Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Review

Recent advances in large-scale


production of monoclonal antibodies
and related proteins
Abhinav A. Shukla1 and Jörg Thömmes2
1
Manufacturing Sciences & Technology, Bristol-Myers Squibb, Co., 6000 Thompson Road, East Syracuse, NY 13057
2
Technical Development, Biogen Idec, 5200 Research Place, San Diego, CA 92122

The rapid development of high-yielding and robust man- variants) and non-product-related (e.g. host cell proteins)
ufacturing processes for monoclonal antibodies is an impurities to acceptable levels. Batch chromatography,
area of significant focus in the biopharmaceutical land- which is traditionally used as the core purification method
scape. Advances in mammalian cell culture have taken in industrial bioprocesses, has long been labeled as a slow
titers to beyond the 5 g/l mark. Platform approaches to process that requires large volumes of mobile phase and
downstream process development have become widely results in very large pool volumes. Recent advances in the
established. Continuous evolution of these platforms is design of chromatographic stationary phases available for
occurring as experience with a wider range of products is purification of biomolecules have provided some solutions to
accrued. The increased cell culture productivity has the throughput dilemma, and have facilitated reasonably
shifted the attention of bioprocess development to oper- productive platforms for industrial protein purification. As
ations downstream of the production bioreactor. This multiple biopharmaceutical companies have become
has rejuvenated interest in the use of non-chromato- increasingly involved with the development of mAb pro-
graphic separation processes. Here, we review the cur- ducts, platform approaches to their production have become
rent state-of-the-art industrial production processes, well established in industry. These platforms leverage sim-
focusing on downstream technologies, for antibodies ilarities in the biochemical properties and chromatographic
and antibody-related products and discuss future ave- behavior of this product class as well as experience gained
nues for evolution. from the first mAbs and Fc fusion proteins that came on the
market. The platforms have enabled significant efficiencies
Large-scale production of monoclonal antibodies
Monoclonal antibodies (mAbs) and related proteins have
Glossary
been shown to successfully target a wide range of extra-
cellular targets with high specificity. This has led to their Capture step: the first downstream processing step that captures the product
from the harvested cell broth, concentrates the product, and achieves
introduction to a variety of disease therapies, including separation from impurities that are most unlike the product (e.g. cells, cell
several forms of cancer, multiple sclerosis and immuno- debris, DNA, most proteins).
logical disorders such as rheumatoid arthritis and psor- Cell specific productivity (Qp): productivity of a cell culture expressed in terms
of amount of protein produced per cell per unit time. A conventional unit for
iasis (Table 1). Significant advances have been made over mammalian cell culture is pg/cell/day.
the years in the design of mAbs as therapeutics that have Downstream process: process steps associated with the purification of a
improved bioavailability, optimized affinity, improved recombinant protein and removal of impurities.
Fed-batch cell culture: a production process based upon feeding a growth
binding specificity, and human antibody sequences to limiting nutrient to the culture. This allows the achievement of a high cell
reduce any immunogenic side effects [1]. Further advances density in the production bioreactor and facilitates metabolic control of the
in protein engineering are improving the therapeutic pro- cells to avoid generation of side products.
Follow-on biologics (also known as ‘biosimilars’): biopharmaceuticals that are
files of mAbs [2–5]. Several cases for which mAb thera- deemed comparable in quality, safety and efficacy to a reference product from
peutics have been approved serve a large patient an innovator company.
population and involve chronic therapy with high doses. NGNA: N-glycolylneuraminic acid that can form the terminal saccharide unit
for an N-linked oligosaccharide structure. Presence of NGNA has been linked to
This has led to the need for production processes that can possible immunogenicity. The usual terminal sugar unit is typically NANA (N-
be developed rapidly to produce consistently and reprodu- acetylneuraminic acid).
Polishing steps: steps that occur after the initial capture step that are aimed at
cibly large quantities of pharmaceutical-grade mAbs at
removal of smaller levels of impurities left in the product stream that have
moderate costs. Process platforms in both upstream cell more similarity to the product (e.g. aggregated forms of the product, protein
culture and downstream purification have become widely structural and sequence variants).
Sanitization: procedures that destroy contaminating microorganisms on a
established in industry to meet this need. piece of equipment used for bioprocessing. For chromatographic columns,
Large-scale production of mAbs utilizes mammalian pro- mild alkaline solutions or high concentrations of chaotropes (e.g. urea) are
duction systems followed by cell removal and purification used for sanitization to achieve bioburden control while preserving the resin.
Transient transfection: process of introducing DNA into a host cell by non-viral
through sequential chromatographic and membrane means such that the genetic material is not inserted into the nuclear genome.
filtration steps, to consistently reduce product- (e.g. protein As a result, the genetic material is lost over time through mitosis.
Upstream process: process steps associated with the production of a
recombinant protein by culture and propagation of the host cells.
Corresponding author: Shukla, A.A. (abhinav.shukla@bms.com).

0167-7799/$ – see front matter ß 2010 Elsevier Ltd. All rights reserved. doi:10.1016/j.tibtech.2010.02.001 Available online 19 March 2010 253
Review Trends in Biotechnology Vol.28 No.5

Table 1. Monoclonal antibodies and related proteins on the market


Name Target Indication Company Year Antibody type
Monoclonal antibodies
Orthoclone OKT3 CD3 Acute kidney transplant rejection Ortho Biotech 1986 Murine
ReoPro Platelet GP IIb/IIIa Prevention of blood clot Centocor 1994 Murine
Rituxan CD20 Non-Hodgkin’s Lymphoma Genentech/ Biogen-Idec 1997 Chimeric
Panorex 17A-1 Colorectal cancer GlaxoSmithKline 1995 Murine
Zenapax IL2Ra (CD25) Acute kidney transplant rejection Hoffman-LaRoche 1997 Humanized
Simulect IL2R Prophylaxis of acute organ rejection Novartis 1998 Chimeric
Synagis RSV Respiratory Synctial Virus Medimmune 1998 Humanized
Remicade TNFa Rheumatoid arthritis Centocor 1998 Chimeric
Herceptin Her2 Metastatic breast cancer Genentech 1998 Humanized
Mylotarg CD33 Acute mylogenous lymphoma Wyeth-Ayerst 2000 Humanixed
Campath CD52 B cell chronic lymphocytic leukemia Takeda 2001 Humanized
Zevalin CD20 Non-Hodgkins Lymphoma Biogen-Idec 2002 Murine
Humira TNFa Rheumatoid arthritis Abbott 2002 Human
Bexxar CD20 Non-Hodgkins Lymphoma Corixa/GSK 2003 Murine
Xolair IgE Allergy Genentech/Novartis 2003 Humanized
Erbitux EGFR/Her1 Colorectal cancer Bristol-Myers 2004 Humanized
Squibb/Imclone (Eli Lilly)
Avastin VEGF Colorectal cancer Genentech 2004 Humanized
Raptiva CD11a Psoriasis Genentech/Xoma 2004 Humanized
Tysabri A4 integrin Multiple sclerosis Biogen-Idec/Elan 2004 Humanized
Vectibix EGFR Colorectal cancer Amgen 2006 Human
Soliris C5 complement PNH – paroxysmal nocturnal Alexion 2007 Humanized
hemoglobinuria
Stelara IL12 and IL23 Psoriasis Centocor 2008 Human
Simponi TNFa Rheumatoid arthritis Centocor 2008 Human
Actemra IL-6 Rheumatoid arthritis Roche 2009 Humanized
Monoclonal antibody fragments
Lucentis VEGF-A Age related macular degeneration Genentech 2006 Fab’
Cimzia TNFa Crohn’s disease UCB 2008 Pegylated Fab fragment
Fc fusion proteins
Enbrel TNFa Rheumatoid arthritis, psoriasis, Amgen 1998 Soluble TNFa receptor
ankylosing spondilitis fused to IgG1 Fc
Amevive CD 2 Psoriasis Biogen-Idec 2003 LFA3 fused to IgG1 Fc
Orencia CD80/86 Rheumatoid arthritis Bristol-Myers Squibb 2005 CTLA4 fused to IgG1 Fc
Arcalyst IL-1 CAPS – Cryopyrin Associated Regeneron 2007 IL-1 receptor fused to Fc
Periodic Syndrome

in process development from time and resource perspect- a templated process; indeed, this has proven difficult even
ives. Allied areas in biopharmaceutical enterprises, such as within the same company because of differences in proper-
quality and manufacturing, have also benefited through ties and purification behavior between various mAbs [6].
templating their documents and procedures. A few publi- What the platform achieves is the creation of a common
cations have provided details on the process platforms in use philosophy and alignment over the types of unit operations
at various companies [6–8], and further details have been to include in the downstream process. Figure 2 shows some
shared at conferences. The sections below provide details on of the downstream process platform schemes adopted at
current platforms and review recent developments that are biopharmaceutical companies involved in mAb process
leading to their evolution. The use of these production plat- development. The schematic diagram for a typical down-
forms has enabled the rapid and efficient introduction of stream process, including other filtration and viral-clear-
large numbers of mAbs into clinical trials. The process ance-oriented process steps, is depicted in Figure 3.
platform continues to evolve as further experience is gained All of these schemes rely on the use of Protein A affinity
with a wider set of mAbs. chromatography as the capture step in the process. This
mode of affinity chromatography is based on the specific
Downstream process platform for mAb production binding affinity between the Fc region of mAbs and the
Upstream process platforms in terms of cell line, clone Protein A ligand. This specificity allows host cell proteins,
selection methodologies, fed-batch cell culture operating DNA and other impurities from the cell culture process to
conditions and media have evolved significantly in the last flow through while the product binds to the stationary
decade. While these two sections focus on downstream phase, providing >98% purity in a single step. Elution
process platforms, Box 1 provides details about process from Protein A affinity adsorbents is effective under con-
operating conditions for large-scale cell culture production ditions of low pH. In many ways, the high specificity of the
of mAbs. Protein A step has facilitated the adoption of a platform
The downstream purification process has evolved approach for downstream purification of mAbs. The polish-
toward a common framework across industry in recent ing steps in the downstream process aim to reduce process-
years [6–8,28,29]. The platform approach does not imply and product-related impurities, particularly host cell

