Preface
Page: i-ii (2)
Author: Igor José dos Santos Nascimento*
DOI: 10.2174/9789815179934123010001
PDF Price: $15
Ligand and Structure-Based Drug Design (LBDD and SBDD): Promising Approaches to Discover New Drugs
Page: 1-32 (32)
Author: Igor José dos Santos Nascimento* and Ricardo Olimpio de Moura
DOI: 10.2174/9789815179934123010003
PDF Price: $15
Abstract
The drug discovery and development process are challenging and have
undergone many changes over the last few years. Academic researchers and
pharmaceutical companies invest thousands of dollars a year to search for drugs
capable of improving and increasing people's life quality. This is an expensive, time-consuming, and multifaceted process requiring the integration of several fields of
knowledge. For many years, the search for new drugs was focused on Target-Based
Drug Design methods, identifying natural compounds or through empirical synthesis.
However, with the improvement of molecular modeling techniques and the growth of
computer science, Computer-Aided Drug Design (CADD) emerges as a promising
alternative. Since the 1970s, its main approaches, Structure-Based Drug Design
(SBDD) and Ligand-Based Drug Design (LBDD), have been responsible for
discovering and designing several revolutionary drugs and promising lead and hit
compounds. Based on this information, it is clear that these methods are essential in
drug design campaigns. Finally, this chapter will explore approaches used in drug
design, from the past to the present, from classical methods such as bioisosterism,
molecular simplification, and hybridization, to computational methods such as docking,
molecular dynamics (MD) simulations, and virtual screenings, and how these methods
have been vital to the identification and design of promising drugs or compounds.
Finally, we hope that this chapter guides researchers worldwide in rational drug design
methods in which readers will learn about approaches and choose the one that best fits
their research.
Quantitative Structure-activity Relationship (QSAR) in Studying the Biologically Active Molecules
Page: 33-56 (24)
Author: Serap ÇETINKAYA, Burak TÜZÜN* and Emin SARIPINAR
DOI: 10.2174/9789815179934123010004
PDF Price: $15
Abstract
Recently, many new methods have been used in the research and
development of a new drug. In this article, QSAR, which is one of the usable areas of
artificial intelligence during molecule research, and the analysis and formulation
studies related to the suitability of this area are discussed. It is explained how a model
to be created is prepared and calculation formulas for how to verify this model are
shown. Examples of the most recent 4D-QSAR calculations are given.
Pharmacophore Mapping: An Important Tool in Modern Drug Design and Discovery
Page: 57-115 (59)
Author: Dharmraj V. Pathak, Abha Vyas, Sneha R. Sagar, Hardik G. Bhatt and Paresh K. Patel*
DOI: 10.2174/9789815179934123010005
PDF Price: $15
Abstract
Computer-Aided Drug Design (CADD) has become an integral part of drug
discovery and development efforts in the pharmaceutical and biotechnology industry.
Since the 1980s, structure-based design technology has evolved, and today, these
techniques are being widely employed and credited for the discovery and design of
most of the recent drug products in the market. Pharmacophore-based drug design
provides fundamental approach strategies for both structure-based and ligand-based
pharmacophore approaches. The different programs and methodologies enable the
implementation of more accurate and sophisticated pharmacophore model generation
and application in drug discovery. Commonly used programmes are GALAHAD,
GASP, PHASE, HYPOGEN, ligand scout etc. In modern computational chemistry,
pharmacophores are used to define the essential features of one or more molecules with
the same biological activity. A database of diverse chemical compounds can then be
searched for more molecules which share the same features located at a similar distance
apart from each other. Pharmacophore requires knowledge of either active ligands
and/or the active site of the target receptor. There are a number of ways to build a
pharmacophore. It can be done by common feature analysis to find the chemical
features shared by a set of active compounds that seem commonly important for
receptor interaction. Alternately, diverse chemical structures for certain numbers of
training set molecules, along with the corresponding IC50 or Ki values, can be used to
correlate the three-dimensional arrangement of their chemical features with the
biological activities of training set molecules. There are many advantages in
pharmacophore based virtual screening as well as pharmacophore based QSAR, which
exemplify the detailed application workflow. Pharmacophore based drug design
process includes pharmacophore modelling and validation, pharmacophore based
virtual screening, virtual hits profiling, and lead identification. The current chapter on
pharmacophores also describes case studies and applications of pharmacophore
mapping in finding new drug molecules of specific targets.
