ChapterPDF Available

Discovery and optimization of lead molecules in drug designing

Authors:

Abstract

During this decade, increasing the demand for drugs with higher specificity and efficacy generates new challenges with drug designing. Lead molecule discovery is associated with the development of novel pharmacological active candidates with therapeutical applicability. The first step involves the determination of chemical structure and their modification, which requires a better fit in the target site to improve their structure affinity relationship with higher efficiency, potency, efficacy, selectivity, thermodynamic and kinetic parameters, and their pharmaceutical properties. Computational drug designing plays a pivotal role in the discovery of novel lead molecules with their structural, functional, and metabolic information. It involves target site identification, hit identification, and hit-to-lead-to-candidate optimization. After these steps, preclinical and clinical trials are applied. Optimization of lead improves the functioning of the developed lead molecule with respect to structure modification, binding affinity, target to lead interaction, toxicity, thermodynamic and pharmacokinetics parameters. However, in silico tools give a new direction and approach to the pharmaceutical industry.
Chapter 16
Discovery and optimization of lead
molecules in drug designing
Shivani Verma
1
and Rajesh Kumar Pathak
2
1
Department of Chemistry, College of Basic Sciences & Humanities, G. B. Pant University of Agriculture and Technology, Pantnagar, India,
2
School
of Agricultural Biotechnology, Punjab Agricultural University, Ludhiana, India
16.1 Introduction
In 1948, the World Health Organization defined health as the complete status of physical, mental, and social well-being
status, not only the absence of disease or infirmity (Sartorius, 2006). The abnormal condition displayed by a group of
living organisms in association to represent a specified disorder or abnormalities of structure and function is called dis-
ease (Scully, 2004). Drug molecules are used to overcome the abnormal condition or function of an organism to the
action on their particular target site. The discovery of drugs creates a driving force for the medicinal chemist to design
new pharmaceutical active ingredients with therapeutic properties. In this whole scenario, pharmaceutical industry plays
a crucial role in new drug development with understanding their biological chemistry (Durrant, 2001; Khanna, 2012;
Mignani, Huber, Tomas, Rodrigues, & Majoral, 2016; Taylor, 2015). The discovery and development of a drug is a
very vast, intense, time-consuming, and interdisciplinary area. The efficiency, specificity, and efficacy of drugs depend
on their site of action, metabolic pathway, and pharmacokinetics parameters. Moreover, many drugs are failed due to
less efficacy, poor selectivity, and unsuitable pharmacokinetics (Pajouhesh & Lenz, 2005). This chapter describes the
role of computer-aided drug design (CADD) in lead molecule discovery and strategies of optimization in drug discovery
processes. It also focused on different types of natural, synthetic, and semisynthetic lead molecules to design and create
novelty for the effective and efficient drug library.
Conventional methods for drug discovery involve lead molecule identification, synthesis, and characterization to
see the relevance between target sites and pharmaceutical action. It also involves clinical trials and approval. These
processes take much time and cost for the synthesis of many compounds and experimental validation. Resolving these
factors, the movement toward in silico chemistry and molecular modeling for computational drug designing are inter-
esting approaches for the pharmaceutical industry (Rai, Pathak, Singh, Bhatt, & Baunthiyal, 2021; Wadood et al.,
2013). CADD attracts the main attention due to their higher selectivity, efficiency, efficacy, lesser time-consuming
process, lesser toxicity, and better to fit with different pharmacokinetics parameters (Makrynitsa, Lykouras, Spyroulia,
& Matsoukas, 2018). The balance between their pharmaceutical chemistry and biological activity is the essential crite-
ria for effective drug designing. With increasing the demand for drugs, the selection of biological active structure
pays much attention. With today’s increasing knowledge of structural biology and the power of computers; it becomes
possible to use computational methods for the identification of active libraries to resolve the molecular obesity
problem.
CADD technology helps to reduce the number of ligands for experimental assay via virtual screening (Pathak,
Gupta, Shukla, & Baunthiyal, 2018; Tropsha & Bajorath, 2016). The high potential of virtual screening toward CADD
gives an immense role in the finding of novel therapeutics molecules. It involves the investigation of the entire library
based on scientific and literature knowledge to select a small set of active molecules. This leads to Hit-to-Lead mole-
cule discovery. It also helps to decrease the molecular obesity for the discovery of novel biologically active lead mole-
cules (Grzybowski, Ishchenko, Shimada, & Shakhnovich, 2002; Sliwoski, Kothiwale, Meiler, & Lowe, 2014). Fig. 16.1
shows the steps involved in the computational drug discovery process.
253
Bioinformatics. DOI: https://doi.org/10.1016/B978-0-323-89775-4.00004-3
©2022 Elsevier Inc. All rights reserved.
16.2 Principles of CADD
The CADD methods are depending on bioinformatics tools and databases and are based on structure- and ligand-based
drug discovery approaches using computational and experimental techniques. It plays a vital role in lead identification
and optimization, which leads to the discovery of novel molecules in very little time as compared to traditional methods
(Fig. 16.2).
We use the structure-based drug designing (SBDD) approach when the structure of the target protein is available in
PDB, or it can be model using sequence information (Schmidt, Bergner, & Schwede, 2014). In SBDD, novel small
molecules are designed based on the 3D structure of the target binding site, and the binding affinity or interaction ener-
gies of ligands with the binding site are also kept in mind. The aim of SBDD involves molecular docking and binding
affinity parameters (Anderson, 2003). First, molecular docking gives information about the orientation and geometry of
both partners in which they coordinate in better form. Second, it gives the information regarding their binding para-
meters in which they bind efficiently to each other with noncovalent interaction, that is, H-bonding and hydrophobic
bonding (Zoete, Grosdidier, & Michielin, 2009). Several computational programs, such as AutoDock, MOE, Discovery
Studio, and Glide, are used for docking study (Morris et al., 2009; Trott & Olson, 2010). These software provide an
estimated value of binding-free energy, which presents in the form of a score. The concept of drug designing is comple-
mentary to protein structure in terms of their geometry and distribution of charge. Recognition of efficient binding
FIGURE 16.1 Process of drug discovery.
FIGURE 16.2 Computer-aided drug design techniques used in lead discovery and optimization.
254 Bioinformatics
affinity between two parties gives the better route for in silico designing and selection of drug libraries virtually. The
designed combination should have improved drug and pharmacokinetics, and pharmacodynamics, that is, absorption dis-
tribution, metabolism, excretion, and toxicity (ADMET) (Jorgensen, 2010). The SBDD involves protein structure determi-
nation, comparative modeling, binding pocket identification, scoring function, knowledge-based scoring, force-based
scoring function, proteinligand docking algorithms, and structure-based virtual screening visualization of pro-
teinligand interaction diagram (Grant, 2009; Leelananda & Lindert, 2016; Pathak, Baunthiyal, Taj, & Kumar, 2014).
Saquinavir, amprenavir, dorzolamide, and ibuprofen drugs were developed based on SBDD (Jorgensen, 2004)(Fig. 16.3).
Ligand-based designing approaches in the identification of small molecule, which binds to target site, efficiently
play a key role in drug discovery. It is the alternative to SBDD. In the absence of knowledge of target structure, the
ligand-based approaches are helpful. It involves pharmacophore modeling, QSAR (quantitative structureactivity rela-
tionship) and molecular similarity approaches (Acharya, Coop, Polli, & MacKerell, 2011). A pharmacophore is a small
chemical entity that exhibits pharmaceutical activity. Pharmacophore modeling involves the study of small lead mole-
cule interaction with the target site. These molecules are designed based on the hydrogen bond donor/acceptor capabil-
ity, a residue of acid/base, and flexibility with the target pocket (Lin, 2000). It consists of the following processes: (1)
based on database knowledge, identifying the lead molecule having the same type of interaction with target site; (2)
designing of 2D and 3D structure of ligand; (3) building of pharmacophore model (4) based on the score giving the
ranking of designed molecules and select the best structure which interacts with 3D structure of target protein; and (5)
validating the molecule for further process (Mason, Good, & Martin, 2001). Norfloxacin, zolmitriptan, and losartan are
the FDA-approved drugs that were designed via ligand-based drug discovery (Clark, 2006)(Fig. 16.4).
16.2.1 Case study: inhibition of lipoxygenases
Lipoxygenases (LOXs) (EC 1.13.11.12) are oxygen oxidoreductase nonheme iron-containing dioxygenase enzyme. It
helps in the oxygenation process of polyunsaturated fatty acids like arachidonic acid (AA). AA is further metabolized
FIGURE 16.3 List of drugs
designed based on the structure-
based approach.
FIGURE 16.4 List of drugs
designed based on the ligand-based
approach.
Discovery and optimization of lead molecules in drug designing Chapter | 16 255
via LOX, epoxygenase, and cyclooxygenase pathways. This pathway is differentiated based on the position of oxygen-
ation. Based on the insertion position (5, 8, 9, 12, and 15) of oxygen, LOXs divided into 5-, 8-, 9-, 12-, and 15-LOXs.
Out of these, 5-LOXs predominant in nature and form 5-hydroperoxy eicosatetraenoic acid (5-HpETE). It acts as a pre-
cursor of peptide and nonpeptidoleukotriens synthesis. It acts as the main cause of asthma, rhinitis, and ulcerative coli-
tis. Furthermore, it induces the growth of tumor cells and mitogenic processes. So, the inhibition of 5-LOXs is an
essential criterion for the treatment of these disorders.
