The Use of Analytics in Biopharmaceuticals: An Introduction

News
Article
BioPharm InternationalBioPharm International May 2025
Volume 38
Issue 4
Pages: 19-23

This article provides an overview and introduction to the use of analytics in biopharmaceutical development and manufacturing.

Science, woman and man looking at test tube in laboratory, research in vaccine results and medical innovation. Healthcare, biotech lab analytics and medicine, scientist team in pharmaceutical study. | Image Credit: ©Talia Mdlungu/peopleimages.com – stock.adobe.com

Science, woman and man looking at test tube in laboratory, research in vaccine results and medical innovation. Healthcare, biotech lab analytics and medicine, scientist team in pharmaceutical study. | Image Credit: ©Talia Mdlungu/peopleimages.com – stock.adobe.com

In the rapidly evolving field of biopharmaceuticals, the application of analytics has become a cornerstone of research, development, and production. The biopharmaceutical industry, which focuses on developing drugs derived from biological sources such as proteins, cells, and genetic material, relies heavily on complex scientific processes. The complexity of biopharmaceuticals—ranging from monoclonal antibodies to gene therapies—demands sophisticated analytical techniques to ensure product quality, efficacy, and safety.

As the industry continues to innovate, with new therapies being developed for diseases once deemed untreatable, the role of analytics has expanded from simply ensuring regulatory compliance to facilitating cutting-edge breakthroughs. Analytics in biopharmaceuticals spans a wide range of areas, from pre-clinical discovery to clinical trials and large-scale manufacturing. It involves the use of various quantitative and qualitative techniques to evaluate the properties of drug candidates, identify potential issues in production, and monitor patient outcomes during clinical studies.

Overview of analytics in biopharmaceuticals

The biopharmaceutical industry leverages a broad spectrum of analytical tools and methodologies to address challenges across different stages of drug development and manufacturing. Analytics is not a monolithic field but rather an integrated framework that combines multiple disciplines, including chemistry, biology, physics, and data science. These tools are essential for making informed decisions that ensure the highest standards of product quality, regulatory compliance, and therapeutic success.

Discovery and early-stage development

In the early stages of biopharmaceutical development, analytics plays a critical role in identifying promising drug candidates and characterizing their properties. High-throughput screening (HTS), mass spectrometry, chromatography, and other molecular profiling techniques are employed to identify potential therapeutic targets and validate their biological activity. This phase is crucial for narrowing down the vast array of possibilities into a manageable set of drug candidates with the highest likelihood of success.

Analytics in this stage also involves the detailed analysis of proteins, nucleic acids, and other biologics, including their structural integrity and functional performance. Bioinformatics tools are used extensively to model and predict how these biologics will behave in biological systems, providing valuable insights for optimizing drug design.

Clinical development

As a promising drug candidate moves into clinical trials, analytics is used to monitor patient safety, pharmacokinetics (how the drug is absorbed, distributed, metabolized, and excreted), and pharmacodynamics (the drug’s effects on the body). In clinical settings, real-time data collection through biomarkers and advanced diagnostic tools allows researchers to better understand how the drug interacts with patients at the molecular and cellular levels.

Bioanalytical techniques, such as enzyme-linked immunosorbent assays (ELISAs), flow cytometry, and polymerase chain reaction (PCR)-based assays, are crucial for quantifying drug concentrations in biological samples. These assays are integral in assessing not only drug efficacy but also potential adverse effects, guiding the adjustment of doses or the early termination of a trial when necessary.

Manufacturing and process control

Once a biopharmaceutical product advances to large-scale production, analytics continues to play a pivotal role in ensuring product quality and consistency. Biomanufacturing processes, which often involve complex biological systems such as mammalian cell cultures, require rigorous monitoring to maintain optimal conditions. Advanced analytics is employed to track critical process parameters (CPPs) such as temperature, pH, nutrient levels, and dissolved oxygen.

Techniques such as chromatography, electrophoresis, and protein purification are used to isolate, purify, and validate biologics at various stages of production. Additionally, analytics plays a crucial role in monitoring and controlling batch-to-batch consistency, ensuring that the final product meets strict quality standards. The application of process analytical technology (PAT) enables real-time monitoring and control of the manufacturing process, which is essential for optimizing yields, reducing waste, and ensuring product safety and efficacy.

Regulatory compliance and quality assurance

Regulatory bodies such as FDA, the European Medicines Agency (EMA), and the International Council for Harmonisation (ICH) require biopharmaceutical companies to provide robust analytical data to demonstrate the safety, efficacy, and consistency of their products. Analytics is integral to this process, as companies must conduct a variety of tests to assess the stability, sterility, potency, and purity of their drug products. Analytical methods must be validated to ensure they can accurately measure the required parameters, and compliance with good manufacturing practices (GMP) must be maintained throughout the production process.

