Monoclonal Antibody Manufacturing Processes

monoclonal



The monoclonal antibody manufacturing process plays an important role in producing therapeutic antibodies widely used in treating diseases such as cancer, autoimmune disorders and other infectious diseases. Monoclonal antibodies are laboratory-created molecules that mimic the immune system’s ability to target specific antigens. They are designed to bind to specific proteins or cells in the body, facilitating targeted therapy.

This article will delve into the intricate details of mAb manufacturing processes, considering the key steps involved, such as cell line development, upstream and downstream processing, purification and quality control. We will also discuss the challenges and advancements in the field, highlighting the importance of these manufacturing processes in developing life-saving therapeutics.

Importance of monoclonal antibodies in biopharma

Monoclonal antibodies have gained immense significance in research and medicine due to their specificity, efficacy and versatility in targeting various diseases and conditions. One of the primary reasons for the importance of monoclonal antibodies is their high specificity. Unlike traditional drugs that often interact with multiple targets in the body, monoclonal antibodies can be engineered to bind to a specific antigen with exceptional precision. This specificity enables targeted therapy, minimizes potential side effects and increases patient therapeutic benefit.

Monoclonal antibodies are extensively used in research as valuable tools for understanding disease mechanisms. They enable scientists to identify and study specific proteins, receptors and pathways involved in various diseases. By targeting these molecular targets, scientists can gain insights into disease progression, develop diagnostic tests and discover new therapeutic strategies.

In medicine, monoclonal antibodies have revolutionized the treatment of numerous diseases. They have been successfully employed in managing cancer, autoimmune disorders, infectious diseases and more. For example, monoclonal antibody therapies such as trastuzumab and rituximab have significantly improved the survival rates and quality of life for patients with breast cancer and non-Hodgkin lymphoma, respectively.

Monoclonal antibodies can act through various mechanisms to exert their therapeutic effects. They can block the activity of specific molecules involved in disease progression, modulate the immune response, deliver toxic payloads to target cells or enhance the body’s immune system to combat pathogens. This versatility makes them highly adaptable to different disease targets and treatment strategies.

Recently, monoclonal antibodies have also helped combat global health crises. During the COVID-19 pandemic, monoclonal antibody therapies such as casirivimab/imdevimab and bamlanivimab/etesevimab have been authorized for emergency use in high-risk patients, offering a targeted approach to neutralize the SARS-CoV-2 virus.

Overview of monoclonal antibody manufacturing processes

The manufacturing process for monoclonal antibodies involves cell line development, upstream cell culture, antibody production, harvest and clarification, purification, formulation, fill and finish, and rigorous quality control testing. Each step requires careful optimization and adherence to strict regulatory guidelines to ensure the production of high-quality monoclonal antibodies for therapeutic use.

  • Identification and Selection of Antibody

The first step in manufacturing monoclonal antibodies is identifying and selecting the specific antibody with desired binding properties. This antibody is usually derived from a hybridoma cell line or generated using recombinant DNA technology.

  • Cell Line Development

Once the antibody is identified, a cell line capable of producing the antibody is established. This involves growing the selected hybridoma cells or transfected host cells, such as Chinese hamster ovary (CHO) cells, in culture and screening for high antibody-producing clones.

  • Upstream Process

The upstream process involves growing the selected cell line in bioreactors under controlled conditions. The cells are cultivated in a suitable growth medium, which provides the necessary nutrients for cell growth and antibody production. Cell growth is monitored, and various parameters such as temperature, pH, dissolved oxygen and nutrient concentrations are optimized to achieve maximum cell density and antibody productivity.

  • Antibody Production

During the upstream process, the cells produce and secrete the monoclonal antibody into the culture medium. Antibody production is monitored, and the culture is periodically harvested to obtain the desired antibody concentration.

  • Harvest and Clarification

Once the desired antibody concentration is reached, the culture medium is harvested and subjected to a series of clarification steps to remove cellular debris, cell fragments and other impurities. This typically involves centrifugation, filtration and/or precipitation techniques.

  • Purification

The clarified harvest is then subjected to a purification process to isolate the monoclonal antibody from other contaminants. Multiple chromatography steps, such as protein A affinity chromatography, ion exchange chromatography, hydrophobic interaction chromatography and size exclusion chromatography, are typically employed to achieve high purity and remove impurities. Each purification step exploits specific characteristics of the antibody, such as its affinity for certain ligands or charge properties.

