Introduction
The growing therapeutic use of proteins has
created an increasing need for practical and economical processing
techniques. As a result, biotechnological production methods have
advanced significantly over the last decade. Also, single-use
production technology which has the potential to mitigate many of
the economic and quality issues arising from manufacturing these
products has evolved rapidly (Hodge 2004).
When producing proteins for therapeutic use, a
number of issues must be considered related to the manufacturing,
purification, and characterization of the products.
Biotechnological products for therapeutic use have to meet strict
specifications especially when used via the parenteral route
(Walter and Werner 1993).
In this chapter several aspects of production
(upstream processing) and purification (downstream processing) will
be dealt with briefly. For further details, the reader is referred
to the literature mentioned.
Upstream Processing
Expression Systems
General Considerations
Expression systems for proteins of therapeutic
interest include both pro- and eukaryotic cells (bacteria, yeast,
fungi, plants, insect cells, mammalian cells) and transgenic
animals. The choice of a particular system will be determined to a
large extent by the nature and origin of the desired protein, the
intended use of the product, the amount needed, and the cost.
In principle, any protein can be produced using
genetically engineered organisms, but not every type of protein can
be produced by every type of cell. In the majority of cases, the
protein is foreign to the host cells that have to produce it, and
although the translation of the genetic code can be performed by
the cells, the posttranslation modifications of the protein might
be different as compared to the original product.
About 5 % of the proteome are thought to comprise
enzymes performing over 200 types of posttranslation modifications
of proteins (Walsh 2006).
These modifications are species and/or cell-type specific. The
metabolic pathways that lead to these modifications are genetically
determined by the host cell. Thus, even if the cells are capable of
producing the desired posttranslation modification, like
glycosylation, still the resulting glycosylation pattern might be
different from that of the native protein. Correct N-linked
glycosylation of therapeutically relevant proteins is important for
full biological activity, immunogenicity, stability, targeting, and
pharmacokinetics. Prokaryotic cells, like bacteria, are sometimes
capable of producing N-linked glycoproteins. However, the N-linked
structures found differ from the structures found in eukaryotes
(Dell et al. 2011). Yeast
cells are able to produce recombinant proteins like albumin, and
yeast has been engineered to produce glycoproteins with humanlike
glycan structures including terminal sialylation (reviewed by Celik
and Calik 2011). Still most
products on the market and currently in development use cell types
that are as closely related to the original protein-producing cell
type as possible. Therefore, human-derived proteins, especially
mammalian cells, are chosen for production. Further developments in
the field of engineering (e.g., glycosylation) may allow bacteria
to reproduce some of the posttranslation modification steps common
to eukaryotic cells (Borman 2006). Although still to be further
developed, bacteria and yeast may play a role as future production
systems given their ease and low cost of large-scale
manufacturing.
Generalized features of proteins expressed in
different biological systems are listed in Table 3.1 (see also Walter
et al. 1992). However, it
should be kept in mind that there are exceptions to this table for
specific product/expression systems.
Table
3.1 ■
Generalized features of proteins of
different biological origin.
Protein feature
|
Prokaryotic bacteria
|
Eukaryotic yeast
|
Eukaryotic mammalian cells
|
---|---|---|---|
Concentration
|
High
|
High
|
High
|
Molecular weight
|
Low
|
High
|
High
|
S-S bridges
|
Limitation
|
No limitation
|
No limitation
|
Secretion
|
No
|
Yes/no
|
Yes
|
Aggregation state
|
Inclusion body
|
Singular, native
|
Singular, native
|
Folding
|
Risk of misfolding
|
Correct folding
|
Correct folding
|
Glycosylation
|
Limited
|
Possible
|
Possible
|
Impurities: retrovirus
|
No
|
No
|
Possible
|
Impurities: pyrogen
|
Possible
|
No
|
No
|
Cost to manufacture
|
Low
|
Low
|
High
|
Transgenic Animals
Foreign genes can be introduced into animals like
mice, rabbits, pigs, sheep, goats, and cows through nuclear
transfer and cloning techniques. Using milk-specific promoters, the
desired protein can be expressed in the milk of the female
offspring. During lactation the milk is collected, the milk fats
are removed, and the skimmed milk is used as the starting material
for the purification of the protein.
The advantage of this technology is the
relatively cheap method to produce the desired proteins in vast
quantities when using larger animals like cows. Disadvantages are
the long lead time to generate a herd of transgenic animals and
concerns about the health of the animal. Some proteins expressed in
the mammary gland leak back into the circulation and cause serious
negative health effects. An example is the expression of
erythropoietin in cows. Although the protein was well expressed in
the milk, it caused severe health effects and these experiments
were stopped.
The purification strategies and purity
requirements for proteins from milk can be different from those
derived from bacterial or mammalian cell systems. Often the
transgenic milk containing the recombinant protein also contains
significant amounts of the nonrecombinant counterpart. To separate
these closely related proteins poses a purification challenge. The
“contaminants” in proteins for oral use expressed in milk that is
otherwise consumed by humans are known to be safe for
consumption.
The transgenic animal technology for the
production of pharmaceutical proteins has progressed within the
last few years. The US and EU authorities approved recombinant
antithrombin III (ATryn®, GTC Biotherapeutics) produced
in the milk of transgenic goats. More details about this technology
are presented in Chap.
8.
Plants
Therapeutic proteins can also be expressed in
plants and plant cell cultures (see also Chap. 1). For instance, human
albumin has been expressed in potatoes and tobacco. Whether these
production vehicles are economically feasible has yet to be
established. The lack of genetic stability of plants was sometimes
a drawback. Stable expression of proteins in edible seeds has been
obtained. For instance, rice and barley can be harvested and easily
kept for a prolonged period of time as raw material sources.
Especially for oral therapeutics or vaccines, this might be the
ideal solution to produce large amounts of cheap therapeutics,
because the “contaminants” are known to be safe for consumption
(see also Chap.
21). A better understanding of the plant
molecular biology together with more sophisticated genetic
engineering techniques and strategies to increase yields and
optimize glycan structures resulted in an increase in the number of
products in development including late-stage clinical trials
(reviewed by Orzaez et al. 2009, and Peters and Stoger 2011). Removal of most early bottlenecks
together with the regulatory acceptance of plants as platforms to
produce therapeutic proteins has resulted in a renewed interest by
the industry.
More details about the use of plant systems for
the production of pharmaceutical proteins are presented in
Chap.
7.
Cultivation Systems
General
In general, cells can be cultivated in vessels
containing an appropriate liquid growth medium in which the cells
are either immobilized and grow as a monolayer, attached to
microcarriers, free in suspension, or entrapped in matrices
(usually solidified with agar). The culture method will determine
the scale of the separation and purification methods.
Production-scale cultivation is commonly performed in fermentors,
used for bacterial and fungal cells, or bioreactors, used for
mammalian and insect cells. Bioreactor systems can be classified
into four different types:
-
Stirred tank (Fig. 3.1a)Figure 3.1 ■(a) Schematic representation of stirred-tank bioreactor (Adapted from Klegerman and Groves 1992). (b) Schematic representation of airlift bioreactor (Adapted from Klegerman and Groves 1992). (c) Schematic representation of fixed-bed stirred-tank bioreactor (Adapted from Klegerman and Groves 1992). (d) Schematic representation of hollow fiber perfusion bioreactor (Adapted from Klegerman and Groves 1992)
-
Airlift (Fig. 3.1b)
-
Fixed bed (Fig. 3.1c)
-
Membrane bioreactors (Fig. 3.1d)
Because of its reliability and experience with
the design and scaling up potential, the stirred tank is still the
most commonly used bioreactor. This type of bioreactor is not only
used for suspension cells like CHO, HEK293, and PER.C6®
cells, it is also used for production with adherent cells like Vero
and MDCK cells. In the latter case the production is performed on
microcarriers (Van Wezel et al. 1985).
Single-Use Systems
In the last decade the development of single-use
production systems was boosted. Wave Biotech AG in Switzerland (now
Sartorius Stedim Biotech) is regarded as a visionary player in the
nurture of single-use technologies for mammalian cell culture. With
the development of 3D single-use bags by companies like Hyclone,
Xcellerex, and Sartorius, the number of single-use systems used has
increased. Single-use bioreactors are nowadays used for the
manufacturing of products in development and on the market. Shire
(Dublin, Ireland) was the first company that used single-use
bioreactors up to 2,000 L for the manufacturing of one of their
products. The advantages of the single-use technology are:
-
Cost-effective manufacturing technologyBy introducing single-use systems, the design is such that all items not directly related to the process can be removed from the culture system, like clean-in-place (CIP) and steam-in-place (SIP) systems. Furthermore, a reduction in capital costs is achieved by introducing single-use systems.
