAS: 3.9 ± 1.6
P2B001 (0.6/0.75 mg) (N = 19)
PPX: 765 ± 227
RAS: 537 ± 256
PPX: 7.7 ± 2.9
RAS: 2.5 ± 0.5
PPX: 18,480 ± 5182
RAS: 3000 ± 715
PPX: 14.6 ± 3.9
RAS: 12.5 ± 7.3
All data are mean ± SD
Thus, both drug components in P2B001 are slowly released simultaneously throughout the day. This profile reflects the desired change to an ER rasagiline formulation in which Cmax is significantly lower (important for safety), a sharp peak is avoided, and the half-life is extended while maintaining a comparable AUC. Moreover, the findings support the safety profile of P2B001 as the overall exposure is lower than the component products with established safety profiles. At steady state, the Cmax and AUC of both rasagiline and pramipexole are approximately twofold higher than after the first dose of P2B001. In a 7-day multidose study also conducted in healthy volunteers (n = 20), both the pramipexole and rasagiline components of P2B001 reached steady state after 5 days of administration (Fig. 1B).

P2B001 Clinical Efficacy

Phase II Clinical Study of Two Doses of P2B001 Versus Placebo

The efficacy and safety of the current P2B001 formulation (0.6 mg pramipexole-ER/0.75 mg rasagiline) and a lower pramipexole dose formulation (0.3 mg ER-pramipexole/0.75 mg ER-rasagiline) were tested in a Phase II 12-week multicenter double-blind, placebo-controlled clinical trial [18]. In this study, untreated patients with early PD were randomized to once-daily treatment with P2B001 (0.3 mg ER-pramipexole/0.75 mg ER-rasagiline), P2B001 (0.6 mg ER-pramipexole/0.75 mg ER-rasagiline) or placebo [18].
The placebo-adjusted mean change from baseline to Week 12 in Total Unified Parkinson’s Disease Rating Scale (UPDRS) score (parts I + II + III, primary end point) was − 4.7 points for the P2B001 0.6/0.75 mg group (P = 0.0004) and − 3.8 points for the P2B001 0.3/0.75 mg group (P = 0.003). Differences versus placebo were seen as early as Week 4 for both dosages of P2B001 (Fig. 2). The placebo-adjusted magnitude of change for the 0.6/0.75 mg dose group is considered clinically relevant [49] and was higher (5.7 points in the P2B001 0.6/0.75 mg group and 4.6 points in the P2B001 0.3/0.75 mg group) when an outlier site that recorded marked placebo effects was excluded in post hoc analysis [18]. Moreover, the symptomatic benefits observed with P2B001 compare well with the efficacy reported in randomized, placebo-controlled trials of the marketed higher dose of the separate components. For example, treatment with pramipexole (1.5 mg/day) improved Total UPDRS scores by 5.2 points versus placebo at Week 10 in the STEP-UP clinical trial [50] and by 4.8 points at 9 months in the PROUD study [51]. Similarly, in the pramipexole PRAMI-BID study, patients treated with pramipexole (1–1.5 mg/day) showed placebo-adjusted improvements of 4.4–4.7 Total UPDRS points at Week 12 [52]. Treatment with rasagiline (1 mg/day) improved Total UPDRS scores by 4.2 points versus the placebo group (which showed significant worsening over time) at Week 26 of the TEMPO study [53] and by 3.0 points at ~ Week 36 in the ADAGIO study [13].
The significant effects of the 0.6/0.75 mg dose of P2B001 on quality of life also compares favorably with the results seen with pramipexole monotherapy in the PRAMI-BID study [52]. Whereas treatment with P2B001 (0.6/0.75 mg) significantly improved PDQ-39 Total (treatment effect of − 3.3 ± 1.2 points vs. placebo; P = 0.01), ADL (− 7.2 ± 2.0 points vs. placebo, P = 0.0005) and Mobility (-4.7 ± 1.8 points vs. placebo P = 0.01) scores [18], these benefits were only achieved in the PRAMI-BID study when pramipexole was given at the higher daily dose of 1.5 mg/day (treatment effect of 2.1 points on PDQ-39 Total scores vs. placebo when given 0.75 mg BID or 0.5 mg TID, P ≤ 0.05). Effects on PDQ-39 Total scores were not significantly different from placebo for the pramipexole 1.0 mg/day (0.5 mg BID) dose group (P = 0.32) [52].

Phase III Clinical Study Comparing P2B001 with its Individual Components

The efficacy and safety of the 0.6/0.75 mg dose have recently undergone further evaluation in a Phase III randomized, pivotal study (NCT03329508). To answer the question of how P2B001 compares to its individual ER components, 544 eligible subjects with early untreated PD were randomized (2:2:2:1) to 12-week double-blind treatment with once-daily P2B001, pramipexole-ER (0.6 mg once daily) alone, rasagiline-ER (0.75 mg once daily) alone or currently marketed pramipexole-ER titrated to optimal dose (active calibration arm; 1.5, 3.0 or 4.5 mg) (Fig. 3). The primary efficacy endpoint compares the change from baseline to Week 12 in UPDRS-Total score (Parts II + III) for P2B001 with its individual components. The first secondary endpoint compares the change from baseline in Epworth Sleepiness Scale (ESS) for P2B001 versus titrated pramipexole-ER. Sample sizes were estimated separately for the primary and secondary endpoints, with a smaller sample required for the secondary endpoint.
At the time of this manuscript's submission, the sponsors had just announced that the study met its primary objective showing superiority of P2B001 0.6/0.75 mg compared to its individual components in the change of Total UPDRS (parts II + III) score. Secondary objectives were also met, including significantly less daytime sleepiness (somnolence) compared to a currently marketed pramipexole-ER [54]. Full peer-reviewed details of this study have not yet been published.

