Skip to main content

Rapid Antibody Glycoengineering in CHO Cells Via RNA Interference and CGE-LIF N-Glycomics

  • Protocol
  • First Online:
Glycosylation

Abstract

The impact of the glycan distribution on the in vivo function and half-life of monoclonal antibodies has long motivated the genetic engineering of producer cells to achieve structures that enhance efficacy, safety and stability. To facilitate glycoengineering of IgG-producing Chinese hamster ovary cells, we present a rapid protocol that involves the use of RNA interference for the knockdown of genes of interest coupled with capillary gel electrophoresis and laser-induced fluorescence detection (CGE-LIF) for fast, high-throughput glycan analysis. We apply this methodology to the Fut8 gene, responsible for the addition of core fucose, which is a typical target for increasing antibody-dependent cellular cytotoxicity.

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

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 109.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 139.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Dalziel M, Crispin M, Scanlan CN et al (2014) Emerging principles for the therapeutic exploitation of glycosylation. Science 343:37–45

    Google Scholar 

  3. Rathore AS, Winkle H (2009) Quality by design for biopharmaceuticals. Nat Biotechnol 27:26–34

    Article  CAS  PubMed  Google Scholar 

  4. Eon-Duval A, Broly H, Gleixner R (2012) Quality attributes of recombinant therapeutic proteins: an assessment of impact on safety and efficacy as part of a quality by design development approach. Biotechnol Prog 28:608–622

    Google Scholar 

  5. Walsh G (2018) Biopharmaceutical benchmarks 2018. Nat Biotechnol 36:1136–1145

    Article  CAS  PubMed  Google Scholar 

  6. Liu L (2015) Antibody glycosylation and its impact on the pharmacokinetics and pharmacodynamics of monoclonal antibodies and Fc-fusion proteins. J Pharm Sci 104:1866–1884

    Article  CAS  PubMed  Google Scholar 

  7. Clynes R, Takechi Y, Moroi Y et al (1998) Fc receptors are required in passive and active immunity to melanoma. Proc Natl Acad Sci U S A 95:652–656

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Clynes RA, Towers TL, Presta LG, Ravetech J v. (2000) Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat Med 6:443–446

    Article  CAS  PubMed  Google Scholar 

  9. Yu X, Baruah K, Harvey DJ et al (2013) Engineering hydrophobic protein-carbohydrate interactions to fine-tune monoclonal antibodies. J Am Chem Soc 135:9723–9732

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gerngross TU (2004) Advances in the production of human therapeutic proteins in yeasts and filamentous fungi. Nat Biotechnol 22:1409–1414

    Article  CAS  PubMed  Google Scholar 

  11. Iida S, Misaka H, Inoue M et al (2006) Nonfucosylated therapeutic IgG1 antibody can evade the inhibitory effect of serum immunoglobulin G on antibody-dependent cellular cytotoxicity through its high binding to FcgammaRIIIa. Clin Cancer Res 12:2879–2887

    Article  CAS  PubMed  Google Scholar 

  12. Ito A, Ishida T, Yano H et al (2009) Defucosylated anti-CCR4 monoclonal antibody exercises potent ADCC-mediated antitumor effect in the novel tumor-bearing humanized NOD/Shi-scid, IL-2Rgamma (null) mouse model. Cancer Immunol Immunother 58:1195–1206

    Article  CAS  PubMed  Google Scholar 

  13. Kanda Y, Imai-Nishiya H, Kuni-Kamochi R et al (2007) Establishment of a GDP-mannose 4,6-dehydratase (GMD) knockout host cell line: a new strategy for generating completely non-fucosylated recombinant therapeutics. J Biotechnol 130:300–310

    Article  CAS  PubMed  Google Scholar 

  14. Li H, Sethuraman N, Stadheim TA et al (2006) Optimization of humanized IgGs in glycoengineered Pichia pastoris. Nat Biotechnol 24:210–215

