Sie sind auf Seite 1von 17

SCALEDOWN OF BIOPHARMACEUTICAL • process characterization to support process valida-

PURIFICATION OPERATIONS tion (15–18). These include range finding studies to


set the acceptable ranges for the operating param-
ANURAG S. RATHORE and eters, spiking studies to establish clearance of host
VARSHA S. JOSHI cell impurities [host cell proteins (HCPs) and DNA]
Chemical Engineering and other product-related impurities, and establish-
Department, Indian ing viral clearance capability (19–23). The latter is a
Institute of Engineering, requirement for regulatory approval;
Hauz Khas, New Delhi, • establishing lifetime for reusable media for
India chromatography and membrane separation steps
(19,24,25);
INTRODUCTION • manufacturing support for licensed commercial
processes to address manufacturing investigations,
Biotech-based therapeutics have emerged as the growth postapproval process change for comparability, and
engine for the pharmaceutical industry (1). Successful evaluate raw material changes postlicensure.
development and commercialization of biotech processes
require us to understand the relationships between the
various attributes of the product and the product’s clinical
GENERAL CONSIDERATIONS
safety and efficacy, as well as the effect of the differ-
ent process parameters on the product’s attributes (2).
Use of Real Feed Streams and Raw Materials
Considering the complexity of the biotech products, under-
standing the latter requires a lot of experimentation. When performing scale-down experiments, it is recom-
Working at small scales offers a potential solution to this mended that we use feed streams that are representative
problem. of manufacturing scale (24). It is best to use process inter-
There are many drivers for working at small scales mediates produced at the manufacturing scale itself. If
(3–5): this is not possible then feed streams should be produced
at pilot scale that is representative of the manufactur-
• Smaller the scale, lesser the requirement of feed ing scale. Furthermore, the raw materials used for the
material. This is often an issue during early stages experiments (buffer, resin, etc.) should be taken from the
of product development. cGMP-released inventory. This ensures that any addi-
• Parallelization and automation offer an enormous tional variability in the experimentation originating from
increase in productivity when working at small scale use of noncomparable raw materials or feed materials is
when compared to manufacturing scale operations. avoided. Knowledge of the storage and stability of the feed-
This allows us to examine a lot more parameters stream material is acquired beforehand. Storage-related
than what we can otherwise. degradation of the inventory could affect the outcome of
• Higher productivity may also result in accelerated the small-scale runs (24).
timelines, something that can be very critical in the
pharmaceutical industry. In case of a blockbuster Qualification of a Scale-Down Model
drug (sales greater than $1 billion/year), each extra
The qualification of the scale-down model must be carried
day delay in development can result in excess of $1
out as per current good manufacturing practice (cGMP)
million in lost sales revenue (3).
validation guidelines that require a written qualification
• Newer initiatives such as Quality by Design (QbD) protocol describing qualification studies to be conducted
(6–8) and Process Analytical Technology (PAT) and should include preapproved acceptance criteria for
(9–11) demand a deeper understanding of the key output parameters. For qualification, the runs are
process and the product. This in turn requires more
performed in triplicate (or more) to assess the repro-
extensive experimentation than what has been
ducibility of the scale-down model and for meaningful
traditionally done.
comparison to full-scale. In most cases, multiple perfor-
When using small-scale models for process development, it mance parameters need to be compared across scales to
is important that the small-scale model performs similar establish comparability of performance and qualify the
to the corresponding manufacturing scale unit operation. scale-down model. Compared to the univariate compari-
This ensures that the data from experimentation and son that is usually done, the application of multivariate
the conclusions reached from the data are applicable to analysis (MVA) tools in performing this comparison has
the manufacturing scale (12–14). Such small-scale models been recently proposed (26). This allows for a more com-
may be used for a variety of purposes: prehensive comparison of the data and an examination
of all important inputs and output variables at the same
• process development and optimization; time. This allows for correlations between variables to

Encyclopedia of Industrial Biotechnology: Bioprocess, Bioseparation, and Cell Technology, edited by Michael C. Flickinger
© 2013 John Wiley & Sons, Inc. Published 2013 by John Wiley & Sons, Inc.

1
2 SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS

be more easily identified. In addition, MVA projection value is kept constant. The cell culture solution is continu-
tools condense complex multivariate data sets into easily ously fed to the outside of the centrifuge and flows through
interpretable graphical projections (26,27). the spaces between the stacked discs toward the center of
In the following sections, we discuss major unit the centrifuge. Cells and particles are thrown radially out-
operations currently used in biopharmaceutical industry ward by the centrifugal force toward the underside of the
for downstream processing (Fig. 1) with respect to the upper disc and slide down the disc surface to the sediment
scale-down methodology and the review literature. holding space. The collected solids are either continuously
or discontinuously discharged from the centrifuge. The
liquid flows continuously to the top of the centrifuge and
CENTRIFUGATION is discharged under pressure. 
Unique expressions for exist for each centrifuge
With the recent developments in mammalian and micro- design. For a disc stack centrifuge (28),
bial cell culture, solids concentrations of up to 30% (w/v)  2π nd ω2 (r32 − r31 )
are readily achievable (28). For such cases, centrifugation = f1 (1)
is often the method of choice for liquid–solid separation 3g tan θ
because of its cost effectiveness, robust operation, and where, nd is the number of active discs, r1 and r2 are the
applicability to continuous operation (29,30). Harvesting inner and outer disc radius (in meters), g is acceleration
of bacterial and mammalian cells is commonly done using due to gravity (in meters per square second), and θ is
stacked disc centrifuges (31). the half disc angle. Furthermore, ω = 2π N, where ω is the
In this unit operation, the centrifugal force is used to angular velocity and N the rotational speed (in revolutions
separate mixtures of particles of varying masses or densi- per second). Also, f1 is the correction factor for spacer
ties suspended in a liquid. When a vessel is rotated at high caulks determined using the following expression:
speeds, the angular momentum yields an outward force to ⎛
2 ⎞
each particle that is proportional to its mass. This force   r1
3ZL BL ⎜ 1 − r2 ⎟
is countered by the so-called ‘‘drag’’ force that the liquid f1 = 1 − ⎝
3 ⎠ (2)
exerts on the particle. The net effect of ‘‘spinning’’ the 4π r2 r
1 − r1
2
sample in a centrifuge is that larger/denser particles move
outward faster than smaller/lighter particles. The rate of where ZL is the number of caulks on a disc and BL the
centrifugation is determined by the angular acceleration caulk width (25).
applied to the sample, typically measured in comparison For the laboratory centrifuge,
with the gravitational force (g). The sedimentation rate of
a particle is proportional to the liquid flow rate  through Vω2 (3 − 2x − 2y)
=
(3)
the centrifuge (Q) divided by the quantity sigma ( ). The 6g ln R2R o
o +R i
latter is the area of a simple gravity settling tank of equiva- 
lent sedimentation characteristics to that of the centrifuge. where is the equivalent settling area of a laboratory cen-
Theoretically, the performance of two centrifuges process-  trifuge, V is the sample volume, ω is the angular velocity,
ing the same feed material will be equivalent if the Q/ x and y are fractional times required for acceleration and

Mammalian or microbial cell culture

Microfiltration

Cell removal and concentration Centrifugation

Coagulation/flocculation
Ultrasonic vibrations
Cell disruption
Homogenizer (e.g. in case of intracellular expression)
Bead or ball mill
Microfiltration
Removal of cell debris
Centrifugation

Solubilization and refolding of IB proteins


Traditionally done by dilution
(e.g. in E.coil fermentation)

Protein precipitation/extraction Figure 1. Process flow diagram for


a typical biotech process used for
manufacturing of a biotech-based
therapeutics. The diagram is meant
Combination of UF/DF and to be illustration and not extensive,
chromatographic purification step as it only shows some of the options.
SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS 3

deceleration, respectively (measured for different centrifu- recovery in continuous high speed centrifuges. The
gation speeds using a stopwatch), Ri is the inner radius authors concluded that such breakup can lead to over
(the distance between the center of rotation and the top 10-fold reduction in separator capacity. Mannweiler and
of the liquid), and Ro is the outer radius (the distance Hoare (36) have described the methodology for 10-fold
between the center of rotation and the bottom of the tube) scale-down of a disc stack centrifuge. Scale-down was
(32). achieved by the reduction of the number of discs available
The performance of a continuous full-scale centrifuge for separation purposes and by careful positioning of
is often quantified by the amount of the solids remain- these discs in the overall disc stack. The authors used a
ing after centrifugation and can be described using the combination of dye tracer and particle separation studies
following relationship (4): to optimize the disc stack configuration. The resulting
  grade efficiency curve was an accurate reflection of that
Qc
S=f  (4) for the full-scale centrifuge, especially in the critical
c design region where near complete particle recovery has
been proposed. To date, several models that make good
the solids remaining, Qc the volumetric flow
where S is
predictions at the 10–100 mL scale have been proposed
rate, and c the equivalent settling area.
(37–39). However, in recent years, there has been an
increased interest in models that are based on microtiter
Scale-Down Modeling Approach
plate technology (40–45). The use of a high speed rotating
Scale-downof the centrifugation step can be achieved by shear device to reproduce shear conditions in combination
keeping Q/ constant (33,34). Sigma theory modeling can with benchtop centrifugation has successfully been used to
be generically applied to a wide number of centrifuge types demonstrate that an ultra-scale-down (USD) model using
using calibration factors to account for changes in flow 15-mL centrifuge tubes can give an accurate prediction
patterns and centrifuge geometry (35). The feed flow rate, of the expected clarification of a mammalian cell culture
centrifuge angular velocity, and system back pressure can in industrial disc stack centrifuges (46). Tustian et al.
be suitably controlled. The temperature of the feed and developed and verified a methodology in which USD
centrate is kept identical to that used in the manufacturing curves for centrifugal clarification were corrected by
scale. The smallest diluting the feed material by 2%. After dilution, they
 disc stack centrifuges available for
scale-down have values of 1800 m2 and can process found that the USD curves accurately predicted pilot-scale
feed volumes of 2–20 L at flow rates of 10–60 L/h with clarification performance of high cell density broths of
a 0.5 L solid holding volume. Production-scale disc stack Saccharomyces cerevisiae and Escherichia coli cells (28).
centrifuges can have sigma values as high as 1.3 × 105 m2 . Zaman et al. (47) have developed a USD-based approach
Thus, scale-down factors of 70-fold are achievable (12). accounts for both gravitational and shear effects on
clarification performance.
Scale-Down Qualification In summary, despite advancements in scale-down of
centrifugation, the models that exist at present are suit-
The performance of the centrifugation step can be mea- able for knowledge generation and preliminary examina-
sured in terms of step yield, the clarity of the centrate, tion of the effects of the various process parameters. Final
filterability of the centrate, particle size distribution, and determination of the acceptable ranges or the process
extent of cell lysis. The clarity of the centrate can be validation acceptance criteria still requires experimenta-
measured using particle counters, ultraviolet (UV) spec- tion with the manufacturing scale centrifuge. This is due
troscopy, or turbidimeter. Yield can be determined by to the complexity of the fluid dynamics in an industrial
analyzing the feed material and the output for method by centrifuge.
high pressure liquid chromatography (HPLC).

