1 / 18

Evaluation of Viral Clearance Studies

Evaluation of Viral Clearance Studies. Mahmood Farshid, Ph.D. Div. Of Hematology OBRR/ CBER/FDA. Biologics . Monoclonal antibodies and recombinant products produced in cell culture Blood and blood products and other human derived products Animal derived products.

lilia
Télécharger la présentation

Evaluation of Viral Clearance Studies

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. Evaluation of Viral Clearance Studies Mahmood Farshid, Ph.D. Div. Of Hematology OBRR/ CBER/FDA

  2. Biologics • Monoclonal antibodies and recombinant products produced in cell culture • Blood and blood products and other human derived products • Animal derived products

  3. Risk Reduction Strategies • Donor Screening: • donor history assessment, • written and oral questionnaire • Donors Testing: • Anti- HIV-1/2, HIV-1 p24 Ag ,anti-HCV, HBsAg , anti HBc, anti-HTLV-1/2, syphilis • (NAT for HCV and HIV) • Pharmacovigilance/ look back studies • Inactivation/Removal • Validating the manufacturing processes for removal / inactivation of viruses

  4. The Aim of Viral Validation • To provide evidence that the production process will effectively inactivate/remove viruses which could potentially be transmitted by the product • To provide indirect evidence that the production process has the capacity to inactivate/remove novel or yet undetermined virus contamination

  5. Virus inactivation: Chemical:organic solvents; pH extremes; solvent detergent; alcohol Physical: Heat treatment (dry heat or pasteurization) Virus removal Precipitation: ammonium sulfate etc. Chromatography: ion exchange; gel filtration; affinity; reverse phase Membrane filtration: Omega, Planova, DV50 Common Virus Clearance Methods

  6. Validation of Virus Removal/inactivation Include: • Scaling down process steps • Spiking appropriate steps with high titer of infectious virus (relevant or model) • Determining virus reduction factors for each step • Summing reduction factors to give a total log10 reduction value (LRV)

  7. Evaluation of the Effectiveness of Viral Clearance Step • Test viruses used • The design of the validation studies • The validity of scaled-down process • The kinetics of inactivation • Susceptibility to small variations in process parameters (robustness) • The limits of assay sensitivity • The log reduction achieved

  8. Virus Selection • Viruses that can potentially be transmitted by the product (relevant or specific model viruses) • Viruses with a wide range of physicochemical properties to evaluate robustness of the process (non-specific model viruses)

  9. Virus Selection • The nature of starting material • Cell lines • Human derived • Animal derived • Feasibility • Availability of a suitable culture system • Availability of high-titer stocks • Reliable methods for quantification

  10. Model viruses for human Blood-Derived Products Virus Model Envelope/ Size Resistance Genome (nm) HIV/HTLV HIV-1 Yes / RNA 80-130 Low HBV DHBV Yes / DNA ~ 40 Medium HCV BVDV Yes / RNA 40-50 Medium HAV HAV No / RNA 28-30 High CMV CMV/HSV Yes / DNA 150-200 Low-Med /PRV B19 PPV No / DNA 18-26 Very high

  11. Viruses Used to Validate Product Derived from Cell Lines Virus Genome Size(nm) Enveloped Resistance MVM ss-DNA 18-26 No Very high Reo-3 ds-RNA 60-80 No High MuLV ss-RNA 80-130 Yes Low PRV ds-DNA 150-200 Yes Low-med

  12. Virus Selection • DNA and RNA genome (single and double-stranded) • Lipid-enveloped and nonenveloped • Large, intermediate, and small size • From very highly resistant to inactivation to very • easily inactivated

  13. Scale-Down of Purification Steps • Usually 1/10 to 1/100 scale • Must keep buffers, pH, protein concentration, and product the same as full scale manufacturing • Must keep operation parameters as close to full scale as possible (e.g., bed height, flow rate) • Must show product is identical to production scale

  14. Criteria for An Effective Virus Clearance Step • Significant viral kill • Reproducible and controllable at process scale and model-able at the laboratory scale • Should have minimal impact on product yield and activity • Not generate neo-antigens or leave toxic residues

  15. GENERAL CONSIDERATIONS • Manufacturing processes for blood derived products should contain two effective steps for removal/inactivation of viruses • At least one step should be effective against non-enveloped viruses

  16. GENERAL CONSIDERATIONS(cont.) • At least one stage in a production process must inactivate rather than remove viruses • A single step having a large effect gives more assurance of viral safety than several steps having the same overall effect

  17. Limitations of Viral Validation Studies • Laboratory strains may behave differently than native viruses • Source plasma or Igs may have neutralizing antibodies • There may exist in any virus population a fraction that is resistant to inactivation • Scale-down processes may be differ from full-scale

  18. Limitations of Viral Validation Studies • Total virus reduction may be overestimated because of repeated and similar process steps • The ability of steps to remove virus after repeated use may vary

More Related