View Article

Abstract

Drug design is a multidisciplinary process aimed at developing new therapeutic agents by understanding molecular targets and mechanisms of action. This review summarizes key strategies in drug design, including structure-based, ligand-based, and computational approaches, while emphasizing the role of molecular modeling and bioinformatics in modern drug discovery. Unlike traditional discovery methods that relied on trial-and-error, modern drug design focuses on understanding the structure and function of biological targets such as proteins, enzymes, or receptors. Using this knowledge, researchers design small molecules or biologics that can interact with these targets in a specific way, similar to how a key fits into a lock. A key element of contemporary pharmaceutical research is drug design, which combines the fields of chemistry, biology, and computational sciences to produce potent therapeutic agents. Drug discovery has significantly accelerated while lowering costs and failures thanks to strategies like structure-based and ligand-based design, which are backed by computational tools like molecular docking, pharmacophore modeling, QSAR, and ADMET analysis. Even though problems like toxicity, drug resistance, and high attrition rates still exist, improvements in high-throughput screening, machine learning, and artificial intelligence are improving precision and effectiveness. All things considered, drug design is still crucial to creating novel, secure, and focused medications that will influence the direction of individualized healthcare in the future.

Keywords

CADD, Pharmacophore , Docking , QSAR , ADMET

Introduction

Drug design is the scientific process of creating new medicines by identifying chemical structures that can interact with specific biological targets (such as proteins or enzymes) in the body to treat diseases effectively and safely. It combines knowledge of chemistry, biology, and pharmacology to design molecules with the desired therapeutic effect.

The Identification of novel molecular entities that could be useful in treating illnesses that meet the criteria for presenting unmet medical needs is known as a discovery campaign needs. These diseases do not have definitively useful therapies and are actually or potentially life-threatening. Marketed drugs at this point represent are relatively small number of drug target types.1

DEFINE OF DRUG DESIGN :    

Drug design involves the identification and optimization of biologically active molecules that can interact with specific targets to produce therapeutic effects. The process integrates medicinal chemistry, pharmacology, and computational tools to enhance efficacy and reduce toxicity.2

SCOPE OF DRUG DESIGN :    

It includes:  

  1. Target Identification & Validation   
  2. Lead Compound Discovery   
  3. Optimization   
  4. Computer-Aided Drug Design (CADD)   
  5. Biotechnology & Advanced Therapies –   
  6. Interdisciplinary Role   
  7. Future Prospects  

OBJECTIVE :    

  1. To find and create novel chemical compounds that have the ability to interact with particular biological targets.   
  2. To maximize effectiveness and minimize toxicity while achieving the intended therapeutic effect.  
  3. To maximize the drug candidates' pharmacodynamic and pharmacokinetic characteristics.  
  4. To reduce negative effects while improving safety and selectivity.  

DRUG DESIGN AS A MULTI-DISCIPLINARY APPROACH:    

Drug design is not limited to just chemistry; it requires the integration of several scientific fields. It is a multidisciplinary approach because:   

  1. Medicinal Chemistry   
  2. Biology & Molecular Biology   
  3. Pharmacology   
  4. Computational Sciences  
  5. Pharmaceutics
  6. Toxicology 3

Difference between Rational and Empirical Drug Design:  

Aspect of Rational Drug Design :   

  • Basis/Approach : Based on knowledge of the biological target (enzyme, receptor, protein).  
  • Method : Uses molecular modeling, structure–activity relationships (SAR/QSAR), docking, and computational tools.  
  • Efficiency: More efficient, time-saving, and cost-effective. 
  • Specificity: Produces highly specific drugs with fewer side effects.  
  • Example : Designing ACE inhibitors by knowing the structure of angiotensin-converting enzyme 4

Aspect Empirical Drug Design :   

  • Basis/Approach : Based on trial-and-error screening of many compounds without full knowledge of the target.  
  • Method : Synthesizes and tests large numbers of compounds experimentally to check activity.  
  • Efficiency: Less efficient, more time-consuming, and expensive.  
  • Specificity : Less specific, often leads to compounds with unwanted effects.  
  • Example : Early antibiotics like penicillin discovered by chance observation.5

HISTORICAL PERSPECTIVE:

Natural Product-Based Drug Discovery:-   

Natural products (compounds derived from plants, microorganisms, or marine organisms) have been a foundation of drug discovery throughout history. They represent some of the earliest and most successful sources of medicines.6

1) Ancient Use :   

Traditional systems like Ayurveda, Chinese medicine, and Egyptian medicine used plant extracts for treating diseases.  

Example: Willow bark was used for pain relief in ancient Greece (later leading to aspirin).  

2) 19th Century Milestones   

Morphine (1806): Isolated from opium poppy (Papaver somniferum), used as a strong analgesic.  

Quinine (1820): Derived from Cinchona bark, a treatment for malaria.  

Cocaine (1859): Isolated from coca leaves, used as a local anesthetic.  

3) 20th Century Developments  

Penicillin (1928): Discovered from Penicillium fungus by Alexander Fleming, revolutionized antibacterial therapy.

Paclitaxel (Taxol®) (1967): Derived from Pacific yew tree (Taxus brevifolia), used in cancer treatment.  

Cyclosporine (1970s): From fungus Tolypocladium inflatum, a breakthrough in organ transplantation.  

4) Modern Perspective  

Natural products remain vital for novel drug scaffolds.  

Over 50% of approved drugs (especially anticancer and anti-infectives) are either natural products or their derivatives.7  

Role of natural products in traditional medicine and early pharmacology  

In Traditional Medicine-  

1. Backbone of Ancient Healthcare:

Plants, minerals, and animal   products were the primary source of remedies in Ayurveda, Traditional Chinese Medicine, Egyptian, and Greek systems.  

2. Examples:   

Willow bark → used for fever and pain (later source of aspirin).  

Neem, turmeric, garlic → used in Ayurveda for antimicrobial and healing effects. ? Cinchona bark → used for malaria in South America.  

3. Holistic approach:

Treatments were based on natural mixtures, focusing on balance and wellness, not just disease cure.4 

Notable Success Stories of Natural Products in Drug Discovery  

1. Penicillin  

Source: Penicillium mold (fungus).  

Discovery: In 1928, Alexander Fleming observed that the mold inhibited the growth of Staphylococcus bacteria.  

Importance: First antibiotic ever discovered; effective against bacterial infections such as pneumonia, syphilis, and wound infections.  

Impact: Launched the antibiotic era, saving millions of lives, especially during World War II, and remains the basis for many β-lactam antibiotics today.8

2. Morphine   

Source: Opium poppy (Papaver somniferum).  

Discovery: Isolated in 1806 by Friedrich Sertürner, marking the first time an active principle was purified from a plant.  

Importance: Potent analgesic far more powerful than raw opium.  

Impact: Opened the door to the field of alkaloid chemistry and established the scientific basis of pharmacology by linking pure compounds to biological activity.  

3. Artemisinin   

Source: Artemisia annua (sweet wormwood), a plant used in Traditional Chinese Medicine.  

Discovery: Isolated in the 1970s by and her team during China’s Project 523.  

Importance: Highly effective against malaria, especially drug-resistant strains.  