    Article  CAS  PubMed  Google Scholar 

  15. Mori K, Kuni-Kamochi R, Yamane-Ohnuki N et al (2004) Engineering Chinese hamster ovary cells to maximize effector function of produced antibodies using FUT8 siRNA. Biotechnol Bioeng 88:901–908

    Article  CAS  PubMed  Google Scholar 

  16. Natsume A, Wakitani M, Yamane-Ohnuki N et al (2005) Fucose removal from complex-type oligosaccharide enhances the antibody-dependent cellular cytotoxicity of single-gene-encoded antibody comprising a single-chain antibody linked the antibody constant region. J Immunol Methods 306:93–103

    Article  CAS  PubMed  Google Scholar 

  17. Okazaki A, Shoji-Hosaka E, Nakamura K et al (2004) Fucose depletion from human IgG1 oligosaccharide enhances binding enthalpy and association rate between IgG1 and Fc-gammaRIIIa. J Mol Biol 336:1239–1249

    Article  CAS  PubMed  Google Scholar 

  18. Rothman RJ, Perussia B, Herlyn D, Warren L (1989) Antibody-dependent cytotoxicity mediated by natural killer cells is enhanced by castanospermine-induced alterations of IgG glycosylation. Mol Immunol 26:1113–1123

    Article  CAS  PubMed  Google Scholar 

  19. Satoh M, Iida S, Shitara K (2006) Non-fucosylated therapeutic antibodies as next-generation therapeutic antibodies. Expert Opin Biol Ther 6:1161–1173

    Article  CAS  PubMed  Google Scholar 

  20. Shields RL, Lai J, Keck R et al (2002) Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human FcγRIII and antibody-dependent cellular toxicity. J Biol Chem 277:26733–26740

    Article  CAS  PubMed  Google Scholar 

  21. Yamane-Ohnuki N, Kinoshita S, Inoue-Urakubo M et al (2004) Establishment of FUT8 knockout Chinese hamster ovary cells: an ideal host cell line for producing completely defucosylated antibodies with enhanced antibody-dependent cellular cytotoxicity. Biotechnol Bioeng 87:614–622

    Article  CAS  PubMed  Google Scholar 

  22. Winkelhake JL, Nicolson GL (1976) Aglycosylantibody. Effects of exoglycosidase treatments on autochthonous antibody survival time in the circulation. J Biol Chem 251:1074–1080

    Article  CAS  PubMed  Google Scholar 

  23. Wright A, Sato Y, Okada T et al (2000) In vivo trafficking and catabolism of IgG1 antibodies with Fc associated carbohydrates of differing structure. Glycobiology 10:1347–1355

    Article  CAS  PubMed  Google Scholar 

  24. Boyd PN, Lines AC, Patel AK (1995) The effect of the removal of sialic acid, galactose and total carbohydrate on the functional activity of Campath-1H. Mol Immunol 32:1311–1318

    Article  CAS  PubMed  Google Scholar 

  25. Hodoniczky J, Zheng YZ, James DC (2005) Control of recombinant monoclonal antibody effector functions by Fc N-glycan remodelling in vitro. Biotechnol Prog 21:1644–1652

    Article  CAS  PubMed  Google Scholar 

  26. Peipp M, Dechant M, Valerius T (2008) Effector mechanisms of therapeutic antibodies against ErbB receptors. Curr Opin Immunol 20:436–443

    Article  CAS  PubMed  Google Scholar 

  27. Kumpel BM, Rademacher TW, Rook GA et al (1994) Galactosylation of human IgG monoclonal anti-D produced by EBV-transformed B-lymphoblastoid cell lines is dependent on culture method and affects Fc receptor-mediated functional activity. Hum Antib Hybrid 5:143–151

    CAS  Google Scholar 

  28. Kumpel BM, Wang Y, Griffiths HL et al (1995) The biological activity of human monoclonal IgG anti-D is reduced by β-galactosidase treatment. Hum Antib Hybrid 6:82–88

    CAS  Google Scholar 

  29. Leader KA, Kumpel BM, Hadley AG, Bradley BA (1991) Functional interactions of aglycosylated monoclonal anti-D with Fc gamma RI+ and Fc gamma RIII+ cells. Immunology 72:481–485

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Thomann M, Reckermann K, Reusch D et al (2016) Fc-galactosylation modulates antibody-dependent cellular cytotoxicity of therapeutic antibodies. Mol Immunol 73:69–75

    Article  CAS  PubMed  Google Scholar 

  31. Chung CH, Mirakhur B, Chan E et al (2008) Cetuximab-induced anaphylaxis and IgE specific for galactose-α-1,3-galactose. N Engl J Med 358:1109–1126

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. O’Neil BH, Allen R, Spigel DR et al (2007) High incidence of cetuximab-related infusion reactions in Tennessee and North Carolina and the association with atopic history. J Clin Oncol 25:3644–3648

    Article  PubMed  Google Scholar 

  33. Umaña P, Jean-Mairet J, Moudry R et al (1999) Engineered glycoforms of an antineuro-blastoma IgG1 with optimized antibody-dependent cellular cytotoxic activity. Nat Biotechnol 17:176–180

    Article  PubMed  Google Scholar 

  34. Shinkawa T, Nakamura K, Yamane N et al (2003) The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. J Biol Chem 278:3466–3473

    Article  CAS  PubMed  Google Scholar 

  35. Jones AJ, Papac DI, Chin EH et al (2007) Selective clearance of glycoforms of a complex glycoprotein pharmaceutical caused by terminal N-acetylglucosamine is similar in humans and cynomolgus monkeys. Glycobiology 17:529–540

    Article  CAS  PubMed  Google Scholar 

  36. Goetze AM, Liu YD, Zhang Z et al (2011) High-mannose glycans on the Fc region of therapeutic IgG antibodies increase serum clearance in humans. Glycobiology 21:949–959

    Article  CAS  PubMed  Google Scholar 

  37. Kanda Y, Yamada T, Mori K et al (2007) Comparison of biological activity among nonfucosylated therapeutic IgG1 antibodies with three different N-linked Fc oligosaccharides: the high-mannose, hybrid, and complex types. Glycobiology 17:104–118

    Article  CAS  PubMed  Google Scholar 

  38. Liu L, Stadheim A, Hamuro L et al (2011) Pharmacokinetics of IgG1 monoclonal antibodies produced in humanized Pichia pastoris with specific glycoforms: a comparative study with CHO produced materials. Biologicals 39:205–210

    Article  CAS  PubMed  Google Scholar 

  39. Wright A, Morrison SL (1994) Effect of altered CH2-associated carbohydrate structure on the functional properties and in vivo fate of chimeric mouse-human immunoglobulin G1. J Exp Med 180:1087–1096

    Article  CAS  PubMed  Google Scholar 

  40. Yu M, Brown D, Reed C et al (2012) Production, characterization, and pharmacokinetic properties of antibodies with N-linked mannose-5 glycans. mAbs 4:475–487

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  42. Zhou Q, Shankara S, Roy A et al (2008) Development of a simple and rapid method for producing non-fucosylated oligomannose containing antibodies with increased effector function. Biotechnol Bioeng 99:652–665

    Article  CAS  PubMed  Google Scholar 

  43. Hiatt A, Bohorova N, Bohorov O et al (2014) Glycan variants of a respiratory syncytial virus antibody with enhanced effector function and in vivo efficacy. Proc Natl Acad Sci U S A 111:5992–5997

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Anthony RM, Nimmerjahn F, Ashline DJ et al (2008) Recapitulation of IVIG anti-inflammatory activity with a recombinant IgG Fc. Science 320:369–373

    Article  Google Scholar 

  45. Flesher AR, Marzowski J, Wang WC, Raff HV (1995) Fluorophore-labelled carbohydrate analysis of immunoglobulin fusion proteins: correlation of oligosaccharide content with in vivo clearance profile. Biotechnol Bioeng 47:405

    Article  CAS  PubMed  Google Scholar 

  46. Kim HJ, Kim HJ (2007) The glycosylation and pharmacokinetics of CTLA4Ig produced in rice cells. Biol Pharm Bull 30:1913–1917