  • Formulation and Final Product Preparation

After purification, the monoclonal antibody is formulated into a final dosage form suitable for administration. This may involve adjusting the pH, concentration and buffer composition of the antibody solution. Stabilizers and excipients may also be added to enhance stability and shelf life.

  • Fill and Finish

The final formulated monoclonal antibody is then filled into vials or other appropriate containers. This process includes labeling, sealing and packaging the finished product under sterile conditions.

  • Quality Control

Throughout the manufacturing process, extensive quality control tests are conducted to ensure the safety, identity, purity and potency of the monoclonal antibody. These tests include analysis of the antibody’s physical and chemical properties, such as protein concentration, size, charge and binding affinity. Microbiological tests, sterility testing and endotoxin assays are also performed to ensure product safety.

Hybridoma Technology

Hybridoma technology is a technique used to generate monoclonal antibodies from immune cells called B cells. The process begins with immunizing an animal, usually a mouse, with the target antigen. This stimulates the mouse’s immune system to produce a diverse population of B cells that produce different antibodies.

Once an immune response is obtained, B cells are isolated from the animal’s spleen and fused with immortal myeloma cells (cancerous cells that can divide indefinitely to provide a continuous source of antibodies), resulting in hybrid cells called hybridomas.

The hybridomas are then cultured in a selective medium that allows only the hybridomas to survive while inhibiting the growth of unfused cells. These cultures are screened to identify the hybridomas that produce the desired monoclonal antibodies. This screening process typically involves various techniques like enzyme-linked immunosorbent assay (ELISA) or flow cytometry.

Once the hybridomas producing the desired antibodies are identified, they can be expanded in culture to produce large quantities of monoclonal antibodies. These antibodies can be harvested from the culture medium and purified for further use in therapeutic or diagnostic applications.

Recombinant DNA Technology

Recombinant DNA technology, also known as genetic engineering, offers an alternative method for producing monoclonal antibodies. Instead of relying on hybridomas, this approach involves introducing genes encoding the antibody’s variable regions into host cells, such as bacteria, yeast or mammalian cells.

The genes encoding the antibody’s variable regions, which determine its specificity, are isolated from hybridomas or synthesized through DNA synthesis techniques. These genes are then inserted into expression vectors, which act as vehicles to transfer the genes into host cells.

The host cells are transfected or transformed with the expression vectors, allowing them to produce the monoclonal antibodies. The host cells are selected based on their ability to correctly express and secrete the antibodies. The recombinant antibodies can be produced on a larger scale by growing the host cells in bioreactors or fermentation tanks.

Once the recombinant antibodies are produced, they undergo purification processes to isolate and remove impurities, ensuring high-quality and homogeneous antibody products. The purified monoclonal antibodies can be used in various applications, including targeted therapies, diagnostic assays and research studies.

Cell culture techniques for monoclonal antibody production

Monoclonal antibody (mAb) production involves the use of cell culture techniques to grow and maintain mammalian cells, such as Chinese hamster ovary (CHO) cells, which are commonly used for large-scale mAb production. These techniques are essential for achieving high yields of mAbs. Here is an overview of the cell culture techniques employed in monoclonal antibody production:

Cell Line Selection and Maintenance

Cell line selection is a critical step in monoclonal antibody production. It involves choosing a suitable cell line that can efficiently produce the desired antibodies. The commonly used cell lines include CHO cells, NS0 myeloma cells and HEK293 cells. The selection criteria typically include productivity, growth characteristics, stability and regulatory compliance.

Once a cell line is selected, it needs to be maintained in optimal conditions. This involves regular subculturing, where a small number of cells are transferred to fresh culture vessels to ensure continuous cell growth. The maintenance medium usually contains nutrients, growth factors and supplements necessary for cell viability and growth. The cells are incubated at controlled temperature, humidity and carbon dioxide levels to provide an ideal environment for their proliferation.

Cell line stability is also crucial to ensure consistent monoclonal antibody production over time. Regular monitoring and characterization of the cell line, such as genetic stability assessments and phenotypic analysis, are performed to confirm its stability and uniformity.