Impact: Became the cornerstone of Artemisin -based Combination Therapies (ACTs), saving millions of lives in malaria-endemic countries. To was awarded the 2015 Nobel Prize in Medicine for this discovery.9  

EMERGENCE OF COMPUTATIONAL DRUG DESIGN:   

The development of computer technology and molecular biology in the latter half of the 20th century gave rise to the idea of computational drug design, or CDD. Trial-and-error screening of synthetic and natural products has been a major component of drug discovery in the past.But when three-dimensional protein and nucleic acid structures were made available by molecular biology methods (like X-ray crystallography and NMR spectroscopy), scientists started modeling drug-target interactions on computers.10 Today, computational drug design is widely used in:  

Virtual screening of large chemical libraries  

Structure-based drug design (SBDD) using 3D protein structures  

Ligand-based drug design (LBDD) using QSAR and pharmacophore models  

Predictive modeling for ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity)  

AI-driven drug discovery for accelerating candidate identification.11

Shift from Trial-and-Error to In Silico Methods:   

1. Traditional Trial-and-Error Approaches  

For centuries, drug discovery was empirical, based mainly on natural products, herbal remedies, and random screening of compounds. Researchers tested thousands of compounds in bioassays to find one with therapeutic activity. While this approach led to  some major discoveries (e.g., penicillin, morphine, aspirin), it was: Time-consuming ,Expensive , Low success rate 12  

Timeline of major computational advancements (QSAR, molecular modeling, Al Integration) 

  • 1960s: QSAR (Hansch–Fujita)   
  • 1970s: Molecular modeling (Levitt & Warshel)   
  • 1980s: Docking (Kuntz)   
  • 1990s: 3D-QSAR & CoMFA (Cramer et al.)   
  • 2000s: Virtual screening & MD (Kitchen et al.)   
  • 2010s: Machine learning (Vamathevan et al.)   
  • 2020s: AI & AlphaFold (Jumper et al.) 14

Advantages of Integrating Computational and Natural Product–Based 

Approaches  

  • Efficient Identification of Bioactive Compounds  
  • Optimization of Lead Structures   
  • Target Identification and Mechanistic Insights  
  • Reduction of Experimental Costs and Time 7 

Natural Products as Lead Compounds in Drug Discovery  

Historical Importance – Natural products have been the oldest source of medicines, from traditional remedies to modern drugs.  

Examples: Morphine 

Chemical Diversity and Structural Complexity-    

Natural products provide unique, stereochemically rich, and complex scaffolds often difficult to design synthetically.  

High Biological Relevance - Many natural products evolved as defense molecules in plants, fungi, or microbes, making them biologically active against human disease targets.13

Overview of Bioactive Natural Products  

Bioactive natural products are a rich source of drugs and lead compounds, spanning antibiotics, anticancer agents, antimalarials, and immunosuppressants.  

a. Classification of Alkaloids –   

Classification Based on Chemical Structure  

  1. True Alkaloids   
  2. Protoalkaloids (Aminoalkaloids)  
  3. Pseudoalkaloids  

Classification Based on Biosynthetic Origin  

  1. Indole Alkaloids  
  2. Isoquinoline Alkaloids  
  3. Tropane Alkaloids  
  4. Pyrrolizidine Alkaloids  
  5. Quinoline Alkaloids 14

b. Classification of Terpenoids   

  1. Hemiterpenoids (C?)   
  2. Monoterpenoids (C??)   
  3. Sesquiterpenoids (C??)  
  4. Diterpenoids (C??)  
  5. Sesterterpenoids (C??)15

c. Classification of flavonoids   

  1. Flavones   
  2. Flavonols  
  3. Flavanones  
  4. Flavanols  
  5. Isoflavone 16

Mechanisms of Action in Therapeutics  

The mechanism of action (MOA) refers to the specific biochemical or physiological interaction through which a drug produces its therapeutic effect.  

  1. Receptor-Mediated Mechanisms  
  2. Enzyme Inhibition or Activation  
  3. Ion Channel Modulation  
  4. Transporter Modulation  
  5. Immune System Modulation 17  

Examples of Natural Products Used as Drugs  

Paclitaxel, Quinine, Aspirin, Morphine  

Limitations of Natural Products   

Low bioavailability – low bioavailability is a major limitation because it prevents many natural products (like curcumin, resveratrol, paclitaxel) from reaching effective concentrations in the body, even though they show strong therapeutic activity in the lab.   

Poor solubility - is a major limitation because it      reduces absorption, lowers bioavailability, and complicates dosage form development, even for highly potent natural products like curcumin, paclitaxel, and camptothecin.  

Synthesis challenges -Synthetic challenges arise because many natural products are too complex to be efficiently synthesized in the lab. This limits supply, increases cost, and delays clinical development — as seen with paclitaxel, artemisinin, and vancomycin.18  

Need for Optimization:    

Rational Derivatization Strategies    

Rational derivatization strategies are needed to optimize natural products by enhancing pharmacokinetics, reducing toxicity, improving selectivity, overcoming resistance, and facilitating production — thereby converting natural scaffolds into successful drugs.  

Scaffold modification and hybrid molecule design   

The need for optimization arises because raw natural products often have drawbacks. Scaffold modification makes them more drug-like by fine-tuning their structure, while hybrid molecule design creates novel compounds that combine the benefits of multiple pharmacophores, enhancing potency, selectivity, and resistance management.19 

Role of QSAR in Lead Optimization  

Introduction to QSAR   

QSAR is a computational method in medicinal chemistry that establishes a quantitative relationship between the chemical structure of compounds and their biological activity.   

2D-QSAR : Uses 2D descriptors derived from molecular structure without considering 3D conformation.   

Mathematical Basis of 2D-QSAR   

  1. Hansch Equation
  2. Free–Wilson Analysis

3D-QSAR : Uses three-dimensional spatial information of molecules and their interaction with a biological target.   

Mathematical Basis of 2D-QSAR   

CoMFA Equation.20 

Types of descriptors :  

Physicochemical Descriptors : Describe physical and chemical properties of molecules that influence their interaction with biological systems.   

Topological Descriptors : Describe molecular connectivity and shape without requiring 3D coordinates. Derived from graph theory representation of molecules.   

Electronic Descriptors : Describe electron distribution and reactivity in molecules. These influence binding affinity, reaction rates, and interactions with biological targets.21   

Key Steps in QSAR Modeling  

1. Dataset Preparation :   

Goal: Collect and organize reliable biological and chemical data for QSAR analysis.   

Steps :    

  1. Data collection   
  2. Standardization of structures   
  3. Activity scaling   
  4. Dataset division   

2. Descriptor Selection : Choose descriptors that significantly correlate with activity using statistical methods:  

3. Model validation :

  1. Internal validation  
  2. External validation   
  3. Statistical checks 22

Case Studies Demonstrating QSAR

QSAR analysis of anti-cancer compounds :   

a) Quinazoline Derivatives as EGFR Inhibitors   

Details: Quinazoline scaffolds are widely used as Epidermal Growth Factor Receptor (EGFR) inhibitors (e.g., Gefitinib, Erlotinib).  

QSAR Findings: Electronic descriptors (Hammett σ values) of substituents on the aniline moiety strongly influenced activity.    

Outcome: Improved understanding of how electron-donating groups at the para-position enhance EGFR inhibition.  

b) Flavonoid Analogues as Anti-Cancer Agents   

Details: Flavonoids are natural products with anti-oxidant and cytotoxic effects.  

QSAR Findings: Topological descriptors showed hydroxyl groups at 3′ and 4′ enhanced DNA intercalation and apoptosis induction.  

Outcome: QSAR provided modification rules for enhancing activity in synthetic flavonoid analogs.  

QSAR analysis of Anti-Tubercular Compounds:   

a) Isoniazid Derivatives   

Details: Isoniazid is a first-line TB drug, acting through inhibition of mycolic acid biosynthesis.   

QSAR Findings: Lipophilicity within an optimum range improved activity (too high → reduced solubility; too low → poor cell penetration).Steric bulk near the isonicotinoyl group reduced activity.  

Outcome: QSAR helped rationalize resistance and optimize analog design.  

b) Diarylquinoline Derivatives (e.g., Bedaquiline)   

Details: Diarylquinoline target ATP synthase in M. tuberculosis.  

QSAR Findings: 3D-QSAR (CoMFA/CoMSIA) showed steric and electrostatic contour maps predicting where bulky or electropositive groups improved binding.