Safety and Tolerability of P2B001

In terms of safety and tolerability, as reported in the Phase II study, P2B001 was well tolerated with an adverse event profile similar to placebo, with the exception of mild and transient nausea and somnolence, known adverse events of pramipexole [18], although somnolence was not significantly different than for placebo when evaluated using the ESS.
Somnolence, which can lead to falls, injury and difficulties driving [33], is a common occurrence in patients receiving pramipexole at doses > 1.5 mg/day (0.5 mg TID) for PD [55]. While somnolence was experienced more frequently with P2B001 than placebo in the Phase II study [18], its incidence was lower than experienced across the pramipexole pivotal studies in early PD (16% with P2B001 [18] and 22% with pramipexole [55]). Indeed, placebo-adjusted changes from baseline in ESS score with P2B001 were comparable to placebo and less than has been reported with higher doses of pramipexole in early PD (0.5 vs. 1.0–2.1) in early PD [52, 56, 57]. While the proportion of patients who shifted to having an ESS score > 10 was similar for P2B001 (0.6/0.75 mg) to placebo (both 6%), it was less than the 16% reported with pramipexole-IR and 21% with pramipexole-ER (versus 8% with placebo) across earlier studies with pramipexole [58].
Table 2 compares the safety and tolerability of P2B001 (0.6/0.75 mg) in the phase II trial [18] with the safety of pramipexole in previous placebo-controlled studies of similar duration [50, 52]. There were no reports of ICDs, hallucinations or psychosis with P2B001 in the 12-week Phase II study [18]. While hallucinations were already seen in > 5% of patients treated with pramipexole 1.5 mg within 10 weeks of the STEP-UP study [50] and in 16.5% of patients receiving pramipexole (up to 4.5 mg/day) across the early PD pivotal trials [55], these psychiatric adverse events often develop over a longer time frame with higher doses of pramipexole, and a longer duration of observation is required before drawing firm conclusions. Headache was one of the most common adverse events leading to study discontinuation in the pramipexole trials [55], but was only experienced by one patient receiving P2B001 (0.6/0.75 mg) in the Phase II study [18].
Table 2
Adverse events reported with P2B001 in the Phase II study compared with data from previous randomized, placebo-controlled trials of pramipexole
Adverse event
P2B001 Phase II Study [18]
STEP-UP Pramipexole Study [50]
Pramipexole PRAMI-BID Study [52]
Placebo
P2B001
(0.6/0.75 mg)
Placebo
n = 51
Pramipexole (1.5 mg)
n = 54
Pramipexole groups combined (1.5–6 mg)
N = 213
Placebo
N = 77
Pramipexole (0.5 mg BID)
n = 81
Pramipexole (0.75 mg BID)
n = 73
Pramipexole (0.5 mg TID)
n = 80
Nausea
1 (2.0)
10 (20.4)
5 (9.8)
9 (16.7)
42 (19.7)
8 (10.4)
18 (22.2)
11 (15.1)
15 (18.8)
Somnolence
0
8 (16.3)
7 (13.7)
9 (16.7)
58 (27.2)
5 (6.5)
14 (17.3)
16 (21.9)
20 (25.0)
Dizziness
4 (8.0)
5 (10.2)
10 (19.6)
10 (18.5)
39 (18.3)
7 (9.1)
9 (11.1)
5 (6.8)
9 (11.3)
Fatigue
1 (2.0)
4 (8.2)
5 (9.8)
4 (7.4)
14 (6.8)
3 (3.9)
6 (7.4)
7 (9.6)
4 (5.0)
Insomnia
2 (4.0)
3 (6.1)
4 (7.8)
2 (3.7)
16 (7.5)
4 (5.2)
3 (3.7)
11 (15.1)
6 (7.5)
Orthostatic hypotension
4 (8.0)
2 (4.1)
NR
NR
NR
NR
NR
NR
NR
Headache
4 (8.0)
1 (2.0)
5 (9.8)
5 (9.2)
24 (11.3)
8 (10.4)
7 (8.6)
6 (8.2)
4 (5.0)
Hallucination
0
0
0
4 (7.4)
14 (6.6)
NR
NR
NR
NR
Constipation
1 (2.0)
1 (2.0)
3 (5.9)
4 (7.4)
23 (10.8)
2 (3.9)
8 (9.9)
7 (9.6)
5 (6.3)
Confusion
0
0
0
3 (5.6)
9 (4.2)
NR
NR
NR
NR
Abnormal dreams
0
0
NR
NR
NR
3 (3.9)
3 (3.7)
2 (2.7)
5 (6.3)
Peripheral edema
0
0
NR
NR
NR
0
2 (2.5)
4 (5.5)
3 (3.8)
NR, not reported
These data suggest that a low-dose, extended-release combination of pramipexole and rasagiline may be a safe and effective way to initiate therapy for patients with early PD. Patients who do not yet require levodopa therapy may prefer to take a once daily medication that offers good symptom control with an adverse event profile similar to placebo.

Regulatory Affairs

Following the positive findings of the Phase III study [54], a new drug application for P2B001 as a treatment for patients with early PD (newly diagnosed and early-stage disease) will be submitted to the US Food and Drug Administration (FDA) in 2022. Marketing approval will then be sought in Europe and the rest of the world.

