Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Clinical Trial
. 2021 Mar;35(3):331-344.
doi: 10.1007/s40263-021-00797-x. Epub 2021 Mar 23.

First-in-Human Randomized Trial to Assess Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of the KDM1A Inhibitor Vafidemstat

Affiliations
Clinical Trial

First-in-Human Randomized Trial to Assess Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of the KDM1A Inhibitor Vafidemstat

Rosa María Antonijoan et al. CNS Drugs. 2021 Mar.

Abstract

Background: Vafidemstat, an inhibitor of the histone lysine-specific demethylase KDM1A, corrects cognition deficits and behavior alterations in rodent models. Here, we report the results from the first-in-human trial of vafidemstat in healthy young and older adult volunteers. A total of 110 volunteers participated: 87 were treated with vafidemstat and 23 with placebo.

Objectives: The study aimed to determine the safety and tolerability of vafidemstat, to characterize its pharmacokinetic and pharmacodynamic profiles, to assess its central nervous system (CNS) exposure, and to acquire the necessary data to select the appropriate doses for long-term treatment of patients with CNS disease in phase II trials.

Methods: This single-center, randomized, double-blind, placebo-controlled phase I trial included a single and 5-day repeated dose-escalation and open-label CNS penetration substudy. Primary outcomes were safety and tolerability; secondary outcomes included analysis of the pharmacokinetics and pharmacodynamics, including chemoprobe-based immune analysis of KDM1A target engagement (TE) in peripheral blood mononuclear cells (PBMCs) and platelet monoamine oxidase B (MAOB) inhibition. CNS and cognitive function were also evaluated.

Results: No severe adverse events (AEs) were reported in the dose-escalation stage. AEs were reported at all dose levels; none were dose dependent, and no significant differences were observed between active treatment and placebo. Biochemistry, urinalysis, vital signs, electrocardiogram, and hematology did not change significantly with dose escalation, with the exception of a transient reduction of platelet counts in an extra dose level incorporated for that purpose. Vafidemstat exhibits rapid oral absorption, approximate dose-proportional exposures, and moderate systemic accumulation after 5 days of treatment. The cerebrospinal fluid-to-plasma unbound ratio demonstrated CNS penetration. Vafidemstat bound KDM1A in PBMCs in a dose-dependent manner. No MAOB inhibition was detected. Vafidemstat did not affect the CNS or cognitive function.

Conclusions: Vafidemstat displayed good safety and tolerability. This phase I trial confirmed KDM1A TE and CNS penetration and permitted characterization of platelet dynamics and selection of phase IIa doses.

Trial registration: EUDRACT No. 2015-003721-33, filed 30 October 2015.

PubMed Disclaimer

Conflict of interest statement

RMA has received personal fees from Centre d’Investigació del Medicament, Institut de Recerca de l’Hospital de la Santa Creu© Sant Pau (IIB-Sant Pau) and Hospital de la Santa Creu i Sant Pau, Barcelona, Spain. JMF-C, JC, MP, and JM-C have received personal fees from Centre d’Investigació del Medicament, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau (IIB-Sant Pau), Barcelona, Spain. MIA, CMa, CMo, CB, and TM have received personal fees from Oryzon Genomics, during the conduct of the study. In addition, CMa is listed as inventor on patent application WO2017/158136 (pending), assigned to Oryzon Genomics; CMo owns company shares on the stock exchange (Oryzon Genomics); and CB reports patent WO2012/013728 (issued in multiple countries), assigned to Oryzon Genomics. CB is executive director and shareholder of Oryzon Genomics. TM has received other compensation from the Alzheimer’s Drug Discovery Foundation, outside the submitted work; is listed as inventor in WO2017/158136 (pending), WO2017/212061 (issued in multiple countries), and WO2019/025588 (pending) and further reports patent WO2012/013728 (issued in multiple countries), all assigned to Oryzon Genomics; is executive director and shareholder of Oryzon Genomics; and is a member of the Scientific Review Board of the Alzheimer’s Drug Discovery Foundation, for which no compensation is received.