In this whole scenario, CADD plays an important role in lead molecule discovery and their optimization. The devel-
opment of a therapeutic agent for LOXs inhibition has many challenges. The two principles of drug designing, such as
structure-based and ligand-based tools, were used. In this study, the structure of LOXs was solved, and novel LOXs
inhibitors were designed via in silico approaches. Zileuton acts as 5-LOX inhibitor, which is FDA approved and used in
the treatment of asthma (Aparoy, Kumar Reddy, & Reddanna, 2012).
16.3 Discovery of the lead molecule
Any chemical compound that exhibits biological or pharmacological properties with therapeutic characteristics is called
a lead molecule. The discovery of the lead molecule with good selectivity, efficiency, efficacy, and pharmacokinetic
parameters is a challenging task. Sometimes, identified lead molecule wants some structural modification to enhance
their biological properties. This process is called lead optimization. The investigated lead molecule must satisfy five
characteristics to work as a bioactive drug molecule. These are potency, bioavailability, duration, safety, and pharma-
ceutical acceptability. Potency involves the capability of any molecule to exhibit desirable pharmaceutical properties in
very few quantities. The transportation of drug molecules with multiple barriers to reach the target site is called bio-
availability. Duration is the time between the entry of drug molecules and their pharmacological response. Safety rules
include their toxicity parameters which indicate no side effects on the organism. Pharmaceutical action related with rea-
sonable synthetic pathway, solubility, chemical stability, dissolution rate, etc. So, these all factors are important in drug
discovery programs and their knowledge is necessary for optimization programs (Cheng, Korfmacher, White, &
Njoroge, 2007).
Lead molecule discovery is based on traditional library screening, fragment-based screening, and virtual screening
(Jain, 2004; Lavecchia & Di Giovanni, 2013). The aim of the rapid discovery of drugs involves target identification and
validation. Following the target validation process, hit identification and lead discovery phases are developed for novel
drug discovery. The physical and biochemical parameters are used to decide for change in the structural property of
compounds to synthesize an effective lead molecule for drug development.
16.4 Types of lead molecules
Several natural leads are available in the databases as well as in the literature that possesses some biological activity
against a biological target and may be used as a drug after some chemical modification. In pharmaceutical sciences, the
lead molecules are divided into three categories, that is, natural, synthetic, and semisynthetic (Leisner, 2020).
16.4.1 Natural lead compounds
Nature gives a wide number of biologically active secondary metabolites that were used to cure a disease. Natural lead
compounds were derived from natural products (NPs). NPs are secondary metabolites that are synthesized by the plants
animal and microbes. These natural compounds are diverse in their structural properties and have several active ingredi-
ents. It gives new opportunities to develop a chemical library (Butler, 2004; Gorlenko, Kiselev, Budanova, Zamyatnin,
& Ikryannikova, 2020; Mishra & Tiwari, 2011). It has been found that more than 80% of drugs were used to cure a dis-
ease, which are derived from natural compounds. Out of these, 40% of drugs based on NPs are approved by FDA
(Maridass & De Britto, 2008; Newman & Cragg, 2012).
The pharmaceutical industry pays much attention to the discovery of natural lead molecules because of their lesser
toxicity and herbal characteristics (Pan et al., 2013). In this process, plant, animal extract, microbial products, marine
organisms, and fungi were used to isolate the biologically active molecule (Beutler, 2009; Dias, Urban, & Roessner,
2012). These isolated active molecules are further optimized and screened by using combinatorial chemistry to get effi-
cient drug candidates (Cragg & Newman, 2013). From the approved data, it has been found that only 6% of unmodified
drugs based on NPs were used and 28% of drugs are used with NP-based modified drugs. This survey concludes that
structural modification of NPs based on the suitability of their target site creates new challenges in the pharmaceutical
256 Bioinformatics
industry (Khazir, Mir, Mir, & Cowan, 2013; Lachance, Wetzel, & Waldmann, 2010). Molecule weight and partition
coefficient are two key descriptors that plays important role in designing of new structure drug molecule (Pascolutti &
Quinn, 2014). The discovery of natural lead molecules includes multidisciplinary collaboration involving computational
chemistry and combinatorial chemistry (Singh, 2018). In the 21st century, CADD plays an immense role in designing
effective and efficient drug libraries with therapeutic properties. It gives information about perfect coordination between
the lead and target site interaction followed by pharmacodynamics parameters (Sidhu, Bhangu, Pathak, Yadav, &
Chhuneja, 2020; Thomford et al., 2018). Fig. 16.5 represents the 2D structure of some natural lead compounds used for
the treatment of various diseases.
16.4.2 Synthetic lead molecules
Synthetic lead molecules are those that design synthetically with the combination of organic synthetic chemistry and
combinatorial chemistry. FDA-approved 36% of drugs derived synthetically were used for medical purposes. The
design of lead molecules depends on their suitability, flexibility, affinity, stability, and toxicity at the target site (Flick
et al., 2017). These parameters also help to synthesize the multitarget ligand. Designing a molecule having similar
structural properties with NPs is more trending in CADD technology. With these molecules, drug likeliness properties
FIGURE 16.5 List of natural drugs and
their applicability.
Discovery and optimization of lead molecules in drug designing Chapter | 16 257
with efficiency and toxicity parameters were satisfied simultaneously. First, the chemical diversity is used to improve
the selectivity against the target. Sometimes, the minute difference in structure creates a problem in interaction with the
target, and a drug does not bind selectively to a target for which it was designed, which in turn leads to undesired
response and toxicity. To overcome this problem, combinatorial chemistry is used to select the perfect library of lead
molecules via chemical structural diversity. The target-oriented designing of molecules has also improved the effective-
ness of the drug (Clemons et al., 2011; Gerry & Schreiber, 2018; Schreiber, 2000; Trosset & Carbonell, 2015).
Recently, the pharmaceutical industry focused on some drugs, such as sulfonamide and sulfones, to improve pharmaco-
logical potential and to minimize toxicity (Fox et al., 2017; Greenwood, 2008). A list of some synthetic drugs that
approved based on their ADMET parameters is shown in Fig. 16.6.
FIGURE 16.6 List of some synthetic drugs and their applicability.
258 Bioinformatics
16.4.3 Semisynthetic drugs
Semisynthetic drugs are those that start from a natural source as starting material and end with chemical products.
These are hybrid of natural and synthetic drug. From ancient times, NP-based drugs are efficient and effective for the
treatment of diseases; but they have become less effective with respect to time, a due mutation in the target structure.
To overcome this problem, the modification in their structure is essential to give a new direction in pharmaceutical
chemistry. It generates semisynthetic drugs and maintains their natural properties. The designing of these drugs is more
focused on because it directly associated with natural compounds. The modification can be done based on the structural
properties nature of the functional group present in the molecules. Generally, the modification occurs in macrolides,
polycyclic glycopeptide, anthracyclines, oligomycin, heliomycin, and other entities. Macrolide-based antibiotic drugs
are clarithromycin and azithromycin exhibiting antimicrobial properties via inhibition of protein synthesis.
Glycopeptide-based semisynthetic antibiotic drugs are telavancin, oritavancin, and dalbavancin (Olsufyeva &
Yankovskaya, 2020). The first semisynthetic drug penicillin was discovered in 1928 by Alexander Flaming. It is derived
from Staphylococcus bacteria and acts as an efficient antibacterial agent (Clarke, 2015). Besides, antibacterial semisyn-
thetic drugs were developed for the treatment of bacterial infection caused by Mycobacterium tuberculosis (Lee, 2016).
16.5 Lead optimization and strategies
Lead optimization involves in which any lead molecule (biologically active) can be modified structurally and maintains
its physicochemical properties, thermodynamic, pharmacokinetic parameters, and toxicity (Jorgensen, 2009). When any
chemical compounds to be biological or pharmacological active with some chemical and physical properties present
active oral drug as per Lipinski rule. Lipinski rule of five is used to evaluate the drug-likeness of newly discovered or
designed molecules. According to this rule, for oral active drugs, the molecule should contain 5 or less than 5 donor
hydrogen bonds and 10 acceptor hydrogen bonds. Molecular mass should be less than 500 Da and its partition coeffi-
cient (log P-value) not higher than 5 (Lipinsk, Lombardo, Dominy, & Feeney, 2001; Wenlock, Austin, Barton, Davis,
& Leeson, 2003). Lead optimization not only depends on only binding parameters that exhibit information about drug
efficiency but also depends on the metabolic process occurs in living organism after consumption of a drug
(Korfmacher, 2003). The optimization of lead molecules was carried out by using organic synthetic chemistry and
dynamic/kinetics parameters. Organic synthetic chemistry involves structure simplification, structure modification,
functional group interconversion, and bonding strength/selectivity. Dynamic/kinetics parameters include thermodynam-
ics, pharmacodynamics/kinetics, and improvement of ADMET properties (Fig. 16.7). The major approaches used in the
optimization of lead molecules are discussed in the following sections.