Literature Review HOMOGENIZATION

Researchers have proposed the use of a benchtop E. coli is among the most frequently used microbial hosts
centrifuge as a scale-down model for pilot-scale equipment to produce recombinant therapeutic products. An intrinsic
(3). The focus of the investigation was on properties of the characteristic of E. coli and some other similar expres-
protein precipitate, the fraction of solids recovered, and sion systems is that they retain the expressed proteins
the extent of dewatering achieved. Good agreement was within the cell (48). Protein release in such cases requires
obtained at bench scale (a 1000-fold scale-down factor) cell lysis induced chemically (49,50), enzymatically (51),
for all these parameters when compared to pilot-scale, or mechanically. A robust and rapid way to achieve the
batch-feed operation. In addition, the methodology latter is by mechanical cell disruption via homogeniza-
developed here allowed for identification of the extent of tion by performing mechanical cell disruption followed by
breakup that occurs in continuous-feed centrifuges when cell debris removal by centrifugation (52,53). Options for
processing shear-sensitive materials such as protein industrial-scale mechanical cell disruption include high
precipitates. Other researchers have used the laboratory speed bead mills (54–57), high pressure homogenization
scale-down model to predict the performance of disc stack (HPH) (58–61), and ultrasonication. Most popular of these
centrifuge for the recovery of yeast cell debris particles in manufacturing of biotech-based therapeutics is HPH.
(34). The scale-down model was also used to investigate In this unit operation, cell suspension is forced by
the extent of shear-sensitive protein aggregates during pressures of several hundred bars to pass through an
4 SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS

adjustable discharge valve, which has a restricted orifice. In summary, like centrifugation, homogenization too
The combination of high shear, the high pressure drop, is a challenging unit operation to scale-down. While the
and the impact forces in the discharge valve results in cell existing models can be used to generate some fundamental
disruption (62). Homogenizers may use discrete multiple knowledge about the behavior of cells under shear stress,
passes through a recycle loop to effect the multiple passes the flow dynamics in most scale-down systems is quite
needed for continuous processing. Disruption occurs via a different from that of their large scale counterparts. This
first-order process with respect to the number of passes necessitates that the final experimentation be done on
with the rate constant being a strong function of pressure the manufacturing equipment itself for setting process
(61) and the valve and valve seat design (63). In addition, validation criteria and acceptable ranges.
the rate constant is affected by the conditioning of the cell
suspension (e.g. chemical pretreatment) and the nature of
the fermentation growth media. Cells grown in complex REFOLDING
media are more difficult to disrupt compared with those
grown in defined media (64). Protein refolding includes solubilization and reduction
of inclusion bodies (IBs) followed by renaturation (slow
removal of denaturant). In most cases, performance of the
Scale-Down Modeling Approach
refolding step is critical to overall recovery of the process.
Scale-down of an HPH-based process is complicated by There are many factors, both chemical (67–69) and phys-
the fact that varying the flow rate by orders of magni- ical (70–73), that can affect protein refolding. Refolding
tude results in significant variation in the gap opening of by dilution is still the preferred technology for large-scale
the homogenization valve, with severe consequences on refolding. The first step is solubilization of the IBs in a
the fluid dynamics and mechanical interaction between mixing vessel. After any remaining undissolved IBs are
the valve and cells. A procedure for calculating the pres- removed by centrifugation, the dissolved IBs are further
sure drop as a function of gap opening has been reported. reduced and renatured in an agitated tank. Aggregation
This procedure has been validated for Stansted high pres- is the major issue confronted during refolding and can
sure homogenizers, which exhibit a quadratic dependence be caused by improper mixing and/or high local protein
on process medium flow rate (65). A typical industrial concentration. Rapid and uniform dilution is necessary to
scale homogenizer needs at least 30 L of the feed volume, avoid aggregation.
while a typical scale-down model may need up to 40 mL
of the feed volume. Thus, scale-down factors of 750-fold Scale-Down Modeling Approach
are achievable. Inactivation depends not only on applied
pressure of homogenization and number of passes but also It has been observed that the refolding yield is inversely
on the microbial strain. For instance, in a single homoge- proportional to the protein concentration (67,69,74). Fur-
nization pass at 250 MPa, six log cycles of inactivation can thermore, slower addition of the denatured protein to
be achieved for E. coli, five log cycles for S. cerevisiae but refolding buffer has been known to result in higher yields
only one log cycle for Lactobacillus delbrueckii. The latter as the influence of macroscopic concentration gradients
is characterized by a thicker and more resistant cell mem- is minimized even under poor mixing conditions (75,76).
brane. Lethality can be increased not only by increasing It is recommended that a temperature-controlled and
the pressure but also through multiple-pass treatments agitator-driven closed vessel (such as a bioreactor) be used
(66). Since temperature rises during homogenization, one as scale-down model. This way, engineering considerations
should aim at keeping the temperature rise similar so as can be applied to mimic conditions at the manufacturing
to get similar step performance. scale. Chemical conditions such as amounts and concen-
tration of the various additives, sequence of addition,
temperature, pH, conductivity, and protein concentration
Scale-Down Qualification
should be kept identical (Table 1). Physical aspects such
The desired product recovery and purity after homoge- as mixing and distribution of oxygen in the tank could
nization can be calculated using HPLC. Generation of be maintained similar by keeping the mass transfer coeffi-
aggregates can be monitored using size-exclusion high cient (kL a) same across scales. Similar quenching protocols
pressure liquid chromatography (SE-HPLC). The log cycle followed by proper storage of the refolded protein should
reduction in the feed should be determined at the end of the be followed at both the scales.
homogenization cycle along with particle size distribution
of the cell debris and viscosity of the product. Temperature Scale-Down Qualification
and pressure should be monitored throughout the process.
Product quality can be monitored during and after refold-
ing by a variety of generic (e.g. solubility and folding)
Literature Review
or specific (e.g. activity) methods (77). Functional assays
Literature is scarce on the topic of scale-down model of high (bioassays, immunoassays, etc.) can be very informative.
pressure homogenizers. Donsi et al. (66) have compared Analytical techniques such as intrinsic fluorescence, cir-
the homogenization efficiency at two scales (laboratory cular dichroism (CD, secondary structure), dynamic light
scale and pilot scale) and demonstrated that the scale scattering (DLS), turbidity measurements, or chromato-
of the equipment does not affect the level of inactivation graphic analysis (HPLC) can be used as indicators to mon-
attained. itor performance of the refolding step (77–80). SE-HPLC
SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS 5

