You are on page 1of 8

Journal of Biotechnology 242 (2017) 11–18

Contents lists available at ScienceDirect

Journal of Biotechnology
journal homepage: www.elsevier.com/locate/jbiotec

Transfer of a three step mAb chromatography process from batch to


continuous: Optimizing productivity to minimize consumable
requirements
Xhorxhi Gjoka, Rene Gantier, Mark Schofield ∗
Pall Life Sciences, 20 Walkup Dr., Westborough, MA, USA

a r t i c l e i n f o a b s t r a c t

Article history: The goal of this study was to adapt a batch mAb purification chromatography platform for continu-
Received 29 August 2016 ous operation. The experiments and rationale used to convert from batch to continuous operation are
Received in revised form 2 December 2016 described. Experimental data was used to design chromatography methods for continuous operation that
Accepted 2 December 2016
would exceed the threshold for critical quality attributes and minimize the consumables required as com-
Available online 6 December 2016
pared to batch mode of operation. Four unit operations comprising of Protein A capture, viral inactivation,
flow-through anion exchange (AEX), and mixed-mode cation exchange chromatography (MMCEX) were
Keywords:
integrated across two Cadence BioSMB PD multi-column chromatography systems in order to process
Continuous chromatography
Monoclonal antibody (mAb)
a 25 L volume of harvested cell culture fluid (HCCF) in less than 12 h. Transfer from batch to continu-
Process intensification ous resulted in an increase in productivity of the Protein A step from 13 to 50 g/L/h and of the MMCEX
Productivity step from 10 to 60 g/L/h with no impact on the purification process performance in term of contaminant
Cadence BioSMB PD removal (4.5 log reduction of host cell proteins, 50% reduction in soluble product aggregates) and overall
chromatography process yield of recovery (75%). The increase in productivity, combined with continuous
operation, reduced the resin volume required for Protein A and MMCEX chromatography by more than
95% compared to batch. The volume of AEX membrane required for flow through operation was reduced
by 74%. Moreover, the continuous process required 44% less buffer than an equivalent batch process. This
significant reduction in consumables enables cost-effective, disposable, single-use manufacturing.
© 2016 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).

1. Introduction geting the same disease have renewed focus upon manufacturing
cost (Konstantinov and Cooney, 2015). Many current mAb or Fc
Over 50 mAbs have been approved by the FDA and another 50 fusion protein therapies on the market are facing patent expiration
are currently in phase three clinical trials (Reichert, 2016). The over the next few years (Strohl et al., 2012), enabling manufactur-
tremendous success of this class of drugs does come at a price, with ers of biosimilars greater access to market share. Along with cost,
mAb sales recently estimated to be close to $75 billion (Ecker et al., flexibility in manufacturing is also critical as the dynamic landscape
2015). However, manufacturing costs are estimated to only be a makes it difficult to predict the quantities of drug required through-
small fraction of the sales price (Kelley, 2009). This has tradition- out product lifetime and it takes many years to build a new mAb
ally led pharmaceutical industries to focus on time to market and manufacturing facility (Kamarck, 2006).
adopt a conservative approach to manufacturing, (Castilho, 2014; To address these combined challenges of cost and flexibility,
Gottschalk, 2013; Warikoo et al., 2012). With continuous process- many biopharmaceutical companies are pursuing process inten-
ing only being employed to reduce the overall purification time sification via continuous manufacturing (Horowitz, 2010). This
for protein therapies whose stability throughout the purification endeavor has been actively encouraged by the FDA, which iden-
process is limiting (Vogel et al., 2012). tifies continuous manufacturing as an opportunity to increase the
However, the biopharmaceutical landscape is changing rapidly. flexibility, agility and robustness of production (Lee et al., 2015).
Growing competition from biosimilars and multiple therapies tar- Process intensification through continuous manufacturing
enables the elimination of hold steps which are not value added and
further simplifies the process (Warikoo et al., 2012). It also reduces
process foot print using smaller and single use equipment which is
∗ Corresponding author.
E-mail address: Mark Schofield@pall.com (M. Schofield).

