PF-06649751 efficacy and safety in early Parkinson's disease: a randomized, placebo-controlled trial

Robert Riesenberg, John Werth, Yao Zhang, Sridhar Duvvuri, David Gray, Robert Riesenberg, John Werth, Yao Zhang, Sridhar Duvvuri, David Gray

Abstract

Background: PF-06649751 is a novel, oral, non-catechol-based, D1/D5 dopamine receptor partial agonist under investigation for the treatment of motor symptoms associated with Parkinson's disease.

Methods: A 15-week, phase II, double-blind, placebo-controlled clinical trial was conducted to assess the efficacy and safety of flexible-dose PF-06649751 in subjects with early stage Parkinson's disease (ClinicalTrials.gov identifier: NCT02847650).

Results: Enrollment was terminated early for reasons unrelated to the trial. Overall, 57 subjects received study medication (PF-06649751 = 29; placebo = 28) and 47 completed the study (PF-06649751 = 25; placebo = 22). Despite early termination, the study met its primary endpoint with the PF-06649751 group showing statistically significant improvement from baseline in the Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) Part III score at week 15 compared with placebo. Mean (SE) change in MDS-UPDRS Part III score was -9.0 (1.54) for PF-06649751 and -4.3 (1.65) for placebo. This corresponds to an improvement versus placebo of 4.8 for the PF-06649751 group (two-sided p = 0.0407; 90% CI = 1.0, 8.6). Statistically significant improvement in MDS-UPDRS-III score was also observed at all assessment time points prior to week 15. The safety profile of PF-06649751 was similar to that observed in prior studies, with the majority of adverse events (AEs) reported as mild or moderate. The most common AEs in the PF-06649751 group were nausea, headache, dry mouth, somnolence, and tremor.

Conclusions: Once-daily dosing of oral PF-06649751 resulted in significant improvement of motor symptoms and was generally well tolerated in subjects with early stage Parkinson's disease.

Keywords: D1/D5 receptor; PF-06649751; Parkinson’s disease; partial agonist; phase II clinical trial.

Conflict of interest statement

Conflict of interest statement: JW and YZ are full-time employees of, and own stock or stock options, in Pfizer Inc. DG and SD were full-time employees of Pfizer Inc. at the time the study was conducted, and own stock in Pfizer Inc. DG is named on the patent for PF-06649751 (though all rights are assigned to Pfizer Inc). RR has no potential conflicts to disclose. Medical writing support provided by Matt Soulsby of Engage Scientific was funded by Pfizer. RR reports no financial relationships with commercial interests. JW, and YZ have received salary as full-time employees of Pfizer Inc. and own stock in Pfizer Inc. SD and DG have received salary as full-time employees of Pfizer Inc., and subsequently Cerevel Therapeutics, and own stock in Pfizer Inc.

© The Author(s), 2020.

Figures

Figure 1.
Figure 1.
Design of the study. aMaximum allowed dose at each interval of the dose optimization period is shown. Subjects initially received 0.25 mg PF-06649751 or matching placebo, with a mandatory increase to 0.75 mg at Visit 2. At Visits 3–10, the dose could be increased 1 level, decreased 1 level, or remain unchanged to achieve an optimum balance of tolerability and control of motor symptoms. Subjects received their optimized dose (up to 15 mg) from Visit 10–14. See methods for details. All visits were clinic except for V5, V7, V9, V11, V13, and V16, which were by phone. There was a +3 day window for Visits 2–9 and a ±3 day window for Visits 10–16, with the exception of Visit 14 which had a −3 day window.
Figure 2.
Figure 2.
Subject disposition.
Figure 3.
Figure 3.
Change in MDS-UPDRS Part III score over the course of the study. Data shown are mean ± SE. 2-sided p ⩽ 0.1 versus placebo; 2-sided p value at week 15 (primary endpoint) was 0.0407. MDS-UPDRS, Movement disorder society-unified Parkinson’s disease rating scale; SE, standard error.

