You are on page 1of 13

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

net/publication/7017951

Bioreactor systems for the production of biopharmaceuticals from ani mal


cells

Article  in  Biotechnology and Applied Biochemistry · August 2006


DOI: 10.1042/BA20050233 · Source: PubMed

CITATIONS READS

102 3,590

2 authors:

James N Warnock Mohamed Al-Rubeai


University of Georgia University College Dublin
58 PUBLICATIONS   1,303 CITATIONS    304 PUBLICATIONS   10,815 CITATIONS   

SEE PROFILE SEE PROFILE

All content following this page was uploaded by James N Warnock on 17 July 2020.

The user has requested enhancement of the downloaded file.


Biotechnol. Appl. Biochem. (2006) 45, 1–12 (Printed in Great Britain) doi:10.1042/BA20050233 1

REVIEW
Bioreactor systems for the production of biopharmaceuticals
from animal cells

James N. Warnock* and Mohamed Al-Rubeai†1

*Department of Agricultural and Biological Engineering, Mississippi State University, Mississippi State, MS 39762, U.S.A., and
†School of Chemical and Bioprocess Engineering, University College Dublin and Center for Synthesis and Chemical Biology,
Belfield, Dublin 4, Ireland

The demand for biopharmaceutical products is set to be aware of the challenges faced with commercializing a
see a significant increase over the next few years. As product.
a consequence, the processes used to produce these Production capacity has significantly increased in the
products must be able to meet market requirements. last few years. Currently, many companies have a capacity
The present paper reviews the current technologies greater than 75 000 litres. In 2002–2003, only three compan-
available for animal cell culture and highlights the ad- ies had greater than 50 000 litres of mammalian cell culture
vantages and disadvantages of each method, while also capacity. According to an industry survey, the produc-
providing details of recent case studies. Processes are tion capacity for biopharmaceutical manufacturing will ex-
described for both suspension and anchorage-depen- pand by an average of 48 % over the next 5 years for mam-
dent cell lines. malian and microbial production systems. Approximately 35
biopharmaceutical products are currently on the market,
and an estimated 700 biotherapeutics are in clinical develop-
Introduction ment, with 150–200 in late-stage trials [1]. Obviously, the
requirements in terms of process development and bio-
The key component for the commercial success of any reactor capacity are tremendous. There is a total of approx.
biopharmaceutical product is the ability to achieve large- 92 companies operating Good-Manufacturing-Practice faci-
scale manufacture. Regardless of the efficacy of a drug, or the lities manufacturing biopharmaceutical products using mam-
efficiency of the downstream product purification, if there malian cell culture for clinical trials and in-market supply
is no process in place to produce the required amount of located in U.S.A. and Europe. The worldwide mammalian
the product to meet market demand, then it will fail. This cell culture capacity for Phase III and in-market supply of
issue was recently highlighted by Anthony Fauci, Director biopharmaceuticals was estimated to be 1.7 million litres at
of the National Institute of Allergy and Infectious Diseases the beginning of 2004, rising significantly in 2008 [2].
(Bethesda, MD, U.S.A.), in his announcement that scientists The present review will discuss the traditional and
had successfully tested a human vaccine towards avian flu. newly developed bioreactor technologies available to pro-
During his otherwise upbeat announcement, he remarked, duce biologics using animal cell culture on a large scale. Tra-
“The critical issue now is can we make enough vaccine, ditional systems are tailored towards anchorage-dependent
given the well-known inability of the vaccine industry to cell lines, but in most cases can be used for suspension cells.
make enough vaccine?” The problem lies in the fact that As more and more cell lines are adapted to serum-free
there has been an increase in the number and demand suspension culture, recent advancements have focused on
for biopharmaceutical products produced from animal cell cell retention in bioreactors to allow for perfusion methods
cultures in recent years. This has led to an increase in the
number of contract manufacturing companies; however, this
is still insufficient to meet market demand, and the problem Key words: animal cell biotechnology, biopharmaceuticals, bioreactor system,
will worsen as the number of approved biotherapeutics filtration system, microcarrier, perfusion culture.
increases. A part of the solution to this problem is to Abbreviations used: ATF, alternating tangential flow; BHK cell, baby-hamster
kidney cell; CHO cell, Chinese-hamster ovary cell; ECS, extracapillary
increase awareness and understanding of the complex pro- space; EPO, erythropoietin; HFBR, hollow-fibre bioreactor; ICS,
cess of producing biologics on a large scale. This will allow intracapillary space; IFN-γ, interferon-γ; IL-2, interleukin-2; IP, infectious
those currently engaged in this field to exploit areas of particles; mAb, monoclonal antibody; OUR, oxygen uptake rate;
STR, stirred tank reactor.
current weakness, while providing insight to those who are 1
To whom correspondence should be addressed (email
not actively involved in process scale-up so that they may m.al-rubeai@ucd.ie).


C 2006 Portland Press Ltd
2 J. N. Warnock and M. Al-Rubeai

Table 1 Characteristics of solid microcarriers divided into solid and macroporous carriers. Solid micro-
Particle
carriers have a lower surface area than macroporous car-
Microcarrier Manufacturer Material diameter (µm) riers, and can be susceptible to bridging when cells reach
high densities. Bridging occurs when cells attached to one
Cytodex 1 Amersham DEAE-dextran 147–248
Biosciences
carrier come into contact with another carrier and form a
Cytodex 2 Amersham THMAP–dextran 135–200 bond between the two. As a consequence, carrier aggre-
Biosciences gates form, which inhibits the efficiency of the reaction.
Cytodex 3 Amersham Gelatin–dextran 141–211
Biosciences
Macroporous carriers have a higher surface area and
FACT III SoloHill Cross-linked polystyrene 90–125 therefore have a greater potential for cell growth. It has been
Hillex (H) SoloHill Modified polystyrene 90–212 shown that Vero cells reach densities of 1.05 × 106 cells/ml
Biosilon Nunc Polystyrene 160–300
2D MicroHex Nunc Polystyrene 125 × 25
and CHO cells (Chinese-hamster ovary cells) can reach
densities of 2.16 × 106 cells/ml [6]. Similarly, V79 cells that
were genetically designed to express human cytochrome
P450 reached a density of 107 cells/ml after 11 days, resulting
to be used. Finally, several manufacturers have developed in a total of 3.6 × 1010 cells [7]. Unfortunately, cell densities
disposable bioreactors to simplify regulatory processes. are often limited by mass transport into the particles. As
the cell numbers increase, nutrients are unable to diffuse
into the centre of the carrier, while toxic metabolites, such
Anchorage-dependent versus as CO2 and lactate, are unable to diffuse out into the bulk
suspension cells medium. This leads to the evolution of a necrotic zone within
the particle. In addition, cell products are trapped within the
At the laboratory scale, or for high-throughput screening particle and cannot be harvested, which also leads to a
or monitoring purposes, it is often more desirable to use loss in product yield [8]. As a result, product titres from
anchorage-dependent cells than suspension cells. However, solid microcarrier cultures are often higher than those for
this poses problems with scale-up, as the amount of available macroporous cultures, as illustrated by Berry et al. [9]. The
surface area becomes limited. Several strategies have been characteristics of commonly used microcarriers are pre-
developed to maximize the amount of available surface area. sented in Tables 1 and 2.
To produce the required amounts of product for clinical In order to overcome the problems associated with
trials, scale-up is often performed in roller bottles. However, large-scale cultivation of anchorage-dependent cells, many
roller bottles have a finite surface area and scale-up can only cell lines have been successfully adapted to grow in suspen-
be achieved by increasing the unit number of bottles, which is sion culture. Scale-up of these systems is relatively simple,
extremely labour-intensive and susceptible to contamination and the maximum cell densities achieved in fed-batch
[3]. Modern facilities have overcome these problems by the cultures can be as high as 2 × 107 cells/ml. Suspension cells
use of expensive robots; however, it is still not possible to can be grown in either STRs (stirred tank reactors), similar
control environmental conditions such as pH or dissolved to those used in microbial fermentations, which are relatively
oxygen levels [4]. The most successful method for the scale- easy to scale-up beyond 10 000 litres, or airlift reactors,
up of anchorage-dependent cells has been the use of micro- which are more commonly used in wastewater treatment.
carriers, first reported by van Wezel [5], which provide a The simple design and operation, along with flexibility and
high surface-area-to-volume ratio and can be used in trad- ease of licensing, make these systems popular for manu-
itional stirred-tank bioreactors. Microcarriers can be sub- facturing.

Table 2 Characteristics of macroporous microcarriers

NA, data not available.

Microcarrier Manufacturer Matrix Diameter (mm) Pore size (µm) Porosity (%)

Cultisphere-G, S, GL Percell Biolytica Gelatin 0.17–0.5 ∼ 50 50


Cytoline 1, 2 Amersham Biosciences Polyethylene and silica (1.7–2.5) × (0.4–1.1) 10–400 65
Cytopore 1, 2 Amersham Biosciences Cellulose 0.2–0.28 30 95
Fibra-cel New Brunswick Scientific Polyester non-woven fibre and polypropylene 6 NA NA
ImmobaSil FS, D, HD Ashby Scientific Silicone (HD + 316L stainless steel) 0.8 × 0.3 (FS, HD) 50 × 150 > 40
10 × 1 (D)
Microsphere Cellex Collagen 0.5–0.6 20–40 75
Siran Schott Glasswerke Glass 0.3–5 10–400 60


C 2006 Portland Press Ltd
Bioreactors for cell culture 3

Figure 1 Factors that cause cell death in large-scale animal cell culture [97]
Figure 2 Scanning electron micrograph of CHO cells immobilized on
ImmobaSil FS macroporous microcarriers (scale bar, 50 µm)

Stirred tank bioreactors


Although many alternatives have been developed, the tradi- through stresses created by bubble entrainment and bubble
tional STR has emerged as the industry’s technology of bursting [13]. Highly agitated cultures will form vortices
choice [10]. The attractiveness of STRs stems from their which will increase the rate of bubble burst; thus the
simplicity, and the ease of monitoring and controlling scale- hydrodynamic forces that result from increased agitation
up. They are able to provide efficient gas transfer to cells have an indirect effect on cell viability [14].
and they have a long history of use with a variety of micro- Many cell lines that are cultivated in STRs have been ad-
organisms. Mixing in an STR is achieved by means of an impel- apted to serum-free suspension culture. Regulatory author-
ler, which is typically axial flow as opposed to a Rushton tur- ities have encouraged the development of processes that do
bine, the common choice for microbial fermentations. The not include materials of animal origin and, therefore, many
impeller speed will be sufficiently high enough to ensure that companies have developed defined serum- and protein-
each phase of the vessel contents is of uniform composition, free media. In the absence of serum, and therefore fibro-
i.e. they do not vary with position inside the reactor. Con- nectin, cells do not attach to a solid substrate and can be
sequently, STRs are homogenous systems when used at adapted to grow in suspension. Suspension cultures can
smaller scales. When the system is scaled up, difficulties in be easily scaled-up as the surface area to volume ratio is
homogeneity within the reactor can occur. The main mech- no longer a factor. Although STRs are commonly used for
anism of cell death in bioreactor culture is apoptosis. It can suspension cell culture, the same bioreactors can be used
be induced by exhaustion of nutrients, an absence or an for anchorage-dependent cell lines that do not survive
excess of dissolved oxygen, gross excursions from the ideal in suspension. These cells can be cultivated on solid or
physiological environment (e.g. extreme pH) and physical macroporous microcarriers. It is also possible to entrap sus-
abuse (see Figure 1). The mixing regimen has to be optimized pension cells within macroporous beads to increase the cell
in large-scale systems to alleviate these environmental pro- density. Macroporous microcarriers offer protection from
blems and ensure the dissolved oxygen concentration is the hydrodynamic forces, provided that cells are able to migrate
same throughout the liquid bulk phase [11]. This prevents towards the centre of the particle. Many fibroblasts can
the development of localized areas of either high or low move as quickly as 1 cm a day on a smooth surface [4], but
dissolved oxygen concentrations, which are detrimental to the internal surface of these matrices is rarely flat or smooth
cells, and the formation of dead zones. The disadvantage of and is quite tortuous for cells. However, once cells are pro-
such rigorous agitation is that high mechanical forces, such as tected from external forces, the mixing of the stirred tank
shear stresses, exist within the vessel. The eddies generated can be increased. The resulting high cell densities range from
by mixing do not cause direct damage to animal cells in 1 to 6 × 107 cells/ml [15–17], depending on cell line and
suspension, provided that their microscale of turbulence is microcarrier used. Figure 2 shows CHO cells that have been
greater than the cell diameter. Direct gas sparging is generally grown to a high density on ImmobaSil FS microcarriers.
the most damaging thing to cells, the extent of the damage Stirred tanks have been successfully used in the produc-
decreasing with reduced sparging rate until it reaches below tion of several biotherapeutic products, including mem-
0.2 vvm (volume · volume−1 · min−1 ), where it becomes equi- brane-anchored recombinant proteins, urokinase-type
valent to unsparged conditions [12]. Cell damage can occur plasminogen activator, prourokinase, IgG and IgA mAbs

C 2006 Portland Press Ltd
4 J. N. Warnock and M. Al-Rubeai

contact between cells and the impeller blades, the second


is contact between cells and other stationary bioreactor
components, such as the baffles, and the third arises from
bubble burst caused by the vortices created by the impeller,
as mentioned in the previous section. In an airlift bioreactor,
these problems are eliminated by the absence of an impeller.
Furthermore, the recirculation of the cells and medium
reduces the exposure of cells to surface bubble burst.
As with STRs, airlift bioreactors can be used to cultivate
either suspension cells [23] or anchorage-dependent cells
cultivated on microcarriers [24]. It has been recently shown
that BHK-21 cell (baby-hamster kidney 21 cell) cultivation
in protein-free medium is feasible in 1000-litre airlift bio-
reactors. The cells had a higher metabolic rate than in a
Figure 3 Schematic representation of an airlift bioreactor with internal bubble-free aerated STR; however, this may have been due
recirculation (left panel) and external recirculation (right panel)
to an increase in cell stress, as specific IL-2 (interleukin-
2) productivity was significantly lower in the airlift reactor
(monoclonal antibodies) and vesicular stomatitis virus cultures, relative to the STR, and lactate dehydrogenase was
[15,18–22]. higher in airlift bioreactors when compared with the STR
The largest stirred tank bioreactors available are [25]. Airlift bioreactors have been scaled-up to 2000 litres
20 000 litres, which are operated by Lonza Biologics in and have been routinely used in a fed-batch mode to
Portsmouth, NH, U.S.A. To our knowledge, fed-batch mode produce mAbs for therapeutic and diagnostic use. Further
is the most common approach in industry for the production information about the recent advances in airlift bioreactors
of biopharmaceuticals from mammalian cells. can be found in the review by Merchuk [26].

Airlift bioreactors
Filtration systems
Although STRs are considered to be the industrial standard
for animal cell biotechnology, airlift bioreactors have been STRs can be run in three different modes, namely batch,
used in a number of large-scale processes. Most notably, fed-batch or perfusion, and each has its own advantages and
airlift reactors are used by Lonza Biologics, the world’s disadvantages [27]. Fed-batch systems are widely used in in-
largest contract manufacturer of mAbs. Airlift bioreactors dustry as they can provide greater cell densities and product-
can take on two configurations, namely internal-loop or ivity compared with batch reactions. The advantage of per-
external-loop, as shown in Figure 3. These reactors offer fusion cultures is that cells are retained in the reactor
some advantages over STRs, such as being more energy- while the product is continuously harvested; therefore high
efficient, not having a mixer shaft and having a less harsh productivities can be achieved in a smaller bioreactor than
hydrodynamic environment. The lack of a mixer shaft eli- for batch or fed-batch systems [28]. However, for perfusion
minates one source of potential contamination, albeit one reactions to be efficient, and a viable option for industrial
of low risk. However, the lack of an impeller is a distinct production, a good separation method is required to retain
advantage. The rigorous agitation required to provide cells in the vessel while simultaneously harvesting the pro-
a homogeneous environment in an STR presents three duct. A comparison between batch and perfusion cultures
possible mechanisms of cell damage. The first is direct is presented in Table 3. Separation of cells and product on a

Table 3 Comparison of batch, fed-batch and perfusion culture systems

Stirred batch (batch/fed-batch) Hollow fibre Spin-filter and dialysis membrane

Culture type Suspension Stationary Suspension


Cell concentration Low/medium High Medium
Productivity Low/medium High High
Specific cell productivity Low/medium Insufficient data High
Scale-up Easy Impossible Easy
Operation Simple Complex Relatively simple
Monitoring and control Direct and easy Indirect and complex Direct and easy
Operating cost Low High Low


C 2006 Portland Press Ltd
Bioreactors for cell culture 5

large scale is based on the physical properties of the parti- Dialysis perfusion
cles, and the different methods utilize either size or The use of a dialysis membrane is a simple yet effective way
density. of performing a perfusion culture. The membrane allows
for the retention of high-molecular-mass components, such
as antibodies, as well as cells, while low-molecular-mass
Spin filters components and metabolic waste products can be conti-
Of all the cell retention methods currently used, the spin nuously removed. Dialysis operates on the principle of
filter is probably the simplest and the oldest device. The mechanical or osmotic pressure differences being present,
spin filter retains cells by use of a wire mesh that rotates allowing for the net transport of molecules up to a certain
in the centre of the reactor vessel. The steric effect of the size to cross the membrane. The molecular cut-off of the
rotating screen allows cells to be retained when the screens membrane allows for the selectivity of the system. The sys-
have pore sizes larger than the average cell [28]. Spin filters tem has several advantages over other perfusion methods;
typically are made of 316L stainless steel and have pore sizes the product can be retained in the bioreactor and is sub-
that range from 10 to 25 µm [29–31], although larger pore sequently recovered at a far greater density, which simplifies
sizes of 50 µm and even 120 µm have been successfully the purification process. Dialysis tubing can be easily re-
used to retain cell aggregates [32] and microcarrier cultures trofitted to standard STRs, and expensive consumables,
[33]. In addition to allowing cell separation from product in special equipment or technically challenging operations are
perfusion cultures, the use of a spin filter has been proven not required. Finally, system homogeneity allows for simple
to reduce hydrodynamic damage to cells; consequently, cell reactor control and assessment of the cellular physiology
viability and productivity are improved when compared with [37].
cultures performed in the absence of a spin filter [34]. The Several groups have evaluated the dialysis perfusion
primary concern when using a spin filter is fouling or clogging system as either an internal or external culture system.
of the screen. There are two different mechanisms behind The internal method cultures hybridoma cells inside the
clogging [29]; the first is when the filtration flux exceeds the dialysis tubing, which is suspended in medium [38,39]. Un-
retention capacity of the filter screen and cells accumulate in fortunately, the cell compartment is not well mixed and
the pores. This can occur suddenly. The second mechanism consequently the maximum usable volume is limited. The
occurs at a much lower rate and is only apparent in long- external method effectively turns the system inside out
term cultures that exceed 40 days. Cells attach to the screen with the cells being cultured in the reactor vessel and the
and begin to grow. Once they reach a certain density, they perfusate flowing through the dialysis tubing. This provides
will block the pores, preventing the product from being a large, well-mixed cell compartment and a small medium
removed from the vessel. compartment that is attached to a larger medium reservoir
Spin filters have been successfully used in numerous [37]. These systems have been scaled up to 1000 litres [40].
studies that have investigated biotherapeutic production The length (and therefore the surface area) of the tubing
from animal cells. The performance of a spin filter can be has been shown to be an important parameter that affects
enhanced by optimizing the culture conditions and perfusion the maximum viable cell number. Despite the potential
parameters. Spin filter rotation speed and perfusion rate do application of dialysis perfusion systems, they have not been
not affect the retention of viable cells; however, enlarging cell utilized in industry. Very few recent developments have
diameters and the formation of aggregates during opera- been made and they are considered an old-fashioned techno-
tion of a perfusion culture increase cell-retention rates logy.
from 75 to 95 % [30]. Additionally, the cell retention per-
formance of a spin filter has been shown to be comparable
with that of an ultrasonic filter. When a hybridoma bcl-2 cell HFBRs (hollow-fibre bioreactors)
line was cultivated in both systems, the maximum cell num- HFBRs operate on the same principle as dialysis perfusion
bers achieved were 1.21 × 107 and 1.58 × 107 cells/ml for the systems. In this bioreactor, cells attach to the outer surface
ultrasonic filter and the spin filter respectively [35]. Corres- of semi-permeable fibres, growing in the ECS (extracapil-
pondingly, when hybridoma cells were cultivated using a lary space) while medium is circulated through the ICS
medium supplemented with 1 % newborn-calf serum and (intracapillary space) or lumen. Nutrients diffuse through
peptone, the maximum cell number maintained in steady the fibres, usually made of cellulose acetate, while toxic
state was 1.3 × 107 cells/ml [36]. Another consideration is metabolites diffuse into the ECS and are carried away from
scale; the largest ‘off-the-shelf’ spin filter is suitable for a the cells. A unit consists of thousands of fibres traditionally
50-litre bioreactor. Consequently, the use is limited to pro- housed in a cylinder, but recent devices house fibres in a
duct research and development studies and they are not cartridge. The product accumulates within the ECS and can
used for commercial manufacture. be harvested intermittently.

C 2006 Portland Press Ltd
6 J. N. Warnock and M. Al-Rubeai

There are numerous advantages to using HFBRs and presented by Gorenflo et al. [56] were able to demonstrate
they have received attention for the commercial production that an air-back-flush system was able to eliminate recircu-
of mAbs and recombinant proteins [41–43]. The main ad- lation pumping of cells, thus eliminating the potential of shear
vantage of HFBRs is the high cell densities that can be stress damage, while maintaining separation efficiencies
achieved. Numerous studies have reported near tissue above 90 % at perfusion rates of 10 litres/day and 107 cells/
densities of > 108 cells/ml [41,44]. These high densities in ml. An additional drawback of acoustic filtration systems
turn lead to increased production and concentration of is that they are an external device and, when cells are in
products and enhance the specific productivity in a relatively the external loop, they are exposed to uncontrolled con-
small bioreactor [45–47]. However, HFBR systems are ditions and nutrient concentrations for short periods of
associated with large diffusional gradients that limit scale- time [58]. It is not known how the cells respond to these
up and affect product quality. As medium enters the lumen, relatively short periods in the external loop and it is not
it encounters a positive transmembrane pressure and is able thought to be of great detriment. Nonetheless, air back-
to permeate into the ECS. As this pressure decreases along flush eliminates the need to pump cells and allows the selec-
the length of the fibre, it becomes negative towards the tion of a separator residence time that is suited to the sens-
outlet, causing medium to flow back into the ICS. Despite itivity of the particular cell line [56]. Most of the studies on
the superior cell densities, HFBR systems are not used in acoustic filters have been conducted on a small/laboratory
commercial processes. However, they have shown potential scale. Success at the industrial scale would be dependent on
in the development of a bioartificial liver assist device [48]. their ability to achieve high separation rates under high flow
conditions [49]. Filters can have up to 95 % efficiency with
perfusion rates of 200 litres/day. Beyond this, separation
Acoustic filters efficiency shows a sharp decline. To overcome this, larger
It has been suggested that acoustic retention devices have retention devices are needed, which in turn require a higher
the greatest potential of all the cell separation systems [49]. power input [54]. Further details of acoustic filter retention
Unlike several other separation devices, acoustic filters have technology can be found in the recent review by Shirgaonkar
no physical barrier and no mechanical parts and are there- et al. [49].
fore less susceptible to fouling or mechanical failure [50].
Acoustic cell separators generate a fixed ultrasonic wave
field that causes cells to form reversible aggregates at the Cross-flow filtration
pressure node of the field [51,52]. Differences in size, density Cross-flow filtration has been widely tested in industry, and
and compressibility allow the separation of cells from the may offer a good alternative to spin filters or acoustic devices
product or medium [53] or the retention of viable versus on a larger scale. The principle behind cross-flow filtration
non-viable cells [51,54]. On the small scale, the acoustic is to force the cell suspension to flow tangentially to a mem-
filter has been shown to be a reliable and simple cell reten- brane with the product crossing the membrane to allow
tion device for long-term cultures [55]. Furthermore, the for harvest. Cross-flow filtration was originally developed
system has been successfully scaled-up to 200 litres/day to reduce the fouling of membranes by particle deposition
perfusion cultures, showing comparable performance with in pores and concentration gradients of macromolecules,
small-scale systems. On a large scale, the performance of which causes aggregation and gel formation [28]. Ironically,
the separator was optimized and provided 96 % separation despite membrane rinsing or back-flushing, fouling and
efficiency [54]. clogging problems have been frequently reported [59–61].
The main concerns associated with acoustic cell reten- However, by incorporating an ATF (alternating tangential
tion devices are the increased shear stress that cells are flow) device, fouling is avoided by washing cell aggregates
exposed to during pumping and the pressure differences back into the bioreactor. The system works by alternatively
present in the separator [50,56]. These can lead to a de- pulling culture medium into the ATF inner chamber via the
crease in cell viability, cell-specific productivity or product movement of a diaphragm at the top of the unit and pushing it
quality, especially in shear-sensitive insect cells [57]. To over- back into the reactor. The back flow part of the cycle causes
come the problem of shear stress, it has been proposed a small back-flush to occur across the filter membrane,
that cells be back-flushed, allowing shear-sensitive cells to cleaning the filter pores and surface. Furthermore, the
be perfused with the separator. The original method of ATF uses a low-shear, large-surface-area diaphragm, which
back-flushing used clarified medium [57]. However, once reduces cell stress. This system was successfully used for
the medium has been used, it needs to be clarified again, the production of recombinant proteins in a microcarrier-
causing an unfavourable increase in the harvest pump based culture. By using such a device, Bleckwenn et al. [62]
flow to maintain a constant average perfusion rate [56]. reported that the exponential growth phase of the cells
The alternative method is to use air back-flush. Results was increased by 50 h when compared with batch culture.

C 2006 Portland Press Ltd
Bioreactors for cell culture 7

This had the effect of increasing the maximum cell density


from 1.5 × 106 to 4.4 × 106 cells/ml [62]. The ATF system
has been used at the 1000-litre scale.

Fixed and fluidized-bed systems


STRs and airlift bioreactors are well suited to suspension cell
cultures. However, not all industrial cell lines can be adapted
to suspension culture, and these cells are strictly adherent.
Thus, when using anchorage-dependent cells to produce
biologics, it may be more appealing to use high-density/low-
volume systems as opposed to high-volume/low-density
systems such as microcarrier STRs. The alternative methods
for the production of biologics are the fixed and fluidized-
bed systems. Figure 4 Schematic representation of the CelliGen Plus packed-bed
bioreactor (courtesy of New Brunswick Scientific; www.nbsc.com)

Packed-bed bioreactors
Packed-bed bioreactors have been used for the cultivation
and production of a wide range of cell lines and biologics. through silicone tubing. This simple but effective method has
Some recent examples include mAbs, anti-leukaemia factor been used for the successful culture of hepatocytes and the
from stromal cells, recombinant proteins (e.g. recombinant production of retrovirus particles from the human packaging
Ca2+ -binding receptor and HIV-1 gp120), and retrovirus cell lines FLYRD18 and TEFLYRD [71,72].
vectors for use in gene therapy [15,63–67]. A disadvantage of An alternative system is the CelliGen® packed-bed
microcarrier culture in stirred tanks is that cells detach from bioreactor, developed by New Brunswick Scientific (Edison,
carriers at high densities and, along with cell debris, cause NJ, U.S.A.), which is shown in Figure 4. Conventional
complications in product purification during downstream packed beds require a conditioning vessel to control culture
processing. Packed-bed bioreactors have the advantage that parameters such as temperature, dissolved oxygen and pH.
they are capable of generating high cell densities with a low The culture medium is circulated through the packed bed
concentration of free cells in suspension. This is possible and the product may be concentrated and continuously
due to the low shear forces present in the system. Cells or periodically harvested from the conditioning vessel for
are immobilized within porous carriers that may be porous prolonged periods. An important parameter in the optimiz-
ceramic beads [68], porous glass beads [69] or polyester ation of these arrangements is the circulation rate; if the rate
discs [15,64,67], which are packed and retained in a is too low, then gradients in pH, nutrients and metabolites
cylindrical vessel through which culture medium is recircu- will appear along the bed, affecting cell viability, productivity
lated. A characteristic problem in intensive production and product quality, as can be seen in HFBRs [73]. If the
systems is the reduced transport rate of limiting nutrients, circulation rate is too high, it may have an adverse effect
such as oxygen to immobilized cells, which restricts their on cell attachment or product formation. This problem is
final density. Packed-bed technology has overcome this eliminated in the CelliGen® packed-bed bioreactor by the
problem by the use of intraparticle convective flow. Through fact that the bed is contained in a basket within a stirred tank
the development of a simple hydrodynamic model based bioreactor. Mixing in the bioreactor is achieved with either
on the Blake–Kozeny equation, Park and Stephanopoulos a cell-lift impeller, which allows bubble-free medium to flow
[70] significantly improved the transport of oxygen in a through the packed bed [67] or a double screen concentric
packed bed, allowing the maintenance of cell viability and cylindrical cage impeller. Shi et al. [74] have shown that the
productivity while sustaining a low-shear environment. In latter impeller is able to increase convective mass transfer,
their evaluation of the model, they achieved cell densities of cell concentration and mAb product concentration. The
5.1 × 108 cells/ml. They also calculated the specific insulin significant improvements were attributed to the increased
productivity to be 0.88 × 10−5 microunit · cell−1 · h−1 . The surface area, allowing for convective oxygen transfer and
model developed allows packed-bed bioreactors to be protection of cells from hydrodynamic stresses. Both
scaled-up severalfold before oxygen becomes limiting. Any methods create a homogeneous environment for cells to
oxygen limitations that do occur primarily exist close to grow and product can be directly removed from the culture
the bioreactor outlet. Another development to overcome vessel, which allows for easier downstream processing and
this problem has been to provide oxygenation to the bed product purification.

C 2006 Portland Press Ltd
8 J. N. Warnock and M. Al-Rubeai

A disadvantage of packed-bed bioreactors is the inac-


cessibility of carriers, making it difficult to monitor viable
cell number. Cellular metabolism can be followed by online
measurements of pH and dissolved oxygen and offline deter-
minations of glucose, lactate, ammonia and glutamine. The
substrate uptake rates and production rates are con-
ventionally used as cell growth markers [75]. In general,
the cell concentration has a linear correlation with glucose
uptake rate at high cell densities. Thus it is possible to calcu-
late the latter from the former [76]. However, this yield
coefficient is not a true constant and will vary for each cell
line and for each bioreactor system. Therefore it should
be determined before being used to estimate cell number.
Alternatively, cell number can be determined through
measuring the OUR (oxygen uptake rate). By placing oxygen
probes upstream and downstream of the packed bed and
measuring the percentages of dissolved oxygen, it is possible
to determine the OUR, which can be directly correlated
with cell number. The density of cells within an immobilized
culture is an important parameter for many purposes, includ-
ing the calculation of the specific productivity of a system.
Figure 5 Schematic representation of the Cytopilot mini fluidized-bed
bioreactor
Fluidized-bed bioreactors
Abbreviation: dO2 , dissolved oxygen; Temp, temperature.
The most widely used culture system for porous micro-
carriers is the fluidized-bed bioreactor [77]. The principle
of their operation is that microcarriers of higher density
than the culture medium are suspended by the upward flow stirred culture. In contrast, the production rate of hy-
of the medium, which is circulated through the bed. The bridoma cells cultured in the fluidized bed remained appre-
height of the bed will increase as fluid flow increases. Medium ciably higher than that in stirred-tank culture. The difference
circulation and fluid flow through the bed are achieved by was assumed to be caused by BHK cells changing their
using either a peristaltic pump or agitation. The advantage of production characteristics during long-term cultivation. This
this system is that carriers are separated from direct aer- phenomenon had previously been reported in hybridoma
ation and agitation, which avoids local high shear forces. In cells [80]. An alternative reason is that, unlike hybrid-
addition, fluidized-bed bioreactors provide a very high mass- oma cells, specific cell productivity is related to specific
transfer interface, since both cells and fluid are moved [78], growth rate. It has been shown by Leelavatcharamas et al.
making it a suitable production system on the larger scale. [81] that there is a linear relationship between the pro-
Commercially available systems are available up to 400 litres, duction of IFN-γ (interferon-γ ) and the specific growth
which can produce cell densities of 2 × 108 cells/ml. There rate of CHO cells. Therefore, in long-term cultures where
are not currently any licensed production processes that use the cells remain in the stationary phase over an extended
fluidized-bed bioreactors, as is also the case with packed-bed period, productivity will be low as the specific growth rate
bioreactors. is essentially zero.
Earlier studies into the potential of fluidized-bed react- Another substrate for the immobilization of cells
ors for the production of biologics focused on the use grown in fluidized-bed bioreactors is polyethylene and silica
of porous glass beads. These studies conducted by Kratje microcarriers such as Cytoline (Amersham Biosciences,
et al. [77,79] examined the potential of fluidized-bed react- Uppsala, Sweden). These have been successfully used in the
ors as production systems using hybridomas suspension cells production of recombinant human proteins, especially hu-
and anchorage-dependent BHK cells for the production of man IFN-γ and EPO (erythropoietin) [82,83]. These micro-
IL-2 (interleukin-2). In these studies, cell densities of 3.8 × carriers have been specifically designed for use in the Cyto-
108 cells/ml intrasphere volume were reached, while the IL-2 pilot fluidized-bed bioreactor (Figure 5) and have proved to
production rate was 0.75 mg · l−1 · day−1 . In spite of the cell offer great potential for the production of biologics. Kong
density being over 18-fold higher, the productivity showed et al. [84] compared CHO cell cultures in this system with
a 1.9-fold decrease when compared with a homogeneous solid microcarrier culture in a stirred-tank bioreactor and

C 2006 Portland Press Ltd
Bioreactors for cell culture 9

reported that the product output rate was approximately also examined the productivity and quality of EPO from a
three times higher in the fluidized bed, with the specific recombinant CHO cell line, cultured in a Cytopilot fluidized-
production rate being 5.5 times higher. The volumetric pro- bed bioreactor. EPO is particularly vulnerable to a decline
ductivity of the stirred tank reached 5.6 mg · l−1 · day−1 during in sialylation due to the extensive level of glycosylation and
the growth period. However, after the maximum cell heterogeneity. It was therefore important to show that the
density was reached, the volumetric productivity decreased glycosylation profile of EPO in the Cytopilot did not differ
and was maintained around 1.65 mg · l−1 · day−1 . In contrast, from other culture systems. Results have shown that no
the fluidized-bed bioreactor maintained a consistent volu- difference was observed in electrophoretic product profiles
metric productivity of 22.8 mg · l−1 · day−1 . It was suggested between fluidized bed, stirred tank and stationary cultures.
that the isolated aeration and agitation in the fluidized- In addition, the significantly higher productivity rate in the
bed bioreactor reduced the amount of hydrodynamic and fluidized-bed bioreactor, when compared with STRs, makes
mechanical stress imposed on the cells. Thus more cellular it an attractive option for the large-scale production of re-
energy was utilized in product formation, resulting in the combinant proteins.
higher and more consistent productivity. The total product
yield was reported as 34.2 and 13.2 mg/day for the 2-litre
fluidized-bed bioreactor and the 10 litre stirred tank bio- Disposable bioreactors
reactor respectively. The Cytopilot has also demonstrated
good retroviral vector production, with a maximum virus Disposable bioreactors, such as the CellCube (Corning),
titre of 107 IP (infectious particles)/ml, and a total viral yield the Cell Factory (Nunclon) and the Wave bioreactor (Wave
of 5.03 × 1010 IP in 11.4 litres. In roller-bottle cultures, daily Biotech), are gaining increasing popularity for industrial
titres of 5 × 106 IP/ml were achieved from 50 ml per bottle applications. This is because they offer many benefits, which
(total yield: 2.5 × 108 IP per bottle). Therefore the total yield include low initial investment costs, reduced lifetime oper-
obtained from the Cytopilot over 19 days was equivalent to ating costs, reduced validation, simplified scale-up and low
the yield from ten daily harvests from 20 roller bottles [85]. risk of cross-contamination. The CellCube culture system
It was observed that virus titre increased with an increasing has been successfully used for the production of retrovirus
cell number. Therefore, as the Cytopilot has the potential for vectors, mAbs and recombinant proteins [66,86,87]. The
higher cell numbers in longer runs, it also has the potential performance of the CellCube is reported to be comparable
for greater virus titres. with the CelliGen® packed-bed reactor, with each system
When comparing the number of cells in each square producing an average of 2.1 × 107 and 0.55 × 107 IP/ml res-
metre of microcarrier surface area, the fluidized-bed bio- pectively [88]. The yield from the CellCube was also
reactor had a value five times lower than that of the STR. reported to be comparable with that from the automated
This shows that there is further potential to increase the cell roller-bottle system, RollerCell 40 [87]. When the CellCube
number and improve the productivity of the system. In order was run under Good-Manufacturing-Practice conditions, the
to unlock this potential, it is necessary to ascertain the limit- virus titre was between 1.2 × 106 and 3.0 × 106 IP/ml. This
ing factor for growth and how to overcome this limitation. was lower than titres recorded under test conditions, owing
Investigations carried out by Preissmann et al. [78] deter- to a higher perfusion rate. While the higher perfusion rate
mined that the transport of oxygen to cells within the reduced the vector concentration, it did increase the titre
macroporous matrix cultivated in a fluidized-bed bioreactor stability and thus the quality of the vector preparation.
was severely limited, leading to poor process performance. The disadvantage of the CellCube bioreactor is that some
Although no limitations in nutrient concentration were components, such as the oxygenator and the probes, must
detected, the beads were not believed to be an ideal sub- be cleaned and re-sterilized between batches. Cleaning
strate to grow cells to tissue-like densities. validation as well as process validation is relatively difficult
Efficient glycosylation is an important factor in the [87]. Additionally, the largest system available is the module
development of a bioprocess production method. The exi- 400, which has a surface area of 340 000 cm2 . The Cell
stence of gradients that cause decreases in dissolved oxygen, Factory possesses a number of attractive features, which
nutrients or pH may have implications for product quality, include unaltered growth kinetics from laboratory scale,
as well as cell densities. In a recent study, the relative pro- low risk of contamination and compact design. A number of
portion of glycosylation site occupancy for IFN-γ was not recent reports have shown the potential of the Cell Factory
seen to change over time during a 500 h culture. Hence, it for the production of vaccines and recombinant proteins
was concluded that long-term perfusion cultures of CHO [89–92]. However, the largest size module available is the
cells in a fluidized-bed bioreactor could produce IFN-γ with 40-tray system, which has a surface area of 25 280 cm2 . To
a consistent and highly comparable degree of glycosylation put these surface areas into perspective, a 12-litre working
[82]. Further studies, carried out by Wang et al. [83], have volume of Cytodex microcarriers at a concentration of 5 g/l

C 2006 Portland Press Ltd
10 J. N. Warnock and M. Al-Rubeai

would have a greater surface area than the CellCube or the Decisions (Moraga, CA, U.S.A.), Biopharmaceutical Con-
Cell Factory. tract Manufacturing 2005: Improved Processes and New
The Wave bioreactor can be used to culture suspension Capacity for Pipeline to Commercial Production (February
cells or microcarrier cultures and is marketed as an alter- 2005; http://www.hightechdecisions.com/), 51 directors of
native to the traditional stainless-steel bioreactor. Cells are biomanufacturing at pharmaceutical and biotechnology com-
cultured in pre-sterilized plastic bags (called Cellbags® ), panies worldwide were surveyed. More than half of these
which are single-use. The bags are filled with medium and directors are either using, or are interested in using, dis-
cells and inflated to form a gas-permeable chamber. It is then posable bags in-house or through a contract manufactur-
placed on a rocking platform, which allows for excellent ing organization (http://www.contractpharma.com/articles/
oxygenation and mixing with low shear stress. The largest 2005/06/disposable-bioprocessing.php). Finally, choosing an
size currently available is 500 litres. Bag sizes of 1500– appropriate production method for any biologic will be of
2500 litres are becoming available, and HyClone reports paramount importance for its commercial success.
offering a 10 000-litre bag through its HyNetics option. The
performance of the Wave bioreactor compares favourably
with conventional cultivation vessels such as stirred-tank
References
bioreactors and roller cultures with respect to simplicity of
operation and cost [93]. They have been successfully used to 1 Anscomb, A. (2004) in Kalorama Information Market
produce adeno-associated viruses from baculovirus/insect Intelligence (Heffner, S., ed.), Kalorama Information, New York
cell culture [94], and adenovirus vectors using PER.C6 cells 2 Sinclair, A. (2004) in Advances in Large-scale Biopharma-
[95,96]. ceutical Manufacturing and Scale-up Production (Langer, E. S.,
ed.), ASM Press and the Institute for Science and Technology
Management, Washington, DC
Conclusion 3 Butler, M. (1987) Adv. Biochem. Eng. Biotechnol. 34,
57–84
During the past 40 years, several strategies have been 4 Hu, W. S. and Peshwa, M. V. (1991) Can. J. Chem. Eng. 69,
formulated to produce biopharmaceutical products from 409–420
animal cells. As the demand for such products increases over 5 van Wezel, A. L. (1967) Nature (London) 216, 64–65
the coming years, the efficiency of the production methods 6 Nikolai, T. J. and Hu, W. S. (1992) Enzyme Microb. Technol.
will become even more important. The STR and the airlift 14, 203–208
reactor have become industry standards, and, owing to their 7 Onderwater, R. C., Goeptar, A. R., Levering, P. R., Vos, R. M.,
ease of operation and ease of licensing, homogeneous reac- Konings, P. N., Doehmer, J., Commandeur, J. N. and Vermeulen,
tors will almost certainly remain at the forefront of animal N. P. (1996) Protein Expression Purif. 8, 439–446
cell biotechnology. Furthermore, cell retention systems are 8 Warnock, J. N. and Al Rubeai, M. (2005) in Applications of
now enabling STRs to run in perfusion mode for ex- Cell Immobilisation Biotechnology (Nedovic, V. and Willaert,
tended duration, which increases their efficiency relative R., eds.), pp. 423–438, Springer, Berlin
to either batch or fed-batch cultures, which is the current 9 Berry, J. M., Barnabe, N., Coombs, K. M. and Butler, M. (1999)
industry standard. Of all the separation techniques currently Biotechnol. Bioeng. 62, 12–19
available, the acoustic filter and the ATF filter have the best 10 Chu, L. and Robinson, D. K. (2001) Curr. Opin. Biotechnol. 12,
potential, although the power requirements for large-scale 180–187
acoustics filters are an issue. The recent interest in these 11 Bailey, J. E. and Ollis, D. F. (1986) Biochemical Engineering
devices also suggests that they may be set to become an Fundamentals, 2nd edn, McGraw-Hill, Jurong, Singapore
industrial standard. Fixed and fluidized-bed systems have 12 Oh, S. K., Nienow, A. W., Al Rubeai, M. and Emery, A. N.
been used as high-density methods of culturing anchorage- (1992) J. Biotechnol. 22, 245–270
dependent cells. Results for CellCube disposable reactors 13 Al Rubeai, M., Emery, A. N., Chalder, S. and Goldman, M. H.
and packed-bed reactors have shown great promise, but (1993) J. Biotechnol. 31, 161–177
their suitability for commercial-scale production has been 14 Kioukia, N., Nienow, A. W., Emery, A. N. and Al Rubeai, M.
brought into question. Therefore fluidized-bed bioreactors (1992) Trans. Inst. Chem. Eng. 70C, 143–148
may be a better option. The Wave bioreactor is currently 15 Hu, Y. C., Kaufman, J., Cho, M. W., Golding, H. and Shiloach, J.
attracting a lot of attention, but there is a lack of published (2000) Biotechnol. Prog. 16, 744–750
information about its performance owing to its newness. 16 Kennard, M. L. and Piret, J. M. (1994) Biotechnol. Bioeng. 44,
Initial studies do suggest that it has great potential for the 45–54
Good-Manufacturing-Practice manufacture of biopharma- 17 Yamaji, H. and Fukuda, H. (1994) Appl. Microbiol. Biotechnol.
ceutical products. In the latest report by HighTech Business 42, 531–535


C 2006 Portland Press Ltd
Bioreactors for cell culture 11

18 Kennard, M. L. and Piret, J. M. (1995) Biotechnol. Bioeng. 47, 43 Williams, S. N. O., Callies, R. M. and Brindle, K. M. (1997)
550–556 Biotechnol. Bioeng. 56, 56–61
19 Nikolai, T. J. and Hu, W. S. (1992) Enzyme Microb. Technol. 44 Koska, J., Bowen, B. D. and Piret, J. M. (1997) Chem. Eng. Sci.
14, 203–208 52, 2251–2263
20 Schweikart, F., Jones, R., Jaton, J.-C. and Hughes, G. J. (1999) 45 Fassnacht, D. and Portner, R. (1999) J. Biotechnol. 72, 169–184
J. Biotechnol. 69, 191–201 46 Glacken, M. W., Fleischaker, R. J. and Sinskey, A. J. (1983)
21 Xiao, C., Huang, Z., Li, W., Hu, X., Qu, W., Gao, L. and Liu, G. Ann. N.Y. Acad. Sci. 413, 355–373
(1999) Cytotechnology 30, 143–147 47 Knight, P. (1989) Bio/Technology 7, 459–461
22 Yamaji, H. and Fukuda, H. (1997) J. Ferment. Bioeng. 83, 48 Bratch, K. and Al Rubeai, M. (2001) Biotechnol. Lett. 23,
489–491 137–141
23 Cosgrove, L., Lovrecz, G. O., Verkuylen, A., Cavaleri, L., Black, 49 Shirgaonkar, I. Z., Lanthier, S. and Kamen, A. (2004) Biotechnol.
L. A., Bentley, J. D., Howlett, G. J., Gray, P. P., Ward, C. W. and Adv. 22, 433–444
McKern, N. M. (1995) Protein Expression Purif. 6, 789–798 50 Dalm, M. C., Jansen, M., Keijzer, T. M., van Grunsven, W. M.,
24 Crowley, J., Marsden, W. L. and Gray, P. P. (1991) in Animal Cell Oudshoorn, A., Tramper, J. and Martens, D. E. (2005)
Culture and Production of Biologicals (Sasaki, R. and Ikura, H., Biotechnol. Bioeng. 91, 894–900
eds.), pp. 275–281, Kluwer Academic, Dordrecht 51 Pui, P. W., Trampler, F., Sonderhoff, S. A., Groeschl, M., Kilburn,
25 Hesse, F., Ebel, M., Konisch, N., Sterlinski, R., Kessler, W. and D. G. and Piret, J. M. (1995) Biotechnol. Prog. 11, 146–152
Wagner, R. (2003) Biotechnol. Prog. 19, 833–843 52 Trampler, F., Sonderhoff, S. A., Pui, P. W., Kilburn, D. G. and
26 Merchuk, J. C. (2003) Can. J. Chem. Eng. 81, 324–337 Piret, J. M. (1994) Bio/Technology 12, 281–284
27 Kadouri, A. and Spier, R. E. (1997) Cytotechnology 24, 89–98 53 Coakley, W. T., Whitworth, G., Grundy, M. A., Gould, R. K. and
28 Voisard, D., Meuwly, F., Ruffieux, P. A., Baer, G. and Kadouri, A. Allman, R. (1994) Bioseparation 4, 73–83
(2003) Biotechnol. Bioeng. 82, 751–765 54 Gorenflo, V. M., Smith, L., Dedinsky, B., Persson, B. and Piret,
29 Deo, Y. M., Mahadevan, M. D. and Fuchs, R. (1996) J. M. (2002) Biotechnol. Bioeng. 80, 438–444
Biotechnol. Prog. 12, 57–64 55 Dalm, M. C., Cuijten, S. M., van Grunsven, W. M., Tramper,
30 Iding, K., Lütkmeyer, D., Fraune, E., Gerlach, K. and Lehmann, J. J. and Martens, D. E. (2004) Biotechnol. Bioeng. 88,
(2000) Cytotechnology 34, 141–151 547–557
31 van Wezel, A. L., van der Velden-de Groot, C. A. M., de 56 Gorenflo, V. M., Angepat, S., Bowen, B. D. and Piret, J. M.
Haan, H. H., van den Heuvan, N. and Schasfoort, R. (1985) (2003) Biotechnol. Prog. 19, 30–36
Dev. Biol. Stand. 60, 229–236 57 Merten, O. W. (2000) Cytotechnology 34, 175–179
32 Avgerinos, G. C., Drapeau, D., Socolow, J. S., Mao, J. I., Hsiao, 58 Dalm, M. C., Jansen, M., Keijzer, T. M., van Grunsven, W. M.,
K. and Broeze, R. J. (1990) Bio/Technology 8, 54–58 Oudshoorn, A., Tramper, J. and Martens, D. E. (2005)
33 McLimans, W. F., Kwasniewski, B., Robinson, F. O., Chu, T. M., Biotechnol. Bioeng. 91, 894–900
Sufrin, G. and Gailani, S. (1977) Cancer Treat. Rep. 61, 59 de la Broise, D., Noiseux, M. and Lemieux, R. (1991)
161–165 Biotechnol. Bioeng. 38, 781–787
34 Jan, D. C., Emery, A. N. and Al Rubeai, M. (1993) Biotechnol. 60 de la Broise, D., Noiseux, M., Massie, B. and Lemieux, R. (1992)
Tech. 7, 351–356 Biotechnol. Bioeng. 40, 25–32
35 Bierau, H., Perani, A., Al Rubeai, M. and Emery, A. N. (1998) 61 Kawahara, H., Mitsuda, S., Kumazawa, E. and Takeshita, Y.
J. Biotechnol. 62, 195–207 (1994) Cytotechnology 24, 89–98
36 Jan, D. C., Jones, S. J., Emery, A. N. and Al Rubeai, M. (1994) 62 Bleckwenn, N. A., Golding, H., Bentley, W. E. and Shiloach, J.
Cytotechnology 16, 17–26 (2005) Biotechnol. Bioeng. 90, 663–674
37 Amos, B., Al Rubeai, M. and Emery, A. N. (1994) Enzyme 63 Kadouri, A. and Zipori, D. (1989) in Advances in Animal Cell
Microb. Technol. 16, 688–695 Biology and Technology for Bioprocesses (Spier, R. E., Griffiths,
38 Adamson, S., Fitzpatrick, S., Behie, L. A., Gaucher, G. and J. B., Stephenne, J. and Grooy, P. J., eds.), pp. 327–330, Courier
Lesser, B. (1983) Biotechnol. Lett. 5, 573–578 International, Tiptree, Essex, U.K.
39 Sjorgren-Jansson, E. and Jeansson, S. (1985) J. Immunol. 64 Kang, S.-H., Lee, G. M. and Kim, B.-G. (2000) Cytotechnology
Methods 84, 359–364 34, 151–158
40 Comer, M. J., Kearns, M. J., Wahl, J., Munster, M., Lorenz, T., 65 Kaufman, J., Wang, G., Zhang, W., Valle, M. A. and Shiloach, J.
Szperalski, B., Koch, S., Behrendt, U. and Brunner, H. (1990) (2000) Cytotechnology 33, 3–11
Cytotechnology 3, 295–299 66 Merten, O. W., Cruz, P. E., Rochette, C., Geny-Fiamma, C.,
41 Kessler, N., Thomas, G., Gerentes, L., Delfosse, G. and Aymard, Bouquet, C., Goncalves, D., Danos, O. and Carrondo, M. J.
M. (1997) Cytotechnology 24, 109–119 (2001) Biotechnol. Prog. 17, 326–335
42 Thelwell, P. E. and Brindle, K. M. (1999) Cytotechnology 30, 67 Wang, G., Zhang, W., Jacklin, C., Freedman, D., Eppstein, L.
121–132 and Kadouri, A. (1992) Cytotechnology 9, 41–49


C 2006 Portland Press Ltd
12 J. N. Warnock and M. Al-Rubeai

68 Park, S. and Stephanopoulos, G. (1993) Biotechnol. Bioeng. 85 Warnock, J. N., Price, T., Slade, A. and Al Rubeai, M. (2004)
41, 25–34 Bioprocess. J. 3, 41–45
69 Chiou, T. W., Murakami, S. and Wang, D. I. C. (1991) 86 Kotani, H., Newton, III, P. B., Zhang, S., Chiang, Y. L., Otto, E.,
Biotechnol. Bioeng. 37, 755–761 Weaver, L., Blaese, R. M., Anderson, W. F. and McGarrity, G. J.
70 Park, S. and Stephanopoulos, G. (1993) Biotechnol. Bioeng. (1994) Hum. Gene Ther. 5, 19–28
41, 25–34 87 Wikström, K., Blomberg, P. and Islam, K. B. (2004)
71 McTaggart, S. and Al Rubeai, M. (2000) Biotechnol. Prog. 16, Biotechnol. Prog. 20, 1198–1203
859–865 88 Merten, O. W. (2004) J. Gene Med. 6 (Suppl. 1), S105–S124
72 Warnock, J. N. (2002) Ph.D. Thesis, University of Birmingham 89 Davis-Sproul, J. M., Harris, M. P., Davidson, N. E., Kobrin, B. J.,
73 Kafri, T. (2001) Curr. Opin. Mol. Ther. 3, 316–326 Jaffee, E. M. and Emens, L. A. (2005) Cytotherapy 7, 46–56
74 Shi, Y., Ryu, D. D. Y. and Park, S. (1992) Biotechnol. Bioeng. 40, 90 Ho, L., Greene, C. L., Schmidt, A. W. and Huang, L. H. (2004)
260–270 Cytotechnology 45, 117–123
75 Rodrigues, M. T. A., Vilaça, P. R., Garbuio, A., Takagi, M., Barbosa, 91 Ni, Y. J., Sun, S. S., Oparaocha, T., Humeau, L., Davis, B.,
Jr, S., Léo, P., Laignier, N. S., Silva, A. A. P. and Moro, A. M. (1999) Cohen, R., Binder, G., Chang, Y. N., Slepushkin, V. and Dropulic,
Bioprocess Eng. 21, 543–546 B. (2005) J. Gene Med. 7, 818–834
76 Portner, R., Bohmann, A., Ludemann, I. and Markl, H. (1994) 92 Tuyaerts, S., Noppe, S. M., Corthals, J., Breckpot, K., Heirman,
J. Biotechnol. 34, 237–246 C., De Greef, C., Van Riet, I. and Thielemans, K. (2002) J.
77 Kratje, R. B. and Wagner, R. (1992) Biotechnol. Bioeng. 39, Immunol. Methods 264, 135–151
233–242 93 Weber, W., Weber, E., Geisse, S. and Memmert, K. (2002)
78 Preissmann, A., Wiesmann, R., Buchholz, R., Werner, R. G. and Cytotechnology 38, 77–85
Noe, W. (1997) Cytotechnology 24, 121–134 94 Chen, H. F., Zhou, S. Z., Pierce, G. P. and Colosi, P. (2004)
79 Kratje, R. B., Reimann, A., Hammer, J. and Wagner, R. (1994) Mol. Ther. 9, S160–S160
Biotechnol. Prog. 10, 410–420 95 Ekstrom, D., Cheng, W., Andersson, R., Mitra, G. and Zhu, J. W.
80 Frame, K. K. and Hu, W. S. (1990) Biotechnol. Bioeng. 35, (2004) Abstracts of Papers of the American Chemical Society
469–476 227, U202
81 Leelavatcharamas, V., Emery, A. N. and Al Rubeai, M. (1994) 96 Mikola, M. R., Burden, E. C., Jug-Dujakovic, M., Pearre, C. L.,
Cytotechnology 15, 65–71 Herber, W. K. and Amanullah, A. (2004) Abstracts of Papers
82 Goldman, M. H., James, D. C., Rendall, M., Ison, A. P., of the American Chemical Society 227, U222
Hoare, M. and Bull, A. T. (1998) Biotechnol. Bioeng. 60, 97 Singh, R. P., Al Rubeai, M., Gregory, C. D. and Emery, A. N.
596–607 (1994) Biotechnol. Bioeng. 44, 720–726
83 Wang, M. D., Yang, M., Huzel, N. and Butler, M. (2002)
Biotechnol. Bioeng. 77, 194–203 Received 6 December 2005/21 March 2006; accepted 28 March 2006
84 Kong, D., Cardak, S., Chen, M., Gentz, R. and Zhang, J. (1999) Published on the Internet 20 June 2006, doi:10.1042/BA20050233
Cytotechnology 29, 215–220


C 2006 Portland Press Ltd

View publication stats

You might also like