fischer2019
fischer2019
Monoclonal antibodies (mAbs) are still the most frequently developed class of
biologics [10]. However, the number of multispecific formats as well as highly
potent fusion proteins (in the following sections, referred to as “multispecifics”)
have also increased dramatically over the past few years [11, 12]. Certainly, these
molecules have never undergone an evolutionary process and thus represent new
territories for production cells in terms of translation, intracellular processing,
folding, and secretion. Thus, it seems little surprising that these novel and com-
plex therapeutics often turn out to be difficult-to-express for CHO cells [13–15].
Although the state-of-the-art industrial cell line development (CLD) workflows
frequently deliver clonal cell lines exceeding product yields of 5 g/l, also stan-
dard IgGs were found to be challenging to be expressed by CHO cells [16, 17],
e.g. if the molecule design is suboptimal. For instance, if the primary amino acid
sequence of an IgG exhibits aggregation-prone regions on the surface or very
unusual amino acid residues at certain positions, compared to a wide range of
known IgG sequences, it can also have a dramatic influence productivity [18–20].
Consequently, there is an urgent demand to steadily improve industrial CHO host
cell lines in order to be prepared for future bioprocessing challenges. In addi-
tion, health care systems are already facing tremendous costs associated with
the increasing demand for therapeutic proteins to address unmet medical needs
[21]. Hence, industrial manufacturing processes of biopharmaceuticals are highly
dependent on nonexpensive and high-yielding production platforms in order to
maximize production yields but also to reduce associated costs. Therefore, host
cell engineering of CHO cells represents a valuable strategy to overcome limita-
tions in the production of biologics.
There are different opportunities to counteract limitations of mammalian cell
factories. In the following chapters, we have summarized the most relevant
cell line and state-of-the-art engineering techniques currently applied for CHO
cell engineering including overexpression or knockout of genes, as well as the
usage of noncoding RNAs. Furthermore, we provide an overview on applications
of cell line engineering approaches in CHO cells to enhance recombinant
protein production, repress cell death and accelerate growth, and modulate
posttranslational modifications (PTMs).
cells are then subjected to antibiotic selection pressure to generate cell pools
with the plasmid DNA stably integrated into their genome. The expression of
the gene of interest (GOI) is often driven by strong viral or cellular promoter
and enhancer sequences to ensure high expression levels [23], while the selective
gene is normally controlled by weak promoters to increase the overall expression
level [24]. After the selection process, the resulting cell culture represents
a heterogeneously mixed pool of cells showing various extent of transgene
overexpressions. This procedure, however, results in phenotypic differences
between individual cells, and thus, cell lines derived from a single progenitor
cell subsequently have to be established (process is called “single cell cloning”)
to obtain a homogenous host cell line exhibiting a strong and stable engineered
phenotype.
identical seed sequences are grouped into families [43]; however, miRNAs from
the same seed family can have surprisingly different roles in vivo. The function of
miRNAs highly depends on the composition of the cellular transcriptome, which
impedes a clear functional classification for individual miRNAs [47, 57].
The lack of genomic sequence information before 2011 substantially hindered
miRNA research in CHO cells [58], and different strategies for functional miRNA
analysis had been pursued such as transient transfections of either miRNA mim-
ics or artificial expression vectors encoding chimeric miRNA precursor hairpins
[59, 60]. However, chimeric miRNA expression vectors were shown to be infe-
rior to vectors coding for the endogenous Cricetulus griseus miRNA sequence
[47, 61]. Taking advantage of next-generation RNA sequencing technology, 307
mature miRNAs and 200 pre-miR sequences were initially found in total RNA
samples of different CHO cell lines cultivated under various culture conditions
[62, 63]. These miRNA sequences were subsequently annotated as cgr-miRNAs
in miRBase [64, 65]. Recently, thorough in silico re-analysis of the CHO genome
revealed the presence of an additional 71 mature miRNAs in CHO cells [66].
Cellular Origin of
pathway engineering gene Target gene Engineered phenotype References
Protein Not indicated Protein disulfide isomerase (PDI) Increased productivity of monoclonal Borth et al. [67]
synthesis (overexpression) antibodies
CHO (overexpression) ERp57 (an isoform of PDI) 2.1-Fold increase in specific thrombopoietin Hwang et al. [68]
(TPO) productivity without decreasing cell
growth
CHO (overexpression) Calnexin (CNX) and calreticulin (CRT) 1.9-Fold increase in specific TPO productivity Chung et al. [69]
without negatively influencing cell growth and
biological activity of the recombinant TPO
Bovine Nonphosphorylatable version of the Enhanced transient expression of recombinant Underhill et al. [70]
(overexpression) eukaryotic translation initiation factor proteins
2 α (eIF2α)
Artificial Artificial zinc finger protein 10-Fold increase in IgG titer Kwon et al. [71]
(overexpression) transcription factor (ZFP-TF)
CHO (overexpression) Activating transcription factor 4 (ATF4) Increase in human antithrombin III (AT-III) Ohya et al. [72]
titer
CHO (overexpression) Growth arrest and DNA 40% increase in human AT-III titer Omasa et al. [73]
damage-inducible protein 34 (GADD34)
Human Mammalian target of rapamycin Increased cell growth, viability, apoptosis Dreesen and
(overexpression) (mTOR) resistance, and specific productivity of Fussenegger [74]
glycoproteins
Human Heat-shock 70 kDa protein 5 (BIP), Increased expression of difficult-to-express Pybus et al. [17]
(overexpression) activating transcription factor 6C monoclonal antibodies and reduced cell growth
(ATF6C), and X-box binding protein 1
(XBP1)
CHO (overexpression) Ying Yang 1 (YY1) Increased production of several product genes Tastanova et al. [75]
(SEAP, VEGF165, and IgG including rituximab)
CHO (overexpression) Glutamate-cysteine ligase modifier Increased specific productivity, mAb titer, and Orellana et al. [76]
subunit (GCLM) frequency of high producer clones by 70%
CHO (knockout) Dihydrofolate reductase (DHFR) Transgene amplification for increased protein Urlaub et al. [5]
production Page and Sydenham
[77]
CHO (knockout) Glutamine synthetase (GS) Transgene amplification for increased protein Sanders et al. [8]
production Cockett et al. [7]
CHO (knockout) DHFR Rapid establishment of DHFR−/− cells within Santiago et al. [29]
one month
CHO (knockout) GS Increased selection efficiency of Fan et al. [9]
high-producing CHO cells
CHO (knockdown) DHFR >100% increase in specific IgG productivity Wu et al. [78]
and 30% improved stability of transgene
expression
CHO (knockout) Insulin-like growth factor 1 receptor Increased production of a difficult-to-express Romand et al. [79]
(IGF1R) protein (IGF1)
CHO (knockout) FAM60A Increased expression stability of mAb Ritter et al. [80]
production clones
CHO (knockout) C12orf35 Several fold increased mAb productivity of Ritter et al. [81]
stable pools and clones
CHO (knockout) Activating transcription factor 6 β Improved mAb productivity Pieper et al. [82]
(ATF6β)
CHO Ceramide synthase 2 (CerS2) and Rab1 Improved mAb productivity Pieper et al. [83]
(knockout) GAP Tbc domain family member 20
(Tbc1D20)
CHO Breast cancer 1 (BRCA1) Increased mAb productivity of up to 5.3-fold Matsuyama et al.
(knockdown) [84]
Table 9.1 (Continued)
Cellular Origin of
pathway engineering gene Target gene Engineered phenotype References
Metabolism Vitreoscilla Vitreoscilla hemoglobin (VHb) 40–100% increase in specific human tPA Pendse and Bailey
(overexpression) productivity [85]
Rat (overexpression) Carbamoyl phosphate synthetase I (CPS 25–33% Decreased accumulation of Park et al. [86]
I) and ornithine transcarbamoylase ammonium and 15–30% increased cell growth
(OTC)
Yeast (overexpression) Pyruvate carboxylase 2 (PYC2) 35% Decrease in lactate production and Fogolin et al. [87]
twofold increase in product titer
Human Pyruvate carboxylase (PC) Increased viability because of 21–39% Kim and Lee [88]
(overexpression) decreased lactate production
Mouse Glucose transporter protein 5 (GLUT5) Less lactate production and higher cell Wlaschin and Hu
(overexpression) densities in fructose fed-batch processes [89]
Mouse GLUT5 Less lactate production, increased growth rate, Le et al. [90]
(overexpression) prolonged culture duration, and higher product
titer
CHO (overexpression) Malate dehydrogenase II (MDH2) Increased intracellular levels of ATP and Chong et al. [91]
NADH led to an 1.9-fold improvement in
integral viable cell number
CHO (overexpression) Taurine transporter (TAUT) Improved viability and increased IgG titer Tabuchi et al. [92]
CHO (overexpression) TAUT and alanine aminotransferase 1 Higher IgG yield in shorter cultivation time Tabuchi and
(ALT1) Sugiyama [93]
CHO (knockout) Lactate dehydrogenase A (LDHA) 45–79% Reduced lactate concentrations and Kim and Lee [94]
diminished glucose consumption
CHO (knockout) LDHA Diminished medium acidification because of Jeong et al. [95]
decreased lactate production leading to less
apoptosis
CHO (knockout) Enolase 1 (ENO1) Increase in viable cell density Doolan et al. [96]
CHO (knockout) LDHA and pyruvate dehydrogenase 68–90% Increase in IgG titer Zhou et al. [97]
kinase (PDHK)
CHO (knockdown) Knockdown of LDHA combined with Improved culture longevity because of Jeong et al. [98]
BCL2 overexpression decreased lactate production and increased
apoptosis resistance
Secretion Human X-box binding protein 1 (XBP1) Higher endoplasmic reticulum content and Tigges and
(overexpression) increase in product titer Fussenegger [99]
Becker et al. [100]
Human Spliced form of XBP-1 (XBP1s) Fourfold increase in specific IgG productivity Ku et al. [101]
(overexpression) Gulis et al. [102]
Human Suppressor of loss of YPT1 protein 1 15-Fold increase in IgG production Peng and
(overexpression) (SLY1) and syntaxin binding protein 3 Fussenegger [103]
(MUNC18C)
Human Tricystronic expression of SLY1, 20-Fold increase in IgG production Peng and
(overexpression) MUNC18C, and XBP1 Fussenegger [103]
Human Ceramide transfer protein (CERT) Increase in specific productivity of human Florin et al. [104]
(overexpression) serum albumin (HSA) and monoclonal
antibodies
Human Mutant form of CERT (S132A) 35% Increase in specific t-PA productivity Rahimpour et al.
(overexpression) [105]
Human Synaptosome-associated protein of Increase in SEAP productivity by enhanced Peng et al. [106]
(overexpression) 23 kDa (SNAP-23) and secretory capacity
vesicle-associated membrane protein 8
(VAMP8)
Human Human signaling receptor protein 14 Improved secretion and production of Le Fourn et al. [107]
(overexpression) (SRP14) difficult-to-express proteins
Table 9.1 (Continued)
Cellular Origin of
pathway engineering gene Target gene Engineered phenotype References
Cell cycle Human Cyclin-dependent kinase inhibitor 1A Growth arrest and 10- to 15-fold increase in Fussenegger et al.
(overexpression) (p21CIP1 ) and CCAAT/enhancer-binding specific SEAP productivity [108]
protein 𝛼 (C/EBP-𝛼)
Human Tricystronic expression of p21CIP1 , Growth arrest and 30-fold increase in specific Fussenegger et al.
(overexpression) C/EBP-𝛼, and BCL-xL SEAP productivity [108]
Astley et al. [109]
Human Cyclin-dependent kinase inhibitor 1B Increased specific SEAP productivity Mazur et al. [110]
(overexpression) (p27KIP1 )
Human p21CIP1 Fourfold increase in IgG production Bi et al. [111]
(overexpression)
Human Myelocytomatosis oncogene (C-MYC) >70% Increase in maximal cell density without Kuystermans and
(overexpression) additional supply of nutrients Al-Rubeai [112]
CHO (knockout) Ataxia telangiectasia and Rad3 related Fourfold increase in specific IgG productivity Lee et al. [113]
(ATR) and threefold improved IgG titer
microRNA CHO (inhibition) miR-7a-5p (inhibition) Reduced growth and enhanced SEAP Barron et al. [102]
productivity Meleady et al. [56]
Sanchez et al. [114]
CHO (overexpression) miR-30a, c, d, e Enhanced mAb and SEAP productivity Fischer et al. [47]
CHO (overexpression) miR-2861 Enhanced mAb and SEAP productivity Fischer et al. [115]
CHO (overexpression) miR-17-5p Enhanced growth and EPO-Fc productivity Jadhav et al. [59]
Clarke et al. [116]
Jadhav et al. [117]
Loh et al. [118]
CHO (overexpression) miR-19b Enhanced mAb productivity Loh et al. [118]
Clarke et al. [116]
CHO (overexpression) miR-20a Enhanced mAb productivity Loh et al. [118]
Clarke et al. [116]
CHO (overexpression) miR-17-92a Enhanced mAb and EPO-Fc productivity Jadhav et al. [117]
Loh et al. [118]
CHO (overexpression) miR-92a Enhanced mAb productivity Loh et al. [119]
Human miR-1287 Enhanced mAb productivity Strotbek et al. [60]
(overexpression)
Human mitosRNA-1978 Enhanced mAb productivity Strotbek et al. [60]
(overexpression)
CHO (inhibition) miR-34a Enhanced SEAP productivity Kelly et al. [120]
CHO (overexpression) miR-483-3p Enhanced mAb and rAAV productivity Emmerling et al.
[121]
CHO (inhibition) miR-23 Enhanced SEAP productivity Kelly et al. [122]
CHO (overexpression) miR-143-3p Enhanced productivity of difficult-to-express Schoellhorn et al.
proteins [123]
Human miR-557 Twofold increase in difficult-to-express mAb in Strotbek et al. [60]
(overexpression) fed-batch cultivation Fischer et al. [124]
218 9 CHO Cell Engineering for Improved Process Performance and Product Quality
Cellular Origin of
pathway engineering gene Target gene Engineered phenotype References
Apoptosis Human (overexpression) B-cell lymphoma 2 (BCL2) 75% Increase in maximum viable cell density Tey et al. [140]
Human (overexpression) BCL2 and BCL2-like 1 (BCL-xL) Improved viability and enhanced apoptosis Mastrangelo et al.
resistance [141]
Meents et al. [142]
Human (overexpression) X-linked inhibitor of apoptosis Improved apoptosis resistance Sauerwald et al.
(XIAP) [143]
Human (overexpression) Apoptosis, caspase inhibitor (AVEN) Improved apoptosis resistance Figueroa et al. [144]
and BCL-xL
Human (overexpression) BCL-xL 88% Increase in viability and enhances IgG Chiang and Sisk
titer by >2-fold [145]
Human (overexpression) Myelocytomatosis oncogene (c-myc) Improved apoptosis resistance and increased Infandi and
and BCL2 viable cell density Al-Rubeai (2005)
CHO (overexpression) Fas apoptotic inhibitory molecule Improved apoptosis resistance leading to 80% Wong et al. [146]
(FAIM) increased VCD and 2.5-fold enhanced
interferon 𝛾 (IFN𝛾) titer
Bombyx mori silkworm Apoptosis-inhibiting 30K protein Increased viable cell density and enhanced Choi et al. [147]
hemolymph (30Kc6) erythropoietin (EPO) titer of up to 10-fold
(overexpression)
Human (overexpression) Telomerase reverse transcriptase Increased apoptosis resistance resulting in Crea et al. [148]
(TERT) higher viable cell density
Human and adenoviral AVEN and control protein E1B 19K Increased viable cell density and viability; 50% Figueroa et al. [149]
(overexpression) (E1B-19K) enhanced IgG titer
Mouse (overexpression) Murine double-mutant 2 (MDM2) Increased apoptosis resistance Arden et al. [150]
Human (overexpression) E2F transcription factor 1 (E2F1) 20% increased viable cell density and Majors et al. [151]
improved proliferation
Human and adenoviral AVEN, E1B-19K and a mutant form 60% Increase in viable cell density and 80% Dorai et al. [152]
(overexpression) of XIAP (EAX197) enhanced IgG titer
CHO (overexpression) Heat-shock proteins 27 and 70 Extended culture longevity and 2.5-fold Lee et al. [153]
(HSP27 and HSP70) increase in IFN𝛾 titer
Human (overexpression) Myeliod cell leukemia 1 (MCL1) Improved viability and 20–35% increased IgG Majors et al. [154]
titer
Human (overexpression) RAC-𝛼 serine/threonine-protein Delayed onset of apoptosis and autophagy Hwang and Lee
kinase (AKT1) during batch culture [155]
Human (overexpression) Mutated form of BCL-xL (Asp29Asn Improved apoptosis resistance Majors et al. [156]
variant)
CHO (overexpression) Chinese hamster heat-shock protein 2.2-Fold higher peak VCD, sustained viability, Tan et al. [157]
27 (HSP27) and 2.3-fold increase in mAb titer
CHO (knockout) BCL2-associated X protein (BAX) Increased apoptosis resistance because of Cost et al. [158]
and BCL2-antagonist/killer (BAK) inhibition of caspase activation leading up to
fivefold increase in IgG titer
CHO (knockout) BAX, BAK, and FUT8 Prolonged culture longevity because of Grav et al. [159]
diminished apoptosis; engineered clones
produced afucosylated IgGs
CHO (knockdown) Caspase 3 Extended culture longevity by more than two Kim and Lee (2002)
days in batch cultures
CHO (knockdown) Caspase 3 and 7 Enhanced cell viability and 55% increase in Sung et al. [160]
hTPO titer
CHO (knockdown) Caspase 8 and 9 Enhanced viability in batch and fed-batch Yun et al. (2007)
cultivations
CHO (knockdown) Alpha-1,3/1,6-mannosyltransferase Elevated cell density and culture longevity; Wong et al. [146]
(ALG2), Requiem (REQ), Fas 1.2- to 2.5-fold increase in IFN𝛾 titer
(TNFRSF6)-associated via death
domain (FADD), and Fas apoptotic
inhibitory molecule (FAIM)
Table 9.2 (Continued)
Cellular Origin of
pathway engineering gene Target gene Engineered phenotype References
CHO (knockdown) Bax and Bak Enhanced cell viability and 35% increase in Lim et al. [161]
IFN𝛾 titer
Autophagy and CHO (overexpression) BCL-xL Delayed onset of autophagy and apoptosis Kim et al. [34]
apoptosis
Human (BCL-2) and CHO BCL-2 and Beclin-1 Extended culture longevity and higher viable Lee et al. [162]
(Beclin-1) (overexpression) cell density because of decreased apoptosis
and autophagy
Proliferation Human (overexpression) Cyclin-dependent kinase like 3 Increased maximum viable cell density Jaluria et al. [163]
(CDKL3) and cytochrome c oxidase
subunit (COX15)
CHO (overexpression) Valosin-containing protein (VCP) Increased cell proliferation and viability Doolan et al. [96]
CHO (knockdown) Cofilin (CFL1) 65% (SEAP) and 47% (tPA) increase in Hammond and Lee
specific productivity [134]
miRNA Human (overexpression) miR-557 Enhanced cell growth Strotbek et al. [60]
Fischer et al. [124]
CHO (overexpression) miR-30a Enhanced cell growth Fischer et al.
[47, 115]
9.3 Applications of Cell Line Engineering Approaches in CHO Cells 225
Origin of
Cellular pathway engineering gene Target gene Engineered phenotype References
Glycosylation Human (overexpression) Alpha 2,6 sialyltransferase (ST6GAL) Expression of partly 𝛼2,6-sialylated Lee et al. [199]
recombinant proteins Zhang et al. [200]
Bragonzi et al. [201]
Rat (overexpression) ST6GAL Expression of 𝛼2,6-sialylated recombinant Minch et al. [202]
human tissue plasminogen activator (tPA)
Bovine (GnT-IV ) and 𝛼-1,3-d-mannoside 𝛽 1,4 56.2% Increase in tetra-antennary sugar Fukuta et al. [203]
human (GnT-V ) N-acetylglucosaminyltransferase chains on recombinant IFN𝛾
(overexpression) (GnT-IV) and 𝛼-1,6 d-mannoside 𝛽-1,6
N-acetylglucosaminyltransferase
(GnT-V)
Mouse (ST3GAL), rat ST3GAL, ST6GAL, and GnT-V Increase in the extent of sialylation of Fukuta et al. [204]
(ST6GAL) and human human recombinant IFN𝛾 of up to 80%
(GnT-V )
(overexpression)
Rat (overexpression) ST6GAL Improved sialylation and therapeutic Jassal et al. [205]
activity of recombinant IgG3
Human (overexpression) 𝛼-2,3 sialyltransferase (ST3GAL) and 𝛽 Expression of homogeneously distributed Weikert et al. [206]
1,4 galactosyltransferase (GalT) and >90% sialylated glycoproteins
Human (overexpression) ST6GAL and GalT Increased sialylation level of recombinant Jeong et al. [98]
human EPO
CHO (overexpression) CMP-sialic acid transporter (CMP-SAT) 4–16% Increased sialylation of human IFN𝛾 Wong et al. [207]
Human (overexpression) CMP-sialic acid synthetase (CMP-SAS), Further enhanced sialylation of Jeong et al. [208]
CMP-SAT, and ST3GAL recombinant human EPO
CHO (CMP-SAT) and Mutant uridine diphosphate-N-acetyl >10-Fold increase in CMP-sialic acid Son et al. [209]
human (ST3GAL) glucosamine 2-epimerase (GNE), concentration leading to a 32% increase in
(overexpression) CMP-SAT, and ST3GAL sialylation of human recombinant EPO
Rat (overexpression) 𝛽-1,4 N-acetylglucosaminyltransferase Expression of antibodies with increased Davies et al. [210]
III (GnT-III) bisecting glycan chains that resulted in a
20-fold lower antibody dosage with high
ADCC
Rat (GnT-III) and human GnT-III and Golgi 𝛼-mannosidase II Expression of nonfucosylated antibodies Ferrara et al. [211]
(ManII) (overexpression) (ManII) possessing N-glycans of the complex type
Human (overexpression) 𝛽-1,6 N-acetylglucosaminyl-transferase Redirection of the O-glycosylation pathway Prati et al. [212]
(C2GnT) in CHO cells
CHO (knockout) 𝛼-1,6-fucosyltransferase (FUT8) Production of completely nonfucosylated Yamane-Ohnuki
antibodies resulting in 100-fold enhanced et al. [213]
antibody-dependent cellular cytotoxicity
(ADCC)
CHO (knockout) FUT8 Production of completely nonfucosylated Malphettes et al.
antibodies with enhanced ADCC [214]
CHO (knockout) FUT8 Genomic knockout of FUT8 with Cristea et al. [215]
simultaneous integration of an antibody
expression cassette
CHO (knockout) N-acetylglucosaminyl-transferase 1 Production of recombinant proteins with Zhong et al. [216]
(MGAT1) Man5 as the predominant N-linked Sealover et al. [217]
glycosylation species
CHO (knockout) GDP-fucose transporter (SLC35C1) and Production of recombinant antibodies Zhang et al. [218]
CMP-sialic acid transporter (SLC35A1) lacking both fucose and sialic acid to
increase ADCC
CHO (knockout) MGAT4A, 4B, and 5 Expression of EPO with almost Yang et al. [219]
homogeneous biantennary N-glycans with a
minor amount of
poly-N-acetyl-lactosamine (poly-LacNAc)
CHO (knockout) MGAT4A/4B/5 and 𝛽-1,4 >90% Reduction in galactosylation of Yang et al. [219]
galactosyltransferase 1 (B4GALT1) recombinant EPO
CHO (knockout) 𝛽-1,3 N-acetylglucosaminyltransferase 2 Expression of recombinant EPO lacking Yang et al. [219]
(B3GNT2) poly-LacNAc
Table 9.3 (Continued)
Origin of
Cellular pathway engineering gene Target gene Engineered phenotype References
CHO (knockout) ST3GAL4/6 and MGAT4A/4B/5 Expression of recombinant EPO with Yang et al. [219]
heterogeneous tetra-antennary N-glycans
without sialylation
CHO (knockout) FUT8 and B4GALT1 Expression of an IgG1 with homogenous Yang et al. [219]
biantennary N-glycans without fucose and
almost no galactose
CHO (knockdown) Sialidase (NEU2) 60% Decrease in sialidase activity led to Ferrari et al. (1998)
increased sialic acid content in IFN𝛾 Ngantung et al.
[176]
CHO (knockdown) Sialidases NEU1 and 3 98% Decrease in sialidase activity led to Zhang et al. [182]
26–33% increase in sialic acid content of
IFN𝛾
CHO (knockdown) 𝛼-1,6-fucosyltransferase (FUT8) Reduction in core fucose by 60–88% Mori et al. [175]
resulted in 100-fold improved ADCC of the Beuger et al. [168]
produced IgG
CHO (knockdown) GDP-fucose 4,6-dehydratase (GMD) Production of 100% defucosylated Kanda et al. [172]
recombinant antibodies if culture medium
lacks l-fucose
CHO (knockdown) FUT8 and GMD Production of fully nonfucosylated Imai-Nishiya et al.
antibodies with improved ADCC [220]
CHO (knockdown) GDP-fucose transporter (GFT) 10–40% Increase in defucosylated AT-III Omasa et al. [73]
Posttranslational CHO (knockout) Peptidylglycine 𝛼-amidating Reduction in C-terminal amidated species Skulj et al. [221]
modifications monooxygenase (PAM) of recombinant monoclonal antibodies
Drug product CHO (knockout) Lipoprotein lipase (LPL) Removal of LPL host cell protein decreased Chiu et al. [222]
stability polysorbate-20 and -80 degradation and
increased drug product stability
Viral resistance CHO (knockout) CMP sialic acid transporter (SLC35A1) Increased resistance to MVM virus Mascarenhas et al.
infection [223]
9.3 Applications of Cell Line Engineering Approaches in CHO Cells 231
9.4 Conclusions
Isolated more than 60 years ago, various sublines of the original CHO cell line
have found a solid place both within the biopharmaceutical industry and in
academic research laboratories all over the world. There are certainly numerous
key factors that have eventually contributed to the great success of CHO cells
234 9 CHO Cell Engineering for Improved Process Performance and Product Quality
References
1 Kunert, R. and Reinhart, D. (2016). Advances in recombinant antibody man-
ufacturing. Appl. Microbiol. Biotechnol. 100: 3451–3461.
2 Lalonde, M.E. and Durocher, Y. (2017). Therapeutic glycoprotein production
in mammalian cells. J. Biotechnol. 251: 128–140.
3 Wells, E. and Robinson, A.S. (2017). Cellular engineering for therapeu-
tic protein production: product quality, host modification, and process
improvement. Biotechnol. J. 12 (1): https://doi.org/10.1002/biot.201600105.
4 Urlaub, G. and Chasin, L.A. (1980). Isolation of Chinese hamster cell
mutants deficient in dihydrofolate reductase activity. Proc. Natl. Acad. Sci.
U.S.A. 77: 4216–4220.
5 Urlaub, G., Kas, E., Carothers, A.M., and Chasin, L.A. (1983). Deletion of
the diploid dihydrofolate reductase locus from cultured mammalian cells.
Cell 33: 405–412.
6 Wurm, F.M. and Hacker, D. (2011). First CHO genome. Nat. Biotechnol. 29:
718–720.
7 Cockett, M.I., Bebbington, C.R., and Yarranton, G.T. (1990). High level
expression of tissue inhibitor of metalloproteinases in Chinese hamster
References 235
58 Bratkovic, T., Glavan, G., Strukelj, B. et al. (2012). Exploiting microRNAs for
cell engineering and therapy. Biotechnol. Adv. 30: 753–765.
59 Jadhav, V., Hackl, M., Bort, J.A. et al. (2012). A screening method to assess
biological effects of microRNA overexpression in Chinese hamster ovary
cells. Biotechnol. Bioeng. 109: 1376–1385.
60 Strotbek, M., Florin, L., Koenitzer, J. et al. (2013). Stable microRNA expres-
sion enhances therapeutic antibody productivity of Chinese hamster ovary
cells. Metab. Eng. 20: 157–166.
61 Klanert, G., Jadhav, V., Chanoumidou, K. et al. (2014). Endogenous
microRNA clusters outperform chimeric sequence clusters in Chinese
hamster ovary cells. Biotechnol. J. 9: 538–544.
62 Hackl, M., Jakobi, T., Blom, J. et al. (2011). Next-generation sequencing
of the Chinese hamster ovary microRNA transcriptome: identification,
annotation and profiling of microRNAs as targets for cellular engineering.
J. Biotechnol. 153: 62–75.
63 Johnson, K.C., Jacob, N.M., Nissom, P.M. et al. (2011). Conserved microR-
NAs in Chinese hamster ovary cell lines. Biotechnol. Bioeng. 108: 475–480.
64 Griffiths-Jones, S., Grocock, R.J., van Dongen, S. et al. (2006). miRBase:
microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 34:
D140–D144.
65 Kozomara, A. and Griffiths-Jones, S. (2014). miRBase: annotating high con-
fidence microRNAs using deep sequencing data. Nucleic Acids Res. 42:
D68–D73.
66 Diendorfer, A.B., Hackl, M., Klanert, G. et al. (2015). Annotation of addi-
tional evolutionary conserved microRNAs in CHO cells from updated
genomic data. Biotechnol. Bioeng. 112 (7): 1488–1493.
67 Borth, N., Mattanovich, D., Kunert, R., and Katinger, H. (2005). Effect of
increased expression of protein disulfide isomerase and heavy chain binding
protein on antibody secretion in a recombinant CHO cell line. Biotechnol.
Progr. 21: 106–111.
68 Hwang, S.O., Chung, J.Y., and Lee, G.M. (2003). Effect of
doxycycline-regulated ERp57 expression on specific thrombopoietin pro-
ductivity of recombinant CHO cells. Biotechnol Prog. 19 (1): 179–184.
69 Chung, J.Y., Lim, S.W., Hong, Y.J. et al. (2004). Effect of
doxycycline-regulated calnexin and calreticulin expression on specific
thrombopoietin productivity of recombinant Chinese hamster ovary cells.
Biotechnol Bioeng. 85 (5): 539–546.
70 Underhill, M.F., Coley, C., Birch, J.R. et al. (2003). Engineering mRNA trans-
lation initiation to enhance transient gene expression in chinese hamster
ovary cells. Biotechnol Prog. 19 (1): 121–129.
71 Kwon, R.J., Kim, S.K., Lee, S.I. et al. (2006). Artificial transcription factors
increase production of recombinant antibodies in Chinese hamster ovary
cells. Biotechnol. Lett. 28: 9–15.
72 Ohya, T., Hayashi, T., Kiyama, E. et al. (2008). Improved production of
recombinant human antithrombin III in Chinese hamster ovary cells by
ATF4 overexpression. Biotechnol. Bioeng. 100: 317–324.
References 239
87 Fogolin, M.B., Wagner, R., Etcheverrigaray, M., and Kratje, R. (2004). Impact
of temperature reduction and expression of yeast pyruvate carboxylase on
hGM-CSF-producing CHO cells. J. Biotechnol. 109: 179–191.
88 Kim, S.H. and Lee, G.M. (2007). Down-regulation of lactate
dehydrogenase-A by siRNAs for reduced lactic acid formation of Chinese
hamster ovary cells producing thrombopoietin. Appl. Microbiol. Biotechnol.
74: 152–159.
89 Wlaschin, K.F. and Hu, W.S. (2007). Engineering cell metabolism for
high-density cell culture via manipulation of sugar transport. J. Biotechnol.
131: 168–176.
90 Le, H., Vishwanathan, N., Kantardjieff, A. et al. (2013). Dynamic gene
expression for metabolic engineering of mammalian cells in culture. Metab.
Eng. 20: 212–220.
91 Chong, W.P., Reddy, S.G., Yusufi, F.N. et al. (2010). Metabolomics-driven
approach for the improvement of Chinese hamster ovary cell growth: over-
expression of malate dehydrogenase II. J. Biotechnol. 147: 116–121.
