Abstract

The present work exploits the potential of in silico approaches for minimizing attrition of leads in the later stages of drug development. We propose a theoretical approach, wherein ‘parallel’ information is generated to simultaneously optimize the pharmacokinetics (PK) and pharmacodynamics (PD) of lead candidates. β-blockers, though in use for many years, have suboptimal PKs; hence are an ideal test series for the ‘parallel progression approach’. This approach utilizes molecular modeling tools viz. hologram quantitative structure activity relationships, homology modeling, docking, predictive metabolism, and toxicity models. Validated models have been developed for PK parameters such as volume of distribution (log Vd) and clearance (log Cl), which together influence the half-life (t1/2) of a drug. Simultaneously, models for PD in terms of inhibition constant pKi have been developed. Thus, PK and PD properties of β-blockers were concurrently analyzed and after iterative cycling, modifications were proposed that lead to compounds with optimized PK and PD. We report some of the resultant re-engineered β-blockers with improved half-lives and pKi values comparable with marketed β-blockers. These were further analyzed by the docking studies to evaluate their binding poses. Finally, metabolic and toxicological assessment of these molecules was done through in silico methods. The strategy proposed herein has potential universal applicability, and can be used in any drug discovery scenario; provided that the data used is consistent in terms of experimental conditions, endpoints, and methods employed. Thus the ‘parallel progression approach’ helps to simultaneously fine-tune various properties of the drug and would be an invaluable tool during the drug development process.

Highlights

  • The pharmacological properties of a drug depend on its biological affinity at the receptor sites, and on its ADMET profile

  • This study reveals that the re-engineered β-blockers are predicted to be nontoxic for the endocrine receptor and the hERG K+ channel

  • The hologram quantitative structure activity relationships (HQSAR) tool was selected for this task

Read more

Summary

Introduction

The pharmacological properties of a drug depend on its biological affinity at the receptor sites, and on its ADMET profile. We have applied a fusion of methodologies to yield a ‘parallel progression approach’ to simultaneously optimize the pharmacokinetics (PK) and pharmacodynamics (PD) using structural information, which limits expending valuable resources and time while awaiting the outcome of iterative experimental evaluations. The advantage of this approach is that the modified molecules can be further evaluated for PK and PD, and during each progressive cycle further modifications can be made for lead optimization. Other studies have reported lipophilicity, solubility descriptors, polarity, and molecular size descriptors to be important parameters (del Amo et al, 2013; Karalis, Tsantili-Kakoulidou, & Macheras, 2002)

Objectives
Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.