    Article  CAS  PubMed  Google Scholar 

  47. Liu L, Gomathinayagam S, Hamuro L et al (2013) The impact of glycosylation on the pharmacokinetics of a TNFR2:Fc fusion protein expressed in glycoengineered Pichia pastoris. Pharm Res 30:803–812

    Article  CAS  PubMed  Google Scholar 

  48. Meier W, Gill A, Rogge M et al (1995) Immunomodulation by LFA3TIP, an LFA-3/IgG1 fusion protein: cell line dependent glycosylation effects on pharmacokinetics and pharmacodynamic markers. Ther Immunol 2:159–171

    CAS  PubMed  Google Scholar 

  49. Stefanich EG, Ren S, Danilenko DM et al (2008) Evidence for an asialoglycoprotein receptor on nonparenchymal cells for O-linked glycoproteins. J Pharmacol Exp Ther 327:308–315

    Article  CAS  PubMed  Google Scholar 

  50. Suen KF, Turner MS, Gao F et al (2010) Transient expression of an IL-23R extracellular domain Fc fusion protein in CHO vs. HEK cells results in improved plasma exposure. Protein Expr Purif 71:96–102

    Article  CAS  PubMed  Google Scholar 

  51. Anthony RM, Ravetch J v. (2010) A novel role for the IgG Fc glycan: the anti-inflammatory activity of sialylated IgG Fcs. J Clin Immunol 30:9–14

    Article  Google Scholar 

  52. Anthony RM, Kobayashi T, Wermeling F, Ravetch J v. (2011) Intravenous gammaglobulin suppresses inflammation through a novel T(H)2 pathway. Nature 475:110–113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kaneko Y, Nimmerjahn F, Ravetch J v. (2006) Anti-inflammatory activity of immunoglobulin G resulting from fc sialylation. Science 313:670–673

    Article  CAS  PubMed  Google Scholar 

  54. Nimmerjahn F, Ravetch J v. (2008) Anti-inflammatory actions of intravenous immunoglobulin. Annu Rev Immunol 26:513–533

    Article  CAS  PubMed  Google Scholar 

  55. Samuelsson A, Towers TL, Ravetch JV (2001) Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science 291:484–486

    Article  CAS  PubMed  Google Scholar 

  56. Sondermann P, Pincetic A, Maamary J et al (2013) General mechanism for modulating immunoglobulin effector function. Proc Natl Acad Sci U S A 110:9868–9872

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Scallon BJ, Tam SH, McCarthy SG et al (2007) Higher levels of sialylated Fc glycans in immunoglobulin G molecules can adversely impact functionality. Mol Immunol 44:1524–1534

    Article  CAS  PubMed  Google Scholar 

  58. Ghaderi D, Taylor RE, Padler-Karavani V et al (2010) Implications of the presence of N-glycolylneuraminic acid in recombinant therapeutic glycoproteins. Nat Biotechnol 28:863–867

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ghaderi D, Zhang M, Hurtado-Ziola N, Varki A (2012) Production platforms for biotherapeutic glycoproteins. Occurrence, impact, and challenges of non-human sialylation. Biotechnol Genet Eng Rev 28:147–175

    Article  CAS  PubMed  Google Scholar 

  60. Tangvoranuntakul P, Gagneux P, Diaz S et al (2003) Human uptake and incorporation of an immunogenic nonhuman dietary sialic acid. Proc Natl Acad Sci U S A 100:12045–12050

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Tejwani V, Andersen MR, Nam JH, Sharfstein ST (2018) Glycoengineering in CHO cells: advances in systems biology. Biotechnol J 13:e1700234

    Article  PubMed  Google Scholar 

  62. Amann T, Schmieder V, Kildegaard HF et al (2019) Genetic engineering approaches to improve posttranslational modification of biopharmaceuticals in different production platforms. Biotechnol Bioeng 116:2778–2796

    Article  CAS  PubMed  Google Scholar 

  63. Xu X, Nagarajan H, Lewis NE et al (2011) The genomic sequence of the Chinese hamster ovary (CHO)-K1 cell line. Nat Biotechnol 29:735–741

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Rupp O, MacDonald ML, Li S et al (2018) A reference genome of the Chinese hamster based on a hybrid assembly strategy. Biotechnol Bioeng 115:2087–2100

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Stolfa G, Smonskey MT, Boniface R et al (2018) CHO-omics review: The impact of current and emerging technologies on Chinese hamster ovary based bioproduction. Biotechnol J 13:e1700227

    Article  PubMed  Google Scholar 

  66. Fire A, Xu S, Montgomery MK et al (1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391:806–811

    Article  CAS  PubMed  Google Scholar 

  67. Hammond SM, Bernstein E, Beach D, Hannon GJ (2000) An RNA-directed nuclease mediates post-transcriptional gene silencing in drosophila cells. Nature 404:293–296

    Article  CAS  PubMed  Google Scholar 

  68. Bernstein E, Caudy AA, Hammond SM, Hannon GJ (2001) Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 409:363–366

    Article  CAS  PubMed  Google Scholar 

  69. Szigeti M, Chapman J, Borza B, Guttman A (2018) Quantitative assessment of mAb Fc glycosylation of CQA importance by capillary electrophoresis. Electrophoresis 39:2340–2343

    Article  CAS  PubMed  Google Scholar 

  70. Zhang X, Lok SHL, Kon L (1998) Stable expression of human alpha-2,6-sialyltransferase in Chinese hamster ovary cells: functional consequences for human erythropoietin expression and bioactivity. Biochim Biophys Acta 1425:441–452

    Article  CAS  PubMed  Google Scholar 

  71. Weikert S, Papac D, Briggs J et al (1999) Engineering Chinese hamster ovary cells to maximize sialic acid content of recombinant glycoproteins. Nat Biotechnol 17:1116–1121

    Article  CAS  PubMed  Google Scholar 

  72. Fukuta K, Yokomatsu T, Abe R et al (2000) Genetic engineering of CHO cells producing human interferon-gamma by transfection of sialyltransferases. Glycoconj J 17:895–904

    Article  CAS  PubMed  Google Scholar 

  73. Ngantung FA, Miller PG, Brushett FR et al (2006) RNA interference of sialidase improves glycoprotein sialic acid content consistency. Biotechnol Bioeng 95:106–119

    Article  CAS  PubMed  Google Scholar 

  74. Jeong YT, Choi O, Lim HR et al (2008) Enhanced sialylation of recombinant erythropoietin in CHO cells by human glycosyltransferase expression. J Microbiol Biotechnol 18:1945–1952

    CAS  PubMed  Google Scholar 

  75. Zhang M, Koskie K, Ross JS et al (2010) Enhancing glycoprotein sialylation by targeted gene silencing in mammalian cells. Biotechnol Bioeng 105:1094–1105

    CAS  PubMed  Google Scholar 

  76. Son YD, Jeong YT, Park SY, Kim JH (2011) Enhanced sialylation of recombinant human erythropoietin in Chinese hamster ovary cells by combinatorial engineering of selected genes. Glycobiology 21:1019–1028

    Article  CAS  PubMed  Google Scholar 

  77. Chung C-Y, Yin B, Wang Q et al (2015) Assessment of the coordinated role of ST3GAL3, ST3GAL4 and ST3GAL6 on the α2,3 sialylation linkage of mammalian glycoproteins. Biochem Biophys Res Commun 463:211–215

    Article  CAS  PubMed  Google Scholar 

  78. Raymond C, Robotham A, Spearman M et al (2015) Production of α2,6-sialylated IgG1 in CHO cells. mAbs 7:571–583

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Yin B, Gao Y, Chung C-Y et al (2015) Glycoengineering of Chinese hamster ovary cells for enhanced erythropoietin N-glycan branching and sialylation. Biotechnol Bioeng 112:2343–2349

