HEK293 Cell Line: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

The HEK293 cell line has earned its place as a producer of biotherapeutics. In addition to its ease of growth in serum-free suspension culture and its amenability to transfection, this cell line’s most important attribute is its human origin, which makes it suitable to produce biologics intended for human use. At the present time, the growth and production properties of the HEK293 cell line are inferior to those of non-human cell lines, such as the Chinese hamster ovary (CHO) and the murine myeloma NSO cell lines. However, the modification of genes involved in cellular processes, such as cell proliferation, apoptosis, metabolism, glycosylation, secretion, and protein folding, in addition to bioprocess, media, and vector optimization, have greatly improved the performance of this cell line.

  • HEK293
  • recombinant protein production
  • cell line engineering

1. Introduction

The human embryonic kidney cell line (HEK293) was created by transforming human embryonic kidney (HEK) cells with sheared fragments of adenovirus type 5 (Ad5) DNA, immortalizing it. Seventeen percent of the extreme left hand of the Ad5 genome is believed to have been integrated into chromosome 19 of the transformed HEK cells [1]. Since its creation, this cell line has been widely characterized and numerous derivative cell lines possessing unique characteristics have been developed. These cell lines include HEK293T [2], HEK293E [3], HEK293-6E [4], HEK293F and its derivatives HEK293FT and HEK293FTM [2][5][6][7], HEK293S [2], and HEK293H [8]. These derivatives and their properties are summarized in Table 1.
Table 1. HEK293 cell line derivatives.
HEK 293 Variant Derivation Commercially
Available?
Desired Characteristic(s) Year of Derivation
HEK293 Transformation of Human embryonic kidney cells with sheared fragments of adenovirus type 5 (Ad5) DNA, selected for immortalization. Yes Parental HEK293 cell line 1973
HEK293S Adapted for suspension growth Yes Grows in suspension in modified minimal Eagle’s medium 1984
HEK293T Stable transfection of the HEK 293 cell line with a plasmid encoding a temperature-sensitive mutant of the SV40 large T antigen Yes Amplification of vectors containing the SV40 ori, considerably increasing the protein expression levels during transient transfection. Before 1985
HEK293FT Fast-growing variant of HEK293T.
Expresses the SV40 large T antigen from the pCMVSPORT6TAg.neo plasmid.
  Designed for lentiviral production.  
HEK293F Cloned from the HEK293 cell line and adapted to commercial medium Yes Fast growth and high transfectivity. Growth in chemically defined medium 2014
HEK293H Cloned from HEK293 to select a clone with good adherence during plaque assays. Later adapted to growth in serum-free medium (SFM) (GIBCO 293H) Yes Fast growth in SFM, good adherence during plaque assays, superior transfection efficiencies and a high level of protein expression 1993
HEK293E(EBNA) Expresses the EBNA-1 protein for episomal replication of oriP harboring plasmids Yes Amplification of vectors containing oriP, considerably increasing the protein expression levels during transient transfection.  
HEK2936E This cell line is transfected with EBNA1t, a truncated version of EBV EBNA1 lacking the Gly-Gly-Ala repeats region   Has an enhanced ability to produce recombinant protein compared to HEK293-EBNA  
HEK293FTM Derived from 293 cells by stable transfection of an FRT-site containing plasmid and of a TetR expression plasmid.   Used for fast and easy generation of a stably transfected cell pool by co-transfecting a Flp-InTM expression vector containing a gene of interest and a Flp recombinase expression vector. Before 2001
HEK293SG Ricin toxin-resistant clone derived from HEK293S by ethylmethanesulfonate (EMS). Yes Lacks N-acetylglucosaminyltransferase I activity (encoded by the MGAT1 gene) and accordingly predominantly modifies glycoproteins with the Man5GlcNAc2 N-glycan. HEK293SG is used to produce homogenously N-glycosylated proteins 2001-2002
HEK293SGGD (Glycodelete) Derives from 293SG through expression of a Golgi targeted form of endoT, an endoglycosidase from the fungus Trichoderma reesei   HEK293SGGD is mainly used to produce proteins for glycosylation studies and structural analysis 2010
HEK293A Subclone of the HEK293 cells with a relatively flat morphology   Facilitates the initial production, amplification and titering of replication-incompetent adenovirus  
HEK293MSR Genetically engineered from HEK293 to express the human macrophage scavenger receptor   Strongly adheres to standard tissue culture plates for dependable results  
Data adapted from [9][10].
