Call for Abstract

16th World Congress on Pharmacoclogy & Drug Discovery, will be organized around the theme “Next-Generation Therapeutics: Innovations in Drug Discovery”

PHARMACOLOGY CONGRESS 2026 is comprised of keynote and speakers sessions on latest cutting edge research designed to offer comprehensive global discussions that address current issues in PHARMACOLOGY CONGRESS 2026

Submit your abstract to any of the mentioned tracks.

Register now for the conference by choosing an appropriate package suitable to you.

Advancements in drug discovery and development continue to reshape therapeutic approaches across various diseases. This area emphasizes the entire process of identifying novel bioactive compounds, optimizing their chemical and pharmacological properties, and progressing them through preclinical and clinical development. Integrative strategies, including rational drug design, combinatorial chemistry, and computer-aided molecular modeling, enhance efficiency and success rates. Collaborative approaches between academia, biotech, and pharmaceutical industries facilitate the translation of innovative research into clinically viable therapies. Additionally, contemporary strategies address the challenges of drug resistance, off-target effects, and regulatory compliance, ensuring safe and effective therapeutics reach patients. By leveraging multidisciplinary methodologies, researchers are now capable of streamlining target validation, lead optimization, and formulation, ultimately accelerating the path from discovery to market. This evolving landscape underscores the importance of integrating novel technologies, predictive models, and translational research, fostering a robust framework for next-generation therapeutics. The focus on strategic planning, project management, and scientific innovation equips stakeholders with the knowledge to navigate complex challenges in modern drug development, ensuring impactful outcomes in global healthcare.

Pharmacogenomics bridges the gap between genetic variability and therapeutic response, enabling personalized medicine that maximizes efficacy while minimizing adverse effects. By analyzing genetic polymorphisms in drug-metabolizing enzymes, transporters, and receptors, pharmacogenomic approaches guide tailored treatment regimens. Personalized medicine integrates this genetic information with clinical, environmental, and lifestyle factors, providing precision therapies for diverse populations. This field is transforming disease management across oncology, cardiology, neurology, and infectious diseases, offering predictive models for drug response and toxicity. Advances in high-throughput sequencing, bioinformatics, and biomarker discovery have accelerated the identification of patient-specific targets and actionable mutations. Implementation of pharmacogenomic data in clinical practice allows healthcare providers to optimize drug selection, dosing, and monitoring, reducing adverse drug reactions and improving therapeutic outcomes. Beyond clinical applications, pharmacogenomics drives drug discovery by informing the design of compounds with improved efficacy and safety profiles. Ethical, regulatory, and economic considerations are critical to adopting personalized medicine globally, ensuring equitable access to genomically informed healthcare solutions. Through these innovations, pharmacogenomics and personalized medicine are poised to redefine therapeutic strategies and enhance patient-centered care.

High-throughput screening (HTS) has revolutionized the early stages of drug discovery by enabling rapid evaluation of thousands to millions of compounds against biological targets. HTS platforms combine robotics, sensitive detection technologies, and computational analytics to identify promising lead molecules efficiently. Once potential hits are identified, lead optimization strategies refine chemical structures to improve potency, selectivity, pharmacokinetics, and safety profiles. Structure-activity relationship (SAR) studies, medicinal chemistry approaches, and computational modeling guide iterative optimization, ensuring high-quality drug candidates advance to preclinical testing. Integration of phenotypic screening and target-based assays enhances predictive accuracy and identifies compounds with novel mechanisms of action. Advances in miniaturization, microfluidics, and multiplexed assay formats increase throughput and reduce costs, accelerating the drug discovery pipeline. Furthermore, HTS facilitates the repurposing of existing compounds and natural products for new therapeutic indications. The combination of HTS and rational lead optimization provides a robust framework for identifying and developing high-value drug candidates, bridging the gap between initial discovery and clinical development while mitigating risks associated with late-stage failures.

