Manufacturing Interleukin-2 For In Vivo Use: Key Considerations
By Carlos BaƱado, CEO, ARScience Bio

Recombinant human IL-2 (rH IL-2) in particular is widely used to enhance the efficacy of adoptive cell therapies, including TILs and natural killer (NK) cell therapies. I discussed development considerations for IL-2 in my first article.
By promoting cell expansion, survival, and functionality, interleukins help optimize therapeutic outcomes in cancer immunotherapy and regenerative medicine. However, their clinical application — especially in vivo use of rH IL-2 — requires stringent control over production, formulation, and stability to ensure safety and efficacy under GMP standards.
Regulatory Compliance And GMP Requirements
Manufacturing biologics under GMP conditions involves a rigorous, traceable documentation package that includes risk assessments throughout the development process, process performance qualification, and robust analytical methods to monitor the quality attributes of materials and products. Unlike small molecules, biologics such as rH IL-2 are highly sensitive to production conditions, making consistency a significant challenge. The FDA and EMA require that each batch meets strict specifications regarding identity, potency, purity and stability, necessitating advanced analytical methodologies and robust quality assurance programs to mitigate potential variability.
Regulatory compliance requires adherence to good documentation practices starting from master cell bank creation through all process development activities. As part of life cycle management, process validation includes upstream and downstream manufacturing steps, from cell culture and protein expression to purification and formulation. Ensuring batch-to-batch consistency demands tight control over raw materials, bioprocess parameters, environmental conditions, and final product testing. Moreover, GMP compliance extends beyond the manufacturing site to include suppliers and CDMOs, all of which must meet rigorous quality and documentation standards.
Fully characterized cell banking for bacterial products presents several challenges that can impact the consistency and quality of the final product, including genetic stability, viability and recovery post-thaw, contamination risk, and storage conditions.
Developing a robust manufacturing process for IL-2 is crucial for ensuring its efficacy and safety in clinical applications. Key considerations include the intended use of IL-2, which will drive formulation requirements. Formulation stability is paramount to maintain the integrity of IL-2 across its shelf life; this includes temperature control and the selection of suitable excipients. Additionally, dosing considerations can significantly impact product development, as IL-2 might be administered as a fixed single dose or adjusted based on patient weight. Such variability necessitates careful calibration of manufacturing processes to ensure consistent quality and compliance with cGMP standards. By addressing these factors early in the process development, manufacturers can streamline the transition to full-scale cGMP production while minimizing the risk of regulatory setbacks.
Transitioning to cGMP manufacturing for initial batches of IL-2 is a pivotal step in preparing for clinical testing, including Phase 1 and 2 trials. Key considerations during this phase involve determining batch sizes based on anticipated clinical needs, which requires careful planning of trial participant numbers and dosing regimens. Additionally, manufacturers must forecast potential commercial sales volumes to ensure scalability post-approval. This includes evaluating in-house production capabilities versus the availability and capacity of CDMOs. A thorough analysis is necessary to weigh the trade-offs between production costs and the risks associated with scaling up too quickly. Increasing batch sizes may lead to economies of scale, but it also intensifies the potential for batch failures or regulatory noncompliance, which can have significant financial implications. Therefore, a strategic approach that anticipates market conditions and regulatory landscapes is essential for successful cGMP manufacturing transitions.
Complexity Of Physicochemical Characterization
One of the most critical and technically demanding aspects of biologic development is the comprehensive physicochemical characterization of the molecule. rH IL-2, like other cytokines and recombinant proteins, exhibits a complex tertiary structure, potential isoforms, and post-translational modifications — such as glycosylation or oxidation — that can influence its biological activity, stability, and immunogenicity. A robust analytical strategy is essential not only for regulatory approval but also for ensuring product consistency and long-term viability.
Characterization typically involves a suite of high-resolution and orthogonal analytical techniques, including mass spectrometry, high-performance liquid chromatography, capillary electrophoresis, and cell-based bioassays, to confirm identity, purity, potency, and structural integrity. In particular, the propensity of IL-2 to aggregate or degrade under certain conditions poses a significant challenge, requiring tight control over formulation, process conditions, and storage parameters. To meet evolving regulatory expectations — particularly from agencies like the FDA and EMA — developers often rely on specialized GMP-compliant laboratories and contract testing organizations with deep expertise in cytokine analytics. These partnerships are instrumental in navigating the intricate landscape of biologic characterization, enabling the generation of comprehensive data packages that support clinical development, marketing authorization, and life cycle management.
Aseptic Manufacturing And Fill/Finish Challenges
The final stages of biologic manufacturing, particularly aseptic fill/finish, pose significant technical and regulatory challenges. Ensuring sterility throughout the process is critical to patient safety, as biologics cannot be terminally sterilized like traditional pharmaceuticals.
Aseptic filling requires state-of-the-art cleanroom environments, advanced filling technologies, and highly trained personnel to minimize contamination risks. Factors such as vial integrity, particulate control, and container-closure interactions must be meticulously monitored. The use of isolator technology and robotic filling systems can reduce human intervention and enhance sterility assurance but requires significant investment and validation. Additionally, ensuring product stability during the fill/finish process is essential. rH IL-2 is highly sensitive to shear forces, temperature fluctuations, and surface interactions, which can affect its potency and structural integrity.
Conclusion
Developing and manufacturing in vivo rH IL-2 present a distinctive array of challenges that span the entire product life cycle. These include stringent adherence to GMP, intricate physicochemical characterization due to IL-2’s structural complexity, and the need for robust aseptic processing and cold chain logistics to maintain product stability and bioactivity. Moreover, variability in biological activity, sensitivity to aggregation, and potential immunogenicity further complicate development. Overcoming these hurdles demands deep expertise in biologics manufacturing, early engagement with regulatory authorities, and strategic partnerships with CDMOs experienced in cytokine and cell therapy production. As interleukin-based therapies — including IL-2 variants and engineered analogs — continue to evolve as critical components of in vivo and ex vivo cell and gene therapy protocols, investment in scalable, reproducible, and regulatory-compliant manufacturing platforms will be key. Streamlining these processes not only accelerates time to the clinic but also ensures broader patient access to these transformative immunotherapies.
About The Author:
Carlos Bañado is CEO and board member of ARScience Biotherapeutics. Previously, he served as founder CEO of mAbxience, a biotech company focused on development, manufacturing, and commercialization of biosimilar monoclonal antibodies. Bañado is a pharmaceutical entrepreneur, being partner at Laboratorios Copahue SA, a pharmaceutical company focused on dermatology; board member and investor in Nanox Release Technologies, a nanotechnology company; and also worked with Celnova Pharma Inc., a pharmaceutical company specialized in complex therapeutic classes. He has served on the board of several companies and has broad international experience.