Skip to main content

Advertisement

Log in

An optimized approach to the rapid assessment and detection of sequence variants in recombinant protein products

  • Research Paper
  • Published:
Analytical and Bioanalytical Chemistry Aims and scope Submit manuscript

Abstract

The development of sensitive techniques to detect sequence variants (SVs), which naturally arise due to DNA mutations and errors in transcription/translation (amino acid misincorporations), has resulted in increased attention to their potential presence in protein-based biologic drugs in recent years. Often, these SVs may be below 0.1 %, adding challenges for consistent and accurate detection. Furthermore, the presence of false-positive (FP) signals, a hallmark of SV analysis, requires time-consuming analyst inspection of the data to sort true from erroneous signal. Consequently, gaps in information about the prevalence, type, and impact of SVs in marketed and in-development products are significant. Here, we report the results of a simple, straightforward, and sensitive approach to sequence variant analysis. This strategy employs mixing of two samples of an antibody or protein with the same amino acid sequence in a dilution series followed by subsequent sequence variant analysis. Using automated peptide map analysis software, a quantitative assessment of the levels of SVs in each sample can be made based on the signal derived from the mass spectrometric data. We used this strategy to rapidly detect differences in sequence variants in a monoclonal antibody after a change in process scale, and in a comparison of three mAbs as part of a biosimilar program. This approach is powerful, as true signals can be readily distinguished from FP signal, even at a level well below 0.1 %, by using a simple linear regression analysis across the data set with none to minimal inspection of the MS/MS data. Additionally, the data produced from these studies can also be used to make a quantitative assessment of relative levels of product quality attributes. The information provided here extends the published knowledge about SVs and provides context for the discussion around the potential impact of these SVs on product heterogeneity and immunogenicity.

Histogram of 97 sequence variants detected in pilot and final scale mixing study of Antibody A. All Y>F adn Y>H sequence variants had an R2 value above 0.95, while several other sequence variants confirmed in both sample types had lower R2 values. The majority of false positive and unclassified sequence variants had R2 values below 0.3

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
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

References

  1. Scott RA, Rogers R, Balland A, Brady LJ (2014) Rapid identification of an antibody DNA construct rearrangement sequence variant by mass spectrometry. mAbs 6(6):1453–1463

    Article  Google Scholar 

  2. Zhang T, Huang Y, Chamberlain S, Romeo T, Zhu-Shimoni J, Hewitt D, Zhu M, Katta V, Mauger B, Kao Y-H (2012) Identification of a single base-pair mutation of TAA (Stop codon) > GAA (Glu) that causes light chain extension in a CHO cell derived IgG1. mAbs 4(6):694–700

    Article  Google Scholar 

  3. Meert C, Brady LJ, Guo A, Balland A (2010) Characterization of antibody charge heterogeneity resolved by preparative immobilized pH gradients. Anal Chem 82:3510–3518

    Article  CAS  Google Scholar 

  4. Dorai H, Sauerwald T, Campbell A, Kyung YS, Goldstein J, Magill A, Lewis MJ, Tang QM, Jan D, Ganguly S (2007) Investigation of product microheterogeneity: a case study in rapid detection of mutation in mammalian production cell lines. Bioprocess Int 5(8):66

    CAS  Google Scholar 

  5. Harris RJ, Murnane AA, Utter SL, Wagner KLC ET, Polastri GD et al (1993) Assessing genetic heterogeneity in production cell lines: detection by peptide mapping of a low level Tyr to Gln sequence variant in a recombinant antibody. Nat Biotechnol 11:1293–1297

    Article  CAS  Google Scholar 

  6. Feeney L, Carvalhal V, Yu XC, Chan B, Michels DA, Wang YJ, Shen A, Ressl J, Dusel B, Laird MW (2012) Eliminating tyrosine sequence variants in CHO cell lines producing recombinant monoclonal antibodies. Biotechnol Bioeng 110(4):1087–1097

    Article  Google Scholar 

  7. Huang Y, O’mara B, Conover M, Ludwig R, Fu J, Tao L, Li ZJ, Rieble S, Grace MJ, Russell RJ (2012) Glycine to glutamic acid misincorporation observed in a recombinant protein expressed by Escherichia coli cells. Protein Sci 21:625–632

    Article  CAS  Google Scholar 

  8. Khetan A, Y-m H, Dolnikova J, Pederson NE, Wen D, Yusef-Makagiansar CP, Ryll T (2010) Control of misincorporation of serine for asparagine during antibody production using CHO cells. Biotechnol Bioeng 107(1):116–123

    Article  CAS  Google Scholar 

  9. Drummond DA, Wilke CO (2009) The evolutionary consequences of erroneous protein synthesis. Nat Genet 10:715–724

    Article  Google Scholar 

  10. Yang Y, Strahan A, Li C, Shen A, Liu H, Ouyang J, Katta V, Francissen K, Zhang B (2010) Detecting low level sequence variants in recombinant monoclonal antibodies. mAbs 2(3):285–298

    Article  Google Scholar 

  11. Bern M, Ki Y, Becker C (2012) Byonic: advanced peptide and protein identification software. Curr Protoc Bioinforma 40:13.20.1–13.20.14

  12. Zhang Z (2009) Large-scale identification and quantification of covalent modifications in therapeutic proteins. Anal Chem 82(20):8354–8364

    Article  Google Scholar 

  13. Zhang Z, Shah B, Bondarenko P (2013) G/U and certain wobble position mismatches as possible main causes of amino acid misincorporations. Biochemistry 52(45):8165–8176

    Article  CAS  Google Scholar 

  14. Zeck A, Regula JT, Larraillet V, Mautz B, Popp O, Gopfert U, Wiegeshoff F, Vollertsen UEE, Gorr IH, Koll H et al (2012) Low level sequence variant analysis of recombinant proteins: an optimized approach. PLoS One 7(7):1–10

    Article  Google Scholar 

  15. Fodor S, Zhang Z (2006) Rearrangement of terminal amino acid residues in peptides by protease-catalyzed intramolecular transpeptidation. Anal Biochem 356(2):282–290

    Article  CAS  Google Scholar 

  16. Bi V, Jawa V, Jourbert M, Kaliyaperumal A, Eakin C, Richmond K, Pan O, Sun J, Hokom M, Goletz T et al (2013) Development of a human antibody tolerant mouse model to assess the immunogenicity risk due to aggregated biotherapeutics. J Pharm Sci 102:3545–3555

    Article  CAS  Google Scholar 

  17. Ponce R, Abad L, Amaravadi L, Gelzleichter T, Gore E, Green J, Gupta S, Herzyk D, Hurst C, Ivens I et al (2009) Immunogenicity of biological-derived therapeutics: assessment and interpretation of nonclinical safety studies. Regul Toxicol Pharmacol 54:164–182

    Article  CAS  Google Scholar 

Download references

Acknowledgments

The authors would like to acknowledge the editorial input of Zhongqi Zhang (Amgen Product and Process Development), Gino Grampp (Amgen Strategic Planning and Operations), and Victor Fung (Amgen Biosimilar Business Unit) during the review of this paper.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Rebecca A. Scott.

Additional information

Lowell J. Brady and Rebecca A. Scott contributed equally to this work.

Electronic supplementary material

Below is the link to the electronic supplementary material.

ESM 1

(PDF 78 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brady, L.J., Scott, R.A. & Balland, A. An optimized approach to the rapid assessment and detection of sequence variants in recombinant protein products. Anal Bioanal Chem 407, 3851–3860 (2015). https://doi.org/10.1007/s00216-015-8618-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00216-015-8618-1

Keywords

Navigation