Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

GlycoDelete engineering of mammalian cells simplifies N-glycosylation of recombinant proteins

Abstract

Heterogeneity in the N-glycans on therapeutic proteins causes difficulties for protein purification and process reproducibility and can lead to variable therapeutic efficacy. This heterogeneity arises from the multistep process of mammalian complex-type N-glycan synthesis. Here we report a glycoengineering strategy—which we call GlycoDelete—that shortens the Golgi N-glycosylation pathway in mammalian cells. This shortening results in the expression of proteins with small, sialylated trisaccharide N-glycans and reduced complexity compared to native mammalian cell glycoproteins. GlycoDelete engineering does not interfere with the functioning of N-glycans in protein folding, and the physiology of cells modified by GlycoDelete is similar to that of wild-type cells. A therapeutic human IgG expressed in GlycoDelete cells had properties, such as reduced initial clearance, that might be beneficial when the therapeutic goal is antigen neutralization. This strategy for reducing N-glycan heterogeneity on mammalian proteins could lead to more consistent performance of therapeutic proteins and modulation of biopharmaceutical functions.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: GlycoDelete strategy and cell-line characterization.
Figure 2: GlycoDelete glycan characterization.
Figure 3: Functional and immunological characterization of GlycoDelete anti-CD20.

Similar content being viewed by others

References

  1. Ferrara, C. et al. Modulation of therapeutic antibody effector functions by glycosylation engineering: influence of Golgi enzyme localization domain and co-expression of heterologous β1, 4-N-acetylglucosaminyltransferase III and Golgi α-mannosidase II. Biotechnol. Bioeng. 93, 851–861 (2006).

    Article  CAS  Google Scholar 

  2. Elliott, S. et al. Control of rHuEPO biological activity: the role of carbohydrate. Exp. Hematol. 32, 1146–1155 (2004).

    Article  CAS  Google Scholar 

  3. Jacobs, P.P., Geysens, S., Vervecken, W., Contreras, R. & Callewaert, N. Engineering complex-type N-glycosylation in Pichia pastoris using GlycoSwitch technology. Nat. Protoc. 4, 58–70 (2008).

    Article  Google Scholar 

  4. Hamilton, S.R. et al. Humanization of yeast to produce complex terminally sialylated glycoproteins. Science 313, 1441–1443 (2006).

    Article  CAS  Google Scholar 

  5. Herter, S. et al. Preclinical activity of the type II CD20 antibody GA101 (obinutuzumab) compared with rituximab and ofatumumab in vitro and in xenograft models. Mol. Cancer Ther. 12, 2031–2042 (2013).

    Article  CAS  Google Scholar 

  6. Kanda, Y. et al. Comparison of cell lines for stable production of fucose-negative antibodies with enhanced ADCC. Biotechnol. Bioeng. 94, 680–688 (2006).

    Article  CAS  Google Scholar 

  7. Reeves, P.J., Callewaert, N., Contreras, R. & Khorana, H.G. Structure and function in rhodopsin: high-level expression of rhodopsin with restricted and homogeneous N-glycosylation by a tetracycline-inducible N-acetylglucosaminyltransferase I-negative HEK293S stable mammalian cell line. Proc. Natl. Acad. Sci. USA 99, 13419–13424 (2002).

    Article  CAS  Google Scholar 

  8. Stals, I. et al. Identification of a gene coding for a deglycosylating enzyme in Hypocrea jecorina. FEMS Microbiol. Lett. 303, 9–17 (2010).

    Article  CAS  Google Scholar 

  9. Grundmann, U., Nerlich, C., Rein, T. & Zettlmeissl, G. Complete cDNA sequence encoding human β-galactoside α-2,6-sialyltransferase. Nucleic Acids Res. 18, 667 (1990).

    Article  CAS  Google Scholar 

  10. Verstraete, K. et al. Structural insights into the extracellular assembly of the hematopoietic Flt3 signaling complex. Blood 118, 60–68 (2011).

    Article  CAS  Google Scholar 

  11. Bernales, S., Papa, F.R. & Walter, P. Intracellular signaling by the unfolded protein response. Annu. Rev. Cell Dev. Biol. 22, 487–508 (2006).

    Article  CAS  Google Scholar 

  12. Lee, F. et al. Isolation of cDNA for a human granulocyte-macrophage colony-stimulating factor by functional expression in mammalian cells. Proc. Natl. Acad. Sci. USA 82, 4360–4364 (1985).

    Article  CAS  Google Scholar 

  13. Forno, G. et al. N- and O-linked carbohydrates and glycosylation site occupancy in recombinant human granulocyte-macrophage colony-stimulating factor secreted by a Chinese hamster ovary cell line. Eur. J. Biochem. 271, 907–919 (2004).

    Article  CAS  Google Scholar 

  14. Crispin, M. et al. Inhibition of hybrid- and complex-type glycosylation reveals the presence of the GlcNAc transferase I-independent fucosylation pathway. Glycobiology 16, 748–756 (2006).

    Article  CAS  Google Scholar 

  15. Ericsson, U.B., Hallberg, B.M., DeTitta, G.T., Dekker, N. & Nordlund, P. Thermofluor-based high-throughput stability optimization of proteins for structural studies. Anal. Biochem. 357, 289–298 (2006).

    Article  CAS  Google Scholar 

  16. Kitamura, T. et al. Establishment and characterization of a unique human cell line that proliferates dependently on GM-CSF, IL-3, or erythropoietin. J. Cell. Physiol. 140, 323–334 (1989).

    Article  CAS  Google Scholar 

  17. Mössner, E. et al. Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell–mediated B-cell cytotoxicity. Blood 115, 4393–4402 (2010).