Media Optimization

Media optimization plays a significant role in maximizing the production of monoclonal antibodies. Cell culture media contain nutrients, vitamins, amino acids and growth factors necessary for cell growth and antibody production. The media composition can be optimized to enhance cell growth, productivity and product quality.

Optimization techniques may involve adjusting the concentration of specific nutrients, supplements or growth factors to support optimal cell growth and antibody production. Additionally, media optimization can also include the addition of chemically defined components to minimize batch-to-batch variation and ensure reproducibility.

The optimization process often requires iterative experiments, where different media formulations and culture conditions are tested and evaluated. Various analytical techniques, such as high-performance liquid chromatography (HPLC) and enzyme-linked immunosorbent assay (ELISA), are used to quantify antibody production and assess product quality.

Scale-Up Strategies

Scale-up is the process of transitioning from small-scale laboratory cultures to large-scale production systems. It involves strategies to increase the culture volume and cell density while maintaining high antibody productivity and product quality.

There are different scale-up strategies employed in the production of monoclonal antibodies. One approach is to increase the culture vessel size, such as by using stacked cell culture flasks or disposable bioreactors. Another strategy involves optimizing culture conditions, such as oxygen and nutrient supply, pH control and agitation or mixing.

Process automation and control systems are implemented to ensure reproducibility and consistency during scale-up. Monitoring and control parameters, such as temperature, pH, dissolved oxygen levels and nutrient concentration, are closely regulated to maintain optimal conditions for cell growth and antibody production.

Scale-up also involves downstream processing considerations, including harvest and purification techniques, to efficiently recover and purify the monoclonal antibodies from the culture medium.

Purification methods for monoclonal antibodies

Purification methods play a crucial role in the manufacturing process of monoclonal antibodies (mAbs) to ensure the production of pure, high-quality antibodies. These methods are designed to separate the target mAb from other impurities present in the cell culture supernatant or crude antibody mixture. By employing various purification techniques, such as affinity chromatography, ion exchange chromatography, hydrophobic interaction chromatography and size exclusion chromatography, the desired mAb can be isolated and purified to a high degree.

Affinity Chromatography

Affinity chromatography is a powerful technique used for the purification of mAbs due to its high specificity and selectivity. This method relies on the specific interaction between the mAb and a ligand immobilized on a solid support. The ligand used is typically a protein or peptide that specifically binds to the target mAb. The most common ligand used is Protein A or Protein G, which selectively binds to the Fc region of immunoglobulins, including mAbs. The cell culture supernatant or the crude antibody mixture is passed through a column packed with the affinity resin, allowing the target mAb to bind while impurities pass through. Subsequently, the column is washed to remove non-specifically bound impurities, and the bound mAb is eluted using a buffer that disrupts the interaction between the ligand and mAb. Affinity chromatography can achieve high purification levels and is often used as the initial step in mAb purification.

Ion Exchange Chromatography

Ion exchange chromatography is another widely employed method in the purification of mAbs. It relies on the charge differences between the mAb and impurities for separation. The chromatographic matrix is typically composed of charged groups that interact with the oppositely charged molecules. For example, a cation exchange column with negatively charged groups can bind positively charged impurities while the negatively charged mAb passes through. Conversely, an anion exchange column with positively charged groups can retain negatively charged impurities while allowing the mAb to pass through. By optimizing the buffer conditions, the mAb can be selectively eluted from the column. Ion exchange chromatography can effectively remove charged impurities, such as host cell proteins, DNA and aggregates from the mAb solution.

Size Exclusion Chromatography

Size exclusion chromatography (SEC), also known as gel filtration chromatography, separates molecules based on their size. In mAb purification, SEC is commonly used as a polishing step to further purify the mAb and remove aggregates or fragments. The chromatographic matrix used in SEC contains porous beads that create a network of different-sized channels. Larger molecules, including aggregates, are excluded from entering the pores and elute earlier, while the smaller mAb molecules enter the pores and elute later, and the fragments elute after mAb molecules. This separation based on size allows for the removal of high molecular weight species and impurities, resulting in a purified and homogeneous mAb product.

