You are on page 1of 11

A Single Nutrient Feed Supports Both Chemically Defined NS0 and CHO

Fed-Batch Processes: Improved Productivity and Lactate Metabolism


Ningning Ma, JoAnn Ellet, Centy Okediadi, Paul Hermes, Ellen McCormick, and Susan Casnocha
Bioprocess R&D, Global Biologics, Pfizer Inc, Chesterfield, MO 63017

DOI 10.1002/btpr.238
Published online July 27, 2009 in Wiley InterScience (www.interscience.wiley.com).

A chemically defined nutrient feed (CDF) coupled with basal medium preloading was
developed to replace a hydrolysate-containing feed (HCF) for a fed-batch NS0 process. The
CDF not only enabled a completely chemically defined process but also increased recombi-
nant monoclonal antibody titer by 115%. Subsequent tests of CDF in a CHO process indi-
cated that it could also replace the hydrolysate-containing nutrient feed in this expression
system as well as providing an 80% increase in product titer. In both CDF NS0 and CHO
processes, the peak lactate concentrations were lower and, more interestingly, lactate metab-
olism shifted markedly from net production to net consumption when cells transitioned from
exponential to stationary growth phase. Subsequent investigations of the lactate metabolic
shift in the CHO CDF process were carried out to identify the cause(s) of the metabolic
shift. These investigations revealed several metabolic features of the CHO cell line that we
studied. First, glucose consumption and lactate consumption are strictly complementary to
each other. The combined cell specific glucose and lactate consumption rate was a constant
across exponential and stationary growth phases. Second, Lactate dehydrogenase (LDH) ac-
tivity fluctuated during the fed-batch process. LDH activity was at the lowest when lactate
concentration started to decrease. Third, a steep cross plasma membrane glucose gradient
exists. Intracellular glucose concentration was more than two orders of magnitude lower
than that in the medium. Fourth, a large quantity of citrate was diverted out of mitochondria
to the medium, suggesting a partially truncated tricarboxylic acid (TCA) cycle in CHO cells.
Finally, other intermediates in or linked to the glycolytic pathway and the TCA cycle, which
include alanine, citrate, isocitrate, and succinate, demonstrated a metabolic shift similar to
that of lactate. Interestingly, all these metabolites are either in or linked to the pathway
downstream of pyruvate, but upstream of fumarate in glucose metabolism. Although the spe-
cific mechanisms for the metabolic shift of lactate and other metabolites remain to be eluci-
dated, the increased understanding of the metabolism of CHO cultures could lead to future
improvements in medium and process development. V C 2009 American Institute of Chemical

Engineers Biotechnol. Prog., 25: 1353–1363, 2009


Keywords: CHO, NS0, chemically defined process, lactate, metabolic shift, metabonomics,
metabolomics

Introduction the cells or are required at much lower levels than supplied.
Third, the chemically undefined nature of hydrolysates hin-
Hydrolysates, especially those of nonanimal origin, are ders the scientists’ understanding process of cell metabolism.
widely used as serum replacements in therapeutic protein For instance, it is not well understood how mammalian cells
production processes using mammalian cells. Hydrolysates metabolize amino acids in peptides, so that amino acid con-
can improve cell growth and protein yield in serum-free sumption analysis based on free amino acids measurements
processes (Burteau et al., 2003; Heidemann et al., 2000; may or may not be accurate (Nyberg et al., 1999).
Sung et al., 2004; Xie et al., 1997). However, there are a
few limitations that complicate hydrolysates’ utilization. First Development of various chemically defined media for
of all, lot-to-lot variation exists for hydrolysates (Luo and NS0 cells and CHO cells has been reported (Chen et al.,
Chen, 2007; Zhang et al., 2003). Prescreening using small- 2000; Hata et al., 1992; Huang et al., 2007; Zhang and Rob-
scale performance testing is typically required to eliminate inson, 2005), and commercial chemically defined media are
underperforming lots. Second, the nutrient composition in readily available from all major mammalian cell culture me-
hydrolysates is not balanced for cell consumption. Various dium manufacturers. However, the development of chemi-
components, such as ash and salts, are either not required by cally defined fed-batch processes that support rapid cell
proliferation, high cell density, and high production rate has
been challenging. Chemically defined fed-batch processes
Correspondence concerning this article should be addressed to N. Ma have been reported recently. Gong et al. (2006) developed a
at ningning.ma@pfizer.com. chemically defined fed-batch process for hybridoma cells.

