Biotechnology Progress - 2016 - Dutta - Performance Optimization of Continuous Countercurrent Tangential Chromatography For

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023].

See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Performance Optimization of Continuous Countercurrent Tangential
Chromatography for Antibody Capture
Amit K. Dutta, Jasmine Tan, and Boris Napadensky
Chromatan Corporation, 200 Innovation Blvd, Suite 260B, State College, PA 16803

Andrew L. Zydney
Dept. of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802
Oleg Shinkazh
Chromatan Corporation, 200 Innovation Blvd, Suite 260B, State College, PA 16803

DOI 10.1002/btpr.2250
Published online March 17, 2016 in Wiley Online Library (wileyonlinelibrary.com)

Recent studies have demonstrated that continuous countercurrent tangential chromatography


(CCTC) can effectively purify monoclonal antibodies from clarified cell culture fluid. CCTC
has the potential to overcome many of the limitations of conventional packed bed protein A
chromatography. This paper explores the optimization of CCTC in terms of product yield,
impurity removal, overall productivity, and buffer usage. Modeling was based on data from
bench-scale process development and CCTC experiments for protein A capture of two clarified
Chinese Hamster Ovary cell culture feedstocks containing monoclonal antibodies provided by
industrial partners. The impact of resin binding capacity and kinetics, as well as staging strat-
egy and buffer recycling, was assessed. It was found that optimal staging in the binding step
provides better yield and increases overall system productivity by 8–16%. Utilization of higher
number of stages in the wash and elution steps can lead to significant decreases in buffer
usage (40% reduction) as well as increased removal of impurities (2 log greater removal).
Further reductions in buffer usage can be obtained by recycling of buffer in the wash and
regeneration steps (35%). Preliminary results with smaller particle size resins show that the
productivity of the CCTC system can be increased by 2.5-fold up to 190 g of mAb/L of resin/
hr due to the reduction in mass transfer limitations in the binding step. These results provide
a solid framework for designing and optimizing CCTC technology for capture applications.
C 2016 American Institute of Chemical Engineers Biotechnol. Prog., 32:430–439, 2016
V
Keywords: continuous chromatography, monoclonal antibody, protein A, optimization, anti-
body purification

Introduction the binding, wash, elution, and regeneration steps occurring


sequentially (one after another). While effective and robust,
Monoclonal antibody (mAb) based treatments of cancer this method of manufacturing has caused major bottlenecks in
and autoimmune diseases have been established as one of downstream processing.4 The problem has become especially
the most successful therapeutic strategies. This class of drugs severe because of increasing bioreactor titers over the past 10
R R
includes such blockbusters as HerceptinV, RemicadeV, years in combination with the increased demands on cost
R R
HumiraV, RituxanV, and many others. There are currently reduction in mAb manufacturing.5 Additional drawbacks of
over 300 mAbs in various stages of clinical trials. These batch chromatography include low productivity (5–15 g of
products have driven much of the growth in the biotechnol- mAb/L of resin/hr is typical), high pressure operation, nonlin-
ogy industry over the last 20 years.1,2 ear scalability for large scale operations, and inability to inter-
Protein A column chromatography is currently used for ini- face with continuous production systems such as perfusion
tial capture of nearly all mAb products from clarified cell cul- bioreactors. Because of these inefficiencies, affinity columns
ture fluid (CCF). Protein A provides a highly robust method use very large volumes of resin that cost $10,000–$17,000 per
for use in platform processes, and it is currently considered liter. A single large scale protein A column designed for pri-
the most practical capture methodology for commercial pro- mary mAb capture can therefore cost manufacturers $15 mil-
duction of monoclonal antibodies.3 Protein A capture systems lion for the resin alone.6
typically use large stainless steel columns in batch mode with There is considerable interest throughout the biopharma-
ceutical industry in moving from batch to continuous proc-
Additional Supporting Information may be found in the online ver- esses. Continuous processing is currently the standard mode
sion of this article.
Correspondence concerning this article should be addressed to O. of operation in the chemical and food industries. There are
Shinkazh at oleg.shinkazh@chromatan.com. several advantages of continuous processing including: (1)

430 C 2016 American Institute of Chemical Engineers


V
15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Biotechnol. Prog., 2016, Vol. 32, No. 2 431

better quality control, (2) much lower capital cost, (3) lower via the retentate exit (the lumen side outflow). A similar
equipment footprint, and (4) enhanced flexibility in manufac- design is used in the subsequent steps.
turing.7,8 Another trend in bioprocessing is the application of The number of stages in the binding, washing, elution,
single-use equipment which can remove some of the major stripping, and regeneration steps can be adjusted to optimize
hurdles associated with biopharma manufacturing, such as system performance—this is discussed in more detail in the
cleaning, column packing, and validation while reducing the body of this paper. A two stage binding step is shown in Fig-
overall cost of production. ure 1C. In this case, the fresh resin is introduced via the first
Several efforts have been reported to develop continuous stage and is combined with partially bound mAb solution that
bioprocessing platforms. For example, Warikoo et al.9 and is recycled from the second stage. Fresh CCF is loaded on to
Godawat et al.10 used periodic counter-current (PCC) multi- the second stage and combined with partially loaded resin
column chromatography for continuous capture of mAb from from the first stage. This creates a unique countercurrent bind-
a perfusion bioreactor. They reported approximately five times ing configuration that is unavailable in conventional column
higher volumetric productivity than a fed batch process with chromatography. The resin from the second stage continues to
comparable yield and purity while the PCC column size was the “after binder” which binds all residual mAb before enter-
smaller than that used in the fed-batch process.9 In another ing the wash-1 step, while the CCF waste leaves the system
study, Pollock et al.11 studied the design and optimization of as first stage permeate. The impact of the after binder on
a PCC system. Muller-Spath et al.12 have used multicolumn product yield is discussed subsequently.
countercurrent solvent gradient purification (MCSGP) chroma- CCTC overcomes many of the limitations of conventional
tography with anion and cation exchange resins for antibody packed bed chromatography. The system operates at low
purification. One problem with multicolumn chromatography pressure using a fully disposable flow path, while the product
is that these systems require complex valve switching and is loaded and purified in a truly continuous steady state fash-
packing of multiple columns (unless operated with more ion. This is a major advantage vs. multicolumn chromatogra-
expensive prepacked columns). Also, they operate in a cyclic phy systems which operate in a continuous mode, but have a
rather than true steady state mode, which can lead to greater cyclic product discharge, causing eluted product concentra-
variability in product quality while creating challenges for tions to vary widely. While the CCTC system discharges a
subsequent downstream operations.13 A number of companies relatively dilute mAb concentration in comparison with that
have recently commercialized prepacked columns that could obtained from a multicolumn system, an in-line ultrafiltration
be applied in single-use applications. However, in manufactur- system can be used in a straightforward manner to control
ing, these are mostly targeted for multibatch clinical cam- the final product pool concentration to any desired level.
paigns in order to maximize utilization of the expensive Another key advantage of the CCTC system is that the pres-
resins, particularly protein A, to their full life cycles. Rosa sure drop is independent of the particle size, in sharp con-
et al.14 have examined aqueous two-phase extraction while trast to the behavior of packed columns. Thus, small particle
Hammerschmidt et al.15 have explored the use of precipitation size resins (10 mm) with low crosslinking densities can be
as alternatives to chromatography for steady state capture of used in the CCTC system; these small particles will have
product. However, unlike protein A affinity resins, these sys- much faster mass transfer rates, leading to greater overall
tems typically cannot provide robust operation and high productivity.
purity. Note that the membrane adsorbers developed by Natrix
Shinkazh et al.17 used CCTC for the separation of bovine
Separations Inc.16 are single-use, but they currently can only
serum albumin (BSA) and myoglobin using an anion
perform batch separations.
exchange resin. CCTC has also been used for purification of
The continuous countercurrent tangential chromatography an IgG4 from a mixture of BSA and myoglobin using a
(CCTC) system developed by Chromatan Corporation con- protein A affinity resin.18 More recently, Dutta et al.19 dem-
sists of a series of chromatographic steps, with each step onstrated the ability of CCTC to purify monoclonal antibod-
consisting of several stages. A CCTC stage is defined as a ies produced in CHO cells. Two different mAbs, with low
combination of a single hollow fiber membrane module and and high titers, were purified directly from clarified cell cul-
a static mixer.17 A schematic of the system is shown in Fig-
ture fluid using the CCTC system, with the overall produc-
ure 1A and includes six steps: binding, wash-1, wash-2, elu-
tivity nearly five times the productivity of a packed column
tion, stripping, and equilibration. The inset shows
with similar yield and purity.
countercurrent staging employed in the elution step. Similar
designs are used for the other steps (discussed in more detail The objective of this study was to examine the design and
subsequently). The static mixer provides contacting between optimization of the CCTC system focusing on the mAb
the resin slurry and buffer/cell culture fluid (CCF) with very yield, the system productivity, and buffer utilization. Model
uniform residence time distribution. The outlet from the calculations were used to examined the effects of staging,
static mixer is connected to a hollow fiber membrane module particle size, and resin capacity on the performance of the
(shown schematically in Figure 1B). The pore size of the CCTC system.
membrane (0.5 mm) is much smaller than that of the resin
(10–50 mm), but much larger than the mAb and any molecu- Model
lar impurities (5–20 nm). The resin slurry flows through the
lumen (retentate) side while the product (or impurities) are The CCTC system consists of a series of static mixers and
removed in the permeate that is collected through the mem- hollow fiber membrane modules. A diagram showing all the
brane. In the binding step, fresh resin is mixed with CCF streams for a single static mixer and membrane are shown in
which allows for capture of the mAb. The resin slurry with Figure 2. The governing mass balances for each static mixer
bound mAb is then passed to the washing steps where the can be written as:
unbound impurities are removed as waste in the permeate
while the resin with bound mAb is passed to the next step qs;in 1 qw 5 qs;out (1)
15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
432 Biotechnol. Prog., 2016, Vol. 32, No. 2