Data generated through analytical methods are essential for supporting regulatory filings, including investigational new drug (IND) applications and biologics license applications (BLA). In this context, the use of validated analytical techniques is crucial for meeting the stringent standards set by regulatory agencies and for ensuring that products are safe and effective for public use.

Post-marketing surveillance and real-world data

Even after a drug has been approved and reaches the market, analytics continues to play an important role in monitoring its long-term performance and identifying any unforeseen side effects. Post-marketing surveillance often involves the collection and analysis of real-world data (RWD) to assess how the drug performs in diverse patient populations.

Advanced data analytics tools, such as machine learning (ML) algorithms and predictive analytics, are increasingly being used to analyze large datasets from clinical trials, electronic health records, and patient registries. These insights can guide decision-making related to drug formulation adjustments, new indications, or safety measures, helping to improve patient outcomes and enhance the drug’s long-term therapeutic value.

Analytics in continuous biomanufacturing

Continuous biomanufacturing represents a transformative shift in the way biologic drugs are produced. Unlike traditional batch-based manufacturing, where products are made in distinct, time-bound cycles, continuous biomanufacturing involves the ongoing production of biopharmaceutical products in a seamless, uninterrupted process. This approach offers several advantages, including enhanced efficiency, scalability, improved product consistency, and cost reductions over time. However, to ensure these benefits are realized, effective and advanced analytics are crucial in monitoring and controlling the production process in real-time.

Analytics plays a pivotal role in continuous biomanufacturing by enabling the real-time measurement and monitoring of CPPs, ensuring product quality, and allowing for proactive adjustments to be made. Given that the production process is continuous, there is no opportunity for corrective action after the fact, making the role of analytics even more important. Continuous monitoring and data collection are key to understanding and controlling variables that impact product quality, such as temperature, pH, dissolved oxygen, and nutrient levels, as well as critical attributes such as protein concentration and glycosylation patterns.

This expanded overview of analytics in continuous biomanufacturing outlines its applications, the technologies used, and the critical role it plays in ensuring that biopharmaceutical products are produced at the highest quality and efficiency.

In the context of continuous biomanufacturing, analytics refers to a set of tools, techniques, and methods used to monitor, measure, and control the entire production process. Analytics are embedded throughout the continuous production system to provide insights into real-time process behavior, product quality, and the overall system performance. The integration of these analytics enables manufacturers to predict and maintain product quality, streamline production, and respond to potential issues before they impact the final product.

Real-time monitoring and control

Continuous biomanufacturing requires constant, real-time monitoring of various CPPs to ensure the production process remains within optimal conditions. This is in stark contrast to batch processing, where adjustments can be made at various stages of the production cycle. In a continuous system, however, analytical systems must be capable of detecting deviations instantly and providing corrective measures in real time.

Techniques such as PAT and in-line sensors are commonly employed for this purpose. PAT encompasses a range of tools that integrate real-time process monitoring with data analysis techniques. These tools help track and measure critical attributes, such as cell growth, metabolite concentrations, protein folding, and aggregate formation. The data generated by PAT can be used for dynamic adjustments to optimize yield and product quality.

By leveraging real-time analytics, manufacturers can better understand process dynamics and ensure that the system is in control, which is crucial for maintaining consistent product quality over extended production periods.

Predictive analytics and process modeling

Predictive analytics plays a significant role in continuous biomanufacturing by using historical and real-time data to forecast process behavior and predict potential issues before they occur. ML models and artificial intelligence (AI) are increasingly being employed to identify patterns in process data that may not be apparent through traditional methods.

For instance, predictive models can anticipate shifts in culture conditions, such as nutrient depletion or temperature fluctuations, and adjust system parameters to prevent deviations from optimal production conditions. Predictive analytics can also forecast product quality metrics, such as protein yield or purity, based on real-time input from various sensors and controllers.

The integration of predictive models into the continuous manufacturing process allows manufacturers to shift from reactive to proactive control, preventing issues that could compromise product quality, reduce efficiency, or lead to batch failures. In this way, predictive analytics helps improve process stability and reduce downtime, which is essential in continuous manufacturing environments.

Data integration and process optimization

Data generated during continuous biomanufacturing comes from a variety of sources, including sensors, equipment controllers, laboratory information management systems (LIMS), and manufacturing execution systems (MES). Integrating these data sources is key to optimizing production and ensuring that all process variables are aligned for the best possible outcome.

Through data integration, analytics can provide a holistic view of the entire manufacturing process, enabling operators to make informed decisions based on comprehensive, real-time data. This allows for better process optimization, as multiple parameters are adjusted simultaneously to maintain system stability.

