Skip to main content

Development, validation, and implementation of a robust and quality control-friendly focused peptide mapping method for monitoring oxidation of co-formulated monoclonal antibodies

Abstract

Monoclonal antibody (mAb) coformulation containing two therapeutic proteins provides benefits of improved therapeutic efficacy and better patient compliance. Monitoring of the individual mAb stability in the coformulation is critical to ensure its quality and safety. Among post-translational modifications (PTMs), oxidation is often considered as one of the critical quality attributes (CQAs) as it potentially affects the structure and potency. Although hydrophobic interaction chromatography (HIC) and reversed phase liquid chromatography (RPLC) have been used to monitor overall protein oxidation, mass spectrometry of peptide digests resolved by LC methods can afford superior selectivity and sensitivity for specific PTMs. With the advent of the Quadrupole Dalton (QDa) mass spectrometer as an affordable add-on detector, implementation of targeted oxidation assays in development and quality control (QC) laboratories is now feasible. In this study, as the first effort to implement MS-based methods for antibody coformulation in QC laboratories, we developed and validated a high-throughput and robust focused peptide mapping method using QDa for simultaneous site-specific monitoring of oxidation of methionine and tryptophan residues in heavy-chain (HC) complementary determining regions (CDRs) of two co-formulated mAbs. The method was validated in terms of accuracy, precision, linearity, range, quantitation limit (QL), specificity, and solution stability per recommendations in ICH Q2. The method robustness was systematically assessed involving multiple sample preparation and instrument method parameters. The method met the validation criteria in GMP laboratories with excellent robustness and was implemented in both GMP and development environments.

Graphical abstract

This is a preview of subscription content, access via your institution.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

References

  1. Drake CG. Combination immunotherapy approaches. Annals of Oncology. 2012;23(Supplement 8):viii41–6.

  2. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348(6230):56–61.

    Article  CAS  PubMed  Google Scholar 

  3. Ferreira MN, Choe JH. Guiding immunotherapy combinations: who gets what? Adv Drug Deliv Rev. 2021;178: 113962.

    Article  CAS  PubMed  Google Scholar 

  4. Schmidt C. Combinations on trial. Nature. 2007;552:S67–9.

    Article  Google Scholar 

  5. Wang SS, Yan Y, Ho K. US FDA-approved therapeutic antibodies with high-concentration formulation: summaries and perspectives. Antibody Therapeutics. 2021;4(4):262–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhu S, Zhang T, Zheng L, Liu H, Song W, Liu D, et al. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol. 2021;14(1):156.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Allmendinger A. Opportunities in an evolving pharmaceutical development landscape: product differentiation of biopharmaceutical drug products. Pharm Res. 2021;38(5):739–57.

    Article  CAS  PubMed  Google Scholar 

  8. Luo L, Jiang B, Cao Y, Xu L, Shameem M, Liu D. A hydrophobic interaction chromatography method suitable for quantitating individual monoclonal antibodies contained in co-formulated drug products. J Pharm Biomed Anal. 2021;193: 113703.

    Article  CAS  PubMed  Google Scholar 

  9. Queiroz JA, Tomaz CT, Cabral JM. Hydrophobic interaction chromatography of proteins. J Biotechnol. 2001;87(2):143.

  10. Geng X, Wang L. Liquid chromatography of recombinant proteins and protein drugs. J Chromatogr B Analyt Technol Biomed Life Sci. 2008;866(1–2):133.

  11. Boyd D, Kaschak T, Yan B. HIC resolution of an IgG1 with an oxidized Trp in a complementarity determining region. J Chromatogr B Analyt Technol Biomed Life Sci. 2011;879(879):955.

    Article  CAS  PubMed  Google Scholar 

  12. Yang J, Wang S, Liu J, Raghani A. Determination of tryptophan oxidation of monoclonal antibody by reversed phase high performance liquid chromatography. J Chromatogr A. 2007;1156(1):174–82.

    Article  CAS  PubMed  Google Scholar 

  13. Haverick M, Mengisen S, Shameem M, Ambrogelly A. Separation of mAbs molecular variants by analytical hydrophobic interaction chromatography HPLC: overview and applications. mAbs. 2014;6(4):852–8.

  14. Jenkins N, Murphy L, Tyther R. Post-translational modifications of recombinant proteins: significance for biopharmaceuticals. Mol Biotechnol. 2008;39(2):113–8.

    Article  CAS  PubMed  Google Scholar 

  15. Millán-Martín S, Jakes C, Carillo S, Buchanan T, Guender M, Kristensen DB, et al. Inter-laboratory study of an optimised peptide mapping workflow using automated trypsin digestion for monitoring monoclonal antibody product quality attributes. Anal Bioanal Chem. 2020;412(25):6833–48.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Bu X, Yang J, Gong X, Welch CJ. Evaluation of a compact mass spectrometer for routine support of pharmaceutical chemistry. J Pharm Biomed Anal. 2014;94:139–44.

