About

Log in?

DTU users get better search results including licensed content and discounts on order fees.

Anyone can log in and get personalized features such as favorites, tags and feeds.

Log in as DTU user Log in as non-DTU user No thanks

DTU Findit

Journal article

Glyco-engineered CHO cell lines producing alpha-1-antitrypsin and C1 esterase inhibitor with fully humanized N-glycosylation profiles

From

CHO Cell Line Engineering and Design, Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark1

Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark2

CHO in Silico Engineering of Glycosylation and Protein Quality (CiSe), Research Groups, Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark3

CHO Core, Translational Management, Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark4

Pre-Pilot Plant, Translational Management, Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark5

iLoop, Translational Management, Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark6

Section for Synthetic Biology, Department of Biotechnology and Biomedicine, Technical University of Denmark7

Network Engineering of Eukaryotic Cell factories, Section for Synthetic Biology, Department of Biotechnology and Biomedicine, Technical University of Denmark8

Department of Biotechnology and Biomedicine, Technical University of Denmark9

Recombinant Chinese hamster ovary (CHO) cells are able to provide biopharmaceuticals that are essentially free of human viruses and have N-glycosylation profiles similar, but not identical, to humans. Due to differences in N-glycan moieties, two members of the serpin superfamily, alpha-1-antitrypsin (A1AT) and plasma protease C1 inhibitor (C1INH), are currently derived from human plasma for treating A1AT and C1INH deficiency.

Deriving therapeutic proteins from human plasma is generally a cost-intensive process and also harbors a risk of transmitting infectious particles. Recombinantly produced A1AT and C1INH (rhA1AT, rhC1INH) decorated with humanized N-glycans are therefore of clinical and commercial interest. Here, we present engineered CHO cell lines producing rhA1AT or rhC1INH with fully humanized N-glycosylation profiles.

This was achieved by combining CRISPR/Cas9-mediated disruption of 10 gene targets with overexpression of human ST6GAL1. We were able to show that the N-linked glyco-structures of rhA1AT and rhC1INH are homogeneous and similar to the structures obtained from plasma-derived A1AT and C1INH, marketed as Prolastin®-C and Cinryze®, respectively. rhA1AT and rhC1INH produced in our glyco-engineered cell line showed no detectable differences to their plasma-purified counterparts on SDS-PAGE and had similar enzymatic in vitro activity.

The work presented here shows the potential of expanding the glyco-engineering toolbox for CHO cells to produce a wider variety of glycoproteins with fully humanized N-glycan profiles. We envision replacing plasma-derived A1AT and C1INH with recombinant versions and thereby decreasing our dependence on human donor blood, a limited and possibly unsafe protein source for patients.

Language: English
Year: 2019
Pages: 143-152
ISSN: 10967176 and 10967184
Types: Journal article
DOI: 10.1016/j.ymben.2018.11.014
ORCIDs: Arnsdorf, Johnny , Amann, Thomas , Hansen, Anders Holmgaard , Kol, Stefan , Hansen, Henning Gram , Voldborg, Bjørn , Andersen, Mikael Rørdam and Kildegaard, Helene Faustrup

DTU users get better search results including licensed content and discounts on order fees.

Log in as DTU user

Access

Analysis