You are on page 1of 12

Downstream Processing REVIEW 87

Downstream Processing in the Biotechnology Industry


Manohar Kalyanpur

Abstract
The biotechnology industry today employs recombinant bacteria, mammalian cells, and transgenic ani-
mals for the production of high-value therapeutic proteins. This article reviews the techniques employed in
this industry for the recovery of these products. The methods reviewed extend from the centrifugation and
membrane filtration for the initial clarification of crude culture media to the final purification of the products
by a variety of membrane-based and chromatographic methods. The subject of process validation including
validation of the removal of bacterial and viral contaminants from the final products is also discussed with
special reference to the latest regulatory guidelines.
Index Entries : bacteria; mammalian cell cultures; centrifuges; homogenizers ; tangential flow filtration;
ultrafiltration; expanded bed adsorption; chromatography methods; vaccines; bacterial and viral clearance;
endotoxin removal; process validation.

1. Introduction high-value medicinal products. In recent years


As the 21st century begins, we look with awe at geneticists have succeeded in breeding transgenic
the accomplishments of new technologies in the sheep, goats, and cattle (1,2) and methods have
last century, especially during the last 20 years. been developed to get these animals to express the
Here, molecular biology has made truly signifi- desired drug products in their milk. The scientists
cant contributions toward the development of new insert specific DNA sequences for a therapeutic
biopharmaceuticals. We have in our hands today protein into the genetic material of an animal
the entire human genome and this is indeed a embryo. The transgenic offspring of this animal
major milestone in biological sciences. This then produces the protein of interest in its milk
information will provide clues for researchers to from where the product is isolated and purified.
develop new drugs that will cure deficiencies and The technique offers a commercially viable alter-
not simply treat their symptoms, as the older phar- native to the classical methods of manufacuring
maceuticals did. The biotechnology industry will recombinant proteins of therapeutic value to treat
strive harder than ever before to develop new chronic diseases such as rheumatoid arthritis, car-
biotherapeutics, and among these new drugs, pro- diovascular problems, numerous cancers, and cer-
teins and glycoproteins will make up the major tain autoimmune diseases. The method has the
part of the targeted drugs. Since the first mono- added advantage that it eliminates the need for
clonal antibody was licensed for sale in 1986, expensive bioreactors and other capital equipment
eight more antibody products have already required in the traditional methods of manufac-
reached the market, while several more are in dif- turing biotechnology products. Another interest-
ferent stages of development in companies around ing and even more recent development is the
the world. success of some companies who have developed
The science of biotechnology covers the exploi- processes for the large-scale production of recom-
tation of microorganisms and mammalian and binant proteins in genetically transformed plants
insect cell cultures, which are a major source of (3) such as transgenic corn. This method is
*Author to whom all correspondence and reprint requests should be addressed: Bioseparations and Pharmaceutical Validation, 10 Rue Odilon
Redon, 78370 Plaisir, France.
Molecular Biotechnology 2002 Humana Press Inc. All rights of any nature whatsoever reserved. 1073–6085/2002/22:1/87–98/$13.00

MOLECULAR BIOTECHNOLOGY 87 Volume 22, 2002


88 Kalyanpur

expected to give an economic advantage for drug as a soluble protein but it is quite often present in
manufacture. the form of an insoluble refractive mass referred
Most of the biotechnology proteins are present to as “inclusion bodies.” The recovery of the
in complex mixtures of products and this makes entire biomass including the cells is performed by
the task of purifying these molecules very diffi- either preparative centrifugation or by means of
cult. If these were nonprotein molecules, like an- tangential flow filtration systems using micro-
tibiotics, for example, one could use solvent porous membranes of appropriate pore diameters.
extraction to isolate the compounds from solutions The different filter manufacturers such as
in which they are present, making the task of puri- Millipore Corporation (Bedford, MA) (5), Pall
fication much easier. This is quite a challenge to Corporation (Port Washington, N.Y), and other
the biochemists, chemical engineers, and other companies offer membranes with pore diameters
personnel in the downstream processing depart- of 0.22, 0.45, and 0.65 µm, and the process devel-
ments of the industry (4). They use several opment scientists must select the membranes
diverse purification methods in the research labo- suited to their specific needs of biomass concen-
ratory at the bench scale to achieve a high level of tration. The particulates from the process fluids
purity, and these methods are eventually scaled up can get into the membrane pores and cause a sig-
to the pilot and production levels. The techniques nificant drop in filtration rates. However, the phe-
are used in a complementary fashion to develop nomenon can be controlled by fine tuning the
cost effective methods that help the companies to process conditions to obtain the optimum feed and
produce drugs of the level of purity demanded by permeate flow rates and transmembrane pressures.
the regulatory authorities. The industry today During cell harvesting, either intermittant or
manufactures on a commercial scale compounds continuous cell washing (also referred to as
that would otherwise have been difficult, if not diafiltration) can be performed by adding a suit-
impossible, to produce in large enough quantities able solution or a buffer to the cell concentrate,
to meet the needs of the sick population. This which also helps to maintain the desired pH or
article attempts to give the reader an overview of ionic strength of the cell suspension and prevents
the techniques available for downstream purifica- cell lysis. The main advantage of diafiltration is
tion of biotechnology products. that it helps to wash away the soluble impurities
from the process stream. This step is usually
2. Production Methods in the
started when the cell concentration reaches a point
Biotechnology Industry
where rapid flux decay is observed.
As mentioned earlier, the industry manufac-
tures drug products by different methods and their 2.2. Cell Lysis and Clarification of the Lysate
downstream processing varies not only from prod- When host cells such as Escherichia coli
uct to product but also varies depending upon the express the desired protein in high concentration,
expression system used for the product manufac- it is converted into micron size inclusion bodies
ture. Each process therefore needs to be fine tuned within the cytoplasm of the bacterial cells. These
depending on the particular method of drug manu- bodies usually contain other proteins from the
facture, the process stream from which it is recov- cells as well as nucleic acids and portions of the
ered, and the chemical properties of the product. cell envelope. Valax and Georgion (6) report that
these contaminants adhere to the surface of the
2.1. Products Made by Recombinant inclusion bodies. They are released from the cyto-
Bacterial Fermentation plasm in subsequent processing steps. The forma-
The first step in these processes is the separa- tion of these bodies is a specific process that, in
tion of the biomass from its surrounding broth. fact, helps to ease the downstream purification of
The protein is expressed within the bacterial cell the protein of interest, since it exists in a relatively