Analytics can identify opportunities for improving yield, reducing waste, and optimizing resource use. For example, integrating process data can lead to adjustments in cell culture feeding strategies or better allocation of nutrients in real time, ultimately improving overall efficiency and product consistency. Moreover, data analytics helps monitor the system’s energy and resource usage, ensuring a more sustainable and cost-effective production process.

Product quality control and assurance

In continuous biomanufacturing, ensuring consistent product quality is paramount, as any variation in the process could result in deviations in the final product. Analytical techniques are essential for monitoring critical quality attributes (CQAs) of the biologic, such as protein titer, glycosylation patterns, and product purity. These quality attributes must be continuously assessed to ensure that the biopharmaceutical product meets the required specifications.

Advanced analytical techniques such as spectroscopy, chromatography, and mass spectrometry are used for real-time characterization of the bioproducts. For example, near-infrared (NIR) spectroscopy and Raman spectroscopy can be used in-line to monitor parameters such as protein concentration, moisture content, and secondary structure. These methods enable manufacturers to monitor product quality continuously without needing to wait for off-line analytical results, which is a major advantage over traditional batch processes.

Automating the quality control process through analytics also reduces the risk of human error, ensures compliance with regulatory standards, and increases throughput. Additionally, the implementation of quality-by-design (QbD) principles within continuous manufacturing helps integrate product quality control into every step of the process, ensuring higher consistency and reducing the likelihood of product failures.

Automation and AI integration

The integration of automation and AI in continuous biomanufacturing is a significant trend that enhances the role of analytics. Automated systems can collect, analyze, and respond to data in real time, reducing the need for manual intervention and increasing process efficiency. Machine learning algorithms can identify subtle trends in data that might not be apparent through traditional monitoring methods and can make adjustments accordingly.

AI can also predict maintenance needs, detect process anomalies, and recommend corrective actions, which is crucial in maintaining the continuity and efficiency of operations. For instance, predictive maintenance models can forecast when equipment or sensors will require maintenance, preventing costly downtime and ensuring that the production process remains uninterrupted.

Regulatory compliance and documentation

Regulatory compliance is an essential component of biomanufacturing, particularly for biologics, which must meet stringent safety and efficacy standards. Analytics ensures that continuous biomanufacturing processes remain compliant with regulations set by agencies such as FDA and EMA. Real-time data and robust documentation capabilities enable manufacturers to demonstrate control over their processes and provide the necessary evidence to support regulatory submissions.

The use of analytics to track and document every aspect of the manufacturing process ensures transparency and traceability, which are crucial for meeting regulatory requirements. It also facilitates the implementation of an effective quality management system (QMS) that supports consistency and safety in drug production.

Analytics in cell and gene therapy manufacturing

Cell and gene therapies (CGTs) represent a transformative approach to treating a wide range of diseases, including genetic disorders, cancer, and other chronic conditions. Unlike traditional small molecule drugs, CGTs involve complex biological materials, such as cells or genes, which are often engineered to target the root causes of disease. The manufacture of these therapies is an intricate process that demands precise, real-time monitoring and control to ensure the safety, efficacy, and consistency of the final product. In this context, analytical techniques play a critical role in the development, production, and release of cell and gene therapies.

Analytics in cell and gene therapy manufacturing encompasses a wide array of techniques and methodologies designed to assess, monitor, and validate various parameters throughout the manufacturing process. These techniques are essential not only for quality control but also for optimizing the entire manufacturing workflow, ensuring compliance with regulatory standards, and enabling continuous improvement in product development.

The complexity of cell and gene therapies arises from the need to handle living cells or genetic material in a way that maintains their therapeutic potential. The manufacturing processes for CGTs include cell collection, genetic modification, cell expansion, formulation, and delivery, each of which requires robust analytical tools to ensure that the product remains safe, stable, and effective.

Types of analytics in CGT manufacturing

The following are analytics commonly used in CGT manufacturing:

  • Quality control and assurance. This involves routine testing to ensure that each batch of therapy meets predefined specifications for identity, purity, potency, and sterility. Techniques such as flow cytometry, quantitative polymerase chain reaction (qPCR), enzyme linked immunosorbent assay (ELISA), and mass spectrometry are commonly used to verify the quality of the final product.
  • Process monitoring. Throughout the manufacturing process, it is critical to monitor CPPs such as temperature, pH, oxygen levels, and cell viability. Technologies such as real-time PCR, sensor-based systems, and automated platforms enable continuous monitoring to ensure optimal conditions for cell growth and gene transfer.
  • Genomic and proteomic analysis. Given the genetic modifications central to many cell and gene therapies, advanced genomic sequencing and proteomic profiling are used to verify gene edits and protein expression profiles. These tools ensure that the genetic material is correctly inserted and that the intended therapeutic proteins are being produced.
  • Cell characterization. The functional attributes of cells used in therapy—such as potency, transfection efficiency, and differentiation potential—must be carefully assessed. High-dimensional flow cytometry, single-cell RNA sequencing, and imaging technologies are commonly employed to characterize cellular phenotypes and assess the functionality of engineered cells.