http://dx.doi.org/10.1016/j.jbiotec.2016.12.005
0168-1656/© 2016 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
12 X. Gjoka et al. / Journal of Biotechnology 242 (2017) 11–18

fundamental to flexible manufacturing and the accommodation of Life Sciences, Port Washington, NY). Tests were repeated using four
dynamic market demand (Fromison, 2009; Hodge, 2004). different load conditions, pH 8 & 6 mS/cm, pH 8 & 10 mS/cm, pH 8.4
A number of companies are now highlighting their continuous & 6 mS/cm, and pH 8.4 & 10 mS/cm at residence times of 1.5 and
purification strategies (Brower et al., 2014; Godawat et al., 2012; 3 min. The breakthrough curves were used to measure the 10% DBC
Hernandez, 2015; Kaltenbrunner et al., 2016; Mahajan et al, 2012; at each residence time. Operating binding capacity was calculated
Pollock et al., 2013; Palmer, 2015; Vogel et al., 2012), but what is using the method described in Gjoka et al. (2015).
largely absent from the literature is how to transfer a batch pro-
cess to continuous operation (Girard et al., 2015). Here we take 2.4. Screening conditions for operation of Mustang Q membrane
a batch mAb purification and describe the experiments and ratio- and CMM HyperCel sorbent
nale required to transfer it to a continuous chromatography process
that can be operated with two Cadence BioSMB PD systems. We The impact of pH and conductivity on binding and separation of
show it is possible to achieve higher loading capacities with shorter the target mAb versus host cell protein and aggregate contaminants
residence times by loading two or more columns in series. This was determined using a DoE based approach with response sur-
increase in capacity results from overloading the first column and face analysis via Minitab. The AEX membrane, Mustang Q (Pall Life
capturing unbound product in the flow through effluent on sub- Sciences, Port Washington, NY) and MMCEX sorbent, CMM Hyper-
sequent columns. As buffer consumption is inversely proportional Cel (Pall Life Sciences, Port Washington, NY), were evaluated for
to capacity, the increased capacity offered by multi column chro- binding over a broad range of conductivity and pH. CMM Hyper-
matography results in significantly reduced buffer consumption. Cel sorbent was transferred into an AcroprepTM Advance 96 Filter
Along with increased capacity, residence times are reduced and so Plate (PN 8129, Pall Life Sciences, Port Washington, NY). A post
is the cycle time. Increased capacity and decreased cycle both lead Protein A mAb sample at 1 mg/mL in the appropriate buffer was
to higher process step productivity expressed in gram of product added to each well (Pezzini et al., 2011; Toueille et al., 2011). Fil-
produced per liter of chromatography media and per hour (g/L/h). ter plates were incubated at room temperature while shaking for
This can considerably reduce the resin volume required for a purifi- 2 h. Flow through was collected and analyzed for recovery of mAb
cation process. Furthermore, by leveraging the increased capacity and level of contaminants. For Mustang Q membrane screening of
that is offered by continuous chromatography, the loading condi- flow through operating conditions, AcroPrepTM Advance 96-Well
tions going from batch to continuous can be modified, increasing Filter Plates with Mustang Q membrane (PN 8171, Pall Life Sciences,
the robustness of purification. Port Washington, NY) were washed with binding buffer and loaded
directly with 1 mg/mL post Protein A mAb samples. Flow through
2. Materials and methods fraction was analyzed for HCP concentration. The best perform-
ing conditions from high throughput screening were verified in
2.1. Analytical methods dynamic mode on 1 mL PRC pre-packed columns (Pall Life Sciences,
Port Washington, NY).
Host cell protein (HCP) was quantified using 3G CHO ELISA
(Cygnus Technologies, Southport, NC) assay. Soluble aggregates 2.5. Cadence BioSMB PD chromatographic system
were measured by HPLC-SEC using a TSK-GEL, Super SW3000,
4.6 mm × 30 cm, 4 ␮m column (Tosoh Haas, King of Prussia, PA). Two Cadence BioSMB PD systems were employed to oper-
Concentration of mAb in HCCF was measured using Protein A ate the multi-column chromatography experiments performed in
biosensors (ForteBio, Menlo Park, CA) and an Octet Red 96 BLI sys- this study. Each system is capable of operating up to 16 columns
tem. Concentration of mAb in post Protein A samples was measured simultaneously through software that enables the operation of sim-
using a NanoDrop 8000 spectrophotometer (Thermo Scientific, ulated moving bed chromatography. The core technology of the
Waltham, MA). machine is the disposable valve cassette which contains 240 valves
and features eight inlets, and six outlets. Valves are activated by
2.2. Impact of operating parameters on purity of protein A elution air pressure applied by solenoid actuators onto a fluoroelastomer
sheet. When air pressure is applied the valves are closed, when
A five factor, two level DoE was designed using Minitab to deter- the pressure is released pressure in the liquid flow-path forces the
mine the impact of overloading (loading the columns to saturation) valves open. The system is equipped with seven pumps capable of
on% recovery of mAb, % mAb aggregate, elution volume and host cell operating from 1 to 200 mL/min. The standard valve cassette flow
protein (HCP) concentration. The factors tested included residence path is 1 mm in diameter and can accommodate flow rates up to
time (0.6 min vs. 4.5 min), mass loaded (10% DBC- dynamic binding 70 mL/min. A larger 3 mm valve cassette is available for flow rates
capacity- vs. 90% DBC), load concentration (2 mg/mL vs. 5 mg/mL), above 70 mL/min.
and wash volume (10 column volumes (CV) versus 30 CV). The system has four UV sensors with wavelengths in
200–850 nm range, four conductivity sensors with a range of
2.3. Capacity determination for multi-column bind/elute 1 ␮S/cm to 200 mS/cm, and two pH probes with a range from 2
processes to 12.

HCCF containing mAb at titer 2, 6, and 10 mg/mL was loaded 3. Results and discussion
at residence times of 0.5, 1,1.5, 2, 3, and 4.5 min onto KANEKA
KanCapA 1 mL Protein A PRC columns (Pall Life Sciences, Port Wash- Streamlining of the purification process began by assessing the
ington, NY) using an AKTA Avant 25 (GE Healthcare, Uppsala, SWE). order of unit operations. The batch process had previously been
The breakthrough curves generated from these experiments were operated with the sequence of operations, Protein A, MMCEX in
used to calculate the 10% DBC. Operating binding capacity (OBC) for bind and elute mode to AEX in flow through mode, Fig. 1. The
continuous mode was calculated by the method described in Gjoka MMCEX sorbent is eluted via an increase in conductivity, however,
et al. (2015). the AEX membrane demonstrates improved HCP reduction at low
Protein A purified mAb (8 mg/mL concentration) titrated to pH conductivity. To accommodate this, the elution from the MMCEX
8.2 and 8 mS/cm was loaded at residence times of 0.75, 1.5, 2.25, 3, sorbent has to be diluted three-fold to lower the conductivity from
4.5, and 6 min onto CMM HyperCel MMCEX 1 mL PRC columns (Pall 22 mS/cm to 7 mS/cm. The dilution requires additional buffer and
X. Gjoka et al. / Journal of Biotechnology 242 (2017) 11–18 13

Mixed Mode Anion Exchange


Viral
Protein A Ca on
va on Membrane
Exchange

Load to 30 mg/mL
Load to 30 mg/mL at Hold for 30 minutes Dilute and flow through at
Bind at pH 7.6 and 7 mS/cm
4 minutes residence between pH 3.5 – 3.7 pH 7.6 and 7 mS/cm
Elute at pH 7.8 and 22 mS/cm

Mixed Mode
Anion Exchange
Ca on
Membrane
Exchange

Load to 30 mg/mL
Flow through at pH 7.6
Bind at pH 7.6 and 7 mS/cm
and 7 mS/cm
Elute at pH 7.8 and 22 mS/cm

Fig. 1. Order of unit operations for a batch process.

