BOC Sciences boasts robust capabilities in drug screening techniques, which enable us to offer efficient and reliable screening services for drug development. We provide a variety of screening methods, combined with automated operating systems and high-sensitivity detection systems to ensure the efficiency of the screening process and the accuracy of the data. Our screening platform is equipped with multiple detection technologies, including optical, radioactive, and fluorescence detection, which can meet the needs of different types of screening. Moreover, BOC Sciences has access to a wide range of compound library resources, such as fragment libraries, natural product libraries, and bioactive compound libraries. These diverse compound libraries offer extensive options for drug screening, facilitating the identification of potential drug candidates.
Drug discovery screening is vital for identifying and optimizing bioactive compounds, yet persistent obstacles continue to limit R&D efficiency and data reliability. A key issue is the lack of physiologically relevant models, many assays fail to mimic in vivo conditions, leading to poor predictability and high attrition in development. Scaling from manual to high-throughput formats also demands costly automation and assay redevelopment. Hit compound specificity poses another major concern. Suboptimal strategies often generate false positives, diverting resources to weak leads. Limited access to integrated tools, such as high-content imaging, 3D cultures, constrains data depth and predictive accuracy. Inadequate cheminformatics and modeling capabilities further hinder interpretation and optimization. In addition, poor cross-team coordination, resource limits, and rising regulatory pressures create fragmented workflows and delays. These challenges call for innovative, scalable, and adaptable screening solutions. BOC Sciences delivers the integrated platforms and expertise to effectively address these barriers and accelerate your drug discovery efforts.
We maintain a diverse and evolving collection of chemical entities to support broad-spectrum screening applications and facilitate efficient identification of active candidates.
Our multidisciplinary team possesses extensive experience in drug discovery workflows, ensuring scientific rigor and operational efficiency throughout the screening process.
We employ a range of modern screening technologies and integrated automation systems to support high-throughput, high-content, and mechanism-based assays.
From initial consultation to compound screening, hit confirmation, and data interpretation, we provide end-to-end support to simplify project coordination and accelerate progress.
Our company provides a wide range of professional screening services tailored to support early-stage drug discovery and development. With expertise in both high-throughput and high-content screening, we combine advanced automation, structural biology, and AI-assisted approaches to deliver high-efficiency, data-rich solutions. Our team ensures rapid hit identification, target validation, and mechanism elucidation through validated and scalable workflows.
High-throughput screening is a rapid and efficient technique that allows screening of large chemical libraries at the molecular or cellular level. Utilizing automated microplate platforms (96-, 384-, or 1536-well formats). It is widely applied in target validation and hit identification.
High-content screening (HCS) integrates high-throughput automation with advanced imaging and analysis to assess multiple cellular parameters. This approach is critical for toxicity profiling, mechanism-of-action studies, and phenotypic screening.
Leveraging known 3D protein structures, this method designs molecules that precisely bind to target proteins. Techniques include molecular docking, and virtual screening, significantly improving the success rate of lead identification.
This technique screens small, low-molecular-weight fragments to discover novel chemical starting points that bind to the target protein. FBDD is instrumental in hit-to-lead optimization and the discovery of unique binding sites.
Employing computational modeling, machine learning, and AI algorithms, this technique accelerates screening through virtual simulations. It improves cost-efficiency and accuracy while narrowing down candidates before wet-lab validation.
This method identifies active compounds by measuring biological responses, such as enzyme activity, fluorescent signaling, or cellular functionality, upon exposure to candidate molecules. It enables precise identification of bioactive agents.
A target-agnostic approach that evaluates compound effects directly on cells or organisms. By observing phenotypic changes, researchers can discover novel compounds without prior knowledge of molecular targets, making it ideal for early-stage discovery.
Using specific biomarker expression levels as indicators, this screening approach is employed for both drug discovery and diagnostic applications. It provides a precise measure of drug efficacy or disease presence.
Focuses on identifying compounds that modulate enzyme activity. Widely used in mechanism-of-action studies and to discover enzyme inhibitors or activators relevant to disease pathways.
This assay monitors the impact of compounds on cellular behavior, including viability, proliferation, and signaling. It plays a pivotal role in identifying toxic compounds and evaluating therapeutic potential.
3D cultures better mimic in vivo tissue architecture and physiology. These models enable more predictive screening results, especially for efficacy and toxicity assessment under physiologically relevant conditions.
This method analyzes large-scale screening datasets to identify drug candidates. Leveraging big data, AI, and machine learning, it helps uncover patterns and correlations that might be overlooked in conventional screening.
When challenges arise during the drug screening process, our dedicated team of experts is on hand to provide comprehensive professional guidance and technical consultation. Utilizing a broad spectrum of advanced screening technologies and drawing on extensive industry experience, we collaborate closely with clients to identify and resolve specific obstacles, optimize assay parameters, and enhance the overall quality and reliability of screening results. Our tailored support is designed to address the unique needs of each project, ensuring that experimental workflows proceed smoothly and efficiently. At BOC Sciences, we are committed to delivering end-to-end assistance that fosters successful drug discovery outcomes, helping clients accelerate their research timelines with confidence.
BOC Sciences has established a comprehensive in vitro and in vivo pharmacological model platform, covering a wide range of classical and advanced models including cell lines, organoids, and disease-specific animal models. With standardized operational systems and expert technical capabilities, we support the entire drug development cycle, from mechanistic studies to efficacy and safety evaluations, ensuring robust, reproducible, and translational research outcomes.
Model Type | Representative Examples |
Tumor Cells | Lung cancer (A549), breast cancer (MCF-7), liver cancer (HepG2), colorectal cancer, prostate cancer |
Stem Cells | Embryonic stem cells, induced pluripotent stem cells (iPSC), mesenchymal stem cells (MSC) |
Neural Cells | Neurons derived from iPSC, neurotoxicity models, neurodegenerative disease-related assays |
Cardiomyocytes | iPSC-derived cardiomyocytes, cardiotoxicity assays, myocardial electrophysiology studies |
Hepatocytes | Primary hepatocytes, drug metabolism, and hepatotoxicity screening |
Pulmonary Cells | Alveolar epithelial cells, bronchial epithelial cells, airway inflammation modeling |
Immune Cells | THP-1, PBMCs, macrophage polarization, cytokine release, immunomodulation assays |
Organoid Models | Patient-derived tumor organoids, liver and gut organoids, personalized drug response |
3D Co-Culture Models | Tumor-stroma co-cultures, immune-tumor interaction models, microfluidic chip models |
Model Type | Representative Examples |
Tumor Models | Subcutaneous xenograft, orthotopic tumors, metastasis models |
Inflammatory Models | DSS-induced colitis, collagen-induced arthritis (CIA), systemic inflammation, cytokine profiling |
Diabetes Models | Type I/II diabetes, STZ-induced models, db/db mice, insulin resistance |
Obesity & Metabolic | High-fat diet-induced obesity, NASH, dyslipidemia, metabolic syndrome |
Neurodegenerative | Alzheimer's disease, Parkinson's, Huntington's disease |
Immunological Models | Autoimmune disease models (EAE, lupus), immunosuppression, graft-vs-host disease |
Liver Injury Models | Acute/chronic liver damage, hepatotoxicity, fibrosis models |
Cardiovascular Models | Myocardial infarction (LAD ligation), hypertension models, ischemia-reperfusion injury |
BOC Sciences is equipped with state-of-the-art technology platforms designed to support a broad spectrum of drug screening services. Our comprehensive infrastructure integrates cutting-edge instruments and automated systems, enabling high-efficiency, high-precision screening across diverse modalities. These platforms facilitate rapid compound evaluation, detailed mechanistic studies, and robust data generation, fully supporting drug discovery and development needs.
At BOC Sciences, we provide extensive compound libraries to facilitate diverse and efficient drug screening campaigns. Our collections cover a wide range of chemical and biological entities, enabling robust evaluation of compound efficacy, selectivity, and mechanism of action. Below are some of the key compound libraries we offer:
High-throughput screening libraries feature drug-like compounds optimized for screening compatibility and physicochemical tractability, supporting large-scale primary screening campaigns in early-stage drug discovery.
Our activity-based libraries contain compounds with experimentally validated pharmacological effects, streamlining hit-to-lead progression by providing active scaffolds against known biological endpoints.
These libraries are strategically constructed around specific target classes or pathways (e.g., kinases, GPCRs, ion channels), facilitating efficient lead identification through mechanism-focused compound sets.
Our diversity-oriented libraries comprise structurally diverse small molecules designed to maximize chemical space coverage, enabling broad screening and initial hit discovery across various biological targets.
Tailored to specific research needs, our custom libraries are designed in close collaboration with clients, incorporating defined structural motifs, physicochemical properties, or bioactivity profiles for specialized screening objectives.
Fragment-based libraries feature low-molecular-weight compounds (<300 Da) that support FBDD by enabling efficient target binding and structural optimization. Ideal for identifying novel scaffolds with high ligand efficiency.
Our natural product library features bioactive molecules isolated from various natural sources, serving as rich reservoirs for novel scaffold discovery and drug leads.
This library includes compounds with well-characterized biological activities, supporting mechanism elucidation and hit validation in screening workflows.
Communicate with the client to clarify project goals, compound features, targets, disease focus, and preferred screening type. This ensures alignment with therapeutic objectives and research direction.
Design tailored protocols with suitable assay formats (e.g., HTS, HCS, enzyme, or cell-based), defining dose ranges, controls, and readouts to ensure reliable and relevant results.
Build and validate models using relevant systems such as cell lines, 3D cultures, or animal models. Apply advanced technologies (e.g., ELISA, imaging, reporter assays) for sensitivity and reproducibility.
Conduct screening under optimized lab conditions, including dose–response or kinetic studies, to evaluate activity, specificity, and off-target effects.
Collect raw data via automated platforms and analyze statistically to determine hit rates, IC50/EC50, SAR, and phenotypic links. Apply QC through controls and repeats.
Provide structured reports with raw data, curves, heatmaps, and expert insights to support lead selection or further screening planning.
Drug screening methods include a diverse range of techniques designed to evaluate compound interactions with biological systems. Common approaches comprise high-throughput screening (HTS) for rapid testing of large compound libraries, high-content screening (HCS) which uses imaging to analyze multiple cellular parameters, structure-based and fragment-based drug discovery that leverage target protein structures, and computer-aided virtual screening for in silico evaluation. Other methods include activity-based assays, phenotypic screening that observes whole-cell or organism responses, biomarker-based assays, enzyme- and cell-based screenings, 3D cell culture systems for physiologically relevant models, and bioinformatics-driven approaches that integrate data analytics to refine screening candidates.
High-throughput screening (HTS) is the most widely used drug screening technique due to its capacity for automated, rapid, and parallel testing of thousands to millions of compounds. HTS enables efficient identification of active compounds against specific biological targets by combining robotics, sensitive detection methods, and data analysis software. Its scalability and adaptability to various assay formats make HTS a cornerstone technology in early-stage drug discovery programs across the pharmaceutical industry.
Target-based screening remains the most common type of drug screening, focusing on testing compounds against a specific molecular target such as a receptor, enzyme, or ion channel. This approach allows researchers to identify modulators that directly influence target activity, providing clear mechanistic insights and facilitating rational drug design. Due to its specificity and ability to streamline lead identification, target-based screening is extensively utilized in preclinical drug development to accelerate the discovery of candidate molecules with therapeutic potential.