Skip to main content
U.S. flag

An official website of the United States government

Glutaminyl cyclase inhibitor PQ912 improves cognition in mouse models of Alzheimer's disease-studies on relation to effective target occupancy

Bibliographic

Year of Publication:
2017
Contact PI Name:
Inge Lues
Contact PI Affiliation:
Probiodrug AG, Weinbergweg, Halle, Germany
Co-Authors:
Torsten Hoffmann, Antje Meyer, Ulrich Heiser, Stephan Kurat, Livia Böhme, Martin Kleinschmidt, Karl-Ulrich Bühring, Birgit Hutter-Paier, Martina Farcher, Hans-Ulrich Demuth, Stephan Schilling
Primary Reference (PubMED ID):
Funding Source:
Not Reported
Study Goal and Principal Findings:

Numerous studies suggest that the majority of amyloid-β (Aβ) peptides deposited in Alzheimer's disease (AD) are truncated and post-translationally modified at the N terminus. Among these modified species, pyroglutamyl-Aβ (pE-Aβ, including N3pE-Aβ40/42 and N11pE-Aβ40/42) has been identified as particularly neurotoxic. The N-terminal modification renders the peptide hydrophobic, accelerates formation of oligomers, and reduces degradation by peptidases, leading ultimately to the accumulation of the peptide and progression of AD. It has been shown that the formation of pyroglutamyl residues is catalyzed by glutaminyl cyclase (QC). Here, they presented data about the pharmacological in vitro and in vivo efficacy of the QC inhibitor (S)-1-(1H-benzo[d]imidazol-5-yl)-5-(4-propoxyphenyl)imidazolidin-2-one (PQ912), the first-in-class compound that is in clinical development. PQ912 inhibits human, rat, and mouse QC activity, with Ki values ranging between 20 and 65 nM. Chronic oral treatment of hAPPSLxhQC double-transgenic mice with approximately 200 mg/kg/day via chow shows a significant reduction of pE-Aβ levels and concomitant improvement of spatial learning in a Morris water maze test paradigm. This dose results in a brain and cerebrospinal fluid concentration of PQ912 which relates to a QC target occupancy of about 60%. Thus, was concluded that >50% inhibition of QC activity in the brain leads to robust treatment effects. Secondary pharmacology experiments in mice indicate a fairly large potency difference for Aβ cyclization compared with cyclization of physiologic substrates, suggesting a robust therapeutic window in humans. This information constitutes an important translational guidance for predicting the therapeutic dose range in clinical studies with PQ912.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Small Molecule
Therapeutic Agent:
PQ912
Therapeutic Target:
Glutaminyl Cyclase (QC)

Animal Model

Model Information:
Species:
Mouse
Model Type:
APPxQC
Strain/Genetic Background:
Not Reported
Species:
Mouse
Model Type:
APPxPS1xhQC
Strain/Genetic Background:
Not Reported

Experimental Design

Is the following information reported in the study?:
Power/Sample Size Calculation
Randomized into Groups
Blinded for Treatment
Blinded for Outcome Measures
Pharmacokinetic Measures
Pharmacodynamic Measures
Toxicology Measures
ADME Measures
Biomarkers
Dose
Formulation
Route of Delivery
Duration of Treatment
Frequency of Administration
Age of Animal at the Beginning of Treatment
Age of Animal at the End of Treatment
Sex as a Biological Variable
Study Balanced for Sex as a Biological Variable
Number of Premature Deaths
Number of Excluded Animals
Statistical Plan
Genetic Background
Inclusion/Exclusion Criteria Included
Conflict of Interest

Outcomes

Outcome Measured
Outcome Parameters
Behavioral
Morris Water Maze
Biochemical
Binding Affinity Measurements
Brain-beta Amyloid Peptide-Total
Brain-beta Amyloid Peptide 42
Testosterone
Thyroid Stimulating Hormone (TSH)
Thyroxine (T4)
Pyroglutamate Modified beta Amyloid Peptides
Cell Biology
beta Amyloid Peptides
Pharmacokinetics
Drug Concentration-CSF
Drug Concentration-Brain
Free Drug Concentration-Brain
Pharmacodynamics
Target Engagement (Reduction N3pE-beta Amyloid-Brain)
Toxicology
Body Weight
Food Intake
Hypothyroidism
Glucose Concentration
General Behavior
General Health
Pharmacology
Binding Affinity
Target Selectivity