GET THE APP

Drug Discovery and In Silico Techniques: A Mini-Review
Enzyme Engineering

Enzyme Engineering
Open Access

ISSN: 2329-6674

+44 1223 790975

Mini Review - (2015) Volume 4, Issue 1

Drug Discovery and In Silico Techniques: A Mini-Review

Mohd Hassan Baig1, Khurshid Ahmad1, Mohd Adil2, Zainul A Khan3, Mohd Imran Khan4, Mohtashim Lohani1, Mohd Sajid Khan1 and Mohammad A Kamal5,6*
1Department of Biosciences, Integral University, Lucknow, India
2Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
3Department of Biosciences, Jamia Millia Islamia, New Delhi, India
4Bioinformatics Division, Institute of Cytology and Preventive Oncology (ICMR), Noida, India
5King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia
6Enzymoic, 7 Peterlee Pl, Hebersham, NSW 2770, Australia
*Corresponding Author: Mohammad A Kamal, King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia Email:

Abstract

Use of Computational (in silico) methods are widely applied in drug discovery. In drug discovery process, identification of the suitable drug target is the first and foremost task. These targets are biomolecules which mainly include DNA, RNA and proteins (such as receptors, transporters, enzymes and ion channels). Validation of such targets is necessary to exhibit a sufficient level of ‘confidence’ and to know their pharmacological relevance to the disease under investigation. The aim of this mini-review is to illustrate some of the in silico methods that are used in drug discovery, and to describe the applications of these computational methods.

Introduction

The drug discovery process involves the identification of the lead structure followed by the synthesis of its analogs, their screening to get candidate molecules for drug development [1]. The goal of the drug discovery process is to search for new drug molecules which can bind to a specific target known to be involved in causing a disease and change the target’s function [2-4].

In the traditional drug discovery process, identification of the suitable drug target is the first and foremost task [5]. These targets are biomolecules which mainly include DNA, RNA and proteins (such as receptors, transporters, enzymes and ion channels). Validation of such targets is necessary to exhibit a sufficient level of ‘confidence’ and to know their pharmacological relevance to the disease under investigation [6]. This can be performed from very basic levels such as cellular, molecular levels to whole animal level. Once the target validation has performed, effective compounds such as inhibitors, modulators or antagonists for such target need to be identified. This process is called lead identification where the design and development of a suitable assay is done to monitor the effect on the target under study [7]. High-Throughput Screening (HTS) plays a crucial role in this phase where large numbers of chemical compounds are exposed to the target. Compounds showing dose-dependent target modulation in terms of a certain degree of confidence are processed further as lead compounds. Subsequently, the experiments are performed on the animal models in the laboratories and the positive results are then optimized in terms of potency and selectivity. Physicochemical properties and their pharmacokinetic and safety features are also assessed before they become candidates for drug development [8]. Even though most of the processes depend on experimental tasks, in silico approaches are playing important roles in every stage of this drug discovery pipeline (Figure 1).

enzyme-engineering-drug-discovery-process

Figure 1: In silico drug discovery process

Drug discovery and development process

The important steps involved in the process of drug discovery are:

Lead identification

A critical factor is the search for lead structures. Leads can be obtained from natural products, structure directed molecular design, modification of natural products, biochemical understanding of the disease process and broad screening of synthetic compounds etc.

Lead optimization

It involves the identification of lead molecule through the synthesis and testing of derivatives of leads to develop Structure Activity Relationships (SARs), calculation of physico-chemical properties and using them for lead refinement from techniques like Quantitative Structure Activity Relationships (QSARs). Modern techniques such as combinatorial chemistry coupled with High Throughput Screening (HTS) provide us an enormous number of New Chemical Entities (NCEs) but these techniques have not been proved cost effective due to high costs of reagents in addition to the involvement of costly modern equipments, hence there is a strong demand for the computer assisted techniques which are fast, reliable and cheap to rationalize these early steps in drug development [9].

Pre-clinical lead development

Drug formulation experiments, in-vivo studies in animals, animal safety studies, drug metabolism studies and large scale synthesis come under the umbrella of pre-clinical lead development [10].