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


Downstream Processing 89

pure form separate from a great majority of the protein of interest within them. Continuous cen-
cellular contaminants. trifugation separates the dense inclusion bodies
The inclusion bodies are released from within from the rest of the contaminants. The method is
the bacterial cells during mechanical disruption of described in detail by Middleberg (12). Both small
the cells using homogenizers of different types. and large production size centrifuges are available
Homogenizers such as those manufactured by from two principal suppliers: the German manu-
Niro-Soavi (Parma, Italy), APV-Rennie (Copen- facturer, Westfalia Separator AG, and Alfa Laval
hagen, Denmark), and APV-Gaullin (Wilmington, Separation AB of Sweden. The centrifuges are
MA) are the principal types in use. Most homog- operated to obtain a good separation of the inclu-
enizers include double-seal designs in order sion bodies. Next the bodies are washed and
to prevent accidental product loss and can be dissolved in a strong denaturing agent such as urea
steam sterilized. They also have a simplified or guanidine sulfate, followed by protein refold-
design to facilitate cleaning and sanitization of the ing. The final purification is accomplished by any
system after use. Middleberg (7) provides a com- number of filtration and/or chromatography steps
prehensive review of this aspect of downstream to recover the protein of interest.
processing.
2.4. Membrane Filtration of Cell Lysate
Homogenization of bacterial cells can be made
more efficient by chemical treatment of the cells In recent years there has been a lot of interest in
prior to homogenization. The strategy is to the use of membrane filtration in the pharmaceu-
weaken the bacterial cell walls by attacking the tical and food industries, and this has been
structural elements that give strength to the cell extended with much success in the biotechnology
walls. Vogels and Kula (8) report that the use of industry. Membrane filters are useful in both clari-
the lytic enzyme cellosyl helps to increase cell dis- fying and sterilizing gases and aqueous solutions
ruption from about 50% to almost 100%. A mix- introduced into bioreactors and during down-
ture of ethylenediaminetetraacetic acid (EDTA) stream recovery and purification of products from
and lysozyme also improves the efficiency of cell a variety of process streams (13). The technolo-
breakage (9). Cell walls of the yeasts Saccharo- gist can today choose from among membranes
myces cerevisiae and Candida utilis can be weak- made from ceramic or organic polymeric materi-
ened by pretreatment with zymolase preparation als, and the membranes can be cast with different
(10,11) available from Seikagaku America of pore geometries and sizes. The ultimate selection
Rockville, MD. However, the use of pretreatment of the membrane for a particular application
enzymes can add to the overall cost of processing. depends on its durability, cost, and suitability for
It is therefore important to calculate the contribu- the intended separation. Also membranes are
tion of the cost increase to the improvement of packed in different filter configurations (14) such
yield. Quite often product yield can be improved as the depth filters, which are operated in the nor-
by repeated homogenization passes to obtain a mal flow filtration (NFF) mode, and the hollow
more complete cell disruption. But this has a hid- fiber, spiral wrap, flat plate, and frame and tubu-
den drawback. The overall rise in temperature dur- lar modules, all of which are operated in the cross
ing repeated homogenization can denature the flow or tangential flow filtration mode (TFF). The
protein, thereby lowering the final product yield. TFF mode is the most commonly used method in
downstream processing applications such as clari-
2.3 Lysate Clarification by Centrifugation fication of process streams using the microporous
The cell lysate is usually clarified by either cen- membranes and in the separation of biomolecules
trifugation or by tangential flow filtration. The of different sizes from each other using the ultra-
lysate contains the cell debris, soluble proteins filtration membranes. The topic of tangential flow
from host cells, and the inclusion bodies with the filtration has been discussed in depth by Michaels