can be used to determine the amount of dimers or aggre- It entails manipulation of the feed solution to favor pro-
gates generated in the refolding process. CD can be used tein precipitation, mixing of the vessel contents as the
to measure protein secondary structure and DLS to mea- precipitation takes place, and subsequent solid–liquid
sure protein aggregation so as to verify protein structure separation to separate the two phases. Mixing is a critical
(77,78). Crystallogenesis offers another approach for prod- aspect of this unit operation and impacts apparent protein
uct quality measurement as only properly folded proteins solubility, protein structure in precipitates, particle size,
with a uniform aggregation state (monodispersed sample) morphology, and recoverable activity (22). Aggregate lev-
yield well-ordered crystals. While the method of choice els and characteristics in the precipitation output impact
will depend on the target protein or project, it is generally the performance of the subsequent centrifugal separation.
recommended that more than one method be employed Protein solubility is affected primarily by the type and
(80). concentration of precipitant, pH, and ionic strength (67,
88–91). The physical properties of the aggregates depend
Literature Review on several factors: the type of precipitation reactor (90),
Mannall et al. (81) have studied the USD approach to pro- choice, concentration, and rate of addition of precipitat-
tein refolding in microwell plates and demonstrated the ing agent (89); initial protein concentration; and nature
applicability and scalability of this microwell screening and extent of mixing and residence time in the reactor
for the development of protein refolding processes. Qoron- (68,69,71).
fleh et al. (82) have suggested the use of high throughput, Batch reactors are the simplest type of precipitation
automated refolding screens. Ordidge et al. (83) have used reactor and have been widely used in the industry. The
an automated robotic platform to develop a dilution refold precipitating agent is slowly added to the protein solution
process-screening platform on which a hierarchical set of under mixing. Since the particles are exposed to a wide
assays rapidly determine optimal refolding conditions at range of shear stresses for a long period of time, the
the microscale. aggregated protein particles tend to be compact, dense,
In summary, the typical bioreactor offers a platform and mechanically stable.
for performing refolding under conditions that mimic
manufacturing conditions. Furthermore, high throughput Scale-Down Modeling Approach
process development (HTPD) platforms for performing Parameters that are likely to impact the performance of
refolding have also been successfully demonstrated. We the precipitation step need to be held constant (Table 1).
expect these to be adopted by the biotech industry to These include pH, conductivity, final precipitant concen-
expedite process development, optimization, and charac- tration, and temperature. Design parameters that need to
terization of protein refolding step. be maintained same across scales include tank geometry
(height-to-diameter ratio), impeller type, diameter of the
PRECIPITATION impeller relative to that of the tank, placement of the
impeller, baffle geometry, mean velocity gradient, aging
Precipitation offers a low cost, high yield unit oper- parameter (Camp number), and residence time (4). In
ation that can be used for separation of the various cases where the small-scale system does not mimic the
host-cell-related and process-related contaminants (84). manufacturing design, dimensionless parameters such as
Although the use of this step is somewhat limited in the mass transfer coefficient (kL a) can be used to target
production of therapeutic proteins because of the concerns identical mixing conditions across scales (Table 1) (92).
related to product stability, it is the step of choice for
certain applications (85). A common application is as Qualification of Scale-Down Model
a primary isolation procedure in which the amounts of Product purity (measured by specific activity or chro-
contaminating proteins (and other species) are decreased matographic methods), product and total protein yield,
along with a significant reduction of process volume (86). and impurity levels [assessed by sodium dodecyl sulfate
The basis of separation in this unit operation is reduc- polyacrylamide gel electrophoresis (SDS-PAGE)] can be
tion in the solubility of the product to induce precipitation. used to compare step performance across scales. Addi-
This can be achieved via several methods: ionic precip- tional measurements could include measurement of ion
itation (e.g., ammonium sulfate and sodium chloride), concentrations (sodium content), turbidity of the super-
temperature, pH, metal ions (e.g. Cu2+ , Zn2+ , and Fe2+ ), natant to gauge the extent of precipitate removal, and
nonionic polymers (e.g. polyethylene glycol), organic sol- the phase ratio and/or fraction of solids in the precipitate.
vents (e.g. ethanol and acetone), tannic acid, heparin, The kinetics of precipitation can be measured to establish
dextran sulfate, cationic polyelectrolytes (e.g. protamines), equivalence across scales (12).
short-chain fatty acids (e.g. caprylic acid), trichloroacetic
acid (TCA), lectins (e.g. concanavalin A), group-specific
Literature Search
dyes (e.g. Procion Blue), and ligand–antibody interac-
tion (87). Of these methods, ionic precipitation utilizing Chhatre et al. have investigated the integrated appli-
inorganic salts is the most commonly used method. cation of USD techniques and economic modeling as a
The precipitation step is generally used at an early means for identifying optimal bioprocess operating con-
stage of the purification procedure, often on the clarified ditions. An economic model of this process was used to
extract, to obtain initial purification and concentration. identify where greatest product loss occurs and hence
6
Table 1. Summary of Scale-Down Approach for Different Purification Unit Operations—Input Parameters That are Held Constant During Scale-Down, Input
Parameters That are Altered During Scale-Down, and Performance Parameters That are Monitored to Ensure Qualification of the Scale-Down Model

Purification Step Operational (Input) Parameters That are Kept Operational (Input) Performance Parameters for Model Qualification
Identical Parameters That are
Altered
Centrifugation Linear flow rate, temperature, sigma factor, feed Process volumes, volumetric Product concentration and recovery, turbidity, cell
characteristics flow rate, centrifugal speed lysis
Homogenization Linear flow rate, temperature, solution pH, Process volumes, volumetric Product concentration and recovery, turbidity, cell
conductivity, protein concentration, feed flow rate, homogenizer lysis, product-related impurities (aggregates)
characteristics pressure, number of passes
Refolding Mass transfer coefficient (kL a), temperature, Agitation rate Product concentration and recovery, conductivity,
chemical stock concentration, feed characteristics pH, product-related impurities (aggregation,
deamidation, methylation, etc.)
Microfiltration Protein loading (g product/m2 membrane area), Process volumes, volumetric Product concentration and recovery, turbidity, filter
number of diavolumes, volumetric concentration flow rate, membrane area capacity
factor, feed characteristic, temperature, linear
flow velocity, cross flow rate per channel,
transmembrane pressure
Precipitation Mass transfer coefficient (kL a), precipitant Mixing speed, precipitant Product concentration and recovery,
concentration, precipitant-to-feed-volume ratio, feed rate, process volume product-related impurities, process-related
feed characteristics impurities, host-cell-related impurities, turbidity
Chromatography Bed height, buffers (pH, conductivity, etc.), resin, Column diameter, column Product concentration and recovery,
linear flow rate, protein loading (g product/L volume, volumetric flow chromatographic profiles, HETP, asymmetry,
resin), pooling criteria, temperature, feed rate conductivity, pH, product-related impurities
characteristics (aggregation, deamidation, methylation etc.),
process-related impurities (protein A, antifoam,
etc.), host-cell-related impurities (host cell
impurities, DNA, etc.)
Ultrafiltration/Diafiltration Protein loading (g product/m2 membrane area), Process volumes, volumetric Product concentration and recovery, turbidity, filter
number of diavolumes, volumetric concentration flow rate, membrane area capacity, pH, conductivity, excipient
factor, feed characteristics, temperature, linear concentration, product-related impurities,
flow velocity, cross flow rate per channel, host-cell-related impurities
transmembrane pressure
Viral inactivation Mass transfer coefficient (kL a), temperature, Agitation rate Product concentration and recovery,
contact time, chemical stock concentration, feed product-related impurities
characteristics
Virus removal filtration Protein loading (g product/m2 membrane area), feed Process volumes, volumetric Product concentration and recovery,
characteristics, temperature, linear flow velocity flow rate, membrane area product-related impurities (aggregates)
Membrane adsorption Bed height, buffers (pH, conductivity, etc.), resin, Column diameter, column Product concentration and recovery, conductivity,
linear flow rate, protein loading (g product/L volume, volumetric flow pH, product-related impurities (aggregation,
resin), temperature, feed characteristics rate deamidation, methylation, etc.), process-related
impurities (protein A, antifoam, etc.),
host-cell-related impurities (host cell impurities,
DNA, etc.)
SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS 7

where the largest opportunity cost was being incurred. Size of Scale-Down Model
Scale-down analysis was used to characterize the
Chromatography is one of the more straightforward unit
influence of the critical steps, identified as precipitation
operations to scale-down (13). The size of the scale-down
and centrifugation, on recovery of lactoperoxidase
system varies with the intended use of the system. For
(LPO). The results demonstrate the power of combin-
performing binding studies, a 96-well format HTPD plat-
ing USD data with models of economic and process
form for chromatography may suffice and is likely to be the
performance in order to establish the best overall
most efficient (5). For performing resin screening studies,
operating strategies for biopharmaceutical manufacture the above mentioned HTPD platform or small columns
(93). Boychyn et al. (4) have used a pilot-scale batch (∼ 2.5 cm height, 1 mL) can be used to screen for var-
stirred-tank reactor and a multichamber-bowl centrifuge ious matrices and mobile phase compositions in order
to predict the performance of larger reactors and to perform preliminary optimization (97). Viral clearance
centrifuges. studies are often done with shorter columns (5–10 cm bed
In summary, the lack of literature on the scale-down height) because of the difficulties and expenses associated
reflects the sparse usage of precipitation in production with creating virus spikes (98,99). Final process character-
of therapeutic proteins. This may gain more interest in ization studies that form the basis for setting the process
the future, as researchers develop new approaches toward validation acceptance criteria and the acceptable ranges
mainstream application of precipitation for achieving cost for operating parameters are often performed using a col-
effective way of protein purification. umn with diameter of at least 1.6 cm and same bed height
as at the manufacturing scale (97). Column diameters of
less than 1.0 cm are generally not used in these studies
since the ratio of system dead volume to column volume
CHROMATOGRAPHY
can result in pool concentrations significantly lower than
those seen at large scale (13). It should be noted that when
Chromatography is the primary workhorse of the biotech compared to the higher end of column size in manufac-
industry for product purification (94). In the commonly turing scale (2 m), the scale-down factors may range from
used bind and elute mode of chromatography, the first 1:100 to over 1:1000000 (12).
step often involves binding of the product to the adsor-
bent. Impurities that do not bind to the column will just
Chromatographic Equipment
flow through and go to waste. Then, a wash is conducted
to wash off remaining unbound and/or weakly bound Automated small-scale chromatography systems, such as
impurities. Finally, elution is performed by altering the the äKTA Explorer, excel at these scale-down studies.
conditions of the mobile phase in the manner that differen- They offer precise, accurate, and reproducible control of
tial binding of the protein of interest and other impurities flow rates and gradients. However, accuracy and precision
is exploited to achieve purification of the product. Alterna- of in-line detectors (pH, conductivity, temperature) should
tively, the column can be operated in flow through mode, be calibrated at reasonable frequency by using off-line
whereby the impurities bind to the column and the product probes (13).
flows through. A variety of chromatographic techniques
are employed in purification processes, mainly differ- Scale-Down Modeling Approach
ing in the nature and structure of the stationary phase Column scale-down is normally performed by maintaining
(resin particles). These techniques exploit differences in bed height while reducing column diameter, which main-
properties of proteins such as (i) size and shape (gel tains the residence time (Table 1). In this way, the linear
filtration/size-exclusion chromatography); (ii) net charge flow rate is maintained, while the volumetric flow rate is
and distribution of charged groups (ion exchange chro- reduced (100). The International Conference on Harmo-
matography); (iii) isoelectric point (chromatofocussing); nization (ICH) guideline on viral safety states that the
(iv) hydrophobicity (hydrophobic interaction chromatogra- validity of the scaling down should be demonstrated and
phy, reversed phase chromatography); (v) metal binding that the column bed height should be shown to be repre-
(immobilized metal ion affinity chromatography); (vi) con- sentative of commercial scale manufacturing (101). Thus,
tent of exposed thiol groups (covalent chromatography); if the bed height is different at small scale, as in case of
and (vii) biospecific affinities for ligands, inhibitors, recep- viral clearance studies, the linear flow velocity should be
tors, and antibodies (affinity chromatography) (95). In adjusted appropriately to keep the residence time constant
recent years, the concept of multimodal, or mixed-mode (102).
adsorption chromatography, is receiving an increasing The operating conditions of the experiments should be
amount of attention. These stationary phases allow for identical to those in manufacturing. This is so as subtle
more than one ‘‘mode’’ of chromatography to be conducted changes in temperature, pH, or conductivity could affect
in a single step. In all cases, separation can be influenced the retention time of proteins on chromatographic resins.
by factors such as the nature of ligand and matrix, solvent Heat exchangers on the inlet tubing and column jack-
pH, temperature, and size of the beads and pores (96). In eting maybe necessary for accurate, consistent, and pre-
the following sections, we discuss some of the key consid- cise temperature control during scale-down. For example,
erations when performing scale-down of chromatography hydrophobic interaction chromatography has been report-
steps. edly sensitive to temperature fluctuations and could give
8 SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS

rise to product retention and selectivity with variation offer and the higher level of comparability that comes in
in temperature (12). Some of the significant operational the laboratory-scale columns is likely to be different for
parameters include the following: different applications. For most cases, however, the best
would be to use the HTPD platforms for examining a large
• Protein loading (g protein/L resin) should be number of process parameters in wide ranges followed
identical. In size-exclusion chromatography, loading by use of the traditional laboratory-scale chromatography
should be based on volume percentage of the column columns for performing the final process characterization
bed volume. experiments that support process validation acceptance
• Gradient lengths (in number of column volumes) and criteria. In the future, the HTPD platforms are likely to
slopes (e.g. increase in percent limit buffer per unit continue to evolve and gradually with increased rigor they
time) should be kept identical. may be able to replace the laboratory-scale experimenta-
• Temperature, pH, and conductivity should be main- tion completely.
tained identical. Differences in temperature across
scales can result in significant changes in conductiv- MICROFILTRATION AND
ity and thus impact step performance. ULTRAFILTRATION/DIAFILTRATION (UF/DF)
• Bed height of the chromatography column and the
linear flow velocity are generally kept constant across Microfiltration
scales.
Microfiltration (MF) membranes can be used to harvest
• Pooling criteria should be the same as in manufactur-
cells from mammalian and bacterial cell culture to provide
ing. They can be online (conductivity or absorbance)
cell-free conditioned medium (containing the product) to
or off-line (gels or HPLC). If the pooling criteria are
the downstream purification steps. The pore sizes of typical
UV based, it is a good practice to ensure that the UV
MF membranes range from 0.1 to 0.8 μm. MF membranes
detectors of the small-scale systems correlate well
retain cells and cellular debris by a sieving mechanism
with those of the large-scale system. UV detectors
at the membrane surface. Pressure is applied to the feed
may be inaccurate or nonlinear over the optical den-
stream to force permeate through the membrane. Pro-
sity (OD) ranges required. If the absorbance range is
teins and small-molecular-weight species (media salts)
nonlinear at 280 nm, it may be prudent to measure
pass through the membrane and are collected in the per-
the online OD at 300 nm or some other wavelength
meate stream (12).
that has a lower extinction coefficient (13).
Ultrafiltration (UF) and diafiltration (DF) are com-
monly used in downstream processing for product con-
Qualification of Scale-Down Model
centration and buffer exchange, respectively. In some
Attributes that need to be compared would depend on the cases, it is also used for fractionation or purification.
objective of the step and could include chromatogram, Membranes are rated by molecular weight cutoffs, called
product concentration, product recovery, quality nominal molecular weight cutoffs (NMWC). Membrane
attributes (concentration of the various product-related, structure can influence the quality and robustness of sep-
process-related, and host-cell-related impurities), and aration. Minimizing macrovoids enables the use of higher
other feed stream characteristics (such as pH and pressures and higher temperatures and gives better sep-
conductivity). arations because of sharp NMWC. Cassettes are the first
Factors at the large scale such as column-header design choice for most protein processing. Hollow fibers are more
and packing can be difficult to control and may result in appropriate when the product is sensitive to shear or
reduced accuracy of the scale-down model (13). Differences a high viscosity solution needs to be processed such as
in column packing at the two scales may have an impact adenoviral vectors and influenza vaccines (113).
on separation and result in differences in chromatograms. Size is the basis of separation in filtration in most
Height equivalent to theoretical plate (HETP) and asym- cases. The solution is contacted with the membrane under
metry factor (As ) are typically used to evaluate quality of an applied pressure (114). The applied pressure forces
column packing (103,104). While these values are likely the solvents or buffer salts and the smaller molecules to
to be different across scales, they should be monitored to migrate through the membrane. The membrane retains
ensure consistency of packing quality during small-scale larger molecules such as proteins. MF and UF operations
experimentation. Furthermore, differences in equipment can be performed in a tangential flow filtration (TFF)
such as tubing length and diameter, monitors, and pumps mode or normal flow filtration (NFF) mode. An example of
should also be carefully examined to ensure comparability NFF would be dead-end or cartridge filters where cells are
of performance across scales (12). retained at the membrane surface or within the membrane
Table 2 summarizes the literature on scale-down or structure. As cells build up at the membrane surface, the
process chromatography. Researchers have used micro- filtration rate, or flux, decreases as the resistance to perme-
liter resin wells (as in 96-well plates) for performing ate flow through the membrane increases. In TFF, the feed
chromatography. A lot of effort has also undergone into solution is passed tangentially over the membrane surface.
modeling of chromatography so that the data generated Cells build up on the membrane surface as the permeate
from scale-down models can be applied toward prediction flows through the membrane and are swept off the mem-
of performance at manufacturing scale. The optimal bal- brane and recirculated back to the feed tank (51). UF is
ance between the high throughput that the microdevices typically conducted in a TFF mode where the feed solution
SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS 9

Table 2. Review of Literature for Development of Scale-Down Models for Process Chromatography

Topic Summary References


General Different formats of microscale devices for scale-down of chromatographic separations. (94)
Advice on how best to operate these devices, examples of use, comparison with
respect to metrics such as cost, usability, and role in QbD
Scale-down experimentation integrated with general rate modeling, a 2.5 cm height, (98)
1 mL column model calibrated against 20 cm height columns from 40 mL to 160 L
volume. Simulations in agreement with experimental results. Overall objective to
reduce the number of experiments, shorten development time, and reduce costs
Novel ultra-scale-down approach that uses empirical correlations derived from (105)
conductivity changes during operation to correct chromatographic profiles for
differences in dispersion and retention. Tested successfully with 1 mL column to
predict elution profiles of a chimeric monoclonal antibody obtained from protein A
chromatography columns of 3 mL and 18.3 L scale
Affinity chromatography Development of software that uses data obtained from scale-down column studies to (106)
relate yield to protein load and the number of matrix reuses to enable decision
making for chromatographic separations of antibodies
Mass balance of protein and Fab fragments, measurement of capacity from batch (107)
uptake kinetics and use of an imaging technique to investigate the impact of varying
column loading and resin reuse on matrix performance. Using a 20-fold scale-down,
number of reuses before recovery starts decreasing was established, as in current
large-scale manufacturing operations
Cation exchange Influence of pH for optimized binding and removal of HCP. Initial screening was done (108)
chromatography by batch-binding capacities, followed by scale-down to 96-well plate format and
proved that assay miniaturization still provided reliable data
Use of scale-down and scouting studies to assist in scaling up a method for isolating (109)
potentially clinically relevant monoclonal antibody C595 from heterogeneous
feedstocks
Expanded Bed chromatography The hydrodynamic model was in agreement with experimental results obtained with 1- (110)
and 5-cm column diameters with buffer solutions of different viscosities. Supported
by visual observations and scale-down model predictions, it was concluded that
operating conditions (liquid viscosity and superficial velocity) resulting in a bed-void
fraction between 0.7 and 0.75 would provide the optimal separation efficiency in
terms of plate number
Suitability of expanded bed chromatography as a scale-down technique to predict the (111)
behavior at large-scale was examined. The performance of three columns of diameters
5.0, 1.0, and 0.5 cm were compared in terms of the bed expansion, hydrodynamics,
and breakthrough for lysozyme adsorption onto STREAMLINE-SPTM. Performance
was similar across a scale-down factor of a 100-fold
Hydrophobic interaction Development and scale-up of a preparative HIC step for a therapeutic protein, removal (97)
chromatography (HIC) of product- and process-related impurities, and direct transferability of the
scale-down optimization experiments via the pilot scale to the final production scale
Annular chromatography Mathematical model to estimate the minimum radius of an annular chromatograph. (112)
The minimal geometry of an annular chromatograph was calculated for a model
system immunoglobulin and bovine serum albumin.