Hydrophobic Interaction Chromatography

Hydrophobic interaction chromatography (HIC) is a purification method based on the differential hydrophobicity of molecules. The chromatographic matrix used in HIC contains ligands with hydrophobic groups. The mAb sample is loaded onto the column under high salt conditions, promoting hydrophobic interactions between the mAb and the stationary phase. The strength of the interaction can be modulated by adjusting the salt concentration or pH. Impurities with lower hydrophobicity elute first, while the mAb of interest is retained. Elution of the mAb is achieved by reducing the salt concentration or changing the pH, disrupting the hydrophobic interactions. HIC is particularly useful for the removal of non-specifically bound impurities, aggregates and variants.

Protein L Affinity Chromatography

Protein L affinity chromatography is an alternative to Protein A or Protein G affinity chromatography for mAb purification. It utilizes a bacterial protein called Protein L, which has a high affinity for the kappa light chain of immunoglobulins, including certain subclasses of mAbs. This method is specifically useful when the mAb does not bind well to Protein A or Protein G, such as with certain humanized or chimeric antibodies. The mAb sample is loaded onto a column containing immobilized Protein L, allowing the mAb to selectively bind. Non-specifically bound impurities are removed through washing, and the bound mAb is subsequently eluted using appropriate buffers.

Mixed-Mode Chromatography

Mixed-mode chromatography involves the use of chromatographic matrices with ligands that possess both hydrophobic and charged functional groups. This allows for the simultaneous exploitation of multiple modes of interaction, such as hydrophobic, ion exchange, or metal affinity interactions. Mixed-mode chromatography can be useful when conventional purification methods do not provide sufficient selectivity or resolution. It offers the flexibility to optimize conditions and select the most appropriate ligands to capture and purify the target mAb while minimizing impurities.

These purification methods, along with others not mentioned, can be employed individually or in combination to achieve the desired purity, potency and yield of monoclonal antibodies. The selection and optimization of purification methods depend on the specific characteristics of the mAb, impurities present, and the desired quality attributes of the final product. Effective purification ensures the production of high-quality mAbs suitable for therapeutic, diagnostic and research applications.

Quality Control

Quality control is a crucial aspect of the manufacturing process for monoclonal antibodies. It involves a series of procedures and tests designed to ensure that the final product meets the required quality standards and specifications. Quality control measures are implemented at various stages of the manufacturing process to identify and address any potential issues that could affect the safety, efficacy and consistency of the monoclonal antibodies.

Structural Analysis

Structural analysis is an essential quality control method employed during monoclonal antibody manufacturing. It involves examining the molecular structure of the antibodies to ensure their integrity and conformation. Various techniques are used for structural analysis, including X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry.

X-ray crystallography allows scientists to determine the three-dimensional structure of the monoclonal antibodies by analyzing the diffraction patterns produced when X-rays pass through the crystallized antibodies. This technique provides valuable insights into the precise arrangement of atoms and the overall conformation of the antibody.

Nuclear magnetic resonance spectroscopy (NMR) uses the magnetic properties of atomic nuclei to study the structure and dynamics of molecules. It can provide information about the folding, flexibility and interactions of the monoclonal antibodies.

Mass spectrometry is used to determine the mass and composition of the monoclonal antibodies. It helps in identifying any modifications or variations in the protein sequence, such as glycosylation or amino acid substitutions, which can impact the antibody’s functionality and stability.

By performing structural analysis, manufacturers can ensure that the monoclonal antibodies have the desired structure, which is crucial for their binding specificity and therapeutic activity.

Functional Assays

Functional assays are another important quality control method used in monoclonal antibody manufacturing. These assays assess the biological activity and functional characteristics of the antibodies, ensuring that they perform their intended therapeutic functions effectively.

Functional assays involve testing the antibodies for specific activities, such as binding to their target antigens, blocking receptor-ligand interactions or inducing immune responses. These assays may include techniques like enzyme-linked immunosorbent assay (ELISA), flow cytometry, and cell-based assays. ELISA is a widely used method for measuring the binding affinity and specificity of antibodies. It involves immobilizing the target antigen on a solid surface and detecting the binding of the antibodies using colorimetric or fluorescent signals.

Flow cytometry is used to analyze and quantify the interactions between antibodies and cells. It can provide information about cell surface markers, antibody binding and cellular functions.

Cell-based assays involve testing the antibodies in relevant biological systems, such as cell lines or primary cells. These assays evaluate the functional effects of the antibodies, including their ability to activate or inhibit specific cellular pathways or immune responses.