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


90 Kalyanpur

and his numerous coworkers from Millipore Cor- typically grown in roller bottles for small-volume
poration (15). applications as in research and product develop-
In the filtration of cell lysate for clarification, ment stages and in bioreactors of varying sizes for
the suspended material is retained at the mem- production purposes. Whatever the batch size, the
brane surface and the clear solution with the first step toward product isolation is the separation
soluble components ends up in the permeate. The of the producing cells, cell debris, and other par-
microporous membranes with smaller pore diam- ticulate impurities from the process stream to ob-
eters such as 0.2 µm perform better than those tain the clear extracellular fluid for product
with larger pores, since the large pores are more isolation. Either TFF or NFF methods are com-
readily plugged by the cell debris. Sometimes monly employed for the clarification process, and
ultrafiltration membranes with even smaller pores the choice of the method depends mainly on the size
perform better than the 0.2 µm microporous mem- of a batch and the frequency of the operation (5).
branes where the debris is prevented from getting With small volume batches and infrequent pro-
into the pores. This helps to avoid a rapid flow cessing, the NFF method is often the preferred
decay. However, the flow rates through the UF method. The filters used here cost much less than
membranes are, in general, lower than those the TFF modules and do not require the capital
obtained with microporous membranes. If the pro- investment in expensive hardware. Moreover,
tein of interest is in the soluble fraction it passes since these are of the disposable type, there is no
through the membrane in the permeate fraction need to perform the routine time consuming clean-
and the solid impurities including the cell debris ing operation nor does one need to validate the
are retained upstream of the membrane filter. filter cleaning and reuse procedure. On the other
If the product is present as inclusion bodies, it hand, TFF modules are expensive but are robust
is in the retentate fraction from the step pre- enough to permit multiple use to justify their cost.
ceeding. It is solubilized by the addition of urea or These are cleaned and sanitized thoroughly after
guanidine sulfate and then separated from the rest each use, but the procedure must be validated to
of the contaminating proteins using ultrafiltration satisfy regulatory requirements. Overall, the TFF
membranes with an appropriate cut-off. There is method is preferred for large and frequent produc-
often the risk that the membranes might retain tion cycles because the costs of the membrane
some large aggregrates of the product of interest modules and the hardware are spread over produc-
and care must be taken to avoid this in order to tion of much larger quantities of the drug product.
prevent product loss. The product yields can be The protein of interest in fermentations with
improved by washing the retentate fraction with a mammalian cell cultures are in the extracellular
buffer or another suitable solvent. The combined fraction. If the cells are lysed during the separa-
pool of the permeate and washings contains the tion phase, the intracellular proteins spill out of
protein of interest, which is then sent to the subse- the cells and contaminate the extracellular fluid.
quent steps of purification where either membrane The fragile mammalian cells therefore need care-
or chromatography methods are employed. If at ful handling if membrane filtration is employed.
this point the process stream is too dilute, it is con- An elevated transmembrane pressure and high fil-
centrated to the desired level, which is adequately tration rate can rupture the fragile cells. An
performed by ultrafiltration, but any other method excellent filtration system for this application was
that is better suited may also be used. developed by Millipore Corporation in the 1980’s.
The system consists of a microporous membrane,
2.5. Harvesting Mammalian Cell Cultures usually with 0.45 µm pore diameters and a recir-
Mammalian cells are grown by several methods culating feed pump as in the conventional TFF
and in small and large batches for the production of systems. But a second pump on the permeate side
diagnostic and therapeutic proteins. The cells are replaces the customary valve used for restricting

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


Downstream Processing 91

the permeate flow. The second pump permits 3.1 Ultrafiltration


accurate control of the permeate flux and helps to This method is commonly employed to concen-
maintain low transmembrane pressures to avoid trate proteins of different molecular weights while
damage to the fragile cells. The method permits removing smaller molecules such as salts, sugars,
recovery of the desired product of good purity in and sometimes small or large peptides from the
high yields (15). Here, too, diafiltration with an product. The concentration of viruses and viral
appropriate buffer improves product yields. antigens in the manufacture of vaccines is a typi-
2.6 Concentration of Viruses for Vaccine cal example. A very wide range of molecular
Production weight cut-offs is available from several suppli-
ers. Prior knowledge of the molecular weight of
The first step in the manufacture of viral vac- the protein of interest helps in choosing the mem-
cines and antigens is the concentration of the brane best suited for the process. Here again
viruses or portions of the viral coat. This is best diafiltration is employed to wash off the impuri-
performed by ultrafiltration with membranes hav- ties. In short, the ultrafiltration step helps to
ing a cut-off of 100 kDa but other cut-offs such as enrich the retentate in the higher-molecular-
30 kDa, or even 10 kDa may be suitable in spe- weight species while the permeate contains all the
cific cases. This is why membrane selection is best smaller molecules and the solvent.
done on a case by case basis. The selected mem-
brane must retain the virus and the viral antigens 3.2. High Resolution Tangential Flow
while permitting the passage into the permeate of Filtration
water, salts, and other impurities with molecular Tangential flow filtration with ultrafiltration
weights smaller than the cut-off of the selected membranes is used to concentrate proteins and
membrane. The use of diafiltration during concen- other large molecules in aqueous streams, with the
tration helps to wash away contaminants of lower simultaneous removal of the lower-molecular-
molecular weights into the permeate with simul- weight species, salts, and water. However, the
taneous purification. Ultrafiltration membranes in method suffers from its inability to fractionate
the TFF mode are employed in the manufacture of mixtures of molecules that have similar molecu-
several vaccines including those used against lar weights. The chromatographic purifications
influenza, Epstein-Barr syndrome, and measles differentiate molecules based on their chemical
(16–18). properties and not simply on their size, and these
3. Further Purification of Biotechnology techniques greatly contribute in a large way to the
manufacture of high-purity drug products in the
Products
biopharmaceutical industry.
Once the desired protein is obtained in a par- The ultrafiltration membranes normally sepa-
ticulate-free process stream, the task of product rate molecules, which differ in their molecular
purification begins and continues until the desired diameters by a factor of between 5X and 10X. But
level of purity is reached. The yield in each step is this factor of separating efficiency can be further
critical because even a small product loss in each improved by employing a method referred to as
purification step adds up to significant losses over high-resolution tangential flow filtration (HRTF)
the entire downstream process, especially if sev- (19). The method aims at separating, in a single-
eral purification steps are required to reach a high stage process, two compounds that differ in
level of purity. The techniques employed usually molecular diameter by a factor of about 3X–5X.
fall into two major groups: membrane-based Diafiltration helps better removal of the smaller
methods such as ultrafiltration and nanofiltration molecule in the permeate and increases its yield
(reverse osmosis) and a variety of chromatogra- while at the same time it improves the purity of the
phy procedures. larger molecule held back in the retentate fraction.