Until two decades ago, the HEK293 cell line was mainly used for transient, small to medium-scale production of research-grade proteins for scientific and pre-clinical studies. Its desirable characteristics have propelled this cell line into a potential producer of biopharmaceuticals [11][12]. These characteristics include suspension growth in serum-free media that enables large-scale production, reproducibility across different batches, rapid reproduction, amenability to different transfection methods, high efficiency of protein production, and—most importantly—its human origin. Five therapeutic proteins produced in HEK293 cells had been approved by the European Medicines Agency or the U.S. Food and Drug Administration for use in humans as of 2015 [13]. This cell line has also been useful in viral vector production as a result of adenoviral DNA in its genome. It has been employed in the production of the influenza virus [14][15], recombinant adenovirus [16], and lentiviruses [17][18]. This cell line is also the preferred host for membrane protein production, where it is most useful as a transient transfection tool for the evaluation of pharmacological properties of multiple receptor subtypes [12][19][20].
The HEK293 cell line is of epithelial origin and adherent in nature. It has been adapted to grow in suspension culture in serum-free or chemically defined media and is currently being utilized for recombinant proteins expression in both adherent and suspension platforms. Adherent platforms include roller bottles [21][22][23], multilayered culture systems (e.g., Nunc Cell Factories, Corning CellStacks and Greiner BIO-ONE CELLdisc™) [24][25][26][27][28], and fixed-bed bioreactors [27][29]. Suspension platforms include batch, fed batch and continuous processes in bioreactors.
Due to its human origin, HEK293 is better suited for production of biotherapeutics for human use since it produces protein with native post-translational modifications (PTMs). Non-human cell lines, such as the Chinese Hamster ovary cells (CHO), have been shown to implement incomplete humanized PTMs and can introduce potentially immunogenic non-human glycosylation patterns, most notably α-gal and NGNA [30][31]. PTMs, especially glycosylation patterns, affect the pharmacokinetics and pharmacodynamics of recombinant biotherapeutics. Although extensive comparative analyses of the PTMs on recombinant proteins produced in numerous cell lines have concluded that the suitability of a cell line for producing human therapeutic proteins should be determined on a case-by case basis [32][33], the HEK293 cell line has been shown to be especially efficient in tyrosine sulfation and glutamic acid γ-carboxylation when compared to other cell lines [34][35][36][37][38]. The impact of the host cell line on glycan profile has been comprehensively reviewed by Goh and Ng [39]. However, clinical experience with the HEK 293 cell line is not as extensive as for non-human other cell lines, although experience is growing, and there is potential susceptibility to human viral contamination due to the absence of species barrier.
Other human cell lines such as the HT-1080, produced from a fibrosarcoma with an epithelial-like phenotype [11] and the PER.C6 cell line created from human embryonic retinal cells, immortalized via transfection with the adenovirus E1 gene [40], have comparable growth, transfectability, production and safety profiles to the HEK293 cell line and produce complex human proteins with humanized PTMS [13]. However, the HEK293 cell line was used more extensively in different research topics and has numerous derivative cell lines which greatly expands the repertoire of recombinant proteins that it can efficiently produce.
