Progress in fedbatch
Progress in fedbatch
https://doi.org/10.1007/s00253-022-12342-x
MINI-REVIEW
Received: 13 September 2022 / Revised: 13 December 2022 / Accepted: 15 December 2022 / Published online: 17 January 2023
© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2023
Abstract
Nearly 80% of the approved human therapeutic antibodies are produced by Chinese Hamster Ovary (CHO) cells. To achieve
better cell growth and high-yield recombinant protein, fed-batch culture is typically used for recombinant protein produc-
tion in CHO cells. According to the demand of nutrients consumption, feed medium containing multiple components in
cell culture can affect the characteristics of cell growth and improve the yield and quality of recombinant protein. Fed-batch
optimization should have a connection with comprehensive factors such as culture environmental parameters, feed com-
position, and feeding strategy. At present, process intensification (PI) is explored to maintain production flexible and meet
forthcoming demands of biotherapeutics process. Here, CHO cell culture, feed composition in fed-batch culture, fed-batch
culture environmental parameters, feeding strategies, metabolic byproducts in fed-batch culture, chemostat cultivation, and
the intensified fed-batch are reviewed.
Key points
• Fed-batch culture in CHO cells is reviewed.
• Fed-batch has become a common technology for recombinant protein production.
• Fed batch culture promotes recombinant protein production in CHO cells.
Keywords CHO cell · Process development · Recombinant therapeutic proteins · Fed-batch culture
Introduction (EMA) and the U.S Food and Drug Administration (FDA)
with thousands of billion dollars in market value (Kaplon
In recent years, with the growing demand for recombinant et al. 2020). A report has estimated that market value about
therapeutic proteins (RTPs) in the pharmaceutical market, monoclonal antibodies (mAbs) will has reached over US$
including recombinant antibody, the production of RTPs has 300 billion by 2025 (Lu et al. 2020). Among the recombinant
developed rapidly. More than 120 recombinant antibodies protein expression system, Chinese hamster ovary (CHO)
have been approved by the European Medicines Agency cells are the preferred cells for RTP production (Ritacco
et al. 2018; Gupta et al. 2021) due to several advantages:
* Tian‑Yun Wang ① not susceptible to human virus infection with high safety
[email protected] (Lalonde and Durocher 2017); ② RTPs with post- transla-
1
International Joint Research Laboratory for Recombinant tional modifications similar to those of human cells (Stach
Pharmaceutical Protein Expression System of Henan, Xinxiang et al. 2019); ③ high-density CHO cells can accommodate
Medical University, Xinxiang 453003, Henan, China suspension culture with the chemically defined serum-free
2
School of Pharmacy, Xinxiang Medical University, medium (CD-SFM) and produce proteins that secrete into
Xinxiang 453003, Henan, China the culture medium (Ritacco et al. 2018; Dahodwala and
3
School of Nursing, Xinxiang Medical University, Lee 2019).
Xinxiang 453003, Henan, China Nowadays, mAbs production in mammalian cell culture
4
School of the First Clinical College, Xinxiang Medical is mainly performed with fed-batch processes in large-scale
University, Xinxiang 453000, Henan, China bioreactors (Schellenberg et al. 2022). CHO cells are usu-
5
School of medicine, Xinxiang University, Xinxiang 453003, ally cultivated in a fed-batch mode to achieve maximum cell
Henan, China density and product titer, with either continuous or bolus
13
Vol.:(0123456789)
1064 Applied Microbiology and Biotechnology (2023) 107:1063–1075
feed addition (Romanova et al. 2022). The initial cellular fed-batch process has become an important production
growth is supported by the basal medium. Concentrated process for the large-scale production of RTPs. However,
feed medium is added in fed-batch culture to replenish maintaining high productivity of cells and quality attributes
nutrients and prolong the culture time (Ritacco et al. 2018; of protein is still a challenge. Therefore, fed-batch culture
Chee Furng Wong et al. 2005). Feed medium can avoid the has shifted from the high-yield recombinant protein merely
nutrients limitation, and it can promote higher production of to the dual goals of high-yield and high-quality production
recombinant proteins (Rish et al. 2022; Bibila and Robinson (Ha et al. 2022).
1995; Lu et al. 2013). The culture time of cells in stationary
phase after fed-batch treatment is longer than that of batch
culture (Sellick et al. 2015). Fed-batch culture requires an Feed composition in fed‑batch culture
understanding of the characteristics of cell growth and cell
metabolism, and the key process parameters that affect the Optimizing the critical feed composition and the concentra-
quality and yield of RTPs (Ghaffari et al. 2020). CHO cell tions of components are crucial to process development (Zou
grows at a high speed in exponential phase, with an acceler- et al. 2020). In fed-batch industry, Raman spectroscopy with
ated consumption of glucose and amino acids, resulting in partial least squares regression (PLSR) or multiple linear
accumulation of lactate and ammonium (Horvat et al. 2020; regression (MLR) are applied to monitor and control the
Zagari et al. 2013). In fed-batch culture, temperature, pH, concentrations of glucose and amino acids (Kozma et al.
and other process parameters also have an impact on anti- 2018). Besides Raman spectroscopy, liquid chromatography-
body expression and quality attributes (Kuwae et al. 2018; mass spectrometry (LC–MS) has been considered to analyze
Pan et al. 2017; Graham et al. 2019). Fed-batch culture can amino acids and inorganic salt (Hoang et al. 2021). For the
improve the expression and production of RTPs in CHO first time, lipidomics analysis has been performed for lipid
cells, but traditional bolus feed delivery also causes accumu- identification and lipid quantitation in fed-batch culture (Ali
lation of metabolic byproducts (Xiao et al. 2021). Therefore, et al. 2018). In addition, feed composition has been linked
a reasonable feeding strategy is quite important (Baik et al. with cell metabolism regulation to demonstrate fed-batch
2015; Zhang et al. 2013; Mellahi et al. 2019). Fed-batch culture (Braasch et al. 2021) (Fig. 1).
culture has focused on reducing the cost of RTP produc- The components of feed medium are similar to SFM,
tion, which has become the research focus in pharmaceutical including amino acids, carbon sources, inorganic salts, trace
industry (Ghaffari et al. 2020). elements, vitamins, lipids, and putrescine and yeast hydro-
lysate etc. (Table 1).
13
Applied Microbiology and Biotechnology (2023) 107:1063–1075 1065
13
1066 Applied Microbiology and Biotechnology (2023) 107:1063–1075
after 160 h and cell viability was decreased (Wilkens et al. According to the glucose content in the culture medium,
2011). During the cell culture, the minimum glucose con- the high concentration glucose solution can be added to the
centration needs to be controlled at 2 ~ 3 g/L (Sun et al. CHO cell culture medium. Because the high concentration
2021). of carbon source also leads to metabolic byproducts accu-
Glutamine is the secondary carbon source in the early mulation, so the concentration of carbon source should be
exponential phase of cells, but it increases lactate levels strictly controlled. Proper alternative carbon sources can
(Kirsch et al. 2022). It was found that the expression of avoid inhibition of cell growth by byproducts after glucose
Fc-fusion protein in CHO cells was increased by 43% and consumption.
the total sialic acid content of cells was increased by 37%
when galactose was supplemented with 40% replacement
ratio and glucose carbon source was replaced (Xiao et al. Inorganic salts and trace elements
2019). CHO cells transfected with GLUT5 fructose trans-
porter were cultivated in fed-batch culture system, and feed Fed-batch culture also requires inorganic salts to regulate
medium containing 14.5 g/L fructose was added. Fructose pH and osmolality in cell culture environment. Zhu et al.
improved cell growth, and it decreased accumulation of lac- used 2L bioreactor for fed-batch culture, and they added Na+
tate. Wlaschin et al. indicated that CHO cells had an ability to control osmolality and partial pressure of CO2 (pCO2).
to replace carbon source through the expression of GLUT5 Both 140 ~ 160 mmHg pCO2 and 400 mOsm/kg osmolality
fructose transporter (Wlaschin and Hu 2007). decreased the viable cell density (VCD) of CHO cells by
13
Applied Microbiology and Biotechnology (2023) 107:1063–1075 1067
18%, but these factors clearly augmented the production of mL and the antibody titer of 378 mg/L, which was threefold
protein B1 (Zhu et al. 2005). In cell culture, selenium (Se) higher than batch culture (Zhang et al. 2013). Therefore, lipid
in the form of selenite can protect cells from oxidative dam- can appropriately supplement in feed medium.
age. Zhang et al. found that the VCD exceeded 1 × 107 cells/
mL and product titer of approximately 3 g/L was achieved Other components
in 14-day fed-batch cultures with the addition of selenite
(Zhang et al. 2006). Putrescine is essential for cell growth (Jänne et al. 2004).