is recirculated over the membrane and then returned to flow velocity, membrane material and pore size, channel
the feed vessel. UF membranes are available in nominal height, flow path geometry, and retentate and filtrate pres-
molecular weight limits of 1000–1,00,000 Da (115–118). sures (Table 1). The membrane area is linearly decreased
A typical system consists of a feed pump, strainer, rack as per the desired feed volume such that the filtrate volume
equipped with filtration modules, filtrate/backwash tank, to membrane surface area ratio stays constant (119,120).
and connecting piping and valves. A separate pump is In the case of hollow fibers, linear scaling with hollow
used to add DF buffer to the recirculation vessel. Control fibers is readily accomplished by producing scale-down
parameters may include feed and retentate pressure (the modules in which fiber length is equal to that of industrial
permeate pressure is usually atmospheric for UF appli- scale systems. Equal flow distribution between fibers and
cations), temperature, and the feed flow rate per feed cartridges is easily accomplished through proper piping
channel. manifold and cartridge housing design. Flat sheet designs,
however, provide a suitable geometric format for a wide
Scale-Down Modeling Approach
range of linear-scale UF.
TFF has traditionally been scaled up by maintaining the The challenge with using scale-down systems for
following parameters constant: filtrate volume to mem- performing MF or UF/DF is that the small-scale devices
brane surface area ratio, transmembrane pressure, cross that are commercially available may differ from their
10 SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS

large-scale counterparts with respect to their design. flow operation by an USD rotating disc filter (RDF) has also
While cassette designs are based on parallel addition of been reported (122). The RDF has been modified by build-
membrane channels, the feed-port channels are added ing in inserts to allow the flexibility of the chamber volume,
in series. This directly impacts the fluid dynamics of so that only 1.5 mL of processing material is required for
the manufacturing scale cassettes and the scale-down each DF experiment. Wall shear rate correlations have
devices, resulting in differences in flow distribution, been established for at both the laboratory-scale cas-
channel height compression, and entrance/exit effects. All sette and the USD device, and identical performance was
these factors impact the performance of the step. Equal achieved by operating both scales under conditions with
flow distribution throughout the system is important to equivalent-averaged shear rates. Predicted flux and trans-
maintain both the system average and system limit values mission data agreed well with the experimental results of
of feed channel pressure drop (PL ), transmembrane a laboratory-scale DF where the cassette resistance was
pressure (TMP; PTM ), filtrate flux (Jf ), and protein considered (122). In another publication, the authors have
concentration at the membrane wall (Cw ) constant at described operation of a continuous mode laboratory fil-
all scales of operation. Channel height compression is ter to prepare filter cakes and filtrate similar to what
dependent on the physical deformation characteristics may be achieved at the industrial scale (123). Analysis of
of membrane, spacer screen, encapsulant, and gaskets. the filtration rate profile indicated the filter cake to have
Variations in channel height will have a significant impact changing properties (compressibility) with time. Results
on both pressure losses and mass transfer coefficients. from the USD filter were used to predict how the cake
Hence, process performance will vary at different scales of compressibility changes with time and thus allowed for
operation if consistent compression is not achieved (119). accurate predictions about the flux profile in large-scale
filtration. Furthermore, the USD filter allowed the pro-
Qualification of Scale-Down Model duction of materials (cake and filtrate) representative of a
Since MF and UF/DF steps do not alter the product purity large-scale continuous filter.
(in most cases) and are generally used for clarification In summary, there has been significant progress in
or concentration or buffer exchange, step recovery may creating scale-down devices for UF/DF unit operation in
be determined using UV absorbance at 280 nm. In cases the past two decades. Two aspects remain that require
where the sample matrix interferes with the UV signal, some consideration from the users. First is to ensure that
use of HPLC or product-specific assays may be required. the design of the scale-down device is comparable to that
Mass balance is required for determining if the product of the large-scale device so that the flow pattern is similar
is being lost into the permeate stream, left behind in if not identical. Second, the dead volume of the system
the holdup volume, or irreversibly bound to membrane. (volume of the connected tubings, pump, and other parts
A comparison of the flux versus TMP curve across scales of the setup besides the cassette) is always significant
provides a good comparison of performance. If the UF sys- when compared to the cassette volume at small scale.
tem is being used for purification, analysis of the impurity Very high dead volume will result in underperformance of
concentration is a key measure of step effectiveness. It is the UF/DF step and limit the usefulness of the scale-down
important that appropriate cleaning is performed on the model. The user should carefully select the equipment that
small-scale system so as to achieve consistent performance has a low dead volume.
over long-term use (12).
VIRAL CLEARANCE VIA CHROMATOGRAPHY AND
Literature Review
FILTRATION
A comprehensive methodology for implementation of lin-
ear scale-up and scale-down of TFF process has been devel- Assuring the viral safety of plasma-derived biologicals and
oped by Reis and coworkers (119). The researchers modi- biopharmaceuticals is critical for safety of health care con-
fied the cassette manufacturing procedures and tolerances sumers. Incidences of contamination of products derived
to achieve linear scale-up of UF processes for recovery for from human plasma in the past have adversely impacted
a ribosomal deoxyribonucleic acid (rDNA)-derived human the health of hundreds of patients and tainted the image of
therapeutic. A 400-fold linear scale-up was achieved with- certain segments of the health care industry (124). Owing
out intermediate pilot-scale tests. In another publication, to stringent measures taken by the industry and regula-
novel pleated scale-down devices (Am = 1.51 to 15.1 × tors to mitigate viral safety risks, similar incidents have
10−3 m2 ) have been designed and fabricated. It was not occurred in the history of biotech-based therapeutics in
shown that these can more accurately predict the per- the recent decades (124). Viral clearance can occur either
formance of industrial scale single-use pleated membrane via process chromatography (the virus not binding to the
cartridges (Am = 1.06 m2 ) that are commonly used within resin while the product binds) or nanofiltration (viruses
biopharmaceutical manufacture (121). The authors found cannot pass through the nanofilters). The aspects related
that single-use scale-down cartridges retain the same to process chromatography are already covered in the
pleat characteristics of the larger cartridges but require above section. In this section, we focus on viral clearance
a reduced feed volume by virtue of a substantially dimin- via nanofiltration.
ished number of active membrane pleats. A 1000-fold Nanofiltration is a pressure-driven process where per-
scale-down was achieved (121). A method to mimic flux meate is forced through a semipermeable membrane with
and transmission performance in a laboratory scale cross typical pore size in the range of 15–50 nm (newer products
SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS 11

have pores in the narrower range of 15–20 nm). This pore correlation. The operation of scale-down viral clearance
size distribution allows for passage of the product and systems can present complications. When using multi-
buffer ingredients while retaining viral particles. Viral layer membrane disks for evaluation, specifically designed
clearance devices are used once and then discarded. Viral holders may have to be used to provide the required seal-
clearance and design considerations have been reviewed ing between the membrane sheets to prevent feed material
in several publications (125–127). from bypassing layers. Scale-down viral clearance devices
Virus clearance studies are always performed with may have the same membrane in the production scale
scale-down models of the manufacturing process steps. devices, but the feed-side flow geometries of the scale-down
It is not feasible to perform viral clearance studies at the devices may also not be consistent with the production
manufacturing scale because it would be inappropriate modules. Feed-side path lengths may not be consistent
to introduce infectious virus into a cGMP manufactur- and feed flows may have to be adjusted to provide equiva-
ing facility. Also, the volumes of virus needed to achieve lent shear forces at the membrane surface. Similar to UF
a satisfactory spiking level at the manufacturing scale application, air–liquid interfaces should be minimized to
would be impractical and prohibitively expensive. Thus, prevent protein denaturation and aggregation (12).
in order for the virus clearance studies to be extrapo-
lated to the manufacturing scale, it is imperative that the Scale-Down Qualification
scale-down model is a true representation of the full-scale
The step recovery and purity for viral clearance can be
manufacturing process. Typically, the virus is spiked into
determined using HPLC or product-specific assays. Aggre-
the relevant intermediate, and the spiked material is pro-
gate generation can be monitored using SE-HPLC. When
cessed across the scaled down unit operation (both for
pure proteins are used, UV absorbance at 280 nm can be
process chromatography and nanofiltration). The reduc-
used for yield measurements. Creating a mass balance
tion in the virus load by the unit operation demonstrates
is useful to estimate losses coming from dead volume in
the effectiveness of the process step for virus removal or
the system or irreversible binding to the membrane. A
inactivation. The virus spike used in viral clearance stud-
comparison of the process flux as a function of time and
ies should be representative of a potential contaminant to
pressure over the whole process is also an indicator of
the extent achievable. It is important not only to select
comparability of performance.
appropriate relevant or model viruses but also to consider
the properties of the virus spike (124). For example, the
Literature Review
presence of serum in a virus spike may be problematic for
a validation study of a serum-free manufacturing scheme. Zhou et al. (128,129) have evaluated different scale-down
It is important that contaminants in the virus spike itself models and proposed a new model to mimic the liquid flow
do not impact key or critical performance parameters in path found in the large-scale capsule. They were able to
a way that makes the scale-down model unrepresentative achieve same capacity at small scale as at larger scale.
of the large-scale process (116). The product fraction post Results from a four-model virus study with a redesigned
filter is then assayed for residual virus infectivity. This Sartobind Q absorber scale-down model at the new pro-
enables a reduction factor to be calculated as follows: cess capacity were presented. The authors have discussed
problems faced with the Q-membrane scale-down model
Viral reduction as well as the proposal of the new model. The proposed
Total virus measured in spiked load Q-membrane scale-down model had a process capacity
= log10
Total virus recovered in the product fraction greater than 3000 g/m2 or 10.7 kg/L with a log reduction
(5) in viability (LRV) over 5 for four-model viruses.
In summary, scale-down modeling for viral clearance
Scale-Down Modeling Approach through process chromatography and nanofiltration is
widely practiced as every biotech company does these
The loading, as measured in volume of load material fil- studies as per the regulatory requirements. The existing
tered per unit surface area of the filter, should be kept models work reasonably well, but as discussed in the ref-
constant across the two scales. A typical load material erences mentioned in this section, there is scope of further
from an earlier processing step obtained from manufac- improvements with respect to the scale of operation. If the
turing should be used for the scale-down studies. Since scale can be further reduced, the cost of the studies will be
this step is usually carried out under constant pressure, significantly lowered as well.
the inlet pressure should be matched for the two scales and
similar flush volumes in terms of liters of buffer flushed to
recover any protein held up in the pores per unit surface VIRAL INACTIVATION
area should be used. An important output parameter is
the average volumetric flux as a function of volumetric Viral inactivation, along with other steps for viral clear-
capacity. ance discussed above, plays a key role in biotech processes
The membranes used for viral clearance can be oper- in demonstrating virus clearance capability. It is a part
ated in direct flow or TFF mode. For viral clearance of the monoclonal antibody platform in most companies
applications, it is required that viral clearance devices (130). Most commonly used approach for viral inactivation
have an integrity test that is correlated to virus removal is acidic pH/low pH inactivation, and this is the focus of
performance. The manufacturer generally provides this this section.
12 SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS

The inactivation of enveloped viruses by low pH is a profile similar to that used in the manufacturing scale
very common step in a monoclonal antibody purification (12).
process. The working principle is that some viruses, when
exposed to a low pH, denature spontaneously. Typically, Literature Review
low pH inactivation step is performed after the protein A
affinity chromatography step. The product from the pro- Not much has been written on the topic of scale-down of
tein A affinity column is titrated to a pH of 3.8 or lower viral inactivation, perhaps because of the simple design of
and incubated for a period of 15–60 min, depending on such a system.
the stability of the monoclonal antibody under considera- In summary, scale-down of viral inactivation is quite a
tion. This brief exposure to low pH effectively inactivates simple exercise. However, care should be taken that the
most lipid-enveloped viruses. Following this incubation, mixing is equivalent so that the end results are comparable
the protein solution is then titrated upward to a pH of across scales.
5.0 or higher to prepare for the next step. While the
inactivation of viruses is favored by low pH, there is an MEMBRANE ADSORBERS
inherent risk of aggregating the target protein at these
low pH conditions. Moreover, a strongly acidic solution Membrane chromatography is increasingly becoming pop-
that is used as a titrating solution for the low pH step can ular as an alternative to certain chromatographic steps,
cause localized low pH conditions, if not adequately mixed. offering higher productivity (131–138). In the membranes,
This could potentially cause aggregation of the protein binding sites are located along the through pores rather
solution. than nestled with long diffusive pores. Even in the newer
chromatography resins with ‘‘through’’ pores or convective
Scale-Down Modeling Approach pores (139), the liquid has the choice to flow through the
Scale-down of a low pH inactivation step is relatively easy. particle or around it. Unlike this, in a stacked membrane,
Care must be taken to ensure adequate mixing at this there is no choice but to flow through the pores. This
step during the addition of the inactivating agent such reduces the mass transport resistance for the solute to the
that no undue aggregation of the target protein results matrix by eliminating pore diffusion, leaving film diffusion
across this step. As mentioned before, the mass transfer from the core of the liquid to the membrane surface in the
coefficient (kL a) can be used to define mixing conditions at interior of a through pore as the only transport resistance.
small scale that will mimic conditions used in the man- As film diffusion is usually orders of magnitude faster
ufacturing scale. The scale-down factors for these types than pore diffusion, mass transport limitations are drasti-
of virus inactivation steps may be large, as there are few cally reduced in membrane chromatography, shifting the
limitations to designing a temperature-controlled contact- limitation of the process more to the properties of the
ing vessel, which simply needs to maintain well-mixed matrix–solute interactions.
solution. Test tubes or small vessels may be used, if
they are immersed in a temperature-controlled heating Scale-Down Modeling Approach
block, and either mixed before or during contact. The
Most of the scale-down systems that are available in the
level of the heating solution should be above the liq-
market do not have a liquid flow path similar to that
uid level in the test vessel to ensure all the test solu-
found in the large-scale capsule and thus tend to generate
tion contents have reached the target temperature. A
extremely high operational backpressure when a high flow
high temperature short-time treatment can be employed
rate is applied (≥ 450 cm/h). The axial velocity of a mobile
using microwave assistance, but this system needs careful
phase is faster in the center of the membrane adsorber
scale-down using miniature model of the process equip-
(MA) unit than near the edges of adsorptive bed (106).
ment, which could limit the scale-down to more modest
The decrease in apparent porosity from the center of the
ratios. The mixing of the vessel contents plays impor-
bed to the gasket area or the edges of the adsorptive bed
tant role in low pH assisted inactivation. Standard mixing
may account for the decrease in velocity observed adjacent
studies must be employed and the kinetics of inactivation
to the gasket or edges (106,140). In contrast, radial flow
should be measured, as it is a regulatory requirement.
adsorbers, prepared by spirally winding a flat sheet mem-
The addition rate of inactivation chemicals should match
brane over a porous cylindrical core, are used in a capsule
the manufacturing process, and the final product con-
for large-scale process (114). Distorted or poor inlet flow
centration of acid should be same as that of full scale.
distribution has less effect in a large-scale capsule, thus
Scale-down variables that should be controlled through-
leading to a smaller operational backpressure drop for
out the inactivation process are pH, temperature, con-
large-scale operation (17,18). Despite being very complex
tact time, and the final inactivation chemical concentra-
to model in a radially outward direction, radial flow adsor-
tions.
bers are claimed to be suitable for large-scale application
(114). The scalability and operating backpressure, there-
Scale-Down Qualification
fore, are main issues for the MA scale-down model. An
The aggregate content of the protein during scale-down extremely high operational backpressure observed with
studies is an important quality attribute to match across the scale-down model can lead to considerably less capacity
scales for qualification of the scale-down model. The chem- and an oversized membrane at full scale. This oversizing
icals used for inactivation should show concentration may lead to an erroneous economic calculation. Larger
SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS 13

scale membranes do not have backpressure issues. How- LIST OF ABBREVIATIONS


ever, the use of a large-scale MA in a scale-down model
is also not practical, especially for a viral clearance study CD circular dichroism
because of the high cost of viruses and feedstock. cGMP current good manufacturing practice
DLS dynamic light scattering
DNA deoxyribonucleic acid
Scale-Down Model Qualification HCP host cell proteins
These aspects are same as in process chromatography. HETP height equivalent to theoretical plate
HPH high pressure homogenization
HPLC high pressure liquid chromatography
Literature Review HTPD high throughput process development
IBs inclusion bodies
Zhou et al. have redesigned scale-down models for
ICH International Conference on
Q-membrane, Q125 and Q40 (128,129). These scale-down
Harmonization
models mimic the liquid flow path found in the large-scale
LPO lactoperoxidase
modules and achieve performance parameters comparable
MA membrane adsorber
to those of the larger Q units. However, even with
MF microfiltration
improved scale-down devices, the operational backpres-
MVA multivariate analysis
sure for Q-membrane chromatography still influences
NFF normal flow filtration
process capacity. Several researchers have focused on
NMWC nominal molecular weight cutoffs
creating an improved scale-down model for membrane
OD optical density
chromatography (128,129).
PAT process analytical technology
In summary, the newer scale-down models seem to
RDF rotating disc filter
have addressed the most critical of the problems that one
rDNA ribosomal deoxyribonucleic acid
faces. However, not all manufacturers offer such devices.
SDS-PAGE sodium dodecyl sulfate polyacrylamide gel
Careful comparison of the performance of the device should
electrophoresis
be done in order to understand the differences between
SE-HPLC size-exclusion high pressure liquid
the two scales, and the resulting data should be suitably
chromatography
interpreted.
TCA trichloroacetic acid
TFF tangential flow filtration
UF ultrafiltration
SUMMARY USD ultra-scale-down
UV ultraviolet
In the era of QbD and PAT, use of scale-down models
has become an integral part of characterizing and vali-
dating industrial processes. Characterization studies form
the backbone of experimentation intended to provide pro- NOMENCLATURE
cess knowledge. These studies are used to identify critical
parameters and to establish acceptable ranges, process 
equivalent settling area of a laboratory centrifuge
validation acceptance criteria, and specifications. Process 2
 (m )
validation studies such as evaluation of viral clearance and 2
c equivalent settling area (m )
impurity clearance require such scale-down models, as it As asymmetry factor
is not feasible from a safety perspective to perform them at BL caulk width (m)
manufacturing scale. This article presents general guide- Ca camp number
lines and relevant parameters affecting scale-down of the Cw protein concentration at the membrane wall
various unit operations involved in purification of proteins. (kg/m3 )
The key parameters, assessment methods, assessment PL feed channel pressure drop (kg/ms2 )
techniques, and challenges involved in scale-down are PTM transmembrane pressure (Pa)
discussed. The quality of the existing scale-down mod- f1 correction factor for spacer caulks
els varies from unit operation to unit operation. Factors g acceleration due to gravity (m/s−2 )
that complicate the scale-down process include difficulties G average value of velocity gradient
in mimicking the hardware design resulting in differ- Jf filtrate flux (m3 /s)
ences in hydrodynamics across scales (mixing, shear, kL a mass transfer coefficient
etc.), higher holdup volumes at smaller scales, and dif- LRV log reduction in viability
ferences in materials used for making the equipment and N rotational speed (rps)
the separation media. A lot of progress has been made nd number of active discs
in each of these areas but with the growing importance ω angular velocity (radians/s)
of scale-down models, scope for more work remains. We P power dissipated (W)
hope the chapter would be valuable to those engaged in Po power number
small-scale development, optimization, and characteriza- Q liquid flow rate through the centrifuge (m3 /s)
tion of biotech processes. Qc volumetric flow rate (m3 /s)
14 SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS

r1 inner disc radius (m) 21. Breece T, Gilkerson E, Schmelzer C. BioPharm 2002; 15:
r2 outer disc radius (m) 16–20.
Re Reynolds number 22. Kelley BD. In: Sofer G, Zabriskie D, editors. Biopharmaceu-
Ri inner radius (the distance between the center of tical process validation. New York: Marcel Dekker; 2000.
rotation and the top of the liquid) (m) pp. 29–59.
Ro outer radius (the distance between the center of 23. Box GEP, Hunter WG, Hunter JS. Statistics of experiments.
rotation and the bottom of the tube) (m) New York: John Wiley and Sons; 1978.
S solids remaining (kg) 24. Seely R, Wright H, Fry H, Rudge S, Slaff G. BioPharm 1994;
θ half disc angle 7: 41–48.
TMP transmembrane pressure (Pa) 25. Rathore AS, Sofer G. In: Rathore AS, Sofer G, editors.
μ dynamic viscosity (Pa·s) Process validation in manufacturing of biopharmaceuticals.
V sample volume (L) Boca Raton (FL): Taylor & Francis Group LLC; 2005. pp.
169–204.
x fractional time required for acceleration (s)
y fractional time required for deceleration (s) 26. Pieracci J, Yusuf-Makagiansar H. In: Rathore AS, Sofer
G, editors. Process validation in manufacturing of biophar-
ZL number of caulks on a disc
maceuticals. Boca Raton, FL, USA CRC press, Taylor and
Francis Group; 2012.
27. Wold S, Geladi P, Esbensen K, Öhman J. J Chemom 1987;
1: 41–56.
REFERENCES 28. Tustian AD, Salte H, Willoughby NA, Hassan I, Rose MH,
1. Wheelwright SM. J Biotechnol 1989; 11: 89–102. Baganz F, Hoare M, Titchener-Hooker NJ. Biotechnol Prog
2007; 23: 1404–1410.
2. Rathore AS. Trends Biotechnol 2009; 27: 698–705.
29. Axelsson H. In: Flickinger MC, Drew SW, editors. Encyclo-
3. Willoughby N, Martin P, Titchener-Hooker N. Biotechnol
pedia of bioprocess technology. New York: John Wiley and
Bioeng 2004; 87: 641–647.
Sons; 1999. pp. 513–531.
4. Boychyn M, Doyle W, Bulmer M, More J, Hoare M. Biotech-
30. Hanle DD. In: Flickinger MC, Drew SW, editors. Encyclo-
nol Bioeng 2000; 69: 1–10.
pedia of bioprocess technology. New York: John Wiley and
5. Bhambure R, Kumar K, Rathore AS. Trends Biotechnol Sons; 1999. pp. 553–559.
2010; 29: 127–135.
31. Kempken R, Preissmann A, Berthold W. J Ind Microbiol
6. Rathore AS, Winkle H. Nat Biotechnol 2009; 27: 26–34. 1995; 14: 52–57.
7. Rathore AS. Trends Biotechnol 2009; 27: 546–553. 32. Maybury JP, Mannweiler K, Titchener-Hooker NJ, Hoare
8. Xu Z, Li J, Zhou JX. Prep Biochem Biotechnol 2012; 42: M, Dunnill P. Bioprocess Biosyst Eng 1998; 18: 191–199.
183. 33. Leung WWF. Industrial centrifugation technology. New
9. Read EK, Park JT, Shah RB, Riley BS, Brorson KA, Rathore York: McGraw Hill; 1998.
AS. Biotechnol Bioeng 2010; 105: 276–284. 34. Maybury JP, Hoare M, Dunnill P. Biotechnol Bioeng 2000;
10. Read EK, Park JT, Shah RB, Riley BS, Brorson KA, Rathore 67: 265–273.
AS. Biotechnol Bioeng 2010; 105: 285–295. 35. Mosqueira FG, Higgins JJ, Dunnill P, Lilly MD. Biotechnol
11. Rathore AS, Bhambure R, Ghare V. Anal Bioanal Chem Bioeng 1981; 23: 335–343.
2010; 398: 137–154. 36. Mannweiler K, Hoare M. Bioprocess Eng 1992; 8: 18–25.
12. Godavarti R, Petrone J, Robinson J, Wright R, Kelley BD, 37. Betts JI, Baganz F. Microb Cell Fact 2006; 5: 21.
Bolton GR. In: Rathore AS, Sofer G, editors. Process valida-
38. Gill NK, Appleton M, Baganz F, Lye GJ. Biochem Eng J
tion in manufacturing of biopharmaceuticals. Boca Raton
2008; 39: 164–176.
(FL): Taylor & Francis Group LLC; 2005. pp. 69–142.
39. Pampel L, Mcnerney T, Wypych J. Pap Am Chem Soc 2004;
13. Rathore AS, Krishnan R, Tozer S, Smiley D, Rausch S,
227: 264–BIOT.
Seely J. BioPharm Int 2005; 18: 58–60.
40. Duetz WA. Trends Microbiol 2007; 15: 469–475.
14. Tait AS, Aucamp JP, Bugeon A, Hoare M. Biotechnol Bioeng
2009; 104: 321–331. 41. Fernandes P, Cabral JMS. Biocatal Biotransformation
2006; 24: 237–252.
15. Cecchini DJ. In: Rathore AS, Mhatre R, editors. Quality by
design for biopharmaceuticals. Hoboken, New Jersey John 42. Islam RS, Tisi D, Levy MS, Lye GJ. Biotechnol Prog 2007;
Wiley and Sons; 2008. pp. 127–142. 23: 785–793.
16. Kozlowski S, Swann P. In: Rathore AS, Mhatre R, editors. 43. Islam RS, Tisi D, Levy MS, Lye GJ. Biotechnol Bioeng 2008;
Quality by design for biopharmaceuticals. Hoboken, New 99: 1128–1139.
Jersey John Wiley and Sons, 2008, pp. 9–30. 44. Micheletti M, Lye GJ. Curr Opin Biotechnol 2006; 17:
17. Points to Consider in the Manufacture and Testing of 611–618.
Monoclonal Antibody Products for Human Use. Center for 45. Micheletti M, Barrett T, Doig SD, Baganz F, Levy MS,
Biologics Evaluation and Research, F.D.A.; 1997. Woodley JM, Lye GJ. Chem Eng Sci 2006; 61: 2939–2949.
18. Banerjee A. Biopharm Int 2010; 23: 26. 46. Hutchinson N, Bingham N, Murrell N, Farid S, Hoare M.
19. Adner N, Sofer G. BioPharm 1994; 7: 44–48. Biotechnol Bioeng 2006; 95: 483–491.
20. Leonard MW, Sefton L, Costigan R. In: Kelley BD, 47. Zaman F, Allan CM, Ho SV. Biotechnol Prog 2009; 25:
Ramelmier RA, editors. Validation of biopharmaceuti- 1709–1716.
cal manufacturing processes. Washington (DC): American 48. Sommer B, Friehs K, Flaschel E, Reck M, Stahl F, Scheper
Chemical Society; 1998. pp. 55–68. T. J. Biotechnol 2009; 140: 194–202.
SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS 15