Table 1 that were tested when compared to a single column operated in


Effect of order of unit operations on purity.
batch mode.
Order of unit operations HCP log reduction % Aggregates The design space for column loading capacity was then used to
KanCapA – CMM HyperCel – Mustang Q 4.1 1.1 select the optimal binding capacity for a continuous multi-column
KanCapA – Mustang Q – CMM HyperCel 3.9 1.5 chromatography process that can accommodate an upstream flow
rate of 12 mL/min at a mAb titer of 4 mg/mL which is imposed on the
capture step by prior unit operations. When the process was devel-
oped only 5 mL pre-packed columns with a 5 cm bed height were
a system for buffer addition and mixing. By modifying the order of
available. Applying 12 mL/min onto these columns would give a lin-
unit operations to Protein A followed by AEX and finally MMCEX we
ear flow rate of 720 cm/h which is in excess of the recommended
can eliminate the dilution step. When we compare the two different
maximum linear velocity for the Protein A sorbent (500 cm/h). To
sequences of operations we can see an overall similar performance,
be able to accommodate this load flow rate, we split the flow across
Table 1. The small advantage in purification performance by oper-
two pairs of columns connected in series, meaning that at any given
ating MMCEX sorbent before AEX membrane is outweighed by the
point in time there are always four columns receiving feed. With
advantages of eliminating the dilution step. This makes integration
this configuration the columns are loaded at 360 cm/h (0.83 min
of the two polishing steps easier and enables the elimination of a
RT per column or 1.66 min total RT as two columns are loaded in
system for buffer addition and mixing from the process. Operation
series). We can see from Fig. 2 that this gives a 35 mg/mL operat-
without a dilution step also reduces the time required to load AEX
ing binding capacity. This compares favorably to the batch process
membrane as only 1/3rd of the volume has to be applied making the
which was loaded using a 4 min residence time and a capacity of
batch process more efficient and less time consuming. This process
27 mg/mL (calculated by taking 60% of the DBC at 10% of protein
sequence, Protein A sorbent followed by intermediate purification
breaking through).
using an AEX membrane and polishing with MMCEX sorbent, is our
To make a complete chromatography cycle, four additional
starting point as we begin to consider transfer of the process from
columns are required to operate the non-load steps (washes, elu-
batch to continuous operation.
tion, CIP and re-equilibration). Thus, the Protein A step is operated
with eight 5 mL columns (configuration of columns shown in Fig. 7)
3.1. Optimization of protein A capture step with a similar overall performance to a predicted four column pro-
cess with 10 mL columns. This flexibility is a feature that is unique
In order to convert the process from batch to continuous oper- to the Cadence BioSMB PD valve block.
ation a DoE was performed on Protein A chromatography media
(KANEKA KanCapA). The effects of overloading, wash volume, load 3.2. Low pH viral inactivation
concentration and residence time were assessed with regards to
the purification performance (data not shown). This experiment In order to perform post Protein A low pH viral inactivation
shows that a short wash (10 CV divided by 3 wash steps) along semi-continuously, a sub-batch cycle to cycle strategy is employed.
with low residence time and overloading has minimal effect on the Eight Protein A elution fractions (the fractions from a complete
HCP concentration post Protein A. cycle) are pooled into a first surge tank over the course of one
To investigate the impact of the multi-column chromatography Cadence BioSMB PD Protein A cycle. At the end of the Protein A
mode of operation on Protein A binding capacity, multiple single cycle, this pool is transferred to a second stirred surge tank where
column breakthrough experiments were employed as described in acidification to pH 3.6 +/− 0.1, 30 min hold, and a neutralization
Gjoka et al. With six single column breakthrough experiments, the step with 0.5 M Tris is performed to bring it to pH 8.2 & 6 mS/cm.
operating binding capacity in multi-column mode of operation can The neutralized viral inactivated material is then transferred into
be approximated over the complete design space. This data pre- third surge tank before the end of the second Cadence BioSMB PD
sented as a contour plot, using the distance method of interpolation, Protein A cycle. These steps were repeated for every Protein A cycle
(Fig. 2) shows a 50% or greater increase in capacity is available in using external programmable pumps to transfer product between
continuous multi-column mode over the range of residence times surge tanks cyclically for the duration of the experiment. Dosing
14 X. Gjoka et al. / Journal of Biotechnology 242 (2017) 11–18