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


92 Kalyanpur

The HRTF system includes two pumps on 3.4. Expanded Bed Adsorption
either side of the membrane module, the conven- Chromatography
tional feed pump and a permeate side pump, which This relatively recent technique is used for whole
is made to operate as a metering pump. The system broth processing and permits the isolation of thera-
is not designed to maximize volumetric flux but to peutic proteins from crude cell culture broths or
maximize solute mass flux by increasing the pas- natural extracts. (20–23). This unique chromato-
sage of the solute through the membrane. The sys- graphic method can often combine into one single
tem performs best with low-binding membranes to operation several steps such as centrifugation, fil-
reduce product loss by absorption to the membrane. tration, concentration, and purification. The versa-
The value of the HRTF system is demonstrated in tile procedure is economical and time saving and
the separation of IgG and IgM from albumin in the can give high yields of the desired protein.
plasma fractionation industry. The system is also In traditional chromatography the adsorbent
used to remove polyethylene glycols from proteins columns are tightly packed. In contrast, expanded
and serum or allantoic proteins from viruses. bed adsorption permits expansion of the adsorbent
3.3. Nanofiltration or Reverse Osmosis due to the upward flow of column fluid and crude
raw materials such as fermentation broths pass
The reverse osmosis membranes can retain freely through the column without clogging,
low-molecular-weight compounds such as salts, which is very often observed in packed bed col-
sugars, and small peptides. These membranes umns. The protein of interest is adsorbed directly
were originally developed for desalination of on the fluidized column adsorbent, which is stabi-
seawater to make potable water, but they have lized toward uncontrolled turbulence and back
found a small niche of applications in the bio- mixing with the optimal design of the column and
pharmaceutical industry. They are often used for the high density of the adsorbent media. The tech-
the concentration of antibiotics, peptides, and nique does not suffer from the problems of back
other molecules with molecular weights between pressure and compression of the adsorbent bed
100 and 3000 Daltons. The newer membranes of when the system is in operation. The method can
this class also permit the desalting of peptide be scaled up to high volume processes for the
solutions by allowing passage of salts from a manufacture of biomolecules. For a complete dis-
solution while retaining the peptides (15). cussion of expanded bed adsorption chromatogra-
3.3. Chromatographic Purification phy and the FastMabs system developed at
Upfront Chromatography of Denmark for the
As mentioned in the Introduction, therapeutic purification of antibodies, refer to an excellent
proteins in the biotechnology industry are isolated review by Lihme et al. (24).
from complex process streams like cell culture
supernatants, bacterial fermentation broths, and 3.5. Ion Exchange Chromatography
animal or plant extracts. The products must be The technique is relatively inexpensive and is
very pure to meet the requirements imposed by widely used in the purification of biomolecules.
regulatory agencies. The possibility of the prod- The method relies on the use of weak binding and
ucts being contaminated with potential disease- elution conditions, which help to retain the bio-
causing viruses have led the regulators to issue logical activity of the compounds being purified.
strict guidelines for their removal during down- The method has a high resolving power and
stream purification. This has pushed the drug capacity and can be used for the purification of
manufacturers to develop very efficient purifica- almost any charged molecule that is soluble in an
tion techniques. The industry can today access aqueous system.
several chromatography methods and media spe- Ion exchange chromatography purifies com-
cially developed for the purpose. These are pounds by ionic interaction between molecules of
described in the following sections. different charge based on either pH or ionic

