Life Sciences
Navigating the Complex Landscape of GMP-Compliant Exosome Manufacturing: Challenges, Regulatory Insights, and Future Directions

By Davide Zocco, Commercial Development Head for Exosomes and mRNA Technologies, Specialized Modalities, Lonza

Scaling GMP-compliant manufacturing for exosome-based therapies presents unique challenges compared to traditional biologics. Could you elaborate on the key hurdles Lonza encounters in this process, particularly regarding standardization and reproducibility? Furthermore, what are the most critical regulatory considerations that developers should prioritize when navigating the approval pathway for exosome therapeutics globally?

Exosomes are heterogeneous nanoparticles that carry a plethora of biomolecules, such as proteins, small RNAs, DNA, and lipids. Their unique ability to cross the blood-brain barrier makes them attractive vectors for delivering therapeutic payloads, such as proteins and nucleic acids, to target specific tissues. If these capabilities are fully realized, the potential applications of exosomes could be vast, spanning disease treatment, immune modulation, and tissue regeneration.

Despite their potential, manufacturing standards for exosome-based therapies are still in the early stages. One key hurdle manufacturers may face in building a Good Manufacturing Practices (GMP)-compliant process for exosome production is the limited understanding of how exosome characteristics influence their biological functions. This knowledge gap can hinder the development of scalable methods for isolating and purifying specific exosome populations from other extracellular vesicles and cellular debris.

Additionally, during manufacturing, specific exosome subpopulations may be selected or excluded, affecting the critical quality attributes of the drug substance. Processes that rely on primary cells (e.g., mesenchymal stem cells [MSCs]) may be difficult to standardize as the exosome cargo may vary from one cell batch to the other. Furthermore, the lack of standardized analytics for exosome therapeutics hinders process development and product approval.

Establishing GMP-compliant processes is an essential part of regulatory strategy and should be addressed in the earliest stages of development. When developing these processes, it is important to focus on defining a product’s critical quality attributes (CQAs), supported by qualified analytics, to enable the manufacture of a product that is safe and, secondly, effective.

Drug developers can collaborate with strategic partners, such as CDMOs, who maintain knowledge of the latest regulatory guidance and apply diverse expertise to commercialize exosome therapies. This helps ensure these investigational candidates are advanced through the clinical pipeline minimizing the risk of delays. 

Exosomes are inherently complex. How does Lonza ensure robust characterization and quality control throughout the manufacturing process, given the variability associated with these vesicles? What specific analytical techniques are proving most effective in ensuring product consistency and safety?

Exosomes are much more complex and heterogeneous compared to antibodies or small molecules. They carry a plethora of macromolecules such as proteins, nucleic acids, and lipids, and their molecular content varies depending on the cell source. Consequently, characterization and control are essential steps in exosome manufacturing to ensure a reproducible, high-quality product.

At Lonza, we provide a broad range of extracellular vesicle (EV) characterization and development services, as well as GMP manufacturing services, for adherent and non-adherent cells, including HEK293, MSCs, fibroblasts, and other cells.

Additionally, we have developed a characterization toolbox that addresses exosomes attributes such as biophysical properties (size, concentration, zeta potential, integrity), molecular properties such as surface biomarkers and cargo (e.g., protein and RNA content) and bioactivity (e.g., cell-based assays). Through this toolbox, we leverage nanoflow cytometry to analyze the biophysical, molecular, and functional attributes of exosomes. This helps overcome the “swarm effect” in standard flow cytometry, enabling quantitative assessment of exosomes at the single-particle level.

Moreover, technologies such as single particle analysis and omics platforms such as mass spectrometry and next-generation sequencing enable quantitative assessment of exosome integrity and molecular cargo, informing about product quality and consistency. Other technologies, such as size exclusion chromatography using ultra-high performance liquid chromatography (SEC-UHPLC) and residual assays, provide insights on process-related impurities and contaminants that must be strictly controlled to help ensure product safety.

By integrating these advanced analytical methods in one end-to-end offering, we aim to ensure robust characterization and quality control throughout the exosome manufacturing process.

Collaborations between biotech companies like Exogenus Therapeutics and CDMOs like Lonza are pivotal in accelerating the development of exosome-based therapies. From Lonza’s perspective, what are the key advantages for developers in collaborating with a CDMO that possesses specialized expertise in exosome manufacturing? How does Lonza support developers in navigating the complexities of process development and scale-up?

Collaborations between biotech companies and CDMOs like Lonza are crucial in accelerating the development of complex exosome-based therapies. For drug developers, this collaboration offers several key advantages, including leveraging specialized expertise and cutting-edge technologies.