Outcome: Supported optimization of Bedaquiline analogs with higher potency and reduced toxicity.23

Integration with Other Tools  

QSAR + docking + ADMET models in optimization pipelines  

1. QSAR (Quantitative Structure–Activity Relationship)  

Function: Correlates chemical descriptors (electronic, hydrophobic, steric, topological) with biological activity.  

Use in pipeline: Virtual screening of compound libraries. Identifies structural features critical for potency.24 

2. Molecular Docking   

Function: Simulates the orientation and interactions of drug candidates in the target’s binding site.  

Use in pipeline: Confirms QSAR-predicted hits by showing how they fit into the protein pocket.   

Identifies hydrogen bonding, hydrophobic interactions, and steric clashes.  

3. ADMET Modeling   

Function: Predicts drug-like behavior—Absorption, Distribution, Metabolism, Excretion, and Toxicity.  

Use in pipeline: Filters compounds with poor solubility, permeability, or high predicted toxicity.   

Ensures optimized candidates are not only potent but also safe and bioavailable.25

Structure-Based Drug Design (SBDD):    

Overview of SBDD   

Structure-Based Drug Design (SBDD) uses the 3D structure of biological targets to design ligands rationally. It combines docking, virtual screening, fragment-based design, pharmacophore modeling, and MD simulations to optimize drug candidates for potency, selectivity, and safety.26  

Difference between structure-based and ligand-based approaches  

 Structure based Drug Design ( SBDD )   

  1. Definition   - Uses the 3D structure of the target protein to design or optimize ligands.   
  2. Requirement- Requires experimentally determined target structure (X-ray crystallography, NMR, cryoEM).  
  3. Strengths - Provides atomic-level insight into ligand–target interactions. <br> - Can guide rational optimization of potency and selectivity.  - Useful when target structure is unknown.  
  4. Limitations - Depends on high-quality target structure. <br> - Computationally intensive (docking, MD). <br> - Scoring functions not always accurate.   

Ligand-Based Drug Design (LBDD)  

  1. Defination - Uses the structural and biological data of known ligands (without target structure) to build predictive models.  
  2. Requirement – Requires a dataset of active ligands with measured biological   
  3. Strengths – Useful when target structure is unknown. <br> - Relies only on experimental activity data.  <br> - Can capture SAR trends across chemical series.   
  4. Limitations – Limited by the quality and size of ligand dataset. <br> - Cannot reveal exact binding mode without target structure .27

Molecular Docking   

Theory :   

Binding Energy Concept - 

The binding energy (ΔG_bind) estimates the stability of the protein–ligand complex. \Delta G_{\text{bind}}= G_{\text{complex}}- (G_{\text{protein}} + G_{\text{ligand}})      ΔG_bind

< 0 → spontaneous and favorable binding.   

Scoring functions - Scoring functions estimate the binding affinity of a ligand for a protein based on the predicted pose.28

PHARMACOPHORE MODELING  

Definition and use in virtual screening   

Define- A pharmacophore is “an ensemble of steric and electronic features that is necessary to ensure the optimal supramolecular interactions with a specific biological target and to trigger (or block) its biological response.”  

Use of Pharmacophore Modeling in Virtual Screening  

  1. Ligand-Based Virtual Screening  
  2. Hit Identification and Optimization  
  3. Scaffold Hopping  
  4. Complement to Docking 29

Docking Workflow and Analysis  

Step-by-step: receptor preparation - ligand preparation – grid generation - docking – analysis.   

1. Receptor Preparation   

Purpose: Ensure the protein structure is suitable for docking.   

Steps:   

  1. Obtain 3D structure from PDB or homology models.   
  2. Remove crystallographic water molecules (unless involved in binding).  
  3. Add missing hydrogen atoms (especially polar hydrogens).  
  4. Assign proper protonation states for residues at physiological pH  
  5. Minimize or relax the structure to relieve steric clashes.  

2. Ligand Preparation   

Purpose: Generate correct ligand structures for docking.  

Steps   

  1. Draw or retrieve ligand structure from PubChem, ChemSpider, or databases.  
  2. Add hydrogen atoms, assign correct bond orders.  
  3. Generate tautomeric and ionization states.  
  4. Optimize 3D geometry (energy minimization).  

3. Grid Generation   

Purpose: Define the region of the receptor where docking will occur.  

Steps:  

  1. If the binding site is known: center the grid box on the active site (ligand co-crystallized position or key residues).  
  2. If the binding site is unknown: use blind docking, where the grid covers the entire protein surface.  
  3. Set grid box dimensions large enough to cover the binding pocket.  

4. Docking Execution   

Purpose: Predict binding poses and affinities.  

Steps:  

  1. Place ligand conformations within the grid.  
  2. Perform conformational sampling (translation, rotation, torsional flexibility).  
  3. Evaluate poses using scoring functions (binding free energy approximation).  
  4. Rank poses based on docking score.  

5. Post-Docking Analysis   

Purpose: Interpret docking results to identify potential leads.  

Steps:  

  1. Analyze docking scores (binding affinity values).  
  2. Inspect binding poses and interactions (hydrogen bonds, hydrophobic contacts, π–π stacking, salt bridges).  
  3. Compare docking poses with reference ligands (if available).  
  4. Visualize complexes using PyMOL, Chimera, Discovery Studio, etc. 30 

ADMET

What is ADMET?   

ADMET stands for Absorption, Distribution, Metabolism, Excretion, and Toxicity. It describes the pharmacokinetic and safety profile of a compound in biological systems.  

Significance of ADMET in Drug Development  

1. Early Identification of Failures  

A large fraction of drug candidates fails in clinical trials due to poor pharmacokinetics or unexpected toxicity.  

2. Improves Success Rate of Lead Compounds  

By integrating ADMET profiling at the hit-to-lead and lead optimization stages, compounds with favorable absorption (bioavailability), metabolic stability, and low toxicity can be prioritized.  

3. Reduces Cost and Time  

Incorporating in silico ADMET models allows virtual filtering of large libraries before synthesis/testing.  

4. Ensures Patient Safety  

Toxicity assessment (hepatotoxicity, cardiotoxicity, mutagenicity, etc.) prevents late-stage failures.  

Common reasons for drug failure in clinical trials   

    1. Poor Absorption & Low Bioavailability  
    2. Unfavorable Distribution  
    3. Metabolic Instability  
    4. Inefficient Excretion  
    5. Toxicity  
    6. Drug-Drug Interaction 31

Importance of early prediction in design phase  

1. Reduces Late-Stage Failures - Most drug candidates fail in Phase II or III clinical trials due to poor ADMET properties (especially toxicity and poor bioavailability).  

2. Saves Time and Development Costs -Clinical trials are extremely costly.

3. Enhances Drug-Like Properties – ensures better oral bioavailability, permeability, and metabolic stability.  

4. Improves Safety and Efficacy – Avoids advancing compounds that may cause severe adverse effects in humans.  

5. Facilitates Rational Drug Design - Integrating computational ADMET models, docking, QSAR, and machine learning in the design phase ensures rational selection of lead molecules.32

IN SILICO ADMET TOOLS AND PARAMETERS –  

Tools 

1. SwissADME   

Description: A free web-based tool used for predicting physicochemical properties, pharmacokinetics, drug-likeness, and medicinal chemistry friendliness of small molecules.  

Significance: Helps in early assessment of oral bioavailability, brain penetration, and metabolic liabilities.  

2. ADMET SAR   

Description: A comprehensive tool for predicting ADMET properties using quantitative structure– activity relationship (QSAR) models.  

Significance: Provides an early toxicity and metabolism profile to filter out unsafe or poorly metabolized compounds.  

3. pkCSM   

Description: Uses graph-based signatures to predict ADMET properties and guide lead optimization.  