Gangwar et al. found that Zn, Cu, Fe, and Mn impact Zhang et al. confirmed that putrescine was designed by PB
charge heterogeneity, and reduce accumulation of meta- design and it sharply increased mAb production. A signifi-
bolic byproducts in cell culture. Fe, Mn, and Ni enhance cant increase in TNFR-Fc production was observed after
the acidic variants while Cu inhibits it. Zn reduces the basic adding feed medium with the concentration of 41.67 mg/L
variants (Gangwar et al. 2021). Chung et al. found that Fer- of putrescine (Zhang et al. 2013). Jardon et al. found that
rous sulfate ( Fe2+) in feed medium stored for 7 weeks can autophagy negatively affected recombinant proteins pro-
reduce the charge heterogeneity, but it improved the yield of duction. The autophagy inhibitor 3-methyl adenine (3-MA)
mAb expressed by CHO cells (Chung et al. 2019). Graham in fed-batch culture resulted in a 2.8-fold increase in t-PA
et al. found that both 50 and 100 μM zinc ions ( Zn2+) added production compared with control group without 3-MA
in bioreactor promoted the production of β-Glucuronidase (Jardon et al. 2012). Autophagy-inducing peptide (AIP),
(GUS) in CHO cells and harvested 2 times increase in spe- a derivative of the autophagy protein Beclin 1, facilitated
cific activity of GUS (Graham et al. 2020). 5 μM copper the production of mAbs in CHO cells by regulating cel-
sulfate (Cu2+) in feed medium obtained the final mAb titer lular autophagy. The addition of 3 µM of AIP in fed-batch
of 11.9 ± 0.6 g/L to express IgG1 in CHO cells, which was culture ameliorated the expression level of human IgG1,
4.8 times higher than the final mAb titer of conventional and IgG1 concentration exceeded 1200 µg/mL (Braasch
fed-batch culture (Xu et al. 2016). et al. 2021). The yield of Fc-fusion protein (Fc) in fed-
batch culture raised twofold by Yeast hydrolysate (YE),
and YE also increased CHO cells size and intracellular
Vitamin nucleotide content (Hu et al. 2018). CHO cells expressing
human mAb and Fab fragments were cultivated in 14-day
Vitamin enhances productivity in CHO cells, and impacts fed-batch culture, and the concentration of 25 mg/L deox-
charge heterogeneity. Gangwar et al. also indicated that yuridine was added at day 2. VCD and protein produc-
vitamins can inhibit the acidic variants while it enhances tion were increased. Takagi et al. harvested 9.2 g/L human
the basic variants (Gangwar et al. 2021). Hou et al. found mAb and over 4 g/L Fab fragment antibody with deoxyu-
that increasing rate of vitamin C and nicotinamide feeding ridine, thymidine, and deoxycytidine (Takagi et al. 2017).
can reduce the phosphorylation level in CHO cells to 3% Since deoxyuridine, thymine, and deoxycytidine are all
and attenuated the hydroxylation level to 9.4% (Hou et al. pyrimidine nucleotides, exogenous nucleoside may give
2019). Adding B vitamins including 0.005 g/L vitamin B2, a platform for efficient antibody production by providing
0.0035 g/L pyridoxine (vitamin B6), 0.03 g/L pyridoxal nucleosides precursors through the remedial synthesis
(vitamin B6), 0.0985 g/L folic acid and 0.024 g/L vitamin pathway of pyrimidine nucleotides. Aki et al. screened
B12 can improve packed cell volume (PCV) and antibody 4-(2,5-dimethyl-1H-pyrrol-1-yl) -N- (2,5-dioxopyrroli-
titer (Vijayasankaran et al. 2013). Lee et al. added 10 g/L din-1-yl) benzamide (MPPB). The mAb concentration
vitamin K solution prepared with 13% polysorbate 20 (PS20) of MPPB treatment was increased to 1098 mg/L, which
in 4-day fed-batch culture for cultured CHO cells expressing was 1.5-fold higher than the mAb concentration of control
recombinant human factor II (rhFII). When the final concen- group (Aki et al. 2021). Further studies in fed-batch culture
tration of vitamin K was 0.1 mg/L, gamma-carboxygluta- facilitate the development of more additives, besides these
mate (Gla) level was decreased (Lee et al. 2017a). above components.
13
1068 Applied Microbiology and Biotechnology (2023) 107:1063–1075
in CHO cells (Schmid et al. 2022). Cells are expanded by galactosylation, and increased the proportion of high-
the cryovial or shake flask, and then grows in bioreactor mannose glycosylation (man5) (Qin et al. 2019). Xiao
(Hernández Rodríguez and Frahm 2020; Schmid et al. et al. found that continuous feeding resulting in osmo-
2022). For fed-batch culture, culture environmental param- lality below 450 mOsm/kg reduced the high-mannose
eters in bioreactor are usually optimized through design-of- glycosylation to less than 5% and increase the yield of
experiments (DoE) or one-factor-at-a-time (OFAT) studies, mAb to 10 g/L (Xiao et al. 2021). Lee et al. investigated
like temperature, dissolved oxygen (DO), and pH (Weng the effect of high osmolality on protein glycosylation in
et al. 2020; Schneider et al. 2019). CHO cells. Osmolality reached over 400 mOsm /kg after
adding betaine, and Fc-fusion protein sialylation in CHO
cells decreased (Lee et al. 2017b). Inorganic salt and high
Culture temperature feed volume can lead to high osmolality in fed-batch cul-
ture. Although high osmolality increased the cell-spe-
Culture temperature downshifting is generally performed cific productivity (qp) of CHO cells, but it increased the
in exponential growth phase. The culture temperature is proportion of high mannose and inhibited the growth of
switched from 37 ℃ to a lower temperature ranged from CHO cells. Romanova et al. launched proteome study of
29 ℃ to 35 ℃ for the sake of decreasing cell apoptosis and hyperosmolality-exposed CHO cells together with control
promoting antibody synthesis (Moore et al. 1997; Kou et al. cells, and harvested cell samples at days 2, 6, and 8 in
2011). Low temperature has a positive impact on integral fed-batch culture, respectively. Whole protein lysates of
cell density, cell viability, and cell-specific productivity hyperosmolality-exposed cell group and control cell group
(McHugh et al. 2020). sampled on days 2, 6, and 8 were analyzed by western
Torres et al. cultured CHO cell expressing human eryth- blot. Septins protein was obviously up-regulated by about
ropoietin (hEPO) using a fed-batch culture process. They 2-threefold at day 6 after treated with hyperosmolality,
observed that low temperature not only had the advantage and up-regulated about 1-threefold at day 8 (Romanova
of improving the specific productivity of CHO cell express- et al. 2022).
ing recombinant proteins, but also significantly increased Exposure of CHO cell to highly concentrated feed
the gene encoding the specific transcription activator of medium causes an increasing osmolality over 300 mOsm/
unfolded protein response, reducing the possibility of protein kg in respect of cell physiology, morphology, and proteome
degradation; Hypothermia also enhanced the expression of (Romanova et al. 2022). In fed-batch culture, appropri-
hEPO by regulating the cell cycle (Torres et al. 2021). Cell ate additives can obtain feasible osmolality. Furthermore,
culture temperature can significantly affect the VCD and an available feeding strategy is also used for avoiding
protein expression in CHO cells. When CHO cells express- hyperosmolality.
ing anti-CD20 mAb were cultured at 35 °C, 33 °C, and 31 °C
with the addition of feed medium at days 3, 6, and 9, 35 °C
can increase cell density and expression of anti-CD20 mAb
(Kong et al. 2020). Stiefel et al. that the effect of shifting pH
temperature from 37 °C to 30 °C on miRNA expression in
CHO cells was investigated. Low temperature condition The pH of cell culture requires optimization to produce
increased IgG production through upregulating the expres- high-quality recombinant protein. The pH of CHO cell
sion of miRNAs related with inhibition of apoptosis, pro- growth ranges from 7.2 to 7.6 (Michl et al. 2019). CHO cells
motion of cell growth and protein production in CHO cells expressing mAb against vascular endothelial growth factor
(Stiefel et al. 2016). Thus, low temperature is one of the (VEGF-MA) were cultured in 3 L bioreactor and the culture
most normal factors of cell culture. conditions were optimized. The pH was adjusted by adding
sodium bicarbonate. When the pH value was set to 7.10,
the yield of VEGF-MA increased to 4.1 g/L, and the charge
Osmolality heterogeneity, glycosylation level and protein purity were
26.1%, 59.1%, and 95.1% respectively (Feng and Shi 2020).