-
Increasing number of batchesBy introduction of single-use systems, it is possible to increase the number of batches that can be produced in 1 year, due to the fact that cleaning and sterilization is not needed anymore. The turnover time needed from batch to batch is shortened.
-
Provides flexibility in facility designWhen stainless steel systems are used, changes to the process equipment might impact the design of the stainless steel tanks, piping, etc. These changes will directly influence the CIP and SIP validation status of the facility. By using single-use systems, process changes can easily be incorporated as the setup of the single-use process is flexible, and CIP and SIP validation are not needed. However, in case a change will influence the process, the validated status of the process must be reconsidered and a revalidation might be needed.
-
Speedup implementation and time to marketDue to the great flexibility of the single-use systems, the speed of product to market is not influenced by process changes that might be introduced during the development process. However, the process needs to be validated during Phase 3 development. When changes are introduced after the process is validated, a revalidation might be needed. There is no difference in this respect to the traditional stainless steel setup.
-
Reduction in water and wastewater costsDue to the fact that the systems are single use, there will be a great reduction in the total costs for cleaning, not only a water reduction but also a reduction in the number of hours needed to clean systems and setup for the next batch of product.
-
Reduction in validation costsNo yearly validation costs for cleaning and sterilization are needed anymore when single-use systems are used.
A disadvantage of the single-use system is that
the operational expenses will increase, storage location for
single-use bags and tubing will increase, and the dependence of the
company to one supplier of single-use systems will increase.
Furthermore, there is not yet a standard set in single-use systems
like it is with stainless steel bioreactor systems. Suppliers are
still developing their own systems.
The advantages of the stainless steel bioreactors
are obvious as this traditional technology is well understood and
controlled, although the stainless steel pathway had major
disadvantages such as expensive design, installation, and
maintenance costs combined with significant expenditures of time in
facilities and equipment qualification and validation
efforts.
Fermentation Protocols
The kinetics of cell growth and product formation
will not only dictate the type of bioreactor used but also how the
growth process is performed. Three types of fermentation protocols
are commonly employed and discussed below:
-
BatchIn a batch process, the bioreactor is filled with the entire volume of medium needed during the cell growth and/or production phase. No additional supplements are added to increase the cell growth or production during the process. Waste products, such as lactate and ammonium, and the product itself accumulate in the bioreactor. The product is harvested at the end of the process. Maximum cell density and product yields will be lower compared to a fed-batch process.
-
Fed-batchIn a fed-batch process, a substrate is supplemented to the bioreactor. The substrate consists of the growth-limiting nutrients that are needed during the cell growth phase and/or during the production phase of the process. Like the batch process, waste products accumulate in the bioreactor. The product is harvested at the end of the process. With the fed-batch process, higher cell densities and product yields can be reached compared to the batch process due to the extension of production time that can be achieved compared to a batch process. The substrate used is highly concentrated and can be added to the bioreactor on a daily basis or as a continuous feed. The fed-batch mode is currently widely used for the production of proteins. The process is well understood and characterized.
-
PerfusionIn a perfusion process, the media and waste products are continuously exchanged and the product is harvested throughout the culture period. A membrane device is used to retain the cells in the bioreactor, and waste medium is removed from the bioreactor by this device (Fig. 3.2). To keep the level medium constant in the bioreactor, fresh medium is supplemented to the bioreactor. By operating in perfusion mode, the level of waste products will be kept constant and generates a stable environment for the cells to grow or to produce. With the perfusion process, much higher cell densities can be reached and therefore higher productivity (Compton and Jensen 2007).Figure 3.2 ■Schematic representation of perfusion device coupled to a stirred-tank bioreactor. ATF alternating tangential flow.
In all these three protocols, the cells go
through four distinctive phases (see also Chap. 1):
1.
Lag phase
In this phase the cells are adapting to the
conditions in the bioreactor and do not yet grow.
2.
Exponential growth phase
During this phase, cells grow in a more or less
constant doubling time for a fixed period. The mammalian cell
doubling time is cell-type dependent and usually varies between 20
and 40 h. Plotting the natural logarithm of cell number against
time produces a straight line. Therefore, the exponential growth
phase is also called the log phase. The growth phase will be
affected by growth conditions like temperature, pH, oxygen
pressure, and external forces like stirring and baffles that are
inserted into the bioreactor. Furthermore, the growth rate is
affected by the supply of sufficient nutrients, buildup of waste
nutrients, etc.
3.
Stationary phase
In the stationary phase, the growth rate of the
cells slows down due to the fact that nutrients are depleted and/or
build up of toxic waste products like lactate and ammonium. In this
phase, constant cell numbers are found due to equal cell growth and
cell death.
4.
Death phase
Cells die due to depletion of nutrients and/or
presence of high concentrations of toxic products like lactate and
ammonium.
Examples of animal cells that are commonly used
to produce proteins of clinical interest are Chinese hamster ovary
cells (CHO), immortalized human embryonic retinal cells
(PER.C6® cells), baby hamster kidney cells (BHK),
lymphoblastoid tumor cells (interferon production), melanoma cells
(plasminogen activator), and hybridized tumor cells (monoclonal
antibodies).
The cell culture has to be free from undesired
microorganisms that may destroy the cell culture or present hazards
to the patient by producing endotoxins. Therefore, strict measures
are required for both the production procedures and materials used
(WHO 2010; Berthold and Walter
1994) to prevent a possible
contamination with extraneous agents like viruses, bacteria, and
mycoplasma. Furthermore, strict measures are needed, especially
with regard to the raw materials used, to prevent contaminations
with transmissible spongiform encephalopathies (TSEs).
Cultivation Medium
In order to achieve optimal growth of cells and
optimal production of recombinant proteins, it is of great
importance not only that conditions such as stirring, pH, oxygen
pressure, and temperature are chosen and controlled appropriately
but also that a cell growth and protein production medium with the
proper nutrients are provided for each stage of the production
process.
The media used for mammalian cell culture are
complex and consist of a mixture of diverse components, such as
sugars, amino acids, electrolytes, vitamins, fetal calf serum,
and/or a mixture of peptones, growth factors, hormones, and other
proteins (see Table 3.2). Many of these ingredients are
pre-blended either as concentrate or as homogeneous mixtures of
powders. To prepare the final medium, components are dissolved in
purified water before filtration. The final medium is filtrated
through 0.2 μm filters or through 0.1 μm filters to prevent
possible mycoplasma contamination. Some supplements, especially
fetal calf serum, contribute considerably to the presence of
contaminating proteins and may seriously complicate purification
procedures. Moreover, the composition of serum is variable. It
depends on the individual animal, season of the year, suppliers’
treatment, etc. The use of serum may introduce adventitious
material such as viruses, mycoplasma, bacteria, and fungi into the
culture system (Berthold and Walter 1994). Furthermore, the possible presence of
prions that can cause transmissible spongiform encephalitis almost
precludes the use of materials from animal origin. However, if use
of this material is inevitable, one must follow the relevant
guidelines in which selective sourcing of the material is the key
measure to safety (EMA 2011).
Many of these potential problems when using serum in cell culture
media led to the development of fully defined, free from
animal-derived material. These medium formulations were not only
developed by the suppliers, there is the trend that the key players
in the biotech industry develop their own fully defined medium for
their specific production platforms. The advantage of this is that
the industry is less dependent on medium suppliers. The fully
defined media have been shown to give satisfactory results in
large-scale production settings for monoclonal antibody processes.
However, hydrolysates from nonanimal origin, like yeast and plant
sources, are more and more used for optimal cell growth and product
secretion (reviewed by Shukla and Thömmes 2010).
Table
3.2 ■
Major components of growth media for
mammalian cell structures.
Type of nutrient
|
Example(s)
|
---|---|
Sugars
|
Glucose, lactose, sucrose, maltose,
dextrins
|
Fat
|
Fatty acids, triglycerides
|
Water (high quality, sterilized)
|
Water for injection
|
Amino acids
|
Glutamine
|
Electrolytes
|
Calcium, sodium, potassium, phosphate
|
Vitamins
|
Ascorbic acid, -tocopherol, thiamine,
riboflavine, folic acid, pyridoxin
|
Serum (fetal calf serum, synthetic
serum)
|
Albumin, transferrin
|
Trace minerals
|
Iron, manganese, copper, cobalt, zinc
|
Hormones
|
Growth factors
|
Downstream Processing
Introduction
Recovering a biological reagent from a cell
culture supernatant is one of the critical parts of the
manufacturing procedure for biotech products, and purification
costs typically outweigh those of the upstream part of the
production process. For the production of monoclonal antibodies,
protein A resin accounts for some 10 % of the cost, while virus
removal by filtration can account for 40 % of the cost (Gottschalk
2006).
