Article
April 29, 2025

Ensuring a Smooth Transition: Tech Transfer Strategies for Monoclonal Antibody Drug Products in Late-Phase Clinical and Commercial Launch

Author

Shawn Cain

Read Bio

The successful transfer of a monoclonal antibody (mAb) drug product from late-stage clinical development to commercial-scale manufacturing is a pivotal milestone in biopharmaceutical development.

Technology transfer (tech transfer) involves the systematic movement of development activities, manufacturing processes, analytical methods, and quality control strategies from a sending site, typically a development or clinical-scale facility, to a receiving site, which is often a large scale or commercial production facility. For Contract Development and Manufacturing Organisations (CDMOs), tech transfer is not just a process of replicating manufacturing steps at a different facility; it is a complex, multidisciplinary effort that ensures process robustness, scalability, regulatory compliance, and supply chain readiness.

At the late-stage clinical phase, the goal shifts from producing limited quantities for clinical trials to establishing a scalable, reproducible, and commercially viable process capable of producing consistent, high-quality batches. This transition requires a strategic approach to process knowledge transfer, analytical method transfer and validation, facility fit assessment, supply chain logistics, and regulatory alignment. Additionally, the challenges of scaling up a mAb process are significant due to their structural heterogeneity, immunogenicity risks, specific formulation requirements, and involve considerations such as purification efficiencies, formulation stability, and aseptic fill-finish operations.

CDMOs must also anticipate regulatory expectations to ensure compliance with global health authorities such as the FDA, EMA, and PMDA. Each jurisdiction has specific regulatory frameworks regarding process validation, comparability studies, and quality assurance – all of which must be thoroughly addressed to support a seamless transition to commercial production.

The successful technology transfer of a monoclonal antibody (mAb) drug product from late-stage clinical development to commercial-scale manufacturing is pivotal.

Establishing a Robust Technology Transfer Strategy

A comprehensive technology transfer strategy begins with defining the scope of the transfer, identifying potential risks, and establishing clear communication channels between the sending and receiving sites. Understanding and proactively addressing the key factors that influence a successful tech transfer can mitigate risks, reduce delays, and ensure a smooth transition to commercial production.

A well-structured technology transfer strategy is essential to ensure process consistency and regulatory compliance. As a CDMO, the tech transfer process is typically initiated with a detailed assessment of the current process, product knowledge and historical data from the sending site, including process performance data, batch records, analytical method history, known product sensitivities (light, oxygen, temperature, and other degradation pathways), together with prior deviations or optimisations.

At the core of the strategy, is a knowledge transfer framework that ensures the intricacies of the mAb production process including all critical process parameters (CPPs) and critical quality attributes (CQAs) are well understood and reproducible at the receiving site. This requires extensive collaboration between the sponsor, sending sites, development teams and receiving sites technical experts in process development, manufacturing sciences, analytical development, quality assurance, and regulatory affairs.

Additionally, the tech transfer plan must define clear acceptance criteria, validation protocols, and comparability requirements to demonstrate that product quality remains unchanged throughout the transfer and potential scale up activities. This often involves generating bridging data between clinical-scale and commercial-scale batches to provide regulatory agencies with assurance that the drug product maintains its safety, efficacy, and stability.

Scaling Up the Manufacturing Process

The transition from clinical-scale production to commercial manufacturing presents several challenges, particularly in downstream purification at the bulk substance stage, and final drug product formulation. The ability to maintain consistency, optimise yield, and ensure process robustness and product homogeneity at larger scales is critical to a successful commercial launch.

Bulk Drug Substance (BDS): Maintaining Yield and Purity at Scale

The purification of monoclonal antibodies usually involves multiple chromatography steps, including Protein A affinity chromatography, ion exchange chromatography, and ultrafiltration/ diafiltration. Each step must be carefully scaled to maintain high recovery rates, impurity clearance, and viral inactivation, without introducing process variability.

During scale-up, one of the main challenges is column packing consistency and resin lifetime management. Ensuring that chromatography resin performance remains consistent across multiple purification cycles is critical to maintaining product consistency and quality. Additionally, buffer preparation and hold times need to be optimised to prevent aggregation, oxidation, or degradation.

To further enhance process robustness, high-throughput process development (HTPD) strategies can be implemented to allow for rapid screening of process conditions and provide a data-driven approach to process optimisation. This enables efficient process scale-up while ensuring that CQAs remain within the established specifications. Attention should also be paid to the target scale and what will be transferred to the formulation and fill-finish process. For instance, should the BDS batch size be specific to the fill-finish batch size or will BDS batches be pooled to the desired fill-finish lot size?

Formulation and Fill-finish: Ensuring Stability and Sterility

The final formulation of the monoclonal antibody must preserve stability, potency, and sterility at large-scale commercial manufacturing conditions. Factors such as buffer composition and capacity, pH control, excipient compatibility, container closure compatibility, and protein concentration play a crucial role in ensuring long-term stability, especially in a liquid presentation.

The fill-finish stage presents unique challenges, particularly in terms of sterility assurance, container-closure integrity (CCI), and particulate contamination control. Over the years, advancements in technology and process control have significantly improved sterility assurance, product quality, and manufacturing efficiency. Innovations such as single-use systems, coated vials and stoppers, closed barrier technologies, and automation have reshaped sterile drug manufacturing, with isolators and robotics emerging as preferred solutions for contamination-free aseptic processing.

Regulatory expectations for sterile drug product manufacturing have also evolved around the globe, particularly with the latest revisions of EU GMP Annex 1, which emphasises enhanced contamination control strategies and risk-based sterility assurance approaches. Adhering to these guidelines ensures that mAb drug products meet the highest sterility and quality standards.

Analytical Method Transfer and Validation

A critical aspect of tech transfer is the successful transfer and validation of analytical methods used for acceptance of incoming Drug Substance, in-process control, release testing, and stability assessment. Analytical method transfer requires ensuring that critical assays such as high-performance liquid chromatography (HPLC), capillary electrophoresis, mass spectrometry, and bioassays used to measure identity, purity, potency, and impurity levels remain robust and reproducible at the receiving site. This is a key focus of all tech transfers.

To facilitate a smooth method transfer, a comparative testing approach is employed, wherein the sending and receiving sites perform side-by-side testing using the same reference standards and control materials. For complex assays, additional co-validation or revalidation studies may be required to demonstrate assay robustness and accuracy.

Regulatory expectations for analytical method validation are guided by ICH Q2 (R1) and ICH Q6B, which require demonstrating assay specificity, accuracy, precision, linearity, and robustness. Ensuring compliance with these guidelines is essential for regulatory approval and long-term product quality assurance.

Regulatory Compliance and Post-approval Considerations

Regulatory agencies such as the FDA, EMA, and PMDA require a well-documented tech transfer process to ensure that product quality is maintained at commercial scale. As part of the transfer, the CDMO should work closely with the sponsor company and support regulatory filing updates to reflect process changes, site changes, and new validation data. This includes updates to the Investigational New Drug (IND) application, Biologics License Application (BLA), or Marketing Authorisation Application (MAA). Sponsors should understand the regulatory standing of the receiving site and ensure their Quality systems will meet the regulatory requirements of the regions they plan to apply for authorisation.

Ensuring cGMP compliance at the receiving CDMO commercial facility is critical, as regulatory agencies expect comprehensive process validation, batch release data, and ongoing post-market surveillance to maintain product integrity. Additionally, change control strategies must be in place to address any future process optimisations, changes in excipients, materials or container closures, or site modifications.

Risk Management and Contingency Planning to Secure Supply Chains

The complexity of mAb tech transfer requires proactive risk management to anticipate and address potential challenges. Process failure mode analysis (FMEA) is used to systematically identify high-risk areas that could impact product quality, yield, or regulatory compliance. By developing robust risk mitigation strategies, such as additional process monitoring, operator training programmes, dual sourcing strategies for critical raw materials and redundancy in equipment and facilities, manufacturers can minimise the likelihood of disruptions.

This risk-based approach allows for targeted process adjustments, and together with technical support from subject matter experts ensures that potential challenges are addressed before they impact largescale production. In addition, disaster recovery plans should also be put in place to ensure business continuity, in the event of supply chain disruptions or facility related incidents to secure supply chains and the delivery of life changing therapies to patients.

Conclusion

The technology transfer of a monoclonal antibody drug product for late-phase clinical and commercial launch is a complex and multifaceted process that requires meticulous planning, robust risk management, and cross functional collaboration. Ensuring process scalability, analytical method robustness, regulatory compliance, and supply chain stability is critical for a seamless transition to commercial production.

By leveraging advanced manufacturing technologies, real-time analytical monitoring, and digital process modelling, biopharmaceutical companies and their partnering CDMOs can enhance tech transfer efficiency and accelerate time-to-market for life changing monoclonal antibody therapies. With careful execution, CDMOs can successfully bridge the gap between clinical development and commercial supply, ensuring that patients receive consistent high-quality, safe, and effective biologic treatments.


We are committed to supporting clients at every stage of the manufacturing cycle, delivering best-in-class services efficiently and effectively.

Find out more about our fully integrated Packaging Services.


Our dedicated team of specialists will be happy to discuss any of your Contract Development needs. Contact us on +1 215-613-3600 (US) or +44 1495 711 222 (UK/EU) or email talkfuture@pci.com

Awards

We take pride in celebrating the accomplishments and accolades that distinguish PCI Pharma Services, our employees, and our services. These achievements are a testament to our commitment to excellence, innovation, and the unwavering dedication of our teams