Nutrient supplementation and metabolic byproducts accu- It is necessary to follow the standard pH range of CHO cells
mulation are able to increase extracellular osmolality at when it comes to designing the pH in fed-batch culture. If
the late stages of culture in fed-batch culture (Alhuthali the pH of cell culture maintains below 6.8 or above 7.6,
et al. 2021). Both nutrient and alkaline supplemented CHO cell growth is negatively affected. The pH of cell cul-
can increase the osmolality of the culture environment ture is usually interactively regulated by C
O2 and inorganic
(Ha et al. 2022). In fed-batch process, NaCl maintained salts to improve CHO cell growth, which can achieve large-
high osmolality. It increased the titer of mAb, reduced scale recombinant proteins production.
13
Applied Microbiology and Biotechnology (2023) 107:1063–1075 1069
Dissolved oxygen 437 mg/L (Yuan et al. 2020). In order to improve the
expression of human mouse chimeric anti epidermal
DO is one of the key environmental parameters in large-scale growth factor receptor variant III (EGFR vIII) antibody
process, which affects cell growth and recombinant protein C12 in CHO cells, continuous feeding strategy was
production (Chotigeat et al. 1994; Restelli et al. 2006). Most adopted and feed medium was added to the bioreactor
of DO values in CHO cells are usually in the range of 20 to from day 3. Compared with bolus feeding methods, the
50% (Fleischaker and Sinskey 1981). Ventini et al. reported continuous feeding method reduced the ratio of cell meta-
that the final recombinant thyroid-stimulating hormone bolic byproducts, culture osmolality and high mannose
(rTSH) titer was 1.6 mg/L when DO value was 20% in fed- type, improved the VCD and viability of CHO cells, and
batch culture mode (Ventini et al. 2011). To improve the the antibody production exceeds 10 g/L (Xiao et al. 2021).
yield and quality of human IgG1 mAb, Mahé et al. adjusted Horvat et al. used continuous feeding to optimize fed-batch
the DO value to 40%, and CHO cells expressing human IgG1 culture process to culture CHO cells. From day 4, glucose
mAb were cultured in fed-batch culture supplied with Cop- solution with a concentration higher than 400 g/L was sup-
per Acetate and Ferric Citrate. The yield of mAb reached plemented to keep final glucose concentration less than
more than 10 g/L in 14 days fed-batch culture, Copper Ace- 5.55 mmol/L. From day 5, feed medium containing serine
tate and Ferric Citrate diminished man5 content and acidic was continuously added (Horvat et al. 2020). Continuous
charge variants of mAb and increased the proportion of G1F feeding can reduce the osmolality and mannosylation ratio
(Mahé et al. 2022). of the cell culture, but it increased the mAb yield. If the
VCD and cell viability of CHO cells show a downtrend in
the late exponential phase or in the early stationary phase,
feed medium should be added in cell culture. Great feed
Feeding strategy strategy has a positive impact on RTP production.
Monitoring and control system is introduced in fed-batch
In addition to adjusting the composition of feed medium culture, which maintains key cellular nutrients at an ideal
and setting reasonable process parameters, it is neces- level. Bolus feed delivery strategy is widely used for fed-
sary to design and develop available feeding strategy to batch culture, but it cannot adjust the medium concentration
produce the high-yield and high-quality of recombinant to the dynamic nutrient consumption rate of cells at different
proteins (Horvat et al. 2020). The control criterion and the metabolic stages (Xie and Wang 1994a, b).
mode of feeding should be taken into consideration in the Raman spectroscopy is used for monitoring nutrient and
aspect of feeding strategy. Keeping the concentration of byproducts (glucose, lactate, and amino acid) (Domján et al.
one or two main nutrients at an ideal level is completed 2022). Dynamic feeding strategies are used to control the deliv-
by control criterion. According to the stoichiometric feed ery of feed medium via Raman spectroscopy. In an early phase
composition, the rest of nutrients are added to culture of the development, Domján et al. established a partial least
system (Costa et al. 2014). Feeding strategy is usually squares (PLS) calibration model that took the cell metabolic
divided into two addition methods: continuous feeding behavior and nutritional consumption into account. Subse-
and bolus feeding. Bolus feeding has the advantages of quently, the nutrients are maintained at the desired level (Dom-
simple operation, low facility requirements and low pro- ján et al. 2022). For mAb production in CHO cells, Eyster et al.
duction costs. Bolus feed delivery strategy increases the developed Raman spectroscopy to control lactate and glucose
expression of mAbs in CHO cells and is the most widely in fed-batch culture, and established PLS model for predict-
used fed-batch culture method for large-scale mammalian ing glucose and lactate concentrations. Three feeding strate-
cell culture. However, the addition of a large amount of gies were evaluated in terms of cell metabolism, productiv-
feed medium leads to accumulation of metabolic byprod- ity, and product quality. Ammonium levels were reduced by
ucts and the increase of osmolality. Continuous feeding 68% through controlling lactate at 2 g/L, while mAb galacto-
can deal with two problems above, and it can adjust the sylation levels were increased by approximately 50% (Eyster
feeding rate to maintain the concentration of nutrients, et al. 2021) .
which is based on the nutrients consumption. Therefore,
continuous feeding has more advantages in the aspect of
cell culture (Xiao et al. 2021; Kamachi and Omasa 2018). Metabolic byproducts in fed‑batch culture
To promote the high expression of anti-HER2 mAb in
CHO cells, Feed 4 was added at 5% of the initial culture In the late stages of cell culture, fed-batch culture usually
volume at days 3, 5, 7, and 9. Peak VCD was 17.1 × 106 stores byproducts such as lactate and ammonium (Brun-
cells/mL in fed-batch culture and mAb yield reached ner et al. 2017; Fan et al. 2015). High concentrations of
13
1070 Applied Microbiology and Biotechnology (2023) 107:1063–1075
metabolic byproducts are detrimental to cell growth and The intensified fed‑batch
antibody expression (Torres et al. 2018; Zhang et al. 2019;
Konakovsky et al. 2016; Karengera et al. 2018). Glucose is In the biomanufacturing industry, intensified processes
transferred to the cytoplasm by transport protein GLUT1 require more time to develop compared with conventional
in CHO cells, and a large amount of pyruvate is generated fed-batch.
through glycolysis. Part of pyruvate using lactate dehydro- Schulze et al. performed standard fed-batch (sFB) and
genase is further converted to lactate (Li et al. 2012). To intensified fed-batch (iFB) processes in 15 mL and 250-
culture CHO cells expressing Fc-fusion protein and reduce mL bioreactors, starting with 4% (vol / vol) feed medium
lactate accumulation in fed-batch culture, galactose is used A and 4% (vol / vol) feed medium B. The final glucose
as a carbon source to reduce accumulation of lactate (Xiao concentration was 5 g/L. Cell concentrations inoculated by
et al. 2019). CHO-K1 cells expressing anti-HIV antibody N-1 perfusion reached 1 × 108 cells/mL, which are used for
VRC01 were cultured in fed-batch culture. Feed medium the iFB. In 15 mL bioreactors, two iFBs inoculated at 2.5
was added daily starting from day 3. After 12 h of cell inocu- and 5 × 106 cells/mL were from N-1 perfusion. Compared
lation, 10 mM and 30 mM NH4Cl were added to the CHO with sFB process, similar titers were reached in shorter
cell culture medium. Compared with the control group, VCD process times, but respectively, the space–time yield
and culture time of group treated with 30 mM NH4Cl were (STY) was dramatically increased by 16% and 36% for the
significantly reduced, and byproducts including lactate, glu- 2.5 and 5 × 106 cells/mL methods. In 250-mL bioreactors,
cose, ammonium, and glutamine were accumulated. Particu- iFB inoculated at 5 × 106 cells/mL added 2.5 mM butyric
larly, ammonium had a negative impact on antibody titer and acid (BA) after 48 h. Compared with iFB inoculated at
cell-specific productivity (Chitwood et al. 2021). 5 × 106 cells/mL without BA, KPI was found by raising
The addition of TCA cycle intermediates including alpha- the STY by 50% after BA treatment. The average cell-
ketoglutarate (α-KG), malate, and succinate to CHO cells specific productivity was increased from 25 to 37 pg/cell/
can reduce accumulation of lactate and ammonium in fed- day through BA supplementation (Schulze et al. 2022a, b).
batch culture and improve cell-specific productivity and
antibody titer (Zhang et al. 2020b). Xiao et al. designed
continuous feeding strategy in fed-batch culture. They found
that continuous feeding reduced accumulation of lactate and Discussion
ammonium because of massive nutritional feed medium in a
7-L bioreactor. Finally, the production of C12 antibody was RTPs are fast developing. About 40 novel antibodies are
promoted (Xiao et al. 2021). In fed-batch culture, byproducts introduced each year. How to implement high-level expres-
should be avoided. It is advisable for fed-batch culture to sion and production of RTPs is a challenge which limits
choose available feeding strategies to reduce the probability RTP drugs development. These are state-of-the-art strat-
and concentration of metabolic byproducts accumulation. egies for boosting RTP production and quality, such as
optimizing gene sequence, expression vector, and glyco-
sylation (Zhang et al. 2022).
Chemostat cultivation A report has shown that the biosimilar development
of pembrolizumab can be directed by Quality by design
For improving cell-specific productivity of RTPs, success- (QbD) (Jaffar-Aghaei et al. 2022). QbD is an efficient but
ful strategies require to manipulate culture temperature and challenging approach for the development of biosimilar
glucose concentration in media. Unfortunately, changes in due to the complex relationship among process, quality,
cell culture affect the specific growth rate, especially in both and efficacy (Zhang et al. 2020c). To ensure critical qual-
operational variables. To produce recombinant human tissue ity attributes (CQAs) about the production of mAbs, an
plasminogen activator (rh-tPA) in CHO cell, Vergara et al. uti- understanding of the production process is highlighted by
lized chemostat cultivation and set three groups about feeding QbD (Gibbons et al. 2022). Jaffar-Aghaei et al. found a
media with 20 mM, 30 mM, and 40 mM glucose concentra- pharmaceutical measurement guided by QbD to under-
tions. A total of 40 mM glucose limited cell growth, but it take the Pembrolizumab biosimilar candidate PSG-024
increased cell-specific productivity of rh-tPA and cells in the (Keytruda®) in fed-batch culture. Currently, Keytruda®
G2/M phase. Furthermore, glucose consumption and lactate has become the ace of therapeutic mAbs (Jaffar-Aghaei
production rates were reduced when temperature condition et al. 2022).
was 33 °C. Vergara et al. indicated that a reduced specific Despite the fact that glucose concentration above 1 g/L
growth rate together with high feed glucose rapidly improves had brought 10 g/L mAb titer, Handlogten et al. still
protein productivity in CHO cell (Vergara et al. 2018). pointed out that bolus feeding was defective (Handlogten
13
Applied Microbiology and Biotechnology (2023) 107:1063–1075 1071
et al. 2018). Recently, the final mAb titer of 4 g/L was manuscript; TYW edited the review content. TYW and YL funded
reached by the glucose concentration of 12 g/L in fed- this manuscript.
batch platform process (Schellenberg et al. 2022); mean- Funding This work was funded by National Natural Science Founda-
while, the final glucose concentration was 5 g/L. This glu- tion of China (No.32101232), and Basic Research Foundation of Key
cose feeding strategy also promoted cell growth efficiently. Scientific Research of Universities in Henan Province (No. 20zx013).
Cysteine greater than 2.5 mM with low cell densities
Data Availability All data included in this study are available upon
resulted in counteracted oxidative stress, but Komuczki request by contact with the corresponding author.
et al. presented that high cysteine concentration with a
higher cell concentration can counteract oxidative stress Declarations
(Komuczki et al. 2022). Wang et al. reported that an upturn
in terms of the product titer was influenced by tempera- Human and animal rights and informed consent This paper does not
contain any studies with human participants or animals performed by
ture shifted to 33℃ in 14-day fed-batch culture, and 100% any of the authors.
normalized titer was set as the highest titer on day 14.
Fed-batch optimization is available and nevertheless, opti- Conflict of interest The authors declare no competing interests.
mizing all process parameters is impossible on account of
limited resources and timeline (Wang et al. 2022).
Biosimilars will enter the markets in the context of
COVID-19 (Schulze et al. 2022a, b; Tichy et al. 2022), References
it depends on a robust and scalable manufacturing with
excellent productivity. PI is a key trend involving process Aki Y, Katsumata Y, Kakihara H, Nonaka K, Fujiwara K (2021)
strategies. Along with the advance of perfusion and other 4-(2,5-Dimethyl-1H-pyrrol-1-yl) -N- (2,5-dioxopyrrolidin-1-yl)
processes, fed-batch culture has been studied increasingly benzamide improves monoclonal antibody production in a Chi-
nese hamster ovary cell culture. PLoS One 16:1–20. https://doi.
(MacDonald et al. 2022; Schulze et al. 2022a, b). Presently, org/10.1371/journal.pone.0250416
there is still some room for improvement in fed-batch culture Alhuthali S, Kotidis P, Kontoravdi C (2021) Osmolality effects on CHO
(Wang et al. 2022). cell growth, cell volume, antibody productivity and glycosyla-
tion. Int J Mol Sci 22:1–19. https://doi.org/10.3390/ijms220732
90
Ali AS, Raju R, Ray S, Kshirsagar R, Gilbert A, Zang L, Karger BL
(2018) Lipidomics of CHO cell bioprocessing: relation to cell
Summary and prospect growth and specific productivity of a monoclonal antibody. Bio-
technol J 13:e1700745. https://doi.org/10.1002/biot.201700745
Overall, fed-batch culture has harvested high-yield and high- Baik JY, Dahodwala H, Oduah E, Talman L, Gemmill TR, Gasimli
quality protein production, prolonging cell culture time. L, Datta P, Yang B, Li G, Zhang F, Li L, Linhardt RJ, Camp-
bell AM, Gorfien SF, Sharfstein ST (2015) Optimization of bio-
Feed medium is replenished continuously or intermittently process conditions improves production of a CHO cell-derived,
in fed-batch culture. In recent years, fed-batch culture has bioengineered heparin. Biotechnol J 10:1067–1081. https://doi.
made considerable progress. Feed components optimization org/10.1002/biot.201400665
is still essential for process development. Simultaneously, Bibila TA, Robinson DK (1995) In pursuit of the optimal fed-batch
process for monoclonal antibody production. Biotechnol Prog
fed-batch culture should consider culture parameters, nutri- 11:1–13. https://doi.org/10.1021/bp00031a001
ent consumption and accumulation of metabolic byproducts, Braasch K, Kryworuchko M, Piret JM (2021) Autophagy-inducing
etc. With in-depth exploration of RTP production in CHO peptide increases CHO cell monoclonal antibody production in
cells, we still take these indicators into account in the future, batch and fed-batch cultures. Biotechnol Bioeng 118:1876–1883.
https://doi.org/10.1002/bit.27703
such as cell growth, the yield and quality of RTPs. However, Brunner M, Doppler P, Klein T, Herwig C, Fricke J (2017) Elevated
fed-batch culture has defects in the aspect of process devel- pCO2 affects the lactate metabolic shift in CHO cell culture pro-
opment, choosing optimal parameters is challenging, includ- cesses. Eng Life Sci 18:204–214. https://doi.org/10.1002/elsc.
ing temperature, pH, dissolved oxygen, basal and feeding 201700131
Butler M (2005) Animal cell cultures: recent achievements and
medium, and additives. Exploring a series of great process perspectives in the production of biopharmaceuticals. Appl
parameters are constrained by limited factors. With the wide Microbiol Biotechnol 68:283–291. https://doi.org/10.1007/
application of multi-omics, new monitoring and analysis s00253-005-1980-8
technology, artificial intelligence together with other meth- Chee Furng Wong D, Tin Kam Wong K, Tang Goh L, Kiat Heng C,
Gek Sim Yap M (2005) Impact of dynamic online fed-batch strat-
ods, intensified fed-batch culture will further improve the egies on metabolism, productivity and N-glycosylation quality in
yield and quality of recombinant protein production. CHO cell cultures. Biotechnol Bioeng 89:164–177. https://doi.
org/10.1002/bit.20317
Chitwood DG, Wang Q, Elliott K, Bullock A, Jordana D, Li Z,
Author contribution WJX and TYW wrote and designed the Wu C, Harcum SW, Saski CA (2021) Characterization of
manuscript; YL and CLM edited the manuscript; JYP revised this metabolic responses, genetic variations, and microsatellite
13
1072 Applied Microbiology and Biotechnology (2023) 107:1063–1075
instability in ammonia-stressed CHO cells grown in fed-batch Gibbons AL, Rafferty C, Robinson K, Abad M, Maslanka F, Le N, Mo
cultures. BMC Biotechnol 21:1–16. https://doi.org/10.1186/ J, Clark K, Madden F, Hayes R, McCarthy B, Rode C, O’Mahony
s12896-020-00667-2 J, Rea R, O’Mahony Y, Hartnett C (2022) Raman based chemo-
Chotigeat W, Watanapokasin Y, Mahler S, Gray PP (1994) Role of metric model development for glycation and glycosylation real
environmental conditions on the expression levels, glycoform time monitoring in a manufacturing scale CHO cell bioreactor
pattern and levels of sialyltransferase for hFSH produced by process. Biotechnol Prog 38:e3223. https://d oi.o rg/1 0.1002/b tpr.
recombinant CHO cells. Cytotechnology 15:217–221. https:// 3223
doi.org/10.1007/BF00762396 Graham RJ, Bhatia H, Yoon S (2019) Consequences of trace metal
Chung S, Tian J, Tan Z, Chen J, Zhang N, Huang Y, Vandermark E, variability and supplementation on Chinese hamster ovary
Lee J, Borys M, Li ZJ (2019) Modulating cell culture oxidative (CHO) cell culture performance: a review of key mechanisms
stress reduces protein glycation and acidic charge variant forma- and considerations. Biotechnol Bioeng 116:3446–3456. https://
tion. Mabs 11:205–216. https://doi.o rg/1 0.1080/1 9420862.2 018. doi.org/10.1002/bit.27140
1537533 Graham RJ, Ketcham S, Mohammad A, Bandaranayake BMB, Cao
Costa AR, Rodrigues ME, Henriques M, Oliveira R, Azeredo J (2014) T, Ghosh B, Weaver J, Yoon S, Faustino PJ, Ashraf M, Cruz
Feed optimization in fed-batch culture. Methods Mol Biol CN, Madhavarao CN (2020) Zinc supplementation improves the
1104:105–116. https://doi.org/10.1007/978-1-62703-733-4_8 harvest purity of β-glucuronidase from CHO cell culture by sup-
Dahodwala H, Lee KH (2019) The fickle CHO: a review of the causes, pressing apoptosis. Appl Microbiol Biotechnol 104:1097–1108.
implications, and potential alleviation of the CHO cell line insta- https://doi.org/10.1007/s00253-019-10296-1
bility problem. Curr Opin Biotechnol 60:128–137. https://doi. Gupta K, Modi D, Jain R, Dandekar P (2021) A Stable CHO K1 Cell
org/10.1016/j.copbio.2019.01.011 line for producing recombinant monoclonal antibody against
Domján J, Pantea E, Gyürkés M, Madarász L, Kozák D, Farkas A, TNF-α. Mol Biotechnol 63:828–839. https://doi.org/10.1007/
Horváth B, Benkő Z, Nagy ZK, Marosi G, Hirsch E (2022) Real- s12033-021-00329-4
time amino acid and glucose monitoring system for the automatic Ha TK, Kim D, Kim CL, Grav LM, Lee GM (2022) Factors affecting
control of nutrient feeding in CHO cell culture using Raman the quality of therapeutic proteins in recombinant Chinese ham-
spectroscopy. Biotechnol J 17(5):e2100395. https://doi.org/10. ster ovary cell culture. Biotechnol Adv 54:1–43. https://doi.org/
1002/biot.202100395 10.1016/j.biotechadv.2021.107831
Eyster WT, Talwar S, Fernandez J, Foster S, Hayes J, Allen R, Rei- Handlogten MW, Lee-O’Brien A, Roy G (2018) Intracellular response
dinger S, Wan B, Ji X, Aon J, Patel P, Ritz DB (2021) Tuning to process optimization and impact on productivity and product
monoclonal antibody galactosylation using Raman spectroscopy- aggregates for a high-titer CHO cell process. Biotechnol Bioeng
controlled lactic acid feeding. Biotechnol Prog 37:e3085. https:// 115:126–138. https://doi.org/10.1002/bit.26460
doi.org/10.1002/btpr.3085 Hecklau C, Pering S, Seibel R, Schnellbaecher A, Wehsling M, Eich-
Fan YZ, Jimenez Del Val I, Müller C, Wagtberg Sen J, Rasmussen SK, horn T, Hagen JV, Zimmer A (2016) S-Sulfocysteine simplifies
Kontoravdi C, Weilguny D, Andersen MR (2015) Amino acid fed-batch processes and increases the CHO specific productivity
and glucose metabolism in fed-batch CHO cell culture affects via anti-oxidant activity. J Biotechnol 218:53–63. https://d oi.o rg/
antibody production and glycosylation. Biotechnol Bioeng 10.1016/j.jbiotec.2015.11.022
112:521–535. https://doi.org/10.1002/bit.25450 Hernández Rodríguez T, Frahm B (2020) Design, optimization, and
Feng W, Shi JS (2020) Culture condition optimization for expression adaptive control of cell culture seed trains. Methods Mol Biol
of anti-human VEGF monoclonal antibody by CHO cell. Indus- 2095:251–267. https://doi.org/10.1007/978-1-0716-0191-4_14
trial Microbiology 50:28–35. https://d oi.o rg/1 0.3 969/j.i ssn.1 001- Hoang D, Galbraith S, Kuang B, Johnson A, Yoon S (2021) Char-
6678.2020.02.005 acterization of Chinese hamster ovary cell culture feed media
Fleischaker RJ, Sinskey AJ (1981) Oxygen demand and supply in cell precipitate. Biotechnol Prog 37:e3188. https://doi.org/10.1002/
culture. Appl Microbiol Biotechnol 12:193–197. https://doi.org/ btpr.3188
10.1007/BF00499486 Hong JK, Nargund S, Lakshmanan M, Kyriakopoulos S, Kim DY, Ang
Fouladiha H, Marashi SA, Torkashvand F, Mahboudi F, Lewis NE, KS, Leong D, Yang Y, Lee DY (2018) Comparative phenotypic
Vaziri B (2020) A metabolic network-based approach for devel- analysis of CHO clones and culture media for lactate shift. J
oping feeding strategies for CHO cells to increase monoclonal Biotechnol 283:97–104. https://doi.org/10.1016/j.jbiotec.2018.
antibody production. Bioprocess Biosyst Eng 43:1381–1389. 07.042
https://doi.org/10.1007/s00449-020-02332-6 Horvat J, Narat M, Spadiut O (2020) The effect of amino acid sup-
Gangwar N, Mishra R, Budholiya N, Rathore AS (2021) Effect of vita- plementation in an industrial Chinese Hamster Ovary process.
mins and metal ions on productivity and charge heterogeneity of Biotechnol Prog 36:e3001. https://doi.org/10.1002/btpr.3001
IgG1 expressed in CHO cells. Biotechnol J 16:e2000464. https:// Hou Y, Su H, Luo Z, Li M, Ma X, Ma N (2019) Nutrient optimization
doi.org/10.1002/biot.202000464 reduces phosphorylation and hydroxylation level on an fc-fusion
Gélinas JF, Azizi H, Kiesslich S, Lanthier S, Perdersen J, Chahal PS, protein in a CHO fed-batch process. Biotechnol J 14:e1700706.
Ansorge S, Kobinger G, Gilbert R, Kamen AA (2019) Produc- https://doi.org/10.1002/biot.201700706
tion of rVSV-ZEBOV in serum-free suspension culture of HEK Hu DD, Zhao L, Wang JQ, Fan L, Liu XP, Wang HB, Tan WS (2018)
293SF cells. Vaccine 37:6624–6632. https://doi.org/10.1016/j. Physiological responses of Chinese hamster ovary cells to a pro-
vaccine.2019.09.044 ductivity-enhancing yeast extract. J Biosci Bioeng 126:636–643.
Ghaffari N, Jardon MA, Krahn N, Butler M, Kennard M, Turner RFB, https://doi.org/10.1016/j.jbiosc.2018.05.005
Gopaluni B, Piret JM (2020) Effects of cysteine, asparagine, or Jaffar-Aghaei M, Khanipour F, Maghsoudi A, Sarvestani R, Mohamma-
glutamine limitations in Chinese hamster ovary cell batch and dian M, Maleki M, Havasi F, Rahmani H, Karagah AH, Kazemali
fed-batch cultures. Biotechnol Prog 36:e2946. https://d oi.org/10. MR (2022) QbD-guided pharmaceutical development of Pem-
1002/btpr.2946 brolizumab biosimilar candidate PSG-024 propelled to industry
Ghorbaniaghdam A, Chen J, Henry O, Jolicoeur M (2014) Analyz- meeting primary requirements of comparability to Keytruda®.
ing clonal variation of monoclonal antibody-producing CHO Eur J Pharm Sci 173:106171. https://d oi.o rg/1 0.1 016/j.e jps.2 022.
cell lines using an in silico metabolomic platform. PLoS ONE 106171
9:e90832. https://doi.org/10.1371/journal.pone.0090832
13
Applied Microbiology and Biotechnology (2023) 107:1063–1075 1073
Jänne J, Alhonen L, Pietilä M, Keinänen TA (2004) Genetic approaches gene expression and N-linked glycan antennary profile. Biotech-
to the cellular functions of polyamines in mammals. Eur J Bio- nol Bioeng 114:1721–1732. https://doi.org/10.1002/bit.26284
chem 271:877–894. https://doi.org/10.1111/j.1432-1033.2004. Li J, Wong CL, Vijayasankaran N, Hudson T, Amanullah A (2012)
04009.x Feeding lactate for CHO cell culture processes: impact on cul-
Jardon MA, Sattha B, Braasch K, Leung AO, Côté HC, Butler M, ture metabolism and performance. Biotechnol Bioeng 109:1173–
Gorski SM, Piret JM (2012) Inhibition of glutamine-dependent 1186. https://doi.org/10.1002/bit.24389
autophagy increases t-PA production in CHO cell fed-batch Li WF, Fan ZL, Lin Y, Wang TY (2021) Serum-free medium for
processes. Biotechnol Bioeng 109:1228–1238. https://doi.org/ recombinant protein expression in Chinese hamster ovary cells.
10.1002/bit.24393 Front Bioeng Biotechnol 9:1–11. https://doi.org/10.3389/fbioe.
Kamachi Y, Omasa T (2018) Development of hyper osmotic resistant 2021.646363
CHO host cells for enhanced antibody production. J Biosci Bio- Lu F, Toh PC, Burnett I, Li F, Hudson T, Amanullah A, Li J (2013)
eng 125:470–478. https://doi.org/10.1016/j.jbiosc.2017.11.002 Automated dynamic fed-batch process and media optimization
Kaplon H, Muralidharan M, Schneider Z, Reichert JM (2020) Antibod- for high productivity cell culture process development. Biotech-
ies to watch in 2020. MAbs 12:1703531. https://d oi.o rg/1 0.1 080/ nol Bioeng 110:191–205. https://doi.org/10.1002/bit.24602
19420862.2019.1703531 Lu RM, Hwang YC, Liu IJ, Lee CC, Tsai HZ, Li HJ, Wu HC
Karengera E, Robotham A, Kelly J, Durocher Y, De Crescenzo G, (2020) Development of therapeutic antibodies for the treat-
Henry O (2018) Concomitant reduction of lactate and ammonia ment of diseases. J Biomed Sci 27:1. https://doi.org/10.1186/
accumulation in fed-batch cultures: impact on glycoprotein pro- s12929-019-0592-z
duction and quality. Biotechnol Prog 34:494–504. https://doi. MacDonald MA, Nöbel M, Roche Recinos D, Martínez VS, Schulz BL,
org/10.1002/btpr.2607 Howard CB, Baker K, Shave E, Lee YY, Marcellin E, Mahler
Kelley B (2009) Industrialization of mAb production technology: the S, Nielsen LK, Munro T (2022) Perfusion culture of Chinese
bioprocessing industry at a crossroads. Mabs 1:443–452. https:// hamster ovary cells for bioprocessing applications. Crit Rev
doi.org/10.4161/mabs.1.5.9448 Biotechnol 42:1099–1115. https://doi.org/10.1080/07388551.
Kirsch BJ, Bennun SV, Mendez A, Johnson AS, Wang H, Qiu H, Li N, 2021.1998821
Lawrence SM, Bak H, Betenbaugh MJ (2022) Metabolic analy- Mahé A, Martiné A, Fagète S, Girod PA (2022) Exploring the limits
sis of the asparagine and glutamine dynamics in an industrial of conventional small-scale CHO fed-batch for accelerated on
Chinese hamster ovary fed-batch process. Biotechnol Bioeng demand monoclonal antibody production. Bioprocess Biosyst
119:807–819. https://doi.org/10.1002/bit.27993 Eng 45:297–307. https://doi.org/10.1007/s00449-021-02657-w
Komuczki D, Stadermann A, Bentele M, Unsoeld A, Grillari J, Muel- McHugh KP, Xu J, Aron KL, Borys MC, Li ZJ (2020) Effective tem-
ler MM, Paul A, Fischer S (2022) High cysteine concentrations perature shift strategy development and scale confirmation for
in cell culture media lead to oxidative stress and reduced bio- simultaneous optimization of protein productivity and quality in
process performance of recombinant CHO cells. Biotechnol J Chinese hamster ovary cells. Biotechnol Prog 36:e2959. https://
17:e2200029. https://doi.org/10.1002/biot.202200029 doi.org/10.1002/btpr.2959
Konakovsky V, Clemens C, Müller MM, Bechmann J, Berger M, Mellahi K, Brochu D, Gilbert M, Perrier M, Ansorge S, Durocher Y,
Schlatter S, Herwig C (2016) Metabolic control in mammalian Henry O (2019) Process intensification for the production of
fed-batch cell cultures for reduced lactic acid accumulation and rituximab by an inducible CHO cell line. Bioprocess Biosyst
improved process robustness. Bioengineering (basel) 3:5. https:// Eng 42:711–725. https://doi.org/10.1007/s00449-019-02075-z
doi.org/10.3390/bioengineering3010005 Michl J, Park KC, Swietach P (2019) Evidence-based guidelines for
Kong JT, Zhuang YP, Guo MJ (2020) Enhancement of Anti-CD20 controlling pH in mammalian live-cell culture systems. Commun
monoclonal antibody expression by CHO based on DOE and Biol 2:144. https://doi.org/10.1038/s42003-019-0393-7
amino acid supplemental strategy. China Biotechnology 40:41– Moore A, Mercer J, Dutina G, Donahue CJ, Bauer KD, Mather JP,
48. https://doi.org/10.13523/j.cb.2009009 Etcheverry T, Ryll T (1997) Effects of temperature shift on cell
Kou TC, Fan L, Zhou Y, Ye ZY, Liu XP, Zhao L, Tan WS (2011) cycle, apoptosis and nucleotide pools in CHO cell batch cul-
Detailed understanding of enhanced specific productivity in Chi- tues. Cytotechnology 23:47–54. https://d oi.o rg/1 0.1 023/A:1 0079
nese hamster ovary cells at low culture temperature. J Biosci Bio- 19921991
eng 111:365–369. https://doi.org/10.1016/j.jbiosc.2010.11.016 Pan X, Streefland M, Dalm C, Wijffels RH, Martens DE (2017) Selec-
Kozma B, Salgó A, Gergely S (2018) Comparison of multivariate data tion of chemically defined media for CHO cell fed-batch culture
analysis techniques to improve glucose concentration prediction processes. Cytotechnology 69:39–56. https://doi.org/10.1007/
in mammalian cell cultivations by Raman spectroscopy. J Pharm s10616-016-0036-5
Biomed Anal 158:269–279. https://doi.org/10.1016/j.jpba.2018. Qin J, Wu X, Xia Z, Huang Z, Zhang Y, Wang Y, Fu Q, Zheng C
06.005 (2019) The effect of hyperosmolality application time on pro-
Kuwae S, Miyakawa I, Doi T (2018) Development of a chemically duction, quality, and biopotency of monoclonal antibodies
defined platform fed-batch culture media for monoclonal produced in CHO cell fed-batch and perfusion cultures. Appl
antibody-producing CHO cell lines with optimized choline Microbiol Biotechnol 103:1217–1229. https://doi.org/10.1007/
content. Cytotechnology 70:939–948. https://doi.org/10.1007/ s00253-018-9555-7
s10616-017-0185-1 Restelli V, Wang MD, Huzel N, Ethier M, Perreault H, Butler M (2006)
Lalonde ME, Durocher Y (2017) Therapeutic glycoprotein production The effect of dissolved oxygen on the production and the glyco-
in mammalian cells. J Biotechnol 125:128–140. https://doi.org/ sylation profile of recombinant human erythropoietin produced
10.1016/j.jbiotec.2017.04.028 from CHO cells. Biotechnol Bioeng 94:481–494. https://d oi.o rg/
Lee JH, Reier J, Heffner KM, Barton C, Spencer D, Schmelzer AE, 10.1002/bit.20875
Venkat R (2017a) Production and characterization of active Rish AJ, Drennen JK, Anderson CA (2022) Metabolic trends of Chi-
recombinant human factor II with consistent sialylation. Biotech- nese hamster ovary cells in biopharmaceutical production under
nol Bioeng 114:1991–2000. https://doi.org/10.1002/bit.26317 batch and fed-batch conditions. Biotechnol Prog 38:e3320.
Lee JH, Jeong YR, Kim YG, Lee GM (2017b) Understanding of https://doi.org/10.1002/btpr.3220
decreased sialylation of Fc-fusion protein in hyperosmotic Ritacco FV, Wu Y, Khetan A (2018) Cell culture media for recombi-
recombinant Chinese hamster ovary cell culture: N-glycosylation nant protein expression in Chinese hamster ovary (CHO) cells:
13
1074 Applied Microbiology and Biotechnology (2023) 107:1063–1075
history, key components, and optimization strategies. Biotechnol Torres M, Akhtar S, McKenzie EA, Dickson AJ (2021) Temperature
Prog 34:1407–1426. https://doi.org/10.1002/btpr.2706 down-shift modifies expression of UPR-/ERAD-related genes
Romanova N, Schelletter L, Hoffrogge R, Noll T (2022) Hyperos- and enhances production of a chimeric fusion protein in CHO
molality in CHO cell culture: effects on the proteome. Appl cells. Biotechnol J 16:1–11. https://doi.org/10.1002/biot.20200
Microbiol Biotechnol 106:2569–2586. https://doi.org/10.1007/ 0081
s00253-022-11861-x Ventini DC, Damiani R, Sousa AP, de Oliveira JE, Peroni CN, Rib-
Schellenberg J, Nagraik T, Wohlenberg OJ, Ruhl S, Bahnemann J, ela MT, Bartolini P, Tonso A, Soares CR, Pereira CA (2011)
Scheper T, Solle D (2022) Stress-induced increase of monoclonal Improved bioprocess with CHO-hTSH cells on higher microcar-
antibody production in CHO cells. Eng Life Sci 22:427–436. rier concentration provides higher overall biomass and productiv-
https://doi.org/10.1002/elsc.202100062 ity for rhTSH. Appl Biochem Biotechnol 164:401–409. https://
Schmid A, Kreidl E, Bertschinger M, Vetsch P (2022) Benchtop bio- doi.org/10.1007/s12010-010-9143-5
reactors in mammalian cell culture: overview and guidelines. Vergara M, Torres M, Müller A, Avello V, Acevedo C, Berrios J, Reyes
Methods Mol Biol 2436:1–15. https://doi.org/10.1007/7651_ JG, Valdez-Cruz NA, Altamirano C (2018) High glucose and low
2021_441 specific cell growth but not mild hypothermia improve specific
Schmidt C, Wehsling M, Le Mignon M, Wille G, Rey Y, Schnellbae- r-protein productivity in chemostat culture of CHO cells. PLoS
cher A, Zabezhinsky D, Fischer M, Zimmer A (2021) Lactoyl One 13:e0202098. https://d oi.o rg/1 0.1 371/j ourna l.p one.0 20209 8
leucine and isoleucine are bioavailable alternatives for canonical Vijayasankaran N, Varma S, Yang Y, Mun M, Arevalo S, Gawlitzek M,
amino acids in cell culture media. Biotechnol Bioeng 118:3395– Swartz T, Lim A, Li F, Zhang B, Meier S, Kiss R (2013) Effect of
3408. https://doi.org/10.1002/bit.27755 cell culture medium components on color of formulated mono-
Schneider A, Gorr IH, Larraillet V, Frensing T, Popp O (2019) Reduc- clonal antibody drug substance. Biotechnol Prog 29:1270–1277.
tion of IL-2 fragmentation during manufacturing of a novel https://doi.org/10.1002/btpr.1772
immunocytokine by DoE process optimization. Biotechnol Bio- Wang Z, Wang C, Chen G (2022) Kinetic modeling: a tool for tem-
eng 116:2503–2513. https://doi.org/10.1002/bit.27090 perature shift and feeding optimization in cell culture process
Schulze M, Niemann J, Wijffels RH, Matuszczyk J, Martens DE development. Protein Expr Purif 198:106130. https://doi.org/10.
(2022a) Rapid intensification of an established CHO cell fed- 1016/j.pep.2022.106130
batch process. Biotechnol Prog 38:e3213. https://doi.org/10. Weng Z, Jin J, Shao C, Li H (2020) Reduction of charge variants by
1002/btpr.3213 CHO cell culture process optimization. Cytotechnology 72:259–
Schulze M, Kumar Y, Rattay M, Niemann J, Wijffels RH, Martens 269. https://doi.org/10.1007/s10616-020-00375-x
DE (2022b) Transcriptomic analysis reveals mode of action of Wilkens CA, Altamirano C, Gerdtzen ZP (2011) Comparative meta-
butyric acid supplementation in an intensified CHO cell fed- bolic analysis of lactate for cho cells in glucose and galactose.
batch process. Biotechnol Bioeng 119:2359–2373. https://doi. Biotechnol Bioprocess Eng 16:714–724. https://d oi.o rg/1 0.1 007/
org/10.1002/bit.28150 s12257-010-0409-0
Sellick CA, Croxford AS, Maqsood AR, Stephens GM, Westerhoff HV, Wlaschin KF, Hu WS (2007) Engineering cell metabolism for high-
Goodacre R, Dickson AJ (2015) Metabolite profiling of CHO density cell culture via manipulation of sugar transport. J Bio-
cells: Molecular reflections of bioprocessing effectiveness. Bio- technol 131:168–176. https://doi.org/10.1016/j.jbiotec.2007.06.
technol J 10:1434–1445. https://d oi.o rg/1 0.1 002/b iot.2 01400 664 006
Spens E, Häggström L (2007) Defined protein and animal component- Xiao Z, Fan L, Tan WS (2019) Galactose feeding in CHO cell culture
free NS0 fed-batch culture. Biotechnol Bioeng 98:1183–1194. process: the impacts on the cell growth, metabolism and glyco-
https://doi.org/10.1002/bit.21509 sylation of fc fusion protein. Biotechnol Bulletin 35:138–145.
Stach CS, McCann MG, O’Brien CM, Le TS, Somia N, Chen X, Lee https://doi.org/10.13560/j.cnki.biotech.bull.1985.2019-0138
K, Fu HY, Daoutidis P, Zhao L, Hu WS, Smanski M (2019) Xiao S, Ahmed W, Mohsin A, Guo M (2021) Continuous feeding
Model-driven engineering of N-linked glycosylation in chinese reduces the generation of metabolic byproducts and increases
hamster ovary cells. ACS Synth Biol 8:2524–2535. https://doi. antibodies expression in Chinese hamster ovary-K1 cells. Life
org/10.1021/acssynbio.9b00215 (basel) 11:1–12. https://doi.org/10.3390/life11090945
Stiefel F, Fischer S, Sczyrba A, Otte K, Hesse F (2016) miRNA profil- Xie L, Wang DI (1994a) Fed-batch cultivation of animal cells using dif-
ing of high, low and non-producing CHO cells during biphasic ferent medium design concepts and feeding strategies. Biotechnol
fed-batch cultivation reveals process relevant targets for host cell Bioeng 43:1175–1189. https://doi.org/10.1002/bit.260431123
engineering. J Biotechnol 225:31–43. https://doi.org/10.1016/j. Xie L, Wang DI (1994b) Applications of improved stoichiometric
jbiotec.2016.03.028 model in medium design and fed-batch cultivation of animal
Sun JJ, Wang G, Zhao QH, Li GL (2021) Research progress on key cells in bioreactor. Cytotechnology 15:17–29. https://doi.org/
technologies of preparing recombinant antibodies from mam- 10.1007/BF00762376
malian CHO cells. Chin Med Biotechnol 16:348–353. https:// Xing Z, Kenty B, Koyrakh I, Borys M, Pan SH, Li ZJ (2011) Opti-
doi.org/10.3969/j.issn.1673-713X.2021.04.010 mizing amino acid composition of CHO cell culture media for
Takagi Y, Kikuchi T, Wada R, Omasa T (2017) The enhancement of a fusion protein production. Process Biochem 46:1423–1429.
antibody concentration and achievement of high cell density https://doi.org/10.1016/j.procbio.2011.03.014
CHO cell cultivation by adding nucleoside. Cytotechnology Xu S, Hoshan L, Chen H (2016) Improving lactate metabolism in an
69:511–521. https://doi.org/10.1007/s10616-017-0066-7 intensified CHO culture process: productivity and product quality
Tichy EM, Hoffman JM, Suda KJ, Rim MH, Tadrous M, Cuellar considerations. Bioprocess Biosyst Eng 39:1689–1702. https://
S, Clark JS, Ward J, Schumock GT (2022) National trends in doi.org/10.1007/s00449-016-1644-3
prescription drug expenditures and projections for 2022. Am J Yang M, Butler M (2000) Effects of ammonia on CHO cell growth,
Health Syst Pharm 79:1158–1172. https://doi.org/10.1093/ajhp/ erythropoietin production, and glycosylation. Biotechnol Bio-
zxac102 eng 68:370–380. https://d oi.o rg/1 0.1 002/( SICI)1 097-0 290(20000
Torres M, Altamirano C, Dickson AJ (2018) Process and metabolic 520)68:4%3c370:AIDBIT2%3e3.0.CO;2-K
engineering perspectives of lactate production in mammalian cell Yao T, Asayama Y (2017) Animal-cell culture media: history, charac-
cultures. Curr Opin Chem Eng 22:184–190. https://doi.org/10. teristics, and current issues. Reprod Med Biol 16:99–117. https://
1016/j.coche.2018.10.004 doi.org/10.1002/rmb2.12024
13
Applied Microbiology and Biotechnology (2023) 107:1063–1075 1075
Yuan Y, Bian YL, Zhang BH, Zhu JW (2020) Establishment of FUT8 Zhang E, Xie L, Qin P, Lu L, Xu Y, Gao W, Wang L, Xie MH, Jiang W,
knockout CHO-S cell line with stable expression of Anti-HER2 Liu S (2020c) Quality by design-based assessment for analytical
monoclonal antibody. Chin J Pharm 51:69–74. https://doi.org/ similarity of adalimumab biosimilar HLX03 to humira®. AAPS
10.16522/j.cnki.cjph.2020.01.008 J 22:69. https://doi.org/10.1208/s12248-020-00454-z
Zagari F, Jordan M, Stettler M, Broly H, Wurm FM (2013) Lactate Zhang JH, Shan LL, Liang F, Du CY, Li JJ (2022) Strategies and con-
metabolism shift in CHO cell culture: the role of mitochondrial siderations for improving recombinant antibody production and
oxidative activity. N Biotechnol 30:238–245. https://doi.org/10. quality in chinese hamster ovary cells. Front Bioeng Biotechnol
1016/j.nbt.2012.05.021 10:856049. https://doi.org/10.3389/fbioe.2022.85604
Zhang J, Robinson D, Salmon P (2006) A novel function for selenium Zhu MM, Goyal A, Rank DL, Gupta SK, Vanden Boom T, Lee SS
in biological system: selenite as a highly effective iron carrier (2005) Effects of elevated pCO2 and osmolality on growth of
for Chinese hamster ovary cell growth and monoclonal antibody CHO cells and production of antibody-fusion protein B1: a case
production. Biotechnol Bioeng 95:1188–1197. https://doi.org/ study. Biotechnol Prog 21:70–77. https://doi.org/10.1021/bp049
10.1002/bit.21081 815s
Zhang H, Wang H, Liu M, Zhang T, Zhang J, Wang X, Xiang W Zou M, Zhou ZW, Fan L, Zhang WJ, Zhao L, Liu XP, Wang HB, Tan
(2013) Rational development of a serum-free medium and fed- WS (2020) A novel method based on nonparametric regression
batch process for a GS-CHO cell line expressing recombinant with a Gaussian kernel algorithm identifies the critical compo-
antibody. Cytotechnology 65:363–378. https://doi.org/10.1007/ nents in CHO media and feed optimization. J Ind Microbiol Bio-
s10616-012-9488-4 technol 47:63–72. https://doi.org/10.1007/s10295-019-02248-5
Zhang L, Castan A, Stevenson J, Chatzissavidou N, Vilaplana F, Chot-
teau V (2019) Combined effects of glycosylation precursors and Publisher's note Springer Nature remains neutral with regard to
lactate on the glycoprofile of IgG produced by CHO cells. J Bio- jurisdictional claims in published maps and institutional affiliations.
technol 289:71–79. https://d oi.o rg/1 0.1 016/j.j biote c.2 018.1 1.0 04
Zhang W, Liu X, Tang H, Zhang X, Zhou Y, Fan L, Wang H, Tan WS, Springer Nature or its licensor (e.g. a society or other partner) holds
Zhao L (2020a) Investigation into the impact of tyrosine on the exclusive rights to this article under a publishing agreement with the
product formation and quality attributes of mAbs in rCHO cell author(s) or other rightsholder(s); author self-archiving of the accepted
cultures. Appl Microbiol Biotechnol 104:6953–6966. https://d oi. manuscript version of this article is solely governed by the terms of
org/10.1007/s00253-020-10744-3 such publishing agreement and applicable law.
Zhang X, Jiang R, Lin H, Xu S (2020b) Feeding tricarboxylic acid
cycle intermediates improves lactate consumption and antibody
production in Chinese hamster ovary cell cultures. Biotechnol
Prog 36:e2975. https://doi.org/10.1002/btpr.2975
13