More than a decade ago, the protein product was
available in a very dilute form, e.g., 10–200 mg/L. At the most
concentrations, up to 500–800 mg/L could be reached (Berthold and
Walter 1994). Developments in
cell culture technology through application of genetics and
proteomics resulted in product titers well above 1 g/L. Product
titers above 20 g/L are also reported (Monteclaro 2010). These high product titers pose a
challenge to the downstream processing unit operations (Shukla and
Thömmes 2010). With the
low-yield processes, a concentration step is often required to
reduce handling volumes for further purification. Usually, the
product subsequently undergoes a series of purification steps. The
first step in a purification process is to remove cells and cell
debris from the process fluids. This process step is normally
performed using centrifugation and/or depth filters. Depth filters
are often used in combination with filter aid or diatomaceous
earth. Often the clarification step is regarded as a part of the
upstream process. Therefore, the first actual step in the
purification process is a capture step. Subsequent steps remove the
residual bulk contaminants, and a final step removes trace
contaminants and sometimes variant forms of the molecule.
Alternatively, the reverse strategy, where the main contaminants
are captured and the product is purified in subsequent steps, might
result in a more economic process, especially if the product is not
excreted from the cells. In the case where the product is excreted
into the cell culture medium, the product will not represent more
than 1–5 % of total cellular protein, and a specific binding of the
cellular proteins in a product-specific capture step will have a
high impact on the efficiency of that step. If the bulk of the
contaminants can be removed first, the specific capture step will
be more efficient and smaller in size and therefore more economic.
Furthermore, smaller subsequent unit operation steps (e.g.,
chromatography columns) could be used.
After purification, additional steps are
performed to bring the desired product into a formulation buffer in
which the product is stabilized and can be stored for the desired
time until further process steps are performed. Before storage of
the final bulk drug substance, the product will be sterilized.
Normally this will be performed by a 0.2 μm filtration step.
Formulation aspects will be dealt with in Chap. 4.
When designing an upstream and purification
protocol, the possibility for scaling up should be considered
carefully. A process that has been designed for small quantities is
most often not suitable for large quantities for technical,
economic, and safety reasons. Developing a purification process,
i.e., the isolation and purification of the desired product also
called the downstream process (DSP), to recover a recombinant
protein in large quantities occurs in two stages: design and scale-up.
Separating the impurities from the product
protein requires a series of purification steps (process design), each removing some of
the impurities and bringing the product closer to its final
specification. In general, the starting feedstock contains cell
debris and/or whole-cell particulate material that must be removed.
Defining the major contaminants in the starting material is helpful
in the downstream process design. This includes detailed
information on the source of the material (e.g., bacterial or
mammalian cell culture) and major contaminants that are used or
produced in the upstream process (e.g., albumin, serum, or product
analogs). Moreover, the physical characteristics of the product
versus the known contaminants (thermal stability, isoelectric
point, molecular weight, hydrophobicity, density, specific binding
properties) largely determine the process design. Processes used
for production of therapeutics in humans should be safe,
reproducible, robust, and produced at the desired cost of goods.
The DSP steps may expose the protein molecules to high physical
stress (e.g., high temperatures and extreme pH) which can alter the
protein properties possibly leading to loss in efficacy. Any
substance that is used by injection must be sterile. Furthermore,
the endotoxin concentration must be below a certain level depending
on the product. Limits are stated in the individual monographs
which are to be consulted (e.g., European Pharmacopoeia: less than
0.2 endotoxin units per kg body mass for intrathecal application).
Aseptic techniques have to be used wherever possible and
necessitate procedures throughout with clean air and microbial
control of all materials and equipment used. During validation of
the purification process, one must also demonstrate that potential
viral contaminants are inactivated and removed (Walter et al.
1992). The purification
matrices should be at least sanitizable or, if possible,
steam-sterilizable. For depyrogenation, the purification material
must withstand either extended dry heat at ≥180 °C or treatment
with 1–2 M sodium hydroxide (for further information, see
Chap.
4). If any material in contact with the product
inadvertently releases compounds, these leachables must be analyzed
and their removal by subsequent purification steps must be
demonstrated during process validation, or it must be demonstrated
that the leachables are below a toxic level. The increased use of
plastic film-based single-use production technology (e.g., sterile
single-use bioreactor bags, bags to store liquids and filter
housings) has made these aspects more significant in the last
decade. Suppliers have reacted by providing a significant body of
information regarding leachables and biocompatibility for typical
solutions used during processing. The problem of leachables is
especially hampering the use of affinity chromatography (see below)
in the production of pharmaceuticals for human use. On small-scale
affinity, chromatography is an important tool for purification and
the resulting product might be used for (animal) toxicity studies,
but for human use the removal of any leached ligands below a toxic
level has to be demonstrated. Because free affinity ligands will
bind to the product, the removal might be cumbersome.
Scale-up is the term used to describe a
number of processes employed in converting a laboratory procedure
into an economical, industrial process. During the scale-up phase,
the process moves from the laboratory scale to the pilot plant and
finally to the production plant. The objective of scale-up is to
produce a product of high quality at a competitive price. Since the
costs of downstream processing can be as high as 50–80 % of the
total cost of the bulk product, practical and economical ways of
purifying the product should be used. Superior protein purification
methods hold the key to a strong market position (Wheelwright
1993).
Basic operations required for a downstream
purification process used for macromolecules from biological
sources are shown in Fig. 3.3.

Figure
3.3 ■
Basic operations required for the
purification of a biopharmaceutical macromolecule.
As mentioned before, the design of downstream
processing is highly product dependent. Therefore, each product
requires a specific multistage purification procedure (Sadana
1989). The basic scheme as
represented in Fig. 3.3 becomes complex. A typical example of a
process flow for the downstream processing is shown in Fig.
3.4. This
scheme represents the processing of a glycosylated recombinant
interferon (about 28 kDa) produced in mammalian cells. The aims of
the individual unit operations are described.

Figure
3.4 ■
Downstream processing of a glycosylated
recombinant interferon, describing the purpose of the inclusion of
the individual unit operations. F filtration, TFF tangential flow filtration,
UF ultrafiltration,
DF diafiltration,
A adsorption (Adapted from
Berthold and Walter 1994).
Once the harvest volume and product concentration
can be managed, the main purification phase can start. A number of
purification methods are available to separate proteins on the
basis of a wide variety of different physicochemical criteria such
as size, charge, hydrophobicity, and solubility. Detailed
information about some separation and purification methods commonly
used in purification schemes is provided below.
Filtration/Centrifugation
Products from biotechnological industry must be
separated from biological systems that contain suspended
particulate material, including whole cells, lysed cell material,
and fragments of broken cells generated when cell breakage has been
necessary to release intracellular products. Most downstream
processing flow sheets will, therefore, include at least one unit
operation for the removal (“clarification”) or concentration, just
the opposite, of particulates. Most frequently used methods are
centrifugation and filtration techniques (e.g., ultrafiltration,
diafiltration, and microfiltration). However, the expense and
effectiveness of such methods is highly dependent on the physical
nature of the particulate material and of the product.
Filtration
Several filtration systems have been developed
for separation of cells from media, the most successful being depth
filtration and tangential flow systems (also referred to as “cross
flow”). In the latter system, high shear across the membrane
surface limits fouling, gel layer formation, and concentration
polarization. In ultrafiltration, mixtures of molecules of
different molecular dimensions are separated by passage of a
dispersion under pressure across a membrane with a defined pore
size. In general, ultrafiltration achieves little purification of
protein product from other molecules with a comparable size,
because of the relatively large pore-size distribution of the
membranes. However, this technique is widely used to concentrate
macromolecules and also to change the aqueous phase in which the
particles are dispersed or in which molecules are dissolved
(diafiltration) to one required for the subsequent purification
steps.
Centrifugation
Subcellular particles and organelles, suspended
in a viscous liquid (e.g., the particles produced when cells are
disrupted by mechanical procedures), are difficult to separate
either by using one fixed centrifugation step or by filtration.
But, they can be isolated efficiently by centrifugation at
different speeds. For instance, nuclei can be obtained by
centrifugation at 400× g for 20 min, while plasma membrane vesicles
are pelleted at higher centrifugation rates and longer
centrifugation times (fractional centrifugation). In many cases,
however, total biomass can easily be separated from the medium by
centrifugation (e.g., continuous disc-stack centrifuge). Buoyant
density centrifugation can be useful for separation of particles as
well. This technique uses a viscous fluid with a continuous
gradient of density in a centrifuge tube. Particles and molecules
of various densities within the density range in the tube will
cease to move when the isopycnic region has been reached. Both
techniques of continuous (fluid densities within a range) and
discontinuous (blocks of fluid with different density) density
gradient centrifugation are used in buoyant density centrifugation
on a laboratory scale. However, for application on an industrial
scale, continuous centrifuges (e.g., tubular bowl centrifuges) are
only used for discontinuous buoyant density centrifugation of
protein products. This type of industrial centrifuge is mainly
applied to recover precipitated proteins or contaminants. For
influenza vaccines, continuous centrifugation is already for
decades the workhorse to purify influenza viruses on an industrial
scale.
Precipitation
The solubility of a particular protein depends on
the physicochemical environment, for example, pH, ionic species,
and ionic strength of the solution (see also Chap. 4). A slow continuous increase
of the ionic strength (of a protein mixture) will selectively drive
proteins out of solution. This phenomenon is known as “salting
out.” A wide variety of agents, with different “salting-out”
potencies are available. Chaotropic series with increasing
“salting-out” effects of negatively (I) and positively (II) charged
molecules are given below:
I.
SCN–, I–, CLO4–, NO3–, Br–, Cl–, CH3COO–, PO43–,
SO42–
II.
Ba2+, Ca2+, Mg2+, Li+, Cs+, Na+, K+, Rb+,
NH4+
Ammonium sulfate is highly soluble in cold
aqueous solutions and is frequently used in “salting-out”
purification.
Another method to precipitate proteins is to use
water-miscible organic solvents (change in the dielectric
constant). Examples of precipitating agents are polyethylene glycol
and trichloroacetic acid. Under certain conditions, chitosan and
nonionic polyoxyethylene detergents also induce precipitation
(Cartwright 1987; Homma et al.
1993; Terstappen et al.
1993). Cationic detergents
have been used to selectively precipitate DNA.
Precipitation is a scalable, simple, and
relatively economical procedure for the recovery of a product from
a dilute feedstock. It has been widely used for the isolation of
proteins from culture supernatants. Unfortunately, with most bulk
precipitation methods, the gain in purity is generally limited and
product recovery can be low. Moreover, extraneous components are
introduced which must be eliminated later. Finally, large
quantities of precipitates may be difficult to handle. Despite
these limitations, recovery by precipitation has been used with
considerable success for some products.
Chromatography
Introduction
In preparative chromatography systems, molecular
species are primarily separated based on differences in
distribution between two phases, one which is the stationary phase
(mostly a solid phase) and the other which moves. This mobile phase
may be liquid or gaseous (see also Chap. 2). Nowadays, almost all
stationary phases (fine particles providing a large surface area)
are packed into a column. The mobile phase is passed through by
pumps. Downstream protein purification protocols usually have at
least two to three chromatography steps. Chromatographic methods
used in purification procedures of biotech products are listed in
Table 3.3
and are briefly discussed in the following sections.
Table
3.3 ■
Frequently used separation processes and
their physical basis.
Separation technique
|
Mode/principle
|
Separation based on
|
---|---|---|
Membranes
|
Microfiltration
|
Size
|
Ultrafiltration
|
Size
|
|
Nanofiltration
|
Size
|
|
Dialysis
|
Size
|
|
Charged membranes
|
Charge
|
|
Centrifugation
|
Isopycnic banding
|
Density
|
Non-equilibrium setting
|
Density
|
|
Extraction
|
Fluid extraction
|
Solubility
|
Liquid/liquid extraction
|
Partition, change in solubility
|
|
Precipitation
|
Fractional precipitation
|
Change in solubility
|
Chromatography
|
Ion exchange
|
Charge
|
Gel filtration
|
Size
|
|
Affinity
|
Specific ligand-substrate interaction
|
|
Hydrophobic interaction
|
Hydrophobicity
|
|
Adsorption
|
Covalent/non-covalent binding
|
Chromatographic Stationary Phases
Chromatographic procedures often represent the
rate-limiting step in the overall downstream processing. An
important primary factor governing the rate of operation is the
mass transport into the pores of conventional packing materials.
Adsorbents employed include inorganic materials such as silica
gels, glass beads, hydroxyapatite, various metal oxides (alumina),
and organic polymers (cross-linked dextrans, cellulose, agarose).
Separation occurs by differential interaction of sample components
with the chromatographic medium. Ionic groups such as amines and
carboxylic acids, dipolar groups such as carbonyl functional
groups, and hydrogen bond-donating and bond-accepting groups
control the interaction of the sample components with the
stationary phase, and these functional groups slow down the elution
rate if interaction occurs.
Chromatographic stationary phases for use on a
large scale have improved considerably over the last decades.
Hjerten et al. (1993) reported
on the use of compressed acrylamide-based polymer structures. These
materials allow relatively fast separations with good
chromatographic performance. Another approach to the problems
associated with mass transport in conventional systems is to use
chromatographic particles that contain some large “through pores”
in addition to conventional pores (see Fig. 3.5). These
flow-through or “perfusion chromatography” media enable faster
convective mass transport into particles and allow operation at
much higher speeds without loss in resolution or binding capacity
(Afeyan et al. 1989; Fulton
1994). Another development is
the design of spirally wrapped columns containing the adsorption
medium. This configuration permits high throughput, high capacity,
and good capture efficiency (Cartwright 1987).

Figure
3.5 ■
The structure of conventional
chromatographic particles (a)
and the perfusion of flow through chromatographic particles
(b) (Adapted from Fulton
1994).
The ideal stationary phase for protein separation
should possess a number of characteristics, among which are high
mechanical strength, high porosity, no nonspecific interaction
between protein and the support phase, high capacity,
biocompatibility, and high stability of the matrix in a variety of
solvents. The latter is especially true for columns used for the
production of clinical materials that need to be cleaned,
depyrogenized, disinfected, and sterilized at regular intervals.
High-performance liquid chromatography (HPLC) systems fulfill many
of these criteria. Liquid phases should be carefully chosen to
minimize loss of biological activity resulting from the use of some
organic solvents. In HPLC small pore-size stationary phases that
are incompressible are used. These particles are small, rigid, and
regularly sized (to provide a high surface area). The mobile liquid
phase is forced under high pressure through the column material.
Reversed-phase HPLC systems, using less polar stationary phases
than the mobile phases, can in a very few cases be effectively
integrated into preparative scale purification schemes of proteins
and can serve both as a means of concentration and purification
(Benedek and Swadesh 1991).
Unfortunately, HPLC equipment and resin costs are high. Moreover,
HPLC is poorly scalable and thus this technology is basically not
applied in large-scale purification schemes.
In production environments, columns which operate
at relatively low back pressure are often used. They have the
advantage that they can be used in equipment constructed from
plastics which, unlike conventional stainless steel equipment,
resists all buffers likely to be employed in the separation of
biomolecules (consider the effect of leachables from plastics).
These columns are commercially available and permit the efficient
separation of proteins in a single run, making this an attractive
unit operation in a manufacturing process. Results can be obtained
rapidly and with high resolution. A new development is the use of
chromatography equipment with fully disposable flow paths that
resists almost all chemicals used in protein purification including
disinfection and sterilization media.
Adsorption Chromatography
In adsorption chromatography (also called “normal
phase” chromatography), the stationary phase is more polar than the
mobile phase. The protein of interest selectively binds to a static
matrix under one condition and is released under a different
condition. Adsorption chromatography methods enable high ratios of
product load to stationary phase volume. Therefore, this principle
is economically scalable.
Ion-Exchange Chromatography
Ion-exchange chromatography can be a powerful
step early in a purification scheme. It can be easily scaled up.
Ion-exchange chromatography can be used in a negative mode, i.e.,
the product flows through the column under conditions that favor
the adsorption of contaminants to the matrix, while the protein of
interest does not bind (Tennikova and Svec 1993). The type of the column needed is
determined by the properties of the proteins to be purified (e.g.,
isoelectric point and charge density). Anion exchangers bind
negatively charged molecules and cation exchangers bind positively
charged molecules. In salt-gradient ion-exchange chromatography,
the salt concentration in the perfusing elution buffer is increased
continuously or in steps. The stronger the binding of an individual
protein to the ion exchanger, the later it will appear in the
elution buffer. Likewise, in pH-gradient chromatography, the pH is
changed continuously or in steps. Here, the protein binds at 1 pH
and is released at a different pH. As a result of the heterogeneity
in glycosylation (e.g., a varying number of sialic acid moieties),
glycosylated proteins may elute in a relatively broad pH range (up
to 2 pH units).