Process and media parameters, such as expression vector, growth media, transfection agent and culture conditions, have been optimized to improved recombinant protein yields from HEK293. Notable innovative approaches include 3D collagen microsphere culture system [41]; the establishment of a glutamine-ammonia ligase (GLUL) mediated gene selection system [42]; glutamine synthase (GS) mediated selection/amplification system, and a dihydrofolate reductase (DHFR) amplification system [43][44], synonymous with the CHO system [45]. Growth and media optimization have led to the development of several formulations of chemically defined growth media and animal-derived component free media additives tailored to HEK293 cell growth and protein expression [46]. Some of the most widely utilized serum-free chemically defined media include ThermoFisher’s FreeStyle™ 293, CD 293, 293 SFM II and Expi293™ Expression Medium, Millipore Sigma’s EX-CELL® 293 Serum-Free Medium for HEK 293 Cells, and Cytiva’s HyClone SFM4Transfx-293 media.
Several transfection reagents have been optimized for use in transfection of HEK293 with plasmid DNA. For example, the Lipofectamine 3000 reagent by Thermo Fisher Scientific is the superior—but cost-prohibitive—option [47], while polyethylenimines (PEI) is the more cost-effective option [48]. Expression vector optimization identified the human cytomegalovirus promoter (CMV) as the most effective promoter for the expression of heterologous proteins in this cell line [49]. Culture parameters, including temperature and carbon dioxide concentration, [50] have also been optimized. Cytostatic agents such as sodium butyrate, trichostatin A, valproic acid, and dimethyl sulfoxide [51], have been used to enhance production. The implementation of these strategies to boost protein yields has resulted in product yields of up to 100–200mg/L in adherent systems, while suspension systems have yielded up to 140–600mg/L of recombinant protein [52][53].
Yet, despite these significant improvements in recombinant protein and virus production, this cell line continues to lag behind the CHO cell line in growth capacity (Maximal cell density of 3-5 × 106/mL for HEK293 vs. 1-2 × 107 for CHO) [42], cultivation time (HEK293 doubling time 33 h vs. 14-17 h for CHO), and product yield in stable production systems- up to 4g/L for CHO cells vs. 600mg/L for HEK 293 cells [54]. Consequently, some innate characteristics of HEK293 cells have been modified, resulting in higher recombinant protein yields, efficient nutrient utilization, and more homogenous glycosylation.

2. Affecting HEK293 Cell Growth and Production Performance by Modifying the Expression of Specific Genes

Although the HEK293 cell line trails non-human cell lines such as CHO and NSO, in recombinant protein production and culture performance [13], it is our opinion that this cell line has a place in the production of specific bioproducts. These cells, in addition to their ability to create native PTMs, efficiently execute tyrosine sulfation and gamma carboxylation which makes them the preferred host for recombinant clotting factors and natural anticoagulants production [13]. The cells also gained regulatory approval and have a place as producer of difficult-to-express antibody fragments or artificial scaffolds, which are forecasted to be the future of antibody therapy [55]. In addition, their use in production of gene therapy vectors will only increase as shown by the number of studies and resources directed towards understanding and improving gene therapy vector production in recent years [56]. The author’s opinion is that rational engineering strategies targeting bottlenecks in proliferation, carbon metabolism, protein processing and maturation with discovery of engineering targets using functional genomics tools have and will continue to enhance the production efficiency of the HEK293 cell line.
Among the targeted engineering strategies discussed in this review, glycosylation is the most critical. Obtaining homogenous glycosylation patterns is practically impossible without genetic manipulation or chemical treatment of the cell line [57]. Two HEK293 cell lines with improved glycosylation profiles are currently in use; the commercially available HEK293SG (HEK293SGnTI- (ATCC® CRL-3022), in which the N-acetyl-glucosaminyltransferase I (GnTI) gene has been knocked out and lacks complex N-glycans, and its derivative cell line HEK293SGGD (293SGlycoDelete (RRID:CVCL_6E38), which expresses a heterologous Golgi endoglycosidase for secretion of de-glycosylated glycoproteins. In addition, treatment of cells with small molecule inhibitors such as kifunensine, that targets α-mannosidase I and swainsonine, that blocks α-mannosidase II, results in relatively simple and chemically uniform glycans [58].