Significance: Provides quantitative predictions for multiple ADMET endpoints, supporting rational lead optimization.  

Parameters    

    1. Absorption   
    2. Distribution   
    3. Metabolism  
    4. Excretion
    5. Ttoxicity 33  

Key Parameters:    

Absorption (LogP, Caco-2)   

LogP (Partition Coefficient): Indicates lipophilicity; moderate LogP (typically 1–5) favors membrane permeability and oral absorption.  

Caco-2 Permeability: Measures transport across human intestinal epithelial cells; predicts intestinal absorption.  

Distribution  

Blood-Brain Barrier (BBB) Permeability: Determines if a compound can cross into the central nervous system (CNS)

Metabolism    

Cytochrome P450 (CYP450) Interactions: Drugs can be substrates, inhibitors, or inducers of  CYP450 enzymes.

Excretion  

Clearance Rate: Volume of plasma cleared of drug per unit time; indicates efficiency of excretion.

Toxicity

LD50 (Lethal Dose 50%): Amount of drug that kills 50% of a test population; indicates acute toxicity.34

Integration of ADMET with Drug Design      

Filtering large libraries    

Role of ADMET in Filtering Libraries   

A. Absorption Screening  

Parameters: LogP, solubility, Caco-2 permeability   

B. Distribution Screening  

Parameters: Blood-brain barrier (BBB)    permeability, plasma protein binding  

C. Metabolism Screening

Parameters: Cytochrome P450 (CYP450) substrate/inhibitor profile  

D. Excretion Screening  

Parameters: Predicted clearance, renal/biliary excretion  

E. Toxicity Screening 

Parameters: LD50, hepatotoxicity, hERG inhibition, AMES toxicity 32 

CASE STUDIES WHERE ADMET SAVED TIME/COST   

1. Pfizer – Early hERG and CYP Screening   

Problem: Many early drug candidates were failing in late-stage clinical trials due to cardiotoxicity (hERG channel inhibition) or metabolic issues (CYP450 interactions).  

Solution: Pfizer integrated early in vitro and in silico ADMET screening for hERG inhibition and CYP metabolism during lead optimization.  

Outcome: Toxic compounds were eliminated before expensive animal studies. Reduced late-stage attrition rates. 35

2. GlaxoSmithKline (GSK) – Solubility and Permeability Filters   

Problem: Poor oral bioavailability due to low solubility and poor intestinal absorption led to multiple failures in early preclinical testing.  

Solution: GSK implemented SwissADME-like in silico filters for solubility, LogP, and Caco-2 permeability during virtual library screening.  

Outcome: Large compound libraries were filtered before synthesis or biological testing. 36

3. Roche – Hepatotoxicity and Toxicity Prediction   

Problem: Several lead compounds failed in preclinical stages due to liver toxicity.  

Solution: Roche integrated in silico hepatotoxicity and LD50 prediction models to prioritize safer compounds during lead optimization.  

Outcome: Compounds with high predicted hepatotoxicity were removed early.  

4. Boehringer Ingelheim – ADMET Modeling in Virtual Screening   

Problem: Screening millions of compounds experimentally was expensive and slow.  

Solution: Boehringer Ingelheim used computational ADMET models (pkCSM, admetSAR equivalents) to filter large libraries.  

Outcome: Only compounds with favorable ADMET profiles were synthesized and tested .37

COMBINATORIAL CHEMISTRY AND HIGH-THROUGHPUT SCREENING (HTS)   

Basics of Combinatorial Chemistry   

Definition- Combinatorial chemistry is a synthetic approach in which large libraries of structurally related compounds are generated rapidly and simultaneously, instead of synthesizing molecules one at a time. This method revolutionized drug discovery by allowing the parallel synthesis and screening of thousands to millions of compounds for biological activity.  

Key Concepts    

Library Design- A “library” is a collection of compounds generated by systematically combining sets of building blocks.  

Parallel vs. Split-and-Mix Synthesis- Parallel synthesis: Compounds are prepared individually in separate reaction vessels (more control, but fewer compounds).  

Solid-Phase Synthesis- Introduced by Merrifield (1963) for peptide synthesis. 38

Principle of generating chemical diversity rapidly   

The central principle of combinatorial chemistry is systematic and parallel synthesis of a large number of structurally related molecules from a small set of starting materials (“building blocks”). This enables rapid exploration of chemical diversity, which is crucial in drug discovery.  

Advantages of Rapid Diversity Generation  

Speed: Thousands of molecules can be made in weeks instead of years.  

Efficiency: Multiple hypotheses about structure–activity relationships can be tested simultaneously.   Exploration of chemical space: Increases probability of identifying novel lead compounds.  

Types

Solid-phase synthesis  

Principle -  

Molecules are assembled while attached to an insoluble support (e.g., resin beads or polymer).  

Reactions occur on the solid phase, while excess reagents and by-products remain in the solution and can be washed away easily.  

Solution-Phase Combinatorial Synthesis -  

Principle:  

Compounds are synthesized in the liquid (solution) phase without a solid support.  

Often uses parallel synthesis in microtiter plates, vials, or automated reactors.39

High-Throughput Screening (HTS)   

Definition:  

High-Throughput Screening (HTS) is an automated experimental approach that allows rapid testing of thousands to millions of compounds for biological activity against a specific target (protein, receptor, enzyme, or cell-based system). It is tightly linked to combinatorial chemistry because it helps identify active compounds (hits) from large libraries.  

Workflow of HTS   

a) Assay Development

Goal: Create a robust, reproducible, miniaturized assay that reflects the biological target of interest.  

Types:  

a. Biochemical assays (enzyme inhibition, receptor binding).  

b. Cell-based assays (cell viability, reporter gene expression, signaling pathway activation).

b) Robotic Screening   

Automation:  

Robotic systems handle liquid dispensing, plate handling, mixing, and detection.  

Allows testing of up to hundreds of thousands of compounds per day.  

Miniaturization:  

Small volumes (microliter/nanoliter scale) reduce reagent use and cost. 

c) Hit Identification   

Primary Screening:  

Identifies compounds with measurable activity (“hits”).  

Typically generates false positives and false negatives.  

Confirmatory Screening (secondary assays):  

Re-testing of hits to validate activity.  

Counter screens against off-targets to ensure selectivity.40  

Integration of HTS with Computational Methods  

High-Throughput Screening (HTS) is powerful but expensive, resource-intensive, and generates massive data. To overcome this, computational methods—especially virtual screening (VS)—are used before HTS to reduce the chemical space and enrich the library with likely actives.  

Virtual Screening (VS) before HTS  

Virtual Screening: Computer-based filtering of large chemical libraries (millions of compounds) to predict which molecules are most likely to interact with the biological target.  

Two main approaches:  

    1. Ligand-based virtual screening (LBVS)  
    2. Structure-based virtual screening (SBVS) 41  

CHALLENGES AND ADVANCEMENTS   

Challenges –   

False positives in HTS   

Compounds appear active in assays but are not true actives.  

Causes include:  

Assay interference: Compounds may absorb/emit light (fluorescence quenchers, colored molecules) and interfere with readouts.  

Aggregation: Some molecules form colloidal aggregates that nonspecifically inhibit proteins.  

Reactive compounds: Electrophiles or redox-active molecules may covalently modify proteins (PAINS – pan-assay interference compounds).  

False Negatives in HTS  

Compounds are active but missed in the screen.   

Causes include:  

Low solubility: Compound precipitates and doesn’t reach effective concentration.  

Instability: Compound degrades during assay conditions.  

Assay sensitivity issues: Signal window too narrow → real actives go undetected. 42 

Advancements to  False Positives/Negatives  

Assay Improvements  

Use of orthogonal assays (different readout methods, e.g., fluorescence + luminescence + label-free) to confirm activity.  