Potential role for P2B001 in early Parkinson’s disease

The therapeutic potential of polytherapy using fixed combinations of agents with established symptomatic efficacy has remained virtually unexplored in the field of PD. In principle, combination therapies offer the potential of mechanistic synergy with efficacy going beyond that provided by individual components. As seen in other fields of medicine [59, 60], combining mechanisms of action offers the opportunity for dose saving, which in itself can improve safety and tolerability. In the case of P2B001, the aim is to leverage the complementary mechanisms of rasagiline and pramipexole in early PD, where the patient’s remaining central dopamine reserves are enhanced by MAO-B inhibition with rasagiline while pramipexole provides prolonged dopamine receptor stimulation. When given at doses that would be considered ‘submaximal’ as separate agents, together they have been shown to provide clinically relevant efficacy with a low propensity for adverse events [18].
The major concern that has limited the use of dopamine agonists in recent years relates to their potential to induce ICDs, which may affect 10–40% of patients depending on patient characteristics, dose and concomitant medications [36]. Since ICDs are particularly common with high-dose agonist therapy [35, 36], the dose savings afforded by a combination with a second dopaminergic drug can be expected to significantly lower the ICD and somnolence risks of pramipexole, and this likely applies to hallucinosis as well. Safety data from the ongoing P2B001 studies have been favorable in this regard, but long-term data will be important for a full risk–benefit assessment.
An important advantage to any approach to initiate therapy in PD is the possibility of a once-daily mode of administration—from both the perspective of convenience and adherence. Given its once-daily formulation, lack of titration and favorable side effect profile, P2B001 may be an attractive option for newly diagnosed patients, who often resist the idea of having to take their medication at least three times a day. In the longer term, initial therapy is expected to require levodopa supplementation to maintain sufficient motor symptom control, although a longer follow-up (than in the current studies) will be needed to understand the amount of time P2B001 can be used as a monotherapy. This will likely require lower doses compared to initial levodopa monotherapy and thus be associated with a lower risk of or longer delay to development of motor complications. Beyond its use as initial therapy in early PD, P2B001 seems worth investigating as an adjunct to levodopa in patients experiencing motor fluctuations. Both components have been shown to reduce motor fluctuations as adjunct therapies at higher doses, and their combined use at low dose might offer similar benefits with improved safety in this indication as well.

Acknowledgements

Funding

This review was supported by Pharma Two B, which employs three of the authors (HF, SO and PL), procured medical writing support and funded the journal’s Rapid Service and Open Access fees. No author received payment for this manuscript.

Medical Writing Assistance

The authors thank Anita Chadha-Patel, PhD, of ACP Clinical Communications Ltd. (Hertfordshire, UK) for providing medical writing support, which was funded by Pharma Two B (Rehovot, Israel) in accordance with Good Publication Practice guidelines.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Author Contributions

Robert A. Hauser, Sheila Oren and Pninit Litman contributed to the first draft equally, and all other authors provided critical review. All authors approved the final version of the submitted manuscript.

Disclosures

Robert A Hauser is supported in part by a Center of Excellence grant from the National Parkinson Foundation and is employed by the University of South Florida (Florida). He received payment from Adamas Pharmaceuticals, Inc. for participating as a Steering Committee member and reports receiving personal fees from Abbvie, Acorda, Adamas, Alterity, Amneal, Cerevance, Curium Pharma, Enterin, Global Kinetics Corp., Inhibikase, Jazz Pharmaceuticals, Kyowa Kirin, Merck, Merz, Neurocrine, NeuroDerm, Orion, Pharmather Inc., Pharma Two B; Sage Therapeutics, Scion, Sio Gene Therapies, Sunovion, Supernus, Tolmar, and Vivifi Biotech. Nir Giladi serves as consultant to Sionara, NeuroDerm, Pharma2B, Denali, Neuron23 and Abbvie, Sanofi-Genzyme and Biogen. He receives royalties from Lysosomal Therapeutics (LTI) and payment for lectures at Abbvie, Sanofi-Genzyme and Movement Disorder Society. He received research support from the Michael J Fox Foundation, the National Parkinson Foundation, the European Union and the Israel Science Foundation as well as from Teva NNE program, Biogen and Ionis. He receives support from the Sieratzki Family Foundation and the Aufzien Academic Center in Tel-Aviv University. Werner Poewe reports receiving personal fees from AbbVie, AFFiRiS, AstraZeneca, BIAL, Boston Scientific, Britannia, Intec, Ipsen, Lundbeck, NeuroDerm, Neurocrine, Denali Pharmaceuticals, Novartis, Orion Pharma, Prexton, Teva, UCB and Zambon. He receives royalties from Thieme, Wiley Blackwell, Oxford University Press and Cambridge University Press and grant support from the Michael J Fox Foundation, EU FP7 and Horizon 2020. Jonathan M. Brotchie has received consultancy payments from Atuka Inc. Hadas Friedman, Sheila Oren MD and Pninit Litman are full-time employees of Pharma Two B.

Compliance with Ethics Guidelines

The Phase I pharmacokinetic study included in this paper was conducted in accordance with the Declaration of Helsinki and International Conference on Harmonization Good Clinical Practice Guidelines, and all participants provided written informed consent.