Figure 1. (A) Schematic of the CCTC system. Each step consists of a combination of static mixers and hollow fiber membrane mod-
ules (see inset for the elution step). The pumps are not shown for simplicity. (B) Schematic of the hollow fiber module. (C)
Schematic of a two stage binding step. BP and BR represent the binding step permeate and retentate streams, respectively.19

qs;in uin 5 qs;out uout (2) Binding step


qs;in uin Cb;in 1 qs;in ð12uin ÞCf;in 1 qw Cw 5 The binding step provides contacting between the clean
(3) resin and the clarified cell culture fluid containing the mAb.
qs;out uout Cb;out 1 qs;out ð12uout ÞCf;out
Thus, in a system with only a single binding stage, Cb,in 5
where qs,in and qs,out are the slurry volumetric flow rates Cf,in 5 0 and Cw 5 CF is the mAb concentration in the CCF
in the inlet and outlet streams; qw is the volumetric fluid feed. Cb,out and Cf,out are determined by the binding kinetics
flow rate into the mixer (CCF, wash buffer, elution buffer, based on the residence time in the static mixer as described
etc.); uin and uout are the resin volume fractions in the in Supporting Information. Additional details on the kinetics
inlet and outlet streams; Cb,in and Cb,out are the concentra- model are available in our previous publication.19 The resi-
tions of bound antibody in the inlet and outlet streams; dence time in the membrane was much smaller than that in
and Cf,in, Cf,out, and Cw are the concentrations of free anti- the static mixer and was neglected in the binding calcula-
body in the inlet, outlet, and fluid streams. The corre- tions; thus, Cb,r 5 Cb,out, and Cf,r 5 Cf,out 5 Cp. The mAb
sponding mass balances for the hollow fiber membrane loading (L) is given as:
modules are:
L5 Cb;out =Qmax 5 cb CF =ðQmax uin Þ (8)
qs;out 5 qr 1 qp (4)
cb 5qw =qs;in (8A)
qs;out uout 5 qr ur (5)
where cb is the ratio of the CCF to resin flow rates which is
qs;out uout Cb;out 1 qs;out ð12uout ÞCf;out one of the key design variables.
(6)
5 qr ur Cb;r 1 qr ð12ur ÞCf;r 1 qp Cp The productivity of the binding step can be increased by
using a two stage (countercurrent) binding step, which pro-
where the subscripts r and p refer to the retentate and perme-
vided equivalent mAb capture with less total residence time
ate streams. Since the membrane is assumed to be fully per-
(i.e., static mixer volume); this is discussed in more detail in
meable to the antibody, host cell proteins, and other
the “Results” section. Calculations were performed using the
impurities:
same basic approach as for single stage binding, but in this
case, the CCF was fed to the second stage and mixed with
Cp 5 Cf;r (7) resin that was preloaded with a small amount of mAb from
the first stage. The permeate from the second stage is then
15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Biotechnol. Prog., 2016, Vol. 32, No. 2 433

Figure 2. Schematic of (A) static mixer and (B) membrane module showing all inlet and outlet streams.

recycled to the first stage, where most of the remaining 1


K5 (12)
unbound mAb is bound by the fresh slurry. Permeate from 12uave ð12xÞ
the first stage then leaves as waste. In addition, the two stage uin ðc12Þ
system included an after binder, a static mixer placed after uave 5 (13)
2c12
the final hollow fiber membrane module, to capture the small
amount of residual mAb remaining in free solution in the where R1 ðor R2 Þ is the fractional impurity removal for
final retentate stream as shown in Figure 1C. wash-1 (or wash-2), N is the number of stages in the given
The binding step capture yield (Ybind) for a single stage wash step, c is the ratio of the wash buffer flow rate to the
system was evaluated from the following equation: concentrated slurry flow rate, S is the average sieving coeffi-
  cient for the species of interest (S 5 1 for CHO cell proteins
Cf ;r for the 0.45 mm pore size membrane used in the experi-
Ybind 5 12 (9)
C0 ments), K is a correction factor that accounts for the pres-
ence of solids in the slurry, uave is the average slurry
since the slurry flow rate out of the hollow fiber module was
volume fraction in the hollow fiber module (evaluated as the
controlled so that it is equal to the slurry flow rate entering
mean of the inlet and outlet values), and x is the liquid frac-
the first static mixer, i.e., qr 5 qs,in. C0 is the mAb concentra-
tion contained in the packed (settled) resin bed. For example,
tion in the feed immediately after mixing between CCF and for uin 5 0.25, c 5 3, and uout 5 uin/(c 1 1) 5 0.0625, one
resin slurry (before any binding occurs): gets uave 50:156: So for S 5 1 and x 5 0.6, Eq. (12) gives
C q K 5 1.067, with a 5 3.20 (from Eq. (11)), N 5 4, giving
C0 5   F w  (9A) R1 5 99.3% (from Eq. (10)). Note that a system operated
qw 1 qs;in ð12uin Þ
with cocurrent staging would only provide 90.5% impurity
removal using otherwise identical conditions. The total
impurity removal from the combination of the two wash
steps will simply be [12{(12R1)(12R2)}].
Wash step The buffer usage (V_ in L/g of mAb) in the wash steps for
The wash step is designed to remove unbound impurities a CCTC system, defined as the volume of buffer required
(e.g., host cell proteins and DNA) in the permeate stream. In per g of mAb product, is given as:
this case, countercurrent staging was used to increase impu- fc 1c g
rity removal and/or reduce the required buffer volume (or V_ 5 w-1 w-2 (14)
cb CF Y
flow rate). Possible binding of impurities was neglected, so
all of the terms involving Cb were assumed to be zero; bind- where Y is the overall mAb yield for the CCTC system, CF
ing of impurities to the base resin or the bound mAb would is the mAb concentration (titer) in the clarified cell culture
reduce the overall level of impurity removal, which is also fluid, and cw-1 and cw-2 are the ratios of buffer flow to slurry
observed in column chromatography. The resin concentration flow rates in the wash-1 and wash-2 steps, respectively.
in the retentate leaving the hollow fiber membrane module
was controlled to be equal to that in the inlet to the static
Elution step
mixer, i.e., ur 5 uin. For a multistage countercurrent system,
the final impurity removal is given as:17 The low pH buffer used in the elution step is assumed to
cause complete release of the bound mAb, i.e., Cb,out 5 Cb,r
aðaN 21Þ 5 0. The resin concentration in the retentate leaving the hol-
R1 ðor R2 Þ5 (10)
aN11 21 low fiber membrane module was again set equal to that in
a5cSK (11) the inlet to the static mixer, i.e., ur 5 uin. The use of coun-
tercurrent staging in the elution step provides greater yield
15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
434 Biotechnol. Prog., 2016, Vol. 32, No. 2