16.5.1 By using organic synthetic chemistry
The response of the drug molecule as a pharmacological agent depends on the target structure and nature of the func-
tional group present in the drug molecule. The suitable relation between binding sites and efficient biological responses
is the effective criteria for the development of drug molecules. The slight changes in the designed molecular structure
may lead to a change in their properties as a drug. Synthetic organic chemistry is the strong pipeline of the CADD tech-
nique (Campos et al., 2019). The proper guidance of structural chemistry for the designing of lead molecules gives an
immense role in the pharmaceutical industry (Blakemore et al., 2018; Nadin, Hattotuwagama, & Churcher, 2012). The
strategy to fit the connection with their structure and functional properties are most important for new drug develop-
ment (Van Arnam & Dougherty, 2014).
To overcome the problem of molecular obesity, the design of the library includes different plans. These are
diversity-oriented synthesis (DOS), target-oriented synthesis (TOS), biological-oriented synthesis (BOS), and function-
ally oriented synthesis (FOS). These libraries help to find out the more active molecule and structure-specific mole-
cules. TOS is used in the discovery of drugs by the synthesis of the small molecule, based on preselected target protein
whereas DOS is used to identify the similar therapeutic protein target sites and more efficient drug molecule. TOS
planned efficiently with retrosynthetic analysis. These are based on stereo selective organic synthetic chemistry. Retro
synthesis involves the study of a structural element of the product rather than the structure of the reactant for the trans-
formation of the synthetic procedure. This gives immense knowledge about the structure simplification by the organic
chemist (Fig. 16.8). In contrast, DOS is related to the collection and study of a large biological active molecule with
complexity and diversity to the target protein site (Fig. 16.9). Complexity and diversity of molecule play essential
dynamics with proteinligand interaction because in many cases simplicity of molecules disturbed binding parameters.
Discovery and optimization of lead molecules in drug designing Chapter | 16 259
FIGURE 16.7 Strategies and approaches used
for the lead optimization.
FIGURE 16.8 Target-oriented synthesis and ret-
rosynthetic analysis of complex molecule (focused
libraries) to simple lead molecules (building
blocks). Example of retrosynthesis: phthalascidin
(anticancer agent) and neocarzinostatin chromo-
phore (antiproliferative actions).
FIGURE 16.9 Simple building
block to the complexity and diverse
structure by diversity-oriented
synthesis.
260 Bioinformatics
It affects the therapeutic properties of the drug molecule. TOS and DOS are complementary to each other. The selection
criteria of techniques depend on the lead molecule structure and target site (Rodriguez, 2012; Tan, 2005). BOS is
related to the study of the identified compound as its biological importance. FOS is associated with structural and chem-
ical biology. FOS aims to design the derivative with a small effect on the biological properties of the parent molecules
(Wender, Verma, Paxton, & Pillow, 2008; Wetzel, Bon, Kumar, & Waldmann, 2011).
16.5.2 Structure simplification
To resolve the problem of molecular obesity, simplification of structure with the same or higher potency leads to atten-
tion in the pharmaceutical industry. The necessary conditions for reoptimization of the drug are synthetic difficulty,
unfavorable physiochemical profile, and higher toxicity. With these difficulties, simplification of structure with favor-
able functional parameters is the important criteria. Simplification of drug structure occurs by structure affinity relation-
ship (SAR)-based design, structure-based design, and pharmacophore-based design. It has been found that structure
simplification with suitable functional group entity improves synthetic accessibility as well as pharmacokinetic para-
meters. Natural products are more complicated in structure. So, these techniques are most useful for natural biologically
active molecules (Wang, Dong, & Sheng, 2019). Some examples of simplified biological active analogs of NPs are
given in Table 16.1.
16.5.3 Structure modification
Modification of the structure of lead molecule leads to optimize the binding affinity with binding site amino acid which
present in the receptors. Hydrophilic compounds are those having a stronger affinity toward polar compounds.
Synthetic chemistry is used to optimize the structure of molecules through modification, in the functional group which
makes the molecule, more efficient than their native structure (Waring, 2010). Another factor associated with rigidifica-
tion makes the molecule stable in one conformation and it helps to find out a better understanding to fit in the binding
pocket. RAF kinases are used as a target protein for the designing of the drug molecule. The study reveals that rigidifi-
cation of pyrazolopyrimidine via ring closure improves the selectivity against RAF kinase and helps in designing effi-
cient anticancer agents (Fig. 16.10)(Assadieskandar et al., 2019).
16.5.4 Functional group interconversion
Functional group interconversion is related to change in structural properties via suitable functionalization to make a
drug more effective, less toxic, and highly efficient. During lead optimization, many functional group interconversion
reactions are used to develop a more effective candidate drug. These are electrophilic reaction, deprotonation, aromatic
nucleophilic substitution, amide formation, SuzukiMiyaura reaction, etc. From the survey, it has been found that, in
the overall process of drug discovery, functional group interconversion plays an 8% role in lead optimization. Click
chemistry, multicomponent reactions, macromolecule synthesis based on NPs, and activity-directed synthesis are
involved in strategies that involve lead molecule identification and optimization (Bostro
¨m, Brown, Young, & Keseru
¨,
TABLE 16.1 Natural product-derived simplified active compounds and their application.
S. no. Natural products Simplified biological active analogs Application
1. Morpholine Butorphanol, pentazocine, pethidine, methadone μand ƙopioid receptor agonist and antagonist
2. Halichondrin B Eribulin, mesilate Antitumor
Activity
3. Myriocin Fingolimod Immunosuppressive
Activity
4. Trichostatin A Vorinostat Histone deacetylase inhibitor
5. Schisandin C Bicyclol Antihepatitis B activity
6. Staurosporine Ruboxistaurin, enzastaurin Protein kinase C inhibitor
7. Asperlicin Devazepide Cholecystokinin receptors antagonist
Discovery and optimization of lead molecules in drug designing Chapter | 16 261
2018). Functional group interconversion correlated with SAR. It (1) improves affinity with target site, (2) increases flex-
ibility, (3) decreases lipophilicity, and (4) enhances the functionality of lead molecule. Based on the SAR, various σ2
receptors are designed to improve their antiproliferative activity. These designed lead molecules are based on morphans,
benzamides, indoles, and N-cyclohexylpiperazines structures (Abate, Perrone, & Berardi, 2012).
16.5.5 Bonding strength and selectivity
Electrostatic interaction, hydrogen bonding, van der Walls force, and hydrophobic interaction are weak type intermolec-
ular interaction, which acts as a stabilizing agent between drugreceptor entities (Barratt et al., 2005; Hubbard &
Haider, 2010; Kenny, 2009; Matthews, 2001) and helps in binding efficiency and efficacy of the drug. It depends on
the orientation and length of the hydrogen bond. The selection of lead molecules and target sites depends on the chemi-
cal nature of both candidates for donor and acceptor accessibility. It gives pharmacological interest to demonstrate their
therapeutic values. The electrostatic potential is the energy associated with the charge around both sides (receptor and
donor entity). It defines the potency of the molecule to attach with the target site (Bhosale, Shaikh, Coutinho, & Saran,
2007). Hydrophobicity is a nonpolar type of interaction that increases the affinity between the drug and the target site.
The parameters of hydrophobicity also play important role in designing the structure of the lead molecule (Patil et al.,
2010).
16.5.6 Using thermodynamic, pharmacodynamics, and pharmacokinetic parameters
Besides synthetic chemistry, the optimization of lead compounds can be done by using thermodynamic-, pharmacody-
namic-, and pharmacokinetic-based parameters. It is also recognized as a key strategy and is discussed in the following
sections.
These parameters are related to Gibbs free energy, binding-free energy, entropy, and enthalpy changes when the
interaction between two entities (drug and target site) occurs (Freire, 2008). Suitable functional groups of a ligand that
exhibit the appropriate efficiency will decide their thermodynamic parameters. These come under rational drug design-
ing (Holdgate & Ward, 2005; Thoppil, Choudhary, & Kishore, 2016). The entropy factor followed with the second law
of thermodynamic maintains the structural property of lead molecule as well as connection suitability (Dini, Guarini,
Morrone, & Marzilli, 2012; Uehara, Sakane, & Bertolotti, 2008) and contributes the favorable condition for binding
(Chaires, 2008). These parameters play an important role to address the interaction based on associated energy and con-
nection between ligand and target for the drug discovery. Binding strength depends on thermodynamics. It will give a
better understanding of the flexibility, strength, and suitability of lead molecules during proteinligand interaction
(Judy, Choudhary, & Kishore, 2016).
Pharmacodynamics act on the lead molecule and provide information about drug toxicity (T) whereas, pharmacoki-
netics gives the information about absorption, distribution, metabolism, and excretion (ADME) of the lead molecule in
biological systems. The action and transportation rate of drug molecules depends on their pharmacodynamics and phar-
macokinetics parameters. The small changes in the structure of the lead molecule will change the whole property of
pharmacokinetics and dynamic. Based on the survey, it has been found that pharmacokinetics parameters play a 39%
role in the failure of a new drug. To improve the efficiency and efficacy of a drug by reducing its toxicity, it is impor-
tant to understand these parameters for the development and optimization of the drug molecule. The coordination
FIGURE 16.10 Rigidification of pyrazolopyri-
midine entity for efficient RAF kinase inhibition:
(A)Nonselective kinase inhibitor and (B) highly
selective and potent B-RAF inhibitor.
262 Bioinformatics
between these parameters gives a better understanding of reactivity, action point, and safety factors with their target
sites (Walker, 2004).