Data Availability

Data for the pharmacokinetic study are available from the corresponding author on reasonable request. Full protocol details for the phase III study are available at clinical trials.gov: https://clinicaltrials.gov/ct2/show/NCT03329508.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Global, regional, and national burden of neurological disorders during 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet Neurol. 2017;16(11):877–97. Global, regional, and national burden of neurological disorders during 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet Neurol. 2017;16(11):877–97.
2.
Zurück zum Zitat Dorsey ER, Elbaz A, Nichols E, Abd-Allah F, Abdelalim A, Adsuar JC, et al. Global, regional, and national burden of Parkinson’s disease, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018;17(11):939–53.CrossRef Dorsey ER, Elbaz A, Nichols E, Abd-Allah F, Abdelalim A, Adsuar JC, et al. Global, regional, and national burden of Parkinson’s disease, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018;17(11):939–53.CrossRef
4.
Zurück zum Zitat Wanneveich M, Moisan F, Jacqmin-Gadda H, Elbaz A, Joly P. Projections of prevalence, lifetime risk, and life expectancy of Parkinson’s disease (2010–2030) in France. Mov Disord. 2018;33(9):1449–55.PubMedCrossRef Wanneveich M, Moisan F, Jacqmin-Gadda H, Elbaz A, Joly P. Projections of prevalence, lifetime risk, and life expectancy of Parkinson’s disease (2010–2030) in France. Mov Disord. 2018;33(9):1449–55.PubMedCrossRef
5.
Zurück zum Zitat Pringsheim T, Jette N, Frolkis A, Steeves TD. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov Disord. 2014;29(13):1583–90.PubMedCrossRef Pringsheim T, Jette N, Frolkis A, Steeves TD. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov Disord. 2014;29(13):1583–90.PubMedCrossRef
6.
7.
Zurück zum Zitat Fox SH, Katzenschlager R, Lim SY, Barton B, de Bie RMA, Seppi K, et al. International Parkinson and movement disorder society evidence-based medicine review: update on treatments for the motor symptoms of Parkinson’s disease. Mov Disord. 2018;33(8):1248–66.PubMedCrossRef Fox SH, Katzenschlager R, Lim SY, Barton B, de Bie RMA, Seppi K, et al. International Parkinson and movement disorder society evidence-based medicine review: update on treatments for the motor symptoms of Parkinson’s disease. Mov Disord. 2018;33(8):1248–66.PubMedCrossRef
8.
Zurück zum Zitat Ferreira JJ, Katzenschlager R, Bloem BR, Bonuccelli U, Burn D, Deuschl G, et al. Summary of the recommendations of the EFNS/MDS-ES review on therapeutic management of Parkinson’s disease. Eur J Neurol. 2013;20(1):5–15.PubMedCrossRef Ferreira JJ, Katzenschlager R, Bloem BR, Bonuccelli U, Burn D, Deuschl G, et al. Summary of the recommendations of the EFNS/MDS-ES review on therapeutic management of Parkinson’s disease. Eur J Neurol. 2013;20(1):5–15.PubMedCrossRef
10.
Zurück zum Zitat Pringsheim T, Day GS, Smith DB, Rae-Grant A, Licking N, Armstrong MJ, et al. Dopaminergic therapy for motor symptoms in early parkinson disease practice guideline summary: a report of the AAN guideline subcommittee. Neurology. 2021;97(20):942–57.PubMedCrossRef Pringsheim T, Day GS, Smith DB, Rae-Grant A, Licking N, Armstrong MJ, et al. Dopaminergic therapy for motor symptoms in early parkinson disease practice guideline summary: a report of the AAN guideline subcommittee. Neurology. 2021;97(20):942–57.PubMedCrossRef
11.
Zurück zum Zitat Holloway RG, Shoulson I, Fahn S, Kieburtz K, Lang A, Marek K, et al. Pramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. Arch Neurol. 2004;61(7):1044–53.PubMed Holloway RG, Shoulson I, Fahn S, Kieburtz K, Lang A, Marek K, et al. Pramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. Arch Neurol. 2004;61(7):1044–53.PubMed
12.
Zurück zum Zitat Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. 056 Study Group. N Engl J Med. 2000;342(20):1484–91.PubMedCrossRef Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. 056 Study Group. N Engl J Med. 2000;342(20):1484–91.PubMedCrossRef
13.
Zurück zum Zitat Olanow CW, Rascol O, Hauser R, Feigin PD, Jankovic J, Lang A, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med. 2009;361(13):1268–78.PubMedCrossRef Olanow CW, Rascol O, Hauser R, Feigin PD, Jankovic J, Lang A, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med. 2009;361(13):1268–78.PubMedCrossRef
14.
Zurück zum Zitat Grosset KA, Bone I, Grosset DG. Suboptimal medication adherence in Parkinson’s disease. Mov Disord. 2005;20(11):1502–7.PubMedCrossRef Grosset KA, Bone I, Grosset DG. Suboptimal medication adherence in Parkinson’s disease. Mov Disord. 2005;20(11):1502–7.PubMedCrossRef
15.
Zurück zum Zitat Rinne UK. Combined bromocriptine-levodopa therapy early in Parkinson’s disease. Neurology. 1985;35(8):1196–8.PubMedCrossRef Rinne UK. Combined bromocriptine-levodopa therapy early in Parkinson’s disease. Neurology. 1985;35(8):1196–8.PubMedCrossRef
16.
Zurück zum Zitat Rinne UK. Combination of a dopamine agonist, MAO-B inhibitor and levodopa—a new strategy in the treatment of early Parkinson’s disease. Acta Neurol Scand Suppl. 1989;126:165–9.PubMedCrossRef Rinne UK. Combination of a dopamine agonist, MAO-B inhibitor and levodopa—a new strategy in the treatment of early Parkinson’s disease. Acta Neurol Scand Suppl. 1989;126:165–9.PubMedCrossRef
17.
Zurück zum Zitat Rinne UK. New strategies in the treatment of early Parkinson’s disease. Acta Neurol Scand Suppl. 1991;136:95–8.PubMedCrossRef Rinne UK. New strategies in the treatment of early Parkinson’s disease. Acta Neurol Scand Suppl. 1991;136:95–8.PubMedCrossRef
18.
Zurück zum Zitat Olanow CW, Kieburtz K, Leinonen M, Elmer L, Giladi N, Hauser RA, et al. A randomized trial of a low-dose Rasagiline and Pramipexole combination (P2B001) in early Parkinson’s disease. Mov Disord. 2017;32(5):783–9.PubMedCrossRef Olanow CW, Kieburtz K, Leinonen M, Elmer L, Giladi N, Hauser RA, et al. A randomized trial of a low-dose Rasagiline and Pramipexole combination (P2B001) in early Parkinson’s disease. Mov Disord. 2017;32(5):783–9.PubMedCrossRef
19.
Zurück zum Zitat Borovac JA. Side effects of a dopamine agonist therapy for Parkinson’s disease: a mini-review of clinical pharmacology. Yale J Biol Med. 2016;89(1):37–47.PubMedPubMedCentral Borovac JA. Side effects of a dopamine agonist therapy for Parkinson’s disease: a mini-review of clinical pharmacology. Yale J Biol Med. 2016;89(1):37–47.PubMedPubMedCentral
20.
Zurück zum Zitat Riederer P, Gerlach M, Muller T, Reichmann H. Relating mode of action to clinical practice: dopaminergic agents in Parkinson’s disease. Parkinsonism Relat Disord. 2007;13(8):466–79.PubMedCrossRef Riederer P, Gerlach M, Muller T, Reichmann H. Relating mode of action to clinical practice: dopaminergic agents in Parkinson’s disease. Parkinsonism Relat Disord. 2007;13(8):466–79.PubMedCrossRef
21.
Zurück zum Zitat Colosimo C, Fabbrini G, Berardelli A. Drug Insight: new drugs in development for Parkinson’s disease. Nat Clin Pract Neurol. 2006;2(11):600–10.PubMedCrossRef Colosimo C, Fabbrini G, Berardelli A. Drug Insight: new drugs in development for Parkinson’s disease. Nat Clin Pract Neurol. 2006;2(11):600–10.PubMedCrossRef
22.
Zurück zum Zitat Newman-Tancredi A, Cussac D, Audinot V, Nicolas JP, De Ceuninck F, Boutin JA, et al. Differential actions of antiparkinson agents at multiple classes of monoaminergic receptor. II. Agonist and antagonist properties at subtypes of dopamine D(2)-like receptor and alpha(1)/alpha(2)-adrenoceptor. J Pharmacol Exp Ther. 2002;303(2):805–14. Newman-Tancredi A, Cussac D, Audinot V, Nicolas JP, De Ceuninck F, Boutin JA, et al. Differential actions of antiparkinson agents at multiple classes of monoaminergic receptor. II. Agonist and antagonist properties at subtypes of dopamine D(2)-like receptor and alpha(1)/alpha(2)-adrenoceptor. J Pharmacol Exp Ther. 2002;303(2):805–14.
23.
Zurück zum Zitat Kvernmo T, Härtter S, Burger E. A review of the receptor-binding and pharmacokinetic properties of dopamine agonists. Clin Ther. 2006;28(8):1065–78.PubMedCrossRef Kvernmo T, Härtter S, Burger E. A review of the receptor-binding and pharmacokinetic properties of dopamine agonists. Clin Ther. 2006;28(8):1065–78.PubMedCrossRef
24.
Zurück zum Zitat Mierau J, Schneider FJ, Ensinger HA, Chio CL, Lajiness ME, Huff RM. Pramipexole binding and activation of cloned and expressed dopamine D2, D3 and D4 receptors. Eur J Pharmacol Mol Pharmacol. 1995;290(1):29–36.CrossRef Mierau J, Schneider FJ, Ensinger HA, Chio CL, Lajiness ME, Huff RM. Pramipexole binding and activation of cloned and expressed dopamine D2, D3 and D4 receptors. Eur J Pharmacol Mol Pharmacol. 1995;290(1):29–36.CrossRef
25.
Zurück zum Zitat Mierau J, Schneider FJ, Ensinger HA, Chio CL, Lajiness ME, Huff RM. Pramipexole binding and activation of cloned and expressed dopamine D2, D3 and D4 receptors. Eur J Pharmacol. 1995;290(1):29–36.PubMedCrossRef Mierau J, Schneider FJ, Ensinger HA, Chio CL, Lajiness ME, Huff RM. Pramipexole binding and activation of cloned and expressed dopamine D2, D3 and D4 receptors. Eur J Pharmacol. 1995;290(1):29–36.PubMedCrossRef
26.