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


Downstream Processing 93

strength. The chemists responsible for methods etrate porous particles in the stationary phase in a
development can choose between the “weak” ion chromatography column. The smaller molecules
exchangers such as diethylaminoethyl (DEAE) or enter the pores of the column material more fre-
carboxymethyl (CM) or the “strong” ion exchngers quently. The pores of the support material with a
such as the quarternary aminoethyl (QAE) or defined diameter do not permit the entry of mol-
quarternary ammonium (Q). The term weak or ecules of a larger diameter and these molecules
strong refers to the effect of pH on the functional flow through the column in the void volume of
group of the ion exchanger and not to the strength the gel. Thus, during elution, the larger proteins
of binding. The choice of the ion-exchange matrix, elute first through the column and the smaller ones
whether anion or cation, strong or weak, is influ- emerge later in the reverse order of their size, the
enced by the property of the compound under puri- smallest ones eluting last.
fication, the effect of pH on its charge 3.8. Affinity Chromatography
characteristics, its solubility and stability. Desai et
al. (25) have reviewed the use of chromatography A number of purification methods employing
in the purification of biotechnology proteins. membrane-based separations and chromatography
procedures are at the disposal of development
3.6. Hydrophobic Interaction chemists in the biotechnology industry. However,
Chromatography most of these are rather nonselective methods that
The molecular weight, structure, and function depend on physicochemical separation of pro-
of a protein is based on its genomic sequence and teins. The regulatory requirements call for a
is exhibited through its amino acid composition greater than 99% purity of the products, particu-
(26). The property of hydrophobicity of proteins larly the injectable proteins, which is not easily
is expressed by certain amino acids and of course, achieved by traditional methods. The highly
by protein molecules that contain these amino selective immunoaffinity chromatography pro-
acids. Quite often the hydrophobic residues are vides the solution to this problem (31).
present as clusters on the surface of the protein Affinity chromatography works on the interac-
molecule. Hydrophobic interaction chromatogra- tion between two molecules just as in the binding
phy is based upon the exploitation of these resi- between an enzyme and its coenzyme or between
dues, which permits the preferential adsorption hormones and their receptors. The interaction
and subsequent elution of the protein in the down- between an antigen and an antibody is a much
stream purification scheme of biotechnology stronger and more specific binding. Monoclonal
products (27,28). A list of the commercially avail- antibodies with low to intermediate affinity are
able hydrophobic matrices and a comparison of effectively employed in the purification of the
their ligand chemistry is presented by Desai et al. rapeutic proteins. The method can be employed
(25). The technique has been used both at the on an industrial scale and has helped the industry
research level and on an industrial scale. Manzke to come up with high-purity therapeutic proteins,
et al. (29) describe the purification of murine which have received regulatory approval.
monoclonal antibodies by this method. A range of matrices of good mechanical
strength and other desirable properties are avail-
3.7. Size Exclusion Chromatography able from several vendors. The general properties
The technique, often referred to as gel chroma- of the ideal matrix for specific applications are
tography or gel filtration (30), relies on the sepa- described by Walters (32) and Jervis (33). Several
ration of proteins based on their molecular weight procedures for immobilizing antibodies on the
differences. The technique is widely employed in matrices are also described by other authors
biochemical research to purify small quantities of (34,35). The choice of the procedure eventually
proteins based on their size difference. The sepa- depends on the functional groups on the matrix
ration depends on the ability of a molecule to pen- and the ligand.

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


94 Kalyanpur

3.9. Removal of Endotoxins ments are to remove from the product certain
Endotoxins, also known as pyrogens, are agents potential contaminants, notably bacterial and
that cause fever when injected intravenously into viral contaminants.
humans or animals. They are lipopolysaccharides 4.1. Sterile Filtration to Remove Bacterial
present in the cell wall of Gram negative bacteria Contaminants
and are released into the surrounding liquid when
they are killed. They are present as aggregrates in The products manufactured by the biotechnol-
solution and their size depends on the composi- ogy industry are mostly injectables and fall in the
tion of the surrounding liquids. general classification of parenteral drugs. These
need to be dispensed in a sterile form. Using asep-
Endotoxins can be effectively removed from
tic manufacturing conditions and good manufac-
process streams by ultrafiltration using membranes
turing practices (GMP) are not sufficient to ensure
with a low-molecular-weight cut off, typically 10
the sterility of these products, since they are
kDa or lower (36–38). The protein being purified,
almost always proteins and they are thermolabile
if it has a molecular weight of less than 10 kDa,
and cannot be sterilized by terminal heating. The
goes through the membrane while the larger endot-
only way to make these products free of bacterial
oxin molecules stay behind in the retentate. But the
contaminants is by filtering the product solution
method is not suitable for depyrogenating large pro-
through sterilizing grade 0.2 µm filters prior to the
teins. However, there are ways to restrict the intro- filling operation. This is an accepted procedure
duction of pyrogens into the products by following that is rigidly followed in the industry, but the pro-
certain procedures as follows: cedure needs to be validated (39) according to the
1. Sterile filtration or heat sterilization of all liq- guidelines for parenteral drugs issued by the U.S.
uids including buffers, salt solutions, and so on, Food and Drug Administration (40).
used in the manufacturing processes to remove
4.2. Virus Removal and Inactivation
the contaminant bioburden.
2. Sterilization of all production equipment in- Many biotechnology products of high therapeu-
cluding membrane filters, chromatography col- tic value are manufactured by cell culture pro-
umns, holding vessels, and other equipment. cesses or derived from different starting materials
3. Maintaining good manufacturing practices in including human plasma, animal tissue, and milk
all production areas to avoid the risk of bacte- of transgenic animals. The products thus carry an
rial contamination, post-use cleaning of all inherent risk of transmitting to the human recipi-
equipment to bring it to an endotoxin-free con- ents potential disease-causing viral contaminants
dition, and maintaining the equipment in that coming from the starting material of their manu-
condition between successive production runs. facturing process. There is also some risk of
contaminating viruses coming from certain down-
The industry is able to keep the endotoxin lev- stream processing steps, e.g., the use of pro-
els of products below the permissible levels by teolytic enzymes of animal origin such as porcine
rigorously following the above procedures. pepsin or trypsin and the monoclonal antibodies
used during purification by immunoaffinity chro-
4. Final Purification of Biotechnology matography. Finally, there always exists the risk
Products of adventitious viruses entering the process stream
The purification procedures described above owing to failure of GMP. Therefore, the regula-
are used in sequences specially adapted to a par- tory authorities have issued clear guidelines for
ticular product or process. At this stage the prod- the removal or inactivation of viruses from
uct is very likely to be between 95% and 99% biotherapeutic proteins (41).
pure. However, it still needs to be put through In 1994, the FDA’s Center for Biologics Evalu-
certain additional steps to further qualify the ation and Research (CBER) issued a directive ask-
product for regulatory approval. The special treat- ing the industries to validate the removal and