Fig. 2. Binding capacity of The Protein A sorbent KANEKA KanCapA in (a) Batch mode and (b) Continuous mode with two columns loaded in series.

Table 2 regenerating a 0.86 mL capsule which can be operated at flow rates


Options considered for flow through operation of AEX membrane adsorber.
up to 10 membrane volumes per minute. These options are feasi-
Strategy # 1 2 3 4 5 ble since the average flow rate directly post viral inactivation is less
Membrane Volume (mL) 10 5 0.86 0.86 0.86 than 5 mL/min. The number of devices required depends on two fac-
# Cartridges 1 2 2 2 3 tors: the load capacity (2.5 g/mL) and the amount of time required
Load capacity (g/mL) 2.5 2.5 1.4 2.8 1.4 to regenerate the device (1 h). Here it would be physically possible
Regeneration Scheme None None <1 h CIP 1 h CIP 1 h CIP to operate with two 0.86 mL capsules and have one device being
loaded while the other device is being washed, regenerated and
re-equilibrated. Strategies 3 and 4 in Table 2 are both options that
was performed using two external pumps that are always running
use 2 smaller capsules for purification. Using two capsules makes
at a fixed flow rate through the Cadence BioSMB PD valve block.
integration with the mixed-mode resin difficult because the cycle
The valves within the cassette are used to direct acid and base to
time for the membrane adsorbers has to be synchronized with the
the second surge tank or to recirculate the acid and base to their
cycle times for the mixed-mode resin. This means the AEX mem-
respective containers.
brane adsorber cycle time has to be a multiple of the MMCEX cycle
time. In strategy 3, one AEX membrane adsorber cycle is exactly
3.3. Evaluation of load conditions – anion exchange membrane
twice as long as the cycle for the MMCEX process. This strategy is
adsorber operated in flow through mode
feasible but requires the CIP step to be shortened to less than 1 h
which increases the possibility of performance loss over time. The
The key metrics used to assess the performance of the polish-
other option is to create a method with a cycle duration equal to
ing steps are HCP and aggregate reduction. Here an AEX membrane
three times that of the mixed-mode cycle which enables us to per-
adsorber (Mustang Q) is employed in flow through mode to lower
form CIP for more than an hour but requires each capsule to be
the HCP concentration. The ability of this membrane to remove HCP
loaded to 2.8 g/mL and increases the risk of failure. Therefore, the
over a broad design space was investigated using a combination of
fifth strategy outlined in Table 2, using 3 capsules each loaded to
96 well membrane plates and a 0.86 mL membrane capsule. First,
1.4 g/mL, was selected because it provided the best compromise in
the HCP removal performance was evaluated at different conduc-
terms of minimizing membrane volume, ensuring the capsules are
tivity and pH load conditions using a fixed load capacity of 0.5 g/mL
regenerated thoroughly, and avoiding operation at high pressure.
of membrane. Fig. 3 shows fold reduction in HCP after passing Pro-
This strategy (5) embraces the concept of continuous processing,
tein A purified mAb through an AEX membrane under a range of
reducing the size of consumables by performing more cycles. This
conditions from pH 7.2–8.4 and conductivity from 5 to 10 mS/cm.
also gives a clear path for how best to operate processes over a
At conductivities ranging from 5 mS/cm to 7 mS/cm and pH 8 to
longer time that would be required to integrate with, for example,
pH 8.4, it is possible to achieve a greater than 60 fold reduction in
a continuous upstream perfusion cell culture process.
HCP. To determine the load capacity of the AEX membrane, up to
5 g of mAb was loaded onto a 0.86 mL capsule at four conditions,
pH 8 & 5 mS/cm, pH 8 & 7 mS/cm, pH 8.4 & 5 mS/cm and pH 8.4 &
7 mS/cm. These experiments (data not shown) indicate that within 3.4. Evaluation of load and elution conditions – CEX mixed-mode
this design space, loading Protein A purified mAb beyond 2.5 g/mL resin operated in bind and elute mode
of membrane results in an exponential increase in pressure across
the device. This pressure increase occurs before breakthrough of A MMCEX sorbent (CMM HyperCel) is employed in the process
HCP. to reduce mAb aggregates and to further reduce HCP concentration.
With this load capacity limitation, multiple strategies listed in Originally, for the batch process a compromise was made between
Table 2 that could accommodate a continuous load onto an AEX capacity and HCP reduction, Fig. 4. Loading at higher pH results in
membrane were considered. An oversized device (a 10 mL capsule) greater HCP reduction at the binding step, but this is at the expense
could be employed that is large enough to polish the complete batch of capacity. The batch process was designed around column vol-
without being regenerated and recycled (Table 2, strategy 1). Con- ume. The strategy was to purify the complete batch (25 L HCCF at
sidering the total load volume to be processed is 25 L of HCCF with 1 g/L mAb titer) via a single cycle with a column of 1 L in volume
a mAb titer of 1 mg/mL, and the AEX membrane loading capacity or less. To accommodate this, loading conditions selected for the
defined in this study is 2.5 g/mL of membrane, this first strategy batch process are pH 7.6 and 7 mS/cm conductivity. The DBC at
could be used to accommodate the load. Alternatively, two AEX 10% breakthrough under these conditions is 45 mg/mL. This allows
5 mL capsule configured in parallel could be loaded and discarded for an operating binding capacity of 27 mg/mL (60% of the DBC at
after use (Table 2, strategy 2). Strategies 3–5 rely on cycling and 10% breakthrough).
X. Gjoka et al. / Journal of Biotechnology 242 (2017) 11–18 15