Zurück zum Zitat Eisenreich W, Sommer B, Hartter S, Jost WH. Pramipexole extended release: a novel treatment option in Parkinson’s disease. Parkinsons Dis. 2010;2010:612619.PubMedPubMedCentral Eisenreich W, Sommer B, Hartter S, Jost WH. Pramipexole extended release: a novel treatment option in Parkinson’s disease. Parkinsons Dis. 2010;2010:612619.PubMedPubMedCentral
27.
Zurück zum Zitat Schapira AHV. Dopamine agonists and neuroprotection in Parkinson’s disease. Eur J Neurol. 2002;9(s3):7–14.PubMedCrossRef Schapira AHV. Dopamine agonists and neuroprotection in Parkinson’s disease. Eur J Neurol. 2002;9(s3):7–14.PubMedCrossRef
28.
Zurück zum Zitat Bennett J, Carvey P, Hinds T, Johnson R, Le W, Phillips W, et al. Mechanisms of action of pramipexole: putative neuroprotective effects. Rev Contemp Pharmacother. 2001;12:33–57. Bennett J, Carvey P, Hinds T, Johnson R, Le W, Phillips W, et al. Mechanisms of action of pramipexole: putative neuroprotective effects. Rev Contemp Pharmacother. 2001;12:33–57.
29.
Zurück zum Zitat Albrecht S, Buerger E. Potential neuroprotection mechanisms in PD: focus on dopamine agonist pramipexole. Curr Med Res Opin. 2009;25(12):2977–87.PubMedCrossRef Albrecht S, Buerger E. Potential neuroprotection mechanisms in PD: focus on dopamine agonist pramipexole. Curr Med Res Opin. 2009;25(12):2977–87.PubMedCrossRef
30.
Zurück zum Zitat Schapira AH, McDermott MP, Barone P, Comella CL, Albrecht S, Hsu HH, et al. Pramipexole in patients with early Parkinson’s disease (PROUD): a randomised delayed-start trial. Lancet Neurol. 2013;12(8):747–55.PubMedPubMedCentralCrossRef Schapira AH, McDermott MP, Barone P, Comella CL, Albrecht S, Hsu HH, et al. Pramipexole in patients with early Parkinson’s disease (PROUD): a randomised delayed-start trial. Lancet Neurol. 2013;12(8):747–55.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Safety and efficacy of pramipexole in early parkinson disease: a randomized dose-ranging study. JAMA. 1997;278(2):125–30. Safety and efficacy of pramipexole in early parkinson disease: a randomized dose-ranging study. JAMA. 1997;278(2):125–30.
32.
Zurück zum Zitat Hubble JP, Koller WC, Cutler NR, Sramek JJ, Friedman J, Goetz C, et al. Pramipexole in patients with early Parkinson’s disease. Clin Neuropharmacol. 1995;18(4):338–47.PubMedCrossRef Hubble JP, Koller WC, Cutler NR, Sramek JJ, Friedman J, Goetz C, et al. Pramipexole in patients with early Parkinson’s disease. Clin Neuropharmacol. 1995;18(4):338–47.PubMedCrossRef
33.
Zurück zum Zitat Avorn J, Schneeweiss S, Sudarsky LR, Benner J, Kiyota Y, Levin R, et al. Sudden uncontrollable somnolence and medication use in Parkinson disease. Arch Neurol. 2005;62(8):1242–8.PubMedCrossRef Avorn J, Schneeweiss S, Sudarsky LR, Benner J, Kiyota Y, Levin R, et al. Sudden uncontrollable somnolence and medication use in Parkinson disease. Arch Neurol. 2005;62(8):1242–8.PubMedCrossRef
34.
35.
Zurück zum Zitat Weintraub D, Siderowf AD, Potenza MN, Goveas J, Morales KH, Duda JE, et al. Association of dopamine agonist use with impulse control disorders in Parkinson disease. Arch Neurol. 2006;63(7):969–73.PubMedPubMedCentralCrossRef Weintraub D, Siderowf AD, Potenza MN, Goveas J, Morales KH, Duda JE, et al. Association of dopamine agonist use with impulse control disorders in Parkinson disease. Arch Neurol. 2006;63(7):969–73.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Corvol JC, Artaud F, Cormier-Dequaire F, Rascol O, Durif F, Derkinderen P, et al. Longitudinal analysis of impulse control disorders in Parkinson disease. Neurology. 2018;91(3):e189–201.PubMedPubMedCentralCrossRef Corvol JC, Artaud F, Cormier-Dequaire F, Rascol O, Durif F, Derkinderen P, et al. Longitudinal analysis of impulse control disorders in Parkinson disease. Neurology. 2018;91(3):e189–201.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Youdim MB, Gross A, Finberg JP. Rasagiline [N-propargyl-1R(+)-aminoindan], a selective and potent inhibitor of mitochondrial monoamine oxidase B. Br J Pharmacol. 2001;132(2):500–6.PubMedPubMedCentralCrossRef Youdim MB, Gross A, Finberg JP. Rasagiline [N-propargyl-1R(+)-aminoindan], a selective and potent inhibitor of mitochondrial monoamine oxidase B. Br J Pharmacol. 2001;132(2):500–6.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Lamensdorf I, Youdim MB, Finberg JP. Effect of long-term treatment with selective monoamine oxidase A and B inhibitors on dopamine release from rat striatum in vivo. J Neurochem. 1996;67(4):1532–9.PubMedCrossRef Lamensdorf I, Youdim MB, Finberg JP. Effect of long-term treatment with selective monoamine oxidase A and B inhibitors on dopamine release from rat striatum in vivo. J Neurochem. 1996;67(4):1532–9.PubMedCrossRef
39.