Improved assay designs with better signal-to-noise ratio and statistical validation (Z-factor ≥ 0.5) Data 

Filtering  

In silico filtering of PAINS (pan-assay interference structures) before screening.  

Confirmatory Screens  

Counter-screens against unrelated targets to test specificity.  

Technological Advancements  

High-content screening (HCS): Imaging-based assays that provide multiparametric readouts, reducing false positives.43 

CASE STUDIES AND SUCCESS STORIES   

Natural Product-Derived Drugs with Computational Support     

Example: Artemisinin derivatives through SBDD

Natural Product: Artemisinin, a sesquiterpene lactone from Artemisia annual.  

Challenge: Original artemisinin had poor solubility and bioavailability, limiting its clinical effectiveness.  

Computational Approach (SBDD):  

  1. Target Identification: Plasmodium falciparum proteins (e.g., PfATP6, heme targets) identified as critical for parasite survival. 
  2. Molecular Docking: Artemisinin and derivatives docked into target binding sites to predict binding affinity and pose. 
  3. Lead Optimization: Semi-synthetic derivatives designed (artemether, artesunate, dihydroartemisinin).  

Outcome:  

Artemisinin derivatives with improved pharmacokinetics and potency.  

Led to WHO-recommended Artemisinin-based Combination Therapies (ACTs), saving millions of lives globally.44  

Curcumin analogs optimized by QSAR   

Natural Product: Curcumin, a polyphenolic compound from Curcuma longa (turmeric).  

Therapeutic Potential: Anti-inflammatory, anticancer, antioxidant, neuroprotective.

QSAR Approach in Curcumin Analog Optimization Stepwise Workflow:  

1. Data Collection: Curcumin and analogs tested in vitro for a biological target (e.g., NF-κB inhibition, anti-cancer activity).   

Activity expressed as IC?? values. 

2. Descriptor Calculation: Physicochemical descriptors: LogP, molecular weight, hydrogen bond donors/acceptors, polar surface area. 

Electronic descriptors: HOMO-LUMO gap, dipole moment.  

3. Model Development:  

2D-QSAR: Correlates structural features with activity.  

3D-QSAR (CoMFA/CoMSIA): Maps steric and electrostatic fields to predict activity hotspots.  

4. Prediction & Design

QSAR models predict which modifications increase potency or stability. 

5. Synthesis & Testing:  

Top predicted analogs synthesized and experimentally validated.  

Iterative QSAR refinement improves predictive accuracy.45

Drugs Discovered Using Integrated Platforms   

HIV protease inhibitors using docking + QSAR + ADMET  

Target: HIV-1 protease, an essential enzyme in viral polyprotein processing.  

Clinical Goal: Inhibit protease to prevent maturation of infectious virus particles.  

Challenges:  

High mutation rate → drug resistance.  

Need for selective inhibitors with good pharmacokinetics and low toxicity.  

Solution: Use an integrated computational approach combining:

  1. Molecular docking → predict binding modes and affinities.  
  2. QSAR modeling → correlate chemical features with activity.  
  3. ADMET prediction → ensure drug-likeness, bioavailability, and safety. 46

Integrated Platform Workflow   

Step 1: Library Preparation  

Large chemical libraries or combinatorial derivatives of known HIV protease inhibitors.  

Pre-filtering using Lipinski’s Rule of 5 and PAINS filters to ensure drug-likeness.  

Step 2: Molecular Docking  

Dock compounds into HIV-1 protease crystal structures (e.g., PDB ID: 1HVR).  

Evaluate binding energies, hydrogen bonding, hydrophobic interactions, and key contacts with catalytic residues (Asp25/Asp25’).  

Shortlist compounds with high predicted affinity for further analysis.  

Step 3: QSAR Modeling  

Build 2D- and 3D-QSAR models correlating descriptors (hydrophobicity, polar surface area, H-bond donors/acceptors, steric parameters) with observed or predicted IC??.  

Predict modifications that improve potency.    

Validate models using cross-validation or external test sets.  

Step 4: ADMET Profiling  

In silico prediction of:  

Absorption: Caco-2 permeability, human intestinal absorption.  

Distribution: Plasma protein binding, blood–brain barrier penetration.  

Metabolism: CYP450 inhibition/induction.  

Excretion & Toxicity: Hepatotoxicity, cardiotoxicity, mutagenicity.  

Step 5: Hit-to-Lead Optimization  

Integrate docking insights (binding interactions) with QSAR-predicted chemical modifications.  

Iterate the cycle to generate lead compounds with high potency, selectivity, and favorable ADMET profiles.  

Key Takeaways  

Docking → identifies potential binders and rationalizes structural features critical for activity.  

QSAR → guides systematic chemical modifications to improve potency and selectivity.  

ADMET → ensures drug-likeness and reduces experimental failure due to poor pharmacokinetics or toxicity.47

CONCLUSION  

Drug design has evolved from traditional trial-and-error methods to modern, rational approaches that combine chemistry, biology, and computation. Computational drug design (CADD), including QSAR, structure-based drug design (SBDD), molecular docking, and ADMET profiling, allows scientists to predict the activity, binding affinity, and pharmacokinetic properties of compounds before synthesis. High-throughput screening (HTS) complements these methods by experimentally testing large libraries of compounds quickly, while virtual screening reduces chemical space and improves efficiency. Integrating these techniques accelerates the hit-to-lead optimization process, as seen in case studies like HIV protease inhibitors, artemisinin derivatives, paclitaxel, camptothecin, and curcumin analogs. QSAR helps identify structural features crucial for activity, docking provides insights into target interactions, and ADMET modeling ensures safety and bioavailability. Natural products, combinatorial libraries, and semi-synthetic analogs benefit greatly from these integrative strategies. This approach reduces cost, time, and experimental failures while increasing the success rate of lead identification. Many FDA-approved drugs, including Imatinib, Darunavir, Atorvastatin, and Sofosbuvir, demonstrate that combining computational methods with experimental validation leads to commercially successful, safe, and effective medicines.  