In order to simplify purification, a specific
amino acid tail can be added to the protein at the gene level to
create a “purification handle.” For example, a short tail
consisting of arginine residues allows a protein to bind to a
cation exchanger under conditions where almost no other cell
proteins bind. However, this technique is only useful for
laboratory-scale isolation of the product and cannot be used for
production scale due to regulatory problems related to the removal
of the arginine or other specific tags from the protein.
(Immuno)Affinity Chromatography
Affinity Chromatography
Affinity chromatography is based on highly
specific interactions between an immobilized ligand and the protein
of interest. Affinity chromatography is a very powerful method for
the purification of proteins. Under physiological conditions, the
protein binds to the ligand. Extensive washing of this matrix will
remove contaminants, and the purified protein can be recovered by
the addition of ligands competing for the stationary phase binding
sites or by changes in physical conditions (such as low or high pH
of the eluent) which greatly reduce the affinity. Examples of
affinity chromatography include the purification of glycoproteins,
which bind to immobilized lectins, and the purification of serine
proteases with lysine binding sites, which bind to immobilized
lysine. In these cases, a soluble ligand (sugar or lysine,
respectively) can be used to elute the required product under
relatively mild conditions. Another example is the use of the
affinity of protein A and protein G for antibodies. Protein A and
protein G have a high affinity for the Fc portions of many
immunoglobulins from various animals. Protein A and G matrices can
be commercially obtained with a high degree of purity. For the
purification of, e.g., hormones or growth factors, the receptors or
short peptide sequence that mimic the binding site of the receptor
molecule can be used as affinity ligands. Some proteins show highly
selective affinity for certain dyes commercially available as
immobilized ligands on purification matrices. When considering the
selection of these ligands for pharmaceutical production, one must
realize that some of these dyes are carcinogenic and that a
fraction may leach out during the process.
An interesting approach to optimize purification
is the use of a gene that codes not only for the desired protein
but also for an additional sequence that facilitates recovery by
affinity chromatography. At a later stage the additional sequence
is removed by a specific cleavage reaction. As mentioned before,
this is a complex process that needs additional purification
steps.
In general, use of affinity chromatography in the
production process for therapeutics may lead to complications
during validation of the removal of free ligands or protein
extensions. Consequently, except for monoclonal antibodies where
affinity chromatography is part of the purification platform at
large scale, this technology is rarely used in the industry.
Immunoaffinity Chromatography
The specific binding of antibodies to their
epitopes is used in immunoaffinity chromatography (Chase and
Draeger 1993). This technique
can be applied for purification of either the antigen or the
antibody. The antibody can be covalently coupled to the stationary
phase and act as the “receptor” for the antigen to be purified.
Alternatively, the antigen, or parts thereof, can be attached to
the stationary phase for the purification of the antibody.
Advantages of immunoaffinity chromatography are its high
specificity and the combination of concentration and purification
in one step.
A disadvantage associated with immunoaffinity
methods is the sometimes very strong antibody-antigen binding. This
requires harsh conditions during elution of the ligand. Under such
conditions, sensitive ligands could be harmed (e.g., by
denaturation of the protein to be purified). This can be alleviated
by the selection of antibodies and environmental conditions with
high specificity and sufficient affinity to induce an
antibody-ligand interaction, while the antigen can be released
under mild conditions (Jones 1990). Another concern is disruption of the
covalent bond linking the “receptor” to the matrix. This would
result in elution of the entire complex. Therefore, in practice, a
further purification step after affinity chromatography as well as
an appropriate detection assay (e.g., ELISA) is almost always
necessary. On the other hand, improved coupling chemistry that is
less susceptible to hydrolysis has been developed to prevent
leaching.
Scale-up of immunoaffinity chromatography is
often hampered by the relatively large quantity of the specific
“receptor” (either the antigen or the antibody) that is required
and the lack of commercially available, ready-to-use matrices. The
use of immunoaffinity in pharmaceutical processes will have major
regulatory consequences since the immunoaffinity ligand used will
be considered by the regulatory bodies as a “second product,” thus
will be subjected to the nearly the same regulatory scrutiny as the
drug substance. Moreover, immunoaffinity ligands can have a
significant effect of the final costs of goods.
Examples of proteins of potential therapeutic
value that have been purified using immunoaffinity chromatography
are interferons, urokinase, erythropoietin, interleukin-2, human
factor VIII and X, and recombinant tissue plasminogen
activator.
Hydrophobic Interaction Chromatography
Under physiological conditions, most hydrophobic
amino acid residues are located inside the protein core, and only a
small fraction of hydrophobic amino acids is exposed on the
“surface” of a protein. Their exposure is suppressed because of the
presence of hydrophilic amino acids that attract large clusters of
water molecules and form a “shield.” High salt concentrations
reduce the hydration of a protein, and the surface-exposed
hydrophobic amino acid residues become more accessible. Hydrophobic
interaction chromatography (HIC) is based on non-covalent and
non-electrostatic interactions between proteins and the stationary
phase. HIC is a mild technique, usually yielding high recoveries of
proteins that are not damaged, are folded correctly, and are
separated from contaminants that are structurally related. HIC is
ideally placed in the purification scheme after ion-exchange
chromatography, where the protein usually is released in high ionic
strength elution media (Heng and Glatz 1993).
Gel-Permeation Chromatography
Gel-permeation or size-exclusion chromatography,
also known as gel filtration, separates molecules according to
their shape and size (see Fig. 3.6). Inert gels with narrow pore-size
distributions in the size range of proteins are available. These
gels are packed into a column and the protein mixture is then
loaded on top of the column and the proteins diffuse into the gel.
The smaller the protein, the more volume it will have available in
which to disperse. Molecules that are larger than the largest pores
are not able to penetrate the gel beads and will therefore stay in
the void volume of the column. When a continuous flow of buffer
passes through the column, the larger proteins will elute first and
the smallest molecules last. Gel-permeation chromatography is a
good alternative to membrane diafiltration for buffer exchange at
almost any purification stage, and it is often used in laboratory
design. At production scale, the use of this technique is usually
limited, because only relatively small sample volumes can be loaded
on a large column (up to one-third of the column volume in the case
of “buffer exchange”). It is therefore best avoided or used late in
the purification process when the protein is available in a highly
concentrated form. Gel filtration is commonly used as the final
step in the purification to bring proteins in the appropriate
buffer used in the final formulation. In this application, its use
has little if no effect on the product purity characteristics.

Figure
3.6 ■
Schematic representation of gel filtration
(Adapted from James 1992).
Expanded Beds
As mentioned before, purification schemes are
based on multistep protocols. This not only adds greatly to the
overall production costs but also can result in significant loss of
product. Therefore, there still is an interest in the development
of new methods for simplifying the purification process. Adsorption
techniques are popular methods for the recovery of proteins, and
the conventional operating format for preparative separations is a
packed column (or fixed bed) of adsorbent. Particulate material,
however, can be trapped near the bed, which results in an increase
in the pressure drop across the bed and eventually in clogging of
the column. This can be avoided by the use of pre-column filters
(0.2 μm) to save the column integrity. Another solution to this
problem may be the use of expanded beds (Chase and Draeger
1993; Fulton 1994), also called fluidized beds (see Fig.
3.7). In
principle, the use of expanded beds enables clarification,
concentration, and purification to be achieved in a single step.
The concept is to employ a particulate solid-phase adsorbent in an
open bed with upward liquid flow. The hydrodynamic drag around the
particles tends to lift them upwards, which is counteracted by
gravity because of a density difference between the particles and
the liquid phase. The particles remain suspended if particle
diameter, particle density, liquid viscosity, and liquid density
are properly balanced by choosing the correct flow rate. The
expanded bed allows particles (cells) to pass through, whereas
molecules in solution are selectively retained (e.g., by the use of
ion-exchange or affinity adsorbents) on the adsorbent particles.
Feedstocks can be applied to the bed without prior removal of
particulate material by centrifugation or filtration, thus reducing
process time and costs. Fluidized beds have been used previously
for the industrial-scale recovery of antibiotics such as
streptomycin and novobiocin (Fulton 1994; Chase 1994). Stable, expanded beds can be obtained
using simple equipment adapted from that used for conventional,
packed bed adsorption and chromatography processes. Ion-exchange
adsorbents are likely to be chosen for such separations.