254
Review Trends in Biotechnology Vol.28 No.5

Box 1. Upstream process platform


The productivity of fed-batch mammalian cell culture processes for generation through the development of stable production cell lines,
mAbs has increased significantly in recent years. Two key factors the use of transient transfectant pools for production is frequently
have underpinned the increase [9]: (i) highly productive cell lines that carried out during pre-clinical developmental stages, to produce
have both the right growth characteristics and a high specific rapidly significant quantities of the product [18,19], but is currently
productivity Qp; and (ii) improved understanding of chemically rarely employed at scales beyond a few hundred liters of cell culture
defined media and feeding strategies to achieve high cell density volume. It is greatly preferred to lock in the most desirable subset of
and sustained viability over the course of the bioreactor run. Figure 1 clones for biopharmaceutical production early in the clinical devel-
is a schematic diagram for an upstream production process that is opment process for a product. This avoids potential changes in
often used for the production of mAbs and other glycoproteins. glycosylation profile later in development, which can create compar-
The creation of a rapid-growth and high-specific-productivity (Qp) ability hurdles if a switch to an alternative cell line is undertaken.
production cell line sets the right foundation for developing the cell In large-scale cell culture processes, the early stages of inoculum
culture production process. Additional requirements include accep- growth after vial thaw are conducted in shake flasks or spinner flasks,
table cell line stability and the ability to grow in chemically defined which progressively increase in size and/or volume. Wave bioreactors
media [10]. The use of effective expression systems [9,10] is a pre- in disposable bags are used extensively at this stage of the process.
requisite to creating a high-producing cell line. Commonly utilized The cell mass is then scaled up through several bioreactor stages
selection markers within the stable cell line development vectors prior to transfer to the production bioreactor. Fed-batch production is
include those based on genes that encode dihydroxy folate reductase the most prevalent and involves the addition of small volumes of feed
and glutamine synthetase. These expression vectors also typically to supplement the nutrients present in the bioreactor as cell growth
utilize strong gene promoters that drive product mRNA transcription, and product production progresses. Dissolved oxygen, pH, tempera-
such as cytomegalovirus or EF-1a, whereas expression of the gene ture and mass transfer of oxygen and CO2 are controlled in the
that encodes the selectable marker is often driven through a weak production bioreactor. Perfusion cell culture, in which the cells are
promoter to maximize selection stringency. A high level of product retained in the bioreactor and new media fed continuously is
synthesis in Chinese hamster ovary (CHO) cells has been linked to employed in some processes, but is less widely adopted because of
increased gene copy number and effective transcription [11]. Loss in challenges with maintaining sterility over long time periods. A key
stability of the cell line in the absence of selective pressure has been development in the achievement of high cell culture titers has been an
linked to decreased transcription rather than loss of copy number, improved understanding of nutrient limitations during cell culture
which is often indicted for instability [10]. Although the complete and the design of chemically defined, serum-free media that can
molecular mechanisms are incompletely understood, transgene support cell growth and product secretion. Hydrolysates from yeast or
silencing has been implicated in the decreased productivity. Cell plant sources are often a component of cell culture media and have
lines used for antibody expression include the most commonly used effectively replaced the use of serum in large-scale processes. Amino
CHO cell lines, murine lymphoid cell lines (e.g. NS0 and SP2/0), acids and trace media constituents such as metals have been shown
human PER.C6, and often murine hybridoma cell lines [12,13]. All of to influence product yields in cell culture [20,21]. Bioreactor processes
these cell lines have been shown to produce product glycosylation also have been developed to optimize gas exchange, to supply
patterns that are compatible with the human immune system, sufficient oxygen to sustain cell growth and productivity, and to
although there have been reports of high NGNA levels in NS0- remove CO2. Maintaining the efficiency of gas exchange is a very
derived expression systems [14]. Cells have also been engineered to important criterion for ensuring the success of scaling up cell culture
increase product expression through coexpression of important platforms [22].
proteins in the secretory pathway [15]. A number of coexpression mAbs are glycosylated products typically with a single N-linked site
strategies have been researched including coexpression of chaper- on each half of the Fc region. Even though the N-linked glycosylation
ones, protein degradation inhibitors and apoptosis inhibitors. Obtain- heterogeneity does not significantly influence pharmacokinetics or
ing more complete knowledge of the CHO proteome will certainly aid half-life of mAbs, because that is predominantly dictated by binding
coexpression efforts to obtain a more productive cell line. to the neonatal Fc receptor (FcRn binding [23]), N-glycosylation does
Production cell lines are screened during their development to play a role in mediating antibody-dependent cellular cytotoxicity or
select for those with a high specific productivity (Qp). However, not all complement-dependent cytotoxicity. Additionally, it is important to
cell lines with a high Qp (generally 20–90 pg/cell/day [16]) may prevent the occurrence of glycosylation structures that can trigger a
perform well in a production process. As a result of this and other human immune response such as galactose a(1,3)-galactose and
factors, including stability profiles and desirable product quality NGNA structures [24]. As cell cultures under suboptimal conditions
metrics, large numbers of clones need to be generated and screened have been shown to produce aberrant oligosaccharide structures
to arrive at the best combination of properties that are most suitable [20,21,25], it is important to link media and bioreactor condition
for biopharmaceutical production. Several automated systems for cell optimization to evaluation of glycosylation quality during cell culture
line screening are in use today to facilitate the identification of the design.
most suitable production line. Fluorescence-activated cell sorting is A typical harvest procedure for mAb cell culture utilizes centrifuga-
one of the techniques used to screen cell lines with high levels of tion followed by depth and membrane filters to remove cells and cell
antibody production from within a heterogeneous population during debris prior to the purification steps in the downstream process [26].
selection. A clone screening technique that utilizes relative mRNA Depth filters consist of a fibrous bed that can trap particles within their
transcript levels to screen predictively for high expressers, using the volume in contrast to membrane filters that reject particulates.
ratio of light to heavy chain mRNA levels as a predictor of aggregated Recently, depth filters have been shown to possess adsorptive
product levels, has been reported to help identify clones most properties for soluble impurities such as host cell proteins and
suitable for antibody production [17]. To monitor clone growth DNA, in addition to their filtration role in removing particulates [27].
characteristics, some level of screening is typically carried out in As a result, the type of depth filter and the choice of operating
shake flasks or even small bioreactors, in addition to early high- conditions need to be studied in an integrative fashion with the
throughput screening in 96-well plates [17]. In addition to material downstream purification process.

proteins, DNA and high-molecular-weight aggregates to filtration (UF/DF) operation to reduce storage volumes and
acceptable levels in the drug substance [30]. To assure viral exchange the product into the formulation buffer to pro-
clearance, the downstream process also includes at least duce the drug substance.
two dedicated, orthogonal steps for viral inactivation and Alternatives to Protein A capture have been an area of
size-based viral removal by viral filtration. The last step in active investigation. As with any other protein, it is
the downstream process is typically an ultrafiltration/dia- possible to design a sequence of purification steps for mAbs

255
Review Trends in Biotechnology Vol.28 No.5

Figure 1. mAb upstream process. The process starts with vial thaw and expansion of cells through a series of inoculum steps. The cells are then further expanded in a series
of seed bioreactors before transfer to the production bioreactor where the mAb is expressed into the medium. Centrifugation and a series of filtration steps are then used to
harvest the cell culture broth from cells and cell debris.

