Safety, Tolerability, and Pharmacokinetics of Mevidalen (LY3154207), a Centrally Acting Dopamine D1 Receptor-Positive Allosteric Modulator (D1PAM), in Healthy Subjects

Darren Wilbraham, Kevin M Biglan, Kjell A Svensson, Max Tsai, William Kielbasa, Darren Wilbraham, Kevin M Biglan, Kjell A Svensson, Max Tsai, William Kielbasa

Abstract

Activation of the brain dopamine D1 receptor has attracted attention because of its promising role in neuropsychiatric diseases. Although efforts to develop D1 agonists have been challenging, a positive allosteric modulator (PAM), represents an attractive approach with potential better drug-like properties. Phase 1 single-ascending-dose (SAD; NCT03616795) and multiple-ascending-dose (MAD; NCT02562768) studies with the D1PAM mevidalen (LY3154207) were conducted with healthy subjects. There were no treatment-related serious adverse events (AEs) in these studies. In the SAD study, 25-200 mg administered orally showed dose-proportional pharmacokinetics (PK) and acute dose-related increases in systolic blood pressure (SBP) and diastolic blood pressure DBP) and pulse rate at doses ≥ 75 mg. AE related to central activation were seen at doses ≥ 75 mg. At 25 and 75 mg, central penetration of mevidalen was confirmed by measurement of mevidalen in cerebrospinal fluid. In the MAD study, once-daily doses of mevidalen at 15-150 mg for 14 days showed dose-proportional PK. Acute dose-dependent increases in SBP, DBP, and PR were observed on initial administration, but with repeated dosing the effects diminished and returned toward baseline levels. Overall, these findings support further investigation of mevidalen as a potential treatment for a range of neuropsychiatric disorders.

Trial registration: ClinicalTrials.gov NCT03305809 NCT03616795 NCT02562768.

Keywords: dopamine; mevidalen (LY3154207); pharmacokinetics; safety; tolerability.

Conflict of interest statement

Darren Wilbraham, Kevin M. Biglan, Kjell A. Svensson, Max Tsai, and William Kielbasa are employees of Eli Lilly and Company Inc., and may own stock in this company.

© 2020 Eli Lilly and Company. Clinical Pharmacology in Drug Development published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.

Figures

Figure 1
Figure 1
(A) Design of single ascending dose. Part A: subjects were randomized to mevidalen (n = 6) or placebo (n = 3) in each cohort in each dosing period; part B: subjects were randomized to mevidalen (n = 8) or placebo (n = 4) in each cohort. aSafety review completed after each dose level prior to escalation. bDose escalation was terminated at 200 mg owing to cardiovascular effects. LY, mevidalen; PBO, placebo. (B) Design of multiple ascending dose. Each cohort contained 12 subjects (mevidalen, n = 9; placebo, n = 3) dosed daily for 14 days. aEach ascending dose cohort commenced only after review of the safety data to at least day 7 from the previous cohort. bThe planned cohort 4 dose of 200 mg was reduced to 150 mg on the basis of emerging safety data from cohort 3. CRU, clinical research unit; CSF, cerebrospinal fluid; LY, mevidalen; PBO, placebo; R, randomized.
Figure 2
Figure 2
(A) Mean plasma concentration‐time profiles following single doses of mevidalen from part A of the SAD study. (B) Mean plasma and CSF concentration‐time profiles following single doses of mevidalen from part B of the SAD study.
Figure 3
Figure 3
Mean plasma concentration‐time profiles on day 14 following once‐daily dosing of mevidalen.
Figure 4
Figure 4
Hourly mean ambulatory blood pressure monitoring (SAD study) change from baseline in (A) systolic blood pressure, (B) pulse rate, and (C) diastolic blood pressure. ABPM, ambulatory blood pressure monitoring; BP, blood pressure.
Figure 5
Figure 5
The initial dose‐related increase in (A) systolic blood pressure, (B) pulse rate, and (C) diastolic blood pressure on day 1 showed accommodation following once‐daily dosing of mevidalen. Data shown as least‐squares mean and 90% confidence interval.

