We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
General Content: White PaperFree Access

Recommendations on qPCR/ddPCR assay validation by GCC

    Mark Wissel

    Eurofins Viracor BioPharma, Lenexa,Kansas, USA

    ,
    Martin Poirier

    Altasciences, Everett, Washington, USA

    ,
    Christina Satterwhite

    Charles River Laboratories, Reno, Nevada, USA

    ,
    John Lin

    Frontage Laboratories, Exton, Pennsylvania, USA

    ,
    Rafiq Islam

    Smithers, Gaithersburg, Maryland, USA

    ,
    Jennifer Zimmer

    Alturas Analytics, Moscow, Idaho, USA

    ,
    Ardeshir Khadang

    Axis Clinical, Dilworth, Minnesota, USA

    ,
    Jennifer Zemo

    BioAgilytix, Durham, North Carolina, USA

    ,
    Todd Lester

    BioAgilytix, Durham, North Carolina, USA

    ,
    Marianne Fjording

    BioAgilytix, Durham, North Carolina, USA

    ,
    Amanda Hays

    BioAgilytix, Durham, North Carolina, USA

    ,
    Nicola Hughes

    Biopharmaservices, Toronto, Ontario, Canada

    ,
    Fabio Garofolo

    BRI - a Frontage Company, Vancouver, British Columbia, Canada

    ,
    Rudolf Guilbaud

    CellCarta, Montreal, Quebec, Canada

    ,
    Elizabeth Groeber

    Charles River Laboratories, Ashland, Ohio, USA

    ,
    Heidi Renfrew

    Charles River Laboratories, Shrewsbury, Massachusetts, USA

    ,
    Kelly Colletti

    Charles River Laboratories, Reno, Nevada, USA

    ,
    Mathilde Yu

    CIRION, Montreal, Quebec, Canada

    ,
    Jenny Lin

    CMIC, Hoffman Estates, Illinois, USA

    ,
    Xinping Fang

    CR Medicon, Piscataway, New Jersey, USA

    ,
    Santosh Shah

    Frontage Laboratories, Exton, Pennsylvania, USA

    , , ,
    Roger Hayes

    ICON Laboratory Services, Whitesboro, New York, USA

    ,
    John Pirro

    KCAS Bioanalytical & Biomarker Services, Shawnee Mission, Kansas, USA

    ,
    Cheikh Kane

    KCAS Bioanalytical & Biomarker Services, Shawnee Mission, Kansas, USA

    ,
    Marsha Luna

    KCAS Bioanalytical & Biomarker Services, Shawnee Mission, Kansas, USA

    ,
    Allan Xu

    Keystone Bioanalytical, Lansdale, Pennsylvania, USA

    , ,
    Mark O'Dell

    Labcorp, Indianapolis, Indiana, USA

    , , ,
    Jennifer Vance

    Nexelis - a Q2 Solutions Company, Seattle, Washington, USA

    ,
    Esme Farley

    Pharmaron, Germantown, Maryland, USA

    ,
    Katie Matys

    PPD Laboratories, Richmond, Virginia, USA

    ,
    Edward Tabler

    PPD Laboratories, Richmond, Virginia, USA

    ,
    William Mylott

    PPD Laboratories, Richmond, Virginia, USA

    ,
    Moucun Yuan

    PPD Laboratories, Richmond, Virginia, USA

    ,
    Shane Karnik

    Pyxant Labs, Colorado Springs, Colorado, USA

    ,
    Troy Voelker

    Pyxant Labs, Salt Lake City, Utah, USA

    ,
    Ira DuBey

    Smithers, Gaithersburg, Maryland, USA

    ,
    Clark Williard

    Syneos Health, Princeton, New Jersey, USA

    ,
    Jing Shi

    WuXi AppTec, Plainsboro, New Jersey, USA

    &
    Jim Yamashita

    WuXi AppTec, Plainsboro, New Jersey, USA

    Published Online:https://doi.org/10.4155/bio-2022-0109

    Gene therapy, cell therapy and vaccine research have led to an increased use of qPCR/ddPCR in bioanalytical laboratories. CROs are progressively undertaking the development and validation of qPCR and ddPCR assays. Currently, however, there is limited regulatory guidance for the use of qPCR and a complete lack of any regulatory guidelines for the use of the newer ddPCR to support regulated bioanalysis. Hence, the Global CRO Council in Bioanalysis (GCC) has issued this White Paper to provide; 1) a consensus on the different validation parameters required to support qPCR/ddPCR assays; 2) a harmonized approach to their validation and 3) a consistent development of standard operating procedures (SOPs) for all the bioanalytical laboratories using these techniques.