Challenges in analytics for CGT manufacturing

The following are some challenges performing analytics in CGT manufacturing:

  • Complexity and variability. The biologically based nature of CGTs means that variability can arise not only from batch to batch but also from patient to patient. The diverse biological sources (e.g., autologous or allogeneic cells) further complicate the standardization and reproducibility of manufacturing processes. Analytical tools must be highly sensitive, robust, and capable of detecting subtle variations.
  • Scale-up and automation. Cell and gene therapies are typically produced in small batches, especially during early-phase clinical trials. However, as the industry shifts toward commercial-scale production, scaling up the manufacturing process without compromising quality becomes a significant challenge. Analytical methods need to evolve to provide accurate and rapid testing during scale-up operations, with automated platforms increasingly being integrated to improve efficiency and reduce human error.
  • Regulatory compliance. CGTs are subject to stringent regulatory scrutiny, with agencies such as FDA and EMA requiring comprehensive analytical data to ensure product safety and efficacy. Manufacturers must adhere to current good manufacturing practices (CGMP) and validate their analytical methods to meet these regulatory standards.

Emerging analytical technologies

The following are some emerging analytical technologies for CGTs:

  • Real-time monitoring and in-situ analytics. Advances in real-time, in-situ monitoring technologies, such as biosensors, miniaturized platforms, and microfluidics, are helping manufacturers gain more immediate insights into the biological processes during cell culture and gene modification. These technologies facilitate quicker adjustments to optimize manufacturing processes and reduce batch failures.
  • AI/ML–AI/ML are increasingly being integrated into the CGT manufacturing process to analyze large datasets generated by multi-parameter analytics. These technologies can predict potential outcomes based on historical data, identify trends, and optimize process control strategies. By integrating AI/ML into analytics, manufacturers can achieve higher process consistency, reduce time to market, and improve overall efficiency.

Future trends

The following are some future trends for analytics in CGTs:

  • Personalized medicine. As CGTs evolve to more personalized treatments, where therapies are tailored to individual patient characteristics, the need for patient-specific analytics becomes critical. New platforms that can analyze patient-specific biomarkers or genetic profiles will play a pivotal role in ensuring that therapies are developed and manufactured for maximum therapeutic benefit.
  • Integration of analytics across the supply chain. For the efficient manufacturing of CGTs, integrating analytics across the entire supply chain—ranging from raw material sourcing to final product release—will be essential. This holistic approach will ensure that each component of the supply chain is tightly controlled, minimizing risks and ensuring consistency.

Conclusion

Analytics is at the heart of continuous biomanufacturing, driving improvements in product quality, process efficiency, and scalability. The ability to monitor and control every aspect of the manufacturing process in real time—coupled with the power of predictive modeling, AI, and process optimization—ensures that continuous biomanufacturing can meet the growing demand for biologic drugs while maintaining the highest standards of quality and compliance. As the biopharmaceutical industry continues to move towards continuous manufacturing, the role of advanced analytics will only become more critical in enabling the production of life-saving therapeutics in a more efficient, cost-effective, and sustainable manner.

The integration of advanced analytics in the biopharmaceutical industry has transformed the way drugs are developed, tested, and manufactured. From accelerating early-stage research to ensuring quality control in large-scale production, analytics enables biopharmaceutical companies to create innovative treatments that are both effective and safe for patients. As the industry continues to embrace new technologies such as artificial intelligence, big data, and personalized medicine, the role of analytics will only grow more critical in driving the next generation of life-saving therapies.

Analytics are indispensable in CGT manufacturing, providing the necessary tools to monitor, control, and optimize the production process. As technologies continue to advance, the scope and impact of analytics will only grow, enabling the industry to meet the increasing demand for these groundbreaking therapies while maintaining the highest standards of quality, safety, and efficacy.u

About the author

Robert Dream is managing director at HDR Company LLC.

Article details

BioPharm International®
Vol. 38, No 4
May 2025
Pages: 19-23

Citation

When referring to this article, please cite it as Dream, R. The Use of Analytics in Biopharmaceuticals: An Introduction. BioPharm International 2025 38, 4.

Recent Videos
Related Content
© 2025 MJH Life Sciences

All rights reserved.