Call for Abstract

13th International Conference and Exhibition on Chromatography and Analytical Techniques, will be organized around the theme “Next-Generation Chromatographic Platforms: Speed, Resolution, and Accuracy”

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

Submit your abstract to any of the mentioned tracks.

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

Gas chromatography (GC) continues to evolve as one of the most powerful tools in analytical chemistry for separating volatile compounds. Recent advances focus on enhanced column efficiency, miniaturization, fast GC, and the development of more selective detectors. These improvements have significantly reduced analysis time while increasing resolution and accuracy. Innovations in sample introduction, such as headspace analysis, solid-phase microextraction (SPME), and thermal desorption, have enhanced GC’s applicability across environmental monitoring, food safety, petrochemical analysis, and forensic sciences. Moreover, modern GC systems integrated with advanced software enable real-time data acquisition and automation, making them indispensable in both R&D and routine testing environments. The integration of gas chromatography with mass spectrometry (GC-MS) has further widened its application scope by providing qualitative and quantitative data with high sensitivity and specificity. With sustainability gaining importance, low-energy GC systems and green carrier gas alternatives like hydrogen are also gaining attention. This session explores the transformative trends, practical challenges, and future pathways in gas chromatography for both academic and industrial applications.

Liquid chromatography (LC) is a cornerstone of analytical separation techniques, used extensively for separating non-volatile and thermally unstable compounds. Advances in high-performance liquid chromatography (HPLC) and ultra-high-performance liquid chromatography (UHPLC) have significantly enhanced speed, resolution, and sensitivity. Current developments include monolithic and superficially porous columns, innovative stationary phases, and ultra-low dispersion systems. These innovations have made LC more robust in handling complex matrices in pharmaceuticals, food analysis, environmental testing, and biological samples. Gradient elution techniques and multidimensional LC are improving peak capacity and allowing better resolution of analytes. Mobile phase modifiers and green solvents are being incorporated to make the processes more environmentally friendly and cost-efficient. The emergence of automated LC systems has also allowed high-throughput workflows in clinical laboratories and quality control settings. The coupling of LC with mass spectrometry (LC-MS) has pushed boundaries in precision and identification, especially in proteomics and metabolomics. This topic focuses on the evolving capabilities of liquid chromatography in solving real-world analytical challenges and enhancing research outcomes.

Hyphenated techniques, such as GC-MS (Gas Chromatography-Mass Spectrometry) and LC-MS (Liquid Chromatography-Mass Spectrometry), represent a pivotal advancement in analytical science by combining the separation power of chromatography with the detection accuracy of mass spectrometry. These hybrid systems offer a comprehensive approach to qualitative and quantitative analysis of complex mixtures, enhancing both selectivity and sensitivity. GC-MS is extensively used for volatile and semi-volatile organic compounds in forensic toxicology, environmental science, and petrochemical industries, whereas LC-MS plays a crucial role in pharmaceutical analysis, biomarker discovery, and food safety monitoring. The rise of triple quadrupole and high-resolution mass analyzers has elevated data quality, enabling lower limits of detection and improved accuracy. Developments in ionization techniques like ESI, APCI, and MALDI have extended applications to a wider array of analytes, including large biomolecules. Automated sample preparation and real-time data analytics now further support regulatory compliance and high-throughput demands. The fusion of these techniques forms the backbone of modern analytical laboratories, bridging the gap between separation and molecular identification.

Chiral separation is essential in the analysis and production of enantiomerically pure compounds, particularly in the pharmaceutical and agrochemical industries. As enantiomers can exhibit markedly different biological activities and toxicological profiles, the ability to separate and quantify each enantiomer is critical. Advances in chromatographic chiral separation methods, including chiral stationary phases (CSPs) and chiral mobile phase additives, have enabled faster, more efficient, and selective separations. Techniques such as supercritical fluid chromatography (SFC), HPLC, and capillary electrophoresis (CE) with chiral selectors have expanded chiral analysis beyond traditional approaches. The emergence of polysaccharide-based CSPs and protein-based phases has increased versatility, allowing high enantioselectivity across diverse analyte classes. Method development and validation remain central in ensuring regulatory compliance, particularly under FDA and EMA guidelines. Innovations in column chemistry, automation, and integration with MS detection are propelling chiral analysis into more dynamic and robust workflows. The importance of chiral separation spans across drug development, quality control, and biosimilar research, making it an indispensable tool in analytical science.