Absorption involves the entering of drug molecules into blood circulation. It enters the body via the lung, skin, or
gastrointestinal tract. Absorption depends on the pathways of transportation in the blood and the concentration of the
drug. After absorption, the transfer of lead molecule occurs from one site to another within the whole body is called dis-
tribution. It depends on the solubility and binding affinity; furthermore, the metabolism of the drug molecule takes
place. Maximum metabolism occurs in the liver (Gunaratna, 2000). It involves various processes, such as dealkylation,
hydration, oxidation, and reduction, and it depends on the metabolic activity of the cell inside the environment.
Excretion is the elimination process of a drug or its metabolic product from the body. Highly polar compounds are
removed easily through urine (Carrillo, 2011). Any drug molecule having lesser toxicity is more efficient to cure dis-
ease. These all factors give an alert toward the success or failure of the designed molecule as a drug. The optimization
of ADMET properties of drugs is a challenging task; it can be optimized during experimental/preclinical and clinical
studies by mixing some other chemicals/preservatives and applied using different concentrations to evaluate their
ADMET nature.
16.6 Computational lead optimization
Computational methods, tools, and database resources hold immense potential in lead discovery and optimization.
Several methods and tools like databases have been developed for this purpose. The scoring functions and search algo-
rithms are used for the identification of lead molecules using different software packages available for molecular dock-
ing and virtual screening, that is, AutoDock vina, Glide, etc. The major database resources are PubChem, ChemSpider,
and ZINC holding the structure and properties of molecules utilized in the drug discovery program. Besides, the
DrugBank database has FDA-approved drugs and plays a key role in drug repurposing as a new lead for the treatment
of other diseases. The ADMET prediction tools are also playing a vital role in evaluating the drug-likeness of mole-
cules. Based on computational evaluation of putative molecules, it will be further subjected to chemical drawing tools
like ChemSketch, MarwinSketch, etc., to modify or replace a targeted functional group for improving their biological
activity (Fig. 16.11), followed by experimental validation. The key disadvantage of any drug molecule is toxicity, many
drugs comeback from the market to the lab for further investigation and optimization due to their toxic nature. The
major toxic effects are carcinogenicity, cytotoxicity, teratogenicity, phospholipidosis, and reproductive toxicity. To pre-
vent toxicity, medicinal chemists should consider the toxicity data during lead optimization. The major strategies used
for minimizing or removing the toxic effect of lead are: (1) eliminate substructures that cause toxic responses; (2) syn-
thetic modifications to eliminate any possibly toxic substructures from the lead molecules; (3) during lead optimization,
prevent the insertion of toxic substructures; (4) screening of toxic compounds through reactive metabolite assays; (5)
obstruct the functional group undergoing bioactivation by a nonbioactive functional group; (6) Insertion of a bulky sub-
stituent near the metabolism site to prevent metabolic activation; and (7) substitution with a metabolism-resistant sub-
stituent or metabolized by a nonreactive species. The structural changes will improve the drug-likeness of molecules
leading to the development of safe drugs (Agnihotry, Pathak, Srivastav, Shukla, & Gautam, 2020; Pathak, Singh, Sagar,
Baunthiyal, & Kumar, 2020).
16.7 Advantages of computational lead designing
In this whole scenario, CADD pays much attention because of three major factors. These are (1) screening of a large
set of the molecule on target structure, which can be further evaluated by computational as well as experimental meth-
ods; (2) guiding the optimization of lead compounds based on their affinity, pharmacokinetic parameters, and toxicity
criteria; and (3) helping in designing of novel compounds based on their structure to improve their functionality as a
potential drug. CADD technique is very applicable for the modeling of the drug. Keeping in mind all the things compu-
tational chemistry and bioinformatics in addition to combinatorial chemistry address the various challenges associated
with the drug discovery pipeline in less time and cost (Singh & Pathak, 2020).
16.8 Future perspectives
As per demanding aspects toward the cure of diseases, the potential drug designing has played more focused attention
right now. The setups of the perfect combination of structural and functional properties are the challenging task to
develop the active pharmaceutical ingredients. The potency of drugs not only depends on their efficiency, selectivity,
Discovery and optimization of lead molecules in drug designing Chapter | 16 263
and efficacy but also depends on toxicity factors. CADD helps to develop new parameters to make the lead molecule
more efficient and selective toward their target. SAR and mode of action also pay much attention to optimization pro-
cesses of lead. Chemical diversity followed by DOS, TOS, BOS, and FOS is important to play role in drug designing to
improve site-specificity and reduce toxicity. The selection of lead for optimization is based on ADMET screening.
These parameters are important for lead optimizations as per our need, and further going for molecular dynamics simu-
lation, binding energy calculation followed by in vitro and in vivo study in drug discovery program. In recent years due
to advances in computational sciences and experimental techniques, it becomes easier and fast in the future. Therefore
coordination within in silico, in vivo, and in vitro technologies plays a vital role in the pharmaceutical industry for the
discovery of well-optimized drugs.
16.9 Conclusion
The computational study based on the identification of lead compounds, that is, compound holds drug-like properties
against particular disease, is a resource taking and complicated process. After the identification of lead compounds, it
takes much time to become a drug. There are multiple pipelines are involved from lead discovery to drug development
via computational and experimental approaches. The optimization of identified lead as per our need in terms of affinity,
efficacy, and potency is one key step in a drug-designing program. This chapter highlights the major strategies used for
lead optimization, that is, structure simplification, structure modification, functional group interconversion, bonding
strength and selectivity, thermodynamic parameters, and pharmacodynamics and pharmacokinetics parameters and their
importance in drug designing for the discovery and development of novel therapeutics.
FIGURE 16.11 Lead optimization strategies for improving absorption, distribution, metabolism, excretion, and toxicity and binding interaction.
264 Bioinformatics
Conflict of interest
The authors declare that they have no conflict of interest.
References
Abate, C., Perrone, R., & Berardi, F. (2012). Classes of Sigma2 (σ2) receptor ligands: Structure affinity relationship (SAfiR) studies and antiprolifera-
tive activity. Current Pharmaceutical Design,18, 938949.
Acharya, C., Coop, A., Polli, J. E., & MacKerell, A. D. (2011). Recent advances in ligand-based drug design: Relevance and utility of the conforma-
tionally sampled pharmacophore approach. Current Comput-Aided Drug Design,7,1022.
Agnihotry, S., Pathak, R. K., Srivastav, A., Shukla, P. K., & Gautam, B. (2020). Molecular docking and structure-based drug design.Computer-aided
drug design (pp. 115131). Singapore: Springer.
Anderson, A. C. (2003). The process of structure-based drug design. Chemistry & Biology,10, 787797.
Aparoy, P., Kumar Reddy, K., & Reddanna, P. (2012). Structure and ligand based drug design strategies in the development of novel 5-LOX inhibi-
tors. Current Medicinal Chemistry,19, 37633778.
Assadieskandar, A., Yu, C., Maisonneuve, P., Kurinov, I., Sicheri, F., & Zhang, C. (2019). Rigidification dramatically improves inhibitor selectivity
for RAF kinases. ACS Medicinal Chemistry Letters,10, 10741080.
Barratt, E., Bingham, R. J., Warner, D. J., Laughton, C. A., Phillips, S. E., & Homans, S. W. (2005). Van der Waals interactions dominate ligand 2-
protein association in a protein binding site occluded from solvent water. Journal of the American Chemical Society,127, 1182711834.
Beutler, J. A. (2009). Natural products as a foundation for drug discovery. Current Protocols in Pharmacology,46,911.
Bhosale, S. S., Shaikh, M. S., Coutinho, E. C., & Saran, A. (2007). Application of molecular electrostatic potentials in drug design. Proceedings of the
Indian National Science Academy,73, 91.
Blakemore, D. C., Castro, L., Churcher, I., Rees, D. C., Thomas, A. W., Wilson, D. M., & Wood, A. (2018). Organic synthesis provides opportunities
to transform drug discovery. Nature Chemistry,10, 383394.
Bostro
¨m, J., Brown, D. G., Young, R. J., & Keseru
¨, G. M. (2018). Expanding the medicinal chemistry synthetic toolbox. Nature Reviews. Drug
Discovery,17, 709727.
Butler, M. S. (2004). The role of natural product chemistry in drug discovery. Journal of Natural Products,67, 21412153.
Campos, K. R., Coleman, P. J., Alvarez, J. C., Dreher, S. D., Garbaccio, R. M., Terrett, N. K., .. . Parmee, E. R. (2019). The importance of synthetic
chemistry in the pharmaceutical industry. Science (New York, N.Y.),363, 6424.
Carrillo, N. J. (2011). Pharmacokinetic process: Does the site of drug action? Excretion of drugs. Revista de Enfermeria (Barcelona, Spain),34, 24.
Chaires, J. B. (2008). Calorimetry and thermodynamics in drug design. Annual Review of Biophysics,37, 135151.
Cheng, K. C., Korfmacher, W. A., White, R. E., & Njoroge, F. G. (2007). Lead optimization in discovery drug metabolism and pharmacokinetics/case
study: The hepatitis C virus (HCV) protease inhibitor SCH 503034. Perspectives in Medicinal Chemistry,1,19.
Clark, D. E. (2006). What has computer-aided molecular design ever done for drug discovery? Expert Opinion on Drug Discovery,1, 103110.