    References

    • 1. Premkumar N, Lowes S, Jersey J et al. Formation of a Global Contract Research Organization Council for Bioanalysis. Bioanalysis 2(11), 1797–1800 (2010).
    • 2. Breda M, Garofolo F, Caturla MC et al. Conference Report: The 3rd Global CRO Council for Bioanalysis at the International Reid Bioanalytical Forum. Bioanalysis 3(24), 2721–2727 (2011).
    • 3. Lowes S, Jersey J, Shoup R et al. Conference Report: 4th Global CRO Council for Bioanalysis: coadministered drugs stability, EMA/US FDA Guidelines, 483s and Carryover. Bioanalysis 4(7), 763–768 (2012).
    • 4. Nicholson R, Lowes S, Caturla MC et al. Conference Report: 6th GCC focus on LBA: critical reagents, positive controls and reference standards; specificity for endogenous compounds; biomarkers; biosimilars. Bioanalysis 4(19), 2335–2342 (2012).
    • 5. Rocci M, Lowes S, Shoup R et al. 7th GCC Insights: incurred samples use; fit-for-purpose validation, solution stability, electronic laboratory notebook and hyperlipidemic matrix testing. Bioanalysis 6(20), 2713–2720 (2014).
    • 6. Hayes R, LeLacheur R, Dumont I et al. 9th GCC closed forum: CAPA in regulated bioanalysis; method robustness, biosimilars, preclinical method validation, endogenous biomarkers, whole blood stability, regulatory audit experiences and electronic laboratory notebooks. Bioanalysis 8(6), 487–495 (2016).
    • 7. Islam R, Briscoe C, Bower J et al. 11th GCC Closed Forum: cumulative stability; matrix stability; immunogenicity assays; laboratory manuals; biosimilars; chiral methods; hybrid LBA/LCMS assays; fit-for-purpose validation; China Food and Drug Administration bioanalytical method validation. Bioanalysis 10(7), 433–444 (2018).
    • 8. Briscoe C, Hughes N, Hayes R et al. 12th GCC Closed Forum: critical reagents; oligonucleotides; CoA; method transfer; HRMS; flow cytometry; regulatory findings; stability and immunogenicity. Bioanalysis 11(12), 1129–1138 (2019).
    • 9. Bower J, Fast D, Garofolo F et al. 8th GCC: Consolidated feedback to US FDA on the 2013 Draft FDA guidance on bioanalytical method validation. Bioanalysis 6(22), 2957–2963 (2014).
    • 10. Cape S, Islam R, Nehls C et al. The 10th GCC Closed Forum: rejected data, GCP in bioanalysis, extract stability, BAV, processed batch acceptance, matrix stability, critical reagents, ELN and data integrity and counteracting fraud. Bioanalysis 9(7), 505–516 (2017).
    • 11. Bower J, Zimmer J, McCown S et al. Recommendations for the content and management of Certificates of Analysis for reference standards from the GCC for bioanalysis. Bioanalysis 13(8), (2021).
    • 12. Nehls C, Buonarati M, Cape S et al. GCC Consolidated Feedback to ICH on the 2019 ICH M10 Bioanalytical Method Validation Draft Guideline. Bioanalysis 11(18s), 1–228 (2019).
    • 13. Islam R, Kar S, Ritzén H et al. Recommendations for classification of commercial LBA kits for biomarkers in drug development from the GCC for bioanalysis. Bioanalysis 11(7), 645–653 (2019).
    • 14. Lowes S, LeLacheur R, Shoup R et al. Recommendations on incurred sample stability (ISS) by GCC. Bioanalysis 6(18), 2385–2390 (2014).
    • 15. Hougton R, Gouty D, Allinson J et al. Recommendations on biomarker bioanalytical method validation by GCC. Bioanalysis 4(20), 2439–2446 (2012).
    • 16. Lowes S, Boterman M, Doig M et al. Recommendations on bioanalytical method stability implications of co-administered and co-formulated drugs by Global CRO Council for Bioanalysis (GCC). Bioanalysis 4(17), 2117–2126 (2012).
    • 17. Boterman M, Doig M, Breda M et al. Recommendations on the interpretation of the new European Medicines Agency Guideline on bioanalytical method validation by Global CRO Council for Bioanalysis (GCC). Bioanalysis 4(6), 651–660 (2012).
    • 18. Sangster T, Maltas J, Struwe P et al. Recommendations on ISR in multi analyte assays, QA/bioanalytical consultants and GCP by Global CRO Council for Bioanalysis (GCC). Bioanalysis 4(14), 1723–1730 (2012).
    • 19. Lowes S, Jersey J, Shoup R et al. Recommendations on: internal standard criteria, stability, incurred sample reanalysis and recent 483s by the Global CRO Council for Bioanalysis. Bioanalysis 3(12), 1323–1332 (2011).
    • 20. European Medicines Agency. EMA General Principles to Address Virus and Vector Shedding (EMEA/CHMP/ICH/449035/2009) (2009). https://www.ema.europa.eu/en/documents/scientific-guideline/international-conference-harmonisation-technical-requirements-registration-pharmaceuticals-human-use_en-10.pdf
    • 21. US FDA Center for biologics evaluation and research. Guidance for industry: design and analysis of shedding studies for virus or bacteria-based gene therapy and oncolytic products (2015). https://www.fda.gov/media/89036/download