References

    1. Svensson KA, Hao J, Bruns RF. Positive Allosteric modulators of the dopamine D1 receptor: a new mechanism for the treatment of neuropsychiatric disorders. In: Witkin JM, ed. Advances in Pharmacology. Academic Press; 2019;86:273‐305.
    1. Klein MO, Battagello DS, Cardoso AR, Hauser DN, Bittencourt JC, Correa RG. Dopamine: functions, signaling, and association with neurological diseases. Cell Mol Neurobiol. 2019;39(1):31‐59.
    1. Arnsten AF, Li BM. Neurobiology of executive functions: catecholamine influences on prefrontal cortical functions. Biol Psychiatry. 2005;57(11):1377‐1384.
    1. Iversen SD, Iversen LL. Dopamine: 50 years in perspective. Trends Neurosci. 2007;30(5):188‐193.
    1. Beaulieu JM, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63(1):182‐217.
    1. Missale C, Nash SR, Robinson SW, Jaber M, Caron MG. Dopamine receptors: from structure to function. Physiol Rev. 1998;78(1):189‐225.
    1. Arnsten AF. The neurobiology of thought: the groundbreaking discoveries of Patricia Goldman‐Rakic 1937−2003. Cereb Cortex. 2013;23(10):2269‐2281.
    1. Puig MV, Rose J, Schmidt R, et al. Dopamine modulation of learning and memory in the prefrontal cortex: insights from studies in primates, rodents, and birds. Front Neural Circuits. 2014;93(8):1‐14.
    1. Goldman‐Rakic PS, Castner SA, Svensson TH, et al. Targeting the dopamine D1 receptor in schizophrenia: insights for cognitive dysfunction. Psychopharmacology. 2004;174(1):3‐16.
    1. Arnsten AF, Girgis RR, Gray DL, et al. Novel dopamine therapeutics for cognitive deficits in schizophrenia. Biol Psychiatry. 2017;81(1):67‐77.
    1. Hall A, Provins L, Valade A. Novel strategies to activate the dopamine D1 receptor: Recent advances in orthosteric agonism and positive allosteric modulation. J Med Chem. 2019;62(1):128‐140.
    1. Lewis MA, Hunihan L, Watson J, et al. Discovery of D1 dopamine receptor positive allosteric modulators: characterization of pharmacology and identification of residues that regulate species selectivity. J Pharmacol Exp Ther. 2015;354(3):340‐349.
    1. Gray DL, Allen JA, Mente, S , et al. Impaired beta‐arrestin recruitment and reduced desensitization by non‐catechol agonists of the D1 dopamine receptor. Nat Commun. 2018;9(1):674.
    1. Gurrell R, Duvvuri S, Sun P, et al. A phase I study of the safety, tolerability, pharmacokinetics, and pharmacodynamics of the novel dopamine D1 receptor partial agonist, PF‐06669571, in subjects with idiopathic Parkinson's disease. Clin Drug Invest. 2018;38(6):509‐517.
    1. Svensson KA, Heinz BA, Schaus JM, et al. An allosteric potentiator of the dopamine D1 receptor increases locomotor activity in human D1 knock‐in mice without causing stereotypy or tachyphylaxis. J Pharmacol Exp Ther. 2017;360(1):117‐128.
    1. Bruns RF, Mitchell SN, Wafford KA, et al. Preclinical profile of a dopamine D1 potentiator suggests therapeutic utility in neurological and psychiatric disorders. Neuropharmacology. 2018;128:351−365.