Clarke, H. T. (Ed.), (2015). Chemistry of penicillin. Princeton University Press.
Clemons, P. A., Wilson, J. A., Danˇ
´k, V., Muller, S., Carrinski, H. A., Wagner, B. K., ... Schreiber, S. L. (2011). Quantifying structure and perfor-
mance diversity for sets of small molecules comprising small-molecule screening collections. Proceedings of the National Academy of Sciences of
the United States of America,108, 68176822.
Cragg, G. M., & Newman, D. J. (2013). Natural products: A continuing source of novel drug leads. Biochimica et Biophysica Acta—General Subjects,
1830, 36703695.
Dias, D. A., Urban, S., & Roessner, U. (2012). A historical overview of natural products in drug discovery. Metabolites,2, 303336.
Dini, F. L., Guarini, G., Morrone, D., & Marzilli, M. (2012). The second law of thermodynamics and the heart. Future Cardiology,8, 697706.
Durrant, C. (2001). The responsibility of the pharmaceutical industry. Wiley, https://doi.org/10.1046/j.1469-0691.7.s6.1.x.
Flick, A. C., Ding, H. X., Leverett, C. A., Kyne, R. E., Jr, Liu, K. K. C., Fink, S. J., & O Donnell, C. J. (2017). Synthetic approaches to the new drugs
approved during 2015. Journal of Medicinal Chemistry,60, 64806515.
Fox, G. J., Schaaf, H. S., Mandalakas, A., Chiappini, E., Zumla, A., & Marais, B. J. (2017). Preventing the spread of multidrug-resistant tuberculosis
and protecting contacts of infectious cases. Clinical Microbiology and Infection,23, 147153.
Freire, E. (2008). Do enthalpy and entropy distinguish first in class from best in class? Drug Discovery Today,13, 869874.
Gerry, C. J., & Schreiber, S. L. (2018). Chemical probes and drug leads from advances in synthetic planning and methodology. Nature Reviews. Drug
Discovery,17, 333.
Gorlenko, C. L., Kiselev, H. Y., Budanova, E. V., Zamyatnin, A. A., & Ikryannikova, L. N. (2020). Plant secondary metabolites in the battle of drugs
and drug-resistant bacteria: New heroes or worse clones of antibiotics? Antibiotics,9, 170.
Grant, M. A. (2009). Protein structure prediction in structure-based ligand design and virtual screening. Combinatorial Chemistry & High Throughput
Screening,12, 940960.
Greenwood, D. (2008). Antimicrobial drugs: Chronicle of a twentieth century medical triumph. Oxford University Press, https://doi.org/10.1093/jac/
dkp066.
Grzybowski, B. A., Ishchenko, A. V., Shimada, J., & Shakhnovich, E. I. (2002). From knowledge-based potentials to combinatorial lead design in sili-
co. Accouts of Chemical Research,35, 261269.
Discovery and optimization of lead molecules in drug designing Chapter | 16 265
Gunaratna, C. (2000). Drug metabolism & pharmacokinetics in drug discovery: A primer for bioanalytical chemists, part I. Current Separations,19,
1724.
Holdgate, G. A., & Ward, W. H. (2005). Measurements of binding thermodynamics in drug discovery. Drug Discovery Today,10(22), 15431550.
Hubbard, R. E., & Haider, M. K. (2010). Hydrogen bonds in proteins: Role and strength. Available from https://doi.org/10.1002/9780470015902.
a0003011.
Jain, A. N. (2004). Virtual screening in lead discovery and optimization. Current Opinion in Drug Discovery & Development,7(4), 396403.
Jorgensen, W. L. (2004). The many roles of computation in drug discovery. Science (New York, N.Y.),303(5665), 18131818.
Jorgensen, W. L. (2009). Efficient drug lead discovery and optimization. Accounts of Chemical Research,42(6), 724733.
Jorgensen, W. L. (2010). Pulled from a protein’s embrace. Nature,466(7302), 4243.
Judy, E., Choudhary, S., & Kishore, N. (2016). Rational drug design and future directions: Thermodynamic perspective. Austin Biomolecules,1(1),
1003.
Kenny, P. W. (2009). Hydrogen bonding, electrostatic potential, and molecular design. Journal of Chemical Information and Modeling,49(5),
12341244.
Khanna, I. (2012). Drug discovery in pharmaceutical industry: Productivity challenges and trends. Drug Discovery Today,17(1920), 10881102.
Khazir, J., Mir, B. A., Mir, S. A., & Cowan, D. (2013). Natural products as lead compounds in drug discovery. Journal of Asian Natural Products
Research,15, 764788.
Korfmacher, W. A. (2003). Lead optimization strategies as part of a drug metabolism environment. Current Opinion in Drug Discovery and
Development,6(4), 481485.
Lachance, H., Wetzel, S., & Waldmann, H. (2010). Role of natural products in drug discovery.Lead Generation Approaches in Drug Discovery
(pp. 187229). Wiley.
Lavecchia, A., & Di Giovanni, C. (2013). Virtual screening strategies in drug discovery: A critical review. Current Medicinal Chemistry,20(23),
28392860.
Lee, R. (2016). Semisynthetic natural products for antibacterial drug discovery. The FASEB Journal,30(1_supplement), 392.
Leelananda, S. P., & Lindert, S. (2016). Computational methods in drug discovery. Beilstein Journal of Organic Chemistry,12(1), 26942718.
Leisner, J. J. (2020). The diverse search for synthetic, semisynthetic and natural product antibiotics from the 1940s and up to 1960 exemplified by a
small pharmaceutical player. Frontiers in Microbiology,11, 976.
Lin, S. K. (2000). Pharmacophore perception, development and use in drug design. edited by osman f. gu
¨ner. Molecules (Basel, Switzerland),5(7),
987989.
Lipinsk, C., Lombardo, F., Dominy, B. W., & Feeney, P. J. (2001). Experimental and computational approaches to estimate solubility and permeability
in drug discovery and development settings. Advanced Drug Delivery Reviews,46(13), 326.
Makrynitsa, G. I., Lykouras, M., Spyroulia, G. A., & Matsoukas, M. T. (2018). Insilico drug design. Wiley, https://doi.org/10.1002/9780470015902.
a0028112.
Maridass, M., & De Britto, A. J. (2008). Origins of plant derived medicines. Ethnobotanical Leaflets,1, 44.
Mason, J. S., Good, A. C., & Martin, E. J. (2001). 3-D pharmacophores in drug discovery. Current Pharmaceutical Design,7(7), 567597.
Matthews, B. W. (2001). Hydrophobic interactions in proteins. Wiley, https://doi.org/10.1038/npg.els.0002975.
Mignani, S., Huber, S., Tomas, H., Rodrigues, J., & Majoral, J. P. (2016). Compound high-quality criteria: A new vision to guide the development of
drugs, current situation. Drug Discovery Today,21(4), 573584.
Mishra, B. B., & Tiwari, V. K. (2011). Natural products: An evolving role in future drug discovery. European Journal of Medicinal Chemistry,46
(10), 47694807.
Morris, G. M., Huey, R., Lindstrom, W., Sanner, M. F., Belew, R. K., Goodsell, D. S., & Olson, A. J. (2009). AutoDock4 and AutoDockTools4:
Automated docking with selective receptor flexibility. Journal of Computational Chemistry,30(16), 27852791.
Nadin, A., Hattotuwagama, C., & Churcher, I. (2012). Lead-oriented synthesis: A new opportunity for synthetic chemistry. Angewandte Chemie
International Edition,51(5), 11141122.
Newman, D. J., & Cragg, G. M. (2012). Natural products as sources of new drugs over the 30 years from 1981 to 2010. Journal of Natural Products,
75(3), 311335.
Olsufyeva, E. N., & Yankovskaya, V. S. (2020). Main trends in the design of semi-synthetic antibiotics of a new generation. Russian Chemical
Reviews,89(3), 339.
Pajouhesh, H., & Lenz, G. R. (2005). Medicinal chemical properties of successful central nervous system drugs. NeuroRx: The Journal of the
American Society for Experimental NeuroTherapeutics,2(4), 541553.
Pan, S. Y., Zhou, S. F., Gao, S. H., Yu, Z. L., Zhang, S. F., Tang, M. K., Sun, J. N., Ma, D. L., Han, Y. F., Fong, W. F., & Ko, K. M. (2013). New
perspectives on how to discover drugs from herbal medicines: CAM’s outstanding contribution to modern therapeutics. Evidence-Based
Complementary and Alternative Medicine. Available from https://doi.org/10.1155/2013/627375.
Pascolutti, M., & Quinn, R. J. (2014). Natural products as lead structures: Chemical transformations to create lead-like libraries. Drug Discovery
Today,19(3), 215221.
Pathak, R. K., Baunthiyal, M., Taj, G., & Kumar, A. (2014). Virtual screening of natural inhibitors to the predicted HBx protein structure of hepatitis
B virus using molecular docking for identification of potential lead molecules for liver cancer. Bioinformation,10(7), 428.
Pathak, R. K., Gupta, A., Shukla, R., & Baunthiyal, M. (2018). Identification of new drug-like compounds from millets as Xanthine oxidoreductase
inhibitors for treatment of Hyperuricemia: A molecular docking and simulation study. Computational Biology and Chemistry,76,3241.