Figure
3.7 ■
Contaminants
For pharmaceutical applications, product purity
mostly is ≥99 % when used as a parenteral (Berthold and Walter
1994; ICH 1999a). Purification processes should yield
potent proteins with well-defined characteristics for human use
from which “all” contaminants have been removed to a major extent.
The purity of the drug protein in the final product will therefore
largely depend upon the purification technology applied.
Table 3.4 lists potential contaminants that may be
present in recombinant protein products from bacterial and
nonbacterial sources. These contaminants can be host-related,
process-related and product-related. In the following sections,
special attention is paid to the detection and elimination of
contamination by viruses, bacteria, cellular DNA, and undesired
proteins.
Table
3.4 ■
Potential contaminants in recombinant
protein products derived from bacterial and nonbacterial
hosts.
Origin
|
Contaminant
|
---|---|
Host-related
|
Viruses
|
Host-derived proteins acid DNA
|
|
Glycosylation variants
|
|
N- and C-terminal variants
|
|
Endotoxins (from Gram-negative bacterial
hosts)
|
|
Product-related
|
Amino acids substitution and deletion
|
Denatured protein
|
|
Conformational isomers
|
|
Dimers and aggregates
|
|
Disulfide pairing variants
|
|
Deamidated species
|
|
Protein fragments
|
|
Process-related
|
Growth medium components
|
Purification reagents
|
|
Metals
|
|
Column materials
|
Viruses
Endogenous and adventitious viruses, which
require the presence of living cells to propagate, are potential
contaminants of animal cell cultures and, therefore, of the final
drug product. If present, their concentration in the purified
product will be very low and it will be difficult to detect them.
Viruses such as retrovirus can be visualized by (nonsensitive)
electron microscopy. For retroviruses, a highly sensitive RT-PCR
(reverse-transcriptase polymerase chain reaction) assay is
available, but for other viruses, a sensitive in vitro assay might
be lacking. The risks of some viruses (e.g., hepatitis virus) are
known (Walter et al. 1991;
Marcus-Sekura 1991), but there
are other viruses whose risks cannot be properly judged because of
lack of solid experimental data. Some virus infections, such as
parvovirus, can have long latent periods before their clinical
effects show up. Long-term effects of introducing viruses into a
patient treated with a recombinant protein should not be
overlooked. Therefore, it is required that products used
parenterally are free from viruses. The specific virus testing
regime required will depend on the cell type used for production
(Löwer 1990; Minor
1994).
Viruses can be introduced by nutrients, by an
infected production cell line, or they are introduced (by human
handling) during the production process. The most frequent source
of virus introduction is animal serum. In addition, animal serum
can introduce other unwanted agents such as bacteria, mycoplasmas,
prions, fungi, and endotoxins. It should be clear that appropriate
screening of cell banks and growth medium constituents for viruses
and other adventitious agents should be strictly regulated and
supervised (Walter et al. 1991; FDA 1993; ICH 1999b; WHO 2010). Validated, orthogonal methods to
inactivate and remove possible viral contaminants are mandatory for
licensing of therapeutics derived from mammalian cells or
transgenic animals (Minor 1994). Viruses can be inactivated by
physical and chemical treatment of the product. Heat, irradiation,
sonication, extreme pH, detergents, solvents, and certain
disinfectants can inactivate viruses. These procedures can be
harmful to the product as well and should therefore be carefully
evaluated and validated (Walter et al. 1992; Minor 1994; ICH 1999b). Removal of viruses by nanofiltration
is an elegant and effective technique and the validation aspects of
this technology are well described (PDA 2005). Filtration through 15 nm membranes
can remove even the smallest non-enveloped viruses like bovine
parvovirus (Maerz et al. 1996). Another common, although less robust,
method to remove viruses in antibody processes is by ion-exchange
chromatography. A number of methods for reducing or inactivating
viral contaminants are mentioned in Table 3.5 (Horowitz et al.
1991).
Table
3.5 ■
Methods for reducing or inactivating viral
contaminants.
Category
|
Types
|
Example
|
---|---|---|
Inactivation
|
Heat treatment
|
Pasteurization
|
Radiation
|
UV-light
|
|
Dehydration
|
Lyophilization
|
|
Cross linking agents, denaturating or
disrupting agents
|
β-propiolactone, formaldehyde, NaOH,
organic solvents (e.g., chloroform), detergents (e.g.,
Na-cholate)
|
|
Neutralization
|
Specific, neutralizing antibodies
|
|
Removal
|
Chromatography
|
Ion-exchange, immuno-affinity,
chromatography
|
Filtration
|
Nanofiltration
|
|
Precipitation
|
Cyroprecipitation
|
Bacteria
Unwanted bacterial contamination may be a problem
for cells in culture or during pharmaceutical purification. Usually
the size of bacteria allows simple filtration over 0.2 μm (or
smaller) filters for adequate removal. In order to further prevent
bacterial contamination during production, the raw materials used
have to be sterilized, preferably at 121 °C or higher, and the
products are manufactured under strict aseptic conditions wherever
possible. Production most often takes place in so-called clean
rooms in which the chances of environmental contamination is
reduced through careful control of the environment, for example,
filtration of air. Additionally, antibiotic agents can be added to
the culture media in some cases but have to be removed further
downstream in the purification process. However, the use of
beta-lactam antibiotics such as penicillin is strictly prohibited
due to oversensitivity of some individuals to these compounds.
Because of the persistence of antibiotic residues, which are
difficult to eliminate from the product, appropriately designed
manufacturing plants and extensive quality control systems for
added reagents (medium, serum, enzymes, etc.) permitting
antibiotic-free operation are preferable.
Pyrogens (usually endotoxins of gram-negative
bacteria) are potentially hazardous substances (see Chap. 4). Humans are sensitive to
pyrogen contamination at very low concentrations (picograms per
mL). Pyrogens may elicit a strong fever response and can even be
fatal. Simple 0.2 μm filtration does not remove pyrogens. Removal
is complicated further because pyrogens vary in size and chemical
composition. However, sensitive tests to detect and quantify
pyrogens are commercially available. Purification schemes usually
contain at least one step of ion-exchange chromatography
(anionic-exchange material) to remove the negatively charged
endotoxins (Berthold and Walter 1994). Furthermore, materials used in process
such as glassware are typically subjected to a depyrogenation step
prior to use often by the use of elevated temperatures (≥180 °C) in
a dry heat oven.
Cellular DNA
The application of continuous mammalian cell
lines for the production of recombinant proteins might result in
the presence of oncogene-bearing DNA fragments in the final protein
product (Walter and Werner 1993; Löwer 1990). A stringent purification protocol
that is capable of reducing the DNA content and fragment size to a
safe level is therefore necessary (Berthold and Walter
1994; WHO 2010). There are a number of approaches
available to validate that the purification method removes cellular
DNA and RNA. One such approach involves incubating the cell line
with radiolabeled nucleotides and determining radioactivity in the
purified product obtained through the purification protocol. Other
methods are dye-binding fluorescence-enhancement assays for
nucleotides and PCR-based methods. If the presence of nucleic acids
persists in a final preparation, then additional steps must be
introduced in the purification process. The question about a safe
level of nucleic acids in biotech products is difficult to answer
because of minimal relevant know-how. Transfection with so-called
naked DNA is very difficult and a high concentration of DNA is
needed. Nevertheless, it is agreed for safety reasons that final
product contamination by nucleic acids should not exceed 100 pg or
10 ng per dose depending on the type of cells used to produce the
pharmaceutical (WHO 2010;
European Pharmacopoeia 2011).
Protein Contaminants and Product Variants
As mentioned before, minor amounts of host-,
process-, and product-related proteins will likely appear in
biotech products. These types of contaminants are a potential
health hazard because, if present, they may be recognized as
antigens by the patient receiving the recombinant protein product.
On repeated use the patient may show an immune reaction caused by
the contaminant, while the protein of interest is performing its
beneficial function. In such cases the immunogenicity may be
misinterpreted as being due to the recombinant protein itself.
Therefore, one must be very cautious in interpreting safety data of
a given recombinant therapeutic protein.
Generally, the sources of host- and
process-related protein contaminants are the growth medium used and
the host proteins of the cells. Among the host-derived
contaminants, the host species’ version of the recombinant protein
could be present (WHO 2010).
As these proteins are similar in structure, it is possible that
undesired proteins are co-purified with the desired product. For
example, urokinase is known to be present in many continuous cell
lines. The synthesis of highly active biological molecules such as
cytokines by hybridoma cells might be another concern (FDA
1990). Depending upon their
nature and concentration, these cytokines might enhance the
antigenicity of the product.