At Lonza specifically, drug developers benefit from our end-to-end exosome manufacturing capabilities, which can significantly de-risk the path to IND submission, clinical development, and commercialization. By leveraging Lonza’s process development expertise, including access to exosome loading technologies and bioactivity assays, both early- and late-stage developers can navigate the complexities of process development and scale up with confidence.

Furthermore, Lonza has developed an innovative standard platform to enable large-scale perfusion-based production of GMP exosomes from HEK293 cells. Through this platform, drug developers can access perfusion-based upstream processes, chromatography-based downstream processes, and proprietary formulation techniques.

Drug developers seeking to manufacture exosomes can leverage our New Product Introduction (NPI) system to design bespoke processes. This NPI system helps companies establish Current Good Manufacturing Practices (cGMP)-compliant manufacturing standards customized for cell and gene therapies. The system also enables oversight throughout a product’s lifecycle, including validation for process robustness, raw materials, analytical methods, sterility, facility, equipment, and tissue acquisition.

In addition to new technologies, CDMOs can offer regulatory guidance for drug developers to align their exosome production with evolving global standards. In this area, CDMOs can help developers navigate process validation, critical quality attributes, and compliance with GMP standards. This helps facilitate a smoother path to clinical and commercial success, especially for emerging modalities like exosomes.

Exosome therapies are increasingly being explored as a means to enhance cell and gene therapies. In your view, how are exosomes revolutionizing these fields, and what are the most promising applications you foresee in the near future?

The exosome therapy field today holds the kind of promise that antibody and protein-based therapeutics did years ago. Exosomes promise to offset, at least partially, some of the shortcomings observed with other cell and gene delivery platforms such as lipid nanoparticles (LNPs) and viral vectors. Specifically, initial clinical studies suggest that exosomes don’t present certain safety risks, including no reports of liver toxicity or adverse inflammatory events, enabling drug redosing and facilitating tissue-specific delivery and target engagement.

Beyond cell and gene indications, exosomes may become a promising platform for vaccine development and immunotherapy approaches, including for cancer, where their drug-loading capacity and tissue-specific targeting can help minimize off-target effects and improve patient outcomes. Additionally, exosomes are currently explored as natural carriers of regenerative molecules (e.g., growth factors), enabling applications in wound healing and tissue regeneration.

Earlier this year, Lonza and Exogenus Therapeutics announced an agreement to develop Exo-101, Exogenus’ exosome-based lead candidate. Could you provide insights into the specific aspects of this collaboration that highlight Lonza’s expertise in exosome development and manufacturing? What are the key milestones you are targeting in this collaboration?

Lonza is leveraging its process and manufacturing expertise, specifically at our site in Siena, Italy, to develop a scalable process for the GMP manufacturing of Exo-101, a complex secretome product derived from cultured umbilical cord blood (UCB) cells. Our team of scientists and state-of-the art facilities draw on extensive experience with cell and gene therapies, providing a key advantage for technology providers, such as Exogenus, who require a bespoke process from upstream to downstream.

Additionally, Lonza´s analytical characterization toolbox will enable Exogenus to identify and select key features of the product that will lead to the definition of its Critical Quality Attributes (CQAs) in GMP production. The manufacturing process has been fully assessed and a gap analysis performed to establish GMP manufacturability and scale. Following the initial feasibility and optimization studies, Lonza will perform confirmation runs with the optimized process to ensure preservation of Exo-101 CQAs and bioefficacy leading to GMP manufacturing. Lonza is very excited to undertake this project with Exogenus Therapeutics and to bring Exo-101 to clinical supply as efficiently as possible.

Looking ahead, what are the critical advancements in exosome manufacturing technologies and regulatory frameworks that you believe will be essential to unlock the full potential of exosome-based therapeutics?

Exosome manufacturers and regulatory agencies must work together, starting in the earliest stages of process development, to ensure alignment on expected critical quality attributes linked to product safety and potency.

On the manufacturing side, we need continued innovation in processes, such as scalable, high-yield production systems that can meet commercial demand while maintaining product integrity. Automated purification workflows and engineered exosome platforms will be crucial for optimizing therapeutic delivery.

From a regulatory perspective, clarity is essential. The industry needs standardized guidelines for exosome characterization, potency, and safety testing. Right now, different regulatory agencies have slightly different expectations, so harmonizing these frameworks will be essential to accelerating approval timelines and making exosome therapies widely available.