BioPharm International - March 2024

BioPharm International - March 2024

Issue link: https://www.e-digitaleditions.com/i/1517675

Contents of this Issue

Navigation

Page 5 of 23

6 BioPharm International ® Quality and Regulatory Sourcebook eBook March 2024 www.biopharminternational.com Cell and Gene Therapies of the lifecycle phase. For clinical-phase and for com- mercial-phase operations, the effectiveness of the cleaning must be validated though swabbing and/or contact plating for microbial growth. It may be effi- cient to combine the line clearance procedure with the cleaning one. It should be noted that traceability means both chain of identity (COI) and chain of cus- tody (COC). These must be implemented throughout the entire lifecycle. Using validated automated bar- code systems is standard as it is much more reliable than any manual process. Pre-sterilized consumable materials and reagents are commonly used, and these are purchased ready to use from qualified suppliers. As sterility assurance is critical, so are the suppliers of these materials. The PQS must have a supplier management procedure in place that allows the risk-based selection, assess- ment, and continuous qualification of suppliers, be- ginning with clinical Phase I. Finding manufacturers or suppliers of the required consumable materials and reagents that are of appropriate quality, manu- factured under suitably controlled conditions, and in stable supply can be a challenge. In analytical testing, as standard, the respective pharmacopeial compendia must be complied with, including microbial and for sterility testing, again sta r t ing w it h product for clin ica l Phase I. Sma l l batch sizes can mean that compendial requirements for sterility test sample size cannot be complied with. A solution may be to use rapid sterility testing meth- ods, which have increasingly become the norm, and are recommended for CGT products due to the speed of obtaining results. Though these are non-compen- dial methods, they can be used if appropriately vali- dated for their intended use. With standard sterility test methods, the results may not be available at the time of product release (determined by the limited shelf-life), suggesting t hat rapid steri lit y testing methods may be the preferred method. It should be noted that some regulator y agencies expect the bioburden and endotoxins testing results to be part of product release. As mentioned, non-compendial methods must be fully validated, which in turn re- quires a PQS that encompasses all the relevant re- quired analytical method validation processes and procedures. Of course, staff need to have the relevant knowledge, training, and expertise to execute these to GMP standards. While qualif ied assays may be used for testing patients during early development phases, samples from pivotal clinical trials (clinical Phase III) need to be tested using fully validated assays. Consideration should be given to the maximum sample storage time as developing some of the necessary assays (e.g., for immunogenicity) may well take several years to fully develop and validate. A s k nowle d ge a nd pr oces s u nder s t a nd i ng i s gained t hroughout t he lifec ycle, changes wi l l be made to the process and the product formulation. From a PQS perspective, a change management pro- cess should be in place; the complexity of which will likely increase closer to the product being commer- cialized. That should meet the expected current GMP requirements. Significant changes to optimize the formulation and its presentation, such as changes to the primar y packaging or the concentration of excipients, should be made prior to the start of piv- otal clinical Phase III studies. Making such changes during Phase III is h igh risk and is discouraged. In any case, as a minimum, analytical data will be needed to demonstrate the comparability of the pre- and post-change materials and to support the stabil- ity of the post-change materials. Such changes may also impact the trial design to demonstrate compa- rability of immunogenicity. Conclusion Throughout early clinical development, the primary focus is on clinical data and positive outcomes. What is often lacking and frequently overlooked is the ap- preciation of the resources needed to establish regu- latory compliance. This can lead to the appropriate evolution of qua lit y systems encompassing GMP being neglected. This results in developers being unprepared for and unsuccessful in regulatory ap- proval, and significant delays to commercialization of CGT. Understanding how to apply phase-appropriate GMP is crucial for achieving successful product ap- proval with any regulatory agency and is critical to- wards patients getting optimal access to their needed therapies. References 1. FDA. Approved Cellular and Gene Therapy Prod- uc t s. F DA .gov (accessed Feb. 29, 2024). ht t ps:// w w w. f d a . g o v/ v a c c i n e s - b l o o d - b i o l o g i c s/c e l - lu l a r-ge ne -t he r a p y-pr o duc t s/a p pr o ve d- c e l lu- lar-and-gene-therapy-products 2. EM A. CAT Qua r terly High lights a nd Approved ATMPs. ema.europa.eu. (accessed Feb. 29, 2024). h t t p s:// w w w.e m a .e u r o p a .e u/e n/d o c u m e n t s/ r e p or t/c a t- q u a r t e rl y-h i g h l i g ht s-a p pr o ve d-a t- mps-may-2023_en.pdf 3. www.clinicaltrials.gov 4. EC. Guidelines on Good Manufacturing Practice Speci f ic to Adva nced T herapy Med ici na l Prod- ucts. EudraLex Vol. 4. https://health.ec.europa.eu/ document/down load/ad33d9dd-03f0-4bef-af53- 21308ce2187d _en?f i lena me=2017_11 _ 22 _ g u ide- lines_gmp_for_atmps.pdf 5. ICH. www.ich.org. ■

Articles in this issue

Links on this page

Archives of this issue

view archives of BioPharm International - March 2024 - BioPharm International - March 2024