References

    1. Zeng XS, Geng WS, Jia JJ, et al. Cellular and molecular basis of neurodegeneration in Parkinson disease. Front Aging Neurosci 2018; 10: 109.
    1. Rizek P, Kumar N, Jog MS. An update on the diagnosis and treatment of Parkinson disease. CMAJ 2016; 188: 1157–1165.
    1. Karimi-Moghadam A, Charsouei S, Bell B, et al. Parkinson disease from Mendelian forms to genetic susceptibility: new molecular insights into the neurodegeneration process. Cell Mol Neurobiol 2018; 38: 1153–1178.
    1. Dickson DW. Neuropathology of Parkinson disease. Parkinsonism Relat Disord 2018; 46(Suppl. 1): S30–S33.
    1. Poewe W, Seppi K, Tanner CM, et al. Parkinson disease. Nat Rev Dis Primers 2017; 3: 17013.
    1. Charvin D, Medori R, Hauser RA, et al. Therapeutic strategies for Parkinson disease: beyond dopaminergic drugs. Nat Rev Drug Discov 2018; 17: 804–822.
    1. Fox SH, Katzenschlager R, Lim SY, 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: 1248–1266.
    1. Ossig C, Reichmann H. Treatment strategies in early and advanced Parkinson disease. Neurol Clin 2015; 33: 19–37.
    1. Connolly BS, Lang AE. Pharmacological treatment of Parkinson disease: a review. JAMA 2014; 311: 1670–1683.
    1. Bastide MF, Meissner WG, Picconi B, et al. Pathophysiology of L-dopa-induced motor and non-motor complications in Parkinson’s disease. Prog Neurobiol 2015; 132: 96–168.
    1. Tran TN, Vo TNN, Frei K, et al. Levodopa-induced dyskinesia: clinical features, incidence, and risk factors. J Neural Transm (Vienna) 2018; 125: 1109–1117.
    1. Vijayakumar D, Jankovic J. Drug-induced dyskinesia, part 1: treatment of levodopa-induced dyskinesia. Drugs 2016; 76: 759–777.
    1. Kim YC, Alberico SL, Emmons E, et al. New therapeutic strategies targeting D1-type dopamine receptors for neuropsychiatric disease. Front Biol (Beijing) 2015; 10: 230–238.
    1. Waddington JL. Therapeutic potential of selective D-1 dopamine receptor agonists and antagonists in psychiatry and neurology. Gen Pharmacol 1988; 19: 55–60.
    1. Mailman R, Huang X, Nichols DE. Parkinson’s disease and D1 dopamine receptors. Curr Opin Investig Drugs 2001; 2: 1582–1591.
    1. Rascol O, Blin O, Thalamas C, et al. ABT-431, a D1 receptor agonist prodrug, has efficacy in Parkinson’s disease. Ann Neurol 1999; 45: 736–741.
    1. Blanchet PJ, Fang J, Gillespie M, et al. Effects of the full dopamine D1 receptor agonist dihydrexidine in Parkinson’s disease. Clin Neuropharmacol 1998; 21: 339–343.
    1. Rascol O, Nutt JG, Blin O, et al. Induction by dopamine D1 receptor agonist ABT-431 of dyskinesia similar to levodopa in patients with Parkinson disease. Arch Neurol 2001; 58: 249–254.
    1. Boehringer Ingelheim Pharmaceuticals Inc. Mirapex US prescribing information, (accessed November 30 2018).
    1. GlaxoSmithKline. Requip US prescribing information, (accessed 30 November 2018). prescribing information
    1. UCB Inc. Neupro US prescribing information, (accessed 30 November 2018).
    1. Dietrichs E, Odin P. Algorithms for the treatment of motor problems in Parkinson’s disease. Acta Neurol Scand 2017; 136: 378–385.
    1. Stowe RL, Ives NJ, Clarke C, et al. Dopamine agonist therapy in early Parkinson’s disease. Cochrane Database Syst Rev 2008: 2: CD006564.
    1. Sohur US, Gray DL, Duvvuri S, et al. Phase 1 Parkinson’s disease studies show the dopamine D1/D5 agonist PF-06649751 is safe and well tolerated. Neurol Ther 2018; 7: 307–319.
    1. Goetz CG, Tilley BC, Shaftman SR, et al. Movement disorder society-sponsored revision of the unified Parkinson’s disease rating scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord 2008; 23: 2129–2170.
    1. Posner K, Brown GK, Stanley B, et al. The Columbia-suicide severity rating scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am J Psychiatry 2011; 168: 1266–1277.
    1. Weintraub D, Mamikonyan E, Papay K, et al. Questionnaire for impulsive-compulsive disorders in Parkinson’s disease-rating scale. Mov Disord 2012; 27: 242–247.
    1. Rickels K, Garcia-Espana F, Mandos LA, et al. Physician withdrawal checklist (PWC-20). J Clin Psychopharmacol 2008; 28: 447–451.
    1. Beck AT, Steer RA, Ball R, et al. Comparison of beck depression inventories -IA and -II in psychiatric outpatients. J Pers Assess 1996; 67: 588–597.
    1. Johns MW. A new method for measuring daytime sleepiness: the Epworth sleepiness scale. Sleep 1991; 14: 540–545.
    1. Peto V, Jenkinson C, Fitzpatrick R. PDQ-39: a review of the development, validation and application of a Parkinson’s disease quality of life questionnaire and its associated measures. J Neurol 1998; 245(Suppl. 1): S10–S14.
    1. Herdman M, Gudex C, Lloyd A, et al. Development and preliminary testing of the new five-level version of EQ-5D (EQ-5D-5L). Qual Life Res 2011; 20: 1727–1736.
    1. Shulman LM, Gruber-Baldini AL, Anderson KE, et al. The clinically important difference on the unified Parkinson’s disease rating scale. Arch Neurol 2010; 67: 64–70.
    1. Goetz CG, Stebbins GT, Tilley BC. Calibration of unified Parkinson’s disease rating scale scores to movement disorder society-unified Parkinson’s disease rating scale scores. Mov Disord 2012; 27: 1239–1242.

Source: PubMed

3
Subskrybuj