We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Perspectives on exploring hybrid LBA/LC–MS approach for clinical immunogenicity testing

    Hao Jiang

    *Author for correspondence: Tel.: +1 609 252 4493;

    E-mail Address: hao.jiang@bms.com

    Bioanalytical Sciences, Bristol-Myers Squibb Co., Princeton, NJ 08543, USA

    ,
    Heather Myler

    PPD Laboratories, Richmond, VA 23230, USA

    ,
    Jianing Zeng

    Bioanalytical Sciences, Bristol-Myers Squibb Co., Princeton, NJ 08543, USA

    ,
    Johanna Mora

    Bioanalytical Sciences, Bristol-Myers Squibb Co., Princeton, NJ 08543, USA

    ,
    Gerry Kolaitis

    Bioanalytical Sciences, Bristol-Myers Squibb Co., Princeton, NJ 08543, USA

    &
    Renuka Pillutla

    Bioanalytical Sciences, Bristol-Myers Squibb Co., Princeton, NJ 08543, USA

    Published Online:https://doi.org/10.4155/bio-2018-0107

    Biological drug products may elicit an antidrug antibody (ADA) response. The current widely used bridging ligand binding assay (LBA) is the gold standard for ADA assessments in drug development, which is a qualitative assay followed by a quasi-quantitative titer analysis but can be prone to interferences from biological matrices, drug targets and circulating drugs. We present our perspectives and findings in exploring a hybrid LBA/LC–MS as an orthogonal bioanalytical tool for clinical immunogenicity assessments. The hybrid LBA/LC–MS is a semiquantitative assay with acceptable specificity, drug tolerance and the capability of multiplexed detection of ADA isotypes. The assay results suggest this technology to be a promising and complementary bioanalytical tool that can provide informative immunogenicity data in drug development.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Mire-Sluis AR, Barrett YC, Devanarayan V et al. Recommendations for the design and optimization of immunoassays used in the detection of host antibodies against biotechnology products. J. Immunol. Methods 289, 1–16 (2004). • A commonly referenced paper for ligand binding assay (LBA) immunogenicity assay design and optimization.
    • 2. Shankar G, Devanarayan V, Amaravadi L et al. Recommendations for the validation of immunoassays used for detection of host antibodies against biotechnology products. J. Pharm. Biomed. Anal. 48(5), 1267–1281 (2008).
    • 3. Shankar G, Devanarayan V, Amaravadi L et al. Recommendations for the validation of immunoassays used for detection of host antibodies against biotechnology products. J. Pharm. Biomed. Anal. 48(5), 1267–1281 (2008).
    • 4. European Medicines Agency. Guideline on immunogenicity assessment of monoclonal antibodies intended for in vivo clinical use (2012). www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2012/06/WC500128688.pdf
    • 5. Zhong ZD, Clements-Egan A, Gorovits B et al. Drug target interference in immunogenicity assays: recommendations and mitigation strategies. AAPS J. 19(6), 1564–1575 (2017).
    • 6. Hart MH, Vrieze H, Wouters D et al. Differential effect of drug interference in immunogenicity assays. J. Immunol. Methods 372(1–2), 196–203 (2011).
    • 7. Chen K, Page JG, Schwartz AM et al. False-positive immunogenicity responses are caused by CD20+ B cell membrane fragments in an anti-ofatumumab antibody bridging assay. J. Immunol. Methods 394(1–2), 22–31 (2013).
    • 8. Lofgren JA, Dhandapani S, Pennucci JJ et al. Comparing ELISA and surface plasmon resonance for assessing clinical immunogenicity of panitumumab. J. Immunol. 178(11), 7467–7472 (2007).
    • 9. Li J, Schantza A, Schwegler M et al. Detection of low-affinity antidrug antibodies and improved drug tolerance in immunogenicity testing by Octet® biolayer interferometry. J. Pharm. Biomed. Anal. 54(2), 286–294 (2011).
    • 10. Bourdage J, Cook CA, Farrington DL et al. An Affinity Capture Elution (ACE) assay for detection of antidrug antibody to monoclonal antibody therapeutics in the presence of high levels of drug. J. Immunol. Methods 327, 10–17 (2007).
    • 11. Smith HW, Butterfield A, Sun D. Detection of antibodies against therapeutic proteins in the presence of residual therapeutic protein using a solid-phase extraction with acid dissociation (SPEAD) sample treatment prior to ELISA. Regul. Toxicol. Pharmacol. 49(3), 230–237 (2007).
    • 12. Zoghbi J, Xu Y, Grabert R et al. A breakthrough novel method to resolve the drug and target interference problem in immunogenicity assays. J. Immunol. Methods. 426, 62–69 (2015).
    • 13. Xu W, Jiang H, Titsch C et al. Development and characterization of a pre-treatment procedure to eliminate human monoclonal antibody therapeutic drug and matrix interference in cell-based functional neutralizing antibody assays. J. Immunol. Methods 1759(416), 318–314 (2014).
    • 14. Niu H, Klem T, Yang J et al. A biotin-drug extraction and acid dissociation (BEAD) procedure to eliminate matrix and drug interference in a protein complex antidrug antibody (ADA) isotype specific assay. J. Immunol. Methods 446, 30–36 (2017).
    • 15. Carrasco-Triguero M, Mahood C, Milojic-Blair M et al. Overcoming soluble target interference in an anti-therapeutic antibody screening assay for an antibody-drug conjugate therapeutic. Bioanalysis 4(16), 2013–2026 (2012).
    • 16. Zeng J, Jiang H, Luo L. Semiquantification and isotyping of antidrug antibodies by immunocapture-LC/MS for immunogenicity assessment. Lee MSJi QC (Eds). Protein Analysis Using Mass Spectrometry: Accelerating Protein Biotherapeutics from Lab to Patient. John Wiley & Sons, Inc., NJ, USA 91–97 (2017).
    • 17. Lee JW. Generic method approaches for monoclonal antibody therapeutics analysis using both ligand binding and LC–MS/MS techniques. Bioanalysis 8(1), 19–27 (2016).
    • 18. Chappell DL, Lassman ME, McAvoy T et al. Quantitation of human peptides and proteins via MS: review of analytically validated assays. Bioanalysis 6(13), 1843–1857 (2014).
    • 19. Wilffert D, Bischoff R, van de Merbel NC. Antibody-free workflows for protein quantification by LC–MS/MS. Bioanalysis 7(6), 763–779 (2015).
    • 20. Jiang H, Zeng J, Titsch C et al. Fully validated LC–MS/MS assay for the simultaneous quantitation of co-administered therapeutic antibodies in cynomolgus monkey serum. Anal. Chem. 85(20), 9859–9867 (2013).
    • 21. Furlong MT, Titsch C, Xu W et al. Evaluation of a universal LC–MS/MS assay for bioanalysis of human IgG4 subclass monoclonal antibody protein drugs and drug candidates in clinical studies. Bioanalysis 6, 1747–1758 (2014).
    • 22. Jiang H, Xu W, Titsch C et al. Innovative use of LC–MS/MS for simultaneous quantitation of neutralizing antibody, residual drug and human immunoglobulin G in immunogenicity assay development. Anal. Chem. 86(5), 2673–2680 (2014). •• A proof-of-concept report to demonstrate that hybrid LBA/LC–MS can detect antidrug antibody.
    • 23. Ackermann B, Neubert H, Hughes N et al. 2015 white paper on recent issues in bioanalysis: focus on new technologies and biomarkers (Part 2 – hybrid LBA/LCMS and input from regulatory agencies). Bioanalysis 7(23), 3019–3034 (2015).
    • 24. Song A, Lee A, Garofolo F et al. 2016 white paper on recent issues in bioanalysis: focus on biomarker assay validation (BAV): (Part 2 – Hybrid LBA/LCMS and input from regulatory agencies). Bioanalysis 8(23), 2457–2474 (2016).
    • 25. Neubert H, Song A, Lee A et al. 2017 white paper on recent issues in bioanalysis: rise of hybrid LBA/LCMS immunogenicity assays (Part 2: hybrid LBA/LCMS biotherapeutics, biomarkers & immunogenicity assays and regulatory agencies’ inputs). Bioanalysis 9(23), 1895–1912 (2017).
    • 26. Chen LZ, Roos D, Philip E. Development of immunocapture-LC/MS assay for simultaneous ADA isotyping and semiquantitation. J. Immunol. Res. 2016, 7682472 (2016). •• Application of hybrid LBA/LC–MS in antidrug antibody isotyping and semiquantitation.
    • 27. Neubert H, Grace C, Rumpel K et al. Assessing immunogenicity in the presence of excess protein therapeutic using immunoprecipitation and quantitative mass spectrometry. Anal. Chem. 80(18), 6907–6914 (2008). • First application of indirect LBA/MS to assess immunogenicity.
    • 28. Dai S, Schantz A, Clements-Egan A et al. Development of a method that eliminates false-positive results due to nerve growth factor interference in the assessment of fulranumab immunogenicity. AAPS J. 16(3), 464–477 (2014).
    • 29. Nesman S, Gokemeijer J, Terryberry J et al. Isotyping therapeutic protein immunogenicity in an automated multiplexed assay. http://sqidiagnostics.com/sites/default/files/resources/pdfs/Isotyping%20Therapeutic%20Protein%20Immunogenicity%20in%20an%20Automated%20Multiplexed%20Assay.pdf
    • 30. Postelnek J, Myler H, Santockyte R et al. MSD U-PLEX® technology differentially measures domain-specific antidrug antibodies. http://abstracts.aaps.org/Verify/NBC15/PosterSubmissions/M1030.pdf
    • 31. GlobalRPH. http://www.globalrph.com/labs_i.htm#
    • 32. Song S, Yang L, Trepicchio WL et al. Understanding the supersensitive antidrug antibody assay: unexpected high antidrug antibody incidence and its clinical relevance. J. Immunol Methods 2016,3072586 (2016).
    • 33. U.S. Department of Health and Human Services Food and Drug Administration. Immunogenicity testing of therapeutic protein products – developing and validating assays for anti-drug antibody detection (2019). www.fda.gov/media/119788/download
    • 34. Neubert H, Song A, Lee A et al. White paper: rise of hybrid LBA/LCMS immunogenicity assays (Part 2: hybrid LBA/LCMS biotherapeutics, biomarkers & immunogenicity assays and regulatory agencies’ inputs). Bioanalysis 9(23), 1895–1912 (2017). •• White paper to summarize consensus and regulatory perspectives on hybrid LBA/LC–MS immunogenicity assays.