Artificial intelligence (AI) and machine learning (ML) have become integral in modern drug design, enhancing prediction accuracy and accelerating therapeutic development. These computational approaches analyze large datasets from chemical libraries, biological assays, and omics studies to identify promising drug candidates efficiently. AI-driven molecular modeling predicts binding affinities, pharmacokinetics, and potential toxicity profiles, reducing reliance on labor-intensive experimental methods. Machine learning algorithms facilitate target identification, structure-based drug design, and optimization of lead compounds. Integration of AI with high-throughput screening and computational chemistry enables the exploration of vast chemical spaces, uncovering novel scaffolds and mechanisms of action. Additionally, AI tools support drug repurposing and personalized therapeutic strategies by predicting patient-specific responses. The incorporation of neural networks, deep learning, and predictive modeling into pharmacology has improved decision-making, minimized late-stage failures, and reduced development costs. As regulatory and ethical frameworks adapt, AI-driven drug design continues to transform research paradigms, promoting innovation in pharmacology and fostering the development of safe, effective, and targeted therapies.

Nanotechnology-based drug delivery systems offer precision in targeting, controlled release, and enhanced bioavailability of therapeutics. Nanocarriers, including liposomes, polymeric nanoparticles, dendrimers, and metallic nanostructures, facilitate efficient transport of drugs to specific tissues, minimizing systemic toxicity and improving therapeutic indices. These delivery systems enable encapsulation of small molecules, biologics, nucleic acids, and imaging agents for diagnostic and therapeutic applications. Surface functionalization and ligand-targeted strategies enhance cellular uptake and specificity, particularly in oncology, infectious diseases, and neurodegenerative disorders. Controlled-release technologies allow sustained or stimuli-responsive drug delivery, optimizing pharmacokinetics and patient compliance. Advances in nanomedicine have also addressed challenges such as solubility, stability, and immune evasion, providing versatile platforms for complex therapeutics. Preclinical and clinical studies demonstrate improved efficacy, reduced adverse effects, and the potential for combination therapies using nanocarriers. As regulatory guidelines evolve, nanotechnology-based drug delivery continues to expand opportunities for precision medicine, targeted therapy, and innovation in pharmacological interventions, redefining treatment paradigms and improving patient outcomes globally.

Biologics and monoclonal antibodies have transformed therapeutic strategies by offering highly specific treatment options for cancer, autoimmune disorders, and chronic diseases. These protein-based therapies leverage advanced biotechnological processes for high specificity and targeted mechanisms of action. Biosimilars provide cost-effective alternatives, improving access without compromising efficacy or safety. Development of biologics involves rigorous characterization of structure, function, immunogenicity, and pharmacokinetics to ensure consistency and clinical effectiveness. Monoclonal antibodies can be engineered for enhanced stability, reduced immunogenicity, or conjugated with cytotoxic agents for targeted therapy. Regulatory frameworks for biosimilars and biologics ensure robust quality control, safety, and therapeutic equivalence. Continuous innovation in antibody-drug conjugates, bispecific antibodies, and fusion proteins expands the scope of personalized treatment, addressing unmet clinical needs.

Translational pharmacology bridges laboratory discoveries with clinical applications, ensuring that novel therapeutics demonstrate safety and efficacy in human populations. Preclinical models, including in vitro cell cultures and in vivo animal models, provide critical insights into pharmacodynamics, pharmacokinetics, and toxicity profiles. Integrative approaches combining molecular biology, genomics, and systems pharmacology improve predictive accuracy and reduce late-stage failures. Translational research emphasizes biomarker identification, mechanism elucidation, and dose optimization to streamline the transition from preclinical studies to clinical trials. Advanced models, such as humanized mice and organ-on-chip systems, replicate human physiology more accurately, facilitating better predictions of therapeutic outcomes. By connecting basic research with clinical development, translational pharmacology accelerates innovation while ensuring patient safety and regulatory compliance.

Neuropharmacology explores the effects of drugs on the central and peripheral nervous systems, addressing neurological and psychiatric disorders such as Alzheimer’s disease, Parkinson’s disease, depression, and epilepsy. Advancements in CNS drug research focus on molecular mechanisms, neurotransmitter modulation, receptor targeting, and synaptic plasticity. Novel small molecules, biologics, and gene therapies are being developed to cross the blood-brain barrier effectively. Preclinical models, neuroimaging, and electrophysiology studies provide insights into efficacy, safety, and neurotoxicity. Precision approaches integrating genomics and biomarkers facilitate patient-specific therapies, improving outcomes for complex neurodegenerative and psychiatric conditions.