The majority of the currently approved therapeutics are being produced in derivative cell lines, underscoring the relevance of their unique properties; the recombinant FVIIIFc and recombinant FIXFc (ALPROLIX®, 2014; ELOCTATE®, respectively) in HEK293-H, the Human-cl rhFVIII (NUWIQ®) produced in HEK293F, the Glucagon-1-like peptide (GLP-1) Fc fusion protein (dulaglutide, TRULICITY®) produced in HEK293 EBNA, while the currently withdrawn drotrecogin alfa (XIGRIS®) was produced in the parental HEK293 cell line [13].
On the VLP and viral vector production front, parental HEK293 and derivatives such as HEK 293T and HEK293FT are the preferred hosts, the latter two due to the presence of the SV40 large T-antigen in their genomes that increases their viral production efficiency. Virus-like particle (VLP for vaccine generation) produced in HEK293 cell line include Rabies virus G protein VLP, Influenza virus A/Puerto Rico/8/34 (H1N1) VLP, Hepatitis B virus small surface antigen (HBaAgS) VLP, Human scavenger receptor class B, member 2 (SCARB2) VLP; Lentiviral vectors for gene therapy: Kymriah™ (Lymphoblastic leukemia) and ZyntegloT™ (Beta-thalassemia); AAV gene therapy vectors: Glybera™ (Lipoprotein lipase deficiency), Luxturna™ (Leber congenital amaurosis), and Zolgensma™ (Spinal muscular atrophy type 1); retroviral gene therapy vectors: Rexin-G™ (Pancreatic cancer), Strimvelis™ (Adenosine deaminase—immunodeficiency), Zalmoxis™ (Leukemia) [59].The choice of HEK cell line for producing viral vectors is usually more straightforward compared to the production of recombinant proteins. For most recombinant proteins produced in the HEK293 cell line and its derivatives, the choice of cell line is assessed on a case-by-case basis, and it is not uncommon for researchers to test different cell lines for productivity before deciding. Notwithstanding, the HEK293H and HEK293F cell lines with their superior transfectivity, higher productivity, and faster growth to high densities in chemically defined media are currently the choice for large scale recombinant protein production.
The HEK 293 cell line, along with a few other human cell lines, currently serves as a valuable niche for the production of biotherapeutics that require human PTMs. It is also the cell line of choice for the production of recombinant gene therapy vectors. Whether or not this cell line will become as widely used as the CHO cell line in biotherapeutic production will depend on the continued technological achievements and research investments in further optimizing this cell line.

This entry is adapted from the peer-reviewed paper 10.3390/cells10071667

References

  1. Graham, F.L.; Russell, W.C.; Smiley, J.; Nairn, R. Characteristics of a Human Cell Line Transformed by DNA from Human Adenovirus Type. J. Gen. Virol. 1977, 36, 59–72.
  2. Lin, Y.-C.; Boone, M.; Meuris, L.; Lemmens, I.; Van Roy, N.; Soete, A.; Reumers, J.; Moisse, M.; Plaisance, S.; Drmanac, R.T.; et al. Genome dynamics of the human embryonic kidney 293 lineage in response to cell biology manipulations. Nat. Commun. 2014, 5, 4767.
  3. Kim, K.-S.; Kim, M.S.; Moon, J.H.; Jeong, M.S.; Kim, J.; Lee, G.M.; Myung, P.-K.; Hong, H.J. Enhancement of recombinant antibody production in HEK 293E cells by WPRE. Biotechnol. Bioprocess. Eng. 2009, 14, 633–638.
  4. Jäger, V.; Büssow, K.; Wagner, A.; Weber, S.; Hust, M.; Frenzel, A.; Schirrmann, T. High level transient production of recombinant antibodies and antibody fusion proteins in HEK293 cells. BMC Biotechnol. 2013, 13, 52.
  5. Geisse, S.; Fux, C. Recombinant Protein Production by Transient Gene Transfer into Mammalian Cells. In Methods in Enzymology; Burgess, R.R., Deutscher, M.P., Eds.; Academic Press: Cambridge, MA, USA, 2009; pp. 223–238.