    1. Meltzer HY, Rajagopal L, Matrisciano F, Svensson, KA , Huang, M . The allosteric dopamine D1 receptor potentiator, DETQ, ameliorates subchronic phencyclidine‐induced object recognition memory deficits and enhances cortical acetylcholine efflux in male humanized D1 receptor knock‐in mice. Behav Brain Res. 2019;361:139‐150.
    1. Hao J, Beck JP, Schaus JM, et al. Synthesis and pharmacological characterization of 2‐(2,6‐Dichlorophenyl)‐1‐((1S,3R)‐5‐(3‐hydroxy‐3‐methylbutyl)‐3‐(hydroxymethyl)‐1‐methyl‐3,4‐dihydroisoquinolin‐2(1H)‐yl)ethan‐1‐one (LY3154207), a potent, subtype selective, and orally available positive allosteric modulator of the human dopamine D1 receptor. J Med Chem. 2019;62(19):8711‐8732.
    1. Smith BP, Vandenhende FR, DeSante KA, et al. Confidence interval criteria for assessment of dose proportionality. Pharm Res. 2000;17(10):1278‐1283.
    1. Jaber M, Robinson SW, Missale C, Caron MG. Dopamine receptors and brain function. Neuropharmacology. 1996;35(11):1503‐1519.
    1. Lin JS, Anaclet C, Sergeeva OA, Haas HL. The waking brain: an update. Cell Mol Life Sci. 2011;68(15):2499‐2512.
    1. Brown RE, Basheer R, McKenna JT, Strecker RE, McCarley RW. Control of sleep and wakefulness. Physiol Rev. 2012;92(3):1087‐187.
    1. Schwartz MD, Kilduff TS. The neurobiology of sleep and wakefulness. Psychiatr Clin North Am. 2015;38(4):615‐644
    1. Zeng C, Zhang M, Asico LD, Eisner GM, Jose PA. The dopaminergic system in hypertension. Clin Sci. 2007;112(12):583–597.
    1. Hahn RA, Wardell JR Jr, Sarau HM, Ridley PT. Characterization of the peripheral and central effects of SK&F 82526, a novel dopamine receptor agonist. J Pharmacol Exp Ther. 1982;223(2):305‐313.
    1. Tayebati SK, Lokhandwala MF, Amenta F. Dopamine and vascular dynamics control:present status and future perspectives. Curr Neurovasc Res. 2011;8(3):246‐257.
    1. Hughes JM, Beck TR, Rose CE Jr, Carey RM. Selective dopamine‐1 receptor stimulation produces natriuresis by a direct tubular action. J Hypertens Suppl. 1986;4(6):S106‐108.
    1. Murphy MB. Dopamine: a role in the pathogenesis and treatment of hypertension. J Hum Hypertens. 2000;14(Suppl 1):S47–S50.
    1. Marwaha A, Lokhandwala MF. Diminished natriuretic response to dopamine D1 receptor agonist, SKF‐38393 in obese Zucker rats. Clin Exp Hypertens. 2003;25(8):509‐515.
    1. Chen KC. Evidence on extracellular dopamine level in rat striatum: implications for the validity of quantitative microdialysis. J Neurochem. 2005;92(1):46‐58.
    1. Zeng C, Jose PA. Dopamine receptors: important antihypertensive counterbalance against hypertensive factors. Hypertension. 2011;57(1):11‐17.
    1. Andersen AD, Blaabjerg M, Binzer M, et al. Cerebrospinal fluid levels of catecholamines and its metabolites in Parkinson's disease: effect of l‐DOPA treatment and changes in levodopa‐induced dyskinesia. J Neurochem. 2017;141(4):614‐625.
    1. Gordan R, Gwathmey JK, Xie LH. Autonomic and endocrine control of cardiovascular function. World J Cardiol. 2015;7(4):204‐214.
    1. Stiefel G, Besag FMC. Cardiovascular effects of methylphenidate, amphetamines and atomoxetine in the treatment of attention‐deficit hyperactivity disorder. Drug Saf. 2010;33 (10):821‐842.

Source: PubMed

3
S'abonner