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


Downstream Processing 95

inactivation of the viruses from all biologicals. lished literature (42– 45). However, an important
Products derived from cell culture processes can point to remember is that no single method among
carry the murine leukemia virus coming from the those listed above can alone give the level of virus
cell lines, while products made by extracting ani- removal expected by the regulatory authorities.
mal tissue or from the milk of transgenic animals Therefore, membrane filtration is used with the
have the potential risk of viruses from the host other methods in a complementary fashion, and
animals. The authorities have recommended that this helps the industry to achieve the overall viral
the biotechnology products must go through dedi- reduction to satisfy regulatory requirements. The
cated virus removal or inactivation steps and that subject of virus contamination in biologicals and
these should achieve an overall virus reduction of the merits and drawbacks of the different methods
at least 12 logs. The blood products industry has employed for their elimination are topics of a
been using for some years different methods for recent review (46). Also Darling (47) has dis-
virus inactivation. These methods are heat inacti- cussed the topic of design and interpretation of
vation (pasteurization), pH inactivation, solvent/ viral clearence studies in the biopharmaceutical
detergent treatment, UV and gamma ray irradia- industry. This is an excellent reference for bio-
tion, and the addition of certain chemical inacti- technology companies planning validation of
vating agents such as ß-propiolactone. Claims virus removal.
have been made by some companies that the chro- 5. Process Validation
matography steps employed in their normal puri-
Process validation permits the drug manufac-
fication procedure can also reduce the virus
turers to assure themselves that the methods set
content of biologicals.
in-place to manufacture a product work as they
However, all these methods have some limita-
are expected to. When performed in a well docu-
tions. For example, the solvent/detergent treat-
mented manner, it also permits the drug manufac-
ment can inactivate the lipid-coated viruses, but
turer to ensure regulatory agencies (48,49) that the
the method is inoccuous against the nonlipid
methods that are employed perform reproducibly
coated viruses. There is a risk of denaturing thera-
and as expected to yield the final product of a con-
peutic proteins if the manufacturer employs heat
sistent quality.
inactivation. With chemical inactivation, the
In the context of downstream processing, pro-
manufacturer has the additional task of removing
cess validation extends to all critical steps, espe-
the added chemical completely from the product,
cially membrane-based separation systems
not to mention the fact that sometimes the chemi-
(50,51) and chromatographic procedures (52),
cals fail to inactivate certain target viruses. The including those used for the clearence of potential
chromatography methods give rather low level of bacterial (39,40) and viral contaminants (47).
virus reduction. Moreover, the results are not Kuwahara and Chuan have published an extensive
always reproducible under different process con- review of the topic of process validation (53),
ditions and are therefore considered as nonrobust which is an excellent reference to personnel in the
methods by the regulatory authorities. validation departments of the industry.
For some years, membrane filtration has
emerged as an effective method of virus removal 6. Conclusions
from biologicals. This inert method removes both The biotechnology industry manufactures
lipid-enveloped and nonenveloped viruses, numerous therapeutic products that are derived
because the removal is based on size exclusion from several source materials. The recovery and
and not on the surface characteristics of the virus, downstream purification of biologicals is a com-
as is the case with the inactivating methods. bination of diverse purification methods. Well-
Larger viruses are removed more effectively than developed processes at the research bench scale
the smaller viruses. The technique is well estab- are carefully scaled up to the production level,
lished and accepted as is evident from the pub- always bearing in mind that fewer the steps used,