REFERENCES

  1. Singh N, Vayer P, Tanwar S, Poyet JL, Tsaioun K, Villoutreix BO. Drug discovery and development: introduction to the general public and patient groups. Front Drug Discovery. 2023 May 24;3:1201419. Doi: 10.3389/fddsv.2023.1201419.  
  2. Mak KK, Pichika MR. Artificial intelligence in drug development: present status and future prospects. Drug Discovery Today. 2019 Mar;24(3):773-780. Doi: 10.1016/j.drudis.2018.11.014.  
  3. Schneider G. Automating drug discovery. Nature Review Drug Discovery. 2018 Feb;17(2):97113. Doi: 10.1038/nrd.2017.232.  
  4. Patrick GL. An introduction to medicinal chemistry. 6th ed. Oxford: Oxford University Press; 2017.  
  5. Bisht D, Arya RKK, Pal GR, Singh RP. A review on recent rational approaches to drug design, development and its discovery. International Journal Pharm Biological Sci. 2020;10(4):96108. Doi: 10.22376/ijpbs/lpr.2020.10.4.P96-108.  
  6. Atanasov AG, Waltenberger B, Pferschy-Wenzig EM, Linder T, Wawrosch C.etal.Discovery and resupply of pharmacologically active plant-derived natural products: a review. Biotechnology Adv. 2015 Dec;33(8):1582-1614. Doi:   10.1016/j.biotechadv.2015.08.001.  
  7. Newman DJ, Cragg GM. Natural products as sources of new drugs over the nearly four decades from 1981 to 2019. J Nat Prod. 2020;83(3):770-803. Doi:   10.1021/acs.jnatprod.9b01285.  
  8. Brunton LL, Hilal-Dandan R, Knollmann BC, editors. Goodman & Gilman’s the pharmacological basis of therapeutics. 13th ed. New York: McGraw-Hill; 2018.  
  9. Vamathevan J, Clark D, Czodrowski P, Dunham I, Ferran E, Lee G, et al. Applications of machine learning in drug discovery and development. Nat Rev Drug Discov. 2019 Jun;18(6):463-477. Doi: 10.1038/s41573-019-0024-5.  
  10. Mouchlis VD, Afantitis A, Serra A, Fratello M, Papadiamantis AG, Aidinis V, et al. Advances in de novo drug design: from conventional to machine learning methods. Int J Mol Sci. 2021 Feb 7;22(4):1676. Doi: 10.3390/ijms22041676.  
  11. Singh N, Vayer P, Tanwar S, Poyet JL, Tsaioun K, Villoutreix BO. Drug discovery and development: introduction to the general public and patient groups. Front Drug Discov. 2023 May 24;3:1201419. Doi: 10.3389/fddsv.2023.1201419.  
  12. Gupta YD, Bhandary S. Artificial intelligence for understanding mechanisms of antimicrobial resistance and antimicrobial discovery: a new age model for translational research. First published 19 June 2024. doi: 10.1002/9781394234196.ch5.  
  13. Koehn FE, Carter GT. The evolving role of natural products in drug discovery. Nat Rev Drug Discov. 2019 Mar;4(3):206-220. Doi: 10.1038/nrd1657.  
  14. Cushnie TP, Cushnie B, Lamb AJ. Alkaloids: an overview of their antibacterial, antibioticenhancing and antivirulence activities. Int J Antimicrob Agents. 2014 Nov;44(5):377-386. Doi: 10.1016/j.ijantimicag.2014.06.001.  
  15. Li, S., et al. (2020). Terpenoids: Natural products with pharmacological relevance. Molecules, 25, 5776.  
  16. Rang HP, Dale MM, Ritter JM, Flower RJ. Rang and Dale’s Pharmacology. 9th Edition. Elsevier; 2019.  
  17. Hauser, A.S., et al. (2018). Pharmacology of receptor-targeted drugs: From discovery to clinical application. Nature Reviews Drug Discovery, 17, 693–710.  
  18. Patrick GL. An Introduction to Medicinal Chemistry. Oxford University Press, 2017.  
  19. Rodrigues T. Harnessing chemical complexity in natural product-based drug discovery. Nat Chem Biol. 2020;16(10):1131–8. Doi:10.1038/s41589-020-00651-6  
  20. Gajewicz A, Cronin MTD, Rasulev B, Leszczynski J, Puzyn T. QSAR modelling: advances and challenges in drug discovery. Int J Mol Sci. 2020;21(17):5939. Doi:10.3390/ijms21175939  
  21. Cherkasov A, Muratov EN, Fourches D, et al. QSAR modeling: where have you been? Where are you going to? J Med Chem. 2020;63(16):8695–722. Doi:10.1021/acs.jmedchem.9b02120  
  22. Khondkaryan L, Tevosyan A, Navasardyan H, et al. Datasets construction and development of QSAR models for predicting micronucleus in vitro and in vivo assay outcomes. Toxics. 2023;11(9):785. Doi:10.3390/toxics11090785  
  23. Bhardwaj V, Rajendran V, Kumar V, et al. Computational approaches in anti-tubercular drug design: QSAR, docking, and molecular dynamics. Molecules. 2020;25(22):5347. Doi:10.3390/molecules25225347  
  24. Koirala M, Yan L, Mohamed Z, DiPaola M. AI-Integrated QSAR Modeling for   Enhanced Drug Discovery: From Classical Approaches to Deep Learning and Structural Insight. Int J Mol Sci. 2025;Â26(19):9384. Doi:10.3390/ijms26199384   
  25. Ghahremanpour MM, Barros EP, Oliveira AS, Chodera JD, Levy RM. In silico ADMET prediction: recent advances and applications. Molecules. 2021;26(10):3001. Doi:10.3390/molecules26103001  
  26. Singh N, Chaput L, Villoutreix BO, Kaur D. Computational approaches in structurebased drug design: an overview. Molecules. 2021;26(12):3675. Doi:10.3390/molecules26123675  
  27. Gillet VJ, Khatib W, Willett P, Fleming PJ, Green DV, et al. Structure- and ligand-based approaches in modern drug discovery: a review. Molecules. 2020;25(21):5063. Doi:10.3390/molecules25215063  
  28. Ferreira LG, Dos Santos RN, Oliva G, Andricopulo AD, De Azevedo WF, et al. Molecular docking and structure-based drug design strategies. Molecules. 2021;26(13):3940. Doi:10.3390/molecules26133940  
  29. Luo L, Zhong A, Wang Q, Zheng T, Liu X, et al. Structure-based pharmacophore modeling, virtual screening, molecular docking, ADMET, and molecular dynamics (MD) simulation of potential inhibitors of PD-L1 from the library of marine natural products. Mar Drugs. 2022;20(1):29. Doi:10.3390/md20010029  
  30. Butt SS, Badshah Y, Shabbir M, Rafiq M, Muhammad A, et al. Molecular docking using Chimera and AutoDock Vina. JMIR Bioinform. 2020;1(1):e14232. doi:10.2196/14232  
  31. Daoud NEH, Owis AI, El-Hawary SS, Ali Z, Efferth T, et al. ADMET profiling in drug discovery and development: perspectives of in silico, in vitro and integrated approaches. Curr Drug Metab. 2021;22(7):503–22. doi:10.2174/1389200222666210707123735  
  32. Ekins S, Clark AM, Williams AJ, Haddad B, Xu JJ, et al. In silico ADMET prediction: recent advances, current challenges and future trends. Expert Opin Drug Metab Toxicol. 2019;15(7):525–36. doi:10.1080/17425255.2019.1626957  
  33. Daina A, Michielin O, Zoete V, et al. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7:42717. doi:10.1038/srep42717  
  34. Wang J, Urban L, Ekins S, Clark AM, Xu JJ, et al. In silico ADMET prediction: recent advances, current challenges and future trends. Expert Opin Drug Metab Toxicol. 2019;15(7):525–36. Doi:10.1080/17425255.2019.1626957  
  35. Ekins S, Clark AM, Williams AJ, Xu JJ, Urban L, et al. In silico ADME/Tox comes of age: twenty years later. Pharm Res. 2023;40(1):1–15. doi:10.1007/s11095-022-03361-z  
  36. Di L, Kerns EH, Carter GT, et al. Drug-like property concepts in drug discovery: rapid in silico screening of ADMET. Curr Opin Chem Biol. 2019;50:57–64. doi:10.1016/j.cbpa.2018.11.017  
  37. Pisani L, Barletta M, Caporuscio F, et al. Computational ADME prediction for early drug discovery: SwissADME as a versatile tool. Molecules. 2020;25(17):3981. doi:10.3390/molecules25173981  
  38. Liu R, et al. Combinatorial chemistry in drug discovery. In: Encyclopedia of Physical Organic Chemistry. Hoboken (NJ): Wiley; 2017. Doi:10.1002/9781118468586.epo093  
  39. Suay-García B, Bueso-Bordils JI, Falcó A, Antón-Fos GM, Alemán-López PA, et al. Virtual combinatorial chemistry and pharmacological screening: a short guide to drug design. Int J Mol Sci. 2022;23(3):1620. Doi:10.3390/ijms23031620  
  40. Olubiyi OO, Olagunju M, Keutmann M, Loschwitz J, Strodel B, et al. High throughput virtual screening to discover inhibitors of SARS-CoV-2 main protease. Front Pharmacol. 2020;11:739698. doi:10.3389/fphar.2020.584823  
  41. Ferreira FJN, Ramos M, Santos R, Pereira D, Oliveira JL, et al. AI-driven drug discovery: a comprehensive review. Front Pharmacol. 2025;13:12177741. doi:10.3389/fphar.2025.12177741  
  42. Shi S, Yang Y, Chen H, Zhang Y, Zhou Y, et al. ChemFH: an integrated tool for screening frequent false positives in high-throughput screening assays. Front Pharmacol. 2024;15:11223804. doi:10.3389/fphar.2024.11223804  
  43. Alam S, Gupta M, Kaur J, Singh N, Sharma R, et al. Pharmacophore and QSAR guided design, synthesis, pharmacokinetics, and in vitro evaluation of curcumin analogs for anticancer activity. Front Pharmacol. 2024;15:12177741. doi:10.3389/fphar.2025.12177741  
  44. Macarron R, Banks MN, Bojanic D, Burns DJ, Cirovic DA, et al. Impact of highthroughput screening in biomedical research. Nat Rev Drug Discovery. 2011;10(3):188–  95. Doi:10.1038/nrd3368  
  45. Walters WP, Namchuk M, et al. Designing screens: how to make your hits a hit. Nat Rev  Drug Discov. 2003;2(4):259–66. doi:10.1038/nrd1060  
  46. Tu Y, et al. Artemisinin—a gift from traditional Chinese medicine to the world (Nobel Lecture). Angew Chem Int Ed Engl. 2016;55(35):10210–26. doi:10.1002/anie.201601967.