Figures

Fig. 1
Fig. 1
Clinical trial design. a Chemical structure of vafidemstat. b Impact of vafidemstat on platelet levels in nonclinical species, represented as % inhibition compared with control animals. All values are expressed as mg/m2/day to allow for direct comparison. Differences in administration (once daily by oral gavage in rats and dogs vs. drinking water in mice) were accounted for. Doses in mg/kg/day were converted to mg/m2/day by multiplication with the Km factor for each species (3, 6, 20, 37 for mouse, rat, dog, human, respectively). Black triangles indicate the SAMP8 efficacy studies in mouse; gray circles indicate the 28-day toxicity in rats; white squares indicate the 28-day toxicity in dogs; black arrows represent the doses chosen for the SAD and MAD cohorts; gray arrows represent those for the CSF cohorts in the phase I trial. The maximum recommended starting dose was 0.2 mg. c Clinical trial doses per cohort. d Study design of the SAD, MAD, and CSF cohorts. Dark gray rectangle indicates the sentinel subject, light gray rectangles indicate subjects in active treatment; white rectangles indicate subjects receiving placebo. CSF cerebrospinal fluid, MAD multiple ascending dose, p.o. oral administration, PLT platelet, QD once daily, SAD single ascending dose
Fig. 2
Fig. 2
Hematological impact of vafidemstat. Impact of vafidemstat on platelet levels at a 1.5 mg/day, b 2.5 mg/day, and (c) the extra dose level of 4 mg/day, represented as % change from baseline levels. Black dots indicate vafidemstat (volunteers analyzed n = 6 in [a/b] or n = 3 in [c]); open circles indicate placebo treatment (volunteers analyzed n = 2 in [a/b] or n = 1 in [c]); black arrows represent dosing occasions. Data are presented as means and standard error of the mean. CFB change from baseline
Fig. 3
Fig. 3
Vafidemstat pharmacokinetics. (a) Plasma concentrations after single and (b) multiple ascending doses in healthy young (full lines) and older adult (dotted line) subjects. Light blue circles 0.2 mg; white squares 0.6 mg; light blue triangles 1 mg; white triangles 1.5 mg; blue diamonds 2.5 mg; white circles 4 mg. Data are presented as mean and standard error of the mean. Volunteers analyzed at each datapoint, n = 6 except for MAD 4 mg, n = 3. (c) Cerebrospinal fluid (full lines) and plasma (unbound, dotted lines) concentrations after a single dose of 2 mg (diamonds) or 4 mg (circles)
Fig. 4
Fig. 4
Vafidemstat pharmacodynamics. a The KDM1A chemoprobe OG-881 used to capture uninhibited protein. b Graphical representation of the chemoprobe and sandwich ELISA used to determine free and total KDM1A. c KDM1A TE after SAD and d MAD in healthy young subjects. Light blue dots 12 h; medium blue dots 108 h; dark blue dots 72 h (SAD) or 192 h (MAD) after the first dose. Data are presented as mean and standard error of the mean. Volunteers sampled at each dose: n = 6 except MAD 4 mg, n = 3. e, f Pharmacokinetic/pharmacodynamic relationship between exposure, as reflected by Cmax,ss or AUCss, and KDM1A TE. g, h Pharmacokinetic/pharmacodynamic relationship between exposure, as reflected by Cmax,ss or AUCss, and platelet dynamics (represented as % CFB levels). All values correspond to independent subjects. Calculated EC50/EC80 values are also shown in the respective graphs. Light blue circles 0.2 mg; white squares 0.6 mg; light blue triangles 1 mg; white triangles 1.5 mg; blue diamonds 2.5 mg; white circles 4 mg; full lines nonlinear fit; dotted lines 95% confidence interval. Volunteers sampled at each dose: n = 6 except MAD 4 mg, n = 3. Samples that did not pass quality control were omitted. AUC,ss area under the plasma concentration–time curve at steady state, CFB change from baseline, Cmax,ss maximum concentration at steady state, EC50 half maximal effective concentration, EC80 80% maximal effective concentration, ELISA enzyme-linked immunosorbent assay, HRP horseradish peroxidase, MAD multiple ascending dose, PBMCs peripheral blood mononuclear cells, SAD single ascending dose, SS steady state, TE target engagement

References

    1. Barter JD, Foster TC. Aging in the brain: new roles of epigenetics in cognitive decline. Neuroscientist. 2018;24(5):516–525. doi: 10.1177/1073858418780971. - DOI - PubMed
    1. Wood IC. The contribution and therapeutic potential of epigenetic modifications in Alzheime’s disease. Front Neurosci. 2018;12:649. doi: 10.3389/fnins.2018.00649. - DOI - PMC - PubMed
    1. Collins BE, Greer CB, Coleman BC. Histone H3 lysine K4 methylation and its role in learning and memory. Epigenet Chromatin. 2019;12(1):7. doi: 10.1186/s13072-018-0251-8. - DOI - PMC - PubMed
    1. Lewis CR, Candace R, Lewis CR, Henderson-Smith A, Reagan S, Breitenstein RS, et al. Dopaminergic gene methylation is associated with cognitive performance in a childhood monozygotic twin study. Epigenetics. 2019;14(3):310–323. doi: 10.1080/15592294.2019.1583032. - DOI - PMC - PubMed
    1. Delgado-Morales R, Agís-Balboa RC, Esteller M. Epigenetic mechanisms during ageing and neurogenesis as novel therapeutic avenues in human brain disorders. Clin Epigenet. 2017;9:67. doi: 10.1186/s13148-017-0365-z. - DOI - PMC - PubMed

Publication types

MeSH terms

Associated data

LinkOut - more resources