  6. Casademunt, E.; Martinelle, K.; Jernberg, M.; Winge, S.; Tiemeyer, M.; Biesert, L.; Knaub, S.; Walter, O.; Schröder, C. The first recombinant human coagulation factor VIII of human origin: Human cell line and manufacturing characteristics. Eur. J. Haematol. 2012, 89, 165–176.
  7. Stepanenko, A.; Dmitrenko, V. HEK293 in cell biology and cancer research: Phenotype, karyotype, tumorigenicity, and stress-induced genome-phenotype evolution. Gene 2015, 569, 182–190.
  8. Bloom, F.R.; Price, P.; Lao, G.; Xia, J.L.; Crowe, J.H.; Battista, J.R.; Helm, R.F.; Slaughter, S.; Potts, M. Engineering mammalian cells for solid-state sensor applications. Biosens. Bioelectron. 2001, 16, 603–608.
  9. Yuan, J.; Xu, W.W.; Jiang, S.; Yu, H.; Poon, H.F. The Scattered Twelve Tribes of HEK293. Biomed. Pharmacol. J. 2018, 11, 621–623.
  10. Malm, M.; Saghaleyni, R.; Lundqvist, M.; Giudici, M.; Chotteau, V.; Field, R.; Varley, P.G.; Hatton, D.; Grassi, L.; Svensson, T.; et al. Evolution from adherent to suspension: Systems biology of HEK293 cell line development. Sci. Rep. 2020, 10, 1–15.
  11. Swiech, K.; Picanco-Castro, V.; Covas, D. Human cells: New platform for recombinant therapeutic protein production. Protein Expr. Purif. 2012, 84, 147–153.
  12. Thomas, P.; Smart, T.G. HEK293 cell line: A vehicle for the expression of recombinant proteins. J. Pharmacol. Toxicol. Methods 2005, 51, 187–200.
  13. Dumont, A.J.; Euwart, D.; Mei, B.; Estes, S.; Kshirsagar, R.R. Human cell lines for biopharmaceutical manufacturing: History, status, and future perspectives. Crit. Rev. Biotechnol. 2016, 36, 1110–1122.
  14. Thompson, C.M.; Petiot, E.; Mullick, A.; Aucoin, M.G.; Henry, O.; Kamen, A.A. Critical assessment of influenza VLP production in Sf9 and HEK293 expression systems. BMC Biotechnol. 2015, 15, 1–12.
  15. Le Ru, A.; Jacob, D.; Transfiguracion, J.; Ansorge, S.; Henry, O.; Kamen, A.A. Scalable production of influenza virus in HEK-293 cells for efficient vaccine manufacturing. Vaccine 2010, 28, 3661–3671.
  16. Grieger, J.C.; Soltys, S.M.; Samulski, R.J. Production of Recombinant Adeno-associated Virus Vectors Using Suspension HEK293 Cells and Continuous Harvest of Vector from the Culture Media for GMP FIX and FLT1 Clinical Vector. Mol. Ther. 2016, 24, 287–297.
  17. Chahal, P.S.; Schulze, E.; Tran, R.; Montes, J.; Kamen, A.A. Production of adeno-associated virus (AAV) serotypes by transient transfection of HEK293 cell suspension cultures for gene delivery. J. Virol. Methods 2014, 196, 163–173.
  18. Ansorge, S.; Lanthier, S.; Transfiguracion, J.; Durocher, Y.; Henry, O.; Kamen, A. Development of a scalable process for high-yield lentiviral vector production by transient transfection of HEK293 suspension cultures. J. Gene Med. 2009, 11, 868–876.
  19. Chaudhary, S.; Pak, J.E.; Pedersen, B.; Bang, L.J.; Zhang, L.B.; Ngaw, S.M.; Green, R.G.; Sharma, V.; Stroud, R.M. Efficient expression screening of human membrane proteins in transiently transfected Human Embryonic Kidney 293S cells. Methods 2011, 55, 273–280.