Fig. 3. AEX membrane, Mustang Q, HCP reduction performance.

In continuous mode a higher capacity is expected because of mAb aggregates, data not shown. From these experiments elu-
columns are loaded in series. This allows the columns to be tion conditions of pH 7.8 and 22 mS/cm conductivity are selected to
overloaded without loss of product as product is captured on a sub- deliver greater than 80% product recovery, >10 fold HCP reduction
sequent column. Therefore, the loading conditions can be modified and a final aggregate clearance of 50%.
when transferring from a batch to continuous process. To find the With this information, along with the process constraints from
best compromise between HCP reduction and operating binding the previous process, the MMCEX process can be defined within
capacity a column based DoE was performed using extended pH the context of continuous operation. To generate a balanced cycle,
conditions, Fig. 4. This DoE enables the identification a new opti- six × 5 mL columns are required in total, where two columns are
mum design space. Using loading conditions of pH 8.1 and 6 mS/cm loaded in series with a residence time of 1.5 min and four columns
conductivity a 45 mg/mL capacity at 1% breakthrough is calculated. are required to operate the non-load steps.
A 40 mg/mL operating loading capacity was used for the actual
MMCEX process to take a modest margin. These buffer conditions 3.5. Implementation of the continuous integrated
used in continuous multi-column mode decrease the HCP level in chromatography platform
elution to 500 ppm compared to 625 ppm obtained in batch mode.
In addition, making the same modification to the loading condi- Two Cadence BioSMB PD systems were connected to operate the
tions onto the membrane adsorber (upstream of MMCEX) results in entire chromatography train. The first Cadence BioSMB PD system
increased HCP reduction from the membrane adsorber (60 fold HCP performed the capture step with eight × 5 mL Protein A columns
reduction, compared to 30 fold with the batch conditions, Fig. 3). along with the dosing for the low pH viral inactivation step. The
Column elution DoEs were performed to determine the optimal elu- second Cadence BioSMB PD operates the combined polishing steps.
tion condition from MMCEX for yield, HCP reduction and removal AEX membrane is operated with three devices with a 120 min cycle