266 Bioinformatics
Pathak, R. K., Singh, D. B., Sagar, M., Baunthiyal, M., & Kumar, A. (2020). Computational approaches in drug discovery and design.Computer-
aided drug design (pp. 121). Singapore: Springer.
Patil, R., Das, S., Stanley, A., Yadav, L., Sudhakar, A., & Varma, A. K. (2010). Optimized hydrophobic interactions and hydrogen bonding at the
target-ligand interface leads the pathways of drug-designing. PLoS One,5(8), 12029.
Rai, S. K., Pathak, R. K., Singh, D. B., Bhatt, A., & Baunthiyal, M. (2021). Chemo-informatics guided study of natural inhibitors targeting rho
GTPase: A lead for treatment of glaucoma. In Silico Pharmacology,9(1), 111.
Rodriguez, R. (2012). Target-oriented and diversity-oriented organic synthesis. Modern Tools for the Synthesis of Complex Bioactive Molecules,9,
513538.
Sartorius, N. (2006). The meanings of health and its promotion. Croatian Medical Journal,47(4), 662.
Schmidt, T., Bergner, A., & Schwede, T. (2014). Modelling three-dimensional protein structures for applications in drug design. Drug Discovery
Today,19(7), 890897.
Schreiber, S. L. (2000). Target-oriented and diversity-oriented organic synthesis in drug discovery. Science (New York, N.Y.),287(5460), 19641969.
Scully, J. L. (2004). What is a disease? Disease, disability and their definitions. EMBO Reports,5(7), 650653.
Sidhu, K. S., Bhangu, S. K., Pathak, R. K., Yadav, I. S., & Chhuneja, P. (2020). Identification of natural lead compounds for leaf rust of Wheat: A
molecular docking and simulation study. Journal of Proteins and Proteomics,11, 283295.
Singh, D. B. (2018). Natural lead compounds and strategies for optimization. Frontiers in Computational Chemistry,4,147.
Singh, D. B., & Pathak, R. K. (2020). Computational approaches in drug designing and their applications. New York, NY: Humana, https://doi.org/
10.1007/978-1-0716-0607-0_6.
Sliwoski, G., Kothiwale, S., Meiler, J., & Lowe, E. W. (2014). Computational methods in drug discovery. Pharmacological Reviews,66(1), 334395.
Tan, D. S. (2005). Diversity-oriented synthesis: Exploring the intersections between chemistry and biology. Nature Chemical Biology,1(2), 7484.
Taylor, D. (2015). The pharmaceutical industry and the future of drug development. RSC. Available from https://doi.org/10.1039/9781782622345-
00001.
Thomford, N. E., Senthebane, D. A., Rowe, A., Munro, D., Seele, P., Maroyi, A., & Dzobo, K. (2018). Natural products for drug discovery in the 21st
century: Innovations for novel drug discovery. International Journal of Molecular Sciences,19(6), 1578.
Thoppil, A. A., Choudhary, S., & Kishore, N. (2016). Competitive binding of anticancer drugs 5-fluorouracil and cyclophosphamide with serum albu-
min: Calorimetric insights. Biochimica et Biophysica Acta—General Subjects,1860(5), 917929.
Tropsha, A., & Bajorath, J. (2016). Computational methods for drug discovery and design. Journal of Medicinal Chemistry,59(1), 1.
Trosset, J. Y., & Carbonell, P. (2015). Synthetic biology for pharmaceutical drug discovery. Drug Design, Development and Therapy,9, 62856302.
Trott, O., & Olson, A. J. (2010). AutoDockVina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization,
and multithreading. Journal of Computational Chemistry,31(2), 455461.
Uehara, M., Sakane, K. K., & Bertolotti, S. A. (2008). Thermodynamics of the heart: Relation between cardiac output and oxygen consumption.
American Journal of Physics,76(6), 566569.
Van Arnam, E. B., & Dougherty, D. A. (2014). Functional probes of drugreceptor interactions implicated by structural studies: Cys-loop receptors
provide a fertile testing ground: Mini perspective. Journal of Medicinal Chemistry,57(15), 62896300.
Wadood, A., Ahmed, N., Shah, L., Ahmad, A., Hassan, H., & Shams, S. (2013). In-silico drug design: An approach which revolutionarised the drug
discovery process. In-Silico Drug Design,1(1), 3.
Walker, D. K. (2004). The use of pharmacokinetic and pharmacodynamic data in the assessment of drug safety in early drug development. British
Journal of Clinical Pharmacology,58(6), 601608.
Wang, S., Dong, G., & Sheng, C. (2019). Structural simplification: An efficient strategy in lead optimization. Acta Pharmaceutica Sinica B,9(5),
880901.
Waring, M. J. (2010). Lipophilicity in drug discovery. Expert Opinon on Drug Discovery,5(3), 235248.
Wender, P. A., Verma, V. A., Paxton, T. J., & Pillow, T. H. (2008). Function-oriented synthesis, step economy, and drug design. Accounts of
Chemical Research,41(1), 4049.
Wenlock, M. C., Austin, R. P., Barton, P., Davis, A. M., & Leeson, P. D. (2003). A comparison of physiochemical property profiles of development
and marketed oral drugs. Journal of Medicinal Chemistry,46(7), 12501256.
Wetzel, S., Bon, R. S., Kumar, K., & Waldmann, H. (2011). Biology-oriented synthesis. Angewandte Chemie International Edition,50(46),
1080010826.
Zoete, V., Grosdidier, A., & Michielin, O. (2009). Docking, virtual high throughput screening and in silico fragment based drug design. Journal of
Cellular and Molecular Medicine,13(2), 238248.
Discovery and optimization of lead molecules in drug designing Chapter | 16 267
... At the final stage in drug discovery, the main goal of lead optimization is to attain a drug candidate that achieves a concentration-time profile in the body that is adequate for the desired efficacy and safety profile [1,2]. During lead optimization, structural modification not only changes pharmacokinetic properties in plasma, but also alters drug exposure/selectivity in disease-targeted tissues/normal tissue [3,4]. ...
... Drug tissue selectivity = Ctissue/ΣCtissue or AUCtissue/ΣAUCtissue (2) A perfect CNS drug candidate is supposed to exhibit high tissue selectivity and exposure in the brain for better therapeutic efficacy, and low tissue selectivity and exposure in other vital organs to reduce toxicity in the meantime. On the contrary, if a CNS drug candidate has low selectivity and low exposure in the brain but high selectivity/exposure in other vital organs, it may not be able to reach its therapeutic concentration in the CNS. ...
Article
Full-text available
The structure–tissue exposure/selectivity relationship (STR) aids in lead optimization to improve drug candidate selection and balance clinical dose, efficacy, and toxicity. In this work, butyrocholinesterase (BuChE)-targeted cannabidiol (CBD) carbamates were used to study the STR in correlation with observed efficacy/toxicity. CBD carbamates with similar structures and same molecular target showed similar/different pharmacokinetics. L2 and L4 had almost same plasma exposure, which was not correlated with their exposure in the brain, while tissue exposure/selectivity was correlated with efficacy/safety. Structural modifications of CBD carbamates not only changed drug plasma exposure, but also altered drug tissue exposure/selectivity. The secondary amine of carbamate can be metabolized into CBD, while the tertiary amine is more stable. Absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters can be used to predict STR. Therefore, STR can alter drug tissue exposure/selectivity in normal tissues, impacting efficacy/toxicity. The drug optimization process should balance the structure–activity relationship (SAR) and STR of drug candidates for improving clinical trials.
... Mwangi et al. [231] discuskey strategies used to optimize the performance of ABPs, including rational design and de novo synthesis as well as predictive computational tools and utilizing artificial intelligence in the design and synthesis of highly efficient lead drug candidates. Research on discovery and optimization of lead molecules, originating from natural sources [232] and nanotechnology-based delivery are some of them, [145]. Luong et al. [233] discuss approaches to improve biological activity of ABPs either by modifying chemical structure or incorporating into delivery systems. ...
Preprint
Full-text available
Conventional antibiotic and multidrug treatments become less and less effective and a discovery of new effective and safe antibacterial agents got a global priority. The returning to the natural antibacterial product is relatively new current trend. Terrestrial biota is a rich source of biologically active substances whose antibacterial potential was not fully utilized. Aim of this review is to present the current state-of-the art of terrestrial biota-derived antibacterial agents and inspired by nature treatments. It is a summary of the most important sources and new identified or modified antibacterial agents and treatments during the last 5 years. It focuses on the significance of plants- animals- and bacteria-derived biologically active agents as powerful alternatives to antibiotics as well as on the advantages of utilizing natural antibacterial molecules alone or in combinations with antibiotics. The main conclusion is that the terrestrial biota derived antibacterial products and substances open variety of new ways to modern improved therapeutic strategies. New terrestrial sources of known antibacterial agents and new antibacterial agents from terrestrial biota were discovered during the last 5 years, which are under investigation together with some long ago known but now surviving their renaissance for the development of new medical treatments. Use of natural antibacterial peptides as well as combinational therapy by commercial antibiotics and natural products outlines as the most promising for treating of bacterial infections. In vivo testing and clinical trials are necessary to reach clinical application.
... Another crucial step in drug design and discovery is lead optimization so as to achieve maximum interaction between the target molecules. Consequently, CADD plays a central role in lead optimization [27]. ...
