Industrial Biotechnology 2022

 

Scaling AAV Production: Easing the Transition from Laboratory Scales to Commercial Manufacturing

Aden associated virus (AAV) has emerged as the leading vector for gene therapy delivery. Compared with options such as lent virus and adenovirus, AAV exhibits a strong safety profile because it has low pathogenicity and requires a helper virus to replicate. AAV is also capable of long-term gene expression, and it can infect both dividing and no dividing cells (1–5). Developers of advanced therapies have found such advantages to be quite attractive. As of January 2021, two gene therapy products have gained US Food and Drug Administration (FDA) approval: Luxturna (voretigene neparvovec) from Spark Therapeutics and Zolgensma (onasemnogene abeparvovec) from AveXis/Novartis. Both products use AAV vectors (serotypes 2 and 9, respectively), as do many candidate therapies moving through clinical studies. Cost-effective AAV manufacturing remains elusive, however. Yields and expression titers from upstream processes continue to be low — an acute problem for a modality that requires doses with high vector concentrations.

Our Conversation
How would you describe Takeda’s typical scale-up process for AAV vector production? We use a proprietary production toolbox to generate our AAV-based gene therapies for clinical trials. It is based on a transient transfection platform, using a traditional three-plasmid approach and a proprietary suspension-adapted human embryonic kidney (HEK293) cell line cultured in chemically defined, protein- and serum-free media. We believe that this system gives us the highest flexibility in producing different AAV vector variants for use in animal models and preclinical studies to identify lead candidates for clinical trials. We have scaled up the entire toolbox from 500-L to 1,000-L scale. Currently, we are using a good manufacturing practice (GMP) facility with 1,000-L capacity to produce clinical trial material (CTM). The facility is equipped for completely single-use operations. Based on our current knowledge, we will use the same facility for commercial manufacturing.

What aspects of AAV production are most difficult to perform at commercial scales? It is well known that certain parameters for some process steps cannot be scaled up linearly — e.g., stirrer speed in a bioreactor. Those are the most difficult parameters to scale. However, an experienced team that knows the underlying physical principles and calculations should not encounter any problems in scaling them.


How can such concerns be minimized during scale-up? Two important considerations can facilitate scale-up, the most important of which is to identify process parameters that will have critical impacts on product quantity and/or quality. Depending on the experience level of a process development team, that task can be quite challenging. For a typical upstream process, critical attributes can include seeding density, pH, partial oxygen pressure (pO2), agitation rate, and maximum cell density before culture split.

During scale-up, quality-critical parameters must be kept within specified ranges such that the quality and quantity of vectors produced at full scale are comparable to those obtained at pilot scale, which is the scale used to produce material for animal studies.

What expression systems are available for AAV production, and how do those systems influence process scalability? Four main platforms are used in the gene therapy industry:

• Transient transfection systems using HEK293 cells (both adherent- and suspension-culture formats are available).
• Stable packaging/producer cell lines (e.g., HeLa cells).
• Herpes simplex virus (HSV) systems.
• Baculovirus expression vector systems (BEVS) using Spodoptera frugiperda (Sf9) cells.

 

How do different cell culture formats influence process scalability? So far as I know, all of the AAV production platforms I mentioned that depend on suspension cell culture (e.g., Sf9 and HEK293 cells) have been brought to scales up to 2,000 L in single-use stirred-tanked reactors.

How much attention should developers give to scale-up during early (pre)clinical phases, and what considerations are most important at that stage? Takeda’s gene therapy process development team works closely with the therapy developer or research team that has designed the vector (capsid and transgene) being produced. Close collaboration helps to align timelines and demands. We on the chemistry, manufacturing, and controls (CMC) team also are involved in selecting a lead candidate, taking into account knowledge gained during preparation of preclinical material.

What else could improve AAV scale-up? The biopharmaceutical industry needs a mixture of new technologies and improved understanding. There is need for excellently educated personnel who have a high level of biological understanding in combination with profound process knowledge; such qualities are key prerequisites for working in biotechnology. And the more experience that a team has gained developing new processes, optimizing existing ones, and troubleshooting GMP operations, then the faster and more effectively it will complete new projects.

What stages of manufacturing-scale AAV production can be optimized most? When and how would you begin to make such adjustments? When a new project enters process development, it is important to balance speed, output, and quality. Some process adjustments will need to be made; otherwise, you might be unable to produce enough material for preclinical or clinical studies, or product quality might not meet specifications. During these first mandatory optimizations, if time permits, further adjustments can be made to increase yield.

The farther that a program moves toward product launch, the less that regulatory authority will welcome process adjustments. Nevertheless, it might still be necessary to carry out further optimizations in phase 3 clinical studies. Around the time of process performance qualification, a process clearly should be optimized, and no further changes should be made.

What other advice do you have for scientists who are seeking to maximize the scalability of their AAV processes — and ultimately, their yields? Just referring to our AAV process toolbox here at Takeda, and thinking only about yield improvement, my first thought, of course, goes to upstream production. It is always tempting and interesting to fine-tune setpoints or to try adding media. However, downstream activities also must be considered. Depending on the vector construct, very low recovery rates might be observed for one or the other purification step. Steps exhibiting the greatest loss of capsids are the low-hanging fruits with the highest probability of yield improvement. Those should be optimized first.

 

Source URL: https://bit.ly/3Q4hkSt

Conference Name: 27th World Congress on Industrial Biotechnology   

Date: November 16-17, 2022     

Venue: Tokyo, japan

URL:  https://bit.ly/3zA6nAP

Whtsapp Number: +44-7360-516157

 




Comments

Popular posts from this blog

Industrial Biotechnology Conference 2022

Industrial Biotech Conference 2022

Molecular Biology Conference