Table 1. Key Data Obtained from Binding Experiments


Antibody Resin Size ðlmÞ Qmax (g/L) k1 (min21) k2 (min21) kab (min21) ks (min21)
mAbl (CCF) 45 26.4 0.64 0.43 0.34 0.49
mAb2 (CCF) 45 21.2 1.49 0.92 0.96 1.22
mAb3 (Pure) 45 37.8 - - - 0.40
mAb3 (Pure) 10 27.2 - - - 1.49
The procedures to estimate rate constants are available in our previous paper (Dutta et al.)19 and are also discussed in Supporting Information.
k1, rate constant for the first stage; k2, rate constant for the second stage; kab, rate constant for "after binder"; ks, rate constant for single stage.

P 5 qelute Celute =RVi ui (16)

where qelute and Celute are the volumetric flow rate and mAb
concentration in the eluate and RViui is the total volume of
resin contained in the CCTC system. Equation (16) can also
be used to calculate the productivity of any given step in the
process by replacing the numerator with the mass flow rate
of product in the stream leaving that step (e.g., the retentate
leaving the after binder in the binding step) with the denomi-
nator evaluated based on the sum of the resin volumes in the
different stages within that particular step.

Results and Discussion


Binding step
The effects of the residence time in the static mixer on the
mAb yield and productivity of the binding step are shown in
Figure 3A for a single stage binding step. The productivity
shown in this plot corresponds to the binding step mAb cap-
ture only, and not the overall productivity of the system.
Calculations were done using the binding parameters for
mAb1 (Table 1) with a resin volume fraction in the inlet
slurry of uin 5 0.25 and an equilibrium loading of 81%
(evaluated by assuming all the mAb is bound to the resin).
Figure 3. (A) Changes in binding step yield and productivity The mAb yield increases with increasing residence time as
with residence time, (B) optimization of yield and expected due to the reduction in free (unbound) mAb as the
productivity. The mAb titer in the CCF was 7.2 g/L binding approaches equilibrium. 99% yield is obtained at a
and the binding capacity of the resin was 26.4 g/L.
The cb was 0.74 corresponding to 81% loading. The residence time of 7.4 min under these conditions. The pro-
binding step was designed as a single stage system. ductivity on the other hand decreases with increasing resi-
The volume fraction of resin slurry was 0.25. dence time since this corresponds to an increase in the
volume of the static mixer (and thus the total resin volume
(recovery) of the mAb while minimizing the amount of elu- in the binding step).
tion buffer, which also leads to a greater mAb concentration The trade-off between the yield and productivity is shown
in the collected eluate. The yield for a multistage elution explicitly in Figure 3B for values of the capture yield >0.95,
step can be evaluated using Eqs. (10–13) with: which is the target range for high value mAb products. The
productivity decreases with increasing yield due to the high
Yelute 5 R (15) residence times (and thus large static mixer volumes) needed
to achieve higher mAb binding. However, the binding step
productivity remains more than 200 g/(L h) at mAb yields
System design up to at least 98%. It is a point to note that the overall sys-
Model calculations were performed for four systems tem productivity will be less than binding step productivity
involving three different monoclonal antibody products: (1) due to the hold-up volume of resin in the static mixers/hol-
mAb1 in CCF using Poros 45 mm resin, (2) mAb2 in CCF low fiber modules in the other steps. The overall system pro-
using Poros 45 mm resin, (3) pure mAb3 using Poros 45 mm ductivity is discussed in the latter part of this article.
resin, and (4) pure mAb3 using a prototype 10 mm resin. The effect of loading on yield and productivity for the
The key parameters for the different systems are summarized binding step at a fixed residence time of 5 min is shown in
in Table 1, with details about the evaluation of these param- Figure 4. The loading was varied by changing the inlet
eters given in Supporting Information. The analysis of the slurry flow rate, qs,in. The yield decreases with increasing
two stage binding step used different binding rate constants loading due to the greater fraction of unbound mAb. Higher
in each stage to account for the fresh resin in the first stage residence times would thus be required at high loadings to
and the partially saturated resin in the second stage. This is maintain high product recoveries. The productivity increases
discussed in more detail in Supporting Information. The pro- with increasing loading due to the reduction in resin volume
ductivity (P) of the system (in units of g mAb/L resin/h) contained within the static mixer and hollow fiber membrane
was calculated as: module; this is a direct result of the reduction in qs,in (a
15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Biotechnol. Prog., 2016, Vol. 32, No. 2 435

Figure 4. Variation in binding step yield and productivity with


resin loading for a single stage binding step. The
mAb titer in the CCF was 7.2 g/L and binding
capacity of the resin was 26.4 g/L. The residence time
was 5 min, and the volume fraction of resin was 0.25.

smaller volume is needed to achieve the desired residence


time at lower flow rates).
The productivity and yield of the binding step can be
improved by using a two stage binding configuration. A
direct comparison of the performance of the one and two
stage binding steps is shown in Figure 5 for the binding
parameters corresponding to mAb1 (7.2 g/L titer) and mAb2
(0.67 g/L titer) with the loading fixed at 81% for mAb1 and
70% for mAb2 (consistent with the experiments performed Figure 5. Comparison of the performances of one stage and
using these mAbs). In both cases, the two stage system two stage binding steps. (A) Results for mAb1
requires less total residence time for a given yield due to the (titer 5 7.2 g/L) with resin binding capacity of
26.4 g/L, cb 5 0.74 (corresponding to 81% loading),
countercurrent configuration in combination with the added and resin volume fraction of 0.25. (B) Results for
product recovery in the after binder. The percent increase in mAb2 (titer 5 0.67 g/L) with resin binding capacity
productivity is more pronounced for the low titer mAb2 of 21.2 g/L, cb 5 3.1 (corresponding to 70% loading),
(Figure 5B) due to greater benefits of countercurrent staging and resin volume fraction of 0.14. The dotted lines
are to guide the eye.
at lower driving force for binding. The addition of the after
binder in the two stage system is critical to achieving high
product yield in the binding step; a two stage system without pressures (<20 psi) and the absence of any compaction
the after binder had very similar performance to a single effects. Note that the small particle size resins examined in
stage system at high yield because of the need to minimize this study did not show higher binding capacities, which is
likely due to the challenges in obtaining the required high
loss in the permeate. For 7.2 g/L titer (mAb1), 81% loading,
pore volume and pore size without further optimization of
u 5 0.25, and 98% binding step capture yield, the total resi-
the particles.
dence time required for a single stage is 5.75 min. This
decreases slightly to 5.70 min for a two stage system without Figure 6 shows the effects of particle size and binding
the after binder, and it drops to 4.92 min for a two stage sys- capacity on the trade-off between the binding step productiv-
tem with the after binder. Note that removal of the after ity and yield for mAb3. Calculations are shown for a single
binder, keeping the static mixers unchanged, would cause stage binding step at a fixed loading of 70% assuming an
the mAb yield to drop from 98% to less than 83% due to inlet resin concentration of uin 5 0.25. The productivity
loss of unbound mAb in the subsequent washing step. decreases with increasing yield, with the performance curves
for the 10 mm resin lying well above those for the 45 mm
Further improvements in system productivity can be
resin. Note that a 10 mm particle with low Qmax 5 27.2 g/L
achieved by using smaller resin particles (10–20 mm in diam- has better productivity than a 45 mm particle with Qmax 5
eter) which will have much faster binding kinetics due to 37.8 g/L. The use of a small particle size resin with high
reduced time required for pore diffusion. The difference in binding capacity (Qmax 5 54.4 g/L) can provide a binding
binding kinetics for the 10 and 45 mm particle size resins is step capture productivity of more than 800 g/(L h) at 98%
shown in Supporting Information. In contrast to column mAb yield which is more than five times the productivity for
chromatography, in which the pressure drop increases dra- a standard 45 mm particle.
matically when using very small particles, the pressure drop
in the CCTC system is essentially independent of particle
size since the viscosity of the flowing slurry depends almost Wash step
entirely on the resin volume fraction. In addition, the resins The effects of the buffer flow rate and number of stages
used in packed columns are highly crosslinked to provide on impurity removal in the wash step are shown in Figure
the mechanical strength needed to withstand high operating 7. It is important to note that the overall removal of
pressures (>100 psi). Resins with lower crosslinking den- unbound impurities in the CCTC system will be greater
sities, which could potentially have much higher binding than that shown in Figure 7 due to additional impurity
capacities,20 can be used in CCTC due to low operating clearance in the permeate collected in the binding step.
15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
436 Biotechnol. Prog., 2016, Vol. 32, No. 2

Figure 6. Effect of particle size on binding step productivity


for 10 and 45 mm particles. Qmax for the 45 mm par-
ticle was 37.8 g/L while simulations for the small
particle were done using Qmax 5 27.2, 37.8, and
54.4 g/L. The load was 70% ðcb 5 2.77, 3.85, and
5.54 for Qmax 5 27.2, 37.8 g/L, and 54.4 g/L, respec-
tively). The mAb3 titer was 1.72 g/L with a resin
volume fraction of 0.25.