Bioanalytical chromatography plays a crucial role in the quantification and characterization of biomolecules such as proteins, peptides, nucleic acids, and metabolites in biological matrices. These techniques are pivotal in pharmacokinetics, pharmacodynamics, bioequivalence studies, and therapeutic drug monitoring. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) is the gold standard in bioanalysis due to its sensitivity, specificity, and throughput. Advances in sample preparation methods like protein precipitation, solid-phase extraction (SPE), and microextraction have streamlined workflows and reduced matrix effects. Furthermore, developments in nano-LC, microfluidic chips, and automation are enabling more accurate and reproducible analysis of low-abundance biomarkers. Regulatory agencies emphasize rigorous method validation, including parameters like selectivity, precision, and stability, making method robustness crucial. In addition to small molecule drugs, bioanalytical chromatography is now central in biologics and biosimilar analysis, including monoclonal antibodies and gene therapies. As personalized medicine and biomarker-driven therapies expand, bioanalytical chromatography continues to provide the analytical backbone for advancing healthcare outcomes and precision diagnostics.

Capillary electrophoresis (CE) is a high-efficiency separation technique widely used for analyzing small ions, biomolecules, and chiral compounds. Its core principle is the differential migration of charged analytes through a capillary under an electric field, offering excellent resolution and rapid analysis with minimal sample and reagent consumption. Recent advancements in CE include microchip-based formats, improved capillary coatings to minimize adsorption, and coupling with mass spectrometry (CE-MS) for enhanced detection capabilities. These innovations have broadened its applications in pharmaceutical analysis, forensic science, clinical diagnostics, and proteomics. CE is especially beneficial for analyzing proteins, peptides, and nucleic acids, making it a key tool in biotechnology and bioanalytical labs. The technique’s compatibility with aqueous buffers and low environmental impact align well with green chemistry principles. Automated systems and multiplexing have further increased throughput and reproducibility, making CE a practical solution for high-volume laboratories. This topic highlights how capillary electrophoresis stands as a cost-effective, robust, and environmentally friendly technique with growing relevance across diverse scientific and industrial sectors.

Chromatographic method validation ensures analytical methods are scientifically sound, reproducible, and suitable for their intended purpose. This critical step in quality assurance aligns with international guidelines such as those from the ICH, FDA, and EMA. Validation parameters include accuracy, precision, linearity, range, detection and quantitation limits, robustness, and specificity. As regulations tighten and analytical demands increase, method validation has become more complex and rigorous. Innovations in software tools, system suitability testing, and real-time validation protocols have streamlined the process and reduced human error. In pharmaceutical industries, validated chromatographic methods are essential for drug development, release testing, and stability studies. Environmental, food, and clinical laboratories also rely on method validation to ensure data integrity and regulatory compliance. With the expansion of hyphenated and automated technologies, validation protocols must now accommodate multi-analyte, multi-matrix environments. This session emphasizes the role of validated chromatography methods in delivering reliable, defensible results and showcases the latest practices and technologies that enhance validation efficiency across analytical workflows.

Environmental sample analysis using chromatography has become indispensable in monitoring and managing pollutants across air, water, soil, and biota. Techniques such as gas chromatography (GC), liquid chromatography (LC), and capillary electrophoresis (CE) enable precise quantification of trace-level contaminants like pesticides, heavy metals, volatile organic compounds (VOCs), and persistent organic pollutants (POPs). Recent advances include high-resolution mass spectrometry (HRMS), two-dimensional chromatography, and automated solid-phase extraction (SPE), which have significantly improved sensitivity, selectivity, and sample throughput. Portable and field-deployable instruments now allow on-site analysis, accelerating decision-making in environmental monitoring. Regulatory bodies such as the EPA and EU REACH require validated chromatographic methods for pollutant detection and compliance reporting, further emphasizing their significance. Green analytical practices, including solvent recycling and miniaturized systems, are being incorporated to align with sustainability goals. This session explores the integration of cutting-edge chromatographic technologies with environmental science, focusing on innovative workflows, data handling, and regulatory standards to better understand and mitigate environmental risks.

Chromatography plays a vital role in ensuring the safety, quality, and authenticity of food and beverages. Techniques like HPLC, GC, and LC-MS/MS are widely applied for detecting contaminants, residues, additives, and nutritional components. Recent innovations in fast chromatography, improved stationary phases, and miniaturized systems have enhanced analytical speed and sensitivity, supporting both regulatory compliance and industry quality control standards. Applications include the detection of pesticide residues, mycotoxins, food allergens, flavor compounds, and preservatives, as well as profiling bioactive nutrients and functional ingredients. Isotope ratio analysis and fingerprinting methods also help in confirming food origin and authenticity, combating food fraud. With increasing demand for trace-level detection and real-time analysis, integrated systems with AI-assisted interpretation and automation are becoming standard in modern food laboratories. Regulatory authorities such as the FDA, EFSA, and WHO mandate stringent testing protocols, which chromatography fulfills reliably. This session delves into the evolving role of chromatographic methods in food testing, highlighting advancements that enable faster, greener, and more comprehensive assessments to protect public health.

Pharmaceutical quality control (QC) relies heavily on chromatography to ensure the purity, potency, and safety of drugs throughout the manufacturing lifecycle. High-performance liquid chromatography (HPLC) is the backbone of QC, used extensively for content uniformity, impurity profiling, dissolution testing, and stability analysis. Emerging technologies like UHPLC, two-dimensional LC, and LC-MS provide enhanced resolution and faster run times, allowing for high-throughput and more detailed analysis. Robust method development and validation are integral to meeting regulatory standards from bodies such as the FDA, EMA, and WHO. Automated sampling, data acquisition, and audit-ready software are becoming essential for compliance with Good Manufacturing Practice (GMP) regulations. Moreover, green and sustainable practices are being adopted to reduce solvent use and operational costs. Chromatographic techniques are also pivotal in detecting nitrosamines and other genotoxic impurities, which are gaining regulatory scrutiny. This session covers the role of chromatography in pharmaceutical QC, with a focus on new technologies, regulatory trends, and quality assurance strategies that support drug safety and efficacy.

Chromatography has become a cornerstone of modern clinical diagnostics, offering high specificity and sensitivity for detecting biomarkers, metabolites, and therapeutic drugs in biological matrices. Techniques such as LC-MS/MS and GC-MS are routinely used for therapeutic drug monitoring, hormone analysis, and toxicology screening. These methods enable rapid and accurate identification of disease states, patient compliance with medications, and early detection of metabolic disorders. The emergence of microfluidic and automated sample preparation systems has streamlined workflows in hospital and clinical laboratories. Additionally, bioanalytical method validation ensures diagnostic accuracy and supports regulatory compliance. In recent years, the demand for personalized medicine has driven the development of multiplexed chromatographic platforms capable of simultaneously measuring multiple analytes in small sample volumes. With integration into laboratory information systems (LIS) and support from artificial intelligence for data interpretation, chromatography is poised to further transform point-of-care testing and centralized lab diagnostics. This session highlights the innovations, regulatory perspectives, and clinical significance of chromatographic methods in diagnostics, reinforcing their indispensable role in advancing precision healthcare.

The characterization and quantification of proteins and peptides are crucial in biopharmaceutical development, proteomics, and biomedical research. Chromatographic techniques such as reversed-phase liquid chromatography (RPLC), ion-exchange chromatography (IEX), size-exclusion chromatography (SEC), and hydrophilic interaction chromatography (HILIC) are extensively used for separating complex protein mixtures, analyzing post-translational modifications, and determining protein purity. Coupling with high-resolution mass spectrometry (LC-MS) further enables in-depth identification and structural elucidation of peptide fragments. Advancements in stationary phases, bioinert columns, and online sample preparation methods have significantly enhanced analytical efficiency and reproducibility. Automation, multi-dimensional chromatography, and micro/nanoflow LC systems have become standard in high-throughput labs. These developments are critical in quality control of biotherapeutics, including monoclonal antibodies and vaccines. Moreover, robust methods are necessary for compliance with regulatory standards governing biologics. This session explores state-of-the-art techniques and applications of chromatography in protein and peptide analysis, focusing on innovations that ensure reproducibility, sensitivity, and comprehensive coverage in complex biological systems.

High-throughput screening (HTS) is pivotal in drug discovery, biomarker identification, and clinical research, where large volumes of samples must be processed rapidly and accurately. Chromatographic techniques—particularly UHPLC and multiplexed LC-MS systems have evolved to meet these demands by enabling ultra-fast separation, minimal sample consumption, and automated operation. With parallel processing, miniaturized platforms, and robotic integration, HTS allows simultaneous analysis of hundreds to thousands of samples per day. The pharmaceutical and biotechnology industries use HTS for lead compound screening, metabolic profiling, and pharmacokinetic evaluations. Moreover, advances in software for data processing and interpretation, including AI and machine learning tools, are accelerating discovery cycles and enhancing decision-making. Integration with other high-throughput modalities such as mass spectrometry imaging and microarrays offers comprehensive profiling capabilities. This session covers the recent advancements, automation strategies, and analytical precision of chromatography in HTS environments, emphasizing how scalable and efficient techniques are reshaping the speed and scope of modern research and development.

As sustainability becomes a core principle in analytical science, green chromatography offers environmentally responsible solutions without compromising analytical performance. This approach focuses on minimizing solvent use, reducing energy consumption, and employing biodegradable materials wherever possible. Techniques such as supercritical fluid chromatography (SFC), miniaturized columns, and aqueous mobile phases are gaining prominence in both academic and industrial settings. Method development now incorporates sustainability metrics, including solvent volume, toxicity, and waste generation, encouraging laboratories to adopt eco-friendly practices. Advances in instrumentation such as energy-efficient detectors and solvent recycling systems—support this transition. Regulatory bodies and quality standards organizations are beginning to recognize the importance of green analytics, creating further momentum. Green chromatography is especially valuable in pharmaceutical, environmental, and food industries, where large-scale analyses can benefit from reduced resource usage and improved safety. This session discusses innovative strategies, instrumentation, and case studies in green chromatography, underlining how sustainability and performance can coexist to shape the future of responsible analytical science.

Nanochromatography is an emerging field that enables high-resolution analysis using ultra-low flow rates and micro- to nano-scale columns. With increased sensitivity and reduced sample requirements, nanochromatography is particularly advantageous in proteomics, metabolomics, and biomarker discovery, where sample volumes are often limited. Coupled with mass spectrometry (nanoLC-MS), it delivers precise quantitation and detailed molecular profiling. Innovations in nano-scale column fabrication, fluidic control systems, and sample injection techniques have significantly improved reproducibility and robustness. These developments make nanochromatography a powerful tool in cutting-edge biomedical and pharmaceutical research. The reduced solvent consumption also aligns with green analytical chemistry principles, offering both environmental and cost benefits. Automated platforms now integrate sample preparation, separation, and detection into miniaturized systems, ideal for high-throughput laboratories and field-based applications. This session explores the latest trends in nanochromatography, from nanoLC-MS instrumentation to its transformative applications in personalized medicine, clinical diagnostics, and next-generation therapeutic research, illustrating its growing impact on precision analytical methodologies.

Chromatography plays a pivotal role in forensic science, offering precise, reproducible, and rapid identification of chemical substances across a variety of complex matrices. Gas chromatography (GC), high-performance liquid chromatography (HPLC), and LC-MS/MS are routinely applied in toxicology, arson investigations, drug testing, and trace evidence analysis. These tools enable forensic experts to detect and quantify substances such as narcotics, explosives, alcohols, and chemical residues, often at minute concentrations. Advancements in portable GC-MS systems have made it possible to perform on-site analyses at crime scenes, improving response time and preserving the integrity of evidence. Derivatization techniques, sample extraction advancements, and microfluidic integration are improving sensitivity and reducing processing times. Additionally, validated chromatographic methods adhere to judicial and regulatory requirements, ensuring the legal admissibility of results. With the integration of machine learning and spectral libraries, data interpretation is becoming faster and more accurate. As forensic science embraces innovation, chromatographic tools continue to evolve as essential instruments for delivering justice through scientifically rigorous and defensible evidence analysis.

