Skip to main content
Log in

Application of Affinity-Capture Self-Interaction Nanoparticle Spectroscopy in Predicting Protein Stability, Especially for Co-Formulated Antibodies

  • Research Paper
  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose

From traditional monoclonal antibodies to more and more complex mAb-based formulations, biopharmaceutical faces one challenge after another. To avoid these issues, identification of therapeutic proteins in the initial discovery process that has high stability and low self-interaction would simplify the development of safe and effective antibody therapeutics.

Method

Affinity-capture self-interaction nanoparticle spectroscopy (AC-SINS) is a new prediction method capable of identifying mAbs with different self-association propensity. In this study, 10 formulated monoclonal antibody (mAb) therapeutics include different mAb isotypes and co-formulated antibodies were measured by AC-SINS and some biophysical methods to predict protein stability. The prediction results of all 10 mAbs were compared to their stability data (Δ%monomer and Δ%HMWs) at accelerated (25°C and 40°C) and long-term storage conditions (4°C) as measured by size exclusion chromatography.

Result

AC-SINS method has a good predictive correlation with each mAbs and co-formulated antibodies. There were no physicochemical, intermolecular, or biological interactions that occurred between the two components of co-formulated antibodies which confirmed by Analytical ultracentrifugation (AUC).

Conclusion

Here we discuss the correlation between each method and protein stability, and also use AC-SINS assay to predict the stability of co-formulated antibodies for the first time. This may be an effective way to predict the stability of these complex mAb-based formulations such as co-formulated mAbs.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Abbreviations

AC-SINS :

Affinity-capture self-interaction nanoparticle spectroscopy

AUC :

Analytical ultracentrifugation

mAb :

Monoclonal antibody

PPI :

Protein-protein interaction

SEC :

Size-exclusion chromatography

SE :

Sedimentation equilibrium experiment

SV :

Sedimentation velocity experiment

References

  1. Kaplon H, Muralidharan M, Schneider Z, Reichert JM. Antibodies to watch in 2020. mAbs. 2020;12(1):1703531.

    Article  Google Scholar 

  2. 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.

    Article  CAS  Google Scholar 

  3. Charles S. The benefits of immunotherapy combinations. Nature. 2017;552(7685):S67–9.

    Article  Google Scholar 

  4. Larkin J, Hodi FS, Wolchok JD. Combined Nivolumab and Ipilimumab or Monotherapy in untreated melanoma. N Engl J Med. 2015;373(13):1270–1.

    Article  Google Scholar 

  5. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369(2):122–33.

    Article  CAS  Google Scholar 

  6. Woodard J, Lau H, Latypov RF. Nondenaturing size-exclusion chromatography-mass spectrometry to measure stress-induced aggregation in a complex mixture of monoclonal antibodies. Anal Chem. 2013;85(13):6429–36.

    Article  CAS  Google Scholar 

  7. Mueller C, Altenburger U, Mohl S. Challenges for the pharmaceutical technical development of protein coformulations. J Pharm Pharmacol. 2018;70(5):666–74.

    Article  CAS  Google Scholar 

  8. Ratanji KD, Derrick JP, Dearman RJ, Kimber I. Immunogenicity of therapeutic proteins: influence of aggregation. J Immunotoxicol. 2014;11(2):99–109.

    Article  CAS  Google Scholar 

  9. Singh SK. Impact of product-related factors on immunogenicity of biotherapeutics. J Pharm Sci. 2011;100(2):354–87.

    Article  CAS  Google Scholar 

  10. Raut AS, Kalonia DS. Effect of excipients on liquid-liquid phase separation and aggregation in dual variable domain immunoglobulin protein solutions. Mol Pharm. 2016;13(3):774–83.

    Article  CAS  Google Scholar 

  11. Saito S, Hasegawa J, Kobayashi N, Kishi N, Uchiyama S, Fukui K. Behavior of monoclonal antibodies: relation between the second virial coefficient (B (2)) at low concentrations and aggregation propensity and viscosity at high concentrations. Pharm Res. 2012;29(2):397–410.