Up-to-Date Developments in Homology Modeling
Page: 116-135 (20)
Author: Muhammed Tilahun Muhammed* and Esin Aki-Yalcin
DOI: 10.2174/9789815179934123010006
PDF Price: $15
Abstract
Homology modeling is used to predict protein 3D structure from its amino
acid sequence. It is the most accurate computational approach to estimate 3D
structures. It has straightforward steps that save time and labor. There are several
homology modeling tools under use. There is no sole tool that is superior in every
aspect. Hence, the user should select the most appropriate one carefully. It is also a
common practice to use two or more tools at a time and choose the best model among
the resulting models.
Homology modeling has various applications in the drug design and development
process. Such applications need high-quality 3D structures. It is widely used in
combination with other computational methods including molecular docking and
molecular dynamics simulation. Like the other computational methods, it has been
influenced by the involvement of artificial intelligence. In this regard, homology
modeling tools, like AlphaFold, have been introduced. This type of method is expected
to contribute to filling the gap between protein sequence release and 3D structure
determination.
This chapter sheds light on the history, relatively popular tools and steps of homology
modeling. A detailed explanation of MODELLER is also given as a case study
protocol. Furthermore, homology modeling’s application in drug discovery is explained
by exemplifying its role in the fight against the novel Coronavirus. Considering the
new advances in the area, better tools and thus high-quality models are expected.
These, in turn, pave the way for more applications of it.
Anticancer Activity of Medicinal Plants Extract and Molecular Docking Studies
Page: 136-158 (23)
Author: Serap ÇETINKAYA and Burak TÜZÜN*
DOI: 10.2174/9789815179934123010007
PDF Price: $15
Abstract
Molecular docking involves the interaction of a molecule with another place,
usually in the protein structure, and simulating the placement of the molecule in the
protein structure with certain score algorithms, taking into account many quantities,
such as the electro-negativity of atoms, their positions to each other, and the
conformation of the molecule to be inserted into the protein structure. Finally, the
activity of the molecule with the highest percentage by mass against various cancer
proteins was investigated according to the GC-MS results made on some medicinal and
aromatic plants in order to set an example of molecular docking calculations.
FBDD & De Novo Drug Design
Page: 159-201 (43)
Author: Anwesha Das, Arijit Nandi, Vijeta Kumari and Mallika Alvala*
DOI: 10.2174/9789815179934123010008
PDF Price: $15
Abstract
Fragment-based drug or lead discovery (FBDD or FBLD) refers to as one of
the most significant approaches in the domain of current research in the pharmaceutical
industry as well as academia. It offers a number of advantages compared to the
conventional drug discovery approach, which include – 1) It needs the lesser size of
chemical databases for the development of fragments, 2) A wide spectrum of
biophysical methodologies can be utilized for the selection of the best fit fragments
against a particular receptor, and 3) It is far more simpler, feasible, and scalable in
terms of the application when compared to the classical high-throughput screening
methods, making it more popular day by day. For a fragment to become a drug
candidate, they are analyzed and evaluated on the basis of numerous strategies and
criteria, which are thoroughly explained in this chapter. One important term in the field
of FBDD is de novo drug design (DNDD), which means the design and development of
new ligand molecules or drug candidates from scratch using a wide range of in silico
approaches and algorithmic tools, among which AI-based platforms are gaining large
attraction. A principle segment of AI includes DRL that finds numerous applicabilities
in the DNDD sector, such as the discovery of novel inhibitors of BACE1 enzyme,
identification and optimization of new antagonists of DDR1 kinase enzyme, and
development and design of ligand molecules specific to target adenosine A2A, etc. In
this book chapter, several aspects of both FBDD and DNDD are briefly discussed.
Molecular Simulation in Drug Design: An Overview of Molecular Dynamics Methods
Page: 202-257 (56)
Author: Fernando D. Prieto-Martínez*, Yelzyn Galván-Ciprés and Blanca Colín-Lozano
DOI: 10.2174/9789815179934123010009
PDF Price: $15
Abstract
Molecular interaction is the basis for protein and cellular function. Careful
inhibition or modulation of these is the main goal of therapeutic compounds. In the
pharmaceutical field, this process is referred to as pharmacodynamics. Over the years,
there have been several hypotheses attempting to describe this complex phenomenon.
From a purely biophysical point of view, molecular interactions may be attributed to
pairwise contributions such as charge angles, torsions, and overall energy. Thus, the
computation of binding affinity is possible, at least in principle. Over the last half of the
past century, molecular simulation was developed using a combination of physics,
mathematics, and thermodynamics. Currently, these methods are known as structure-based drug design (SBDD) and it has become a staple of computer-aided drug design
(CADD). In this chapter, we present an overview of the theory, current advances, and
limitations of molecular dynamics simulations. We put a special focus on their
application to virtual screening and drug development.