  20. Andréll, J.; Tate, C. Overexpression of membrane proteins in mammalian cells for structural studies. Mol. Membr. Biol. 2012, 30, 52–63.
  21. Ayuso, E.; Mingozzi, F.; Montane, J.; Leon, X.; Anguela, X.; Haurigot, V.A.; Edmonson, A.S.; Africa, L.; Zhou, S.; High, K.; et al. High AAV vector purity results in serotype- and tissue-independent enhancement of transduction efficiency. Gene Ther. 2009, 17, 503–510.
  22. Liu, Y.-L.; Wagner, K.; Robinson, N.; Sabatino, D.; Margaritis, P.; Xiao, W.; Herzog, R.W. Optimized Production of High-Titer Recombinant Adeno-Associated Virus in Roller Bottles. BioTechniques 2003, 34, 184–189.
  23. Dalton, A.C.; Barton, W.A. Over-expression of secreted proteins from mammalian cell lines. Protein Sci. 2014, 23, 517–525.
  24. Zolotukhin, S.; Potter, M.; Zolotukhin, I.; Sakai, Y.; Loiler, S.; Fraites, T.J.; Chiodo, A.V.; Phillipsberg, T.; Muzyczka, N.; Hauswirth, W.W.; et al. Production and purification of serotype 1, 2, and 5 recombinant adeno-associated viral vectors. Methods 2002, 28, 158–167.
  25. Potter, M.; Lins, B.; Mietzsch, M.; Heilbronn, R.; Van Vliet, K.; Chipman, P.; Agbandje-McKenna, M.; Cleaver, B.D.; Clément, N.; Byrne, B.J.; et al. A simplified purification protocol for recombinant adeno-associated virus vectors. Mol. Ther. Methods Clin. Dev. 2014, 1, 14034.
  26. Piras, A.B.; Drury, E.J.; Morton, C.L.; Spence, Y.; Lockey, T.D.; Nathwani, A.C.; Davidoff, A.M.; Meagher, M.M. Distribution of AAV8 particles in cell lysates and culture media changes with time and is dependent on the recombinant vector. Mol. Ther. Methods Clin. Dev. 2016, 3, 16015.
  27. Emmerling, V.V.; Pegel, A.; Milian, E.G.; Venereo-Sanchez, A.; Kunz, M.; Wegele, J.; Kamen, A.A.; Kochanek, S.; Hoerer, M. Rational plasmid design and bioprocess optimization to enhance recombinant adeno-associated virus (AAV) productivity in mammalian cells. Biotechnol. J. 2016, 11, 290–297.
  28. Strobel, B.; Zuckschwerdt, K.; Zimmermann, G.; Mayer, C.; Eytner, R.; Rechtsteiner, P.; Kreuz, S.; Lamla, T. Standardized, Scalable, and Timely Flexible Adeno-Associated Virus Vector Production Using Frozen High-Density HEK-293 Cell Stocks and CELLdiscs. Hum. Gene Ther. Methods 2019, 30, 23–33.
  29. Powers, A.; Piras, B.; Clark, R.K.; Lockey, T.D.; Meagher, M.M. Development and Optimization of AAV hFIX Particles by Transient Transfection in an iCELLis®Fixed-Bed Bioreactor. Hum. Gene Ther. Methods 2016, 27, 112–121.
  30. Ghaderi, D.; Taylor, E.R.; Padler-Karavani, V.; Diaz, S.; Varki, A. Implications of the presence of N-glycolylneuraminic acid in recombinant therapeutic glycoproteins. Nat. Biotechnol. 2010, 28, 863–867.
  31. Fliedl, L.; Grillari, J.; Grillari-Voglauer, R. Human cell lines for the production of recombinant proteins: On the horizon. N. Biotechnol. 2015, 32, 673–679.