92 Tabuchi, H., Sugiyama, T., Tanaka, S., and Tainaka, S. (2010). Overexpression
of taurine transporter in Chinese hamster ovary cells can enhance cell viabil-
ity and product yield, while promoting glutamine consumption. Biotechnol.
Bioeng. 107: 998–1003.
93 Tabuchi, H. and Sugiyama, T. (2013). Cooverexpression of alanine amino-
transferase 1 in Chinese hamster ovary cells overexpressing taurine trans-
porter further stimulates metabolism and enhances product yield. Biotechnol.
Bioeng. 110: 2208–2215.
94 Kim, S.H. and Lee, G.M. (2007). Functional expression of human pyruvate
carboxylase for reduced lactic acid formation of Chinese hamster ovary cells
(DG44). Appl. Microbiol. Biotechnol. 76: 659–665.
95 Jeong, D., Kim, T.S., Lee, J.W. et al. (2001). Blocking of acidosis-mediated
apoptosis by a reduction of lactate dehydrogenase activity through antisense
mRNA expression. Biochem. Biophys. Res. Commun. 289: 1141–1149.
96 Doolan, P., Meleady, P., Barron, N. et al. (2010). Microarray and pro-
teomics expression profiling identifies several candidates, including the
valosin-containing protein (VCP), involved in regulating high cellular growth
rate in production CHO cell lines. Biotechnol. Bioeng. 106: 42–56.
97 Zhou, M., Crawford, Y., Ng, D. et al. (2011). Decreasing lactate level and
increasing antibody production in Chinese Hamster Ovary cells (CHO) by
reducing the expression of lactate dehydrogenase and pyruvate dehydroge-
nase kinases. J. Biotechnol. 153: 27–34.
98 Jeong, Y.T., Choi, O., Lim, H.R. et al. (2008). Enhanced sialylation of recom-
binant erythropoietin in CHO cells by human glycosyltransferase expression.
J. Microbiol. Biotechnol. 18: 1945–1952.
99 Tigges, M. and Fussenegger, M. (2006). Xbp1-based engineering of secretory
capacity enhances the productivity of Chinese hamster ovary cells. Metab.
Eng. 8: 264–272.
References 241
100 Becker, E., Florin, L., Pfizenmaier, K., and Kaufmann, H. (2008). An XBP-1
dependent bottle-neck in production of IgG subtype antibodies in chemi-
cally defined serum-free Chinese hamster ovary (CHO) fed-batch processes.
J Biotechnol. 135 (2): 217–223. https://doi.org/10.1016/j.jbiotec.2008.03.008.
101 Ku, S.C., Ng, D.T., Yap, M.G., and Chao, S.H. (2008). Effects of overex-
pression of X-box binding protein 1 on recombinant protein production
in Chinese hamster ovary and NS0 myeloma cells. Biotechnol. Bioeng. 99:
155–164.
102 Gulis, G., Simi, K.C., de Toledo, R.R. et al. (2014). Optimization of heterol-
ogous protein production in Chinese hamster ovary cells under overexpres-
sion of spliced form of human X-box binding protein. BMC Biotechnol. 14:
26. https://doi.org/10.1186/1472-6750-14-26.
103 Peng, R.W. and Fussenegger, M. (2009). Molecular engineering of exocytic
vesicle traffic enhances the productivity of Chinese hamster ovary cells.
Biotechnol. Bioeng. 102: 1170–1181.
104 Florin, L., Pegel, A., Becker, E. et al. (2009). Heterologous expression of the
lipid transfer protein CERT increases therapeutic protein productivity of
mammalian cells. J. Biotechnol. 141: 84–90.
105 Rahimpour, A., Vaziri, B., Moazzami, R. et al. (2013). Engineering the cel-
lular protein secretory pathway for enhancement of recombinant tissue
plasminogen activator expression in Chinese hamster ovary cells: effects of
CERT and XBP1s genes. J. Microbiol. Biotechnol. 23: 1116–1122.
106 Peng, R.W., Abellan, E., and Fussenegger, M. (2011). Differential effect of
exocytic SNAREs on the production of recombinant proteins in mammalian
cells. Biotechnol. Bioeng. 108: 611–620.
107 Le Fourn, V., Girod, P.A., Buceta, M. et al. (2014). CHO cell engineering to
prevent polypeptide aggregation and improve therapeutic protein secretion.
Metab. Eng. 21: 91–102.
108 Fussenegger, M., Schlatter, S., Datwyler, D. et al. (1998). Controlled prolif-
eration by multigene metabolic engineering enhances the productivity of
Chinese hamster ovary cells. Nat. Biotechnol. 16: 468–472.
109 Astley, K., Naciri, M., Racher, A., and Al-Rubeai, M. (2007). The role of
p21cip1 in adaptation of CHO cells to suspension and protein-free culture.
J Biotechnol. 130 (3): 282–290.
110 Mazur, X., Fussenegger, M., Renner, W.A., and Bailey, J.E. (1998). Higher
productivity of growth-arrested Chinese hamster ovary cells expressing the
cyclin-dependent kinase inhibitor p27. Biotechnol. Progr. 14: 705–713.
111 Bi, J.X., Shuttleworth, J., and Al-Rubeai, M. (2004). Uncoupling of cell
growth and proliferation results in enhancement of productivity in
p21CIP1 -arrested CHO cells. Biotechnol. Bioeng. 85: 741–749.
112 Kuystermans, D. and Al-Rubeai, M. (2009). cMyc increases cell number
through uncoupling of cell division from cell size in CHO cells. BMC
Biotechnol. 9: 76.
113 Lee, J.S., Ha, T.K., Park, J.H., and Lee, G.M. (2013). Anti-cell death engi-
neering of CHO cells: co-overexpression of Bcl-2 for apoptosis inhibition,
Beclin-1 for autophagy induction. Biotechnol Bioeng. 110 (8): 2195–2207.
https://doi.org/10.1002/bit.24879.
242 9 CHO Cell Engineering for Improved Process Performance and Product Quality
114 Sanchez, N., Kelly, P., Gallagher, C. et al. (2013). CHO cell culture longevity
and recombinant protein yield are enhanced by depletion of miR-7 activity
via sponge decoy vectors. Biotechnol. J. 9 (3): 396–404.
115 Fischer, S., Paul, A., Wagner, A. et al. (2015). miR-2861 as novel HDAC5
inhibitor in CHO cells enhances productivity while maintaining product
quality. Biotechnol. Bioeng. 112 (10): 2142–2153.
116 Clarke, C., Henry, M., Doolan, P., et al. (2012). Integrated miRNA, mRNA
and proteinexpression analysis reveals the role of post-transcriptional reg-
ulation incontrolling CHO cell growth rate. BMCGenomics. 13:656. doi:10
.1186/1471-2164-13--656.
117 Jadhav, V., Hackl, M., Klanert, G. et al. (2014). Stable overexpression of
miR-17 enhances recombinant protein production of CHO cells. J. Biotech-
nol. 175: 38–44.
118 Loh, W.P., Loo, B., Zhou, L. et al. (2014). Overexpression of microRNAs
enhances recombinant protein production in Chinese hamster ovary cells.
Biotechnol. J. 9 (9): 1140–1151.
119 Loh, W.P., Yang, Y., and Lam, K.P. (2017). miR-92a enhances recombinant
protein productivity in CHO cells by increasing intracellular cholesterol
levels. Biotechnol. J. 12 (4): https://doi.org/10.1002/biot.201600488.
120 Kelly, P.S., Gallagher, C., Clynes, M., and Barron, N. (2015). Conserved
microRNA function as a basis for Chinese hamster ovary cell engineering.
Biotechnol. Lett. 37 (4): 787–798.
121 Emmerling, V.V., Fischer, S., Stiefel, F. et al. (2016). Temperature-sensitive
miR-483 is a conserved regulator of recombinant protein and viral vector
production in mammalian cells. Biotechnol. Bioeng. 113 (4): 830–841.
122 Kelly, P.S., Breen, L., Gallagher, C. et al. (2015). Re-programming CHO
cell metabolism using miR-23 tips the balance towards a highly productive
phenotype. Biotechnol. J. 10: 1029–1040.
123 Schoellhorn, M., Fischer, S., Wagner, A. et al. (2017). miR-143 targets
MAPK7 in CHO cells and induces a hyperproductive phenotype to
enhance production of difficult-to-express proteins. Biotechnol. Progr. 33
(4): 1046–1058.