    Article  Google Scholar 

  18. Nallet, S. et al. Glycan variability on a recombinant IgG antibody transiently produced in HEK-293E cells. N. Biotechnol. 29, 471–476 (2012).

    Article  CAS  Google Scholar 

  19. Jefferis, R. Glycosylation as a strategy to improve antibody-based therapeutics. Nat. Rev. Drug Discov. 8, 226–234 (2009).

    Article  CAS  Google Scholar 

  20. Roopenian, D.C. & Akilesh, S. FcRn: the neonatal Fc receptor comes of age. Nat. Rev. Immunol. 7, 715–725 (2007).

    Article  CAS  Google Scholar 

  21. Lux, A., Yu, X., Scanlan, C.N. & Nimmerjahn, F. Impact of immune complex size and glycosylation on IgG binding to human FcγRs. J. Immunol. 190, 4315–4323 (2013).

    Article  CAS  Google Scholar 

  22. Hamblin, M.W. & Metcalf, M.A. Primary structure and functional characterization of a human 5–HT1D-type serotonin receptor. Mol. Pharmacol. 40, 143–148 (1991).

    CAS  PubMed  Google Scholar 

  23. Tradtrantip, L., Ratelade, J., Zhang, H. & Verkman, A.S. Enzymatic deglycosylation converts pathogenic neuromyelitis optica anti–aquaporin-4 immunoglobulin G into therapeutic antibody. Ann. Neurol. 73, 77–85 (2013).

    Article  CAS  Google Scholar 

  24. Nandakumar, K.S. et al. Dominant suppression of inflammation by glycan-hydrolyzed IgG. Proc. Natl. Acad. Sci. USA 110, 10252–10257 (2013).

    Article  CAS  Google Scholar 

  25. Allhorn, M. & Collin, M. Sugar-free antibodies—the bacterial solution to autoimmunity? Ann. NY Acad. Sci. 1173, 664–669 (2009).

    Article  CAS  Google Scholar 

  26. Anonymous. Biosimilar, biobetter and next generation therapeutic antibodies. MAbs 3, 107–110 (2011).

  27. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010).

    Article  CAS  Google Scholar 

  28. Tada, H., Shiho, O., Kuroshima, K., Koyama, M. & Tsukamoto, K. An improved colorimetric assay for interleukin 2. J. Immunol. Methods 93, 157–165 (1986).

    Article  CAS  Google Scholar 

  29. Ramsland, P.A. et al. Structural basis for Fc γRIIa recognition of human IgG and formation of inflammatory signaling complexes. J. Immunol. 187, 3208–3217 (2011).

    Article  CAS  Google Scholar 

  30. Magistrelli, G. et al. Robust recombinant FcRn production in mammalian cells enabling oriented immobilization for IgG binding studies. J. Immunol. Methods 375, 20–29 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank G. Pontini, Y. Poitevin, L. Bernasconi, D. Schrag and S. Raimondi (NovImmune) for their help with anti-CD20 generation and characterization; S. Savvides (Ghent University) for providing the 293SGnTI(−) clone expressing the Flt3 receptor extracellular domain; E. Van Damme (Ghent University) for providing ConA; and E. Dirksen and K. Nooijen (Merck) for the LC-MS analysis of anti-CD20. L.M. and M.B. are supported by predoctoral fellowships, and N.F. by a postdoctoral fellowship, of the Fund for Scientific Research-Flanders (FWO). F.S. and S.D. are supported by predoctoral fellowships of agentschap voor Innovatie door Wetenschap en Technologie (IWT) Flanders (Strategic Basic Research fellowship nos. 101456 and 111252). This research was supported by VIB, Ghent University Industrial Research Fund (UGent-IOF) Advanced Grant no. 041 (N.C.), FWO research project grant no. G.0.541.08.N.10 (N.C.) and Hercules Foundation grant no. AUGE019 (B.D.).

Author information

Authors and Affiliations

Authors

Contributions

L.M.: took the GlycoDelete technology from concept to reality: cell engineering, cell characterization, GM-CSF analytics, and thermofluor assays and co-wrote the manuscript. F.S.: made key contributions in the full exploration of the GlycoDelete technology: GM-CSF analytics, anti-CD20 analytics, and pharmacokinetics experiment, and co-wrote the manuscript. G.E.: made key contributions to the exploration of GlycoDelete technology for modulating antibody properties: experimental design, and supervision and interpretation of anti-CD20 bioanalytics experiments, and manuscript correction. N.F.: bioactivity, immunogenicity ELISA and pharmacokinetics experiments, contributed to the manuscript. M.B.: gene expression experiments and data processing. A.D.S.: ADCC, ELISA FcγR binding and CD20-binding assays. S.D.: LC-MS/MS experiments and analysis. F.R.: anti-CD20 expression construct. (This plasmid is not explicitly mentioned in the text and is an unpublished resource that was made available to us through the work of F.R.) E.P.: assisted with immunogenicity ELISA experiments. E.H.: pharmacokinetics experiment. P.M. and G.M.: SPR and BLI experiments. L. Cons and L. Chatel: pharmacokinetics experiment. B.D.: supervision of LC-MS/MS experiments. N.C.: conceived the GlycoDelete technology, initiated the project, assisted in experimental design and interpretation, and co-wrote the manuscript.

Corresponding author

Correspondence to Nico Callewaert.

Ethics declarations

Competing interests

L.M. and N.C. are inventors on patent applications covering the GlycoDelete technology (WO/2010/015722).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15, Supplementary Tables 1–8 and Supplementary Notes 1–6 (PDF 4124 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meuris, L., Santens, F., Elson, G. et al. GlycoDelete engineering of mammalian cells simplifies N-glycosylation of recombinant proteins. Nat Biotechnol 32, 485–489 (2014). https://doi.org/10.1038/nbt.2885

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.2885

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research