49. Ariga O, Watari T, Andoh Y, Fujishita Y, Sano Y. J Ferm 81. Mannall GJ, Myers JP, Liddell J, Titchener-Hooker NJ,
Bioeng 1989; 68: 243–246. Dalby PA. Biotechnol Bioeng 2009; 103: 329–340.
50. Zhao FS, Yu JY. Biotechnol Prog 2001; 17: 490–494. 82. Qoronfleh MW, Hesterberg LK, Seefeldt MB. Protein Expr
51. Malamy MH, Horecker BL. Biochemistry 1964; 3: Purif 2007; 55: 209–224.
1889–1893. 83. Ordidge GC, Mannall GJ, Liddell J, Dalby PA, Micheletti
52. Clarkson AI, Lefevre P, Titchener-Hooker NJ. Biotechnol M. Biotechnol Prog 2012; 28: 435444
Prog 1993; 9: 462–467. 84. Fisher RR, Glatz CE. Biotechnol Bioeng 1988; 32: 777–785.
53. van Hee P, Middelberg APJ, van der Lans RGJM, van der 85. Glatz CE, Hoare M, Landa-Vertiz J. AIChE J 1986; 32:
Wielen LAM. Biotechnol Bioeng 2004; 88: 100–110. 1196–1204.
54. Kloosterman J, Vanwassenaar PD, Slater NKH, Vander- 86. Rothstein F. Protein process engineering. New York: Marcel
padt A. Chem Eng J 1988; 37: B47–B54. Dekker; 1994.
55. Limon-Lason J, Hoare M, Orsborn CB, Doyle DJ, Dunnill 87. Bell DJ, Dunnill P. Biotechnol Bioeng 1982; 24: 1271–1285.
P. Biotechnol Bioeng 1979; 21: 745–774.
88. Smoluchowski M. Z Phys Chem 1917; 92: 129–168.
56. Ricci-Silva ME, Vitolo M, Abrahao-Neto J. Process Biochem
89. Rushton JH, Costich EW, Everett HJ. Chem Eng Prog 1950;
2000; 35: 831–835.
46: 467–476.
57. Schutte H, Kula MR. Biotechnol Appl Biochem 1990; 12:
90. Bell DJ, Brunner K-H. Filt Sep 1983; 20: 274–286.
599–620.
91. Mannweiler K, Titchener-Hooker NJ, Hoare M. IChemE
58. Engler CR. Bioprocess Technol 1990; 9: 95–105.
Symp, Adv Biochem Eng/Biotechnol 1989; 1: 105–117.
59. Hetherington PJ, Follows M, Dunnill P, Lilly MD. Trans
92. Garcia-Ochoa F, Gomez E. Biotechnol Adv 2009; 27:
Inst Chem Eng 1971; 49: 142–148.
153–176.
60. Middelberg APJ. Trans IChemE 1993; 71C: 215–219.
93. Chhatre S, Pampel L, Titchener-Hooker NJ. Biotechnol
61. Sauer T, Robinson CW, Glick BR. Biotechnol Bioeng 1989; Prog 2011; 27: 998–1008.
33: 1330–1342.
94. Chhatre S, Titchener-Hooker NJ. J Chem Technol Biotech-
62. Keshavarz-Moore E, Hoare M, Dunnill P. Enzyme Microb nol 2009; 84: 927–940.
Technol 1990; 12: 764–770.
95. Watanabe E, Tsoka S, Asenjo JA. Ann N Y Acad Sci 1994;
63. Kelly WJ, Muske KR. Bioprocess Biosyst Eng 2004; 27:
721: 348–364.
25–37.
96. Errson B, Ryden L, Janson JC. In: Janson JC, editor.
64. Gray PP, Dunnill P, Lilly MD. In: Terui G, editors. Fermen-
Protein purification: principles, high resolution methods,
tation technology today. Japan: Society of Fermentation
and applications. New Jersey: John Wiley and Sons; 2011.
Technology; 1972. pp. 347–351.
pp. 8–9.
65. Floury J, Bellettre J, Legrand J, Desrumaux A. Chem Eng
97. Graumann K, Ebenbichler A. Chem Eng Technol 2008; 28:
Sci 2004; 59: 843–853.
1398–1407.
66. Donsı̀ F, Ferrari G, Lenza E, Maresca P. Chem Eng Sci
98. Gerontas S, Asplund M, Hjorth R, Bracewell DG. J Chro-
2009; 64: 520–532.
matogr A 2010; 1217: 6917–6926.
67. Clark EDB. Curr Opin Biotechnol 1998; 9: 157–163.
99. Rubino M, Bailey M, Baker JC, Boose JA, Metzka L, Moore
68. Hevehan DL, De Bernardez Clark E. Biotechnol Bioeng V, Quertinmont M, Wiler W. In: Rathore AS, Sofer G,
1997; 54: 221–230.
editors. Process validation in manufacturing of biopharma-
69. Yasuda M, Murakami Y, Sowa A, Ogino H, Ishikawa H. ceuticals. Boca Raton (FL): CRC Press; 2005. pp. 545–564.
Biotechnol Prog 1998; 14: 601–606.
100. Smith TM, Wilson E, Scott RG, Misczak JW, Bodek JM,
70. Boyle DM, Buckley JJ, Johnson GV, Rathore AS, Gustafson Zabriskie DW. In: Kelley BD, Ramelmier RA, editors.
MG. Biotechnol Appl Biochem 2009; 54: 85–92. Validation of biopharmaceutical manufacturing process.
71. Buswell AM, Middelberg AP. Biotechnol Bioeng 2003; 83: Washington (DC): American Chemical Society; 1998. pp.
567–577. 80–92.
72. Lee C, Buswell A, Middelberg AP. Chem Eng Sci 2002; 57: 101. Viral safety of biotechnology products derived from cell lines
1679–1684. of human or animal origin, ICH Viral Safety Document.
73. Mannall GJ, Titchener-Hooker NJ, Chase HA, Dalby PA. International Conference on Harmonization; 1998.
Biotechnol Bioeng 2006; 93: 955–963. 102. Yamamoto S, Nomura M, Sano Y. J Chromatogr 1987; 409:
74. Goldberg ME, Rudolph R, Jaenicke R. Biochemistry 1991; 101–110.
30: 2790–2797. 103. Barry A, Chojnacki R. BioPharm 1994; 7: 43–47.
75. Katoh S, Katoh Y. Process Biochem 2000; 35: 1119–1124. 104. Sofer G, Hagel L. Handbook of process chromatography: a
76. Katoh S, Sezai Y, Yamaguchi T, Katoh Y, Yagi H, Nohara guide to optimization, scale-up, and validation. New York:
D. Process Biochem 1999; 35: 297–300. Academic Press; 1997.
77. Cowieson NP, Wensley B, Listwan P, Hume DA, Kobe B, 105. Hutchinson N, Chhatre S, Baldascini H, Davies JL,
Martin JL. Proteomics 2006; 6: 1750–1757. Bracewell DG, Hoare M. Biotechnol Prog 2009; 25:
78. Vincentelli R, Canaan S, Campanacci V, Valencia C, Maurin 1103–1110.
D, Frassinetti F, Scappucini-Calvo L, Bourne Y, Cambillau 106. Chhatre S, Thillaivinayagalingam P, Francis R,
C, Bignon C. Protein Sci 2004; 13: 2782–2792. Titchener-Hooker NJ, Newcombe AR, Keshavarz-Moore E.
79. Willis MS, Hogan JK, Prabhakar P, Liu X, Tsai K, Wei Y, Biotechnol Prog 2007; 23: 888–894.
Fox T. Protein Sci 2005; 14: 1818–1826. 107. Thillaivinayagalingam P, O’Donovan K, Newcombe AR,
80. Rathore AS, Sharma A, Chillin D. BioPharm 2006; 19: Keshavarz-Moore E. J Chromatogr B Analyt Technol
48–57. Biomed Life Sci 2007; 848: 88–96.
16 SCALEDOWN OF BIOPHARMACEUTICAL PURIFICATION OPERATIONS

108. Stein A, Kiesewetter A. J Chromatogr B Analyt Technol Report No. 47. University of Iowa: Parenteral Drug Associ-
Biomed Life Sci 2007; 848: 151–158. ation, Inc.; 2010.
109. Denton G, Murray A, Price MR, Levison PR. J Chromatogr 125. Levy RV, Philips MW, Lutz H. In: Meltzer TH, Jornitz
A 2001; 908: 223–234. MW, editors. Filtration in biopharmaceutical industry. New
110. Fenneteau F, Aomari H, Chahal P, Legros R. Biotechnol York: Marcel Dekker; 1998. pp. 619–646.
Bioeng 2003; 81: 790–799. 126. Kelley BD, Petrone J. In: Wang WK, editor. Membrane
111. Ghose S, Chase H. Bioseparation 2000; 9: 21–28. separations in biotechnology. New York: Marcel Dekker;
2001. pp. 351–396.
112. Uretschlager A, Jungbauer A. J Chromatogr A 2000; 890:
53–59. 127. Huang PY, Peterson J. In: Wang WK, editor. Membrane
separations in biotechnology. New York: Marcel Dekker;
113. Hagel L, Jagschies G, Sofer G. Handbook of process chro-
2001.
matography: development, manufacturing, validation and
economics. Netherland: Academic Press; 2008. 128. Zhou JX, Tressel T, Gottschalk U, Solamo F, Pastor A,
Dermawan S, Hong T, Reif O, Mora J, Hutchison F, Murphy
114. Mulder M. Basic principles of membrane technology. Dor-
M. J Chromatogr A 2006; 1134: 66–73.
drecht, The Netherlands: Kluwer Academic Publishers;
1996. 129. Zhou JX, Tressel T. Biotechnol Prog 2006; 22: 341–349.
115. Ma G, Aucamp J, Gerontas S, Eardley-Patel R, Craig A, 130. Shukla AA, Hubbard B, Tressel T, Guhan S, Low D. J
Hoare M, Zhou Y. Biotechnol Prog 2010; 26: 466–476. Chromatogr B Analyt Technol Biomed Life Sci 2007; 848:
28–39.
116. Reynolds T, Boychyn M, Sanderson T, Bulmer M, More J,
Hoare M. Biotechnol Bioeng 2003; 83: 454–464. 131. Rathore AS, Shirke A. Prep Biochem Biotechnol 2011; 41:
307–315.
117. Englard S, Seifter S. Methods Enzymol 1990; 182:
285–3006. 132. Thommes J, Kula MR. Biotechnol Prog 1995; 11: 357–367.
118. Paul EL, Rosas CB. Chem Eng Prog 1990; 8: 17. 133. Brandt S, Goffe RA, Kessler SB, O’Connor JL, Zale SE.
Bio/Technology 1988; 6: 779–782.
119. Stavrinides S, Ayazi Shamlou P, Hoare M. In: Ayazi
Shamlou P, editor. Processing of solid–liquid suspensions. 134. Briefs KG, Kula MR. Chem Eng Sci 1992; 47: 141–149.
London: Butterworth Heinman; 1993. pp. 118–158. 135. Charcosset C. J Chem Technol Biotechnol 1998; 71: 95–110.
120. Harrison RG. Protein purification process engineering. New 136. Lutkemeyer D, Bretschneider M, Buntemeyer H, Lehmann
York: Marcel Dekker; 1993. J. J Chromatogr A 1993; 639: 57–66.
121. Foster PR, Dunnill P, Lilly MD. Biotechnol Bioeng 1976; 137. Reif O-W, Freitag R. J Chromatogr A 1993; 654:29–41.
18: 545–580. 138. Riese U, Lutkemeyer D, Heidemann R, Buntemeyer H,
122. Iyer HV, Przybycien TM. AIChE J 1994; 40: 349–359. Lehmann J. J Biotechnol 1994; 34: 247–257.
123. Iyer HV, Przybycien TM. Biotechnol Bioeng 1995; 48: 139. Afeyan NB, Fulton SP, Gordon NF, Maszaroff I, Varady L,
324–332. Regnier F. Bio/Technology 1990; 8: 203–206.
124. Asher D, Brorson K, Hotta J, Hughes J, Martin J, Ruppach 140. Fisher RR, Glatz CE, Murphy PA. Biotechnol Bioeng 1986;
H, Sofer G, Wisher M, Willkommen H, Yang B Preparation 28: 1053–1063.
of virus spikes used for virus clearance studies. Technical
Please note that the abstract and keywords will not be included in the printed book, but are required for the
online presentation of this book which will be published on Wiley’s own online publishing platform.
If the abstract and keywords are not present below, please take this opportunity to add them now.
The abstract should be a short paragraph upto 200 words in length and keywords between 5 to 10 words.

Abstract: Biotech-based therapeutics have emerged as the growth engine for the pharmaceutical industry. Successful
development and commercialization of biotech processes require the understanding of the relationships between the
various attributes of the product and the product’s clinical safety and efficacy, as well as, the effect of the different
process parameters on the product’s attributes. The complexity of the biotech products and the requirement for rigorous
experimentation justify working at small scales.
This article presents general guidelines and relevant parameters affecting scale-down of the various unit operations
involved in purification of proteins. The key parameters, assessment methods, assessment techniques, and challenges
involved in scale-down are discussed.

Keywords: scale-down; biopharmaceutical; purification; quality by design (QbD);

Das könnte Ihnen auch gefallen