Cardiovascular pharmacology investigates drug interventions for heart and vascular diseases, including hypertension, heart failure, arrhythmias, and atherosclerosis. Research emphasizes molecular targets, signal transduction pathways, and receptor pharmacology to optimize therapeutic efficacy and safety. Advances include novel small molecules, biologics, and combination therapies aimed at modulating lipid metabolism, endothelial function, and cardiac contractility. Preclinical and clinical studies assess pharmacodynamics, pharmacokinetics, and cardiovascular risk profiles. Integration of precision medicine, biomarkers, and imaging technologies enables personalized treatment strategies, reducing morbidity and mortality while improving patient outcomes globally.

Oncology drug discovery focuses on identifying molecular targets, signaling pathways, and tumor microenvironment interactions to develop precision cancer therapies. Targeted treatments include kinase inhibitors, monoclonal antibodies, immune checkpoint modulators, and antibody-drug conjugates. Integration of genomics, proteomics, and computational modeling accelerates identification of actionable mutations and patient-specific therapies. Preclinical models and high-throughput screening facilitate selection of potent and selective compounds, while clinical trials evaluate efficacy, safety, and resistance mechanisms. Precision oncology strategies aim to optimize therapeutic response, minimize toxicity, and provide tailored interventions for diverse cancer types.

Natural products and phytopharmaceuticals remain a rich source for drug discovery, offering structurally diverse bioactive compounds. Research focuses on isolation, characterization, and optimization of compounds from plants, fungi, and marine organisms for therapeutic applications. These compounds are explored for anticancer, anti-inflammatory, antimicrobial, and metabolic disorder therapies. Advances in analytical techniques, high-throughput screening, and bioinformatics facilitate identification of novel scaffolds. Formulation technologies and pharmacokinetic optimization enhance bioavailability and clinical efficacy. Integration of traditional knowledge with modern pharmacology ensures sustainable and scientifically validated approaches for natural therapeutics.

Clinical trials are essential for evaluating the safety, efficacy, and pharmacological profile of new therapeutics. Rigorous design, ethical conduct, and regulatory compliance ensure data integrity and patient safety. This area encompasses phase I–IV trials, adaptive trial designs, and post-marketing surveillance. Regulatory frameworks, including FDA, EMA, and ICH guidelines, govern trial protocols, documentation, and approval pathways. Integration of real-world data, biomarkers, and patient stratification improves trial outcomes and accelerates approval timelines. Efficient regulatory navigation supports successful translation of discoveries into market-ready therapeutics.

Toxicology and safety assessment evaluate potential adverse effects of therapeutics on biological systems, ensuring risk mitigation before clinical application. Studies include acute, subchronic, and chronic toxicity, genotoxicity, reproductive toxicity, and immunotoxicity assessments. Preclinical and in vitro models, combined with computational toxicology, enhance predictive accuracy. Risk management strategies integrate pharmacovigilance, safety monitoring, and regulatory compliance. Advances in predictive modeling, high-content screening, and biomarker identification reduce late-stage failures and optimize drug safety profiles, supporting safe and effective therapy development.

Understanding pharmacokinetics (PK), pharmacodynamics (PD), and absorption, distribution, metabolism, and excretion (ADME) is critical for drug optimization. PK/PD studies determine dose-response relationships, therapeutic windows, and systemic exposure. ADME profiling ensures appropriate bioavailability, metabolic stability, and elimination, minimizing toxicity. Integration of modeling, simulation, and in vitro/in vivo studies supports rational dosing strategies and informs clinical trial design. Advanced analytical techniques, including mass spectrometry and bioinformatics, facilitate precise measurement of drug behavior. Comprehensive PK/PD and ADME analysis underpins the development of safe, effective, and personalized therapeutics.

Immunopharmacology explores modulation of the immune system to treat autoimmune diseases, inflammation, infections, and cancer. Innovations include immune checkpoint inhibitors, cytokine therapies, vaccines, and adoptive cell therapies. Research focuses on understanding immune signaling pathways, receptor-ligand interactions, and host-pathogen responses. Advances in monoclonal antibodies, biologics, and small molecules enhance specificity and efficacy while minimizing adverse immune reactions. Integration of genomics, proteomics, and computational modeling enables personalized immunotherapeutics. Emerging trends emphasize translational research, combination therapies, and predictive biomarkers to optimize immune modulation and therapeutic outcomes.