Functional assays ensure that the monoclonal antibodies are biologically active and capable of exerting their intended therapeutic effects. They play a crucial role in verifying the potency, specificity and functionality of the antibodies before they are released for clinical use.

Comparability Studies

Comparability studies are conducted during monoclonal antibody manufacturing to assess any changes or modifications in the manufacturing process and evaluate their impact on the final product’s quality and performance. These studies are particularly important when making changes to critical process parameters, manufacturing sites or scaling up production.

Comparability studies involve comparing the product manufactured using the modified process or under different conditions with the reference product manufactured using the established process. They include comprehensive testing and analysis to ensure that the modified product is equivalent or comparable to the reference product in terms of quality, safety and efficacy.

Impurity Testing

Impurity testing is a critical quality control method used to detect and quantify impurities present in monoclonal antibody products. Impurities can arise from various sources, including the manufacturing process, raw materials and degradation of the antibody itself. These impurities can impact the safety, efficacy and stability of the product.

Impurity testing involves the identification and quantification of impurities using analytical techniques such as chromatography (e.g., high-performance liquid chromatography, or HPLC), electrophoresis, spectroscopy and mass spectrometry. These methods can separate and detect impurities based on their physical and chemical properties.

Common impurities tested in monoclonal antibodies include host cell proteins, DNA, aggregates, variants, residual solvents and process-related substances. By monitoring and controlling the levels of impurities, manufacturers can ensure the safety and quality of the monoclonal antibody product.

Batch-to-Batch Consistency Testing

Batch-to-batch consistency testing is performed to ensure that each production batch of monoclonal antibodies meets the established quality specifications and is consistent with previous batches. This testing aims to identify any variations or deviations in the product attributes that could impact its safety, efficacy or quality.

Batch-to-batch consistency testing involves comparing critical quality attributes of the monoclonal antibody, such as purity, potency and structural characteristics, across different production lots. These attributes are assessed using appropriate analytical techniques and assays to determine if the batches are consistent and within acceptable limits. This testing provides confidence in the reproducibility and reliability of the manufacturing process, ensuring that each batch of monoclonal antibodies maintains consistent quality standards and performance.

Sterility Testing

Sterility testing is conducted to ensure that the monoclonal antibody product is free from viable microorganisms, such as bacteria, yeast and fungi. It is a crucial quality control measure to prevent the risk of contamination and ensure the safety of the product.

Sterility testing is typically performed using compendial methods, such as membrane filtration or direct inoculation, in which the product is incubated under appropriate conditions to promote the growth of any potential microorganisms. The absence of microbial growth after the incubation period indicates the sterility of the product.

Sterility testing is conducted on both the final product and the components used in the manufacturing process, including raw materials, containers and closures. It ensures that the monoclonal antibodies are free from microbiological contamination, reducing the risk of adverse effects when administered to patients.

By implementing comprehensive quality control measures, including structural analysis, functional assays, impurity testing, batch-to-batch consistency testing and sterility testing, manufacturers can ensure the safety, efficacy and quality of monoclonal antibody products. These quality control methods provide assurance to healthcare professionals and patients and contribute to the overall success of therapeutic interventions using monoclonal antibodies.

Challenges in monoclonal antibody manufacturing processes

Monoclonal antibody (mAb) manufacturing processes face several challenges that can impact the quality, efficacy and safety of the final product. The three challenges mentioned in the H3 headings are heterogeneity issues, aggregation and degradation problems, and contamination risks. Let’s elaborate on each of these challenges: 

Heterogeneity Issues

Heterogeneity refers to the presence of variability within a population of monoclonal antibodies. It can arise at different stages of the manufacturing process, including cell line development, cell culture and purification. Heterogeneity can manifest as differences in glycosylation patterns, charge variants, amino acid sequence variants or post-translational modifications.

These heterogeneity issues can affect the stability, pharmacokinetics and immunogenicity of the monoclonal antibody. They can also impact the batch-to-batch consistency and regulatory compliance. Managing heterogeneity requires robust process control, analytical techniques and optimization strategies to minimize variations and ensure consistent product quality.

Aggregation and Degradation Problems

Aggregation and degradation are common challenges in monoclonal antibody manufacturing processes. Aggregation refers to the clustering of mAb molecules, resulting in the formation of dimers, trimers or larger aggregates. Degradation, on the other hand, involves the breakdown of the mAb structure, leading to the formation of fragments or loss of activity.