Also, the impurity removal was calculated assuming negli-


gible binding of impurities to the resin or mAb. Similarly
to column chromatography, any impurities that bind to the
product or the resin (and cannot be removed during the
washing stages) would probably coelute with the mAb in
the elution step, which would reduce the overall level of
impurity removal.
As expected, the impurity removal increases with Figure 7. (A) Effect of c on impurity removal and total wash
increasing wash fluid flow rate (or c), but this also leads to buffer usage for a fixed number of stages in wash
an increase in buffer usage. Alternatively, one can signifi- steps, (B) effect of number of stages on wash buffer
cantly reduce buffer usage by increasing the number of usage to achieve a fixed impurity removal.
Titer 5 7.2 g/L, cb 5 0.74, x 5 0.6, and u 5 0.25. The
stages at a fixed impurity removal. For example, a 3-log dotted lines are to guide the eye.
(1000-fold) reduction in unbound impurities can be
achieved using two 4 stage wash steps (wash-1 and wash-
2), corresponding to 0.72 L of wash buffer (total of wash-1
and wash-2) per g of product, which is a 150% reduction in System optimization
buffer requirements compared to 2 stage wash steps. The overall performance of the CCTC system was deter-
Although there are diminishing returns on decreasing buffer mined based on the combined performance of the binding,
requirements by increasing the number of stages (N), signif- wash, elution, and regeneration steps. Calculations were per-
icant savings can be achieved up to N 5 5 stages for most formed for systems with a single stage binding step having
cases (Figure 7B), although this would lead to greater flow Ybind 5 Yelute 5 0.98, 3-log impurity removal (in the combi-
path costs because of the additional static mixers and hol- nation of the two wash steps), and 98% washout in the strip
low fiber modules. and equilibration steps (calculated using Eq. (10)). The resin
concentration (uÞ in the retentate exit was fixed at 25% for
Elution step all hollow fiber modules and the product loading was main-
tained at 81%.
The effect of countercurrent staging on product yield for
The effects of the resin binding capacity (Qmax) on the
the elution step for different values of c is shown in Figure buffer requirements (in L of buffer per g of recovered mAb)
8. Calculations were performed with the resin initially loaded are shown in Figure 9. The buffer usage rapidly decreases
with mAb at a bound concentration of 21.3 g/L (81% of the with increasing Qmax and increasing number of stages
maximum binding capacity). As expected, the elution step (assumed to be the same in all steps after binding). For
yield increases with an increase in the number of stages, par- example, a resin with 25 g/L binding capacity requires
ticularly as N increases from 1 to 4. For low buffer flow 5.8 L/g buffer when using only two stages, but this decreases
rates, e.g., c 5 1.5, more than seven stages are required to to 2.1 L/g for four stages. Although further increases in the
achieve 98% yield. High yields can also be achieved using number of stages would lead to a further reduction in buffer
fewer stages by employing a higher buffer flow rate, usage, this would also increase the number of hollow fiber
although this leads to higher buffer usage and more dilute membrane modules and, in turn, the overall cost of the
product in the elution pool. Note that the mAb concentration CCTC system. An increase in Qmax has tremendous benefit
could be readily increased by using an in-line ultrafiltration as this will directly decrease buffer usage without the need
of the product stream collected from the elution step. The to increase the number of stages. For example, at Qmax 5
buffer usage in the elution step, at a fixed yield of 98%, 25 g/L, a four stage system requires 2.1 L of buffer/g of
decreases from 1.18 L/g with N 5 2 to 0.42 L/g with N 5 4 mAb. This buffer usage can be reduced to 1.2 L/g for a three
(Figure 8B). stage system with Qmax 5 60 g/L. This shows the
15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Biotechnol. Prog., 2016, Vol. 32, No. 2 437

importance of future smaller particle/higher capacity resins


designed specifically for CCTC technology.
Further reductions in buffer requirements can be achieved
by internal recycling of buffers. For example, the low salt
effluent solution used in wash-2 could be recycled to wash-
1, supplemented by appropriate in-line addition of concen-
trated salt to achieve the target ionic strength in the wash-1
step. Similarly, the buffer solution used in the equilibration
step could be recycled and used as feed to the stripping step
after addition of acid to lower the pH to the desired value.
This type of buffer recycling is relatively straightforward to
implement in the CCTC system since all buffers are used
continuously with steady state flows (in contrast to the peri-
odic/cyclic buffer usage in batch and multicolumn chroma-
tography). The effect of buffer recycling on the overall
buffer usage is shown in Figure 9C. For Qmax 5 25 g/L,
u 5 0.25, the buffer requirement was reduced by 37% (from
2.13 to 1.34 L/g) simply by recycling the wash-2 and equili-
bration buffers. In comparison, a column packed with resin
of Qmax 5 25 g/L, packing density of 0.64, 70% loading,
and 12 column volumes (cv) of buffer (3 cv wash-1, 2 cv
wash-2, 3 cv elution, 2 cv stripping, 2 cv equilibration would
require 1.1 L of buffer/g of mAb at the same yield. It is
important to note that increasing the resin slurry volume
fraction (u) in CCTC will proportionally decrease buffer
usage (and vice versa). However, higher resin concentrations
can reduce the critical flux which will require an increase in
Figure 8. Effect of number of stages on (A) elution step yield,
(B) elution buffer usage to achieve 98% elution step
membrane area and may make it more difficult to maintain
yield. Titer 5 7.2 g/L, cb 5 0.74, x 5 0.6, and steady state operation over longer process times.
u 5 0.25. The dotted lines are to guide the eye. The overall productivity of the CCTC system is examined
in Figure 10 for both the high titer (7.2 g/L mAb1) and low

Figure 9. (A) Effect of resin binding capacity and number of stages on total buffer usage assuming 98% yield (or washout) in the
binding, elution, stripping, and equilibration steps with a total of three log combined impurity removal in the wash steps
(using the same number of stages in all the steps except the binding step). (B) Close up of Figure 9A, showing low buffer
usage region. (C) Effect of resin binding capacity and buffer recycle (from wash-2 to wash-1 and from equilibration to strip-
ping) on total buffer usage. Titer 5 7.2 g/L, cb 5 0.74, x 5 0.6, and u 5 0.25. The dotted lines are to guide the eye.
15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
438 Biotechnol. Prog., 2016, Vol. 32, No. 2

multiple static mixers that were required to achieve the neces-


sary residence time. The net result is that the experimental
productivity of 67 g/(L h) was approximately 10% lower than
the predicted value of 73 g/(L h). The experimental yield was
also slightly lower (90% compared to 96% in the model) due
to additional losses in the wash-1 and strip steps.
Figure 10B shows results from a similar analysis for the
low titer mAb2. In this case, a two stage binding step with a
loading of 70% and a resin concentration of 14% was used.
The two stage binding was used to increase the productivity
of the system. The productivity for the low titer mAb2 is
significantly lower than that for the high titer mAb1 due to
the greater residence time required to achieve high levels of
binding. The productivity again decreases with increasing
product yield, although the dependence on the yield is con-
siderably weaker than that seen for the high titer mAb1 due
to the use of a two stage binding step for mAb2. In this
case, the overall productivity for 96% product yield is 32 g/
(L h), which is in good agreement with the experimental
value obtained under these conditions [29 g/(L h)].19

