GET THE APP

A Guide for in Bioinformatic Drug Design
..

Medicinal Chemistry

ISSN: 2161-0444

Open Access

Perspective - (2022) Volume 12, Issue 10

A Guide for in Bioinformatic Drug Design

Yiqun Lai*
*Correspondence: Yiqun Lai, Department of Pharmaceutical Sciences, University of Oklahoma, Norman, USA, Email:
Department of Pharmaceutical Sciences, University of Oklahoma, Norman, USA

Received: 03-Oct-2022, Manuscript No. mccr-22-84890; Editor assigned: 05-Oct-2022, Pre QC No. P-84890; Reviewed: 17-Oct-2022, QC No. Q-84890; Revised: 21-Oct-2022, Manuscript No. R-84890; Published: 27-Oct-2022 , DOI: 10.37421/2161-0444.2022.12.647
Citation: Lai, Yiqun. “A Guide for in Bioinformatic Drug Design." J Med Chem 12 (2022): 647.
Copyright: © 2022 Lai Y. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Introduction

The drug discovery process is a difficult one, with few candidates making it from hit compound to commercially available product, often due to factors such as poor binding affinity, off-target effects, or physicochemical properties such as solubility or stability. This process is further complicated by high costs and time constraints. As a result, it is critical to optimise each step of the process in order to maximise the chances of success. As a result of recent advances in computer power and technology, computer-aided drug design has become an essential component of modern drug discovery, guiding and accelerating the process [1].

Description

New drugs with improved efficacy and lower toxicity are always in high demand; however, the process of drug discovery and development is costly, time-consuming, and fraught with difficulties. Aside from the pitfalls of target validation and hit identification, clinical trials frequently fail due to poor pharmacokinetics, efficacy, and toxicity. Wong, et al. found that the probability of success for all drugs (marketed and in development) was only 13.8% in a study that examined 406,038 trials from January 2000 to October 2015. Based on data from 106 randomly selected new drugs developed by 10 pharmaceutical companies, One possible explanation for the rise in R&D costs is that regulators, such as the FDA, have become more risk-averse, tightening safety requirements, resulting in higher trial failure rates and higher drug development costs. To maximise the chances of success, it is critical to optimise every aspect of the R&D process.

To summarise, drug targets can be identified using techniques such as data mining, phenotype screening, and bioinformatics. After that, potential targets must be validated to see if they are rate limiting for disease progression or induction. Establishing a strong link between the target and the disease increases confidence in the scientific hypothesis, resulting in greater success and efficiency later in the drug discovery process. Once the targets have been identified and validated, compound screening assays are used to find new hit compounds (hit-to-lead). This screening can employ a variety of strategies involving physical methods such as mass spectroscopy, fragment screening, nuclear magnetic resonance screening, and DNA encoded chemical libraries [2,3].

Following the identification of hit compounds, properties such as absorption, distribution, metabolism, excretion, and toxicity should be considered and optimised early in the drug discovery process. One of the obstacles that frequently leads to clinical trial failure is a drug candidate's unfavourable pharmacokinetic and toxicity profile. Despite the fact that physical and computational screening techniques are distinct in nature, they are frequently integrated in the drug discovery process to complement one another and maximise the potential of the screening results. This information and knowledge is used in computer-aided drug design to screen for new drug candidates. CADD has proven to be a tool that reduces the time and resources required in the drug discovery pipeline as technology and computer power have advanced in recent years.

A protein's functionality is determined by its structure, and structure-based drug design is based on 3D structural information obtained from experimental methods such as X-ray crystallography, NMR spectroscopy, and cryo-electron microscopy. SBDD aims to predict the Gibbs free energy of binding, as well as the binding affinity of ligands to the binding site, by simulating their interactions. Molecular dynamics simulations, molecular docking, fragment-based docking, and de novo drug design are some examples of SBDD.

The protein sequence is first obtained, either experimentally or from databases such as the Universal Protein Resource, and then modelling templates with high sequence similarity and resolution are identified by performing a blast search against the Protein Data Bank. Because protein functions are primarily determined by structural arrangement rather than amino acid sequence, using profile-based methods to identify patterns of residue conservation can be more useful and accurate than simply comparing raw sequences. One of the most significant limitations of homology modelling is its reliance on the availability of suitable templates and accurate sequence alignment. A high degree of sequence identity between the query protein and the template protein typically increases confidence in the homology model. Generally, a minimum of 30% sequence identity is considered to be a threshold for successful homology modelling, as approximately 20% of the residues are expected to be misaligned for sequence identities below 30%, leading to poor homology model.

Although all of the components in a polyurethane formulation are likely to influence the above responses, reducing the number of experimental factors reduces the number of experiments required to explore the experimental space and allows for more detailed modelling of responses with an equal number of formulations. Only the amine catalysts and surfactants were chosen as factors to reduce the number of experiments. PU foam catalysts are essential in the production of polyurethane foams. It has been demonstrated that changing the catalyst compositions and loading alters the relative rates of the blowing and gelling reactions, which has a significant impact on PU foam properties.

Historically, the homology modelling approach has been the "go-to" method for protein structure prediction because it is less computationally expensive and produces more accurate predictions. One of the most significant limitations is that it is based on previously known structures, making prediction of more complex targets, such as membrane proteins with little known structural data, nearly impossible. Another solution to this problem is to use a template-free approach, also known as ab initio modelling, free modelling, or de novo modelling.This method, as the name suggests, predicts a protein structure from amino acid sequences without the use of a template. Furthermore, the ab initio approach can model protein complexes and provide information on complex formation and protein-protein interaction [4,5].

Conclusion

Recent advances in computational software and hardware have transformed the use of in silico methods in drug design, with access to highperformance computers enabling the processing of more complex calculations and larger data sets. We have highlighted a variety of in silico methods that are commonly used in the hit identification and lead optimization stages of the drug design process in this review; however, computational methods are also used in other areas of the pipeline. Drug repurposing, protein-protein docking, de novo protein design, inverse docking, adverse event prediction, physiologically-based pharmacokinetic modelling, and guiding chemical synthesis are some examples.

References

  1. Wouters, Olivier J., Martin McKee and Jeroen Luyten. "Estimated research and development investment needed to bring a new medicine to market, 2009-2018." Jama 3232020): 844-853.
  2. Google Scholar, Crossref, Indexed at

  3. Wong, Chi Heem, Kien Wei Siah and Andrew W. Lo. "Estimation of clinical trial success rates and related parameters." Biostatistics 20 (2019): 273-286.
  4. Google Scholar, Crossref, Indexed at

  5. DiMasi, Joseph A., Ronald W. Hansen and Henry G. Grabowski. "The price of innovation: New estimates of drug development costs." J Health Econ 22 (2003): 151-185.
  6. Google Scholar, Crossref, Indexed at

  7. Dalvit, Claudio. "NMR methods in fragment screening: Theory and a comparison with other biophysical techniques." Drug Discov Today14 (2009): 1051-1057.
  8. Google Scholar, Crossref, Indexed at

  9. Shoichet, Brian K., Susan L. McGovern, Binqing Wei and John J. Irwin, et al. "Lead discovery using molecular docking." Curr Opin Chem Biol 6 (2002): 439-446.
  10. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 6627

Medicinal Chemistry received 6627 citations as per Google Scholar report

Medicinal Chemistry peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward