We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Published Online:https://doi.org/10.4155/bio-2022-0215

Over the past two decades, we have seen an increase in the complexity and diversity of biotherapeutic modalities pursued by biopharmaceutical companies. These biologics are multifaceted and susceptible to post-translational modifications and in vivo biotransformation that could impose challenges for bioanalysis. It is vital to characterize the functionality, stability and biotransformation products of these molecules to enable screening, identify potential liabilities at an early stage and devise a bioanalytical strategy. This article highlights our perspective on characterization and bioanalysis of biologics using hybrid LC–MS in our global nonregulated bioanalytical laboratories. AbbVie's suite of versatile, stage-appropriate characterization assays and quantitative bioanalytical approaches are discussed, along with guidance on their utility in answering project-specific questions to aid in decision-making.

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

References

  • 1. Zhu L, Glick J, Flarakos J. Bioanalytical challenges in support of complex modalities of antibody-based therapeutics. AAPS J. 22(6), 130 (2020). •• This review provides a very good synopsis of the landscape of complex modalities and their respective bioanalytical challenges.
  • 2. Kaur S, Bateman KP, Glick J et al. IQ consortium perspective: complementary LBA and LC–MS in protein therapeutics bioanalysis and biotransformation assessment. Bioanalysis 12(4), 257–270 (2020). • This perspective article is a great reference for the two bioanalytical platform comparisons for biotherapeutic bioanalysis.
  • 3. Durbin KR, Nottoli MS, Catron ND, Richwine N, Jenkins GJ. High-throughput, multispecies, parallelized plasma stability assay for the determination and characterization of antibody–drug conjugate aggregation and drug release. ACS Omega 2(8), 4207–4215 (2017). •• This article is published by the AbbVie internal team from Drug metabolism pharmacokinetics and bioanalysis (DMPK-BA) and a routine, high-throughput, two-in-one assay platform used to assess the stability of biologics and drug conjugates.
  • 4. Sydow JF, Lipsmeier F, Larraillet V et al. Structure-based prediction of asparagine and aspartate degradation sites in antibody variable regions. PLOS ONE 9(6), e100736 (2014).
  • 5. Jarasch A, Koll H, Regula JT, Bader M, Papadimitriou A, Kettenberger H. Developability assessment during the selection of novel therapeutic antibodies. J. Pharm. Sci. 104(6), 1885–1898 (2015).
  • 6. Liu YD, Van Enk JZ, Flynn GC. Human antibody Fc deamidation in vivo. Biologicals 37(5), 313–322 (2009).
  • 7. Varshavsky A. The N-end rule pathway and regulation by proteolysis. Protein Sci. 20(8), 1298–1345 (2011).
  • 8. Shadid M, Bowlin S, Bolleddula J. Catabolism of antibody drug conjugates and characterization methods. Bioorg. Med. Chem. 25(12), 2933–2945 (2017).
  • 9. Bessire AJ, Ballard TE, Charati M et al. Determination of antibody–drug conjugate released payload species using directed in vitro assays and mass spectrometric interrogation. Bioconjug. Chem. 27(7), 1645–1654 (2016).
  • 10. Sang H, Liu J, Zhou F et al. Target-responsive subcellular catabolism analysis for early-stage antibod–drug conjugates screening and assessment. Acta Pharm. Sin. B 11(12), 4020–4031 (2021).
  • 11. Tumey LN, Leverett CA, Vetelino B et al. Optimization of tubulysin antibody–drug conjugates: a case study in addressing ADC metabolism. ACS Med. Chem. Lett. 7(11), 977–982 (2016).
  • 12. Cini E, Faltoni V, Petricci E et al. Antibody drug conjugates (ADCs) charged with HDAC inhibitor for targeted epigenetic modulation. Chem. Sci. 9(31), 6490–6496 (2018).
  • 13. Bessire AJ, Subramanyam C. LC/MS methods for studying lysosomal ADC catabolism. Methods Mol. Biol. 2078, 341–351 (2020).
  • 14. Austin CD, De Mazière AM, Pisacane PI et al. Endocytosis and sorting of ErbB2 and the site of action of cancer therapeutics trastuzumab and geldanamycin. Mol. Biol. Cell 15(12), 5268–5282 (2004).
  • 15. Durbin KR, Phipps C, Liao X. Mechanistic modeling of antibody–drug conjugate internalization at the cellular level reveals inefficient processing steps. Mol. Cancer Ther. 17(6), 1341–1351 (2018). • This is an AbbVie internal publication that emphasizes the power of mechanistic modeling used with cellular disposition assay to determine antibody–drug conjugate internalization.
  • 16. Durbin KR, Nottoli MS, Jenkins GJ. Effects of microtubule-inhibiting small molecule and antibody–drug conjugate treatment on differentially-sized A431 squamous carcinoma spheroids. Sci. Rep. 10(1), 907 (2020).
  • 17. Bhat J, Narayan A, Venkatraman J, Chatterji M. LC–MS based assay to measure intracellular compound levels in Mycobacterium smegmatis: linking compound levels to cellular potency. J. Microbiol. Methods 94(2), 152–158 (2013).
  • 18. Wu C, Wang X, Xu M, Liu Y, Di X. Intracellular accumulation as an indicator of cytotoxicity to screen hepatotoxic components of Chelidonium majus L. by LC–MS/MS. Molecules 24(13), 2410 (2019).
  • 19. Singh AP, Shah DK. Measurement and mathematical characterization of cell-level pharmacokinetics of antibody–drug conjugates: a case study with trastuzumab-vc-MMAE. Drug Metab. Dispos. 45(11), 1120–1132 (2017).
  • 20. Wei C, Zhang G, Clark T et al. Where did the linker-payload go? A quantitative investigation on the destination of the released linker-payload from an antibody–drug conjugate with a maleimide linker in plasma. Anal. Chem. 88(9), 4979–4986 (2016).
  • 21. Shen BQ, Bumbaca D, Saad O et al. Catabolic fate and pharmacokinetic characterization of trastuzumab emtansine (T-DM1): an emphasis on preclinical and clinical catabolism. Curr. Drug Metab. 13(7), 901–910 (2012).
  • 22. Kaur S, Alley SC, Szapacs M et al. 2021 white paper on recent issues in bioanalysis: mass spec of proteins, extracellular vesicles, CRISPR, chiral assays, oligos; nanomedicines bioanalysis; ICH M10 section 7.1; non-liquid & rare matrices; regulatory inputs (part 1A – recommendations on endogenous compounds, small molecules, complex methods, regulated mass spec of large molecules, small molecule, PoC & part 1B – regulatory agencies' inputs on bioanalysis, biomarkers, immunogenicity, gene & cell therapy and vaccine). Bioanalysis 14(9), 505–580 (2022).
  • 23. Van De Merbel NC. Protein quantification by LC–MS: a decade of progress through the pages of Bioanalysis. Bioanalysis 11(7), 629–644 (2019). •• This review article very well captures the progress and technology advancements in using LC–MS for protein quantification.
  • 24. Mehl JT, Landry F, Discenza L et al. Quantification of surrogate monoclonal antibodies in mouse serum using LC–MS/MS. Bioanalysis 13(3), 147–159 (2021).
  • 25. Sucharski FK, Meier S, Miess C et al. Development of an automated, interference-free, 2D-LC–MS/MS assay for quantification of a therapeutic mAb in human sera. Bioanalysis 10(13), 1023–1037 (2018).
  • 26. Jordan G, Onami I, Heinrich J, Staack RF. Evaluation of the potential use of hybrid LC–MS/MS for active drug quantification applying the ‘free analyte QC concept’. Bioanalysis 9(21), 1705–1717 (2017).