Zurück zum Zitat Bar-Am O, Weinreb O, Amit T, Youdim MB. The neuroprotective mechanism of 1-(R)-aminoindan, the major metabolite of the anti-parkinsonian drug rasagiline. J Neurochem. 2010;112(5):1131–7.PubMedCrossRef Bar-Am O, Weinreb O, Amit T, Youdim MB. The neuroprotective mechanism of 1-(R)-aminoindan, the major metabolite of the anti-parkinsonian drug rasagiline. J Neurochem. 2010;112(5):1131–7.PubMedCrossRef
40.
Zurück zum Zitat Brotchie J, Johnston TH, Visanji NP, Pires D, Thiele S, Fitzer-Attas C, et al. 1-Aminoindan, a main metabolite of rasagiline, enhances dopamine release and provides symptomatic benefit in an animal model of Parkinson's disease. Parkinson Relat Disord. 2007;13((Suppl 2)):102 (Abstract). Brotchie J, Johnston TH, Visanji NP, Pires D, Thiele S, Fitzer-Attas C, et al. 1-Aminoindan, a main metabolite of rasagiline, enhances dopamine release and provides symptomatic benefit in an animal model of Parkinson's disease. Parkinson Relat Disord. 2007;13((Suppl 2)):102 (Abstract).
41.
Zurück zum Zitat Olanow CW. Rationale for considering that propargylamines might be neuroprotective in Parkinson’s disease. Neurology. 2006;66(10 Suppl 4):S69-79.PubMedCrossRef Olanow CW. Rationale for considering that propargylamines might be neuroprotective in Parkinson’s disease. Neurology. 2006;66(10 Suppl 4):S69-79.PubMedCrossRef
42.
Zurück zum Zitat Jenner P, Langston JW. Explaining ADAGIO: a critical review of the biological basis for the clinical effects of rasagiline. Mov Disord. 2011;13:2316–23.CrossRef Jenner P, Langston JW. Explaining ADAGIO: a critical review of the biological basis for the clinical effects of rasagiline. Mov Disord. 2011;13:2316–23.CrossRef
43.
Zurück zum Zitat A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch Neurol. 2002;59(12):1937–43. A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch Neurol. 2002;59(12):1937–43.
44.
Zurück zum Zitat Lew MF, Hauser RA, Hurtig HI, Ondo WG, Wojcieszek J, Goren T, et al. Long-term efficacy of rasagiline in early Parkinson’s disease. Int J Neurosci. 2010;120(6):404–8.PubMedCrossRef Lew MF, Hauser RA, Hurtig HI, Ondo WG, Wojcieszek J, Goren T, et al. Long-term efficacy of rasagiline in early Parkinson’s disease. Int J Neurosci. 2010;120(6):404–8.PubMedCrossRef
45.
Zurück zum Zitat Lecht S, Haroutiunian S, Hoffman A, Lazarovici P. Rasagiline - a novel MAO B inhibitor in Parkinson’s disease therapy. Ther Clin Risk Manag. 2007;3(3):467–74.PubMedPubMedCentral Lecht S, Haroutiunian S, Hoffman A, Lazarovici P. Rasagiline - a novel MAO B inhibitor in Parkinson’s disease therapy. Ther Clin Risk Manag. 2007;3(3):467–74.PubMedPubMedCentral
46.
Zurück zum Zitat Thébault JJ, Guillaume M, Levy R. Tolerability, safety, pharmacodynamics, and pharmacokinetics of rasagiline: a potent, selective, and irreversible monoamine oxidase type B inhibitor. Pharmacotherapy. 2004;24(10):1295–305.PubMedCrossRef Thébault JJ, Guillaume M, Levy R. Tolerability, safety, pharmacodynamics, and pharmacokinetics of rasagiline: a potent, selective, and irreversible monoamine oxidase type B inhibitor. Pharmacotherapy. 2004;24(10):1295–305.PubMedCrossRef
47.
Zurück zum Zitat Youdim MB, Bar Am O, Yogev-Falach M, Weinreb O, Maruyama W, Naoi M, et al. Rasagiline: neurodegeneration, neuroprotection, and mitochondrial permeability transition. J Neurosci Res. 2005;79(1–2):172–9.PubMedCrossRef Youdim MB, Bar Am O, Yogev-Falach M, Weinreb O, Maruyama W, Naoi M, et al. Rasagiline: neurodegeneration, neuroprotection, and mitochondrial permeability transition. J Neurosci Res. 2005;79(1–2):172–9.PubMedCrossRef
48.
Zurück zum Zitat Bar-Am O, Amit T, Youdim MB. Aminoindan and hydroxyaminoindan, metabolites of rasagiline and ladostigil, respectively, exert neuroprotective properties in vitro. J Neurochem. 2007;103(2):500–8.PubMedCrossRef Bar-Am O, Amit T, Youdim MB. Aminoindan and hydroxyaminoindan, metabolites of rasagiline and ladostigil, respectively, exert neuroprotective properties in vitro. J Neurochem. 2007;103(2):500–8.PubMedCrossRef
49.
Zurück zum Zitat Shulman LM, Gruber-Baldini AL, Anderson KE, Fishman PS, Reich SG, Weiner WJ. The clinically important difference on the unified Parkinson’s disease rating scale. Arch Neurol. 2010;67(1):64–70.PubMedCrossRef Shulman LM, Gruber-Baldini AL, Anderson KE, Fishman PS, Reich SG, Weiner WJ. The clinically important difference on the unified Parkinson’s disease rating scale. Arch Neurol. 2010;67(1):64–70.PubMedCrossRef
50.
Zurück zum Zitat Safety and efficacy of pramipexole in early Parkinson disease. A randomized dose-ranging study. Parkinson Study Group. JAMA. 1997;278(2):125–30. Safety and efficacy of pramipexole in early Parkinson disease. A randomized dose-ranging study. Parkinson Study Group. JAMA. 1997;278(2):125–30.