    Article  CAS  PubMed  Google Scholar 

  80. Chung CY, Wang Q, Yang S et al (2017) Integrated genome and protein editing swaps α-2,6 sialylation for α-2,3 sialic acid on recombinant antibodies from CHO. Biotechnol J 12:1–12

    Google Scholar 

  81. Amann T, Hansen AH, Kol S et al (2018) CRISPR/Cas9-multiplexed editing of Chinese hamster ovary B4Gal-T1, 2, 3, and 4 tailors N-glycan profiles of therapeutics and secreted host cell proteins. Biotechnol J 13

    Google Scholar 

  82. Bydlinski N, Maresch D, Schmieder V et al (2018) The contributions of individual galactosyltransferases to protein specific N-glycan processing in Chinese hamster ovary cells. J Biotechnol 282:101–110

    Article  CAS  PubMed  Google Scholar 

  83. Lee CG, Oh MJ, Park SY et al (2018) Inhibition of poly-LacNAc biosynthesis with release of CMP-Neu5Ac feedback inhibition increases the sialylation of recombinant EPO produced in CHO cells. Sci Rep 8:1–9

    Google Scholar 

  84. Marx N, Grünwald-Gruber C, Bydlinski N et al (2018) CRISPR-based targeted epigenetic editing enables gene expression modulation of the silenced beta-galactoside alpha-2,6-sialyltransferase 1 in CHO cells. Biotechnol J 13:1–11

    Google Scholar 

  85. Amann T, Hansen AH, Kol S et al (2019) Glyco-engineered CHO cell lines producing alpha-1-antitrypsin and C1 esterase inhibitor with fully humanized N-glycosylation profiles. Metab Eng 52:143–152

    Article  CAS  PubMed  Google Scholar 

  86. la Cour Karottki KJ, Hefzi H, Xiong K et al (2020) Awakening dormant glycosyltransferases in CHO cells with CRISPRa. Biotechnol Bioeng 117:593–598

    Article  Google Scholar 

  87. Ferrara C, Grau S, Jäger C et al (2011) Unique carbohydrate-carbohydrate interactions are required for high affinity binding between FcγRIII and antibodies lacking core fucose. Proc Natl Acad Sci U S A 108:12669–12674

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Imai-Nishiya H, Mori K, Inoue M et al (2007) Double knockdown of α1,6-fucosyltransferase (FUT8) and GDP-mannose 4,6-dehydratase (GMD) in antibody-producing cells: a new strategy for generating fully non-fucosylated therapeutic antibodies with enhanced ADCC. BMC Biotechnol 7:84

    Article  PubMed  PubMed Central  Google Scholar 

  89. Beuger V, Künkele K-P, Koll H et al (2009) Short-hairpin-RNA-mediated silencing of fucosyltransferase 8 in Chinese-hamster ovary cells for the production of antibodies with enhanced antibody immune effector function. Biotechnol Appl Biochem 53:31–37

    Article  CAS  PubMed  Google Scholar 

  90. Malphettes L, Freyvert Y, Chang J et al (2010) Highly efficient deletion of FUT8 in CHO cell lines using zinc-finger nucleases yields cells that produce completely nonfucosylated antibodies. Biotechnol Bioeng 106:774–783

    Article  CAS  PubMed  Google Scholar 

  91. von Horsten HH, Ogorek C, Blanchard V et al (2010) Production of non-fucosylated antibodies by co-expression of heterologous GDP-6-deoxy-D-lyxo-4-hexulose reductase. Glycobiology 20:1607–1618

    Article  Google Scholar 

  92. Zhang P, Haryadi R, Chan KF et al (2012) Identification of functional elements of the GDP-fucose transporter SLC35C1 using a novel Chinese hamster ovary mutant. Glycobiology 22:897–911

    Article  CAS  PubMed  Google Scholar 

  93. Ronda C, Pedersen LE, Hansen HG et al (2014) Accelerating genome editing in CHO cells using CRISPR Cas9 and CRISPy, a web-based target finding tool. Biotechnol Bioeng 111:1604–1616