Conclusions
The results presented in this study provide the first
detailed analysis of the design and optimization of CCTC
systems for continuous product capture, focusing on the
Figure 10. (A) Optimization of productivity and yield for a effects of countercurrent staging and buffer flow rates on
high titer (7.2 g/L) CCF for cb 5 0.74 with c 5 3.0 system performance. Countercurrent staging is essential for
in all other steps. The volume fraction of resin was
0.25. Number of stages in wash-1, wash-2, and elu- achieving high degrees of impurity removal while minimiz-
tion steps was three, while that for stripping and ing buffer requirements in the CCTC system. Staging also
equilibration steps was two. The binding step con- provides high product yield in the elution step. The calcu-
tained only a single stage. (B) Optimization of pro- lated productivity of the CCTC system for a high titer mAb
ductivity and yield for a low titer (0.67 g/L) CCF
for cb 5 3.1 with c 5 3.0 in all other steps. The vol- purification from an industrial cell culture was 73 g of mAb/
ume fraction of resin was 0.14. Number of stages in L of resin/h with 96% product yield and 3.4-log reduction in
wash-1, wash-2, and elution steps was three, while impurity levels; this productivity was in good agreement
that for stripping and equilibration steps was two. with experimental data and is more than four times that of
The binding step was designed with a two stage
system. The dotted lines are to guide the eye. comparable Protein A columns.
The CCTC system does require higher amounts of buffer
than packed column chromatography; however, buffer usage
titer (0.67 g/L mAb2) products. In both cases, the wash-1, in the CCTC system can be significantly reduced through
wash-2, and elution steps employed three stages while the strip- enhancements in system design and resin characteristics. For
ping and equilibration steps had two stages, consistent with the example, a packed column requires 1.1 L of buffer/g of
experimental studies performed by Dutta et al.19 Figure 10A mAb while a CCTC system with two stages in each step
shows results for the high titer mAb, with the resin concentra- requires 5.8 L of buffer/g of mAb. However, the use of a
tion in the retentate exit fixed at 25% using a single stage bind- four stage system provides more than a 60% reduction in
ing step with a loading of 81%. The overall productivity buffer with respect to a two stage system (2.1 L/g), while
decreases with increasing mAb yield due to the additional vol- recycling the buffer in the wash and equilibration steps leads
ume of the static mixers needed to achieve the required resi- to an additional 37% reduction (1.3 L/g). In addition, the use
dence time to capture higher levels of mAb. The calculated of a smaller particle size resin allows the binding step to
value of the productivity for an overall yield of 96% is 73 g/(L operate with much smaller residence times, which could
h) which is 4–5 times greater than a typical packed protein A potentially provide an additional 150–200% increase in sys-
column. Scale up modeling shows that a CCTC system with tem productivity. Further increases in productivity, and
this productivity would be able to purify a 2,000 L batch of reductions in buffer usage, could be achieved using a protein
mAb1 using only 17 L of protein A resin in a single shift. This A resin with higher binding capacity. Based on the results
will result in tremendous cost savings, both for the expensive presented in Figures 6–9, it should be possible to design a
protein A media and the overall process (detailed economic CCTC system with an overall productivity greater than
modeling will be the subject of future publications). 190 g/(L h) that uses less than 1.1 L/g of buffer. This is an
The solid circle symbol in Figure 10A shows the experi- order of magnitude greater productivity than current Protein
mental results obtained by Dutta et al. for the above condi- A columns with similar buffer requirements. Future studies
tions.19 It is important to note that the experimental data used will examine the impact of key design variables on the eco-
a 28% resin concentration instead of 25%. The experimental nomics of mAb capture/purification, including comparisons
system also had additional dead volume in the binding step with more recently developed multicolumn chromatography
due to a large amount of extra tubing used to connect the systems. The CCTC platform will also be assessed for other
15206033, 2016, 2, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.2250 by CAPES, Wiley Online Library on [15/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Biotechnol. Prog., 2016, Vol. 32, No. 2 439