Article
Full-text available
Computer-aided drug design and discovery methods have been essential in developing small molecules with therapeutic properties over the last decades. Application of computational resources includes drug target identification, hit discovery, and lead optimization. Accordingly, with tremendous research efforts and the availability of financial support from government agencies across the world, and multinational drug companies, the overall research level in this area will continue to advance. The methodology used in this review paper entailed a thorough examination of research studies on relevant literature on drug design and development using computational resources. Extensive searches using Scopus, International Pharmaceutical Abstracts (OvidSp, WHO Global Health Library, Cochrane, Google Scholar, Web of Science, Science Direct, ProQuest dissertation & theses, Worldwide Political Science Abstracts (CSA), and PubMed was carried out. A standardized template was used to ensure that the selected papers met the inclusion criteria, and relevant to the review. Ultimately, there are robust technologies developed to enhance the drug discovery process. Therefore, this review provides insights into computational resources in Silico and ab initio methods and algorithms, not restricted to drug metabolism predictions for drug design, and the practical applications of artificial intelligence (AI) in drug discovery. Computational tools and methods for drug design and development such as molecular dynamics (MD), molecular docking, quantum mechanics (QM), hybrid quantum mechanics/molecular mechanics (QM/MM), and Density functional theory (DFT) have been reviewed. Accordingly, the emerging technique of synergistically employing these techniques influences the fundamental challenges of conventional medicines for complex diseases. Herein, we discuss ligand-based and structure-based drug discoveries, force field models in MD simulations, docking algorithms, subtractive and additive QM/MM coupling. Nonetheless, as computer-aided drug (CADD) approaches continue to evolve with significant improvements, the focus areas will be on docking and virtual screening, scoring functions, optimization of hits, and assessment of adsorption, distribution, metabolism, excretion, and toxicity (ADMET) properties. With the current success, the present computational resources will aid in the future discovery of novel compounds with high therapeutic performance. The ongoing oncology research efforts will also significantly contribute to UN sustainable development goals – good health and well-being, sustainable innovation and industrialization.
Chapter
The health of animals, humans, and the environment are intrinsically linked. It is estimated that 60% of all human diseases originate from animals. Globally, diseases affecting food-producing animals result in a 20% decrease in production. Increasing human activity and the resulting stress on ecosystems have created novel opportunities for the emergence and spread of diseases. The rapidly growing human population also raises significant concerns for the scientific community about protecting livestock from disease and increasing livestock production to meet the growing demand. Veterinarians play a vital role in disease prevention, which is crucial for preserving the health of our planet. Veterinary medicines are essential in maintaining animal health, which is directly linked to human and environmental health. However, due to the challenges posed by multi-drug resistance and mutations in pathogen genomes, new medicines are periodically required. Drug discovery, however, is a time-consuming process that demands substantial funds, resources, and manpower. The R&D departments of most pharmaceutical companies focus on human diseases; in recent years, however, researchers have begun employing novel approaches in drug discovery for livestock due to their significant impact on human health, food security, and farmers’ livelihoods. The advent of computational methods has facilitated the investigation, design, comparison, modeling, and prediction of binding energy, as well as pharmacokinetic and pharmacodynamics predictions and led compound optimization in drug discovery programs. Therefore, harnessing the potential of vetinformatics is akin to discovering an elixir that will strengthen the One Health agenda and aid in the identification of new veterinary drugs or vaccine candidates. This chapter aims to highlight various computational resources and approaches, and their applications in veterinary medicine.
Chapter
Worldwide problems, including increasing population, depletion of natural resources, and climate change, have led to a food crisis. Moreover, various diseases affecting livestock are impacting humans. Hence, the scientific community has been advised to adopt new and innovative approaches, and the use of bioinformatics in veterinary science is one such useful technique. In the current artificial intelligence (AI) era, several problems can be solved by decoding livestock systems using bioinformatic tools. Bioinformatics is a vast subject that has applications in various fields, and the approach incorporating it with veterinary and animal science is called “vetinformatics.” It can help understand the complex molecular mechanisms in farm animals that in turn would aid in developing practices for caring, breeding, and disease management, leading to robust livestock productivity. This chapter seeks to introduce the concept behind vetinformatics, the related challenges and opportunities, and its applications in veterinary and animal sciences.
Chapter
The variety of animal health issues and species poses particular challenges for veterinary medicine in the context of drug development. This book chapter delves further into the significance of machine learning in developing veterinary medication discovery and design methodologies. It defines how machine learning algorithms, in combination with data-driven methods, could accelerate drug discovery processes, improve therapeutic strategies, and enhance animal welfare. This chapter offers a detailed insight into the combination of machine learning and veterinary pharmacology, providing a look into the promising future of drug discovery in veterinary medicine through a careful assessment of applications, methodology, and case studies for the benefit of the veterinary science community.
Chapter
The first step in the drug discovery and development process is often the identification of new active scaffolds or fundamental chemical structures with the desired biological effect, besides serendipitous findings and outcomes from ethnopharmacology/traditional medicine. In contrast to the widespread usage of target-based approaches within the pharmaceutical industry over the past three decades, phenotypic drug discovery (PDD) methods do not depend on an understanding of a therapeutic target’s identity or a theory of how it contributes to disease. However due to recent significant advancements in the instruments for cell-based phenotypic screening, as well as their potential to address the incompletely understood complexity of diseases and their promise of producing first-in-class medications, PDD techniques have seen a renaissance of interest. On the other hand, PDD methodologies face significant obstacles such as hit validation and target deconvolution. Target-based drug discovery (TBDD) has been the standard method used by the pharmaceutical industry over the past three decades, owing to the explosion of knowledge facilitated by developments in molecular biology, recombinant technology, and genomics. Despite increasing investment from the pharmaceutical industry, the number of new molecular entities and biological product discoveries has not increased alongside the benefits of TBDD in throughput and costs. This chapter introduces the concept of comparing target-based versus phenotypic screening, including opportunities and challenges in current drug discovery. We also discussed approaches, evidence-based PDD in different diseases, and commonly used artificial intelligence techniques. In TBDD, we have highlighted different biological targets, methods of HIT and lead identification in silico design, and the advantages of TBDD. Though PDD and TBDD approaches are generally framed as antagonistic, they can be complementary, and combining them could increase the likelihood of successfully generating a new drug entity.
Article
Molecular docking is a structure-based computational technique that plays a major role in drug discovery. Molecular docking enhances the efficacy of determining the metabolic interaction between two molecules, i.e., the small molecule (ligand) and the target molecule (protein), to find the best orientation of a ligand to its target molecule with minimal free energy in forming a stable complex. By stimulating drug-target interactions, docking helps identify small molecules that might inhibit cancer-promoting proteins, aiding in the development of novel targeted therapies. Molecular docking enables researchers to screen vast reorganization, identifying potential anti-cancer drugs with enhanced specificity and reduced toxicity. The growing importance of molecular docking underscores its potential to revolutionize cancer treatment by accelerating the identification of novel drugs and improving clinical outcomes. As a wide approach, this computational drug design technique can be considered more effective and timesaving than other cancer treatment methods. In this review, we showcase brief information on the role of molecular docking and its importance in cancer research for drug discovery and target identification. Therefore, in recent years, it can be concluded that molecular docking can be scrutinized as one of the novel strategies at the leading edge of cancer-targeting drug discovery.
Article
Full-text available
Glaucoma, the most perilous disease leading to blindness is a result of optical neuropathy. Accumulation of aqueous humor in the posterior chamber due to a large difference in the rate of formation and its drainage in the anterior chamber causes an increase in intraocular pressure (IOP) leading to damage of nerve cells. A literature survey has revealed that inhibition of the Rho guanosine triphosphatases (rho GTPase) pathway by specific inhibitors leads to the relaxation of contractile cells involved in the aqueous outflow pathway. Relaxation of the strained contractile cells results in increased outflow thereby releasing IOP. In the present study molecular docking has been used to screen twenty seven bioactive (17 natural compounds and 10 conventional drugs) compounds that may play a significant role in relaxing contractile cells by inhibiting rho-GTPase protein. Docking results showed that among all-natural bioactive compounds Cyanidin and Delphinidine have a good binding affinity (− 8.4 kcal/mol) than the top screened conventional drug molecule Mitomycin, (− 6.3 kcal/mol) when docked with rho-GTPase protein. Cyanidin and Delphinidin belong to anthocyanidin, a glycoside form of anthocyanins from Vaccinium myrtillus L. and Punica granatum. The resembling potential of Cyanidin and Delphinidin concerning the drug Mitomycin was confirmed through simulation analysis. Molecular dynamics study (MDS) for 100 ns, showed that the rho GTPase-Delphinidine complex structure was energetically more stable than rho GTPase-Cyaniding complex in comparison to rho GTPase-Mitomycin complex. The comparative study of both the selected hits (Cyanidin and Delphinidin) was assessed by RMSD, RMSF, Rg, SASA, H-bond, PCA MM/PBSA analysis. The analysis revealed that Delphinidine is more potent to inhibit the rho GTPase as compare to Cyaniding and available conventional drugs in terms of stability and binding free energy. Based on the results, these molecules have good pharmacokinetic and pharmacodynamics properties and will prove to be a promising lead compound as a future drug for Glaucoma.