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


96 Kalyanpur

higher is the eventual yield. This is because even 7. Middleberg, A.P.J. (2000). Microbial cell disruption
if a particular step loses only 5% of the product, by high pressure homogenization. in Methods in Bio-
technology, Vol 9 : Downstream Processing of Pro-
the losses add up when the product goes through a teins : Methods and Protocols (M. A. Desai, Ed.) Pub.
multistep procedure for eventually providing a Humana Press Inc., Totowa, N.J.
product of the desired level of purity. The produc- 8. Vogels G. and Kula, M.R. (1992). Combination of
tion must also proceed following GMP while pay- enzymatic and/or thermal pretreatment with mechani-
ing attention to keeping all the procedures within cal cell disintegration. Chem. Eng. Sci . 47, 123–131.
9. Lutzer, R.G., Robinson, C.W., and Glick, B.R. (1994).
the limits specified in the validation reports of the Two stage process for increasing cell disruption of E.
company. Coli for intracellular products recovery, in Proceed-
Early batches are used to make product for dif- ings of the 6th European Congress of Biotechnology,
ferent phases of clinical trials for the drug prod- (Alberghina, A., Frontali, L and Sensi, P., eds).
Elsevier Sciences B.V., Amsterdam, 111–121.
uct. By paying attention to all details, the
10. Baldwin, C. V. and Robinson, C.W. (1990). Disrup-
manufacturing personnel can help the company to tion of Saccharomyces cerevisiae using enzymatic
secure regulatory approval to market the drug lysis combined with high pressure homogenization.
ahead of its closest rivals. In today’s highly com- Biotechnol. Tech., 4, 329–334.
petitive environment in the biopharmaceutical 11. Baldwin, C.V. and Robinson, C.W. (1994). Enhanced
disruption of Candida utilis using enzymatic pretreat-
industry, the first company that gets the market- ment and high pressure homogenization, Biotechnol.
ing approval for a new drug stands to benefit Bioeng., 43, 46–56.
immensely from its sales and takes a significant 12. Middleberg, A.P.J. (2000). Large scale recovery of
share of the market for that drug and can, in most protein inclusion bodies by continuous centrifugation
cases, prevent its competitors from putting a simi- in Methods in Biotechnology, Vol. 9 : Downstream
Processing of Proteins : Methods and Protocols.
lar product on the market by virtue of its patent (Desai, M.A., ed.), Humana Press Inc., Totowa, NJ.
protection. Therefore, downstream processing of 13. Strathman, H. (1985). Membranes and membrane pro-
new drug products is critical to a biotechnology cesses in biotechnology, Trends in Biotechnol. 35,
company in achieving a premium position in the 112–118.
industry. 14. Belfort, G. (1987). in Advanced Biochemical Engi-
neering (Bungay H.R. and Belfort, G., eds) , Wiley
6. References and Sons, New York, NY., U.S.A.
1. Lehrman, S.N. (2000) in Annual Report of Genzyme 15. Michaels, M., Michaels, A.S., Antoniou, C., et al.
Transgenics, Framingham, MA., U.S.A. (1985). Tangential flow filtration. in Separations
2. Jack, K. (2001) Using transgenic cattle for the produc- Technology : Pharmaceutical and Biotechnology Ap-
tion of biologicals. Paper presented at the Conference plications, (Olson, W.P., ed), Interpharm Press, Inc.,
on Production of Biopharmaceuticals (IBC Global Buffalo Grove, IL, U.S.A.
Conferences), Hamburg, Germany. 16. Vaheri, A., Gazzei, G. and Genna, G. (1981). Tangen-
3. Theisen, M. (2001) Production of therapeutic proteins tial flow filtration of Bordetella pertusis submerged
in transgenic plants . Paper presented at the Confer- cultures. Experentia, 35, 1535–1536.
ence on Production of Biopharmaceuticals (IBC Glo- 17. Shibley, G.P., Manousas, M., Munch,K., Zelljadt, I.,
bal Conferences), Hamburg, Germany. Fisher, L., and Mayyas, S. (1980). New method for large
4. Hamel, J-F.P. and Hunter, J.B. (1990). Modeling and scale growth and concentration of Epstein Barr viruses.
applications of downstream processing - a survey of Appl. Environ. Microbiol. 40, 633–639.
innovative strategies. in Downstream Processing and 18. Bellini, W.J., Trudgett, A., and McFarlin, D.E. (1979).
Bioseparation, ACS Symposium Series, (Hamel, J- Purification of measles virus with preservation of
F.P., Hunter, J.B. and Sikdar S.K., eds), ACS Chemi- infectivity and antigenicity. J. Gen. Virol. 43, 633–639.
cal Society, Washingtion D.C., U.S.A. 19. Zydney, A.L. and Kuriyel, R. (2000). Protein concen-
5. Millipore Corporation (2000). Prefiltration and sterile tration and buffer exchange using ultrafiltration. in
filtration product selection guide : Optimised econom- Methods in Biotechnology , Vol. 9 : Downstream Pro-
ics for biopharmaceutical applications. Lit.# SG 1 cessing of Proteins : Methods and Protocols,
500EN00. (M.A.Desai, ed.), Humana Press Inc., Totowa, NJ.
6. Valax, P, and Georgion, G. (1993). Molecular charac- 20. Wells, C.M., Lyddiatt, A. and Patel, K. (1987). Liquid
terization of ( -lactamase inclusion bodies produced in fluidized bed adsorption in biochemical recovery from
E coliM. 1. Composition., Biotechnol. Prog. 9, 539–547. biological suspensions, in Separations for Biotechnol.,