separations, such as anion exchange flow through, ion 8. Zydney AL. Perspectives on integrated continuous bioprocess-
exchange capture and mixed-mode separations. ing—opportunities and challenges. Curr Opin Chem Eng. 2015;
10:8–13.
Another aspect that requires thorough investigation is the 9. Warikoo V, Godawat R, Brower K, Jain S, Cummings D,
scale up of the CCTC system. A large commercial scale sys- Simons E, Johnson T, Walther J, Yu M, Wright B, McLarty J,
tem would need to employ large static mixers to accommo- Karey KP, Hwang C, Zhou W, Riske F, Konstantinov K. Inte-
date greater volumetric flow rates. The mixing characteristics grated continuous production of recombinant therapeutic pro-
of these large mixers will be studied in detail as part of our teins. Biotechnol Bioeng. 2012;109:3018–3029.
10. Godawat R, Brower K, Jain S, Konstantinov K, Riske F,
future development efforts. In addition, larger membrane sur- Warikoo V. Periodic counter-current chromatography—design
face area would be scaled based on load and flux require- and operational considerations for integrated and continuous
ments. On-line process analytical technology (PAT) would purification of proteins. Biotechnol J. 2012;7:1496–1508.
also need to be incorporated as part of any commercial con- 11. Pollock J, Bolton G, Coffman J, Ho SV, Bracewell DG, Farid
tinuous process. Watson et al.21 recently showed how at line SS. Optimising the design and operation of semi-continuous
affinity chromatography for clinical and commercial manufac-
PAT could be incorporated in the scale up of a microfiltra- ture. J Chromatogr A. 2013;1284:17–27.
tion unit; a similar approach could potentially be used to 12. Muller-Sp€ath M, Aumann L, Str€ohlein G, Kornmann H, Valax
incorporate PAT in a CCTC system. P, Delegrange L, Charbaut E, Baer G, Lamproye A, J€ohnck M,
Schulte M, Morbidelli M. Two step capture and purification of
IgG2 using multicolumn countercurrent solvent gradient purifi-
Acknowledgments cation (MCSGP). Biotechnol Bioeng. 2010;107:974–983.
13. Langer ES, Rader RA. Introduction to continuous manufactur-
The authors would like to thank Andrew D. Tustian, ing: technology landscape and trends. Available at: http://www.
Philip A. Ropp, Regeneron Pharmaceuticals, and Fujifilm biopharma.com/continuous.pdf. Accessed November 18, 2015.
14. Rosa PAJ, Azevedo AM, Sommerfeld S, Mutter M, B€acker W,
Diosynth Biotechnologies for their assistance with the exp- Aires-Barros MR. Continuous purification of antibodies from
erimental studies. We would also like to thank the National cell culture supernatant with aqueous two-phase systems: from
Institute of General Medical Sciences (part of NIH, Grant concept to process. Biotechnol J. 2013;8:352–362.
No.9R44GM108259-02) and Ben Franklin Technology Part- 15. Hammerschmidt N, Tscheliessnig A, Sommer R, Helk B,
ners of Central PA for financial support of this project. Jungbauer A. Economics of recombinant antibody production
processes at various scales: industry-standard compared to con-
tinuous precipitation. Biotechnol J. 2014;9:766–775.
LITERATURE CITED 16. Hou Y, Brower M, Pollard D, Kanani D, Jacquemart R,
Kachuik B, Stout J. Advective hydrogel membrane chromatogra-
1. Aggarwal S. What’s fueling the biotech engine? Nat Biotechnol. phy for monoclonal antibody purification in bioprocessing. Bio-
2007;25:1097–1104. technol Prog. 2015;31:974–982.
2. Otto R, Santagostino A, Schrader U. Rapid growth in bio- 17. Shinkazh O, Kanani D, Barth M, Long M, Hussain D, Zydney
pharma: challenges and opportunities. Available at: http://www. AL. Countercurrent tangential chromatography for large-scale
mckinsey.com/insights/health_systems_and_services/rapid_grow- protein purification. Biotechnol Bioeng. 2011;108:582–591.
th_in_biopharma. Accessed November 18, 2015. 18. Napadensky B, Shinkazh O, Teella A, Zydney AL. Continuous
3. Protein A capture and its alternatives. Genet Eng News. 2014; countercurrent tangential chromatography for monoclonal anti-
34(18). Available at: http://www.genengnews.com/gen-articles/ body purification. Sep Sci Technol. 2013;48:1289–1297.
protein-a-capture-and-its-alternatives/5334/. Accessed November 19. Dutta AK, Tran T, Napadensky B, Teella A, Brookhart G, Ropp
18, 2015. PA, Zhang AW, Tustian AD, Zydney AL, Shinkazh O. Purifica-
4. Langer ES. Downstream processing: promising technologies are tion of monoclonal antibodies from clarified cell culture fluid
on the horizon. Biopharm Int. 2010;23(23). Available at: http:// using Protein A capture continuous countercurrent tangential
www.biopharminternational.com/downstream-processing-promisi chromatography. J Biotechnol. 2015;213:54–64.
ng-technologies-are-horizon. Accessed November 18, 2015. 20. Whitney D, McCoy M, Gordon N, Afeyan N. Characterization
5. Gronemeyer P, Ditz R, Strube J. Trends in upstream and down- of large-pore polymeric supports for use in perfusion biochro-
stream process development for antibody manufacturing. Bioen- matography. J Chromatogr A. 1998;807:165–184.
gineering. 2014;1:188–212. 21. Watson DS, Kerchner KR, Gant SS, Pedersen JW, Hamburger JB,
6. Kelley B. Very large scale monoclonal antibody purification: Ortigosa AD, Potgieter TI. At-line process analytical technology
the case for conventional unit operations. Biotechnol Prog. (PAT) for more efficient scale up of biopharmaceutical microfiltra-
2007;23:995–1008. tion unit operations. Biotechnol Prog. 2016;32:108–115.
7. Zydney AL. Continuous downstream processing for high value
biological products: a review. Biotechnol Bioeng. 2016;113: Manuscript received Dec. 8, 2015, and revision received Feb. 11,
465–475. 2016.

You might also like