Reference

  1. Singh N, Vayer P, Tanwar S, Poyet JL, Tsaioun K, Villoutreix BO. Drug discovery and development: introduction to the general public and patient groups. Front Drug Discovery. 2023 May 24;3:1201419. Doi: 10.3389/fddsv.2023.1201419.  
  2. Mak KK, Pichika MR. Artificial intelligence in drug development: present status and future prospects. Drug Discovery Today. 2019 Mar;24(3):773-780. Doi: 10.1016/j.drudis.2018.11.014.  
  3. Schneider G. Automating drug discovery. Nature Review Drug Discovery. 2018 Feb;17(2):97113. Doi: 10.1038/nrd.2017.232.  
  4. Patrick GL. An introduction to medicinal chemistry. 6th ed. Oxford: Oxford University Press; 2017.  
  5. Bisht D, Arya RKK, Pal GR, Singh RP. A review on recent rational approaches to drug design, development and its discovery. International Journal Pharm Biological Sci. 2020;10(4):96108. Doi: 10.22376/ijpbs/lpr.2020.10.4.P96-108.  
  6. Atanasov AG, Waltenberger B, Pferschy-Wenzig EM, Linder T, Wawrosch C.etal.Discovery and resupply of pharmacologically active plant-derived natural products: a review. Biotechnology Adv. 2015 Dec;33(8):1582-1614. Doi:   10.1016/j.biotechadv.2015.08.001.  
  7. Newman DJ, Cragg GM. Natural products as sources of new drugs over the nearly four decades from 1981 to 2019. J Nat Prod. 2020;83(3):770-803. Doi:   10.1021/acs.jnatprod.9b01285.  
  8. Brunton LL, Hilal-Dandan R, Knollmann BC, editors. Goodman & Gilman’s the pharmacological basis of therapeutics. 13th ed. New York: McGraw-Hill; 2018.  
  9. Vamathevan J, Clark D, Czodrowski P, Dunham I, Ferran E, Lee G, et al. Applications of machine learning in drug discovery and development. Nat Rev Drug Discov. 2019 Jun;18(6):463-477. Doi: 10.1038/s41573-019-0024-5.  
  10. Mouchlis VD, Afantitis A, Serra A, Fratello M, Papadiamantis AG, Aidinis V, et al. Advances in de novo drug design: from conventional to machine learning methods. Int J Mol Sci. 2021 Feb 7;22(4):1676. Doi: 10.3390/ijms22041676.  
  11. Singh N, Vayer P, Tanwar S, Poyet JL, Tsaioun K, Villoutreix BO. Drug discovery and development: introduction to the general public and patient groups. Front Drug Discov. 2023 May 24;3:1201419. Doi: 10.3389/fddsv.2023.1201419.  
  12. Gupta YD, Bhandary S. Artificial intelligence for understanding mechanisms of antimicrobial resistance and antimicrobial discovery: a new age model for translational research. First published 19 June 2024. doi: 10.1002/9781394234196.ch5.  
  13. Koehn FE, Carter GT. The evolving role of natural products in drug discovery. Nat Rev Drug Discov. 2019 Mar;4(3):206-220. Doi: 10.1038/nrd1657.  
  14. Cushnie TP, Cushnie B, Lamb AJ. Alkaloids: an overview of their antibacterial, antibioticenhancing and antivirulence activities. Int J Antimicrob Agents. 2014 Nov;44(5):377-386. Doi: 10.1016/j.ijantimicag.2014.06.001.  
  15. Li, S., et al. (2020). Terpenoids: Natural products with pharmacological relevance. Molecules, 25, 5776.  
  16. Rang HP, Dale MM, Ritter JM, Flower RJ. Rang and Dale’s Pharmacology. 9th Edition. Elsevier; 2019.  
  17. Hauser, A.S., et al. (2018). Pharmacology of receptor-targeted drugs: From discovery to clinical application. Nature Reviews Drug Discovery, 17, 693–710.  
  18. Patrick GL. An Introduction to Medicinal Chemistry. Oxford University Press, 2017.  
  19. Rodrigues T. Harnessing chemical complexity in natural product-based drug discovery. Nat Chem Biol. 2020;16(10):1131–8. Doi:10.1038/s41589-020-00651-6  
  20. Gajewicz A, Cronin MTD, Rasulev B, Leszczynski J, Puzyn T. QSAR modelling: advances and challenges in drug discovery. Int J Mol Sci. 2020;21(17):5939. Doi:10.3390/ijms21175939  
  21. Cherkasov A, Muratov EN, Fourches D, et al. QSAR modeling: where have you been? Where are you going to? J Med Chem. 2020;63(16):8695–722. Doi:10.1021/acs.jmedchem.9b02120  
  22. Khondkaryan L, Tevosyan A, Navasardyan H, et al. Datasets construction and development of QSAR models for predicting micronucleus in vitro and in vivo assay outcomes. Toxics. 2023;11(9):785. Doi:10.3390/toxics11090785  
  23. Bhardwaj V, Rajendran V, Kumar V, et al. Computational approaches in anti-tubercular drug design: QSAR, docking, and molecular dynamics. Molecules. 2020;25(22):5347. Doi:10.3390/molecules25225347  
  24. Koirala M, Yan L, Mohamed Z, DiPaola M. AI-Integrated QSAR Modeling for   Enhanced Drug Discovery: From Classical Approaches to Deep Learning and Structural Insight. Int J Mol Sci. 2025;Â26(19):9384. Doi:10.3390/ijms26199384   
  25. Ghahremanpour MM, Barros EP, Oliveira AS, Chodera JD, Levy RM. In silico ADMET prediction: recent advances and applications. Molecules. 2021;26(10):3001. Doi:10.3390/molecules26103001  
  26. Singh N, Chaput L, Villoutreix BO, Kaur D. Computational approaches in structurebased drug design: an overview. Molecules. 2021;26(12):3675. Doi:10.3390/molecules26123675  
  27. Gillet VJ, Khatib W, Willett P, Fleming PJ, Green DV, et al. Structure- and ligand-based approaches in modern drug discovery: a review. Molecules. 2020;25(21):5063. Doi:10.3390/molecules25215063  
  28. Ferreira LG, Dos Santos RN, Oliva G, Andricopulo AD, De Azevedo WF, et al. Molecular docking and structure-based drug design strategies. Molecules. 2021;26(13):3940. Doi:10.3390/molecules26133940  
  29. Luo L, Zhong A, Wang Q, Zheng T, Liu X, et al. Structure-based pharmacophore modeling, virtual screening, molecular docking, ADMET, and molecular dynamics (MD) simulation of potential inhibitors of PD-L1 from the library of marine natural products. Mar Drugs. 2022;20(1):29. Doi:10.3390/md20010029  
  30. Butt SS, Badshah Y, Shabbir M, Rafiq M, Muhammad A, et al. Molecular docking using Chimera and AutoDock Vina. JMIR Bioinform. 2020;1(1):e14232. doi:10.2196/14232  
  31. Daoud NEH, Owis AI, El-Hawary SS, Ali Z, Efferth T, et al. ADMET profiling in drug discovery and development: perspectives of in silico, in vitro and integrated approaches. Curr Drug Metab. 2021;22(7):503–22. doi:10.2174/1389200222666210707123735  
  32. Ekins S, Clark AM, Williams AJ, Haddad B, Xu JJ, et al. In silico ADMET prediction: recent advances, current challenges and future trends. Expert Opin Drug Metab Toxicol. 2019;15(7):525–36. doi:10.1080/17425255.2019.1626957  
  33. Daina A, Michielin O, Zoete V, et al. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7:42717. doi:10.1038/srep42717  
  34. Wang J, Urban L, Ekins S, Clark AM, Xu JJ, et al. In silico ADMET prediction: recent advances, current challenges and future trends. Expert Opin Drug Metab Toxicol. 2019;15(7):525–36. Doi:10.1080/17425255.2019.1626957  
  35. Ekins S, Clark AM, Williams AJ, Xu JJ, Urban L, et al. In silico ADME/Tox comes of age: twenty years later. Pharm Res. 2023;40(1):1–15. doi:10.1007/s11095-022-03361-z  
  36. Di L, Kerns EH, Carter GT, et al. Drug-like property concepts in drug discovery: rapid in silico screening of ADMET. Curr Opin Chem Biol. 2019;50:57–64. doi:10.1016/j.cbpa.2018.11.017  
  37. Pisani L, Barletta M, Caporuscio F, et al. Computational ADME prediction for early drug discovery: SwissADME as a versatile tool. Molecules. 2020;25(17):3981. doi:10.3390/molecules25173981  
  38. Liu R, et al. Combinatorial chemistry in drug discovery. In: Encyclopedia of Physical Organic Chemistry. Hoboken (NJ): Wiley; 2017. Doi:10.1002/9781118468586.epo093  
  39. Suay-García B, Bueso-Bordils JI, Falcó A, Antón-Fos GM, Alemán-López PA, et al. Virtual combinatorial chemistry and pharmacological screening: a short guide to drug design. Int J Mol Sci. 2022;23(3):1620. Doi:10.3390/ijms23031620  
  40. Olubiyi OO, Olagunju M, Keutmann M, Loschwitz J, Strodel B, et al. High throughput virtual screening to discover inhibitors of SARS-CoV-2 main protease. Front Pharmacol. 2020;11:739698. doi:10.3389/fphar.2020.584823  
  41. Ferreira FJN, Ramos M, Santos R, Pereira D, Oliveira JL, et al. AI-driven drug discovery: a comprehensive review. Front Pharmacol. 2025;13:12177741. doi:10.3389/fphar.2025.12177741  
  42. Shi S, Yang Y, Chen H, Zhang Y, Zhou Y, et al. ChemFH: an integrated tool for screening frequent false positives in high-throughput screening assays. Front Pharmacol. 2024;15:11223804. doi:10.3389/fphar.2024.11223804  
  43. Alam S, Gupta M, Kaur J, Singh N, Sharma R, et al. Pharmacophore and QSAR guided design, synthesis, pharmacokinetics, and in vitro evaluation of curcumin analogs for anticancer activity. Front Pharmacol. 2024;15:12177741. doi:10.3389/fphar.2025.12177741  
  44. Macarron R, Banks MN, Bojanic D, Burns DJ, Cirovic DA, et al. Impact of highthroughput screening in biomedical research. Nat Rev Drug Discovery. 2011;10(3):188–  95. Doi:10.1038/nrd3368  
  45. Walters WP, Namchuk M, et al. Designing screens: how to make your hits a hit. Nat Rev  Drug Discov. 2003;2(4):259–66. doi:10.1038/nrd1060  
  46. Tu Y, et al. Artemisinin—a gift from traditional Chinese medicine to the world (Nobel Lecture). Angew Chem Int Ed Engl. 2016;55(35):10210–26. doi:10.1002/anie.201601967.

