1 / 40

Manufacturing of Cellular & Gene Therapies: Ensuring Product Safety and Quality

Manufacturing of Cellular & Gene Therapies: Ensuring Product Safety and Quality. Dan Takefman, Ph.D. Chief, Gene Therapy Branch Division of Cellular and Gene Therapies Center for Biologics Evaluation and Research FDA. Before You Begin Manufacturing….

rockwell
Download Presentation

Manufacturing of Cellular & Gene Therapies: Ensuring Product Safety and Quality

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. Manufacturing of Cellular & Gene Therapies: Ensuring Product Safety and Quality Dan Takefman, Ph.D. Chief, Gene Therapy Branch Division of Cellular and Gene Therapies Center for Biologics Evaluation and Research FDA

  2. Before You Begin Manufacturing… • Guidance for Review Staff and Sponsors: Content and Review of Chemistry, Manufacturing, and Control (CMC) Information for • Human Gene Therapy Investigational New Drug Applications (INDs) • Human Somatic Cell Therapy Investigational New Drug Applications (INDs) • http://www.fda.gov/cber/genadmin/octgtprocess.htm

  3. Step-wise Approach to Application of Regulatory Requirements • For phase I submission, product safety is the focus of the CMC assessment • Microbial contamination, freedom from adventitious agents, other assessments • Characterization as directly related to safety will need to be documented • Minimal characterization expected • Assurance of product quality increases with clinical development • Characterization (CFR 610) • cGMPs (CFR 210, 211)

  4. Product Safety and Quality • Components used in Product Manufacture • Final Product Testing and Characterization • Control of Manufacturing Process • cGMP Practices • In process controls

  5. CMC Information – How Much is Enough? • Important to describe your manufacturing process in enough detail for FDA to assess safety • If safety tests done by contract lab, submit SOPs and available information on assay sensitivity & specificity. If regulatory file available submit cross reference • Flow charts with narrative • When cross-referencing manufacturing information, be specific in what you are referencing.

  6. Components Used in Manufacture of Product • Vector • Cells • Allogeneic & autologous cell components • Cell bank system • Master cell bank/working cell bank • Master viral bank/working viral bank • Ancillary products/reagents • Growth factors, cytokines, MoAb

  7. Vector • Description, history and details on derivation of construct • Vector diagram • Sequence analysis (from MVB) • Full sequence for vectors <40KB • Vectors >40kb: sequence inserts, flanking regions, modified regions • Description of unexpected sequences • Do not submit only raw data

  8. Cells • Cell substrates for production of gene therapies • History, source, general characteristics • Autologous and allogeneic cells used for cell therapies or ex vivo modified gene therapies • Source (tissue and cell type) • Collection procedure (mobilization, surgery, leukapheresis, devices used) • Donor Eligibility (infectious disease screening & testing, 21 CFR 1271)

  9. Donor Eligibility – Testing • Allogeneic • HIV 1&2, HBV, HCV, Treponema pallidum • For viable, leukocyte-rich cells/tissues, additional test for HTLV I & II • For reproductive cells/tissues, additional tests for Chlamydia trachomatis and Neisseria gonorrhea • FDA licensed or approved kits or description of test methods, controls, sensitivity • Testing on donor mothers for cord blood or maternally derived tissue (not banked)

  10. Donor Eligibility – Testing • Autologous • DE testing recommended, not required • Determine if cell culture methods could propagate viruses

  11. Donor Eligibility – Screening • Allogeneic • Risk-factors for, and clinical evidence of relevant communicable disease agents or diseases • Communicable disease risk associated with xenotransplantation

  12. Master Cell Bank and Master Virus Bank • Safety Testing • Sterility • Mycoplasma

  13. Master Cell Bank and Master Virus Bank • Safety Testing (continued) • Adventitious Virus • In vitro and in vivo adventitious virus assays • Bovine and porcine viruses (not needed if reagents tested) • Human cell lines: EBV, HBV, HCV, CMV, HIV 1&2, HTLV 1 & 2, B19, (others) • Murine cell lines: MAP, retroviruses • RCV (GT products, typically on MVB) • Refer to guidance for assays and limits

  14. Master Cell Bank – Characterization • Identity • Cell therapies: phenotype, genotype, other markers • Gene therapies: isoenzyme, others • Purity • Contaminating cells (GT products) • Charactering cell types (CT products) • Tumorigenicity • May be considered a safety test for certain cellular products (e.g. stem cell products)

  15. Master Cell Bank – Characterization • Other- processes critical to safety • Document culture conditions, medium, cryopreservation, storage, genetic & phenotypic stability after multiple passages

  16. Working Cell Bank and Working Viral Bank • Safety • Sterility • Mycoplasma • In vitro adventitious virus assay • Replication competence virus (WVB) • Characterization • Identity • Stability • Others

  17. Reagents Used in Manufacturing • Tabulation of reagents used • Final concentration • Vendor • Source (human, bovine, etc.) • Licensed product, clinical grade, reagent grade • Certificates of Analysis • Cross reference letter • Qualification program • Safety testing and quality assessment

  18. Product Manufacturing • Vector production/purification • Cell products/ex vivo transduced cells • Method of collection/processing • Culture/transduction procedures • Other modifications (irradiation) • Final harvest