“Known” or expected contaminants should be
monitored at the successive stages in a purification process by
suitable in-process controls, e.g., sensitive immunoassay(s).
Tracing of the many “unknown” cell-derived proteins is more
difficult. When developing a purification process, other
less-specific analyses such as SDS-PAGE are usually used in
combination with various staining techniques.
Product-related contaminants/variants may pose a
potential safety issue for patients. These contaminants can be
aggregated forms of the product, a product with heterogeneity in
the disulfide bridges, N- and C-terminal variants, glycosylation
variants, deamidated product, etc. Some of these
contaminants/variants are described in the following
paragraphs.
N- and C-Terminal Heterogeneity
A major problem connected with the production of
biotech products is the problem associated with the amino
(NH2)-terminus of the protein, e.g., in E. coli systems, where
protein synthesis always starts with methylmethionine. Obviously,
it has been of great interest to develop methods that generate
proteins with an NH2-terminus as found in the authentic protein.
When the proteins are not produced in the correct way, the final
product may contain several methionyl variants of the protein in
question or even contain proteins lacking one or more residues from
the amino terminus. This is called the amino-terminal
heterogeneity. This heterogeneity can also occur with recombinant
proteins (e.g., α-interferon) susceptible to proteases that are
either secreted by the host or introduced by serum-containing
media. These proteases can clip off amino acids from the C-terminal
and/or N-terminal of the desired product (amino- and/or
carboxy-terminal heterogeneity). Amino- and/or carboxy-terminal
heterogeneity is not desirable since it may cause difficulties in
purification and characterization of the proteins. In case of the
presence of an additional methionine at the N-terminal end of the
protein, its secondary and tertiary structure can be altered. This
could affect the biological activity and stability and may make it
immunogenic. Moreover, N-terminal methionine and/or internal
methionine is sensitive to oxidation (Sharma 1990).
Conformational Changes/Chemical Modifications
Although mammalian cells are able to produce
proteins structurally equal to endogenous proteins, some caution is
advisable. Transcripts containing the full-length coding sequence
could result in conformational isomers of the protein because of
unexpected secondary structures that affect translational fidelity
(Sharma 1990). Another factor
to be taken into account is the possible existence of equilibria
between the desired form and other forms such as dimers. The
correct folding of proteins after biosynthesis is important because
it determines the specific activity of the protein (Berthold and
Walter 1994). Therefore, it is
important to determine if all molecules of a given recombinant
protein secreted by a mammalian expression system are folded in
their native conformation. In some cases, it may be relatively easy
to detect misfolded structures, but in other cases, it may be
extremely difficult. Apart from conformational changes, proteins
can undergo chemical alterations, such as proteolysis, deamidation,
and hydroxyl and sulfhydryl oxidations during the purification
process. These alterations can result in (partial) denaturation of
the protein. Vice versa, denaturation of the protein may cause
chemical modifications as well (e.g., as a result of exposure of
sensitive groups).
Glycosylation
Many therapeutic proteins produced by recombinant
DNA technology are glycoproteins (Sharma 1990). The presence and nature of
oligosaccharide side chains in proteins affect a number of
important characteristics, like the proteins’ serum half-life,
solubility, and stability, and sometimes even the pharmacological
function (Cumming 1991).
Darbepoetin, a second-generation, genetically modified
erythropoietin, has a carbohydrate content of 80 % compared to 40 %
for the native molecule, which increases the in vivo half-life
after intravenous administration from 8 h for erythropoietin to 25
h for darbepoetin (Sinclair and Elliott 2005). As a result, the therapeutic profile
may be “glycosylation” dependent. As mentioned previously, protein
glycosylation is not determined by the DNA sequence. It is an
enzymatic modification of the protein after translation and can
depend on the environment in the cell. Although mammalian cells are
very well able to glycosylate proteins, it is hard to fully control
glycosylation. Carbohydrate heterogeneity can occur through the
size of the chain, type of oligosaccharide, and sequence of the
carbohydrates. This has been demonstrated for a number of
recombinant products including interleukin-4, chorionic
gonadotropin, erythropoietin, and tissue plasminogen activator.
Carbohydrate structure and composition in recombinant proteins may
differ from their native counterparts, because the enzymes required
for synthesis and processing vary among different expression
systems (e.g., glycoproteins in insect cells are frequently smaller
than the same glycoproteins expressed in mammalian cells) or even
from one mammalian system to another.
Proteolytic Processing
Proteases play an important role in processing,
maturation, modification, or isolation of recombinant proteins.
Proteases from mammalian cells are involved in secreting proteins
into the cultivation medium. If secretion of the recombinant
protein occurs co-translationally, then the intracellular
proteolytic system of the mammalian cell should not be harmful to
the recombinant protein. Proteases are released if cells die or
break (e.g., during cell break at cell harvest) and undergo lysis.
It is therefore important to control growth and harvest conditions
in order to minimize this effect. Another source of proteolytic
attack is found in the components of the medium in which the cells
are grown. For example, serum contains a number of proteases and
protease zymogens that may affect the secreted recombinant protein.
If present in small amounts and if the nature of the proteolytic
attack on the desired protein is identified, appropriate protease
inhibitors to control proteolysis could be used. It is best to
document the integrity of the recombinant protein after each
purification step.
Proteins become much more susceptible to
proteases at elevated temperatures. Purification strategies should
be designed to carry out all the steps at 2–8 °C (Sharma
1990) if proteolytic
degradation occurs. Alternatively, Ca2+ complexing agents (e.g.,
citrate) can be added as many proteases depend on Ca2+ for their
activity. From a manufacturing perspective, however, providing
cooling to large-scale chromatographic processes, although not
impossible, is a complicating factor in the manufacturing
process.
Bacteria: Protein Inclusion Body Formation
In bacteria soluble proteins can form dense,
finely granular inclusions within the cytoplasm. These “inclusion
bodies” often occur in bacterial cells that overproduce proteins by
plasmid expression. The protein inclusions appear in electron
micrographs as large, dense bodies often spanning the entire
diameter of the cell. Protein inclusions are probably formed by a
buildup of amorphous protein aggregates held together by covalent
and non-covalent bonds. The inability to measure inclusion body
proteins directly may lead to the inaccurate assessment of recovery
and yield and may cause problems if protein solubility is essential
for efficient, large-scale purification (Berthold and Walter
1994). Several schemes for
recovery of proteins from inclusion bodies have been described. The
recovery of proteins from inclusion bodies requires cell breakage
and inclusion body recovery. Dissolution of inclusion proteins is
the next step in the purification scheme and typically takes place
in extremely dilute solutions which tend to have the effect of
increasing the volumes of the unit operations during the
manufacturing phases. This can make process control more difficult
if, for example, low temperatures are required during these steps.
Generally, inclusion proteins dissolve in denaturing agents such as
sodium dodecyl sulfate (SDS), urea, or guanidine hydrochloride.
Because bacterial systems generally are incapable of forming
disulfide bonds, a protein containing these bonds has to be
refolded under oxidizing conditions to restore these bonds and to
generate the biologically active protein. This so-called
renaturation step is increasingly difficult if more S-S bridges are
present in the molecule and the yield of renatured product could be
as low as only a few percent. Once the protein is solubilized,
conventional chromatographic separations can be used for further
purification of the protein.
Aggregate formation at first sight may seem
undesirable, but there may also be advantages as long as the
protein of interest will unfold and refold properly. Inclusion body
proteins can easily be recovered to yield proteins with >50 %
purity, a substantial improvement over the purity of soluble
proteins (sometimes below 1 % of the total cell protein).
Furthermore, the aggregated forms of the proteins are more
resistant to proteolysis, because most molecules of an aggregated
form are not accessible to proteolytic enzymes. Thus the high yield
and relatively cheap production using a bacterial system can offset
a low-yield renaturation process. For a non-glycosylated, simple
molecule, this still is the production system of choice.