    Article  CAS  Google Scholar 

  12. Thiagarajan G, Semple A, James JK, Cheung JK, Shameem M. A comparison of biophysical characterization techniques in predicting monoclonal antibody stability. mAbs. 2016;8(6):1088–97.

    Article  CAS  Google Scholar 

  13. Roberts CJ, Das TK, Sahin E. Predicting solution aggregation rates for therapeutic proteins: approaches and challenges. Int J Pharm. 2011;418(2):318–33.

    Article  CAS  Google Scholar 

  14. Larson R. The structure and rheology of complex fluids; 1998.

  15. Sule SV, Sukumar M, Weiss WF, Marcelino-Cruz AM, Sample T, Tessier PM. High-throughput analysis of concentration-dependent antibody self-association. Biophys J. 2011;101(7):1749–57.

    Article  CAS  Google Scholar 

  16. Sule SV, Dickinson CD, Lu J, Chow CK, Tessier PM. Rapid analysis of antibody self-association in complex mixtures using immunogold conjugates. Mol Pharm. 2013;10(4):1322–31.

    Article  CAS  Google Scholar 

  17. Estep P, Caffry I, Yu Y, Sun T, Cao Y, Lynaugh H, et al. An alternative assay to hydrophobic interaction chromatography for high-throughput characterization of monoclonal antibodies. mAbs. 2015;7(3):553–61.

    Article  CAS  Google Scholar 

  18. Liu Y, Caffry I, Wu J, Geng SB, Jain T, Sun T, et al. High-throughput screening for developability during early-stage antibody discovery using self-interaction nanoparticle spectroscopy. mAbs. 2014;6(2):483–92.

    Article  Google Scholar 

  19. Eftink MR. Intrinsic fluorescence of proteins. In: Lakowicz JR, editor. Topics in fluorescence spectroscopy: volume 6: protein fluorescence. Boston: Springer US; 2000. p. 1–15.

    Google Scholar 

  20. Chennamsetty N, Helk B, Voynov V, Kayser V, Trout BL. Aggregation-prone motifs in human immunoglobulin G. J Mol Biol. 2009;391(2):404–13.

    Article  CAS  Google Scholar 

  21. Kumar V, Dixit N, Zhou LL, Fraunhofer W. Impact of short range hydrophobic interactions and long range electrostatic forces on the aggregation kinetics of a monoclonal antibody and a dual-variable domain immunoglobulin at low and high concentrations. Int J Pharm. 2011;421(1):82–93.

    Article  CAS  Google Scholar 

  22. Howlett GJ, Minton AP, Rivas G. Analytical ultracentrifugation for the study of protein association and assembly. Curr Opin Chem Biol. 2006;10(5):430–6.

    Article  CAS  Google Scholar 

  23. Liu J, Andya JD, Shire SJ. A critical review of analytical ultracentrifugation and field flow fractionation methods for measuring protein aggregation. AAPS J. 2006;8(3):E580–9.

    Article  CAS  Google Scholar 

  24. Roberts C, Das T, Sahin E. Predicting solution aggregation rates for therapeutic proteins: approaches and challenges. Int J Pharm. 2011;418:318–33.

    Article  CAS  Google Scholar 

  25. Elcock AH, McCammon JA. Calculation of weak protein-protein interactions: the pH dependence of the second virial coefficient. Biophys J. 2001;80(2):613–25.

    Article  CAS  Google Scholar 

  26. Curtis RA, Prausnitz JM, Blanch HW. Protein-protein and protein-salt interactions in aqueous protein solutions containing concentrated electrolytes. Biotechnol Bioeng. 1998;57(1):11–21.

    Article  CAS  Google Scholar 

  27. Yadav S, Liu J, Shire SJ, Kalonia DS. Specific interactions in high concentration antibody solutions resulting in high viscosity. J Pharmaceutic ences. 2010;99(3):1152–68.

    CAS  Google Scholar 

  28. Neergaard MS, Kalonia DS, Parshad H, Nielsen AD. M?Ller EH, Marco VDW. Viscosity of high concentration protein formulations of monoclonal antibodies of the IgG1 and IgG4 subclass - prediction of viscosity through protein-protein interaction measurements. Eur J Pharmaceutic ences Official J Eur Federation Pharmaceutic ences. 2013;49(3):400–10.

    CAS  Google Scholar 

  29. Schuck P. On the analysis of protein self-association by sedimentation velocity analytical ultracentrifugation. Anal Biochem. 2003;320(1):104–24.

    Article  CAS  Google Scholar 

  30. Rizzo JM, Shi S, Li Y, Semple A, Esposito JJ, Yu S, et al. Application of a high-throughput relative chemical stability assay to screen therapeutic protein formulations by assessment of conformational stability and correlation to aggregation propensity. J Pharm Sci. 2015;104(5):1632–40.

    Article  CAS  Google Scholar 

  31. Jacobs SA, Wu SJ, Feng Y, Bethea D, O'Neil KT. Cross-interaction chromatography: a rapid method to identify highly soluble monoclonal antibody candidates. Pharm Res. 2010;27(1):65–71.

    Article  CAS  Google Scholar 

  32. Kohli N, Jain N, Geddie ML, Razlog M, Xu L, Lugovskoy AA. A novel screening method to assess developability of antibody-like molecules. mAbs. 2015;7(4):752–8.

    Article  CAS  Google Scholar 

  33. Wu W, Giese RF, Oss CJV. Linkage between ζ-potential and electron donicity of charged polar surfaces 1. Implications for the mechanism of flocculation of particle suspensions with plurivalent counterions. Colloids Surf A Physicochem Eng Aspects. 1994;89(2–3):241–52.

    Article  CAS  Google Scholar 

  34. Lehermayr C, Mahler HC, Mäder K, Fischer S. Assessment of net charge and protein-protein interactions of different monoclonal antibodies. J Pharm Sci. 2011;100(7):2551–62.

    Article  CAS  Google Scholar 

  35. Chari R, Jerath K, Badkar AV, Kalonia DS. Long- and short-range electrostatic interactions affect the rheology of highly concentrated antibody solutions. Pharm Res. 2009;26(12):2607–18.

    Article  CAS  Google Scholar 

  36. Scherer TM, Liu J, Shire SJ, Minton AP. Intermolecular interactions of IgG1 monoclonal antibodies at high concentrations characterized by light scattering. J Phys Chem B. 2010;114(40):12948–57.

    Article  CAS  Google Scholar 

  37. Wang X, Singh SK, Kumar S. Potential aggregation-prone regions in complementarity-determining regions of antibodies and their contribution towards antigen recognition: a computational analysis. Pharm Res. 2010;27(8):1512–29.

    Article  CAS  Google Scholar 

  38. Perchiacca JM, Bhattacharya M, Tessier PM. Mutational analysis of domain antibodies reveals aggregation hotspots within and near the complementarity determining regions. Proteins. 2011;79(9):2637–47.

    Article  CAS  Google Scholar 

Download references

Acknowledgments

This research was funded by Shanghai Hengrui Pharmaceutical Co., Ltd. The authors declare no conflicts of interest

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Xun Liu or Piaoyang Sun.

Additional information

Publisher’s Note

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

Supplementary Information

ESM 1

(DOCX 108 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, M., Yan, Z., Li, H. et al. Application of Affinity-Capture Self-Interaction Nanoparticle Spectroscopy in Predicting Protein Stability, Especially for Co-Formulated Antibodies. Pharm Res 38, 721–732 (2021). https://doi.org/10.1007/s11095-021-03026-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11095-021-03026-8

Key Words

Navigation