  32. Böhm, E.; Seyfried, B.K.; Dockal, M.; Graninger, M.; Hasslacher, M.; Neurath, M.; Konetschny, C.; Matthiessen, P.F.; Mitterer, A.; Scheiflinger, F. Differences in N-glycosylation of recombinant human coagulation factor VII derived from BHK, CHO, and HEK293 cells. BMC Biotechnol. 2015, 15, 1–15.
  33. Kronman, C.; Chitlaru, T.; Elhanany, E.; Velan, B.; Shafferman, A. hierarchy of post-translational modifications involved in the circulatory longevity of glycoproteins. Demonstration of concerted contributions of glycan sialylation and subunit assembly to the pharmacokinetic behavior of bovine acetylcholinesterase. J. Biol. Chem. 2000, 275, 29488–29502.
  34. Berkner, K. Expression of recombinant vitamin K-dependent proteins in mammalian cells: Factors IX and VII. In Methods in Enzymology; Academic Press: Cambridge, MA, USA, 1993; Volume 222, pp. 450–477.
  35. Kannicht, C.; Ramström, M.; Kohla, G.; Tiemeyer, M.; Casademunt, E.; Walter, O.; Sandberg, H. Characterisation of the post-translational modifications of a novel, human cell line-derived recombinant human factor VIII. Thromb. Res. 2013, 131, 78–88.
  36. Peters, R.T.; Toby, G.; Lu, Q.; Liu, T.; Kulman, J.D.; Low, S.C.; Bitonti, A.J.; Pierce, G.F. Biochemical and functional characterization of a recombinant monomeric factor VIII–Fc fusion protein. J. Thromb. Haemost. 2013, 11, 132–141.
  37. McCue, J.; Osborne, D.; Dumont, J.; Peters, R.; Mei, B.; Pierce, G.F.; Kobayashi, K.; Euwart, D. Validation of the manufacturing process used to produce long-acting recombinant factor IX Fc fusion protein. Haemophilia 2014, 20, e327–e335.
  38. McCue, J.; Kshirsagar, R.; Selvitelli, K.; Lu, Q.; Zhang, M.; Mei, B.; Peters, R.; Pierce, G.F.; Dumont, J.; Raso, S.; et al. Manufacturing process used to produce long-acting recombinant factor VIII Fc fusion protein. Biologicals 2015, 43, 213–219.
  39. Goh, J.B.; Ng, S.K. Impact of host cell line choice on glycan profile. Crit. Rev. Biotechnol. 2018, 38, 851–867.
  40. Havenga, M.; Holterman, L.; Melis, I.; Smits, S.; Kaspers, J.; Heemskerk, E.; van der Vlugt, R.; Koldijk, M.; Schouten, G.; Hateboer, G.; et al. Serum-free transient protein production system based on adenoviral vector and PER.C6 technology: High yield and preserved bioactivity. Biotechnol. Bioeng. 2008, 100, 273–283.
  41. Wong, H.-L.; Wang, M.-X.; Cheung, P.-T.; Yao, K.-M.; Chan, B.P. A 3D collagen microsphere culture system for GDNF-secreting HEK293 cells with enhanced protein productivity. Biomaterials 2007, 28, 5369–5380.
  42. Chin, C.L.; Goh, J.B.; Srinivasan, H.; Liu, K.I.; Gowher, A.; Shanmugam, R.; Lim, H.L.; Choo, M.; Tang, W.Q.; Tan, A.H.-M.; et al. A human expression system based on HEK293 for the stable production of recombinant erythropoietin. Sci. Rep. 2019, 9, 1–16.
  43. Mensah, E.O.; Guo, X.-Y.; Gao, X.-D.; Fujita, M. Establishment of DHFR-deficient HEK293 cells for high yield of therapeutic glycoproteins. J. Biosci. Bioeng. 2019, 128, 487–494.
  44. Lee, S.Y.; Baek, M.; Lee, G.M. Comprehensive characterization of dihydrofolate reductase-mediated gene amplification for the establishment of recombinant human embryonic kidney 293 cells producing monoclonal antibodies. Biotechnol. J. 2021, 16, e2000351.