    Article  CAS  PubMed  Google Scholar 

  17. Malcolm A, Wright S, Syms RRA, Moseley R, O’Prey S, Dash N, et al. A miniature mass spectrometer for liquid chromatography applications. Rapid Commun Mass Spectrom. 2011;25:3281.

    Article  CAS  PubMed  Google Scholar 

  18. Bu X, Regalado EL, Hamilton SE, Welch CJ. The emergence of low-cost compact mass spectrometry detectors for chromatographic analysis. TrAC, Trends Anal Chem. 2016;82:22–34.

    Article  CAS  Google Scholar 

  19. Xu W, Jimenez RB, Mowery R, Luo H, Cao M, Agarwal N, et al. A Quadrupole Dalton-based multi-attribute method for product characterization, process development, and quality control of therapeutic proteins. mAbs. 2017;9:1186.

  20. Cao X, Flagg SC, Li X, Chennamsetty N, Balakrishnan G, Das TK. Quadrupole Dalton-based controlled proteolysis method for characterization of higher order protein structure. Anal Chem. 2019;91(8):5339–45.

    Article  CAS  PubMed  Google Scholar 

  21. Evans AR, Hebert AS, Mulholland J, Lewis MJ, Hu P. ID–MAM: a validated identity and multi-attribute monitoring method for commercial release and stability testing of a bispecific antibody. Anal Chem. 2021;93(26):9166–73.

    Article  CAS  PubMed  Google Scholar 

  22. Cao M, De Mel N, Shannon A, Prophet M, Wang C, Xu W, et al. Charge variants characterization and release assay development for co-formulated antibodies as a combination therapy. mAbs. 2019;11(3):489–99.

  23. Delmar JA, Buehler E, Chetty AK, Das A, Quesada GM, Wang J, et al. Machine learning prediction of methionine and tryptophan photooxidation susceptibility. Molecular Therapy - Methods & Clinical Development. 2021;21:466–77.

    Article  CAS  Google Scholar 

  24. Saveliev S, Bratz M, Zubarev R, Szapacs M, Budamgunta H, Urh M. Trypsin/Lys-C protease mix for enhanced protein mass spectrometry analysis. Nat Methods. 2013;10(11):i–ii.

    Article  Google Scholar 

  25. Huesgen PF, Lange PF, Rogers LD, Solis N, Eckhard U, Kleifeld O, et al. LysargiNase mirrors trypsin for protein C-terminal and methylation-site identification. Nat Methods. 2015;12(1):55–8.

    Article  CAS  PubMed  Google Scholar 

  26. Shibue M, Mant CT, Hodges RS. Effect of anionic ion-pairing reagent concentration (1–60mM) on reversed-phase liquid chromatography elution behaviour of peptides. J Chromatogr A. 2005;1080(1):58–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Xu C, Guo H, Breitbach ZS, Armstrong DW. Mechanism and sensitivity of anion detection using rationally designed unsymmetrical dications in paired ion electrospray ionization mass spectrometry. Anal Chem. 2014;86(5):2665–72.

    Article  CAS  PubMed  Google Scholar 

  28. Toole EN, Dufresne C, Ray S, Schwann A, Cook K, Ivanov AR. Rapid highly-efficient digestion and peptide mapping of adeno-associated viruses. Anal Chem. 2021;93(30):10403–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Li X, Rawal B, Rivera S, Letarte S, Richardson DD. Improvements on sample preparation and peptide separation for reduced peptide mapping based multi-attribute method analysis of therapeutic monoclonal antibodies using lysyl endopeptidase digestion. J Chromatogr A. 2022;1675: 463161.

    Article  CAS  PubMed  Google Scholar 

  30. Nie S, Greer T, Huang X, Zheng X, Li N. Development of a simple non-reduced peptide mapping method that prevents disulfide scrambling of mAbs without affecting tryptic enzyme activity. J Pharm Biomed Anal. 2022;209: 114541.

    Article  CAS  PubMed  Google Scholar 

  31. Bults P, Bischoff R, Bakker H, Gietema JA, van de Merbel NC. LC-MS/MS-based monitoring of in vivo protein biotransformation: quantitative determination of trastuzumab and its deamidation products in human plasma. Anal Chem. 2016;88(3):1871–7.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Dr. Michiel Waterbeemd, Dr. Alexander J. Pavon, and Dr. Petra Bennington for their support and helpful discussions.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Chengdong Xu.

Ethics declarations

Conflict of interest

The authors declare no competing interests.

Additional information

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 158 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and Permissions

About this article

Verify currency and authenticity via CrossMark

Cite this article

Xu, C., Khanal, S., Pierson, N.A. et al. Development, validation, and implementation of a robust and quality control-friendly focused peptide mapping method for monitoring oxidation of co-formulated monoclonal antibodies. Anal Bioanal Chem 414, 8317–8330 (2022). https://doi.org/10.1007/s00216-022-04366-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00216-022-04366-z

Keywords

  • Co-formulated monoclonal antibodies
  • LC–MS
  • Bioanalytical methods
  • Protein oxidation
  • Focused peptide mapping
  • Method robustness