without the use of an affinity step [31,32]. However, these This ligand also has improved the homogeneity of antibody
approaches are more difficult to template. In the past, interactions with Protein A, thus facilitating the use of a
significant attention has focused on the development of generic set of operating buffers for the Protein A step [37].
small-molecule ligands that could offer similar selectivity
to Protein A. However, comprehensive evaluation of hydro- Recent advances in Protein A capture chromatography
phobic charge induction chromatography (HCIC) [33] and Given the key significance of Protein A chromatography,
protein A mimetic ligands [34], using multiple antibody recent efforts have focused on improving its understanding
process streams, has indicated markedly lower selectivity and mitigating some of the limitations it poses for large-scale
than the Protein A ligand. Although the success of non- mAb production. Process throughput is constrained by the
proteinaceous ligands in replacing Protein A chromatog- maximum loading possible on Protein A; therefore, it is
raphy has been low, these alternative modes of chromatog- important to understand why binding is limited. Using a
raphy are being employed as polishing steps in antibody panel of mAbs and Fc fusion proteins, it has been determined
purification. Alternatively, other affinity ligands such as that multiple product molecules can bind to the Protein A
single-chain antibodies, against the Fc region, derived from ligand. Inter- and intra-ligand steric effects have been shown
camelid species have been shown to offer promising selec- to limit the amount of mAb that can bind per unit volume of
tivity for mAbs [35,36]. Improvements in the Protein A the resin [38]. Dynamic binding capacity on the Protein A
ligand itself have also occurred, with one of the recent affinity step is significantly influenced by flow rate and
improvements being the development of a base-stable ligand residence time during loading [39]. The capture step, which
that can resist alkaline conditions, which are often preferred involves the loading of relatively dilute cell culture harvest
for sanitization of large-scale chromatography columns. on the resin, is generally throughput-limited. To address this

Box 2. Non-chromatographic technologies aimed at improving downstream process throughput


Renewed interest in non-chromatographic separation has been caprylic acid or poly-ether sulfone. Some efforts have focused on
sparked by some of the recent discussions about process throughput removing impurities by flocculation or precipitation. Promising
limitations faced by batch chromatography. A number of such avenues for future research include the use of selective polymers
techniques have been investigated ever since the advent of protein for product precipitation. Modified PEG molecules have been used for
production on a large scale, but have largely not been adopted for selective capture of mAbs from CHO cell supernatants. Crystallization
therapeutic protein production. Aqueous two-phase separations can result in highly pure proteins and is often used for smaller
using polymer–salt [poly(ethylene glycol) (PEG) phosphate and PEG proteins and peptides. Although this area elicited some early interest
citrate] systems have been applied successfully for purification of for mAb production, it is now realized that the phase behavior of
mAbs from cell culture harvest streams [65]. Expanded bed chroma- mAbs makes this a less robust process, with relatively narrow
tography has been left behind during the past decade, despite operating ranges [70]. Charged UF membranes can provide separa-
promise as an integrative technology between cell culture and tion based on size (hydrodynamic radius is strongly influenced by pH
purification. Recently, interest has returned through the introduction and ionic strength) and charge [71,72]. Membrane adsorbers are
of improved resins and flow distribution systems that allow large- already a reality in the large-scale downstream processing of some
scale implementation [66]. Precipitation of proteins using salts has antibodies [73]. Anion exchange membrane adsorbers provide high
long been a mainstay of laboratory-level protein isolation. It has been capacity and throughput but are effective for minimal polishing only
realized that application to mAb production will require improvement (DNA removal, some host cell protein clearance, and for providing
in the selectivity of this technique [67–69]. Efforts focus on precipitat- additional viral clearance capability). Ability to bind to large quantities
ing the product using negatively charged polyelectrolytes such as of aggregates or host cell protein is minimal.

256
Review Trends in Biotechnology Vol.28 No.5

Figure 2. Downstream process platforms in use in several biopharmaceutical companies (a)–(c). The typical downstream platform starts with mAb capture by Protein A
chromatography. The commonly used platforms differ in the number and type of polishing steps. AEX, anion exchange; CEX, cation exchange; HIC, hydrophobic
interaction chromatography.

issue, when ample binding sites are available, a modulated acetic acid and benzyl alcohol [42] have been found to be
flow rate with rapid initial flow, followed by a progressive effective sanitization solutions that maintain ligand func-
decline in flow rate, has been developed [40]. tionality. Overall, adsorbents based on Protein A have
Another area of concern for large-scale Protein A been shown to be remarkably stable over many hundred
chromatography is the lifetime of the resin, with re-use cycles of use, thus making Protein A chromatography a
over multiple cycles in operation. The Protein A ligand is very reliable tool in the antibody purification platform [43].
known to be sensitive to extreme conditions of pH and can Host cell proteins constitute an important class of pro-
lose its ability to bind to products. The base-stable ligand cess-related contaminants that need to be reduced to low
mentioned earlier is still not in widespread use because of levels to assure product safety. While the Protein A capture
its higher cost. A progressive decline in binding capacity step is highly selective for mAbs over soluble host cell
has been shown to occur upon exposure to the mild alkaline protein impurities, the levels of host cell protein that often
conditions often used for regenerating agarose-based persist after Protein A purification can still pose a clear-
Protein A resins [41]. Protective additives in 0.1 N NaOH ance challenge for non-affinity polishing steps. It has been
[41] or use of a combination solution of phosphoric acid, shown that host cell protein species that persist through

Figure 3. Monoclonal antibody downstream process. The process depicted in this figure starts with Protein A chromatographic capture and includes two subsequent
polishing chromatographic steps for impurity removal. The process also includes two dedicated orthogonal steps for viral clearance: low pH viral inactivation after Protein A
chromatography and viral filtration. The final process step is ultrafiltration/diafiltration (UF/DF) to formulate and concentrate the product.