Clinical lead development

It involves the small scale safety and dose ranging test in healthy human volunteers (phase I) and development of clinical study protocols employing clinical investigations on patients (phase II) and comparative double blind studies on patients’ studies (phase III).

Several sequence and structure-based approaches have been proposed for target identification. In the sequence-based method, functional information about the target and its positioning in the biological networks is provided to detect unique targets from the disease causing pathogens (e.g. bacteria or viruses) by comparing functional genomics of humans with corresponding genomics of pathogens [11].

The general methodology followed for finding a novel targetspecific- drug for a given disorder involves random high-throughput screening (HTS) [8]. In this process, large chemical-libraries are checked for their capacity to block and/or modify the target. For instance, if the target is a novel beta lactamase enzyme, compounds will be screened for their ability to enhance or reduce the activity of that receptor or altogether block it [12]. In this manner, novel pharmacophores are being discovered at a significant pace nowadays.

Furthermore, an important aspect of such screenings is that the selectivity of the compounds for the target is also deduced by concurrently screening the hits against other targets. This is called as cross-screening. The significance of cross-screening lies in the fact, that in case the compound hits frequently at non-related targets; it is expected to cause significant ‘toxicity’ in the human body (in case it reaches to the stage of human clinical trials) [3].

At this juncture, it is noteworthy to mention about another screening method referred to as Virtual High Throughput Screening (vHTS). In this type of screening method, computer-generated models are used. These computer programs attempt to dock virtual libraries consisting of 3-D structures of chemicals to a target. This method is quick, reliable and economic as well [13-15].

Through these methods the scientists discover ‘a lead molecule series’. The chemical structures included in this series are expected to possess adequate target specificity and favorable properties for ‘druglikeness’. Following this, one or two compounds may then be selected for drug development. The best compound of these is termed as the "lead" compound [14,15]. The lead compound is further optimized by random trials or more judiciously by rational drug design methods. The leads are examined for whether these compounds share some common features, chemists/modelers then use Structure-Activity Relationships (SARs) to improve certain structural features of the lead molecules in order to (i) increase activity against the chosen target, (ii) reduce activity against unrelated targets, and (iii) improve the "druglike" or ADME properties of the molecule.

A major target in the drug discovery process is to develop a high degree of “rationality.” This would represent an intellectually rigorous approach incorporating Computer-Assisted Molecular Design (CAMD), limited but highly focused chemical synthetic effort, and sophisticated biological assays. Thus, the drug design process may become significantly less risky and more resource efficient [13].

The rational drug design approaches are maturing in the sense that they are moving from being only a theoretical promise based on their use in the retrospective analysis of known compounds, to innovative design and discovery of Novel Chemical Entities (NCEs) and providing solutions to novel problems. There are a number of successful examples in the literature using rational design approaches for design and discovery of NCEs [16-18]. The advances in computer hardware and software have made this process more effective and time efficient. The systematic evolution of drug discovery research is outlined in Table 1.

Time Materials Test systems
Ancient time Plants, venoms, minerals (Natural Products) Humans
-1806 Morphine  
- 1850 Chemicals  
- 1890 synthetics, dyes animals
- 1920 synthetics, dyes animals, isolated organs
- 1970 synthetics, dyes enzymes, membranes
- 1990 Combinatorial libraries Human proteins, HTS
- 2000 onwards Focused libraries CAMD, vHTS, virtual screening

Table 1: The Evolution of Drug Research

Conclusion

In silico techniques significantly contribute to early drug discovery and are important in target and lead discovery. It can be anticipated that the contribution of in silico techniques in drug discovery will increase substantially in the future. Ultimately, to have a much broader impact, the use of in silico methods will need to become a part of every researcher involved in drug discovery. The need for timely adaptation and application of in silico approaches in pharmaceutical research has clearly been recognized and is expected to improve further the overall efficiency of drug discovery.