Chapter
Full-text available
Drug discovery is an expensive and complicated process. The drug must fulfill some criteria of being nontoxic, bioavailable, and potent. In the view of evermore stringent demands about efficacy, potency, and safety, the finding of the new drug-like molecule has become a complex and resource-intensive undertaking. Now, the availability of 3D structures of molecular drug targets and advances in computational approaches and bioinformatics speed up the application of molecular modeling in discovery. In this chapter, several molecular modeling strategies employed in modern drug discovery program are discussed. The concepts of structure- and ligand-based drug designing, protein modeling and visualization, molecular docking, virtual screening, molecular dynamics simulation, pharmacophore modeling, and QSAR approaches have been explained. Besides, we also provide important database resources and tools available for drug research. Finally, we present case studies conducted in our lab, showing how computational approaches can be implemented in reality for the discovery and designing of novel drugs from natural sources.
Article
Full-text available
Leaf rust of wheat, incited by fungus Puccinia triticina is an economically important disease. It is currently highly destructive leading to significant yield losses. Many disease resistant wheat varieties have been developed but unable to maintain resistance over a long time due to rapidly evolving effector proteins by the pathogen, which escapes interaction with plant R-genes over time. Synthetic fungicides are available for control of the disease but they are biohazardous and eco-unfriendly. Mitogen Activated Protein Kinases (MAPKs) have been found in all organisms, including fungi, and are involved in the transmission of a variety of extracellular signals to manage growth and differentiation processes. The present study aimed to identify natural fungicide like molecules that can inhibit MAPKs machinery involved in growth, mating, and virulence of pathogen through molecular docking and dynamics simulation study. A high-quality structural model of MAPK1 protein was predicted and used to dock 110 natural compounds. Two compounds viz. Cynaroside and Prodelphinidin showed a more binding affinity with MAPK1. Therefore, the docked complex structure of these two compounds and MAPK1 was further subjected to molecular dynamics simulation to evaluate its stability and binding nature during protein–ligand interaction. Based on the result analysis, it was predicted that they have efficient and stable binding nature, and better cell permeability properties. It may be used as a natural fungicide against leaf rust and other fungal diseases, only after further validation in lab and field conditions.
Article
Full-text available
The 1940s and 1950s witnessed a diverse search for not just natural product antibiotics but also for synthetic and semisynthetic compounds. This review revisits this epoch, using the research by a Danish pharmaceutical company, LEO Pharma, as an example. LEO adopted a strategy searching for synthetic antibiotics toward specific bacterial pathogens, in particular Mycobacterium tuberculosis, leading to the discovery of a new derivative of a known drug. Work on penicillin during and after WWII lead to the development of associated salts/esters and a search for new natural product antibiotics. This led initially to no new, marketable compounds, but concluded with the serendipitous discovery of fusidic acid, an antibiotic used to treat infections by Staphylococcus aureus, in 1960. The discovery process included contemporary approaches such as open innovation; targeting specific pathogens and/or specific organs in the patient; examining the effects of antimicrobial compounds on bacterial virulence as well as on antibiotic-resistant variants, and searching for antibiotic producers among microorganisms not previously well explored. These activities were promoted by the collaboration with a renowned Danish clinical microbiologist, K. A. Jensen, as well as company expertise in fermentation technologies, chemical synthesis and purification of bioactive compounds from organic materials.
Article
Full-text available
Infectious diseases that are caused by bacteria are an important cause of mortality and morbidity in all regions of the world. Bacterial drug resistance has grown in the last decades, but the rate of discovery of new antibiotics has steadily decreased. Therefore, the search for new effective antibacterial agents has become a top priority. The plant kingdom seems to be a deep well for searching for novel antimicrobial agents. This is due to the many attractive features of plants: they are readily available and cheap, extracts or compounds from plant sources often demonstrate high-level activity against pathogens, and they rarely have severe side effects. The huge variety of plant-derived compounds provides very diverse chemical structures that may supply both the novel mechanisms of antimicrobial action and provide us with new targets within the bacterial cell. In addition, the rapid development of modern biotechnologies opens up the way for obtaining bioactive compounds in environmentally friendly and low-toxic conditions. In this short review, we ask the question: do antibacterial agents derived from plants have a chance to become a panacea against infectious diseases in the “post-antibiotics era”.
Article
Full-text available
The trend toward designing large hydrophobic molecules for lead optimization is often associated with poor drug-likeness and high attrition rates in drug discovery and development. Structural simplification is a powerful strategy for improving the efficiency and success rate of drug design by avoiding "molecular obesity". The structural simplification of large or complex lead compounds by truncating unnecessary groups can not only improve their synthetic accessibility but also improve their pharmacokinetic profiles, reduce side effects and so on. This review will summarize the application of structural simplification in lead optimization. Numerous case studies, particularly those involving successful examples leading to marketed drugs or drug-like candidates, will be introduced and analyzed to illustrate the design strategies and guidelines for structural simplification.
Article
The pressing need for new treatments for multi‐drug resistant bacterial infections including MDR and XDR tuberculosis has led us to re‐explore the use of natural product antibiotics not normally used to treat target infections, especially those with unique mechanisms of action. Being the product of combinatorial evolution natural products have many advantages over purely synthetic antibacterial leads, but also some notable disadvantages often in regard to spectrum and human metabolism. This presentation will go over our efforts to advance under‐explored antibiotic scaffolds with semi‐synthetic approaches to tackle these disadvantages. Using spectinomycin as an example, synthetic modifications have been applied to overcome native bacterial efflux leading to potent leads with better than predicted in vivo efficacy in complex infection models. These observations have led us to examine active uptake and slow‐growing pathogen targeting hypotheses that are hard wired into many natural products, which are very hard to replicate by direct purely synthetic de novo antibacterial drug design. Support or Funding Information This work was supported by research grant R01AI090810 by the National Institutes of Health and the American Lebanese Syrian Associated Charities (ALSAC)
Chapter
Computer-aided drug designing (CADD) relates to drug discovery, also characterized as a cost-effective and active tool that manages or creates theoretical models that would be used by large databases for discovery and virtual screening. Till now, several algorithms have been developed and managed through CADD to study different prospects like protein structure and function prediction, identification of ligands interaction, residues of the active site, and study of protein–ligand interactions, which possibly leads to the discovery of newer therapeutic agents or drugs. As per in terms of new medicine discovery, designing and binding of small molecules (ligand) with DNA, RNA, or protein (target) is the key step, defined as docking. Docking actively identifies specific hit from large data libraries through simple rigid or flexible docking approaches with the receptor to maximize hit rates in virtual screening. The calculated scores of free energy of binding (poses) define the active compounds involved in interactions. Different new prospects in docking programs are now being used that more focuses accuracy on molecular interaction energy calculation without stringent parameters. The quantum-chemical methods, implicit solvent models, and new global optimization algorithms are now being used to improve flexibility and mobility of ligands and proteins, respectively. This chapter presents some basic algorithms, molecular docking programs based on rigid and flexible receptor/ligand-based on various machine learning techniques used in CADD and molecular docking.
Article
This review summarizes main advances achieved by Russian researchers in the synthesis and characterization of semi-synthetic antibiotics of a new generation in the period from 2004 to 2019. The following classes of compounds are considered as the basis for modification: polycyclic antibacterial glycopeptides of the vancomycin group, classical macrolides, antifungal polyene macrolides, the antitumour antibiotic olivomycin A, antitumour anthracyclines and broad-spectrum antibiotics, in particular, oligomycin A, heliomycin and some other. Main trends in the design of modern anti-infective and antitumour agents over this period are considered in relation to original natural antibiotics, which have been independently discovered by Russian researchers. It is shown that a new type of hybrid structures can, in principle, be synthesized based on glycopeptides, macrolides and other antibiotics, including heterodimers containing a new benzoxaborole pharmacophore. The review addresses the influence of the length of the spacer between two antibiotic molecules on the biological activity of hybrid structures. A combination of genetic engineering techniques and methods of organic synthesis is shown to be useful for the design of new potent antifungal antibiotics based on polyenes of the amphotericin B group. Many new semi-synthetic analogues exhibit important biological properties, such as a broad spectrum of activity and low toxicity. Emphasis is given to certain aspects related to investigation of a broad range of biological activity and mechanisms of action of new derivatives. The bibliography includes 101 references.
Article
One effective means to achieve inhibitor specificity for RAF kinases, an important family of cancer drug targets, has been to target the monomeric inactive state conformation of the kinase domain, which, unlike most other kinases, can accommodate sulfonamide-containing drugs such as vemurafenib and dabrafenib because of the presence of a unique pocket specific to inactive RAF kinases. We previously reported an alternate strategy whereby rigidification of a nonselective pyrazolo[3,4-d]pyrimidine-based inhibitor through ring closure afforded moderate but appreciable increases in selectivity for RAF kinases. Here, we show that a further application of the rigidification strategy to a different pyrazolopyrimidine-based scaf-fold dramatically improved selectivity for RAF kinases. Crystal structure analysis confirmed our inhibitor design hypothesis revealing that 2l engages an active-like state conformation of BRAF normally associated with poorly discriminating inhibitors. When screened against a panel of distinct cancer cell lines, the optimized inhibitor 2l primarily inhibited the proliferation of the expected BRAFV600E-harboring cell lines consistent with its kinome selectivity profile. These results suggest that rigidification could be a general and powerful strategy for enhancing inhibitor selectivity against protein kinases, which may open up therapeutic opportunities not afforded by other approaches.