257
Review Trends in Biotechnology Vol.28 No.5

the Protein A step are often associated with the product retention size) that were employed in the past [53].
species [44]. These protein–protein interactions have been Although the smaller pore size rating provides added viral
found to be disrupted by chaotrope combinations at high retention, these filters can be clogged by small levels of
pH at which mAb–Protein A interactions are still strong. particulates that are otherwise not detectable by analytical
This has allowed the development of selective wash con- size exclusion chromatography (SEC) or particle size
ditions that can reduce the host cell protein levels after measurements in the product stream [54,55]. Viral filters
Protein A chromatography. are typically single-use filters and can be among the most
In addition to a better mechanistic understanding of expensive consumables in the process based on their cost
host cell protein clearance during mAb purification, better per gram of product. In addition to appropriate placement
methodologies have been developed to monitor host cell of these filters in the downstream process, pre-filters can be
protein (HCP) clearance and to validate the capability of used to protect the viral filters from flux decay arising from
the downstream process to clear them. 2D difference gel a high particulate load [56]. Viral filters from multiple
electrophoresis has been used to create a map of host cell vendors are typically screened to arrive at the right selec-
proteins to monitor their clearance through the down- tion for a process, and significant innovation has taken
stream process [45]. Multiple strategies, including worst- place in recent years in the development of low-fouling
case studies, bypass and spiking, have been employed to viral filters that can still provide excellent viral clearance
demonstrate robust clearance of host cell proteins through [57]. These filters are made of hydrophilic materials that
a mAb downstream process [46]. The low-pH conditions reduce the tendency for proteins to unfold at the surface,
used for Protein A elution often result in precipitation thus minimizing nonspecific fouling of the pores.
issues for the product species. These have been shown to Additional improvements have been made in pore
be mitigated by judicious buffer selection or by selecting morphology, with a reduction in polydispersity of pore
stabilizing additives for the elution buffer [47]. In some sizes. These have led to progressive improvement in the
cases, the turbidity has been shown to be caused by the maintenance of high fluxes with mAb process streams.
precipitation of host cell protein contaminants, rather than The large production quantities of mAbs create another
the product. This can still be undesirable from a process challenge for the handling of the large volumes of drug
reproducibility and filtration perspective [48] and has been substance produced. Several ultra-high-concentration
shown to be prevented by appropriate sizing and design of antibody formulations with protein concentrations exceed-
the harvest depth filtration step [27]. ing 100 mg/ml are in use for mAbs, particularly for sub-
cutaneous delivery. The generation of such high protein
Recent advances in unit operations following Protein A concentrations requires special attention to the design of
capture the UF/DF step that is typically used for buffer exchange
As shown in Figure 2, some platforms adopt a completely into the formulation buffer [58]. Air–water interfaces cre-
templated approach to the polishing steps (predominantly ated during pumping of such highly concentrated protein
through the use of anion (AEX) and cation (CEX) exchange solutions can cause product aggregation during large-scale
chromatography), whereas others have a molecule-specific production [59]. The flowpath in the UF system is designed
approach based on the particular impurity clearance chal- to minimize system hold-up volumes and to decrease the
lenge. Hydrophobic interaction chromatography (HIC) is incidence of zones where air entrainment might occur. For
often selected for its particular ability to clear high-mol- high-concentration formulations, the concentration at
ecular-weight aggregates [6], and mixed-mode ion- which DF is conducted is selected to minimize the number
exchange chromatography has found recent application of pump passes to which an average protein molecule
as a result of its unique selectivity [49,50]. Several new might be exposed during the step. Even exposure to stain-
mixed-mode resins have been launched (e.g. Captoadhere1 less steel surfaces for significant lengths of time can result
and CaptoMMC1 from GE Healthcare) and are finding in aggregation [60]. If the drug substance requires frozen
increasing application as polishing steps for host cell conditions during long-term storage, design of the freezing
protein and aggregate clearance. Cation-exchange chroma- and thawing steps needs to be carefully approached to
tography column loading is often carried out at low salt avoid product quality changes caused by cryo-concen-
concentrations. mAbs have a basic pI, and the initial tration and exclusion effects caused by slow freezing rates
molecules that adsorb on the resin are often at the entrance in large volumes, and product denaturation on high-sur-
to pores in the resin structure, thus repelling more protein face-area ice crystals caused by rapid freezing rates [61].
molecules and restricting column load capacities [51]. The The adoption of a platform approach has created greater
anion-exchange column is usually operated under flow- efficiencies in process development and manufacturing for
through conditions at fairly high load capacities. As mAbs. Evolution of the platform through experiences accu-
described later, this step can sometimes be replaced with mulated while dealing with challenges faced with atypical
a membrane adsorber step. If impurity resolution, particu- antibody processes is an equally essential part of creating a
larly HCP and aggregate is needed, the anion-exchange robust and widely applicable platform.
chromatographic step can be operated under a weak par-
titioning mode as opposed to true flow-through conditions, Non-chromatographic downstream processing
which results in retention of the impurities [52]. Chromatographic steps have formed the mainstay of mod-
A majority of recently developed mAb production pro- ern-day biopharmaceutical purification processes because of
cesses employ parvoviral grade viral filters (20 nm reten- their high selectivity and resolution. However, throughput
tion size) in contrast to the retroviral grade filters (50 nm for conventional preparative chromatography is inherently