References

  1. Khan MS, Baig MH, Ahmad S, Siddiqui SA, Srivastava AK, et al.(2013) Design, Synthesis, Evaluation and Thermodynamics of 1-Substituted Pyridylimidazo [1,5-a] Pyridine Derivatives as Cysteine Protease Inhibitors. PloS one 8: e69982.
  2. Khan, AU, Baig MH, Wadhwa G (2011). Molecular docking analysis of new generation cephalosporins interactions with recently known SHV-variants. Bioinformation, 5(8):331-5.
  3. Shan Y, Kim ET, Eastwood MP, Dror RO, Seeliger M A, Shaw DE (2011). How does a drug molecule find its target binding site?. Journal of the American Chemical Society 133(24): 9181-9183.
  4. Merad M, Soufi W, Ghalem S, Boukli F, Baig MH, et al. (2014). Molecular Interaction of Acetylcholinesterase with Carnosic Acid Derivatives: A Neuroinformatics Study. CNS & neurological disorders drug targets. 13(3), 440-446.
  5. Terstappen G, Reggiani A (2001). In silico research in drug discovery. Trends in pharmacological sciences 22(1): 23-26.
  6. Blake RA (2007). Target validation in drug discovery. Methods Molecular Biology 356:367-77.
  7. Ekins S, Freundlich JS, Hobrath JV, White EL, Reynolds RC (2014). Combining computational methods for hit to lead optimization in Mycobacterium tuberculosis drug discovery. Pharmaceutical research 31(2): 414-435.
  8. Smith C (2003) Drug target validation: Hitting the target. Nature 422: 341-345.
  9. Green DV, Segall M (2014). CHEMOINFORMATICS IN LEAD OPTIMIZATION. Chemoinformatics for Drug Discovery 149-178.
  10. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ (2012) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advanced drug delivery reviews 64: 4-17.
  11. Vacca J P, Condra J H (1997) Clinicaly effective HIV-1 protease inhibitors. Drug Discovery Today 2: 261-72.
  12. Baig MH, Shakil S, Khan AU (2012). Homology modeling and docking study of recent SHV type ß-lactamses with traditional and novel inhibitors: an in silico approach to combat problem of multiple drug resistance in various infections. Medicinal Chemistry Research 21(9): 2229-2237.
  13. Doman TN, McGovern SL, Witherbee BJ, Kasten TP, Kurumbail R, et al. (2002) Molecular docking and high-throughput screening for novel inhibitors of protein tyrosine phosphatase-1B. Journal of Medicinal Chemistry 45: 2213-2221.
  14. Danishuddin M, Khan A, Faheem M, Kalaiarasan P, Hassan Baig M, et al. (2013). Structure-based screening of inhibitors against KPC-2: designing potential drug candidates against multidrug-resistant bacteria. Journal of Biomolecular Structure and Dynamics 32(5):741-50
  15. Baig MH, Danishuddin M, Khan S, Khan AU. (2012). Screening of inhibitors for S130G inhibitor resistant mutants of TEM type beta-lactamase. Bioinformation 8(24): 1225.
  16. Laskowski RA, Hutchinson EG, Michie AD, Wallace AC, Jones ML, et al. (1997) PDBsum: a Web-based database of summaries and analyses of all PDB structures. Trends Biochemical Sciences 22: 488-490.
  17. Traxler P M (1997) Protein tyrosine kinase inhibitors in cancer treatment. Expert Opinion on Therapeutic Patents 7: 571-88.
  18. Brady SF, Stauffer KJ, Lumma WC, Smith GM, Ramjit HG, et al. (1998) Discovery and development of the novel potent orally active thrombin inhibitor N-(9-hydroxy-9-fluorenecarboxy) prolyl trans-4-aminocyclohexylmethyl amide (L-372,460): coapplication of structure-based design and rapid multiple analogue synthesis on solid support. Journal of Medicinal Chemistry 41: 401-406.
Citation: Baig MH, Ahmad K, Adil M, Khan ZA, Khan MI, et al. (2015) Drug Discovery and In Silico Techniques: A Mini-Review. Enz Eng 4:123.

Copyright: © 2015 Baig MH, et al. 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.
Top