Quantum Chemistry in Drug Design: Density Function Theory (DFT) and Other Quantum Mechanics (QM)-related Approaches
Page: 258-309 (52)
Author: Samuel Baraque de Freitas Rodrigues, Rodrigo Santos Aquino de Araújo, Thayane Regine Dantas de Mendonça, Francisco Jaime Bezerra Mendonça-Júnior, Peng Zhan and Edeildo Ferreira da Silva-Júnior*
DOI: 10.2174/9789815179934123010010
PDF Price: $15
Abstract
Drug design and development are expensive and time-consuming processes,
which in many cases result in failures during the clinical investigation steps. In order to
increase the chances to obtain potential drug candidates, several in silico approaches
have emerged in the last years, most of them based on molecular or quantum
mechanics theories. These computational strategies have been developed to treat a
large dataset of chemical information associated with drug candidates. In this context,
quantum chemistry is highlighted since it is based on the Schrödinger equation with
mathematic solutions, especially the Born-Oppenheimer approximation. Among the
Hartree-Fock-based methods, the Density Functional Theory (DFT) of HohenbergKohn represents an interesting and powerful tool to obtain accurate results for
electronic properties of molecules or even solids, which in many cases are corroborated
by experimental data. Additionally, DFT-related methods exhibit a moderate time-consuming cost when compared to other ab initio methods. In this chapter, we provide
a deep overview focused on the formalism behind DFT, including historical aspects of
its development and improvements. Moreover, different examples of the application of
DFT in studies involving GABA inhibitors, or catalytic mechanisms of enzymes, such
as RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2, and different proteases associated impacting diseases, such as malaria, Chagas disease, human African
trypanosomiasis, and others. Moreover, the role of metal ions in catalytic enzymatic
mechanisms is also covered, discussing iron-, copper-, and nickel-catalyzed processes.
Finally, this chapter comprises several aspects associated with the elucidation of
catalytic mechanisms of inhibition, which could be used to develop new potential
pharmacological agents.
Free Energy Estimation for Drug Discovery: Background and Perspectives
Page: 310-345 (36)
Author: Fernando D. Prieto-Martínez* and Yelzyn Galván-Ciprés
DOI: 10.2174/9789815179934123010011
PDF Price: $15
Abstract
Drug development is a remarkably complex subject, with potency and
specificity being the desired traits in the early stages of research. Yet, these need
careful thought and rational design, which has led to the inclusion of multidisciplinary
efforts and non-chemistry methods in the ever-changing landscape of medicinal
chemistry. Computational approximation of protein-ligand interactions is the main goal
of the so-called structure-based methods. Over the years, there has been a notable
improvement in the predictive power of approaches like molecular force fields.
Mainstream applications of these include molecular docking, a well-known method for
high-throughput virtual screening. Still, even with notable success cases, the search for
accurate and efficient methods for free energy estimation remains a major goal in the
field. Recently, with the advent of technology, more exhaustive simulations are
possible in a reasonable time. Herein, we discuss free energy predictions and
applications of perturbation theory, with emphasis on their role in molecular design and
drug discovery. Our aim is to provide a concise but comprehensive view of current
trends, best practices, and overall perspectives in this maturing field of computational
chemistry.
Subject Index
Page: 346-350 (5)
Author: Igor José dos Santos Nascimento*
DOI: 10.2174/9789815179934123010012
PDF Price: $15
Introduction
Designing and developing new drugs is an expensive and time-consuming process, and there is a need to discover new tools or approaches that can optimize this process. Applied Computer-Aided Drug Design: Models and Methods compiles information about the main advances in computational tools for discovering new drugs in a simple and accessible language for academic students to early career researchers. The book aims to help readers understand how to discover molecules with therapeutic potential by bringing essential information about the subject into one volume. Key Features . Presents the concepts and evolution of classical techniques, up to the use of modern methods based on computational chemistry in accessible format. . Gives a primer on structure- and ligand-based drug design and their predictive capacity to discover new drugs. . Explains theoretical fundamentals and applications of computer-aided drug design. . Focuses on a range of applications of the computations tools, such as molecular docking; molecular dynamics simulations; homology modeling, pharmacophore modeling, quantitative structure-activity relationships (QSAR), density functional theory (DFT), fragment-based drug design (FBDD), and free energy perturbation (FEP). . Includes scientific reference for advanced readers