258
Review Trends in Biotechnology Vol.28 No.5

limited as a result of the batch nature of the process and the manufacturing facilities then becomes the major driver
limitations in fluid velocity for medium-sized particle beads for process innovation.
(40–120 mm) that are used in these columns. With increas- Innovative technologies that can operate within the
ing production amounts per cell culture batch in commercial current plant framework are likely to be the first movers
scale operations (up to 25,000 l working volumes), the in a changing processing landscape. Box 2 lists some of the
achievement of >50 kg drug substance batches per run non-chromatographic technologies that are being investi-
are close to realization. Significant debate has been sparked gated to improve process throughput for mAb downstream
by these changes over whether conventional packed bed processing.
chromatography can continue to meet downstream process
demands in the future. Persuasive arguments have been Production of antibody-related products
made for two-column step processes with high product loads In addition to intact mAbs, of which >20 have been
on Protein A capture, and a single polishing step operated in approved for therapeutic use, there is growing interest
the flow-through mode, which indicate that chromato- in antibody-related proteins as the next wave of biother-
graphic steps with adequate cycling can continue to meet apeutics. These include Fc fusion proteins, which consist of
these demands into the future [7,8]. It has also been argued a fusion between the Fc region of mAbs and a protein
that very few mAb products actually require these large partner that is often the ligand for a target protein [74].
production scales because of limitations on market demand. Four Fc fusion proteins are currently approved for thera-
Others have argued that innovative unit operations are peutic use. Chemical conjugates with antibodies are
worthwhile investigating as alternative means of dealing advancing in clinical trials, particularly in oncology [75].
with ever-increasing bioreactor output [29,62,63]. Unit Three conjugated antibodies are commercially approved
operations such as aqueous two-phase separation, selective (Zevalin, Bexxar and Mylotarg). Although they possess
precipitation with polyelectrolytes or polymers, membrane several advantages, intact mAbs often have limited bioa-
chromatography, and high-resolution UF and crystalliza- vailability because of their large size. Antibody fragments
tion have been cited as possible replacements for chroma- are being developed to circumvent this limitation, while
tography. retaining the binding characteristics of mAbs [76]. A new
In reality, the best non-chromatographic processing generation of products that utilize scaffolds, and are
steps will likely resemble a combination of these two similar to mAbs, have also been developed. These include
seemingly divergent scenarios. It is clear that the biophar- adnectins that are based on the fibronectin III protein and
maceutical industry is slow to adopt novel processing can be engineered to bind to a wide range of targets in
technologies that require significant capital investments similar fashion to mAbs [77,78]. In addition to improved
or modifications to existing facilities. Globally, biopharma- bioavailability to tissues, these engineered proteins can
ceutical manufacturers have made significant investments potentially access clefts or grooves within target antigens
in current processing equipment, particularly centrifuges that bigger mAbs may not.
and large-scale tanks. By contrast, the industry has readily The mAb process platform can serve the needs for some
adopted innovations that fit in within the current facility of these types of products, including Fc fusion proteins,
paradigm, such as novel adsorbents and membrane bispecific antibodies or conjugated antibodies. Antibody
chromatography. fragments are smaller units that are not glycosylated,
Understanding some of the reasons behind cost press- which allows high productivity that is inherent in bacterial
ures is key in predicting what changes could occur in the expression systems [79]. Many of these types of products
future. A pressure that has been cited as a reason for are expressed as intracellular inclusion bodies that require
significant change is the coming advent of follow-on bio- refolding to recover the intact product. Refolding of the
logics [64]. However, follow-on biologics (also known as inclusion bodies is carried out using chaotropes, such as
‘biosimilars’) will be required to face regulatory require- urea, at low protein concentrations to reduce product
ments for comparability, which is a significantly bigger aggregation as the denaturant concentration is decreased.
hurdle for biopharmaceuticals than for small-molecule The polishing steps for these products need to achieve
pharmaceuticals. The burden in demonstrating compar- separation of the misfolded forms in addition to separ-
ability for these complex molecules often will extend ated-out host cell proteins and aggregates. Dedicated viral
beyond biochemical and biophysical comparability to in- clearance steps are not a regulatory requirement for micro-
clude safety and efficacy clinical trials. This is likely to bially expressed proteins. Refolding can often be a low-
diminish the potential for biosimilars to gain a significant yielding unit operation, and, as mammalian cell culture
foothold in the marketplace. As a result, we feel it is titers improve, there could be occasion to revisit the expres-
unlikely that the cost pressures from biosimilars will serve sion system of choice for antibody fragments. A defined
as a major driver for innovation in biologic manufacturing process platform has yet to become widely established for
processes. By contrast, biopharmaceutical companies antibody fragment production but will be a key future
already face significant internal pressures to minimize development as these products mature in the development
extensive investments in new facilities and optimize the pipeline.
usage of existing facilities. A large part of this pressure is
due to the slowing rate of new drug approvals and Challenges and future trends
increased competition among pharmaceutical and biotech- The upstream process platform for mAb production has
nology companies in indications for which new thera- evolved significantly, both on the cell line creation and
peutics are developed. Effective utilization of existing production bioreactor process fronts, which leads to the

259
Review Trends in Biotechnology Vol.28 No.5

achievement of product titers that have exceeded all 11 Jiang, Z. et al. (2006) Regulation of recombinant monoclonal
antibody production in CHO cells – a comparative study of gene
previous experience with cell culture. The downstream
copy number, mRNA level and protein expression. Biotechnol. Progr.
process platform for mAb production still includes several 22, 313–318
chromatographic steps. Improved understanding of pro- 12 Wurm, F. (2004) Production of recombinant protein therapeutics in
cess-scale purification of mAbs has arisen as a result of cultivated mammalian cells. Nat. Biotechnol. 22, 1393–1398
experience with an ever-increasing number of mAbs in 13 Chu, L. and Robinson, D. (2001) Industrial choices for protein
production by large-scale cell culture. Curr. Opin. Biotechnol. 12,
development and large-scale manufacturing. Future devel-
180–187
opments will take place on two fronts: technology that will 14 Wagner-Rousset, E. et al. (2008) The way forward, enhanced
enable incremental advances in the current platforms, and characterization of therapeutic antibody glycosylation: comparison of
developments that will cause more radical change in the three level MS based strategies J. Chromatogr. B 872, 23–37
technology employed for mAb production. 15 Majors, B. et al. (2009) Mcl-1 overexpression leads to higher viabilities
and increased production of humanized monoclonal antibody in CHO
Although the current process platform might appear to cells. Biotechnol. Progr. 25, 1161–1168
be safely ensconced in practice, we feel that several 16 Jain, E. and Kumar, A. (2008) Upstream processes in antibody
drivers will lead to significant evolution over the next production – evaluation of critical parameters. Biotechnol. Adv. 26,
decade. With the high titers now achievable in cell culture, 46–72
the need to construct new facilities with very large 17 Lee, C. et al. (2008) A clone screening method using mRNA levels to
determine specific productivity and product quality for monoclonal
bioreactors will diminish, although current facilities will antibodies. Biotechnol. Bioeng. 102, 1107–1118
continue to be in use. Lower production volumes will 18 Haldankar, R. et al. (2006) Serum free suspension large-scale transient
increase the need for facility flexibility and faster tranfection of CHO cells in WAVE bioreactors. Mol. Biotechnol. 34,
turnaround that will lead to growth in the use of 191–199
19 Ye, J. et al. (2009) High-level protein expression in scaleable CHO
disposables in manufacturing. On the process front, the
transient transfection. Biotechnol. Bioeng. 103, 542–551
productivity driver will remain even with the use of 20 Crowell, C. et al. (2007) Amino acid and manganese supplementation
smaller bioreactors. This will continue to drive innovation modulates the glycosylation state of erythropoietin in a CHO culture
in the development of non-chromatographic downstream system. Biotechnol. Bioeng. 96, 538–549
steps. However, it has to be noted that, while such 21 Wong, D. et al. (2004) Impact of dynamic on-line fed-batch strategies on
metabolism, productivity and N-glycosylation in CHO cell cultures.
technologies have been investigated for a very long time
Biotechnol. Bioeng. 89, 164–177
already, implementation into a biopharmaceutical 22 Matsunaga, N. et al. (2009) Estimation of dissolved carbon dioxide
process platform has not yet been demonstrated. We feel stripping in a large bioreactor using model medium. J. Biosci. Bioeng.
that the alternative technologies that will be the first to 107, 49–424
find application will be those that fit within the current 23 Roopenian, D. and Akilesh, S. (2007) FcRn: the neonatal Fc receptor
comes of age. Nat. Rev. Immunol. 7, 715–725
biomanufacturing facility designs, rather than requiring 24 Walsh, G. and Jefferis, R. (2006) Post-translational modifications
major investment on the facilities front. in the context of therapeutic proteins. Nat. Biotechnol. 24, 1241–
Finally, another key development will be the matu- 1252
ration of antibody fragments and novel scaffolds within 25 Hossler, P. et al. (2009) Optimal and consistent protein glycosylation in
the clinical development pipelines. We anticipate the birth mammalian cell culture. Glycobiology 19, 936–949
26 Shukla, A. and Kandula, J. (2008) Harvest and recovery of monoclonal
of a new generation of platform processes that address antibodies from large-scale mammalian cell culture. Biopharm. Int. 7,
specific manufacturing issues with these therapeutic mod- 34–45
alities, using a combination of traditional and non- 27 Yigzaw, Y. et al. (2006) Exploitation of the adsorptive properties of
traditional methods. depth filters for host cell protein removal during monoclonal antibody
purification. Biotechnol. Progr. 22, 288–296
References 28 Kozlowski, S. and Swann, P. (2006) Current and future issues in the
1 Reichert, J. and Valge-Archer, V. (2007) Development trends for manufacturing and development of monoclonal antibodies. Adv. Drug
monoclonal antibody cancer therapeutics. Nat. Rev. Drug Discov. 6, Deliv. Rev. 58, 707–722
349–356 29 Low, D. et al. (2007) Future of antibody purification. J. Chromatogr. B
2 Carter, P. (2006) Potent antibody therapeutics by design. Nat. Rev. 848, 48–63
Immunol. 6, 343–357 30 Ghose, S. et al. (2009) Integrated polishing steps for monoclonal
3 Ruuls, S. et al. (2008) Novel human antibody therapeutics: the age of antibody production, In Process Scale Purification of Antibodies (1st
the Umabs. Biotechnol. J. 3, 1157–1171 edn) (Gottschalk, U., ed.), pp. 145–167, Wiley
4 Jefferis, R. (2009) Glycosylation as a strategy to improve antibody- 31 Follman, D. and Fahrner, R. (2004) J. Chromatogr. A 1024, 79–85
based therapeutics. Nature 8, 226–234 32 Arunakumari, A. and Wang, J. (2009) Purification of human
5 Weiner, L. (2006) Fully human therapeutic monoclonal antibodies J. monoclonal antibodies – non Protein A strategies, In Process Scale
Immunotherapy 29, 1–9 Purification of Antibodies (1st edn) (Gottschalk, U., ed.), pp. 103–123,
6 Shukla, A. et al. (2007) Downstream processing of monoclonal Wiley
antibodies – application of platform approaches. J. Chromatogr. B 33 Ghose, S. et al. (2005) Protein interactions in hydrophobic
848, 28–39 charge induction chromatography (HCIC). Biotechnol. Progr. 21,
7 Kelley, B. (2007) Very large-scale monoclonal antibody purification – 498–508
the case for conventional unit operations. Biotechnol. Prog. 23, 995– 34 Ghose, S. et al. (2006) Evaluation and comparison of alternatives to
1008 Protein A chromatography – mimetic and hydrophobic charge
8 Kelley, B. (2009) Industrialization of mAb production technology – the induction chromatographic stationary phases. J. Chromatogr. A
bioprocessing industry at a crossroads. MAbs 1, 1–10 1122, 144–152
9 Birch, J. and Racher, A. (2006) Antibody production. Adv. Drug Deliv. 35 Liu, J. et al. (2009) Comparison of camelid antibody ligand to Protein A
Rev. 58, 671–685 for monoclonal antibody purification. Biopharm. Int. 22, 35–43
10 Chusainow, J. et al. (2009) A study of monoclonal antibody producing 36 Zandian, M. and Jungbauer, A. (2009) Engineering properties of a
CHO cell lines – what makes a stable high producer? Biotechnol. camelid antibody affinity sorbent for immunoglobulin G purification. J.
Bioeng. 102, 1182–1196 Chromatogr. A 1216, 5548–5556

260
Review Trends in Biotechnology Vol.28 No.5

37 Ghose, S. et al. (2005) Antibody variable region interactions with 58 Harris, R. et al. (2004) Commercial manufacturing scale formulation
Protein A – implications for the development of generic purification and analytical characterization of therapeutic recombinant antibodies.
processes. Biotechnol. Bioeng. 92, 665–673 Drug Dev. Res. 61, 137–154
38 Ghose, S. et al. (2007) Binding capacity differences for antibodies and 59 Bee, J. et al. (2009) Response of a concentrated monoclonal antibody
Fc-fusion proteins on Protein A chromatographic materials. formulation to high shear. Biotechnol. Bioeng. 103, 936–943
Biotechnol. Bioeng. 96, 768–779 60 Bee, J. et al. (2010) Aggregation of a monoclonal antibody induced by
39 Tugcu, N. et al. (2008) Maximizing productivity of chromatographic adsorption to stainless steel. Biotechnol. Bioeng. 105, 121–129
steps for purification of monoclonal antibodies. Biotechnol. Bioeng. 99, 61 Singh, S. et al. (2009) Large-scale freezing of biologics. Bioprocess Intl.
599–613 7, 32–44
40 Ghose, S. et al. (2004) Use and optimization of a dual flow-rate loading 62 Przybycien, T. et al. (2004) Alternative bioseparation operations – life
strategy for maximizing throughput in Protein A affinity beyond packed-bed chromatography. Curr. Opin. Biotech. 15, 469–478
chromatography. Biotechnol. Progr. 20, 830–840 63 Thommes, J. and Etzel, M. (2007) Alternatives to chromatographic
41 Jiang, C. et al. (2009) A mechanistic study of Protein A chromatography separations. Biotechnol. Progr. 23, 42–45
resin lifetime. J. Chromatogr. A 1216, 5849–5855 64 Grabowski, H. (2008) Follow-on biologics – data exclusivity and the
42 Rogers, M. et al. (2009) Development of a rapid sanitization solution for balance between innovation and competition. Nat. Rev. Drug. Discov. 7,
silica-based protein A affinity adsorbents. J. Chromatogr. A 1216, 478–488
4589–4596 65 Azevedo, A. et al. (2009) Chromatography-free recovery of
43 Brorson, K. et al. (2003) Identification of protein A media performance biopharmaceuticals through aqueous two-phase processing. Trends
attributes that can be monitored as surrogates for retrovirus clearance Biotechnol. 27, 240–247
during extended re-use. J. Chromatogr. A 989, 155–163 66 Noel, R. (2007) Chromatography: industrial processing and
44 Shukla, A. and Hinckley, P. (2008) Host cell protein clearance during biotherapeutics Filtration and Separation 44, 26–28
Protein A chromatography – development of an improved wash step. 67 Wang, J. et al. (2009) Precipitation of process-derived impurities in
Biotechnol. Progr. 24, 1115–1121 non-Protein A purification schemes for antibodies. Biopharm Intl.
45 Jin, M. et al. (2010) Profiling of host cell proteins by two dimensional Special Issue on Downstream Processing 2010 – embracing
difference gel electrophoresis (2D-DIGE) – implications for innovation Oct, 4–10
downstream process development. Biotechnol. Bioeng. 105, 306–316 68 McDonald, P. et al. (2009) Selective antibody precipitation using
46 Shukla, A. et al. (2008) Demonstration of robust host cell protein polyelectrolytes – a novel approach to the purification of monoclonal
clearance in biopharmaceutical downstream processes. Biotechnol. antibodies. Biotechnol. Bioeng. 102, 1141–1151
Progr. 24, 615–622 69 Azevedo, A. et al. (2009) Affinity enhanced purification of human
47 Shukla, A. et al. (2007) Protein aggregation kinetics during Protein A monoclonal antibodies by aqueous two-phase separation. Sepn.
chromatography – case study for an Fc fusion protein. J. Chromatogr. A Purific. Technol. 65, 31–39
1171, 22–28 70 Ahamed, T. et al. (2007) Phase behavior of an intact monoclonal
48 Kandula, S. et al. (2009) Design of a filter train for precipitate removal antibody. Biophys. J. 93, 610–619
in mAb downstream processing. Biotechnol. Appl. Biochem. 54, 149– 71 van Reis, R. and Zydney, A. (2007) Bioprocess membrane technology. J.
155 Membrane Sci. 302, 16–50
49 Gagnon, P. (2009) IgG aggregate removal by charged hydrophobic 72 Rao, S. and Zydney, A. (2006) High resolution protein separations
mixed-mode chromatography. Curr. Pharm. Biotech. 10, 434–439 using affinity ultrafiltration with small charged ligands. J. Membrane
50 Zhao, G. et al. (2009) Ligands for mixed-mode protein chromatography: Sci. 280, 781–789
principles, characteristics and design. J. Biotechnol. 144, 3–11 73 Zhou, J. and Tressel, T. (2006) Basic concepts in Q membrane
51 Zydney, A. et al. (2009) Modeling electrostatic exclusion effects during chromatography for large-scale antibody production. Biotechnol.
ion exchange chromatography of monoclonal antibodies. Biotechnol. Progr. 22, 341–349
Bioeng. 102, 1131–1140 74 Chang, C. et al. (2009) Advances and challenges in developing cytokine
52 Kelley, B. et al. (2008) Weak partitioning chromatography for anion- fusion proteins as improved therapeutics. Expert Opin. Drug. Discov. 4,
exchange purification of monoclonal antibodies. Biotechnol. Bioeng. 181–194
101, 553–566 75 Deonarain, M. (2008) Recombinant antibodies for cancer therapy.
53 Zhou, J. (2008) Viral clearance using disposable systems in monoclonal Expert Opin. Biol. Ther. 8, 1123–1141
antibody commercial downstream processing. Biotechnol. Bioeng. 100, 76 Holliger, P. and Hudson, P. (2005) Engineered antibody fragments and
488–496 the rise of single domains. Nat. Biotechnol. 23, 1126–1136
54 Kern, G. and Krishnan, M. (2006) Virus removal by filtration – points to 77 Gill, D. and Damle, N. (2006) Biopharmaceutical drug discovery using
consider. Biopharm. Int. 19, 32–41 novel protein scaffolds. Curr. Opin. Biotechnol. 17, 653–658
55 Bohonak, D. and Zydney, A. (2005) Compaction and permeability 78 Dineen, S. et al. (2008) The adnectin CT-322 is a novel VEGF receptor
effects with virus filtration membranes. J. Membrane Sci. 254, 71–79 inhibitor that decreases tumor burden in an orthotopic mouse model of
56 Ireland, T. et al. (2005) Optimizing virus filtration performance with pancreatic cancer. BMC Cancer 8 (352), 1–10
prefiltration. Bioprocess Intl. Supplement 3, 44–47 79 Andersen, D. and Reilly, D. (2004) Production technologies for
57 Marques, B. et al. (2009) Virus filtration of high-concentration monoclonal antibodies and their fragments. Curr. Opin. Biotechnol.
monoclonal antibody solutions. Biotechnol. Progr. 25, 483–491 15, 456–462

261

You might also like