51.
Zurück zum Zitat Schapira AH, Barone P, Hauser RA, Mizuno Y, Rascol O, Busse M, et al. Extended-release pramipexole in advanced Parkinson disease: a randomized controlled trial. Neurology. 2011;77(8):767–74.PubMedCrossRef Schapira AH, Barone P, Hauser RA, Mizuno Y, Rascol O, Busse M, et al. Extended-release pramipexole in advanced Parkinson disease: a randomized controlled trial. Neurology. 2011;77(8):767–74.PubMedCrossRef
52.
Zurück zum Zitat Kieburtz K, Parkinson Study Group Prami BIDI. Twice-daily, low-dose pramipexole in early Parkinson's disease: a randomized, placebo-controlled trial. Mov Disord. 2011;26(1):37–44. Kieburtz K, Parkinson Study Group Prami BIDI. Twice-daily, low-dose pramipexole in early Parkinson's disease: a randomized, placebo-controlled trial. Mov Disord. 2011;26(1):37–44.
53.
Zurück zum Zitat A controlled, randomized, delayed-start study of rasagiline in early Parkinson disease. Arch Neurol. 2004;61(4):561–6. A controlled, randomized, delayed-start study of rasagiline in early Parkinson disease. Arch Neurol. 2004;61(4):561–6.
56.
Zurück zum Zitat Hauser RA, Schapira AH, Rascol O, Barone P, Mizuno Y, Salin L, et al. Randomized, double-blind, multicenter evaluation of pramipexole extended release once daily in early Parkinson’s disease. Mov Disord. 2010;25(15):2542–9.PubMedCrossRef Hauser RA, Schapira AH, Rascol O, Barone P, Mizuno Y, Salin L, et al. Randomized, double-blind, multicenter evaluation of pramipexole extended release once daily in early Parkinson’s disease. Mov Disord. 2010;25(15):2542–9.PubMedCrossRef
57.
Zurück zum Zitat Poewe W, Rascol O, Barone P, Hauser RA, Mizuno Y, Haaksma M, et al. Extended-release pramipexole in early Parkinson disease: a 33-week randomized controlled trial. Neurology. 2011;77(8):759–66.PubMedCrossRef Poewe W, Rascol O, Barone P, Hauser RA, Mizuno Y, Haaksma M, et al. Extended-release pramipexole in early Parkinson disease: a 33-week randomized controlled trial. Neurology. 2011;77(8):759–66.PubMedCrossRef
59.
Zurück zum Zitat MacDonald TM, Williams B, Webb DJ, Morant S, Caulfield M, Cruickshank JK, et al. Combination Therapy Is Superior to Sequential Monotherapy for the Initial Treatment of Hypertension: A Double-Blind Randomized Controlled Trial. Journal of the American Heart Association. 2017;6(11):e006986. MacDonald TM, Williams B, Webb DJ, Morant S, Caulfield M, Cruickshank JK, et al. Combination Therapy Is Superior to Sequential Monotherapy for the Initial Treatment of Hypertension: A Double-Blind Randomized Controlled Trial. Journal of the American Heart Association. 2017;6(11):e006986.
60.
Zurück zum Zitat Bayat Mokhtari R, Homayouni TS, Baluch N, Morgatskaya E, Kumar S, Das B, et al. Combination therapy in combating cancer. Oncotarget. 2017;8(23):38022–43.PubMedCrossRef Bayat Mokhtari R, Homayouni TS, Baluch N, Morgatskaya E, Kumar S, Das B, et al. Combination therapy in combating cancer. Oncotarget. 2017;8(23):38022–43.PubMedCrossRef
Metadaten
Titel
P2B001 (Extended Release Pramipexole and Rasagiline): A New Treatment Option in Development for Parkinson’s Disease
verfasst von
Robert A. Hauser
Nir Giladi
Werner Poewe
Jonathan Brotchie
Hadas Friedman
Sheila Oren
Pninit Litman
Publikationsdatum
10.03.2022
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 5/2022
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-022-02097-2

Weitere Artikel der Ausgabe 5/2022

Advances in Therapy 5/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Triglyzeridsenker schützt nicht nur Hochrisikopatienten

10.05.2024 Hypercholesterinämie Nachrichten

Patienten mit Arteriosklerose-bedingten kardiovaskulären Erkrankungen, die trotz Statineinnahme zu hohe Triglyzeridspiegel haben, profitieren von einer Behandlung mit Icosapent-Ethyl, und zwar unabhängig vom individuellen Risikoprofil.

Gibt es eine Wende bei den bioresorbierbaren Gefäßstützen?

In den USA ist erstmals eine bioresorbierbare Gefäßstütze – auch Scaffold genannt – zur Rekanalisation infrapoplitealer Arterien bei schwerer PAVK zugelassen worden. Das markiert einen Wendepunkt in der Geschichte dieser speziellen Gefäßstützen.

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Wie managen Sie die schmerzhafte diabetische Polyneuropathie?

10.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Mit Capsaicin-Pflastern steht eine neue innovative Therapie bei schmerzhafter diabetischer Polyneuropathie zur Verfügung. Bei therapierefraktären Schmerzen stellt die Hochfrequenz-Rückenmarkstimulation eine adäquate Option dar.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.