Chromatography is a cornerstone of metabolomics and proteomics, enabling the separation, identification, and quantification of small molecules and proteins in biological systems. Techniques such as liquid chromatography–mass spectrometry (LC-MS), ion mobility spectrometry, and multidimensional chromatography offer high-resolution insight into cellular pathways, metabolic fluctuations, and disease biomarkers. These approaches are central to systems biology, personalized medicine, and biomarker discovery. In proteomics, chromatography supports peptide mapping, protein digestion analysis, and post-translational modification profiling. In metabolomics, it provides comprehensive coverage of endogenous metabolites across various tissues and fluids. Miniaturization, nanoflow LC, and improved column chemistries have dramatically increased sensitivity and throughput, while stable isotope labeling and advanced sample prep techniques enhance quantification accuracy. Combined with bioinformatics platforms and machine learning, chromatographic data now drives predictive modeling and deeper understanding of cellular function. These integrated workflows not only uncover biological complexity but also contribute to diagnostics, therapeutic development, and nutritional science. Continued innovations in chromatographic systems promise even more refined tools for exploring life at the molecular level.

Columns are the heart of chromatographic separation, and recent developments in column technology have significantly advanced the speed, efficiency, and resolution of analytical workflows. Novel stationary phases, including monolithic, core-shell, and sub-2 µm particle columns, have enhanced separation performance while reducing backpressure and analysis time. These innovations allow for more robust and reproducible methods in both liquid and gas chromatography. Functionalized surfaces and advanced bonding chemistries support highly selective separations across diverse sample types, from small molecules to large biomolecules. Temperature- and pH-stable columns have expanded the operational versatility, especially in pharmaceutical, environmental, and bioanalytical applications. Smart columns with embedded sensors and RFID tracking are now emerging, offering real-time performance monitoring and lifecycle tracking to support quality assurance. Additionally, greener column technologies featuring recyclable materials and reduced solvent requirements are gaining traction. As analytical demands grow, especially in high-throughput environments, evolving column technologies are meeting the need for precision, speed, and sustainability in modern chromatography, reshaping the foundation of separation science.

The growing need for efficiency, accuracy, and reproducibility in analytical chemistry has fueled the widespread integration of automation in laboratories. From robotic sample handling and automated injection systems to fully integrated liquid chromatography platforms, automation streamlines workflows, minimizes human error, and boosts throughput. In chromatography, automated systems perform tasks such as sample preparation, filtration, derivatization, and even column switching without manual intervention. These tools are particularly valuable in pharmaceutical quality control, clinical diagnostics, and environmental monitoring, where high sample volumes demand rapid turnaround. Integration with Laboratory Information Management Systems (LIMS) and software-controlled instrumentation ensures traceability and compliance with regulatory standards. Artificial intelligence and machine learning algorithms are being incorporated into automated platforms to optimize method development and predict instrument maintenance. Additionally, cloud-based monitoring allows for remote access and real-time troubleshooting. Automation not only reduces operational costs and analyst burden but also enhances data integrity. As the analytical sciences continue to scale, laboratory automation is playing a critical role in driving productivity and innovation.

The evolution of real-time data analytics in chromatography has transformed traditional analytical workflows by enabling immediate decision-making, faster troubleshooting, and higher-quality outputs. With modern detectors and intelligent software, analytical instruments now generate continuous streams of high-resolution data. Real-time processing allows for immediate peak integration, baseline correction, and anomaly detection, reducing analysis times and improving precision. This dynamic capability is vital in quality control environments where batch release decisions depend on rapid validation. Integrated software platforms now offer adaptive analytics powered by artificial intelligence and machine learning, enabling predictive maintenance, optimization of separation conditions, and method robustness assessments. Data dashboards, cloud-based storage, and mobile-accessible analytics further enhance transparency and collaboration. Additionally, real-time analytics is accelerating method development by offering instant feedback on parameter adjustments, saving time and resources. In research, this capability allows scientists to monitor complex reactions and biological interactions as they occur. As data-driven science becomes the standard, real-time analytics is redefining the future of chromatographic performance and process intelligence.