Herein, we report the hit-to-lead identification of a drug-like pleuromutilin conjugate , based on a triaromatic hit reported in 2020. The lead arose as the clear candidate from a hit-optimization campaign in which Gram-positive antibacterial activity, solubility, and P-gp affinity were optimized. Conjugate was extensively evaluated for its ADMET performance which, apart from solubility, was overall on par with lefamulin. This evaluation included Caco-2 cell permeability, plasma protein binding, hERG inhibition, cytotoxicity, metabolism in microsomes and CYP3A4, resistance induction, and time-kill kinetics. Intravenous pharmacokinetics of proved satisfactory in both mice and pigs; however, oral bioavailability was limited likely due to insufficient solubility. The efficacy was evaluated in mice, systemically infected with , where showed rapid reduction in blood bacteriaemia. Through our comprehensive studies, lead has emerged as a highly promising and safe antibiotic candidate for the treatment of Gram-positive bacterial infections.
Download full-text PDF |
Source |
---|---|
http://dx.doi.org/10.1021/acs.jmedchem.3c02153 | DOI Listing |
Eur J Med Chem
February 2025
Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, PR China; Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24, Uppsala, Sweden; Centre for Cancer Drug Discovery, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK. Electronic address:
The extensive bioactivity data available in public databases, such as ChEMBL, has facilitated in-depth structure-activity relationship (SAR) analysis, which are essential for understanding the impact of molecular modifications on biological activity in a comprehensive manner. A central strategy in SAR analysis is the assessment of molecular similarity. Several approaches preferred by medicinal chemists have been developed to efficiently capture structurally related compounds on a large scale.
View Article and Find Full Text PDFChem Biol Drug Des
October 2024
Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK.
The technological revolutions in computers and the advancement of high-throughput screening technologies have driven the application of artificial intelligence (AI) for faster discovery of drug molecules with more efficiency, and cost-friendly finding of hit or lead molecules. The ability of software and network frameworks to interpret molecular structures' representations and establish relationships/correlations has enabled various research teams to develop numerous AI platforms for identifying new lead molecules or discovering new targets for already established drug molecules. The prediction of biological activity, ADME properties, and toxicity parameters in early stages have reduced the chances of failure and associated costs in later clinical stages, which was observed at a high rate in the tedious, expensive, and laborious drug discovery process.
View Article and Find Full Text PDFEur J Med Chem
December 2024
Laboratório de Bioquímica de Tripanosomatídeos, Instituto Oswaldo Cruz - FIOCRUZ, Av. Brasil, 4365, Rio de Janeiro, 21040-900, Brazil. Electronic address:
Bioorg Med Chem Lett
November 2024
Prelude Therapeutics Incorporated, 175 Innovation Boulevard, Wilmington, DE 19805, USA. Electronic address:
KAT6, a histone acetyltransferase from the MYST family, has emerged as an attractive oncology target due to its role in regulating genes that control cell cycle progression and cellular senescence. Amplification of the KAT6A gene has been seen among patients with worse clinical outcome in ER breast cancers. Although multiple inhibitors have been reported, no KAT6 inhibitors have been approved to date.
View Article and Find Full Text PDFActa Crystallogr D Struct Biol
September 2024
Structural Biology, Nuvisan ICB GmbH, Muellerstrasse 178, 13353 Berlin, Germany.
A key prerequisite for the successful application of protein crystallography in drug discovery is to establish a robust crystallization system for a new drug-target protein fast enough to deliver crystal structures when the first inhibitors have been identified in the hit-finding campaign or, at the latest, in the subsequent hit-to-lead process. The first crucial step towards generating well folded proteins with a high likelihood of crystallizing is the identification of suitable truncation variants of the target protein. In some cases an optimal length variant alone is not sufficient to support crystallization and additional surface mutations need to be introduced to obtain suitable crystals.
View Article and Find Full Text PDFEnter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!