Commercial-Scale Manufacturing and Innovation
A major part of the recombinant proteins on the
market consist of monoclonal antibodies produced in mammalian
cells. Pharmaceutical production processes have been set up since
the early 1980s of the twentieth century. These processes
essentially consist of production in stirred tanks bioreactors,
clarification using centrifugation, and membrane technology,
followed by protein A capture, low-pH virus inactivation,
cation-exchange and anion-exchange chromatography, virus
filtration, and UF/DF for product formulation (Shukla and Thömmes
2010). Such platform processes
run consistently at very large scale (e.g., multiple 10,000 L
bioreactors and higher). Product recovery is generally very high
(>70 %). Since product titers in the bioreactors have increased
to a level where further increases have no or a minimal effect on
the cost of goods, and the cost of goods are a fraction of the
sales price, the focus of these large-scale processes is changing
from process development to process understanding (Kelley
2009). However, it is also
anticipated that the monoclonal antibody demands may decrease due
to more efficacious products like antibody-drug conjugates and the
competition with generic products. A lower demand together with the
increase in recombinant protein yields will lead to a decrease in
bioreactor size, an increase in the need for flexible facilities,
and faster turnaround times leading to a growth in the use of
disposables and other innovative technologies (Shukla and Thömmes
2010).
Self-Assessment Questions
Questions
1.
Name four types of different bioreactors.
2.
Chromatography is an essential step in the
purification of biotech products. Name at least five different
chromatographic purification methods.
3.
What are the major safety concerns in the
purification of cell-expressed proteins?
4.
What are the critical issues in production and
purification that must be addressed in process validation?
5.
Mention at least five issues to consider in the
cultivation and purification of proteins.
6.
True or false: Glycosylation is an important
posttranslational change of pharmaceutical proteins. Glycosylation
is only possible in mammalian cells.
7.
Glycosylation may affect several properties of
the protein. Mention at least three possible changes in case of
glycosylation.
8.
Pharmacologically active biotech protein products
have complex three-dimensional structures. Mention two or more
important factors that affect these structures.
Answers
1.
Stirred-tank, airlift, microcarrier, and membrane
bioreactors
2.
Adsorption chromatography, ion-exchange
chromatography, affinity chromatography, hydrophobic interaction
chromatography, gel-permeation or size-exclusion
chromatography
3.
Removal of viruses, bacteria, protein
contaminants, and cellular DNA
4.
Production: scaling up and design. Purification:
procedures should be reliable in potency and quality of the product
and in the removal of viral, bacterial, DNA, and protein
contaminants
5.
Grade of purity, pyrogenicity, N- and C-terminal
heterogeneity, chemical modification/conformational changes,
glycosylation, and proteolytic processing
6.
False; glycosylation is also possible in yeast
(and to some degree in bacteria)
7.
Solubility, pKa, charge, stability, and
biological activity
8.
Amino acid structure, hydrogen and sulfide
bridging, posttranslational changes, and chemical modification of
the amino acid rest groups
References
Afeyan N, Gordon N,
Mazsaroff I, Varady L, Fulton S, Yang Y, Regnier F (1989)
Flow-through particles of the high-performance liquid
chromatographic separation of biomolecules, perfusion
chromatography. J Chromatogr 519:1–29
Benedek K, Swadesh JK
(1991) HPLC of proteins and peptides in the pharmaceutical
industry. In: Fong GW, Lam SK (eds) HPLC in the pharmaceutical
industry. Dekker, New York, pp 241–302
Borman S (2006)
Glycosylation engineering. Chem Eng News 84:13–22
Cartwright T (1987)
Isolation and purification of products from animal cells. Trends
Biotechnol 5:25–30CrossRef
Celik E, Calik P (2011)
Production of recombinant proteins by yeast cells. Biotechnol Adv.
doi:10.1016/j.biotechadv.2011.09.011
PubMed
Chase H, Draeger N (1993)
Affinity purification of proteins using expanded beds. J Chromatogr
597:129–145
Compton B, Jensen J
(2007) Use of perfusion technology on the Rise – New modes are
beginning to gain ground on Fed-Batch strategy. Genetic Engineering
& Biotechnology News 27(17):48
Dell A, Galadari A,
Sastre F, Hitchen P (2011) Similarities and differences in the
glycosylation mechanisms in prokaryotes and eukaryotes. Int J
Microbiol 2010(2010):148178
EMA (2011) Note for
guidance on minimising the risk of transmitting animal spongiform
encephalopathy agents via human and veterinary medicinal products
(EMA/410/01 rev.3)
European Pharmacopoeia
Seventh Edition. Strasbourg: Council of Europe (2011) 5.2.3. Cell
substrates for the production of vaccines for human use
FD3A, Center for
Biologics Evaluation and Research (1990) Cytokine and growth factor
pre-pivotal trial information package with special emphasis on
products identified for consideration under 21 CFR 312 Subpart E.
Bethesda
FDA, Office of
Biologicals Research and Review (1993) Points to consider in the
characterization of cell lines used to produce biologicals.
Rockville Pike/Bethesda
Gottschalk U (2006) The
renaissance of protein purification. BioPharm Int 19(6):S8–S9
Hjerten S, Mohammed J,
Nakazato K (1993) Improvement in flow properties and pH stability
of compressed, continuous polymer beds for high-performance liquid
chromatography. J Chromatogr 646:121–128CrossRef
Hodge G (2004)
Disposable components enable a new approach to biopharmaceutical
manufacturing. BioPharm Int 2004(15):38–49
Horowitz MS, Bolmer SD,
Horowitz B (1991) Elimination of disease-transmitting enveloped
viruses from human blood plasma and mammalian cell culture
products. Bioseparation 1:409–417
ICH (International
Conference on Harmonization) Topic Q6B (1999a) Specifications: test
procedures and acceptance criteria for biotechnology/biological
products
ICH (International
Conference on Harmonization) Topic Q5A (1999b) Viral safety
evaluation of biotechnology products derived from cell lines of
human or animal origin
International
Conference on Harmonization guideline M7 on assessment and control
of DNA reactive (mutagenic) impurities in pharmaceuticals to limit
potential carcinogenic risk
James AM (1992)
Introduction fundamental techniques. In: James AM (ed) Analysis of
amino acids and nucleic acids. Butterworth-Heinemann, Oxford, pp
1–28
Jones K (1990) Affinity
chromatography, a technology up-date. Am Biotechnol Lab
8:26–30PubMed
Klegerman ME, Groves MJ
(1992) Pharmaceutical biotechnology. Interpharm Press, Inc.,
Buffalo Grove
Marcus-Sekura CJ (1991)
Validation and removal of human retroviruses. Center for Biologics
Evaluation and Research, FDA, Bethesda
Monteclaro F (2010)
Protein expression systems, ringing in the new. Innov Pharm Technol
12:45–49
Note for Guidance (1991)
Validation of virus removal
and inactivation procedure, Ad Hoc Working Party on
Biotechnology/Pharmacy, European Community, DG III/8115/89-EN
PDA Journal of
Pharmaceutical Science and Technology (2005) Technical report No.
41, Virus filtration, 59, No. S-2
Sadana A (1989) Protein
inactivation during downstream separation, part I: the processes.
Biopharm 2:14–25
Sharma SK (1990) Key
issues in the purification and characterization of recombinant
proteins for therapeutic use. Adv Drug Deliv Rev
4:87–111CrossRef
Tennikova T, Svec F
(1993) High performance membrane chromatography: highly efficient
separation method for proteins in ion-exchange, hydrophobic
interaction and reversed phase modes. J Chromatogr
646:279–288CrossRef
Van Wezel AL, van der
Velden-de Groot CA, de Haan HH, van den Heuvel N, Schasfoort R
(1985) Large scale animal cell cultivation for production of
cellular biologicals. Dev Biol Stand 60:229–236PubMed
Walsh C (2006)
Posttranslational modification of proteins: expanding nature’s
inventory, vol xxi. Roberts and Co. Publishers, Englewood, p
490
Walter J, Werner RG
(1993) Regulatory requirements and economic aspects in downstream
processing of biotechnically engineered proteins for parenteral
application as pharmaceuticals. In: Kroner KH, Papamichael N,
Schütte H (eds) Downstream processing, recovery and purification of
proteins, a handbook of principles and practice. Carl Hauser
Verlag, Muenchen
Walter J, Werz W, McGoff
P, Werner RG, Berthold W (1991) Virus removal/inactivation in
downstream processing. In: Spier RE, Griffiths JB, MacDonald C
(eds) Animal cell technology: development, processes and products.
Butterworth-Heinemann Ltd. Linacre House, Oxford, pp 624–634
Walter K, Werz W,
Berthold W (1992) Virus removal and inactivation, concept and data
for process validation of downstream processing. Biotech Forum
Europe 9:560–564
Wheelwright SM (1993)
Designing downstream processing for large scale protein
purification. Biotechnology 5:789–793
WHO (World Health
Organization) (2010) Recommendations for the evaluation of animal
cell cultures as substrates for the manufacture of biological
medicinal products and for the characterization of cell banks.
Technical report series, proposed replacement of 878, annex 1 (not
yet published)