Aggregates and degraded forms of monoclonal antibodies can compromise their therapeutic efficacy and safety. They may induce unwanted immune responses, alter pharmacokinetics or reduce the binding affinity to the target molecule. Manufacturing conditions, such as high protein concentrations, low or high pH, and temperature fluctuations, can promote aggregation and degradation. Implementing appropriate formulation strategies, process optimizations and analytical methods can help mitigate these issues.

Contamination Risks

Contamination risks pose a significant challenge in monoclonal antibody manufacturing processes. Contaminants can originate from various sources, including raw materials, equipment, personnel and the environment. Common contaminants include microbial contaminants (e.g., bacteria, fungi, viruses), adventitious agents (e.g., viruses from cell lines), process-related impurities (e.g., host cell proteins, DNA) and endotoxins.

Contaminants can compromise the safety, efficacy and quality of monoclonal antibody products. They may introduce potential health risks to patients or affect the stability and function of the therapeutic antibodies. Implementing robust quality control measures, adhering to Good Manufacturing Practices (GMP) and employing stringent purification techniques are crucial to minimize contamination risks and ensure product safety.

Scalability and Cost-Effectiveness

Scaling up the manufacturing process of monoclonal antibodies from small-scale research or clinical production to large-scale commercial production can present challenges. The process must be scalable to meet the increased demand while maintaining product quality, consistency and cost-effectiveness.

Scaling up requires optimization of various parameters, such as cell culture conditions, media formulation, purification methods and equipment. Ensuring consistent and reproducible performance at larger volumes can be complex due to factors such as mass transfer limitations, increased shear stress and process dynamics.

Regulatory Compliance

The manufacturing of monoclonal antibodies is subject to stringent regulatory requirements to ensure patient safety and product efficacy. Compliance with regulatory guidelines, such as those provided by regulatory authorities like the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), is essential throughout the manufacturing process.

Meeting regulatory requirements involves comprehensive documentation, adherence to GMP and validation of critical processes and analytical methods. Ensuring batch-to-batch consistency, robust quality control measures and traceability of raw materials are essential components of regulatory compliance.

Time and Cost Efficiency

mAb manufacturing processes often involve multiple steps, complex equipment and extended timelines. The lengthy and resource-intensive nature of the process can impact time to market and increase manufacturing costs.

Reducing manufacturing process time and increasing process efficiency are critical for timely availability of monoclonal antibody therapies and cost-effectiveness. Process optimization, automation and the use of advanced technologies can help streamline operations, minimize cycle times and reduce costs.

Addressing these challenges requires continuous innovation, collaboration and investment in research and development to develop novel manufacturing technologies, process improvements and analytical tools for robust and cost-effective production of monoclonal antibodies.

Advancements in the monoclonal antibody production process

Advancements in the monoclonal antibody production process, particularly in CHO cell-based production, hold significant promise for the future. These advancements aim to improve the efficiency, scalability and cost-effectiveness of producing monoclonal antibodies, thereby addressing the growing demand for these therapeutic molecules.

One area of advancement is the development of novel cell culture media and supplements that enhance cell growth and productivity. Researchers are continuously optimizing the formulation of these media to provide the necessary nutrients and growth factors for CHO cells, resulting in higher antibody yields. Another promising avenue is the implementation of advanced bioreactor technologies. Bioreactors equipped with sensors and control systems enable real-time monitoring and adjustment of critical process parameters such as pH, temperature, dissolved oxygen levels and nutrient availability.

To sum up, future advancements in monoclonal antibody production will focus on optimizing cell culture media, implementing advanced bioreactor technologies and utilizing genetic engineering techniques to enhance CHO cell-based production. These advancements hold great potential for increasing efficiency, scalability and cost-effectiveness, ultimately facilitating the availability of monoclonal antibodies for various therapeutic applications.

If you want to learn more about bioprocessing for mAbs, download the invaluable resources by Avantor or order the products from our ecommerce platform. Their expertise in the field, combined with a wide range of products and services, can assist you in achieving efficient and reliable production of monoclonal antibodies. Take advantage of their comprehensive solutions and collaborate with their knowledgeable teams to enhance your bioprocessing capabilities and accelerate your research and development efforts.