124 Schoellhorn, M., Fischer, S., Wagner, A. et al. (2017). miR-143 targets
MAPK7 in CHO cells and induces a hyperproductive phenotype to
enhance production of difficult-to-express proteins. Biotechnol Prog. 33
(4): 1046–1058. https://doi.org/10.1002/btpr.2475.
125 Neermann, J. and Wagner, R. (1996). Comparative analysis of glucose and
glutamine metabolism in transformed mammalian cell lines, insect and
primary liver cells. J. Cell. Physiol. 166: 152–169.
126 Yip, S.S., Zhou, M., Joly, J. et al. (2014). Complete knockout of the lactate
dehydrogenase A gene is lethal in pyruvate dehydrogenase kinase 1, 2, 3
down-regulated CHO cells. Mol. Biotechnol. 56 (9): 833–838.
127 Kim, J.Y., Kim, Y.G., Han, Y.K. et al. (2011). Proteomic understanding of
intracellular responses of recombinant Chinese hamster ovary cells cul-
tivated in serum-free medium supplemented with hydrolysates. Appl.
Microbiol. Biotechnol. 89: 1917–1928.
References 243
128 Carlage, T., Hincapie, M., Zang, L. et al. (2009). Proteomic profiling of
a high-producing Chinese hamster ovary cell culture. Anal. Chem. 81:
7357–7362.
129 Charaniya, S., Karypis, G., and Hu, W.S. (2009). Mining transcriptome data
for function-trait relationship of hyper productivity of recombinant antibody.
Biotechnol. Bioeng. 102: 1654–1669.
130 Fomina-Yadlin, D., Mujacic, M., Maggiora, K. et al. (2015). Transcriptome
analysis of a CHO cell line expressing a recombinant therapeutic protein
treated with inducers of protein expression. J. Biotechnol. 212: 106–115.
131 Kantardjieff, A., Jacob, N.M., Yee, J.C. et al. (2010). Transcriptome and pro-
teome analysis of Chinese hamster ovary cells under low temperature and
butyrate treatment. J. Biotechnol. 145: 143–159.
132 Yee, J.C., Gerdtzen, Z.P., and Hu, W.S. (2009). Comparative transcriptome
analysis to unveil genes affecting recombinant protein productivity in mam-
malian cells. Biotechnol. Bioeng. 102: 246–263.
133 Hayduk, E.J. and Lee, K.H. (2005). Cytochalasin D can improve heterologous
protein productivity in adherent Chinese hamster ovary cells. Biotechnol.
Bioeng. 90: 354–364.
134 Hammond, S. and Lee, K.H. (2012). RNA interference of cofilin in Chinese
hamster ovary cells improves recombinant protein productivity. Biotechnol.
Bioeng. 109: 528–535.
135 Ritter, A., Rauschert, T., Oertli, M. et al. (2016). Disruption of the gene
C12orf35 leads to increased productivities in recombinant CHO cell lines.
Biotechnol. Bioeng. 113: 2433–2442.
136 Barron, N., Kumar, N., Sanchez, N. et al. (2011). Engineering CHO cell
growth and recombinant protein productivity by overexpression of miR-7.
J. Biotechnol. 151: 204–211.
137 Fischer, S., Handrick, R., and Otte, K. (2015). The art of CHO cell engineer-
ing: a comprehensive retrospect and future perspectives. Biotechnol. Adv. 33:
1878–1896.
138 Al-Rubeai, M., Singh, R.P., Goldman, M.H., and Emery, A.N. (1995). Death
mechanisms of animal cells in conditions of intensive agitation. Biotechnol.
Bioeng. 45: 463–472.
139 Rangan, S., Kamal, S., Konorov, S.O. et al. (2018). Types of cell death and
apoptotic stages in Chinese Hamster Ovary cells distinguished by Raman
spectroscopy. Biotechnol. Bioeng. 115: 401–412.
140 Tey, B.T., Singh, R.P., Piredda, L. et al. (2000). Influence of BCL-2 on cell
death during the cultivation of a Chinese hamster ovary cell line expressing
a chimeric antibody. Biotechnol. Bioeng. 68: 31–43.
141 Mastrangelo, A.J., Hardwick, J.M., Bex, F., and Betenbaugh, M.J. (2000).
Part I. BCL-2 and BCL-XL limit apoptosis upon infection with alphavirus
vectors. Biotechnol. Bioeng. 67: 544–554.
142 Meents, H., Enenkel, B., Eppenberger, H.M. et al. (2002). Impact of coex-
pression and coamplification of sICAM and antiapoptosis determinants
bcl-2/bcl-x(L) on productivity, cell survival, and mitochondria number in
CHO-DG44 grown in suspension and serum-free media. Biotechnol Bioeng.
80 (6): 706–716.
244 9 CHO Cell Engineering for Improved Process Performance and Product Quality
143 Sauerwald, T.M., Betenbaugh, M.J., and Oyler, G.A. (2002). Inhibiting
apoptosis in mammalian cell culture using the caspase inhibitor XIAP and
deletion mutants. Biotechnol. Bioeng. 77: 704–716.
144 Figueroa, B. Jr., Chen, S., Oyler, G.A. et al. (2004). AVEN and BCL-XL
enhance protection against apoptosis for mammalian cells exposed to vari-
ous culture conditions. Biotechnol. Bioeng. 85: 589–600.
145 Chiang, G.G. and Sisk, W.P. (2005). Bcl-x(L) mediates increased produc-
tion of humanized monoclonal antibodies in Chinese hamster ovary cells.
Biotechnol Bioeng. 91 (7): 779–792.
146 Wong, D.C., Wong, K.T., Nissom, P.M. et al. (2006). Targeting early apop-
totic genes in batch and fed-batch CHO cell cultures. Biotechnol. Bioeng. 95:
350–361.
147 Choi, S.S., Rhee, W.J., Kim, E.J., and Park, T.H. (2006). Enhancement of
recombinant protein production in Chinese hamster ovary cells through
anti-apoptosis engineering using 30Kc6 gene. Biotechnol Bioeng. 95 (3):
459–467.
148 Crea, F., Sarti, D., Falciani, F., and Al-Rubeai, M. (2006). Over-expression
of hTERT in CHO K1 results in decreased apoptosis and reduced serum
dependency. J Biotechnol. 121 (2): 109–123.
149 Figueroa, B. Jr., Ailor, E., Osborne, D. et al. (2007). Enhanced cell culture
performance using inducible anti-apoptotic genes E1B-19K and Aven in
the production of a monoclonal antibody with Chinese hamster ovary cells.
Biotechnol Bioeng. 97 (4): 877–892.
150 Arden, N., Majors, B.S., Ahn, S.H. et al. (2007). Inhibiting the apoptosis
pathway using MDM2 in mammalian cell cultures. Biotechnol Bioeng. 97 (3):
601–614.
151 Majors, B.S., Arden, N., Oyler, G.A. et al. (2008). E2F-1 overexpression
increases viable cell density in batch cultures of Chinese hamster ovary cells.
J Biotechnol. 138 (3–4): 103–106.
152 Dorai, H., Kyung, Y.S., Ellis, D. et al. (2009). Expression of anti-apoptosis
genes alters lactate metabolism of Chinese Hamster Ovary cells in culture.
Biotechnol Bioeng. 103 (3): 592–608. https://doi.org/10.1002/bit.22269.
153 Lee, Y.Y., Wong, K.T., Tan, J. et al. (2009). Overexpression of heat shock
proteins (HSPs) in CHO cells for extended culture viability and improved
recombinant protein production. J Biotechnol. 143 (1): 34–43. https://doi.org/
10.1016/j.jbiotec.2009.05.013.
154 Majors, B.S., Betenbaugh, M.J., Pederson, N.E., and Chiang, G.G. (2009).
Mcl-1 overexpression leads to higher viabilities and increased production of
humanized monoclonal antibody in Chinese hamster ovary cells. Biotechnol.
Progr. 25: 1161–1168.
155 Hwang, S.O. and Lee, G.M. (2009). Effect of Akt overexpression on pro-
grammed cell death in antibody-producing Chinese hamster ovary cells.
