1 / 18

Integrated Continuous B iomanufacturing: Quality and Regulatory C onsiderations

Integrated Continuous B iomanufacturing: Quality and Regulatory C onsiderations. Chantal Cazeault Executive Director, Product Quality Integrated Continuous Biomanufacturing, Castelldefels, Spain October 20-24, 2013. Presentation Outline. Drivers for Innovation.

matt
Télécharger la présentation

Integrated Continuous B iomanufacturing: Quality and Regulatory C onsiderations

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Integrated Continuous Biomanufacturing: Quality and Regulatory Considerations Chantal CazeaultExecutive Director, Product Quality Integrated Continuous Biomanufacturing, Castelldefels, Spain October 20-24, 2013

  2. Presentation Outline • Drivers for Innovation • Enablers of Continuous Manufacturing • Integrated Continuous Manufacturing: Key Quality and Regulatory Considerations • Continuous Manufacturing and Single-use Technologies: Opportunities and Challenges • Conclusion .

  3. Innovation in Manufacturing: What are the Drivers from a Regulator’s Perspective? • Speed to clinic/market, especially in areas of unmet medical needs • Cost of drugs • Manufacturing flexibility (maintaining production capacity) • Increased focus on Product Quality and Quality by Design/PAT • e.g. FDA’s 2011 Strategic Plan Overarching goal is patient safety, uninterrupted drug supply and lowering drug costs.

  4. Enablers of Continuous Manufacturing • Increased use of disposable, single-use technologies • Bioreactors • Ready-to-use chromatography columns • Viral filters • TFF • Increased titers using improved media formulations, feed strategies, higher producing clones, etc. • New recovery/purification methods being developed (moving bed and counter current chromatography) • Control system sophistication

  5. Enablers of Continuous Manufacturing • Innovation in real-time measurement and control technologies • Deeper process knowledge; just-in-time product disposition • Increased interest in continuous manufacturing process from beginning to end: less equipment, smaller portable equipment, fewer steps, faster cycle times • FDA looking at effects of continuous manufacturing on product quality (2011 Strategic Plan)

  6. Continuous Manufacturing: Key Quality and Regulatory Considerations • No guidance specific to continuous manufacturing • Strong encouragement from regulators to use QbD principles. Why? • Provides a more systematic approach to product development, based on sound science, Quality Risk Management, patient safety • Emphasizes product/process understanding and process control • It all comes down to understanding your product and establishing a commercial manufacturing process capable of consistently producing a product of the intended quality • How? • Identifying CQAs of the product • Identifying potential sources that lead to variability in the CQAs • Establishing a control strategy to ensure that the CQAs remain consistent and within acceptable limits over the lifecycle of the product

  7. Operational Concepts will Challenge Existing Paradigms • Manufacturing facilities with modular design: standardized, portable and flexible designs • Modular/reconfigurable manufacturing systems • Need for process oversight due to reduced segregation (i.e. ballroom) • Minimally disruptive reconfiguration for NPI • Area classification • Increased use of closed systems • Concurrent Multi-product Production • Segregation through technology • Predominantly single-use systems upstream • Limited CIP/SIP

  8. Operational Concepts will Challenge Existing Paradigms • Reduced size and volume of equipment • “Scale-up” by adding more production lines or extending time as opposed to increasing size of bioreactor • Continuous Harvesting and Purification • Sampling: finding the right balance in sampling frequency, on-line/in-line monitoring, rapid assays • Reduced residence time and process holds • Less opportunity for bioburden proliferation

  9. Connected Purification Process Reduces the Need for Large Hold Tanks FVIP Feed Column 3 Clean Column 3 Wash Column 3 Load Column 3 Equil Surge Tank CEX FT Column 2 Load Column 2 Wash Column 2 Clean Column 2 Elution FT – Viral Filtration Elapsed Time: 1.5-4 hrs. Elapsed Time: 3-7 hrs. Overall Time with Cleaning UF/DF Ultrafiltration Diafiltration Total Process Time: ~10-14 hrs. Elapsed Time: 7-11 hrs Viral Filtration

  10. Continuous Manufacturing: Key Quality and Regulatory Considerations • Continuous processes have to be validated to demonstrate that the process is consistently making product of intended quality • Steady-state: Is the process stable? • Control strategy: Continuous processes are more complex to design and require a higher level of control • Continuous process verification • Continuous fermentation: • Typically done at smaller scale but longer culture time • Focus on in vitro cell age (genetic stability), end-of-production cells (susceptibility to contamination, viral safety) • Use of physical, analytical barriers for prevention and early detection of contamination (e.g. HTST, PCR)

  11. Continuous Manufacturing: Key Quality and Regulatory Considerations • Implementation of a fully continuous process: • Enhanced process understanding, including raw material variability • Understanding of potential interactions between unit operations • Need to build in flexibility; • If a unit operation fails, does the entire process go down? • Process upset conditions: do you discard the portion of the batch affected by the upset? • Can process adjustments be made real-time? • Challenge is achieving control of the overall integrated process not just by unit operations • Batch definition

  12. Implications of PAT strategy • Integration of operational quality control with manufacturing • Facility design should allow for at-line testing • Scaling data-based decision making capabilities • PAT will increase relevant, actionable data acquired • Need for sophisticated data management systems • Increasing real-time quality assurance on the floor

  13. Single-use Technologies: Opportunities and Challenges • Innovation and improvements in single-use technology have led to broader acceptance by industry and regulators • Single-use technology is no longer limited to upstream processing • Emergence of ready-to-use, disposable purification systems • Overall robustness and functionality of single-use systems has improved

  14. Single-use Technologies: Opportunities and Challenges • Single-use technology offers many advantages over traditional stainless steel systems such as: • Increased flexibility • Quicker turnaround between batches, campaigns • Reduced cleaning burden (CIP, SIP, cleaning validation) • Reduced risk of cross-contamination between products (multi-use facilities) • Reduced risk of contamination and less down time • Improved sterile connections and sample handling

  15. Single-use Technologies: Opportunities and Challenges • The main concerns over the use of single-use technology pertain to extractables/leachables and vendor dependency • More information is now available on extractables and leachables to support risk assessments • Many disposables are now available from multiple vendors • Single-use systems from different vendors are not necessarily interchangeable; still have to demonstrate fitness-for-use • Lot to lot variability should also be assessed

  16. Comprehensive Leachables and Extractables Strategy Component Evaluation Process Assessment Analytical Confirmation • L&E profile major component of vendor selection • Vendor documentation evaluation • Compiled in format of risk assessment for many factors: • Temperature • Exposure duration • Surface area • Process step • Total L&E calculated based on cumulative assessment • Clearances for specific compounds of concern determined • Spiking studies to confirm clearance capability • Testing at full scale to confirm clearance Summary Report

  17. Conclusion • Continuous manufacturing clearly presents significant opportunities, not only from a business perspective but from a regulatory compliance perspective. • Product, process understanding and a robust control strategy will be key to successful implementations. • Regulators are open to new approaches in manufacturing and control strategies, however, there is a lack of experience with continuous manufacturing models in biotech. It will be critical to engage regulators early and keep an open dialogue during the development phase.

  18. Acknowledgments • Tony Mire-Sluis • Darrin Cowley • Barry Cherney • Brandon Persinger • Emanuela Lacana • Mark Heintzelman

More Related