  45. Yu, D.Y.; Lee, S.Y.; Lee, G.M. Glutamine synthetase gene knockout-human embryonic kidney 293E cells for stable production of monoclonal antibodies. Biotechnol. Bioeng. 2018, 115, 1367–1372.
  46. Liste-Calleja, L.; Lecina, M.; Cairó, J.J. HEK293 cell culture media study: Increasing cell density for different bioprocess applications. BMC Proc. 2013, 7, P51.
  47. Ooi, A.; Wong, A.; Esau, L.; Lemtiri-Chlieh, F.; Gehring, C. A Guide to Transient Expression of Membrane Proteins in HEK-293 Cells for Functional Characterization. Front. Physiol. 2016, 7, 300.
  48. Reed, S.E.; Staley, E.M.; Mayginnes, J.P.; Pintel, D.J.; Tullis, G.E. Transfection of mammalian cells using linear polyethylenimine is a simple and effective means of producing recombinant adeno-associated virus vectors. J. Virol. Methods 2006, 138, 85–98.
  49. Román, R.; Miret, J.; Scalia, F.; Casablancas, A.; Lecina, M.; Cairó, J.J. Enhancing heterologous protein expression and secretion in HEK293 cells by means of combination of CMV promoter and IFNα2 signal peptide. J. Biotechnol. 2016, 239, 57–60.
  50. Lin, C.-Y.; Huang, Z.; Wen, W.; Wu, A.; Wang, C.; Niu, L. Enhancing Protein Expression in HEK-293 Cells by Lowering Culture Temperature. PLoS ONE 2015, 10, e0123562.
  51. Cervera, L.; Fuenmayor, J.; González-Domínguez, I.; Gutiérrez-Granados, S.; Segura, M.M.; Gòdia, F. Selection and optimization of transfection enhancer additives for increased virus-like particle production in HEK293 suspension cell cultures. Appl. Microbiol. Biotechnol. 2015, 99, 9935–9949.
  52. Loignon, M.; Perret, S.; Kelly, J.; Boulais, D.; Cass, B.; Bisson, L.; Afkhamizarreh, F.; Durocher, Y. Stable high volumetric production of glycosylated human recombinant IFNalpha2b in HEK293 cells. BMC Biotechnol. 2008, 8, 65.
  53. Spidel, J.L.; Vaessen, B.; Chan, Y.Y.; Grasso, L.; Kline, J.B. Rapid high-throughput cloning and stable expression of antibodies in HEK293 cells. J. Immunol. Methods 2016, 439, 50–58.
  54. Frenzel, A.; Hust, M.; Schirrmann, T. Expression of Recombinant Antibodies. Front. Immunol. 2013, 4, 217.
  55. Vazquez-Lombardi, R.; Nevoltris, D.; Luthra, A.; Schofield, P.; Zimmermann, C.; Christ, D. Transient expression of human antibodies in mammalian cells. Nat. Protoc. 2018, 13, 99–117.
  56. Bulcha, J.T.; Wang, Y.; Ma, H.; Tai, P.W.L.; Gao, G. Viral vector platforms within the gene therapy landscape. Signal. Transduct. Target. Ther. 2021, 6, 1–24.
  57. Lepenies, B.; Seeberger, P.H. Simply better glycoproteins: Reducing the glycan heterogeneity of recombinant proteins may improve the efficacy of biopharmaceuticals. Nat. Biotechnol. 2014, 32, 443–445.
  58. Aricescu, A.R.; Owens, R.J. Expression of recombinant glycoproteins in mammalian cells: Towards an integrative approach to structural biology. Curr. Opin. Struct. Biol. 2013, 23, 345–356.
  59. Pulix, M.; Lukashchuk, V.; Smith, D.C.; Dickson, A.J. Molecular characterization of HEK293 cells as emerging versatile cell factories. Curr. Opin. Biotechnol. 2021, 71, 18–24.
More
This entry is offline, you can click here to edit this entry!