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


Downstream Processing 97

(Verral, M.S. and Hudson, M.J., eds.) Ch. 16, 217-224, 37. Gabler, F.R. (1987). Pyrogens and the depyrogenation
Ellis Horwood Ltd., Chichester, U.K. of solutions with ultrafiltration membranes. in Filtra-
21. Draeger, N.M. and Chase, H.A. (1991). Liquid fluid- tion in the Pharmaceutical Industry, (Meltzer, T., Ed.),
ized bed adsorption of proteins in the presence of cells. Marcel Dekker, New York, NY.
Bioseparations, 2, 67–80. 38. Abramson, D., Butler, D. and Chrai, S. (1981).
22. McCormick, D.K. (1993). Expanded bed adsorption : The Depyrogenation of a parenteral solution by ultrafiltra-
first new unit operation in decades. Bio/Technology, 11, 1059. tion. J. Parent. Sci. Technol. 35 (1), 3–7.
23. Chase, H.A. (1994). Purification of proteins by ad- 39. Kalyanpur, M., (2000). Validation of sterilising filters
sorption chromatography in expanded beds. in the biotechnology industry, in Methods in Biotech-
TIBTECH, 12, 296–303. nology, Vol. 9 : Downstream Processing of Proteins :
24. Lihme, A., Hansen, M., Olander, M. and Zafirakos, E. Methods and Protocols, (Desai, M.A., ed.), Humana
(2000). Expanded bed adsorption in the purification of Press, Inc., Totowa, NJ.
biomolecules, in Methods in Biotechnology, Vol. 9 : 40. U.S. Food and Drug Administration (1987). Guideline
Downstream Processing of Proteins : Methods and Proto- on Sterile Drug Products Produced by Aseptic Pro-
cols, (Desai, M.A., ed.), Humana Press Inc., Totowa, NJ. cessing, Center for Drugs and Biologics, Rockville,
25. Desai, M.A., Rayner, M., Burns, M. and Bermingham, MD, U.S.A.
D. (2000). Applications of chromatography in down- 41. ICH Topic Q 5 A : Quality of Biotechnological Prod-
stream processing of biomolecules. in Methods in Bio- ucts - Viral Safety Evaluation of Biotechnology Prod-
technology, Vol. 9 : Downstream Processing of ucts Derived from Cell Lines of Human or Animal
Proteins : Methods and Protocols, (Desai, M.A., ed.), Origin, European Agency of the Evaluation of Medici-
Humana Press Inc., Totowa, NJ. nal Products, Human Medicines Evaluation Unit,
26. Mazek, J. and Kypr, J. (1992). Nucleotide composi- (CPMP/ICH/295/95), March 1997.
tion of genes and hydrophobicity of the encoded pro- 42. Burnouf-Radosevich, M., Appourchaux, P., Huart, J.J.
teins. FEBS Lett., 305, 163–165. and Burnhouf, T. (1994). Nanofiltration , a new spe-
27. Eriksson, K. (1989). in Protein purification (Rydén, cific virus elimination method applied to high purity
J., ed.), VCH, New York, NY. Factor IX and Factor IX concentrates. Vox Sang 67,
28. Hearn, M.T.W. (1989). in Protein purification (Rydén, 132–138.
J., ed), VCH, New York, NY. 43. Hoffer, L., Schwinn, H., Biesert, H., and Josic, D.
29. Manzke, O., Tesch, H ., Diehl, V. and Bohlen, H. (1995). Improved virus safety and purity of a chro-
(1997). Single step purification of biospecific mono- matographically produced Factor IX concentrate by
clonal antibodies for immunotherapeutic use by hy- nanofiltration. J. Chromatogr. 669, 187–196.
drophobic interaction chromatography. J. Immunol. 44. DiLeo, A.J., Vacante, D.A., and Dean, E.F. (1993a).
Methods, 208, 65–73. Size exclusion removal of model mammalian viruses
30. Gel Filtration : Principles and Methods, 6th edition, using a unique membrane system, Part I : Membrane
Pharmacia Biotech., Uppsala, Sweden. qualification. Biologicals, 21, 275–286.
31. Desai, M.A. (2000). Immunoaffinity adsorption in the 45. DiLeo, A.J., Vacante D.A., and Dean, E.F. (1993b).
large scale isolation of biomolecules, in Methods in Size exclusion removal of model mammalian viruses
Biotechnology, Vol. 9 : Downstream Processing of using a unique membrane system, Part II : Module
Proteins : Methods and Protocols, (Desai, M.A., ed.) qualification and process simulation. Biologicals, 21,
Humana Press, Inc., Totowa, NJ. 287–296.
32. Walters, R. (1985). Affinity Chromatography. Anal. 46. Levy, R.V., Phillips, M ., and Lutz, H. (1998) Filtra-
Chem. 57, 1099A–1114A. tion and removal of viruses from biopharmaceuticals,
33. Jervis, L. (1987). Polymers in affinity chromatogra- in Filtration in the Biopharmaceutical Industry,
phy in Developments in Polymer Reagents, Catalysts (Meltzer, T.H. and Jorintz, M.W., eds.), Marcel
and Adsorbents (Hodge, P. and Sherringtoon, D.C., Dekker, New York, NY.
eds.), Wiley, London, U.K., pp.65–96. 47. Darling, A.J. (2000). Design and interpretation of
34. Sada, E., Katoh, S., Sukai, K., Tohma, M. and Kondo, Viral Clearence Studies for Biopharmaceutical Prod-
A. (1986). Adsorption equilibrium in immunoaffinity ucts, in Methods in Biotechnology, Vol. 9 : Down-
chromatography with polyclonal and monoclonal an- stream Processing of Proteins : Methods and Protocols
tibodies. Biotechnol. Bioeng. 28, 1497–1502. (Desai, M.A., ed) Humana Press Inc., Totowa, NJ.
35. Desai, M.A. (1990) Immunoaffinity adsorption : Pro- 48. U. S. Food and Drug Administration, Center for
cess scale isolation of therapeutic grade biochemicals. Drugs, Biologics, Devices and Radiologic Health
J. Technol. Biotechnol. 48, 105–106. (1987). Guidelines on general principles of process
36. Nelsen, L.N. (1978) Removal of pyrogens from validations, Rockville, MD.
parenteral solutions by ultrafiltration. Pharma. 49. Commission of the European Communities (1989).
Technol. 2 (5), 46–49, 80. Guide to good manufacturing of medicinal products,

MOLECULAR BIOTECHNOLOGY Volume 22, 2002


98 Kalyanpur

Vol. IV. The rules governing medicinal products in the 52. Parenteral Drug Association (1992). Industry perspec-
European community, Luxembourg, Office for Offi- tive on the validation of column-based separation pro-
cial Publications of the European Communities. cesses for the purification of proteins. J. Parent. Sci.
50. Michaels, S.L. (1991). Validation of tangential flow and Technol. 46, 87–97.
filtration systems. J. Parent. Sci. and Technol. 45 (5), 53. Kuwahara, S.S. and Chuan, J.H. (1995). Process vali-
218–223. dation of separation systems in Separations Technol-
51. Millipore Corporation, Bedford, MA, U.S.A. (1990). ogy : Pharmaceutical and Biotechnology Applications,
Validation of tangential flow filtration (TFF) systems, (Olson, W.P., ed.), Interpharm Press, Inc., Buffalo
lit. # TB 052. Grove, IL.

MOLECULAR BIOTECHNOLOGY Volume 22, 2002

You might also like