Photo
Mayuri Salve
Corresponding author

Siddhi’s Institute of Pharmacy, Nandgaon, Murbad, Thane- 421401, Maharashtra, India.

Photo
Rutuja Pawar
Co-author

Siddhi’s Institute of Pharmacy, Nandgaon, Murbad, Thane- 421401, Maharashtra, India.

Photo
Shweta Bundhe
Co-author

Siddhi’s Institute of Pharmacy, Nandgaon, Murbad, Thane- 421401, Maharashtra, India.

Photo
Priyanka Suroshe
Co-author

Siddhi’s Institute of Pharmacy, Nandgaon, Murbad, Thane- 421401, Maharashtra, India.

Photo
Avinash Gunjal
Co-author

Siddhi’s Institute of Pharmacy, Nandgaon, Murbad, Thane- 421401, Maharashtra, India.

Mayuri Salve, Rutuja Pawar, Shweta Bundhe, Priyanka Suroshe, Avinash Gunjal, A Comprehensive Review on Drug Design: Principles, Approaches, and Recent Advances, Int. J. of Pharm. Sci., 2026, Vol 4, Issue 1, 339-358. https://doi.org/10.5281/zenodo.18154189

More related articles
Recent Trends on Transdermal Drug Delivery System...
Chetan Mali, Nitin Mali, Srushti Mali, Jayshree Naik, R. R. Patil...
Quality By Design (QbD) in Pharmaceutical Formulat...
Prajwal Gosavi, Akanksha Nirmal, Rutuja Ahire, ...
A Comprehensive Review of Bidens Pilosa: Botanical...
Deepika J. K., T. Balasubramanian, Suresha B. S., ...
Carboxymethyl Starch as a Pharmaceutical Excipient: A Comprehensive Review of it...
Harita Desai, Atharva Zore, Palak Khania, Virti Vora, ...
Review on Green Pharmacy Practice ...
N. B. Patil Thorat, A. S. Kulkarni, S. P. Patil, T. G. Dhobale, A. R. Satale, ...
Related Articles
Design, Synthesis, and Evaluation of Novel Pharmaceutical Co-crystals of an Anti...
Sachin Gholve , Shweta Sarkalwad, Pradnya Kshirsagar, Keshavrao Kulkarni, ...
Overall Review on Development and Validation of RP-HPLC Method for Relugolix in ...
Jyoti Darade , Komal Bankar , Maya Dudhare , Mamata Chavan, Gaytri Fuke, Priya Jadhav, Vaishnavi Pak...
Review on Green Analytical Chemistry...
N. B. Patil Thorat, R. P. Raula, A. S. Kulkarni, V. A. Neje, M. M. Pawar, ...
Comprehensive Review on Analytical Methods of Atorvastatin...
Pradnya Kumbhar, Dr. Sandeep Waghulde, ...
Recent Trends on Transdermal Drug Delivery System...
Chetan Mali, Nitin Mali, Srushti Mali, Jayshree Naik, R. R. Patil, ...
More related articles
Recent Trends on Transdermal Drug Delivery System...
Chetan Mali, Nitin Mali, Srushti Mali, Jayshree Naik, R. R. Patil, ...
Recent Trends on Transdermal Drug Delivery System...
Chetan Mali, Nitin Mali, Srushti Mali, Jayshree Naik, R. R. Patil, ...