  19. Product Manufacturing (cont.) • Formulation of final product • Formulation buffer • Excipients • Vector/cell concentration • Storage

  20. Final Product Testing • Demonstration of product safety • Assessment of product characterization • Maintenance of product lot consistency • Results available prior to patient administration • Listing of tests, methods, acceptance criteria in IND

  21. Final Product Testing – Safety • Sterility (CFR 610.12) • Mycoplasma (CFR 610.30) • Endotoxin (CFR 610.13) • Adventitious Virus (for gene therapy products) • In vitro virus assay • Replication competent virus

  22. Approaches to Release Testing for Non-Cryopreserved Cells • Sterility • Regulation • 14 day sterility assay on final product • Approach • Sample 72-48 hours or after last manipulation - release on “no growth” • Results of Gram stain available-release on negative result • Sample final product & initiate 14 day sterility culture • Action plan in the event of positive for microorganisms

  23. Approaches to Release Testing • Mycoplasma • Recommend testing at cell harvest • Regulation: 21-28 day culture, 5-7 day fluorescent • Approach: PCR (6-8 hour test) • Pyrogenicity • Regulation: rabbit bioassay • Approach: LAL (1-2 hour test) • Equivalent methods (CFR 610.9) • Data demonstrating equivalence to methods in regulation needed for licensure

  24. Final Product Characterization • Final product testing • Purity (CFR 610.13) • Identity (CFR 610.14) • Potency (CFR 610.10) • Stability • Cell viability • Development of test methods and acceptance criteria

  25. Final Product Characterization • Identity • Restriction map, structural characterization, cell phenotype • Purity • Residual contaminants (DNA, protein, culture reagents) • Cell populations for cell therapies • Particle:IU ratio for vectors • Stability • Should be tested throughout all clinical phases

  26. Final Product Characterization • Potency • Indicate biological activity (s) specific/relevant to the product • Provide quantitative readout • Required prior to initiation of phase III • GT products transgene expression and titer acceptable in early phases • Cell therapies, cell surface phenotype in early phases • Guidance in development

  27. Approaches for Potency Measurements • Direct measure of biological activity • In vivo or in vitro assay • Indirect measure of biological activity • Analytical assay methods: non-bioassay method directly correlated to a unique and specific activity of the product • Multiple Assay Approach (Assay Matrix) • May not be possible or feasible to develop a single assay that encompasses all elements of an acceptable potency assay

  28. Product Characterization – Why? • To demonstrate lot-to-lot consistency • To show comparability after manufacturing changes • To generate solid clinical data • For pivotal trials characterization assays will need to be established with appropriate release limits

  29. Product Characterization • Start collecting data early! • Communicate progress with your CMC reviewer

  30. Current Good Manufacturing Practices (cGMP) • Definition • A set of current, scientifically sound methods, practices or principles that are implemented and documented during product development and production to ensure consistent manufacture of safe, pure and potent products • Increase in control of the manufacturing process with clinical trial advancement

  31. cGMPs for Early Development • For detailed guidance, please refer to: “Draft Guidance for Industry: INDs – Approaches to Complying with CGMP During Phase 1” • http://www.fda.gov/cber/gdlns/indcgmp.pdf

  32. GMPs: Early Clinical Phase Critical Questions • Is the process reproducible? • Appropriate testing at critical steps? • Quality of the raw and source materials adequately controlled? • Are the records and record keeping systems adequate?

  33. Examples for Early Development • Procedures to prevent contamination & cross contamination • Aseptic processing • Tracking of autologous products (labeling system) • Patient cell segregation • Methods qualification • Appropriate method specificity, sensitivity, reproducibility

  34. Examples for Early Development • Process qualification • Safety related process • Irradiation of tumor cells/ cell lines, removal of toxic residuals, viral clearance • Performance runs/development lots • Equipment calibration • Irradiators, other critical equipment

  35. Examples for Early Development • Quality (QC) Program • Ideal - separate unit with ultimate reporting to sponsor • Responsibility and authority to accept or reject materials, procedures and specifications • Designed to prevent, detect, and correct deviations and failures

  36. cGMPs that Develop with Clinical Studies – Examples • Process validation • Must first characterize your process • Identify and control sources of variability and understand how variability affects your product • Validate by licensure • Methods validation • Develop and refine • Validate by licensure • Process controls • In-process testing, specifications • SOP development

  37. Characterization and cGMPs • cGMP are a set of current, scientifically sound methods, practices or principles that are implemented and documented during product development and production to ensure consistent manufacture of safe, pure and potent products • Manufacturers often refer to their products as GMP grade, but aren’t assaying for potency or ensuring lot to lot consistency • Need to characterize your product and process!

  38. Summary • Step-wise Approach to Regulatory Requirements • Safety testing • Product characterization • Control of Manufacturing Process • cGMP Practices Ensure a Safe and Quality Product

  39. References and Contact Information • References for the Regulatory Process for OCTGT • http://www.fda.gov/cber/genadmin/octgtprocess.htm • General questions for OCTGT • 301-827-5102

  40. References and Contact Information • CMC Questions for Cellular Therapies • Keith Wonnacott • keithwonnocot@fda.hhs.gov • CMC Questions for Gene Therapies • Dan Takefman • dantakefman@fda.hhs.gov

More Related