J. Biotechnol. 139: 89–94.
156 Majors, B.S., Chiang, G.G., Pederson, N.E., and Betenbaugh, M.J. (2012).
Directed evolution of mammalian anti-apoptosis proteins by somatic hyper-
mutation. Protein Eng. Des. Sel. 25: 27–38.
References 245
157 Tan, J.G., Lee, Y.Y., Wang, T. et al. (2015). Heat shock protein 27 overex-
pression in CHO cells modulates apoptosis pathways and delays activation
of caspases to improve recombinant monoclonal antibody titre in fed-batch
bioreactors. Biotechnol. J. 10: 790–800.
158 Cost, G.J., Freyvert, Y., Vafiadis, A. et al. (2010). BAK and BAX deletion
using zinc-finger nucleases yields apoptosis-resistant CHO cells. Biotechnol.
Bioeng. 105: 330–340.
159 Grav, L.M., Lee, J.S., 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.
160 Sung, Y.H., Lee, J.S., Park, S.H. et al. (2007). Influence of co-down-regulation
of caspase-3 and caspase-7 by siRNAs on sodium butyrate-induced apoptotic
cell death of Chinese hamster ovary cells producing thrombopoietin. Metab.
Eng. 9: 452–464.
161 Lim, S.F., Chuan, K.H., Liu, S. et al. (2006). RNAi suppression of Bax and
Bak enhances viability in fed-batch cultures of CHO cells. Metab. Eng. 8:
509–522.
162 Lee, J.S., Ha, T.K., Park, J.H., and Lee, G.M. (2013). Anti-cell death engi-
neering of CHO cells: co-overexpression of BCL-2 for apoptosis inhibition,
Beclin-1 for autophagy induction. Biotechnol. Bioeng. 110: 2195–2207.
163 Jaluria, P., Betenbaugh, M., Konstantopoulos, K., and Shiloach, J. (2007).
Enhancement of cell proliferation in various mammalian cell lines by gene
insertion of a cyclin-dependent kinase homolog. BMC Biotechnol. 7: 71.
164 Blagosklonny, M.V. (2005). Molecular theory of cancer. Cancer Biol. Ther. 4:
621–627.
165 Kaneko, Y., Sato, R., and Aoyagi, H. (2010). Changes in the quality of anti-
bodies produced by Chinese hamster ovary cells during the death phase of
cell culture. J. Biosci. Bioeng. 109: 281–287.
166 Arden, N. and Betenbaugh, M.J. (2004). Life and death in mammalian cell
culture: strategies for apoptosis inhibition. Trends Biotechnol. 22: 174–180.
167 Simon, L. and Karim, M.N. (2002). Control of starvation-induced apoptosis
in Chinese hamster ovary cell cultures. Biotechnol. Bioeng. 78: 645–657.
168 Beuger, V., Kunkele, K.P., Koll, H. et al. (2009). Short-hairpin-RNA-mediated
silencing of fucosyltransferase 8 in Chinese-hamster ovary cells for the pro-
duction of antibodies with enhanced antibody immune effector function.
Biotechnol. Appl. Biochem. 53: 31–37.
169 Ghaderi, D., Zhang, M., Hurtado-Ziola, N., and Varki, A. (2012). Production
platforms for biotherapeutic glycoproteins. Occurrence, impact, and chal-
lenges of non-human sialylation. Biotechnol. Genet. Eng. Rev. 28: 147–175.
170 Hong, W.W. and Wu, S.C. (2007). A novel RNA silencing vector to improve
antigen expression and stability in Chinese hamster ovary cells. Vaccine 25:
4103–4111.
171 Irani, N., Wirth, M., van Den Heuvel, J., and Wagner, R. (1999). Improve-
ment of the primary metabolism of cell cultures by introducing a new
cytoplasmic pyruvate carboxylase reaction. Biotechnol. Bioeng. 66: 238–246.
172 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
246 9 CHO Cell Engineering for Improved Process Performance and Product Quality
189 Lee, K.H., Onitsuka, M., Honda, K. et al. (2013). Rapid construction of
transgene-amplified CHO cell lines by cell cycle checkpoint engineering.
Appl. Microbiol. Biotechnol. 97: 5731–5741.
190 Muller, D., Katinger, H., and Grillari, J. (2008). MicroRNAs as targets for
engineering of CHO cell factories. Trends Biotechnol. 26: 359–365.
191 Druz, A., Betenbaugh, M., and Shiloach, J. (2012). Glucose depletion acti-
vates mmu-miR-466h-5p expression through oxidative stress and inhibition
of histone deacetylation. Nucleic Acids Res. 40: 7291–7302.
192 Druz, A., Chu, C., Majors, B. et al. (2011). A novel microRNA
mmu-miR-466h affects apoptosis regulation in mammalian cells. Biotechnol.
Bioeng. 108: 1651–1661.
193 Druz, A., Son, Y.J., Betenbaugh, M., and Shiloach, J. (2013). Stable inhibition
of mmu-miR-466h-5p improves apoptosis resistance and protein production
in CHO cells. Metab. Eng. 16: 87–94.
194 Cho, W.C. (2007). OncomiRs: the discovery and progress of microRNAs in
cancers. Mol Cancer. 6: 60.
195 Raver-Shapira, N., Marciano, E., Meiri, E. et al. (2007). Transcriptional acti-
vation of miR-34a contributes to p53-mediated apoptosis. Mol Cell. 26 (5):
731–743.
196 Butler, M. and Spearman, M. (2014). The choice of mammalian cell host
and possibilities for glycosylation engineering. Curr. Opin. Biotechnol. 30:
107–112.
197 Hossler, P., Khattak, S.F., and Li, Z.J. (2009). Optimal and consistent protein
glycosylation in mammalian cell culture. Glycobiology 19: 936–949.
198 Wang, Q., Stuczynski, M., Gao, Y., and Betenbaugh, M.J. (2015). Strate-
gies for engineering protein N-glycosylation pathways in mammalian cells.
Methods Mol. Biol. 1321: 287–305.
199 Lee, E.U., Roth, J., and Paulson, J.C. (1989). Alteration of terminal glycosyla-
tion sequences on N-linked oligosaccharides of Chinese hamster ovary cells
by expression of beta-galactoside alpha 2,6-sialyltransferase. J. Biol. Chem.
264: 13848–13855.
200 Zhang, X., Lok, S.H., and Kon, O.L. (1998). Stable expression of human
alpha-2,6-sialyltransferase in Chinese hamster ovary cells: functional con-
sequences for human erythropoietin expression and bioactivity. Biochim.
Biophys. Acta 1425: 441–452.
201 Bragonzi, A., Distefano, G., Buckberry, L.D. et al. (2000). A new Chinese
hamster ovary cell line expressing alpha2,6-sialyltransferase used as universal
host for the production of human-like sialylated recombinant glycoproteins.
Biochim. Biophys. Acta 1474: 273–282.
202 Minch, S.L., Kallio, P.T., and Bailey, J.E. (1995). Tissue plasminogen activator
coexpressed in Chinese hamster ovary cells with alpha(2,6)-sialyltransferase
contains NeuAc alpha(2,6)Gal beta(1,4)Glc-N-AcR linkages. Biotechnol.
Progr. 11: 348–351.
203 Fukuta, K., Abe, R., Yokomatsu, T. et al. (2000). Remodeling of sugar chain
structures of human interferon-gamma. Glycobiology 10: 421–430.
248 9 CHO Cell Engineering for Improved Process Performance and Product Quality
204 Fukuta, K., Yokomatsu, T., Abe, R. et al. (2000). Genetic engineering of CHO
cells producing human interferon-gamma by transfection of sialyltrans-
ferases. Glycoconjugate J. 17: 895–904.
205 Jassal, R., Jenkins, N., Charlwood, J. et al. (2001). Sialylation of human
IgG-Fc carbohydrate by transfected rat alpha2,6-sialyltransferase. Biochem.
Biophys. Res. Commun. 286: 243–249.
206 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.
207 Wong, N.S., Yap, M.G., and Wang, D.I. (2006). Enhancing recombinant gly-
coprotein sialylation through CMP-sialic acid transporter over expression in
Chinese hamster ovary cells. Biotechnol. Bioeng. 93: 1005–1016.
208 Jeong, Y.T., Choi, O., Son, Y.D. et al. (2009). Enhanced sialylation of recom-
binant erythropoietin in genetically engineered Chinese-hamster ovary cells.
Biotechnol. Appl. Biochem. 52: 283–291.
209 Son, Y.D., Jeong, Y.T., Park, S.Y., and Kim, J.H. (2011). Enhanced sialylation
of recombinant human erythropoietin in Chinese hamster ovary cells by
combinatorial engineering of selected genes. Glycobiology 21: 1019–1028.
210 Davies, J., Jiang, L., Pan, L.Z. et al. (2001). Expression of GnTIII in a recom-
binant anti-CD20 CHO production cell line: expression of antibodies with
altered glycoforms leads to an increase in ADCC through higher affinity for
FC gamma RIII. Biotechnol. Bioeng. 74: 288–294.
211 Ferrara, C., Brunker, P., Suter, T. et al. (2006). Modulation of therapeu-
tic antibody effector functions by glycosylation engineering: influence of
Golgi enzyme localization domain and co-expression of heterologous beta1,
4-N-acetylglucosaminyltransferase III and Golgi alpha-mannosidase II.
Biotechnol. Bioeng. 93: 851–861.
212 Prati, E.G., Matasci, M., Suter, T.B. et al. (2000). Engineering of coordinated
up- and down-regulation of two glycosyltransferases of the O-glycosylation
pathway in Chinese hamster ovary (CHO) cells. Biotechnol. Bioeng. 68:
239–244.
213 Yamane-Ohnuki, N., Kinoshita, S., Inoue-Urakubo, M. et al. (2004). Estab-
lishment 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.
214 Malphettes, L., Freyvert, Y., Chang, J. et al. (2010). Highly efficient dele-
tion of FUT8 in CHO cell lines using zinc-finger nucleases yields cells that
produce completely nonfucosylated antibodies. Biotechnol. Bioeng. 106:
774–783.
215 Cristea, S., Freyvert, Y., Santiago, Y. et al. (2013). In vivo cleavage of trans-
gene donors promotes nuclease-mediated targeted integration. Biotechnol.
Bioeng. 110: 871–880.
216 Zhong, X., Cooley, C., Seth, N. et al. (2012). Engineering novel Lec1 gly-
cosylation mutants in CHO-DUKX cells: molecular insights and effector
modulation of N-acetylglucosaminyltransferase I. Biotechnol. Bioeng. 109:
1723–1734.
References 249
217 Sealover, N.R., Davis, A.M., Brooks, J.K. et al. (2013). Engineering Chinese
hamster ovary (CHO) cells for producing recombinant proteins with sim-
ple glycoforms by zinc-finger nuclease (ZFN)-mediated gene knockout of
mannosyl (alpha-1,3-)-glycoprotein beta-1,2-N-acetylglucosaminyltransferase
(Mgat1). J. Biotechnol. 167: 24–32.
218 Zhang, P., Haryadi, R., Chan, K.F. et al. (2012). Identification of functional
elements of the GDP-fucose transporter SLC35C1 using a novel Chinese
hamster ovary mutant. Glycobiology 22: 897–911.
219 Yang, Z., Wang, S., Halim, A. et al. (2015). Engineered CHO cells for
production of diverse, homogeneous glycoproteins. Nat. Biotechnol. 33:
842–844.
220 Imai-Nishiya, H., Mori, K., Inoue, M. et al. (2007). Double knockdown of
alpha1,6-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 Biotech-
nol. 7: 84.
221 Skulj, M., Pezdirec, D., Gaser, D. et al. (2014). Reduction in C-terminal ami-
dated species of recombinant monoclonal antibodies by genetic modification
of CHO cells. BMC Biotechnol. 14: 76.
222 Chiu, J., Valente, K.N., Levy, N.E. et al. (2017). Knockout of a
difficult-to-remove CHO host cell protein, lipoprotein lipase, for improved
polysorbate stability in monoclonal antibody formulations. Biotechnol. Bio-
eng. 114: 1006–1015.
223 Mascarenhas, J.X., Korokhov, N., Burger, L. et al. (2017). Genetic engineer-
ing of CHO cells for viral resistance to minute virus of mice. Biotechnol.
Bioeng. 114: 576–588.
224 Elliott, S., Lorenzini, T., Asher, S. et al. (2003). Enhancement of therapeu-
tic protein in vivo activities through glycoengineering. Nat. Biotechnol. 21:
414–421.
225 Sinclair, A.M. and Elliott, S. (2005). Glycoengineering: the effect of gly-
cosylation on the properties of therapeutic proteins. J. Pharm. Sci. 94:
1626–1635.
226 Ronda, C., Pedersen, L.E., Hansen, H.G. 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.
227 Chan, K.F., 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.
228 Jenkins, N., Parekh, R.B., and James, D.C. (1996). Getting the glycosyla-
tion right: implications for the biotechnology industry. Nat. Biotechnol.
14: 975–981.
229 Bork, K., Horstkorte, R., and Weidemann, W. (2009). Increasing the sialyla-
tion of therapeutic glycoproteins: the potential of the sialic acid biosynthetic
pathway. J. Pharm. Sci. 98: 3499–3508.
250 9 CHO Cell Engineering for Improved Process Performance and Product Quality
230 Butler, M. (2005). Animal cell cultures: recent achievements and perspectives
in the production of biopharmaceuticals. Appl. Microbiol. Biotechnol. 68:
283–291.
231 Chung, C.Y., Wang, Q., Yang, S. et al. (2017). Integrated genome and protein
editing swaps alpha-2,6 sialylation for alpha-2,3 sialic acid on recombinant
antibodies from CHO. Biotechnol. J. 12 (2): https://doi.org/10.1002/biot
.201600502.
232 Raymond, C., Robotham, A., Spearman, M. et al. (2015). Production of
alpha2,6-sialylated IgG1 in CHO cells. mAbs 7: 571–583.
233 Haryadi, R., Zhang, P., Chan, K.F., and Song, Z. (2013). CHO-gmt5, a novel
CHO glycosylation mutant for producing afucosylated and asialylated
recombinant antibodies. Bioengineered 4: 90–94.
234 Cha, H.M., Lim, J.H., Yeon, J.H. et al. (2017). Co-overexpression of
MGAT1 and MGAT4 in CHO cells for production of highly sialylated
albumin-erythropoietin. Enzyme Microb. Technol. 103: 53–58.
235 Schjoldager, K.T., Joshi, H.J., Kong, Y. et al. (2015). Deconstruction
of O-glycosylation–GalNAc-T isoforms direct distinct subsets of the
O-glycoproteome. EMBO Rep. 16: 1713–1722.
236 Gramer, M.J. (2014). Product quality considerations for mammalian cell cul-
ture process development and manufacturing. Adv. Biochem. Eng. Biotechnol.
139: 123–166.
237 Johnson, K.A., Paisley-Flango, K., Tangarone, B.S. et al. (2007). Cation
exchange-HPLC and mass spectrometry reveal C-terminal amidation of an
IgG1 heavy chain. Anal. Biochem. 360: 75–83.
238 Tsubaki, M., Terashima, I., Kamata, K., and Koga, A. (2013). C-terminal
modification of monoclonal antibody drugs: amidated species as a general
product-related substance. Int. J. Biol. Macromol. 52: 139–147.
239 Hu, Z., Zhang, H., Haley, B. et al. (2016). Carboxypeptidase D is the only
enzyme responsible for antibody C-terminal lysine cleavage in Chinese
hamster ovary (CHO) cells. Biotechnol. Bioeng. 113: 2100–2106.
240 Moody, M., Alves, W., Varghese, J., and Khan, F. (2011). Mouse minute virus
(MMV) contamination–a case study: detection, root cause determination,
and corrective actions. PDA J. Pharm. Sci. Technol. 65: 580–588.
241 Cotmore, S.F. and Tattersall, P. (2007). Parvoviral host range and cell entry
mechanisms. Adv. Virus Res. 70: 183–232.
242 Lopez-Bueno, A., Rubio, M.P., Bryant, N. et al. (2006). Host-selected amino
acid changes at the sialic acid binding pocket of the parvovirus capsid mod-
ulate cell binding affinity and determine virulence. J. Virol. 80: 1563–1573.