    • 22. US FDA Center for biologics evaluation and research. Guidance for industry: long term follow-up after administration of human gene therapy products (2020). https://www.fda.gov/media/113768/download
    • 23. Pharmaceuticals and Medical Devices Agency. PMDA Japan guidance on ensuring the quality and safety of products for gene therapy (2019). https://www.pmda.go.jp/files/000235607.pdf
    • 24. Piccoli S, Mehta D, Vitaliti A et al. 2019 White Paper on Recent Issues in Bioanalysis: FDA Immunogenicity Guidance, Gene Therapy, Critical Reagents, Biomarkers and Flow Cytometry Validation (Part 3-Recommendations on 2019 FDA Immunogenicity Guidance, Gene Therapy Bioanalytical Challenges, St. Bioanalysis (2019).
    • 25. US FDA Center for biologics evaluation and research. Guidance for Industry: Scientific Considerations in Demonstrating Biosimilarity to a Reference Product (2015). https://www.fda.gov/media/82647/download
    • 26. Ma H, Bell KN, Loker RN. qPCR and qRT-PCR analysis: Regulatory points to consider when conducting biodistribution and vector shedding studies. Mol. Ther. – Methods Clin. Dev. 20, 152–168 (2021).
    • 27. Stevenson L, Richards S, Pillutla R et al. 2018 White Paper on Recent Issues in Bioanalysis: focus on flow cytometry, gene therapy, cut points and key clarifications on BAV (Part 3 – LBA/cell-based assays: immunogenicity, biomarkers and PK assays). Bioanalysis 10(24), 1973–2001 (2018).
    • 28. Piccoli S, Mehta D, Vitaliti A et al. 2019 White Paper on Recent Issues in Bioanalysis: FDA Immunogenicity Guidance, Gene Therapy, Critical Reagents, Biomarkers and Flow Cytometry Validation (Part 3 – Recommendations on 2019 FDA Immunogenicity Guidance, Gene Therapy Bioanalytical Challenges, Strategies for Critical Reagent Management, Biomarker Assay Validation, Flow Cytometry Validation & CLSI H62). Bioanalysis 2207–2244 (2019).
    • 29. Corsaro B, Yang T, Murphy R et al. 2020 White Paper on Recent Issues in Bioanalysis: Vaccine Assay Validation, qPCR Assay Validation, QC for CAR-T Flow Cytometry, NAb Assay Harmonization and ELISpot Validation (Part 3 – Recommendations on Immunogenicity Assay Strategies, NAb Assays, Biosimilars and FDA/EMA Immunogenicity Guidance/Guideline, Gene & Cell Therapy and Vaccine Assays). Bioanalysis 13(6), 415–463 (2021).
    • 30. Sugimoto H, Chen S, Minembe JP et al. Insights on droplet digital pcr-based cellular kinetics and biodistribution assay support for CAR-T cell therapy. AAPS J. 23(2), (2021).
    • 31. European Medicines Agency. Guideline on quality, non-clinical and clinical aspects of medicinal products containing genetically modified cells. https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-quality-non-clinical-clinical-aspects-medicinal-products-containing-genetically-modified_en-0.pdf (2020).
    • 32. Martinez M, Moon EK. CAR T cells for solid tumors: new strategies for finding, infiltrating, and surviving in the tumor microenvironment. Front. Immunol. 10 (2019).
    • 33. Roberts CC, Swoyer R, Bryan JT, Taddeo FJ. Comparison of real-time multiplex human papillomavirus (HPV) PCR assays with the linear array HPV genotyping PCR assay and influence of DNA extraction method on HPV detection. J. Clin. Microbiol. 49(5), 1899–1906 (2011).
    • 34. Wang Y, Kang H, Liu X, Tong Z. Combination of RT-qPCR testing and clinical features for diagnosis of COVID-19 facilitates management of SARS-CoV-2 outbreak. J. Med. Virol. 92(6), 538–539 (2020).
    • 35. Bustin SA, Benes V, Garson JA et al. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clin. Chem. 55(4), 611–622 (2009).
    • 36. Whale AS, de Spiegelaere W, Trypsteen W et al. The digital MIQE guidelines update: minimum information for publication of quantitative digital PCR experiments for 2020. Clin. Chem. 66(8), 1012–1029 (2020).
    • 37. Yang TY, Doddareddy R. Considerations in the development and validation of real-time quantitative polymerase chain reaction and its application in regulated bioanalysis to characterize the cellular kinetics of CAR-T products in clinical studies. Bioanalysis 13(2), 115–128 (2021).
    • 38. Rangarajan S, Walsh L, Lester W et al. AAV5-Factor VIII gene transfer in severe hemophilia A. N. Engl. J. Med. 377(26), 2519–2530 (2017).
    • 39. Bustin S, Huggett J. qPCR primer design revisited. Biomol. Detect. Quantif. 14, 19–28 (2017).
    • 40. US FDA Center for biologics evaluation and research. Guidance for industry: bioanalytical method validation (2018). https://www.fda.gov/media/70858/download
    • 41. Zhao Y, Stepto H, Schneider CK. Development of the first World Health Organization lentiviral vector standard: toward the production control and standardization of lentivirus-based gene therapy products. Human Gene Ther. Methods 28(4), 205–214 (2017).
    • 42. Global CRO Council for Bioanalysis (2021). http://www.global-cro-council.org