C 2009 American Institute of Chemical Engineers


V 1353
1354 Biotechnol. Prog., 2009, Vol. 25, No. 5

Peak viable cell density reached about 8  106 cells/mL, but tate metabolic shift from net production to net consumption.
viable cells dropped sharply after a 5-day culture. Burky At the end of the process, lactate concentration in the CDF
et al. (2006) developed a generic chemically defined fed- processes was almost undetectable. Subsequent investigations
batch process for NS0 cells. The process supported peak via- were carried out to better understand this metabolic shift in
ble cell density around 1  107 cells/mL and product titer of the CDF CHO process.
2.64 g/L on Day 13.
Lactic acid, or lactate, has long been recognized as a Materials and Methods
major by-product in cell culture processes (Hu et al., 1987;
Miller et al., 1989; Ozturk and Palsson, 1992). Base addition Cells
to neutralize lactic acid elevates medium osmolarity, which, NS0 and CHO cells producing test recombinant monoclo-
in turn, inhibits cell growth and causes viability to drop. nal antibodies were used in this study. Both cell types used
Reducing lactic acid production had been shown to increase glutamine synthatase as the transfection selection marker.
peak cell density, extend process duration, and increase
recombinant protein yield. Four strategies had been reported
in the literature to reduce lactate production. The first one is Process
to reduce medium glucose concentration to limit glucose NS0 and CHO cells were routinely subcultured in 250 mL
supply (Cruz et al., 1999; Zhou et al., 1995, 1997b). Using shake flasks (Corning, Acton, MA) in chemically defined hy-
an online dynamic feeding protocol, Zhou et al. (1995, bridoma and CHO media, respectively. Both hybridoma and
1997b) successfully maintained glucose at a low level of CHO media are commercially available. The CHO medium was
about 0.5 mM. As a result, lactate production was reduced supplemented with 25 mM methionine sulfoximine (MSX).
and higher peak cell densities and higher viability were Two-liter Applikon bioreactors (Applikon, Foster city,
achieved. A second strategy is to use alternative, slow-con- CA) with a 1-L working volume were used in this study. In
suming carbon sources (Altamirano et al., 2004). Altamirano these bioreactors, two pitched blade impellers provided mixing
et al. (2004) used a feeding strategy in which glucose and and gas bubble dispersion. Temperature was controlled at
galactose feeds were alternated. This strategy forced CHO 36.5 C using a heating blanket. pH was controlled at 7.0 by
cells to consume more residual lactate than the culture fed carbon dioxide sparging through a ring sparger and by adding
with glucose only. As a result, harvest viability was mod- either 0.5 N sodium hydroxide or 7.5% sodium bicarbonate.
estly improved. The third strategy is to downregulate lactate Dissolved oxygen (DO) was controlled at 30% air saturation
dehydrogenase-A (LDH-A) (the LDH subunit that effectively using air overlay and oxygen sparging. The fed-batch process
catalyzes pyruvate to lactate conversion) using a wide vari- for NS0 and CHO cells was very similar. Cells are seeded at
ety of techniques including homologous recombination 2.5  105 viable cells/mL in corresponding NS0 or CHO pro-
(Chen et al., 2001), antisense mRNA (Jeong et al., 2001; duction media. Starting from Day 3, a nutrient feed was added
Paredes et al., 1999), and small interfering RNA (Kim and into the bioreactors at 28 mL/L/day and 16 mL/L/day for NS0
Lee, 2007a). Chen et al. (2001) compared a LDH-A downre- and CHO cells, respectively, till the end of the process. Glucose
gulated hybridoma clone to a control clone. In the culture was fed separately to control glucose concentration around 2 g/
utilizing the LDH-A downregulated cell line, lactate concen- L. Bioreactors were sampled daily for offline measurements.
tration was about 35% lower and monoclonal antibody titer
was about 200% higher. However, although Kim and Lee
(2007a) reduced lactate production rate even further (by 45– Routine bioreactors offline measurements
79%), the recombinant thrombopoietin product rates of three Cell density and viability were measured using a Cedex
LDH-A downregulated clones were not statistically better cell counter (Innovatis, Bielefeld, Germany). Offline DO,
than the control clone. A fourth strategy is to enhance the pH, glucose concentration, and lactate concentration were
flow of glucose carbon into tricarboxylic acid (TCA) cycle. measured with a BioProfile chemical analyzer (Nova Bio-
A focus area has been the overexpression of pyruvate car- medical, Waltham, MA). A portion of the broth from each
boxylase of yeast (Irani et al., 1999, 2002) or human origin daily sampling was filtered through a 0.2 lm polyethersul-
(Kim and Lee, 2007b). In a perfusion culture, Irani et al. fone filter (Pall, Ease Hills, NY) for product titer and amino
(2002) demonstrated that pyruvate carboxylase-transfected acid measurements.
BHK-21 cells produced half of the lactate when compared Monoclonal antibody titer was measured using an Agilent
with the control cells. Cell density was about 50% higher HPLC (Hewlett Packard, Waldbronn, Germany) with a
and recombinant erythropoietin production rate was about POROS protein-A affinity column (Applied Biosystems, Fos-
100% higher. More recently, it was reported that the expres- ter City, CA). The column was calibrated using correspond-
sion of antiapoptosis genes, E1B-19K and Aven (EA167), ing reference standards before titer quantification.
could reduce lactate production of CHO cells (Dorai et al.,
in press). In batch cultures with or without glucose limita-
tion, a CHO clone transfected with antiapoptotic genes gave Intracellular LDH activity measurement
lower lactate concentration compared with the control cell About 2–4 mL of broth was taken out of bioreactors and
line at corresponding conditions. transferred to a centrifuge tube chilled on ice. The cells were
In this study, we developed a chemically defined nutrient washed twice with cold phosphate buffered saline (PBS). In
feed (CDF) with companion production medium preloading each wash, the cells were pelletted in a refrigerated centri-
to replace a hydrolysate-containing feed (HCF) for a fed- fuge at 150g for 5 min, the supernatant was discarded, and
batch NS0 process. The CDF was later investigated in a the cell pellet was gently resuspended in 4 mL of cold PBS.
CHO process. In both NS0 and CHO processes, CDF After the second wash, cells were diluted to 5  105 viable
resulted in higher product titer and lower peak lactate con- cells/mL and lysed using freeze-thaw. In the freeze-thaw
centration. The CDF processes also exhibited a dramatic lac- procedure, cells were first flash frozen in liquid nitrogen and
Biotechnol. Prog., 2009, Vol. 25, No. 5 1355

then thawed overnight at 2–8 C. LDH activity was quantified


using a colorimetric LDH assay kit (CytoTox 96 nonradioac-
tive cytotoxicity assay, Promega, Madison, WI) and a spec-
trophotometer (Molecular Devices, Sunnyvale, CA). Sample
preparation and measurement followed manufacturers’
instructions. The LDH assay kit quantifies LDH activity
based on the conversion rate of lactate and NADþ to pyru-
vate and NADH.

Sample collection for metabonomics assay


Both cell and spent medium samples were collected for
metabonomics analysis. To collect cell samples, cell culture
broth containing 2  107 viable cells was removed from the
bioreactor and immediately chilled in an ice bucket. The
cells were pelleted at 1,000g for 3 min in a refrigerated cen-
trifuge (Bechman Coulter, Fullerton, CA) and then rinsed
twice using cold PBS. After two washes, the cell pellet was
flash frozen in liquid nitrogen and stored at 80 C. A 2 mL
spent medium sample was collected where cell broth was
first centrifuged at 1,000g for 3 min and then the supernatant
was filtered through a 0.2 lm polyethersulfone filter. Spent
medium samples were also stored at 80 C. After all sam-
ples were collected, they were sent on dry ice to the analysis
provider.

Sample preparation and metabonomics assay


Sample preparation and metabonomics analysis were con-
ducted at Metabolon (Durham, NC). The metabonomics
methodology is detailed elsewhere (Lawton et al., 2008) but
briefly, the samples were thawed, small molecules were
extracted, and the reconstituted extracts were split for analy-
sis by GC and LC mass spectrometry. Chromatographic sep-
aration of all ions in each sample was followed by library
matching of these ions to Metabolon’s reference library of
standards. The identity of metabolites was determined by
matching the combination of chromatographic retention
index and mass spectra signatures compared with the refer-
ence library entries.
Figure 1. The top-down approach used in the chemically
defined nutrient feed development.
Results and Discussion
The development included three major steps: (I) obtain an over
Development of a chemically defined nutrient rich nutrient feed, (II) formula simplification, and (III) polish-
ing and formulation.
feed for NS0 process
A CDF was developed to replace a hydrolysate-containing
nutrient feed (HCF) that was previously used for a fed-batch etary chemically defined NS0 medium and a previously
NS0 process. In the hydrolysate-containing fed-batch pro- developed CDF. To ensure that all necessary components
cess, the basal medium is chemically defined, but the nutri- were in ample supply, most components were added at a
ent feed contains a nonanimal-derived hydrolysate. The high concentration. In the next step, nutrients in the over-
limitations of hydrolysates, such as lot-to-lot variation, rich solution were divided into eight rough groups to balance
prompted the development of the CDF to achieve a fully their ratio. Grouping was based on nutritional category (trace
chemically defined process. elements, nucleotides, etc.) or by solubility (low pH, ethanol,
The development took a top-down approach, as illustrated etc.). In the subsequent formula simplification steps, compo-
in Figure 1. There were three major development steps: (1) nents were screened in groups for two cycles, first in rough
develop an over-rich nutrient feed, (2) simplify formula by groups and then in refined groups. In the first cycle, two of
removing unnecessary components and reduce the concentra- eight rough groups were found unnecessary where their
tion of overfeed components, and (3) polishing and formula- exclusion rendered no detectable difference in process per-
tion. The plan for the over-rich solution was that it should formance (cell density, viability, and product titer). Compo-
contain all nutrients that NS0 cells require and all required nents in these two subgroups were subsequently removed
nutrients should be at sufficiently high concentrations. To from the formula. The remaining components were divided
ensure that all necessary components were included in the into 13 refined groups again based on nutritional category
over-rich solution, over 90 components were incorporated. and solubility. Most groups contained less than five compo-
These components came from the formula of a Pfizer propri- nents. In the second cycle of screening, five refined groups
1356 Biotechnol. Prog., 2009, Vol. 25, No. 5

concentration and a 62% increase in cell specific productiv-


ity (Qp). These results demonstrate that CDF not only allows
for removing hydrolysate from the process but also delivers
better cell growth and productivity. Another benefit of the
CDF process is the lower lactate concentration. As shown in
Figure 2b, lactate in the CDF process was lower throughout
the whole process, and the peak concentration was only half
of that in the HCF process. In both processes, cells switched
from net lactate production to net consumption around Day
5. In the CDF process, residual lactate was consumed in 2
days and remained lower than the detection limit until har-
vest. In the HCF process, the lactate concentration was gen-
erally higher and net lactate production resumed, although at
a very slow rate, after Day 8.
The benefit of the top-down approach to nutrient feed de-
velopment is that it eliminates nutrient limitation up front,
thereby allowing the development to focus on nutrient bal-
ancing and simplification. The risk of this strategy is the
inclusion of unnecessary components and overfeeding, which
could be toxic or inhibitory to cell growth or production.
The successful development of a CDF demonstrates that the
risk is manageable. During the development, many compo-
nents, such as vitamins and minerals, were found to be non-
toxic even at a concentration one or two orders of magnitude
higher than what the cells needed. This observation agrees
with Xie and Wang (1994), who found that a high concentra-
tion of vitamins was not harmful to animal cells. Amino
acids have the potential to be inhibitory to cell growth at
high concentrations. Fortunately, amino acid overfeeding can
be avoided using spent medium analysis.

Figure 2. Comparison of the CDF (solid lines) and HCF (dot-


ted lines) NS0 processes. Evaluation of the CDF in CHO processes
(a) Viable cell density and viability. (b) Product titer and lac-
tate concentration. The results shown are the average of three
The CDF developed for NS0 cells was later tested on
replicate batches. Error bar indicates 1 SD. CHO cells and compared with the CHO HCF process. The
medium used in both processes is a commercially available
chemically defined CHO medium. As shown in Figure 3, the
CDF process offered clear advantages over the HCF process.
were found to offer no detectable beneficial (or detrimental) Results shown in Figure 3 were the average of two duplicate
effect on the process performance and were removed. In the bioreactors. Peak viable cell density reached 18  106
final step, the components in the simplified formula, with cells/mL in both processes. However, viability was main-
less than 40 components, were balanced for optimization. tained better in the CDF process. When the processes were
Amino acids were optimized based on spent medium analy- terminated on Day 15, viability for the CDF process was
sis, whereas the other components were optimized relied on around 80%, whereas that for the HCF process had declined
refined group titration study. Finally, all components were to 50%. Product titer was similar in both processes till Day
formulated into a single pH neutral nutrient feed. For com- 10, after which titer started to level off in the HCF process,
ponents with low solubility in the feed, the portion over the whereas that in the CDF process kept rising linearly till har-
solubility limit was preloaded into the basal medium. All de- vest. By Day 15, product titer in the CDF process was 82%
velopment studies were conducted in fed-batch shake flasks. higher than that in the HCF process. Lactate concentration
At each milestone, the performance was confirmed in 2 L was much lower in the CDF process after Day 7. By Day
bench-scale bioreactors. The whole development took 9 10, lactate concentration in the CDF process became close to
months, with each of three major steps for about 3 months. undetectable, whereas that in the HCF process stayed at 1.8–
A comparison of the CDF process and HCF process in 2.6 g/L.
2 L bioreactors is presented in Figure 2, where results are
the average of three bioreactor runs and error bars indicate
1 SD. Cell growth phase was extended in the CDF process Lactate metabolic shift in the CDF processes
where the peak viable cell density reached 15  106 cells/ A major metabolic feature of the CDF processes is the
mL, 50% higher than that in the HCF process. Viability dramatic shift of lactate metabolism from net production to
was modestly improved in the CDF process, the harvest via- net consumption. On one hand, the metabolic shift is benefi-
bility was about 10% higher in the CDF process. Product ti- cial for the process as it significantly reduced the concentra-
ter was much higher in the CDF process after Day 5. At tion of a major cellular metabolic by-product. At the end of
harvest on Day 11, product titer in the CDF process was the process, lactate concentration was almost undetectable.
115% higher than that in the HCF process. This 115% On the other hand, the shift could be due to a glucose carbon
increase was from a 33% increase in the integral viable cell limitation, which may have a detrimental effect on the cell
Biotechnol. Prog., 2009, Vol. 25, No. 5 1357

Figure 4. Cell growth and glucose and lactate metabolism of


Figure 3. Comparison of the CDF (solid lines) and HCF (dot- CHO cells in the CDF process in two duplicate
ted lines) CHO processes. bioreactors.
(a) Viable cell density and viability. (b) Product titer and lac- Bioreactor 1, solid symbols; Bioreactor 2, hollow symbols. (a)
tate concentration. The results shown are the average of two Cell growth and lactate profile. The arrow indicates the correla-
duplicate batches. tion between shift of lactate metabolism and cell growth phase
transition. (b) Weight cumulative glucose consumption, lactate
production, and net glucose consumption. The net glucose con-
sumption is defined as glucose consumption minus lactate pro-
duction. The glucose and lactate metabolism can be divided
into three phases: (I) high glucose consumption and net lactate
growth and productivity. Although glucose supply was not production, (II) low glucose consumption and net lactate
limited in both CDF NS0 and CDF CHO processes (glucose consumption, and (III) high glucose consumption and very low
residual lactate concentration.
concentration [1 g/L), possible limitations could still exist
in the glucose transportation step or the flux through the gly-
colytic pathway. To better understand and to identify the tate production is a hallmark of transformed cells, a phenom-
possible mechanism(s) for the lactate metabolic shift, we enon established more than 70 years ago (Warburg, 1930).
examined carbon metabolisms in the CDF CHO process. The High lactate production has been attributed in the literature
lactate metabolic shift in fed-batch processes has been previ- to the upregulation of various enzymes in the glycolytic
ously reported with different cell expression systems, includ- pathway (Board et al., 1990; Dang and Semenza, 1999;
ing DHFR-CHO (Tsao et al., 2005), GS-NS0 (Zhou et al., Kondoh et al., 2005). As cells exit the rapid proliferating
1997a), and a non-GS-NS0 (Burkey et al., 2006). However, phase, they may concurrently cease high lactate production.
no thorough investigation of this phenomenon has been pub- Alternatively, the lactate metabolism shift could be a result
lished. We focused our investigation on CHO cells, where of glucose carbon limitation because of restricted flux
the cell line used was a subclone of the CHO cell line used through the glycolytic pathway.
in the initial CDF evaluation (Figure 3). The CDF fed-batch Analysis of glucose consumption revealed a strong com-
process was kept the same. plementary relationship between glucose and lactate con-
As indicated in Figure 4a, the lactate metabolic shift took sumption. Cumulative glucose utilization, lactate production,
place in the same period when cells were transitioning from and net glucose consumption (defined as combined glucose
exponential to stationary growth phase. The same timing cor- and lactate consumption) from the two duplicate bioreactors
relation between the lactate metabolic shift and cell growth referred to in Figure 4a are illustrated in Figure 4b. The
phase transition exists in previous publications using differ- slope of the curves in Figure 4b represents either cell-spe-
ent mammalian cell types (Burkey et al., 2006; Tsao et al., cific substrate consumption (glucose or net glucose) or pro-
2005; Zhou et al., 1997a). Whether there is a cause-effect duction (lactate) rate. The glucose consumption and lactate
relationship between lactate metabolic shift and cell growth production rates fluctuated during the process. However, net
slowdown is unknown. One possibility is that the metabolic glucose consumption rate was steady throughout the whole
shift results from the cell growth phase transition. High lac- process. This observation inferred that (1) cell-specific
1358 Biotechnol. Prog., 2009, Vol. 25, No. 5

Figure 5. Major NAD1 and NADH conversion pathways.

pyruvate consumption rate in TCA cycle was a constant


throughout the whole process and (2) glucose and lactate
consumption rate was tightly controlled so that the flux from
glucose to pyruvate and the flux from lactate to pyruvate are
strictly complementary to each other.
NADH homeostasis, as previously being suggested (e.g.,
Zhou et al., 1997a), could be the cause of the tight control
of glucose and lactate consumption. The three major path-
ways related to NADH homeostasis are illustrated in Figure
5. The conversion of pyruvate to lactate recycles NADH pro-
duced in glycolysis back to NADþ. When the conversion is
reversed, it competes with glycolysis for NADþ. The
reduced availability of NADþ could slow down the glycoly-
sis rate. NADH is also recycled back to NADþ inside mito-
chondria through the aspartate–malate shuttle (Figure 5) or
similar shuttle systems. We need to assume that the shuttle Figure 6. Variation of LDH activity in the CHO CDF process.
rate was a constant for NADH homeostasis to exert the com- Results were from two similar bioreactor runs using the same
clone. Bioreactor 1, solid symbols; Bioreactor 2, hollow
plementation effect on glucose and lactate consumption. It symbols.
should be noted that in Stage II and III of Figure 4b, all
NADH needed to be recycled in mitochondria as no NADH
was recycled through pyruvate to lactate conversion.
to lactate production. More studies are needed to fully under-
stand the role of LDH activity variation in lactate metabolic
LDH activity changes during the process shift, such as the ratio between the two major LDH subunits,
Cell-specific LDH activity was monitored in two bioreactor LDH-A and LDH-B, which favor the pyruvate–lactate con-
runs using the same CHO subclone and CDF process. As version in opposite directions, and the ratio between the other
shown in Figure 6, cell-specific LDH activity oscillated in two substrates in the pyruvate–lactate conversion reaction,
both runs. During the exponential growth phase, LDH activ- NADþ and NADH, where a high NADþ/NADH ratio favors
ity first increased and then decreased. It further decreased lactate to pyruvate conversion.
and reached the lowest level during the growth phase transi-
tion from exponential to stationary phase. With the decrease
of cell-specific LDH activity, lactate metabolism shifted from Shift of amino acid metabolism
net production to net consumption. After the lactate meta- In addition to lactate metabolic shift, several amino acids
bolic shift, LDH activity gradually increased and reached its also demonstrated shifts of metabolism during the fed-batch
maximum at the end of the process. Cell size was consistent process, as shown in Figure 7. The results were from a sin-
during the fed-batch process (data not shown), so that the gle bioreactor run. The alanine shift is of particular interest
LDH activity variation was not due to a cell volume change. as similar to lactate, alanine can be derived from pyruvate.
Although there seems to be a linkage between LDH activity Figure 7 showed that the pattern of alanine metabolic shift
and lactate metabolism, the results were hard to interpret. was similar to that of lactate, although the alanine production
First, in the LDH assay that we used, LDH activity is quanti- and consumption rate was much lower than that of lactate.
fied by the rate of lactate to pyruvate conversion, hence it is Asparagine net consumption rate was initially very high, but
counterintuitive to find that the lowest activity measured off- decreased rapidly midway through the process. The decrease
line correlated to the highest lactate to pyruvate conversion was due to asparagine supply limitation. As most asparagine
rate in the process. Second, although decreased LDH activity is likely utilized in the TCA cycle through aspartate and
was correlated with net lactate consumption, subsequent then oxaloacetate, under asparagine limitation, net aspartate
increase in LDH activity did not reverse the metabolism back consumption rate would increased, as observed in Figure 7.
Biotechnol. Prog., 2009, Vol. 25, No. 5 1359

Figure 7. Cumulative consumption of amino acids whose me-


tabolism shifted in the CHO CDF process.
Other amino acids, except for glutamine, showed constant con-
sumption rates. Glutamine consumption was not monitored as
no glutamine was supplied in the process.

It should be noted that asparagine limitation was not the


cause of the lactate metabolic shift. Increasing asparagine
supply did not alter either cell growth or lactate metabolism
(data not shown). For the other two amino acids, glycine
shifted from net production to net consumption and serine
shifted from high to low net consumption during the process.
Glycine can be produced from 3-phosphoglycerate, a glyco-
lytic pathway intermediate, through serine. But whether this
link is related to the metabolic shift of glycine and serine is
unknown.
Figure 8. Concentration of glycolytic pathway metabolites in
Glycolytic pathway and TCA cycle metabolite profiling the CHO CDF process.
(a) Intracellular concentration. (b) Extracellular concentration.
A more broad investigation of metabolites in the glyco- Only metabolites that were detected on consecutive days are
lytic pathway and TCA cycle was conducted as part of a linked with lines. The sporadically detected metabolites are
metabonomics study. In this study, we monitored the change shown as individual data points. Missing data points were
below detection limit.
of intracellular and extracellular metabolite pools in the CDF
CHO fed-batch process in one bioreactor run. One other spe-
cific purpose of the metabonomics study was to verify Figure 8a indicates that the intracellular glucose ion count
whether there were any flux limitation steps in the glycolytic was very low, being detected only sporadically in the first 5
pathway that limit glucose utilization and force cells to use days. In contrast, the extracellular glucose ion count was
lactate. much more abundant (Figure 8b). The difference between
The profiling of glycolytic pathway intermediates and the the intracellular and extracellular glucose concentration is
TCA cycle intermediates are shown in Figures 8 and 9, more than two orders of magnitude, indicating a steep con-
respectively. It should be noted that the results shown in Fig- centration gradient across the plasma membrane. The intra-
ures 8 and 9 are relative ion count results from mass spec- cellular glucose concentration is regulated by two factors,
trometry measurements. As different metabolites have glucose transportation and its subsequent phosphorylation.
different ionization potential, data shown in Figures 8 and 9 Glucose transportation is accomplished through a family of
cannot be used to compare relative concentration among dif- 13 facilitative glucose transporter (GLUT) proteins (Macheda
ferent metabolites. However, for a specific metabolite, ion et al., 2005). GLUT1 (Harrison et al., 1991) and GLUT4
counts are linearly proportional to its concentrations. Hence, (Bogan et al., 2001) are likely expressed on CHO cells. Pho-
the ion count data of one specific metabolite can be used to phorylation of glucose by hexokinase II (HK II) is the first
study its concentration change during the process and to reaction in the glycolysis pathway. In transformed cells, HK
compare its abundance between extracellular and intracellu- II activity was found being upregulated by 100 times
lar samples. Because the intracellular samples were more (Peterson et al., 2002). This upregulation was achieved by
diluted than the medium (extracellular) samples, the intracel- two means: overexpression of HK II enzyme and docking of
lular results were adjusted based on the difference of sample HK II to porins on the mitochondrial membrane. If glucose
dilution factors between intracellular and extracellular sam- transportation was the rate limiting step regulating the intra-
ples, as shown in Eq. 1, so that the intracellular and extracel- cellular glucose concentration, the immediate metabolite of
lular results could be directly comparable. IC indicates ion glucose, glucose-6-phosphate, should be depleted because of
counts in Eq. 1. glucose limitation. However, intracellular glucose-6-phos-
phate ion count was at a relative high level during most of
the process, and its extracellular concentration increased by
Intracelluar sample dilution factor
ICAdjusted ¼ ICOriginal (1) a factor of 10 during the process. These data suggest that the
Extracelluar sample dilution factor glycolytic pathway had an ample supply of glucose-6-
1360 Biotechnol. Prog., 2009, Vol. 25, No. 5

pyruvate are intermediates for biomass formation, slow down


of cell proliferation would reduce their consumption rates.
Intracellular and extracellular TCA cycle intermediates are
shown in Figures 9a,b respectively. One important metabo-
lite, oxaloacetate, was not measured as its volatility makes it
hard to preserve during sample preparation. All other
detected intermediates were maintained at a steady concen-
tration in the cells during the fed-batch process (Figure 9a).
Isocitrate detection was sporadic, because of its concentra-
tion being close to the detection limit. Considerably, more
variation was observed with the extracellular metabolites
(Figure 9b). All metabolite levels changed during the pro-
cess. Citrate, isocitrate, and succinate showed a similar meta-
bolic shift as lactate and alanine. Their concentration
increased till Day 9 or 10 and then decreased. As none of
these intermediates was supplemented during the process,
any extracellular accumulation was due to an efflux of
metabolites from mitochondria to the cytosol and then into
the broth.
Citrate concentration was much higher in the broth. Citrate
must be transported out of the cells against a steep concentra-
tion gradient. Previous studies had revealed that various can-
cer and transformed cells have a truncated TCA cycle where
flux from citrate to a-ketoglutarate was downregulated (Bag-
getto, 1992). One proposed mechanism for the downregula-
tion is a strong efflux of citrate out of mitochondria to the
cytosol. Once in the cytosol, citrate can be used to synthesize
cholesterol and fatty acids, critical building blocks for mem-
brane production in rapidly growing cancer or transformed
cells (Hatzivassiliou et al., 2005). In this study, it was clearly
Figure 9. Concentration of TCA cycle metabolites in the CHO
shown that citrate was not only transported out of mitochon-
CDF process. dria but also out of cells. The high citrate accumulation rate
(a) Intracellular concentration. (b) Extracellular concentration. in the first 9 days suggests that the TCA cycle in CHO cells
Only metabolites that were detected on consecutive days are might be partially truncated in these days. After Day 9, net
linked with lines. The sporadically detected metabolites are citrate accumulation was switched to net consumption.
shown as individual data points. Missing data points were
below detection limit. Because the absolute citrate concentration was not known,
we cannot estimate the percentage of pyruvate carbon that
was diverted out of TCA cycle as citrate. However, consider-
phosphate. Therefore, the low intracellular glucose concen- ing the vast volume difference between broth and cells, the
tration is more likely due to high HK II activity, instead of efflux of citrate out cells must be a considerable drain for the
low glucose transportation rate. TCA cycle. The extracellular isocitrate concentration was
The extracellular lactate concentration profile matches that also higher than that in the cells, indicating a transportation
measured using a conventional chemistry analyzer (BioPro- of isocitrate out of cells as well. It should be noted that the
file of Nova Biomedical). As shown in Figure 8b, the lactate intracellular concentration shown in Figure 9a is neither the
concentration increased 10-fold from Day 2 to 6, after cytosolic concentration nor the mitochondrial concentration,
which the concentration dropped sharply by 30-fold and but an average based on cellular volume. However, the use of
stayed at a low level until the end of the process. It is inter- intracellular average concentration does not change the citrate
esting to observe that on Day 9, when the extracellular lac- and isocitrate cellular efflux conclusion.
tate concentration decreased to a very low level, pyruvate in The extracellular malate and fumarate concentrations
the broth became undetectable (Figure 8b). At the same increased during the process. Their concentrations were
time, the two glycolytic metabolites preceeding pyruvate for- much lower initially, but reached a similar level to their cor-
mation, 3-phosphoglycerate and phosphoenolpyruvate, had responding intracellular concentrations at the end of the pro-
accumulated. These observations seem to suggest that the cess. Malate can be a TCA cycle overflow gate. When extra
flow from phosphoenolpyruvate to pyruvate might be par- malate is produced, it is converted to pyruvate by malate
tially blocked after Day 9, which resulted in the buildup of enzyme (Godia and Cairo, 2006). Results shown in this
metabolites in front of the blockage, and depletion of the study indicate that the conversion of malate to pyruvate
metabolites downstream of it. However, this hypothesis of a might be limited as malate concentration continued increas-
partial glycolytic pathway blockage conflicts with the glu- ing in the medium while pyruvate became undetectable after
cose consumption rate change illustrated in Figure 4b. Figure Day 8. The concentration of a-ketoglutarate varied during
4b showed that after all lactate was consumed, glucose con- the process without a clear trend. Similar metabolic shift
sumption rate went up (stage II to stage III transition). Alter- was observed in a NS0 metabonomics study where lactate,
natively, the accumulation of 3-phosphoglycerate and alanine, citrate, and succinate shifted from net production to
phosphoenolpyruvate could be due to a slow down of cell net consumption (data not shown). Isocitrate was not
proliferation. As both 3-phosphoglycerate and phosphoenol- detected in the NS0 study.
Biotechnol. Prog., 2009, Vol. 25, No. 5 1361

Figure 10. Glycolytic pathway and TCA cycle.


Metabolites that demonstrated a shift from net accumulation to net consumption during CHO CDF process are in bold.

Metabolic shift of glycolytic pathway and TCA link closely to the central metabolic pathway, to understand
cycle intermediates the mechanism(s).
Metabolites discussed in the previous sections are sum-
marized in Figure 10. The intermediates showing a transition Conclusions
from net accumulation to net consumption are in bold, and
the intermediates that were undetectable or not monitored A CDF with companion medium preloading for a fed-
are labeled with an asterisk. It is interesting to note that the batch NS0 process was successfully developed to replace
shifted metabolites are clustered together, downstream of py- a HCF. The new feed not only allowed us to achieve a
ruvate and upstream of fumarate with citrate considered the fully chemically defined process but also offered higher
start of the TCA cycle. Of all the intermediates monitored in peak cell density and higher product titer. When the CDF
this segment, a-ketoglutarate is the only one showing a dif- with its companion medium preloading was tested in a
ferent metabolic profile from lactate. The similar metabolism CHO process, it again demonstrated multiple significant
of a group of closely related metabolites suggests that the benefits. In addition to allowing the removal of hydroly-
metabolic shift was controlled by flux or by a group of sates, both viability and product titer were substantially
highly coordinated enzymes. A possible flux controlling improved. A lower lactate concentration is an additional
point is the synthesis of pyruvate from phosphoenolpyruvate. benefit of the CDF processes in both NS0 and CHO cul-
Again, the flux limitation hypothesis does not agree with the tures. In the CDF processes, there was a rapid shift in lac-
increased glucose consumption rate (Figure 4b from Stage II tate metabolism that resulted in a nearly complete
to Stage III). More studies are needed to elucidate the true depletion of lactate in the second half of the process. It
mechanism of the metabolic shift. First, in this study, we was also observed that the lactate metabolic shift occurred
monitored only a single enzyme (LDH). A more broad pro- concurrently with the transition of cell growth from expo-
teomics study is needed to assist interpreting the metabolo- nential to stationary phase.
mics results. Second, the metabolic analysis was largely Analysis of net glucose consumption by CHO cells indi-
based on trend analysis. A quantitative flux analysis could cated that the glycolytic pathway and pyruvate to lactate
increase the quality of the analysis and might lead to new conversion are closely coordinated. Their flows are strictly
findings. Third, it might be necessary to consider other path- complementary to each other so that the combined cell spe-
ways, such as urea cycle and lipid synthesis pathways that cific glucose and lactate consumption rate is a constant. A
1362 Biotechnol. Prog., 2009, Vol. 25, No. 5

plausible factor that could exert this tight control is cellular Literature Cited
maintenance of the NADþ/NADH balance. Altamirano C, Paredes C, Illanes A, Cairo JJ, Godia F. Strategies
LDH activity oscillated during the process. The lowest ac- for fed-batch cultivation of t-PA producing CHO cells: substitu-
tivity measured was during the lactate metabolic shift from tion of glucose and glutamine and rational design of culture me-
net accumulation to net consumption. However, the results dium. J Biotechnol. 2004;110:171–179.
Baggetto LG. Deviant energetic metabolism of glycolytic cancer
seem to be counterintuitive so that a more in-depth study of cells. Biochimie. 1992;74:959–974.
the LDH subunits and the other pair of substrates in the lac- Board M, Hummt S, Newsholme EA. Maximum activities of key
tate–pyruvate reaction, NADþ and NADH, is needed to enzymes of glycolysis, glutaminolysis, pentose phosphate pathway
determine the link between the LDH activity change and the and tricarboxylic acid cycle in normal, neoplastic and suppressed
lactate metabolic shift. cells. Biochem J. 1990;265:503–509.
Bogan JS, McKee AE, Lodish HF. Insulin-responsive compartments
Additional studies with the CHO process indicated that containing GLUT4 in 3T3-L1 and CHO cells: regulation by
several amino acids demonstrated metabolic shifts as well. amino acid concentrations. Mol Cell Biol. 2001;21:4785–4806.
Alanine, another byproduct of pyruvate, showed a similar Burky JE, Wesson MC, Young A, Farnsworth S, Dionne B, Zhu Y,
net accumulation to net consumption shift as lactate. This Hartman TE, Qu L, Zhou W, Sauer PW. Protein-free fed-batch
demonstrates that the metabolic shift is not limited to pyru- culture of non-GS NS0 cell lines for production of recombinant
vate–lactate conversion only. antibodies. Biotechnol Bioeng. 2006;96:281–293.
Burteau CC, Verhoeye FR, Mols J, Ballez JS, Agathos SN, Schnei-
Finally, profiling of most glycolytic and TCA cycle inter- der YJ. Fortification of a protein-free cell culture medium with
mediates in the CHO CDF process was conducted. In this plant peptones improves cultivation and productivity of an inter-
study, both intracellular and extracellular metabolite levels feron-c -producing CHO cell line. In Vitro Cell Dev Biol Anim.
were measured. A steep glucose concentration gradient was 2003;39:291–296.
observed across the plasma membrane, where intracellular Chen K, Liu Q, Xie L, Sharp PA, Wang DIC. Engineering of a mam-
malian cell line for reduction of lactate formation and high mono-
glucose concentration was more than two orders of magni-
clonal antibody production. Biotechnol and Bioeng. 2001;72:55–61.
tude lower than the extracellular concentration. The low glu- Chen Z, Iding K, Lukemeyer D, Lehmann. A low-cost chemically
cose concentration is likely due to the high phosphorylation defined medium for a recombinant CHO cell line producing pro-
rate of glucose in the cytosol, rather than a glucose transpor- thrombin. Biotechnol Lett. 2000;22:837–841.
tation limitation. Cruz HJ, Moreira JL, Carrondo MJT. Metabolic shifts by nutrient
manipulation in continuous cultures of BHK cells. Biotechnol and
When lactate was nearly depleted, the medium pyruvate
Bioeng. 1999;66:104–113.
became undetectable in the CHO CDF process. Meanwhile, Dang CV, Semenza GL. Oncogenic alterations of metabolism.
two glycolytic intermediates upstream of pyruvate formation, 3- Trends Biochem Sci. 1999;24:68–72.
phosphoglycerate and phosphoenolpyruvate, accumulated in the Dorai H, Kyung YS, Ellis D, Kinny C, Lin C, Jan D, Moore G,
medium. The opposite profile between phosphoenolpyruvate and Betenbaugh MJ. Expression of anti-apoptotic genes alters lactate
pyruvate suggests that the flux from the former to the latter metabolism of Chinese hamster ovary cells in culture. Biotechnol
might be partially blocked around the time at which the lactate Bioeng. In press.
Godia F, Cairo JJ. Cell metabolism. In:Ozturk SS,Hu W-S, editor-
metabolic shift occurred. However, this hypothesis is not sup- s.Cell Culture Technology for Pharmaceutical and Cell-Based
ported by the glucose consumption data, where glucose con- Therapies. New York: Taylor and Francis; 2006. pp 81–112.
sumption rate increased after the lactate metabolic shift. Gong X, Li D, Li X, Fang Q, Han X, Wu Y, Yang S, Shen BQ.
A considerable amount of citrate was exported out of mi- Fed-batch culture optimization of a growth-associated hybridoma
tochondria into the medium in the first 9-day culture, after cell line in chemically defined protein-free media. Cytotechnol-
ogy. 2006;52:25–38.
which citrate metabolism converted from net accumulation Harrison SA, Buxton JM, Czech MP. Suppressed intrinsic catalytic
to net consumption in the CHO CDF process. The extracel- activity of GLUT1 glucose transporters in insulin-sensitive 3T3-
lular citrate concentration increased up to 10-fold higher L1 adipocytes. Proc Natl Acad Sci USA. 1991;88:7839–7843.
than the intracellular average concentration. This observation Hata J, Tamura T, Yokoshima S, Yamashita S, Kabeno S, Matsu-
suggests that the TCA cycle in CHO cells might be partially moto K, Onadera K. Chemically defined medium for the
truncated. Besides citrate, isocitrate and succinate also dem- production of biologically active substances of CHO cells. Cyto-
technology. 1992;10:9–14.
onstrated similar metabolic shift as lactate and alanine. More Hatzivassiliou G, Zhao F, Bauer DE, Andreadis C, Shaw AN, Dhanak D,
interestingly, all the metabolites showing a metabolic shift Hingorani SR, Tuveson DA, Thompson CB. ATP citrate lyase inhibi-
are clustered together, generally downstream of pyruvate and tion can suppress tumor cell growth. Cancer Cell. 2005;8:311–321.
upstream of fumarate. Heidemann R, Zhang C, Qi H, Rule JL, Rozales C, Park S, Chuppa
Although the CDF fed-batch process demonstrated signifi- S, Ray M, Michaels J, Konstantinov K, Naveh D. The use of pep-
tones as medium additives for the production of a recombinant
cant empirical benefits over the previous HCF process and therapeutic protein in high density perfusion cultures of mamma-
provides a platform for the study of cellular metabolism, the lian cells. Cytotechnology. 2000;32:157–167.
cause(s) of the metabolic shifts in the CDF CHO process is Hu WS, Dodge TC, Frame KK, Himes VB. Effect of glucose on
still unknown. It could be a flux restriction, where a possible the cultivation of mammalian cells. Developments in biological
restriction point is the conversion of phosphoenolpyruvate to standardization. 1987;66:279–290.
pyruvate or a systematic enzymatic shift during cell growth Huang EP, Marquis CP, Gray PP. Development of super-CHO pro-
tein-free medium based on a statistical design. J Chem Technol
phase transition. Follow-up studies, especially a companion
Biotechnol. 2007;82:431–441.
proteomics study, will help elucidate the mechanism(s). Irani N, Beccaria AJ, Wagner R. Expression of recombinant cyto-
plasmic yeast pyruvate carboxylase for the improvement of the
production of human erythropoietin by recombinant BHK-21
Acknowledgments cells. J Biotechnol. 2002;93:269–282.
Irani N, Wirth M, van Den Heuvel J, Wagner R. Improvement of
The authors appreciate Dr. Kurt Droms and Dr. Amit Bane- the primary metabolism of cell cultures by introducing a new
rjee for their critical review of the manuscript and Process De- cytoplasmic pyruvate carboxylase reaction. Biotechnol and Bio-
velopment Analytical group for the amino acid analyses. eng. 1999;66:238–246.
Biotechnol. Prog., 2009, Vol. 25, No. 5 1363

Jeong DW, Kim TS, Lee JW, Kim KT, Kim HJ, Kim IH, Kim IY. Peterson PL, Mathupala S, Rempel A, Geschwind JF, Ko YH. Mito-
Blocking of acidosis-mediated apoptosis by a reduction of lactate chondrial bound type II kinase: a key player in the growth and
dehydrogenase activity through antisense mRNA expression. Bio- survival of many cancers and an ideal prospect for therapeutic
chem Biophys Res Commun. 2001;289:1141–1149. intervention. Biochim Biophys Acta. 2002;1555:14–20.
Kim SH, Lee GM. Down-regulation of lactate dehydrogenase-A by Sung YH, Lim SW, Chung JY, Lee GM. Yeast hydrolysate as a
siRNAs for reduced lactic acid formation of Chinese hamster low-cost additive to serum-free medium for the production of
ovary cells producing thrombopoietin. Appl Microbiol Biotechnol. human thrombopoietin in suspension cultures of Chinese hamster
2007a;74:152–159. ovary cells. Appl Microbiol Biotechnol. 2004;63:527–536.
Kim SH, Lee GM. Functional expression of human pyruvate carbox- Tsao YS, Cardoso AG, Condon RGG, Voloch M, Lio P, Lagos JC,
ylase for reduced lactic acid formation of Chinese hamster ovary Kearns BG, Liu Z. Monitoring Chinese hamster ovary cell culture
cells (DG44). Appl Microbiol Biotechnol. 2007b;76:659–665. by the analysis of glucose and lactate metabolism. J Biotechnol.
Kondoh H, Lleonart ME, Gil J, Wang J, Degan P, Peters G, Marti- 2005;118:316–327.
nez D, Carnero A, Beach D. Glycolytic enzymes can modulate Warburg O.The Metabolism of Tumors. London: Arnold Constable;
cellular life span. Cancer Res. 2005;65:177–185. 1930.
Lawton KA, Berger A, Mitchell M, Milgram KE, Evans AM, Guo Xie L, Nyberg G, Gu X, Li H, Möllborn F, Wang DIC. Gammain-
L, Hanson RW, Kalhan SC, Ryals JA, Milburn MV. Pharmacoge- terferon production and quality in stoichiometric fed-batch cul-
nomics. 2008;9:383–397. tures of Chinese hamster ovary (CHO) cells under serum-free
Luo Y, Chen G. Combined approach of NMR and chemometrics for conditions. Biotechnol Bioeng. 1997;56:577–582.
screening peptones used in the cell culture medium for the pro- \Xie L, Wang DIC. Stoichiometric analysis of animal cell growth and its
duction of a recombinant therapeutic protein. Biotechnol Bioeng. application in medium design. Biotechnol Bioeng. 1994;43:1164–1174.
2007;97:1654–1659. Zhang J, Reddy J, Buckland B, Greasham R. Toward consistent and
Macheda ML, Rogers S, Best JD. Molecular and cellular regulation productive complex media for industrial fermentations: studies on
of glucose transporter (GLUT) proteins in cancer. J Cell Physiol. yeast extract for a recombinant yeast fermentation process. Bio-
2005;202:654–662. technol Bioeng. 2003;82:640–652.
Miller WM, Wilke CR, Blanch HW. Transient responses of hy- Zhang J, Robinson D. Development of animal-free, protein-free, and
bridoma cells to nutrient additions in continuous culture: I. Glu- chemically-defined media for NS0 cell culture. Cytotechnology.
cose pulse and step changes. Biotechnol and Bioeng. 1989;33: 2005;48:59–74.
477–486. Zhou W, Chen CC, Buckland B, Aunins J. Fed-batch culture of
Nyberg GB, Balcarcel RR, Follstad BD, Stephanopoulos G, Wang Recombinant NS0 myeloma cells with high monoclonal antibody
DIC. Metabolism of peptide amino acids by Chinese hamster production. Biotechnol Bioeng. 1997a;55:783–792.
ovary cells grown in a complex medium. Biotechnol Bioeng. Zhou W, Rehm J, Hu WS. High viable cell concentration fed-batch
1999;62:324–335. cultures of hybridoma cells through online nutrient feeding. Bio-
Ozturk SS, Riley MR, Palsson BO. Effects of ammonia and lactate technol and Bioeng. 1995;46:579–587.
on hybridoma growth, metabolism, and antibody production. Bio- Zhou W, Rehm J, Europa A, Hu WS. Alteration of mammalian cell
technol and Bioeng. 1992;39:418–431. metabolism by dynamic nutrient feeding. Cytotechnology. 1997b;
Paredes C, Prats E, Cairos JJ, Azorin F, Cornudella Ll, Godia F. 24:99–108.
Modification of glucose and glutamate metabolism in hybridoma
cells through metabolic engineering. Cytotechnology. 1999;30:
85–93. Manuscript received Sept. 15, 2008, and revision received Feb. 28, 2009.

You might also like