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Grav LM, Lee JS, Gerling S et al (2015) One-step generation of triple knockout CHO cell lines using CRISPR/Cas9 and fluorescent enrichment. Biotechnol J 10:1446–1456

    Article  CAS  PubMed  Google Scholar 

  95. Sun T, Li C, Han L et al (2015) Functional knockout of FUT8 in Chinese hamster ovary cells using CRISPR/Cas9 to produce a defucosylated antibody. Eng Life Sci 15:660–666

    Article  CAS  Google Scholar 

  96. Yang Z, Wang S, Halim A et al (2015) Engineered CHO cells for production of diverse, homogeneous glycoproteins. Nat Biotechnol 33:842–844

    Article  CAS  PubMed  Google Scholar 

  97. Chan KF, Shahreel W, Wan C et al (2016) Inactivation of GDP-fucose transporter gene (Slc35c1) in CHO cells by ZFNs, TALENs and CRISPR-Cas9 for production of fucose-free antibodies. Biotechnol J 11:399–414

    Article  CAS  PubMed  Google Scholar 

  98. Chung CY, Wang Q, Yang S et al (2017) Combinatorial genome and protein engineering yields monoclonal antibodies with hypergalactosylation from CHO cells. Biotechnol Bioeng 114:2848–2856

    Article  CAS  PubMed  Google Scholar 

  99. Kelly RM, Kowle RL, Lian Z et al (2018) Modulation of IgG1 immunoeffector function by glycoengineering of the GDP-fucose biosynthesis pathway. Biotechnol Bioeng 115:705–718

    Article  CAS  PubMed  Google Scholar 

  100. Luo C, Chen S, Xu N et al (2017) Glycoengineering of pertuzumab and its impact on the pharmacokinetic/pharmacodynamic properties. Sci Rep 7:46347

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Roy G, Martin T, Barnes A et al (2018) A novel bicistronic gene design couples stable cell line selection with a fucose switch in a designer CHO host to produce native and afucosylated glycoform antibodies. mAbs 10:416–430

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Schmieder V, Bydlinski N, Strasser R et al (2018) Enhanced genome editing tools for multi-gene deletion knock-out approaches using paired CRISPR sgRNAs in CHO cells. Biotechnol J 13:1700211

    Article  Google Scholar 

  103. Chang MM, Gaidukov L, Jung G et al (2019) Small-molecule control of antibody N-glycosylation in engineered mammalian cells. Nat Chem Biol 15:730–736

    Article  CAS  PubMed  Google Scholar 

  104. Yuan Y, Zong H, Bai J et al (2019) Bioprocess development of a stable FUT8−/−-CHO cell line to produce defucosylated anti-HER2 antibody. Bioprocess Biosyst Eng 42:1263–1271

    Article  CAS  PubMed  Google Scholar 

  105. Mishra N, Spearman M, Donald L et al (2020) Comparison of two glycoengineering strategies to control the fucosylation of a monoclonal antibody. J Biotechnol X 5:100015

    Article  CAS  Google Scholar 

Download references

Acknowledgments

PK thanks the Department of Chemical Engineering, Imperial College London, for his scholarship. RD thanks the U.K. Biotechnology and Biological Sciences Research Council for his studentship. MM is funded by the U.K. Biotechnology and Biological Sciences Research Council (Grant reference: BB/S006206/1). IAG thanks the Irish Research Council (Scholarship No. GOIPG/2017/1049) and CONACyT (Scholarship No. 438330).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Cleo Kontoravdi .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2022 Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Kotidis, P. et al. (2022). Rapid Antibody Glycoengineering in CHO Cells Via RNA Interference and CGE-LIF N-Glycomics. In: Davey, G.P. (eds) Glycosylation. Methods in Molecular Biology, vol 2370. Humana, New York, NY. https://doi.org/10.1007/978-1-0716-1685-7_7

Download citation

  • DOI: https://doi.org/10.1007/978-1-0716-1685-7_7

  • Published:

  • Publisher Name: Humana, New York, NY

  • Print ISBN: 978-1-0716-1684-0

  • Online ISBN: 978-1-0716-1685-7

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics