You are on page 1of 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/229465560

Insect Cell Culture

Chapter · January 2010

CITATION READS

1 2,437

1 author:

Spiros N. Agathos
Université Catholique de Louvain - UCLouvain
245 PUBLICATIONS   6,924 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

industrial usage of WRF View project

Oxerom View project

All content following this page was uploaded by Spiros N. Agathos on 17 May 2014.

The user has requested enhancement of the downloaded file.


Insect Cell Culture
SPIROS N. AGATHOS

15
15.1. INTRODUCTION orders and originate from eggs or adult tissues like ovaries,
Insect cells were first isolated and put into culture in the imaginal disks, midgut, etc. (50). The IC-BEVS system
form of continuous cell lines in the late 1950s and early makes use mostly of lepidopteran cell lines, in particular
1960s for the study of insect metabolism and physiology but from Bombyx mori (silkworm), Mamestra brassicae, Spodop-
also for the in vitro synthesis of baculoviruses (a group of tera frugiperda (fall army worm), and Trichoplusia ni (cab-
viruses that infect invertebrates) as biological control agents bage looper). Among them, Sf-9 and Sf-21, isolated from
against insect pests. However, insect cell culture received a S. frugiperda, and Tn-368 and BTI-TN-5B1-4, isolated from
major boost when the genetic manipulation of baculoviruses T. ni, are the cell lines most commonly used in industrial
in the early 1980s made it possible to use them as vectors for applications. These cell lines are very susceptible to infec-
the production of recombinant proteins. Today the in vitro tion by Autographa californica multiple nucleopolyhedrosis
cultivation of insect cells coupled with infection with bacu- virus (AcMNPV) and other baculoviruses that form the
lovirus vectors (the insect cell-baculovirus expression vector basis for the construction of vectors in the IC-BEVS system.
system, or IC-BEVS) constitutes a major biomanufacturing A detailed, comprehensive, and up-to-date reference on
platform for the commercial production of occluded viruses all the techniques and methodologies involving the use of
as biopesticides and of recombinant proteins as human and insect cell culture and baculoviruses for research and indus-
animal vaccines and therapeutics. This is the result of a com- trial applications is given by Murhammer (55).
bination of advantages of both the cell culture component The first line to be intensively used in research and
and the vectors ensuring the transfer of heterologous genes: technological applications was Sf-21, isolated from ovar-
the culture of insect cells in vitro is now mastered to a simi- ian tissues of S. frugiperda. The Sf-9 cell line, derived from
lar extent as mammalian cell culture, and the application of Sf-21 (68), remains probably the most widely used of all
baculoviruses as vectors for recombinant protein production insect cell lines, thanks to its improved growth and high
or for manufacturing of viral biopesticides is safe and readily susceptibility to baculovirus infection, leading to excel-
amenable to scale-up (1, 36, 60). The IC-BEVS is a highly lent viral particle and protein yields. Tn-368 was obtained
versatile system because it can express gene products of from ovarian tissues of T. ni, and BTI-TN-5B1-4 is a clone
practically any origin (from bacteria to human tissue), and in of the embryonic Tn-5 cell line isolated from T. ni (31).
contrast to most industrial mammalian cell culture systems, it BTI-TN-5B1-4, patented by Granados (30), has been
is based on engineering only the vector and not the host cell commercialized under the name High Five™ (Invitrogen,
line. As a result, the development time needed to progress Carlsbad, CA) because of its superior capacity for secreted
from gene cloning to protein overproduction is much shorter glycoprotein production compared to Sf cell lines (20, 66)
(weeks instead of months). Compared to other biomanu- and now rivals Sf-9 as the most popular cell line for het-
facturing platforms, the IC-BEVS offers other significant erologous gene expression. Conversely, the Sf-9 cell line
advantages, including typically high product titers, a range is used more frequently for extracellular virus production
of posttranslational modifications, and the possibility to (e.g., biopesticides) because of its higher yield of assembled
express multimeric proteins or even several distinct proteins viral particles. A genetically modified Sf cell line com-
using the same vector. The ready adaptation of insect cells to mercialized under the name Mimic™ Sf-9 (Invitrogen)
suspension culture and the continuous improvement of cell is used for the stable expression of complex glycosylated
culture media and additives (3) are contributing to reliable heterologous proteins and is the same as the SfSWT-1 cell
and robust scale-up practices for commercial applications. line developed by the group of Jarvis (32).
The Sf lines are adapted to suspension cultivation and
are easily detached from T-flask (or other recipient) surfaces
15.2. CELL GROWTH by gentle agitation without trypsinization (57). Tn lines
were originally anchorage dependent, but today they are
15.2.1. Characteristics of Cell Lines fully adapted to suspension cultivation. Moreover, upon
Several hundreds of insect cell lines have been established microcarrier cultivation, High Five cells are more easily
from more than 100 different species encompassing seven detached than Sf-9 cells (34). Among Sf lines, Sf-21 cells

212

ASM_MIMB3_Ch15.indd 212 12/1/09 5:55:23 PM


15. Insect Cell Culture ■ 213

are more fragile than Sf-9; less tolerant to osmotic, pH, and Insect cells have been cultured for decades in noncom-
shear stress than Sf-9; and have a lower growth rate (57). mercial basal media, such as Grace’s, TNM-FH, TC-100, or
High Five cells are more robust to shear stress and osmotic IPL-41, that have been supplemented with 5 to 20% fetal
shocks than Sf-9 (42), although Sf-9 cells are more resistant bovine serum. In the last decade serum-free media have be-
to thermal shock (28). High-Five cells (15 ␮m) are larger come dominant in insect cell culture in order to overcome
and have higher protein content than Sf-9 cells (13 ␮m), the drawbacks of serum (cost, lot-to-lot variability, poten-
and they have wider cell size distribution than Sf-9 cells tial adventitious agents or contaminants, interference with
(J. C. Drugmand, Y. J. Schneider, and S. N. Agathos, un- product purification, etc.). Serum-free media are formulated
published data). However, the cell size depends on medium from the above basal media (typically IPL-41 or TNM-FH)
osmolarity, shear stress, and cell state (viable, apoptotic, upon supplementation with yeastolate (ultrafiltered yeast
etc.) (58). In general, Sf and Tn cells are bigger when extract), a lipid mixture emulsified in Pluronic F-68 (51)
infected (49). Maximal cell densities in serum-free batch and other, mostly proprietary complements. Commercial
culture can reach up to 9.6 ⫻ 106 cells/ml with High Five serum- and protein-free media, including BD BaculoGold
cells in YPR medium (Drugmand et al., unpublished) and Max-XP (BD Biosciences, Franklin Lakes, NJ), ESF 921
8.1 ⫻ 106 cells/ml with Sf-9 cells (63) in Sf900-II medium. (Expression Systems, Woodland, CA), EX-CELL® 405 and
Insect cell lines can grow over long-term passaging, which, 420 (Sigma-Aldrich, St. Louis, MO), Express Five® and
however, can give rise to morphological and physiological Sf900III™ (Invitrogen), HyClone SFX-Insect™ (Thermo
changes: decrease of productivity, increase of growth rate Scientific, Logan, UT), and Insect-XPRESS™ (Cambrex
and cell diameter (22), and lower susceptibility to growth Bio Science, Walkersville, MD), are used routinely for in- QU2
enhancement by conditioned medium (11, 12). Finally, the sect cell cultivation. These media, although able to support
tendency of baculovirus-infected cultured insect cells to- high cell densities and recombinant protein titers, are typi-
ward production of defective interfering particles (“passage cally expensive, cell line specific, and of proprietary com-
effect”) increases with higher cell passage number (43, 61). position. A comprehensive listing of most serum-free media
It is recommended to renew the working subcultivation currently available for insect cell culture has been compiled
after 30 passages (60). by Agathos (3). A dominant feature of medium design to-
The industrially and biotechnologically important cell day in response to stringent regulations is the elimination
lines of S. frugiperda and T. ni grow in the typical pattern of animal-derived components. Thus, supplements designed
of other animal cells in vitro. Following a short lag phase, to replace serum and other animal-derived ingredients are
the cells grow exponentially with doubling times between protein hydrolysates (peptones) of plant or microbial origin.
18 and 30 h. Starting from seeding densities of 2 to 4 ⫻ Hydrolysates may have a nutritional role if basal media with
105 cells/ml, maximal cell densities between 5 and 10 ⫻ lower amino acid content or no amino acids are used, but
106 cells/ml for Sf-9 and High Five cells are typically ob- mostly they are assumed to perform many serum functions
tained on day 5 to 7 postinoculation in batch shake-flask (protection from oxidative stress, shear damage, or apopto-
culture and in suspension bioreactors, depending on the sis, etc.), as shown by the use of yeastolate, which moreover
media formulation and the passage number of the inoculum. facilitates the purification of the recombinant proteins
These values can easily increase by 100% or more in cases produced in the IC-BEVS system (3, 36).
of fed-batch culture, especially when medium replacement In addition to their high cost, the lack of information on
or specific nutrient supplementation is practiced (36). The the detailed composition of commercial serum-free media
optimal temperature of insect cell cultivation is 27 to 28°C can also limit their attractiveness for specific biotechno-
and it does not require sparging of CO2. The optimal pH for logical applications. Hence, there is a need for formulating
the growth of most insect cells in culture lies between 6.2 serum-free media based on a rational assessment of the
and 6.3, somewhat more acidic compared to the growth pH metabolic requirements of the lepidopteran cells during
required by cultured mammalian cells. both the growth and production phases. The development
The biotechnological exploitation of cultured insect of low-cost, in-house serum- and protein-free media can be
cells is based on their infection with either a recombinant based on empirical, statistical, or advanced genetic algo-
baculovirus to produce a recombinant protein or with a rithm approaches (3).
baculovirus (mostly wild-type but also recombinant) to A typical empirical path toward formulating an in-house,
produce biopesticides in the form of budded virus. The pro- cost-effective medium starts with a basal medium requiring
duction of the recombinant protein or of the viral particles serum supplementation, such as IPL-41 (72), which has a
occurs late in the infection phase, when the infected cells simple composition and is easy to filter. Levels of glucose
are lysed due to the pathogenic effect of the baculovirus. and of glutamine are fixed first, according to the require-
Thus the IC-BEVS production platform is a lytic system ments of the cell line to be cultured. Next, the lipid mixture
and the formation of the viral or protein product is tran- emulsified in Pluronic F-68 (51). Alternatively, if the level
sient, as a new batch of insect cells must be grown and of Pluronic F-68 is to be varied, the lipid mixture (1,000⫻,
infected for a new cycle of production. liquid; Sigma-Aldrich) can be mixed directly with Pluronic QU3
F-68 (10% solution; Sigma-Aldrich or Invitrogen). Serum
15.2.2. Insect Cell Culture Media Development and amino acids are replaced with hydrolysates (or, alter-
Insect cell media contain the same basal ingredients as mam- natively, with different defined peptides and vitamins). All
malian cell media (carbohydrates, amino acids, and salts) components of the serum-free medium under development
but at concentrations adapted to insect cell metabolism. In are mixed together, followed by filtration through a 0.2-µm
contrast to mammalian media, they are supplemented with filter. Each component is tested by individual titration to
specific additives such as a lipid mixture to supply lipids determine its optimal level of supplementation. For this,
that insect cells are unable to synthesize. Other ingredients criteria of growth rate, viability, and growth extent (maxi-
QU1 added include the shear-protective agent Pluronic® F-68 mal or final cell density) are used for the growth phase, and
(typically at 0.1 to 0.2% [wt/vol]) and antifoam for agitated growth extent, viability, and product virus or protein titer
and sparged reactor cultivation. for the infection (production) phase. The last steps after

ASM_MIMB3_Ch15.indd 213 12/1/09 5:55:24 PM


214 ■ FERMENTATION AND CELL CULTURE

serum substitution must be repeated as different compo- T-flasks for adherent culture to shake flasks or spinners up
nents are added, in order to exclude combinations that have to 250 ml for suspension culture. The equipment required is
an adverse effect on growth and/or production. limited to an incubator and an orbital shaker. All addition
A statistical approach involves factorial experimental and removal of medium should be done in a biological safety
design, which enables the rational formulation of media cabinet or laminar-flow hood by using aseptic conditions
through screening of many ingredients at the same time and scrupulously sterilized equipment. The incubator is held
while keeping the number of experimental runs required low at an optimal temperature of 27 ⫾ 0.5°C for Sf-9, Sf-21, and
and manageable (54). This approach can result in great time High Five cells, which, moreover, do not need CO2, since
and cost savings. Ikonomou et al. (33) used a 27-4fractional most media are buffered.
factorial experiment for the screening of seven different Most types of insect cells tend to clump and lose viability
hydrolysates and a subsequent 22 full factorial experiment when they are transferred abruptly to suspension from mono-
for the optimization of the concentrations of the two se- layer culture or to serum-free from serum-supplemented
lected hydrolysates (yeastolate and Primatone RL). This led medium. It is recommended that one adapt the cells to sus-
to the formulation of the serum-free medium YPR, which pension culture before attempting to grow them in serum-free
performed equally well with commercial serum-free media medium. The increased sensitivity of the cells in serum-free
costing 10- to 20-fold more (33). A variation of this serum- culture requires reduced use or total elimination of antibiot-
free medium that we have found to perform equally well and ics, avoidance of trypsinization for dislodging adherent cells
to be as cost-effective is given in Table 1. Designated YSD, from culture vessels, and consistent cryopreservation (freez-
this versatile serum-free medium is an evolution of YPR to ing) and recovery (thawing) procedures. Cells propagated in
which a low level of dextran has been added and in which serum-free medium are also more fragile than cells grown in
Primatone RL, a meat hydrolysate, has been replaced by serum-containing medium. Therefore, upon subculturing,
soy protein hydrolysate to ensure that the medium is free of insect cells grown in serum-free medium must be separated by
animal-derived components. low-speed centrifugation. Dislodging of adherent cells from
In contrast to factorial experiments, which use only culture vessels should be done with mild tapping or shaking
a small number of levels, genetic algorithms may enable or with a gentle stream of medium.
the comprehensive optimization of insect cell media and For routine monolayer culture in polystyrene T-flasks,
cultivation conditions because they allow the screening of dishes, or multiwell plates, cells are inoculated at 2 to 5 ⫻
a wide variety of components and each of them at a wide 105 cells/ml (e.g., 0.8 to 2 ⫻ 106 cells/25-cm2 T-flask). The
range of concentrations. For instance, in the case of a fed- recipient container is incubated at 27 ⫾ 0.5°C for 3 days,
batch culture of Helicoverpa zea insect cells, a feed was de- whereupon spent supernatant medium is removed from the
veloped from 11 different medium components, each used monolayer and the culture is refed with fresh medium gen-
at a range of up to 31 concentrations. The feed was then tly introduced to the side of the recipient container. Sub-
optimized within four sets of 20 experiments (52). culturing to the next passage is done when the monolayer
reaches 90 to 100% confluency. Exponential growth can be
15.2.3. Small-Scale Culture maintained by splitting cells at a 1:5 dilution. After about
Small bench-scale culture involves volumes ranging from a 30 passages (2 to 3 months in culture) and after increased
total of a few milliliters in wells of multiple-well plates or doubling times (⬎28 h) have been reached, insect cells
tend to lose their viability and it is time to thaw new cell
cryovials to initiate new monolayer cultures.
Cell viability should be monitored at each passage and
should exceed 90%. Most insect cell lines do not adhere to
TABLE 1 Formulation of YSD, a generic all-purpose serum- monolayer substrata (e.g., T-flasks, dishes) as strongly as do
free medium for insect cell culture anchorage-dependent mammalian cell lines. In the case of
Component Content Supplier(s) High Five cells, which have already been adapted to suspen-
sion in many laboratories, an occasional tendency toward
QU9 Basal IPL mediuma Hyclone adhering to substratum and clumping can be counteracted
Glucose supplementb 41 mM by addition of heparin or dextran sulfate (20a) in order to
obtain single-cell suspensions. This is not necessary when
Glutamine 3.5 mM an already suspension-adapted cell line is obtained from a
supplementc commercial supplier (e.g., High Five cells from Invitrogen QU4
NaHCO3 4 mM have been adapted to grow individually in suspension).
Pluronic® F-68 0.1 % (vol/vol) Sigma-Aldrich Thus, monolayer cultures of all types of lepidopteran cells
Lipid mixture for 0.25 % (vol/vol) Sigma-Aldrich
can be used to initiate suspension cultures.
insect cell culture
A typical protocol of adaptation requires six to eight
confluent 25-cm2 T-flasks to start a 30-ml suspension culture
Dextran 67 kDa 0.1 % (wt/vol) Sigma-Aldrich (or six to ten 75-cm2 T-flasks to start a 100-ml suspension).
Yeastolate 6 g/liter Sigma-Aldrich After cells are dissociated from the bottom of the flasks with
Soy hydrolysate 5 g/liter DMV International a gentle stream of medium or by a sharp shaking motion,
Nutritionals (SE 50 the suspensions from the various T-flasks are pooled into
MAF), Kerry Bio- a shake flask or spinner and the cells counted. Fresh me-
Science (HySoy) dium is added to the cell suspension to reach 2 to 5 ⫻ 105
cells/ml. The shake flasks are incubated on the shaker at 90
a
Preparation in Milli-Q® water (Millipore), pH 6.2 to 6.3. to 100 rpm and the spinners are operated at a stirring rate
b
Final glucose concentration, 55 mM.
c
Final glutamine concentration, 10 mM. For certain applications a higher
of 40 to 60 rpm in an incubator (or constant-temperature
supplementation with free glutamine may be needed (final concentration, room) at 27 ⫾ 0.5°C. Subculturing into fresh medium is
15 to 17 mM). done when the viable cell count reaches 1 to 2 ⫻ 106 cells/ml,

ASM_MIMB3_Ch15.indd 214 12/1/09 5:55:24 PM


15. Insect Cell Culture ■ 215

and the stirring or shaking speed is increased by increments 15.2.4. Large-Scale Culture
of 5 rpm as long as viability is maintained above 90%. The capacity of insect cells to grow in suspension; the wide
This last step is repeated until a constant agitation speed availability of serum-free media and additives, including hy-
of 90 rpm for spinner flasks or 130 rpm for shake flasks is drodynamic shear-protective agents; together with the vast
reached and a consistent viability above 90% is maintained. prior experience using mammalian cells in biotechnological
Further subculturing from this point can be done at 0.3 to applications have opened the way for the development of
1 ⫻ 106 cells/ml (i.e., every 1 to 3 days). If cell clumping reliable and efficient large-scale insect cell culture processes.
is observed, the larger cell aggregates can be left to settle The successful scale-up from spinners or shake flasks of a few
and only single cells and small clumps are selected from the hundred milliliters to a large bioreactor of several hundred
upper third of the suspension for subsequent subculture. In a liters is done in steps, passing through progressively larger
final step, a sufficient quantity of cells fully adapted to sus- vessels. Scale-up requires a good understanding and control
pension culture should be frozen into cell banks to initiate of fundamental process parameters, such as adequate oxygen
future suspension cultures. delivery, good mixing, and appropriate modes of feeding,
Erlenmeyer shaker flask culture is by far the easiest way i.e., parameters whose consequences are crucial for optimal
to grow S. frugiperda and T. ni insect cells in suspension. The cell growth and efficient baculovirus infection. For instance,
most common configurations are 100-ml (working volume, the crucial phenomena of insect cell damage in an aerated
30 to 40 ml) or 250-ml (working volume, 80 to 100 ml) flasks. and stirred bioreactor can be assessed and mitigated by un-
Loosening the cap approximately one quarter turn enables derstanding and controlling the complex interplay among
unhampered aeration to the cells, which can grow at their the aeration rate through the orifices of a sparger, the agita-
maximum specific growth rate. In a representative sequence tion rate with a mechanical impeller, and the existence (and
of operations, the desired number of 250-ml shake flasks is extent) of gas-liquid interface for air bubble bursting and its
inoculated with 80 to 100 ml of medium containing 3 ⫻ 105 control by additives such as Pluronic F-68 (14).
viable cells/ml at a viability of at least 90%. The flasks are Generally speaking, the culture conditions allowing the
placed on an orbital shaker agitated at 125 to 140 rpm (de- attainment of high cell densities in the growth phase are
pending on the cell line and the medium) and maintained in different from conditions compatible with high production
an environment of 27 ⫾ 0.5°C. When the culture reaches 1 during the infection phase. Although the pH, temperature,
to 3 ⫻ 106 viable cells/ml, the shake flask contents are split dissolved oxygen (DO), and media conditions to sustain
to subculture the cells to about 3 ⫻ 105 viable cells/ml into high cell densities are well established, the corresponding
similar-size shake flasks with fresh medium. The subculturing parameters favoring an optimal outcome of the infection
should be done when the cells are in mid-exponential growth process and hence high recombinant protein productivity
and at maximum viability (i.e., above 90%). Every 3 weeks, are less well documented. In addition to these, productivity
cultures are centrifuged (100 to 200 ⫻ g for 5 min) and the of infected cells depends upon several more factors such
cell pellet is resuspended into fresh medium to reduce cell as virus construction (62), number of passages of the virus
debris and toxic by-product accumulation. (45), cell density at infection (CDI), multiplicity of infec-
Spinner culture is less common than shake flask culture tion (MOI) (16), “culture age” (36), cell state (773), and QU5
but better simulates the environment of a stirred bioreactor state of medium depletion (37). An important phenome-
for eventual scale-up. A spinner flask with a vertical impel- non underlying the distinct optima between the conditions
ler is preferred over that with a hanging stir bar because for growth and the conditions for efficient virus infection
it supplies better aeration. The liquid culture volume in a and productivity is the “cell density effect,” i.e., the consis-
spinner should not exceed one-half the graduated volume of tently observed drop in cell-specific productivity when CDI
the vessel, to ensure proper aeration (e.g., a 500-ml spinner exceeds a certain value (36). Its consequences and practices
should contain no more than 250 ml of culture fluid). A aiming at overcoming it are discussed below.
minimum volume should also be respected, so the impeller
stays submerged (e.g., 200 ml for a 500-ml spinner). In a 15.2.4.1. Selection of Bioreactors
spinner culture protocol for S. frugiperda, exponential-phase A number of bioreactor systems have been investigated and
monolayer culture cells can be inoculated at a cell density of proven their merits in terms of robustness and scalability
0.3 to 1 ⫻ 106 viable cells/ml (four or five confluent 75-cm2 (2, 36, 60). The selection of a production-scale bioreac-
T-flasks can supply this biomass for a 100-ml culture). Each tor system from the point of view of design (stirred-tank,
spinner is incubated at 27 ⫾ 0.5°C with constant stirring airlift, packed-bed, or Wave) and from that of cultivation
at 75 to 90 rpm. The top of the impeller should be slightly mode (batch, fed-batch, or continuous perfusion) should be
above the medium surface to provide additional aeration, and guided by considerations of product titer, overall productiv-
the sidearm caps of the vessels are kept loose to equilibrate ity, and product quality. Although in the 1990s there was a
the headspace with the atmosphere. When the cells reach tendency to experiment with a wide variety of bioreactor
a density of 2 to 3 ⫻ 106 viable cells/ml (i.e., once every designs including prototypes, the last few years have seen a
1 to 3 days), a sufficient quantity of fresh medium is added to reaffirmation of the selection of mainstream reactors, mostly
the spinner(s) to bring the cell density back to 106 cells/ml. of the stirred-tank type. For instance, a side-by-side com-
Alternatively, new, sterile spinners containing fresh medium parison of the bioprocess behavior of Sf-9 cells in an 8-liter
can be reseeded to about 3 ⫻ 105 cells/ml from a growing stirred-tank reactor and in a 6-liter airlift reactor showed
suspension culture. As in all growth protocols, insect cells similar growth characteristics and productivity of a recom-
should be monitored daily for density and viability. binant kinase (65). The flexibility and wide availability of
Growing shaker flask or spinner cultures of sufficient stirred-tank reactors make them more attractive than airlifts,
volume and good physiological characteristics (in mid- despite the latter’s recognized potential for adequate oxygen
exponential phase and with ⬎90% viability) can be used transfer rates with low and homogeneously distributed shear
for maintenance culture or for inoculating a higher-scale compared to conventional stirred vessels. At several hun-
vessel, e.g., a stirred Celligen™ bioreactor (New Brunswick dred liters, airlift scale-up rules impose a constant height-to-
Scientific, Edison, NJ). circumference ratio that can result in very tall, customized

ASM_MIMB3_Ch15.indd 215 12/1/09 5:55:25 PM


216 ■ FERMENTATION AND CELL CULTURE

vessels. In contrast, stirred-tank fermentors widely used in the baculovirus vector’s cytopathic effect on the cells will
microbial applications can be readily retrofitted with low- occur at about 24 h postinfection and the production of the
shear impellers (e.g., marine propellers or hydrofoils) and recombinant protein will increase after that time. Based on
adapted spargers for mammalian and insect cell cultivation. the definition of MOI as the ratio of PFU per cell at the
Packed-bed reactors make use of a cell immobilization time of infection, the volume of baculovirus stock, Vb, is
matrix (nonwoven fibers, foamy polymeric materials, and given by the equation
macroporous microcarriers) compactly loaded into a reten-
tion basket within the vessel (29). The main advantage of Vb ⫽ X ⫻ V ⫻ MOI ⫻ 103
__________________ (1)
the fixed bed is its ability to retain the cells and perfuse the P
medium across the reactor under low shear stress. In this de- where X is the viable cell density (cells/ml), V is the work-
sign, high cell densities can be reached in small bioreactors, ing bioreactor volume (liters), MOI is the target multiplic-
potentially leading to high productivity if the cell density ity of infection (PFU/cell), and P is the baculovirus stock
effect can be managed. However, the density of the immo- titer. However, as a rule, the volume of virus stock added
bilized cells in the bed is difficult to estimate, cell distribu- should not be much higher than 2% of the total culture
tion inside the matrix can be heterogeneous, and problems volume.
of oxygen limitation with gas exchange and metabolic by- A close monitoring of the growth and infection phases
product accumulation are possible. In addition, the packed by a combination of off-line and on-line techniques such as
cells must remain viable and productive for prolonged peri- OUR (see section 15.4) will allow the determination of the
ods during the culture. Although insect cell immobilization best time for harvesting, which, depending on the protein,
studies have some promise, as seen with microcarriers in a may be between 36 and 72 h postinfection.
stirred reactor (34), insect cell cultivation in packed-bed Fed-batch cultivation is today perhaps the most robust
reactors (5, 17, 18, 74) seems potentially more attractive for bioreactor operation mode, and its success seems to be
constitutive rather than transient protein production. linked in large measure to its capacity to overcome the cell
The Wave bioreactor was conceived for the suspension density effect, which depends on the CDI. This effect rep-
culture of shear-sensitive animal cells without the need for resents a consistent reduction of the specific productivity
protective additives like Pluronic F-68 and is part of the when the cell concentration at infection is above 3 to 4 ⫻
current trend toward single-use biomanufacturing systems 106 Sf-9 cells/ml or above 5 to 6 ⫻ 106 High Five cells/ml
(67). It consists of disposable, presterilized polyethylene bags (26). This is thought to be due to nutrient depletion in
(from 1 to 1,000 liters with working volumes from 100 ml the medium during infection, although its understanding
to 500 liters) placed on a rocking platform. The bags are remains incomplete. For instance, it has been shown that
filled with medium and cells and the rest of the chamber is the drop in productivity of Sf-9 cells occurs prior to nutri-
inflated with air (67). The rocking motion of the platform ent depletion (23). Complete medium replenishment or
generates waves, which enable good oxygen transfer and selected feeding of nutrients counteracts the cell density
mixing. A 20-liter Wave bioreactor was used in a side-by- effect (as reviewed in reference 36), lending support to nu-
side comparison with a stirred vessel for the production trient depletion as at least one causative factor. Generally,
of a secreted adhesion molecule (intercellular adhesion infection is more rapid at high cell density and high MOI,
molecule-1) by baculovirus-infected Sf-9 cells (71). Similar but the temporary burst in metabolic activity may not be
product yields were obtained in both the Wave bioreactor sustainable in the absence of fresh nutrients. Cell produc-
and the stirred-tank reactor, but operational costs were 40% tivity is also affected by the physiological state of the cells
lower for the disposable bioreactor. The Wave reactor is well (e.g., apoptotic cells are less productive than exponentially
adapted to the fed-batch mode of operation. On the other growing cells). Finally, the effect of the time of infection
hand, despite generating a constantly renewed liquid-gas (TOI; also known as culture age), i.e., the duration of the
interface, oxygen may be limiting at high-density infections culture from inoculation to infection, depends directly on
where oxygen uptake rate (OUR) increases transiently after the specific growth rate of cells, on the inoculation cell den-
infection (2). The introduction of a floating filter and a sity, and on the target CDI. Hence, as stated above in the
perfusion control system in the Wave bioreactor (56) has batch culture protocol, infections that aim to reach high
opened the way to its use in insect cell perfusion culture. recombinant protein production must be as synchronized as
In recent versions of this system it is possible to fit DO and possible, applied when the cells are highly viable in expo-
pH probes and to connect sample, input, and exit lines to a nential phase, and at that point, medium depletion must be
variety of analyzers for on-line monitoring (41). compensated with appropriate nutrient feeding.
Due to the depletion of medium ingredients and the den-
15.2.4.2. Batch, Fed-Batch, and Continuous sity effect, infection of simple batch cultures is not optimal
Culture for high protein production levels. An initial approach to
Batch culture remains the most common method for large- counteract this problem has been the total or partial me-
scale IC-BEVS processing because of its inherent simplicity dium replacement at the time of infection. Medium replace-
and flexibility in bioreactor equipment. In a typical proto- ment can be applied before or shortly after the infection and
col adapted from Elias et al. (27), the batch bioreactor is has been found to be efficient for recovering high specific
inoculated at a density of between 3 and 5 ⫻ 106 cells/ml production at high cell density (reviewed in reference 36).
by pooling cells from exponentially growing smaller-scale Moreover, the use of a cell retention (or centrifugation)
cultures (see section 15.2.3). The volume of the inoculum system to separate the cells from the exhausted media could
should represent approximately 10% of the working vol- serve to concentrate the cells (37). Partial medium replace-
ume. Cells are grown to 2 to 3 ⫻ 106 cells/ml (a CDI that ment is less costly and allows the retention of potential
is considered safe for infection, i.e., is not linked to a cell growth-promoting factors secreted by the cells (11, 12, 24,
density effect) and are then infected by adding the bacu- 36a). Complete medium replacement, originally proposed in
lovirus stock. If a synchronous infection is desired in the the early 1990s (47), helps to obtain high production with
interest of fast and abundant production, an MOI of 5 to 10 Sf-9 cells. Today, however, this solution is considered nei-
is appropriate. This ensures that a growth cessation due to ther economical nor practical for large-scale applications.

ASM_MIMB3_Ch15.indd 216 12/1/09 5:55:26 PM


15. Insect Cell Culture ■ 217

Fed-batch operation involves essentially the intermittent strategy should be adapted in a manner ensuring that there
addition of nutrients to a batch culture. It enables increased is no nutritional limitation during the infection phase,
cell density and productivity without medium depletion. It is since this may compromise the final production level.
easier and cheaper to implement than perfusion and avoids Continuous culture involving the steady-state feed of
exposure of the cells to high shear stress in a cell retention sterile medium and replacement of culture liquor without
device. However, it requires knowledge of physiological re- insect cell retention has not been used much for produc-
quirements of the cells. Moreover, in fed-batch mode, accu- tion of baculovirus or recombinant protein because the
mulation of by-products may be considerable. Nevertheless, continuous withdrawal of the culture from the continuous
the low sensitivity of insect cells to by-product accumulation, stirred-tank reactor (CSTR) causes the dilution of cells
to osmolarity increases, and to pH variation has made them and product (25). Moreover, the two distinct phases of the
a better candidate for fed-batch processing than mammalian production process (growth and infection) are not easily
cells. Fed-batch cultivation of insect cells has been studied implemented in a continuous mode in the same reactor due
intensively in the last several years, especially in the context of to the transient, lytic nature of BEVS. Thus, continuous
increasing both the cell density and the production yield using operation in a reactor cascade has been proposed, consisting
serum-free media and a suite of feeding strategies. Bédard et al. of two (or more) CSTRs in series, where the first is used for
(6) demonstrated that Sf cells needed a single-pulse addition of cell growth and the next for infection (77). A key limita-
yeastolate and amino acid mixture as supplement. Incidentally, tion in this configuration is the accumulation of defective
the use of a richer supplement consisting of glucose, tyrosine, virus (69).
vitamins, iron, and a trace metal solution led to very high In contrast to classical CSTR operation, continuous
densities of Sf-9 cells (30 ⫻ 106 cells/ml) (7). In a report from perfusion culture in a reactor equipped with a cell retention
the same group, (semi)continuous addition was better than system is better adapted to insect cell propagation because,
single-pulse addition for achieving high cell densities of Sf-9 just as in the case of fed-batch operation, it allows increase
cells (26). The highest cell density reported with lepidopteran in the cell density (16). Among the many cell retention
cells in fed-batch culture was 52 ⫻ 106 cells/ml using Sf-9 cells, systems available, perfusion culture of insect cells makes
but the cell density effect resulted in unsuccessful infection use of membrane-based modules (internal or external). The
at such a cell density (26). The highest reported Sf-9 CDIs lepidopteran cells cultivated in perfusion are reported to
sustaining high production were 11.5 ⫻ 106 cells/ml (7), 12 ⫻ attain very high cell densities and production levels. For ex-
106 cells/ml (15), and 17 ⫻ 106 cells/ml (26). There is less pub- ample, when cultivated in perfusion, Sf-21 cells were found
lished information on the fed-batch cultivation of T. ni cells. to reach 55 ⫻ 106 cells/ml (21), while Sf-9 cells attained 30
Infection of High Five cells at a density of 3.8 ⫻ 106 cells/ml ⫻ 106 cells/ml (13), and about 500 mg/ml of a recombinant
attained in serum-supplemented fed-batch culture allowed a protein was obtained with High Five cells (16).
modest production of ␤-galactosidase (75). In another report,
a serum-free fed-batch strategy was developed based on culture
pH as an indicator of a metabolic switch linked to the require- 15.3. BACULOVIRUS INFECTION
ments of High Five cells for glucose and glutamine (70).
A protocol adapted from the developers of high-cell- 15.3.1. Recombinant Baculovirus Vector
density culture using fed-batch methodology (27) involves Generation
a start-up phase of batch cultivation of Sf-9 cells followed Recombinant AcMNPV is generated by cotransfecting into
by additions of concentrated nutrient solutions together insect cells the linearized AcMNPV genome with a transfer
with close on-line monitoring and control of the integral plasmid carrying the gene of interest in an expression cas-
process. In this sequence, the baculovirus infection is done sette controlled by a late viral promoter such as that of the
routinely at cell densities of 10 ⫻ 106 cells/ml. The initial polyhedrin (polh) gene or of the p10 gene. Following re-
batch is inoculated at a density of 3 to 5 ⫻ 106 cells/ml combination of the expression cassette into the AcMNPV
with an exponentially growing culture of Sf-9 cells. The genome, the recombinant virus is amplified in an insect cell
nutrient concentrate, which includes glucose, amino acids, host and used for productive infections (62). The replace-
yeastolate, lipids, vitamins, and trace metals (6, 7, 26), is ment of either of the above nonessential genes (polh, p10)
added at 10% (vol/vol) to the culture volume at 24-h inter- with the foreign gene of choice and the very strong pro-
vals and the working volume of the culture is maintained moter control allow the production of recombinant protein
constant by removing an equivalent volume of mixed liquor at very high yield (53, 68). Owing to continuous improve-
between two consecutive feeds of nutrient concentrate. Fi- ments in recent years to the flexibility and efficiency of bac-
nally, the culture is infected when the cell density reaches ulovirus vector-based expression, this system is now adapted
10 ⫻ 106 cells/ml. As the cell density is progressively in- for simultaneous expression of several foreign genes using a
creasing, it is important to ensure adequate oxygenation of single recombinant baculovirus (76). The recombinant vi-
the culture by using an appropriate airflow rate or, if need ruses produced using these transfer vectors have facilitated
be, an O2-enriched gas mixture. The efficiency of oxygen the study of complex protein assembly, by coexpressing
supply can be checked by monitoring the DO concentra- protein subunits in the same host cells. Coinfection with
tion, which should not be allowed to drop below 20% several recombinant viruses is also possible to express differ-
air saturation. Both oxygen supply and nutrient adequacy ent genes (44, 64). The incorporation of the recombinant
should be checked throughout both the growth and the expression cassette into the AcMNPV genome by site- and
infection phases. As suggested in the batch culture protocol orientation-specific Tn7-mediated transposition has im-
above, an MOI of 5 to 10 is also recommended here for proved and simplified the selection procedure for amplified
synchronous infection. However, if a lower MOI is chosen recombinant baculovirus (4). In addition, recombinant
based on small-scale trials because of better and prolonged protein yield is significantly enhanced by the deletion of
product formation, the asynchronous infection will enable the genes coding for the baculoviral enzymes chitinase
significant postinfection growth that will contribute to oxy- (chiA) and v-cathepsin (V-CATH), which are involved in QU6
gen and nutrient consumption. Therefore, these substrates insect cell host lysis when the cytopathic effect takes hold
should be carefully monitored, and if need be, the feeding (40). The above improvements have been combined in a

ASM_MIMB3_Ch15.indd 217 12/1/09 5:55:27 PM


218 ■ FERMENTATION AND CELL CULTURE

baculovirus expression system named MultiBac, which is 15.3.3. Infection of Insect Cell Cultures
used mainly for the expression of heterologous multiprotein Important considerations involved in the process of infect-
complexes (8, 10). Many of the components and reagents ing an insect cell culture with a baculovirus are the cell line
needed for the generation of recombinant baculovirus vec- to use, the timing of the infection, the cell density at which
tors are now commercially available in the form of kits (e.g., the cells should be infected (CDI), the MOI to apply, and
from Invitrogen). the time at which to harvest the product.
15.3.2. Vector Amplification and Viral Titration 15.3.3.1. Cell Line Selection
Depending on the amount of viral stock required, The choice of insect cell line for use in the IC-BEVS bioman-
the baculovirus vector can be amplified in monolayer ufacturing platform depends primarily on the product to be
cultures, shaker flasks, or bioreactors, typically using expressed. Lepidopteran insect cells are able to bring about
a routine protocol of small-scale culture (see section eukaryotic posttranslational modification of heterologous
15.2.3). Although S. frugiperda and T. ni cells, in addi- proteins, such as N and O glycosylation, phosphorylation,
tion to several other lepidopteran cell lines, are readily fatty acid acylation, sialylation, ␣-amidation, N-terminal
susceptible to infection by wild-type and recombinant amidation, carboxymethylation, and isoprenylation (9, 32,
AcMNPV baculovirus, the general tendency in practice 38, 39, 59). Moreover, baculovirus-infected insect cells are
is to use Sf-9 or Sf-21 cell cultures for routine production able to effect folding of heterologous proteins and peptide
of baculovirus stock. It has been observed repeatedly that cleavage, although these recombinant proteins exhibit dif-
despite its capacity for overexpression of recombinant ferences from their authentic counterparts. As a rule, Sf-21
proteins (especially secreted ones) T. ni is less prolific and Sf-9 are the cell lines of choice for overproduction of
than S. frugiperda in the production of extracellular virus. whole viruses (e.g., as biopesticides) and also for the pro-
A low MOI of about 0.1 should be used for viral stock duction of intracellular or periplasmic proteins, whereas
production to ensure the purity of the stock and prevent High Five has been repeatedly demonstrated to overexpress
mutant accumulation. Usually a series of amplification many glycosylated and secreted proteins. Specifically, High
steps are carried out until the viral stocks reach 1 ⫻ 108 Five cells are supposed to carry out better posttranslational
or 1 ⫻ 109 PFU/ml. Adding 5 to 10% serum can enhance modifications than Sf-9 cells, with a higher percentage of
the stability of baculoviruses stored at 4°C (used as a complex glycoforms and even some sialylation (38, 39).
working viral stock), but cryopreservation of the viral
stock at −85°C is also possible without loss of stability 15.3.3.2. TOI, CDI, and MOI
for several months (master viral stocks). The fraction of the population of insect cells initially
An accurate baculovirus titer (in terms of PFU/ml) is infected by the virus is a crucial factor affecting cell pro-
essential to determine the strength of the stock for future ductivity. This fraction of the cell population is a function
infections. This effective concentration of infective par- of the MOI, which corresponds to the number of infectious
ticles can be obtained upon infection of insect cells in particles (PFU) divided by the number of cells, and follows
standardized laboratory assays, usually in culture dishes a Poisson distribution given by equation 2. In this relation-
and multiwell plates. The most commonly used method- ship, p(r) is the probability of a cell being infected by r virus
ologies for baculovirus titration are the plaque assay and particles and MOI is expressed in PFU/cell (57). QU7
end-point dilution (57). The plaque assay is carried out MOIr ⭈ exp(⫺MOI)
on tissue culture-treated dishes and involves the infection p(r) ⫽ _________________
r! (2)
of a monolayer of insect cells with a low concentration
of virus, followed by an agarose medium overlay. When The probability p(r) is equivalent to the fraction of cells
the infection is complete, a virus plaque (i.e., an isolated infected with r baculovirus particles at a given MOI. Since,
region of infected cells) is identified by visual inspection, according to equation 2, the fraction of uninfected cells
and an agarose plug is removed at the corresponding plaque (i.e., cells infected with r ⫽ 0 virus particles) at the given
location for generating viral stocks. With the end-point MOI would be p(0) ⫽ exp(⫺MOI), then it follows that the
dilution method, the culture medium is serially diluted to fraction f of cells infected with at least 1 virus particle would
the point at which any infection event is expected to result be f ⫽ 1 ⫺ p(0), i.e.:
from a single infectious viral particle. The direct visual
f ⫽ 1 ⫺ exp(⫺MOI) (3)
identification of positive wells (i.e., wells containing cells
infected by the baculovirus) is difficult for recombinant Various reports have examined the combined effect of TOI
baculovirus vectors lacking the polyhedrin gene. Because and MOI on productivity. Infection using a high MOI (⬎1)
of the exacting and time-consuming nature of both the implies that the infection is synchronized (i.e., all cells are
end-point dilution and plaque assay methods, a number of infected at the same time), while with an MOI of ⬍1 all cells
alternative techniques for determining the viral titer have are not simultaneously infected and several cycles of infection
been proposed. Thermo Scientific has commercialized a are required to infect the entire cell population. High MOI
kit containing the necessary ingredients for plaque assay, is thus predicted to lead to a short production period with
shortening the time of preparation (HyQ BEVS plaKit). high productivity (i.e., product concentration per unit time
Clontech (Mountain View, CA), Expression Systems, and of process duration). On the other hand, in larger bioreactors
Merck Biosciences (Darmstadt, Germany) offer quick and infection at a high MOI would also imply a need for large
efficient enzyme-linked immunosorbent assay kits for titrat- volumes of virus stock (see equation 1). As seen previously
ing the virus based on the detection of the gp64 viral capsid (section 15.3.2), a low MOI is used for virus amplification and
protein with a monoclonal antibody. Finally, viral titration for applications where viral stocks are low (57). In addition,
can also be accomplished through the detection of green infections at low MOI and low CDI can be successful and cost-
fluorescent protein, whose gene is incorporated in the virus effective in cell lines like Sf-9 that are very prolific in extracel-
genome and coexpressed together with the recombinant lular virus production or when it has been empirically found
protein of choice (19). that the accumulation of the product is not adversely affected

ASM_MIMB3_Ch15.indd 218 12/1/09 5:55:27 PM


15. Insect Cell Culture ■ 219

by a long infection phase. However, as a rule, asynchronous another technique for tracking the infection phase. Finally,
infections increase the time of harvest, lead to the produc- protein production can be followed by a variety of assays, in-
tion of defective viral particles, and expose the recombinant cluding fluorometric staining, Western blots, enzyme-linked
protein to proteases (2). Nonetheless, different opinions exist immunosorbent assay, and enzyme activity.
on this point. Some authors consider that an MOI between 1
and 20 leads to essentially similar levels of productivity (51),
while others contend that an MOI close to 1 is better (2). 15.5. PRODUCT RECOVERY
Still others recommend an optimization of both the TOI and The recovery of the recombinant protein or viral product
MOI in an effort to ensure that all cells are infected by the makes use of various well-established downstream processes
baculovirus and that there are sufficient concentrations of nu- that are commonly used in many biomanufacturing plat-
trients left for product (virus or recombinant protein) forma- forms (microbial fermentation, mammalian cell culture,
tion (73). Although previous attempts to predict the effect of etc.) but can vary widely as a function of the nature of the
MOI (and TOI) on recombinant protein production based on product (intracellular or extracellular protein, molecular
probabilistic mathematical simulations (46) have been borne size, state of ionization, hydrophobicity, etc.). The separa-
out experimentally (73), the phenomena are complex and tion of the insect cells may be a prerequisite in the case of
MOI optimization is not straightforward. In conclusion, MOI intracellular, periplasmic, or membrane-associated recom-
optimization depends on the virus, the cell line, the media/nu- binant proteins. This cell recovery step and all associated
tritional conditions, and the physiological state of the insect operations should be carried out at 4°C. Insect cells can be
cells at the time of infection. recovered by centrifugation at low speeds (500 to 1,000 ⫻
g for 5 to 10 min) or by tangential flow membrane filtra-
15.3.3.3. Time of Harvesting tion, e.g., using a Pellicon tangential filtration unit (Mil-
The time of harvesting is an important consideration in the lipore Corp., Billerica, MA) or a Filtron membrane unit
management of any biomanufacturing process involving (Pall Corp., East Hills, NY). An alternative possibility is
IC-BEVS. The recombinant protein must be harvested be- cross-flow microfiltration using hollow-fiber cartridges (GE
fore potential degradation by proteases that are secreted by Healthcare, Piscataway, NJ). The release of the intracellular
the insect cells, which may occur during lysis and disinte- or cell-associated protein may be aided by a prior cell lysis
gration of the cells (35, 48). This is especially critical in the step (mechanical milling or using a detergent mix in buf-
case of a secreted protein product. Therefore, as a general fer). In the case of a protein product secreted in the culture
guideline, a protein product should be harvested when the supernatant, the cells can be centrifuged at 1,500 ⫻ g for
cell viability falls below 80%. 10 min. If further processing is to be done several days after
harvesting, the cell paste and supernatant must be stored
at ⫺80°C until the next phase of separation and purifica-
15.4. QUANTITATIVE ASSESSMENT OF tion. It is recommended that large volumes of supernatant
GROWTH AND INFECTION PHASE, AND be concentrated by ultrafiltration through an appropriate
PROCESS MONITORING membrane cutoff before being frozen for further processing.
Insect cell growth is evaluated in terms of population dou-
bling time, specific growth rate (␮), growth extent (maxi-
mal cell density or CDI), length of the stationary/decline 15.6. SUMMARY
phase, and viability. Infection at a given MOI is assessed by The practice of insect cell culture is becoming more and
the time course of the postinfection cell density and viabil- more widespread as an essential component of the IC-
ity and, most importantly, by the evaluation of the viral or BEVS, perhaps one of the most successful and far-reaching
recombinant protein titer. CDI, MOI, and time of harvest- expression systems available today for the production of
ing must be optimized on an individual basis for different recombinant proteins and viral particles. In addition to
recombinant products, even though some general rules can its established applications in the production of ecological
be deduced (see section 15.3.3.2). biopesticides and of recombinant proteins as diagnostics and
Cell growth and infection in insect cell culture are com- veterinary vaccines, this technology platform is attracting
monly monitored by counting total and viable cell numbers increasing attention because of emerging applications in hu-
with a hemacytometer and using trypan blue staining/exclu- man health, including new-generation vaccines against hu-
sion. A more accurate but more time-consuming alternative man papillomavirus and influenza virus. Furthermore, in the
is the 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium era of proteomics, the IC-BEVS is an obvious choice for the
bromide cell proliferation assay, which can be applied using ample and rapid expression of hitherto unknown or poorly
a commercial kit (e.g., from the American Type Culture understood proteins of interest in fundamental biological
Collection [Manassas, VA]). Other less labor-intensive or research or with potential applications in drug discovery,
invasive techniques are also possible for monitoring cell sustainable agriculture practice, etc. Therefore, the under-
growth and infection, both off-line and on-line (2). Off-line standing of insect cell physiology in vitro and the constant
measurements of growth make use of optical density or of improvement of insect cell culture techniques using some of
a Coulter particle counter. On-line monitoring of growth the tools and concepts outlined above will ensure the indus-
through respiratory activity is based on determinations of trial maturity of the IC-BEVS through further development
cellular OUR or CO2 production rate using mass spectrom- of efficient, scaleable, and safe production processes.
etry or individual analyzers (O2 paramagnetic analyzer or
CO2 infrared analyzer). Specific OUR can also be deter-
mined using a DO electrode. With proper calibration it is REFERENCES
possible to monitor the infection phase via OUR because of 1. Agathos, S. N. 1991. Production scale insect cell culture.
the transient increase in oxygen consumption at the onset of Biotechnol. Adv. 9:51–68.
infection. Epifluorescence microscopy of live and dead cells 2. Agathos, S. N. 1996. Insect cell bioreactors. Cytotechnology
using fluorescence stains (Molecular Probes, Invitrogen) is 20:173–189.

ASM_MIMB3_Ch15.indd 219 12/1/09 5:55:28 PM


220 ■ FERMENTATION AND CELL CULTURE

3. Agathos, S. N. 2007. Development of serum-free media among various insect cell lines expressing a secreted gly-
for lepidopteran insect cell lines, p. 155–185. In D. W. coprotein. In Vitro Cell. Dev. Biol. Anim. 29A:842–846.
Murhammer (ed.), Baculovirus Expression Protocols, 2nd ed. 20a. Dee, K. U., M. L. Shuler, and H. A. Wood. 1997. Induc-
Humana Press, Totowa, NJ. ing single-cell suspension of BRI-TnB1-4 insect cells: 1.
4. Airenne, K. J., E. Peltomaa, V. P. Hytonen, O. H. The use of sulfated polyanions to prevent cell aggregation
Laitinen, and S. Ylä-Herttuala. 2003. Improved gen- and enhance recombinant protein production. Biotechnol.
eration of recombinant baculovirus genomes in Escherichia Bioeng. 54:191–205.
coli. Nucleic Acids Res. 31:e101. 21. Deutschmann, S. M., and V. Jäger. 1994. Optimization of
5. Archambault, J., J. Robert, and L. Tom. 1994. Culture of the growth conditions of Sf21 insect cells for high-density
immobilized insect cells. Bioprocess Eng. 11:189–197. perfusion culture in stirred-tank bioreactors. Enzyme Mi-
6. Bédard, C., A. Kamen, R. Tom, and B. Massie. 1994. crob. Technol. 16:506–512.
Maximization of recombinant protein yield in the 22. Donaldson, M. S., and M. L. Shuler. 1998. Effects of
insect cell/baculovirus system by one-time addition of long-term passaging of BTI-Tn5B1-4 insect cells on
nutrients to high-density batch cultures. Cytotechnology growth and recombinant protein production. Biotechnol.
15:129–138. Prog. 14:543–547.
7. Bédard, C., S. Perret, and A. A. Kamen. 1997. Fed-batch 23. Doverskog, M., J. Ljunggren, L. Öhman, and L.
culture of Sf-9 cells supports 3 ⫻ 107 cells per ml and im- Häggström. 1997. Physiology of cultured animal cells. J.
proves baculovirus-expressed recombinant protein yields. Biotechnol. 59:103–115.
Biotechnol. Lett. 19:629–632. 24. Doverskog, M., E. Bertram, J. Ljunggren, L. Öhman, R.
8. Berger, I., D. J. Fitzgerald, and T. J. Richmond. 2004. Sennerstam, and L. Häggström. 2000. Cell cycle progres-
Baculovirus expression system for heterologous multipro- sion in serum-free cultures of Sf9 insect cells: modulation
tein complexes. Nat. Biotechnol. 22:1583–1587. by conditioned medium factors and implication for prolif-
9. Betenbaugh, M. J., E. Ailor, E. Whiteley, P. Hinderliter, eration and productivity. Biotechnol. Prog. 16:837–846.
and T.-A. Hsu. 1996. Chaperone and foldase coexpres- 25. Drews, M., T. Paalme, and R. Vilu. 1995. The growth
sion in the baculovirus-insect cell expression system. and nutrient utilization of the insect cell line Spodoptera
Cytotechnology 20:149–156. frugiperda Sf9 in batch and continuous culture. J. Biotech-
10. Bienossek, C., T. J. Richmond, and I. Berger. 2008. Multi- nol. 40:187–198.
Bac: multigene baculovirus-based eukaryotic protein complex 26. Elias, C. B., A. Zeiser, C. Bédard, and A. A. Kamen.
production. Curr. Protoc. Protein Sci. 51:5.20.1–5.20.26. 2000. Enhanced growth of Sf-9 cells to a maximum density
11. Calles, K., I. Svensson, E. Lindskog, and L. Häggström. of 5.2 ⫻ 107 cells per mL and production of ␤-galactosi-
2006. Effects of conditioned medium factors and passage dase at high cell density by fed batch culture. Biotechnol.
number on Sf9 cell physiology and productivity. Biotech- Bioeng. 68:381–388.
nol. Prog. 22:394–400. 27. Elias, C. B., B. Jardin, and A. Kamen. 2007. Recombi-
12. Calles, K., U. Eriksson, and L. Häggström. 2006. Effect nant protein production in large-scale agitated bioreactors
of conditioned medium factors on productivity and cell using the baculovirus expression vector system, p. 225–245.
physiology in Trichoplusia ni insect cell cultures. Biotechnol. In D. W. Murhammer (ed.), Baculovirus Expression Proto-
Prog. 22:653–659. cols, 2nd ed. Humana Press, Totowa, NJ.
13. Cavegn, C., H. D. Blasey, M. A. Payton, B. Allet, J. Li, 28. Gerbal, M., P. Fournier, P. Barry, M. Mariller, F. Odier,
and A. R. Bernard. 1992. Expression of recombinant pro- G. Devauchelle, and M. Duonor-Cerutti. 2000. Adapta-
tein in high density insect cell cultures, p. 569–578. In R. tion of an insect cell line of Spodoptera frugiperda to growth
E. Spier, J. B. Griffiths, and C. MacDonald (ed.), Animal at 37°C: characterization of an endodiploid clone. In Vitro
Cell Technology: Developments, Processes and Products. But- Cell. Dev. Biol. Anim. 36:117–124.
terworth-Heinemann, Oxford, United Kingdom. 29. Goosen, M. F. A. 1993. Insect cell immobilization,
14. Chalmers, J. J. 1996. Shear sensitivity of insect cells. p. 69–104. In M. F. A. Goosen, A. J. Daugulis, and P.
Cytotechnology 20:163–171. Faulkner (ed.), Insect Cell Culture Engineering. Marcel
15. Chan, L. C. L., P. F. Greenfield, and S. Reid. 1998. Dekker, New York, NY.
Optimising fed-batch production of recombinant proteins 30. Granados, R. R. •••••••, 1994. Trichoplusia ni cell line
using the baculovirus expression vector system. Biotechnol. which supports replication of baculoviruses. U.S. patent
Bioeng. 59:178–188. 5,300,435. Boyce Thompson Institute for Plant Research,
16. Chico, E., and V. Jäger. 2000. Perfusion culture of bacu- Ithaca, NY. QU8
lovirus-infected BTI-Tn-5B1-4 insect cells: a method to 31. Hink, W. F. 1970. Established insect cell line from the
restore cell-specific -trace glycoprotein productivity at cabbage looper, Trichoplusia ni. Nature 226:466–467.
high cell density. Biotechnol. Bioeng. 70:574–586. 32. Hollister, J. R., E. Grabenhorst, M. Nimtz, H. O.
17. Chiou, T.-W., Y.-C. Wang, and H.-S. Liu. 1998. Utiliz- Conradt, and D. L. Jarvis. 2002. Engineering the pro-
ing the macroporous media for insect cell/baculovirus tein N-glycosylation pathway in insect cells for produc-
expression. Part 1: the entrapment kinetics of insect cells tion of biantennary, complex N-glycans. Biochemistry
within porous packed bed. Bioprocess Eng. 18:45–53. 41:15093–15104.
18. Chiou, T.-W., Y.-C. Wang, and H.-S. Liu. 1998. Utiliz- 33. Ikonomou, L., G. Bastin, Y.-J. Schneider, and S. N.
ing the macroporous media for insect cell/baculovirus Agathos. 2001. Design of an efficient medium for insect
expression. Part 2: the production of human interleukin-5 cell culture and recombinant protein production. In Vitro
in polyurethane foam and cellulose foam packed bed bio- Cell. Dev. Biol. Anim. 37:549–559.
reactors. Bioprocess Eng. 18:91–100. 34. Ikonomou, L., J.-C. Drugmand, G. Bastin, Y.-J. Sch-
19. Dalal, N. G., W. E. Bentley, and H. J. Cha. 2005. Facile neider, and S. N. Agathos. 2002. Microcarrier culture of
monitoring of baculovirus infection for foreign protein ex- lepidopteran cell lines: implications for growth and recombi-
pression under very late polyhedron promoter using green nant protein production. Biotechnol. Prog. 18:1345–1355.
fluorescent protein reporter under early-to-late promoter. 35. Ikonomou, L., C. Peeters-Joris, Y.-J. Schneider, and S.
Biochem. Eng. J. 24:27–30. N. Agathos. 2002. Supernatant proteolytic activities of
20. Davis, T. R., M. L. Shuler, R. R. Granados, and H. A. High-Five insect cells grown in serum-free culture. Biotech-
Wood. 1993. Comparison of oligosaccharide processing nol. Lett. 24:965–969.

ASM_MIMB3_Ch15.indd 220 12/1/09 5:55:29 PM


15. Insect Cell Culture ■ 221

36. Ikonomou, L., Y.-J. Schneider, and S. N. Agathos. 2003. 54. Montgomery, D. C., and G. C. Runger. 2006. Applied
Insect cell culture for industrial production of recombinant Statistics and Probability for Engineers, 4th ed. John Wiley
proteins. Appl. Microbiol. Biotechnol. 62:1–20. & Sons, Inc., New York, NY.
36a. Ikonomou, L., G. Bastin, Y.-J. Schneider, and S. N. 55. Murhammer, D. W. (ed.). 2007. Baculovirus and In-
Agathos. 2004. Effect of partial medium replacement on sect Cell Expression Protocols, 2nd ed. Humana Press,
cell growth and protein production for the High Five™ Totowa, NJ.
insect cell line. Cytotechnology 44:67–76. 56. Ohashi, R., V. Singh, and J.-F. P. Hamel. 2001. Perfusion
37. Jäger, V. 1996. Perfusion bioreactors for the production cell culture in disposable bioreactors, p. 403–409. In E.
of recombinant proteins in insect cells. Cytotechnology Lindner-Olsson, N. Chatzissavidou, and E. Lüllau (ed.),
20:191–198. Animal Cell Technology: from Target to Market. Kluwer
38. Joosten, C. E., and M. L. Shuler. 2003. Production of Academic, Dordrecht, The Netherlands.
a sialylated N-linked glycoprotein in insect cells: role of 57. O’Reilly, D. R., L. K. Miller, and V. A. Luckow. 1992.
glycosidase and effect of harvest time on glycosylation. Baculovirus Expression Vectors: a Laboratory Manual. W. H.
Biotechnol. Prog. 19:193–201. Freeman & Co., New York, NY.
39. Joshi, L., T. R. Davis, T. S. Mattu, P. M. Rudd, R. A. 58. Palomares, L. A., J. C. Pedroza, and O. T. Ramirez.
Dwek, M. L. Shuler, and H. A. Wood. 2000. Influence 2001. Cell size as a tool to predict the production of
of baculovirus-host cell interactions on complex glycosyl- recombinant protein by the insect-cell baculovirus expres-
ation of a recombinant human protein. Biotechnol. Prog. sion system. Biotechnol. Lett. 23:359–364.
16:650–656. 59. Palomares, L. A., C. E. Joosten, P. R. Hughes, R. R.
40. Kaba, S. A., A. M. Salcedo, P. O. Wafula, J. M. Vlak, and Granados, and M. L. Shuler. 2003. Novel insect cell
M. M. van Oers. 2004. Development of a chitinase and v- line capable of complex N-glycosylation and sialylation of
cathepsin negative bacmid for improved integrity of secreted recombinant proteins. Biotechnol. Prog. 19:185–192.
recombinant proteins. J. Virol. Methods 122:113–118. 60. Palomares, L. A., S. Estrada, and O. T. Ramirez. 2006.
41. Kadwell, S. H., and P. I. Hardwicke. 2007. Production Principles and applications of the insect cell-baculovirus
of baculovirus-expressed recombinant proteins in Wave expression vector system, p. 627–692. In S. S. Ozturk
bioreactors, p. 247–266. In D. W. Murhammer (ed.), and W.-S. Hu (ed.), Cell Culture Technology for Pharma-
Baculovirus Expression Protocols, 2nd ed. Humana Press, ceutical and Cell-Based Therapies. Taylor & Francis, New
Totowa, NJ. York, NY.
42. Kioukia, N., A. W. Nienow, A. N. Emery, and M. Al- 61. Pijlman, G. P., E. van den Born, D. E. Martens, and J.
Rubeai. 1995. Physiological and environmental factors M. Vlak. 2001. Autographa californica baculoviruses with
affecting the growth of insect cells and infection with large genomic deletions are rapidly generated in infected
baculovirus. J. Biotechnol. 38:243–251. insect cells. Virology 283:132–138.
43. Kool, M., J.-W. Voncken, F. L. van Lier, J. Tramper, and 62. Possee, R. D. 1997. Baculoviruses as expression vectors.
J. M. Vlak. 1991. Detection and analysis of Autographa Curr. Opin. Biotechnol. 8:569–572.
californica nuclear polyhedrosis virus mutants with defec- 63. Rhiel, M., C. M. Mitchell-Logean, and D. W. Murham-
tive interfering properties. Virology 183:739–746. mer. 1997. Comparison of Trichoplusia ni BTI-Tn-5B1-4
44. Kost, T. A., J. P. Condreay, and D. L. Jarvis. 2005. Bacu- (High Five™) and Spodoptera frugiperda Sf-9 insect cell line
lovirus as versatile vectors for protein expression in insect metabolism in suspension cultures. Biotechnol. Bioeng. 55:
and mammalian cells. Nat. Biotechnol. 23:567–575. 909–920.
45. Krell, P. J. 1996. Passage effect of virus infection in insect 64. Rhodes, D. J. 1996. Economics of baculovirus-insect cell
cells. Cytotechnology 20:125–137. production systems. Cytotechnology 20:291–297.
46. Licari, P., and J. E. Bailey. 1992. Modelling the popula- 65. Rice, J. W., N. B. Rankl, T. M. Gurganus, C. M. Marr,
tion dynamics of baculovirus-infected insect cells: optimiz- J. B. Barna, M. M. Walters, and D. J. Burns. 1993.
ing infection strategies for enhanced recombinant protein A comparison of large-scale Sf9 insect cell growth and
yields. Biotechnol. Bioeng. 39:432–441. protein production: stirred vessel vs. airlift. BioTechniques
47. Lindsay, D. A., and M. J. Betenbaugh. 1992. Quantifica- 15:1052.
tion of cell culture factors affecting recombinant protein 66. Saarinen, M. A., K. A. Troutner, S. G. Gladden, C.
yield in baculovirus-infected insect cells. Biotechnol. Bioeng. M. Mitchell-Logean, and D. W. Murhammer. 1999.
39:614–618. Recombinant protein synthesis in Trichoplusia ni BTI-
48. Lindskog, E., I. Svensson, and L. Häggström. 2006. TN-5B1-4 insect cell aggregates. Biotechnol. Bioeng. 63:
A homologue of cathepsin L identified in conditioned 612–617.
medium from Sf9 insect cells. Appl. Microbiol. Biotechnol. 67. Singh, V. 1999. Disposable bioreactor for cell culture using
71:444–449. wave-induced agitation. Cytotechnology 30:149–158.
49. Lloyd, D. R., P. Holmes, L. P. Jackson, A. N. Emery, 68. Smith, G. E., M. D. Summers, and M. J. Fraser. 1983.
and M. Al-Rubeai. 2000. Relationship between cell size, Production of human interferon in insect cells infected
cell cycle and specific recombinant protein productivity. with a baculovirus expression vector. Mol. Cell. Biol.
Cytotechnology 34:59–70. 3:2156–2165.
50. Lynn, D. E. 2001. Novel techniques to establish new in- 69. van Lier, F. L., J. P. van den Hombergh, C. D. de Gooijer,
sect cell lines. In Vitro Cell. Dev. Biol. Anim. 37:319–321. M. M. den Boer, J. M. Vlak, and J. Tramper. 1996. Long-
51. Maiorella, B., D. Inlow, A. Shauger, and D. Harano. term semi-continuous production of recombinant baculo-
1988. Large-scale insect cell-culture for recombinant pro- virus protein in a repeated (fed-)batch two-stage reactor
tein production. Bio/Technology 6:1406–1410. system. Enzyme Microb. Technol. 18:460–466.
52. Marteijn, R. C. L., O. Jurrius, J. Dhont, C. D. de Gooi- 70. Wang, M.-Y., and S.-R. Doong. 2000. A pH-based fed-
jer, J. Tramper, and D. E. Martens. 2003. Optimization batch process for the production of a chimeric recombinant
of a feed medium for fed-batch culture of insect cells using infectious bursal disease virus (IBDV) structural protein
a genetic algorithm. Biotechnol. Bioeng. 81:269–278. (rVP2H) in insect cells. Process Biochem. 35:877–884.
53. Merrington, C. L., J. E. Bailey, and R. D. Possee. 1997. 71. Weber W., E. Weber, S. Geisse, and K. Memmert.
Protocol: manipulation of baculovirus vectors. Mol. Bio- 2001. Catching the Wave: the BEVS and the Biowave,
technol. 8:283–297. p. 335–337. In E. Lindner-Olsson, N. Chatzissavidou,

ASM_MIMB3_Ch15.indd 221 12/1/09 5:55:30 PM


222 ■ FERMENTATION AND CELL CULTURE

and E. Lüllau (ed.), Animal Cell Technology: from Target to 75. Wu, J., Q. Ruan, and H. Y. P. Lam. 1998. Evaluation of
Market. Kluwer Academic, Dordrecht, The Netherlands. spent medium recycle and nutrient feeding strategies for
72. Weiss, S.A., G. C. Smith, S. S. Kalter, and J. L. Vaughn. recombinant protein production in the insect cell-baculo-
1981. Improved method for the production of insect cell virus process. J. Biotechnol. 66:109–116.
cultures in large volume. In Vitro 17:495–502. 76. Zavodszky, P., and S. Cseh. 1996. Production of multi-
73. Wong, K. T. K., C. H. Peter, P. F. Greenfield, S. Reid, domain complement glycoproteins in insect cells. Cyto-
and L. K. Nielsen. 1996. Low multiplicity infection of technology 20:279–288.
insect cells with a recombinant baculovirus: the cell yield 77. Zhang, J., N. Kalogerakis, and K. Iatrou. 1993. A two
concept. Biotechnol. Bioeng. 49:659–666. stage bioreactor system for the production of recombinant
74. Wu, J.-Y., and M. F. A. Goosen. 1996. Immobilization of proteins using a genetically engineered baculovirus/insect
insect cells. Cytotechnology 20:199–208. cell system. Biotechnol. Bioeng. 42:357–366.

ASM_MIMB3_Ch15.indd 222 12/1/09 5:55:31 PM


QUERIES:
QU1: Please add manufacturer and location.
QU2: Should this be Cambrex, East Rutherford, NJ?
QU3: Is “1,000×, liquid” correct? And should Sigma-Aldrich be here? Not necessary if already introduced at QU1.
QU4: This sentence seems to contradict the previous one; both are talking about High Five cells.
QU5: What reference should “773” be?
QU6: Italicize “V-CATH” if it is the gene name?
QU7: Please confirm accuracy of these equations.
QU8: Please give month of patent.
QU9: Is this related to the HyClone (cap “C”) brand from Thermo Scientific mentioned in the text? Also, need to fill in
“Content” column in this row?

ASM_MIMB3_Ch15.indd
View publication stats Sec1:a 12/1/09 5:55:31 PM

You might also like