Fig. 4. Extension of MMCEX (CMM HyperCel) sorbent design space made possible by continuous operation.
16 X. Gjoka et al. / Journal of Biotechnology 242 (2017) 11–18

Process #1 Process #2
A post KanCapA post VI/depth/sterile post CMM
B post KanCapA post VI/depth/sterile post CMM

Mab concentration (mg/mL)


14 14

Mab Concentration (mg/mL)


12 12

10 10

8 8

6 6

4 4

2 2

0 0
60 180 300 420 540 660 0 120 240 360 480
Time (min) Time (min)

Host Cell Protein Aggregates


C Expt. #1 #2
Process Expt. #2 #1
Process
D Process
Expt. #1 #2 Process
Expt. #2 #1
100 5
90
80 4
70

% Aggregates
HCP (ppm)

60 3
50
40 2
30
20 1
10
0 0
60 160 260 360 460 560 60 160 260 360 460 560
Time (min) Time (min)

Fig. 5. (a) mAb concentration versus time during continuous operation for process #1. (b) mAb concentration versus time during continuous operation for process #2. (c)
Residual HCP levels after polishing steps (d) Residual aggregate levels after polishing steps.

time. The MMCEX sorbent cycle is 40 min and requires six columns.
These two polishing operations are operated on a single Cadence
BioSMB PD system with a single pump used to flow-through AEX
membrane and load onto two MMCEX columns in series. To accom-
modate this on a single Cadence BioSMB PD, three MMCEX sorbent
cycles are performed per AEX membrane cycle. This again shows
the flexibility and intensification offered by this approach. Unit
operations are combined even though they have different number
of columns and cycle times.
Two continuous integrated processes (process 1 and process 2
in Fig. 5) have been performed to test the performance of contin-
uous processing with this configuration. For both processes, prior
to capture step, 20 Liters of clarified CHO supernatant were con-
centrated using an in line concentrator down to 5 Liters and a
final mAb concentration of 4 mg/mL. Each process was operated
for a duration of around 10 h. Each Protein A and MMCEX column
was cycled 15 times, while AEX membrane capsules were cycled
5 times over the course of each experiment. The mAb production Fig. 6. Impact of continuous processing on consumables.
rate, or throughput, was approximately 2 g/h. Sample aliquots were
taken every hour for analysis of critical quality attributes. Aggre-
gate and HCP concentration after the final polishing step are shown of the surge tanks between steps was lowered in process 2 resulting
in Fig. 5. Overall, the continuous purification train resulted in a 4.5 in further reduction of the overall process time.
log reduction in HCP (from 500,000 ppm to 10 ppm), a 50% reduc- Fig. 6 summarizes the benefits of processing a 25 L HCCF sam-
tion in aggregates (from 2 to 3% aggregates in the load down to ple volume in continuous processing mode compared to batch
1–1.5% after an isocratic elution from the mixed-mode resin) and mode. Both of these processes assume the same sequence of unit
75% total yield. There was no significant time dependent perfor- operations being performed under optimized conditions for each
mance observed in the quality of the final polished mAb. The final respective mode of operation. For the batch mode of operation,
HCP concentration was approximately five-fold lower compared it is assumed that the columns are ideally sized to perform the
to the batch process confirming that modifying the design space purification in a single cycle. Under these assumptions, the over-
when transferring from batch to continuous processing can be used all process time from start to finish is similar for both continuous
to enhance purification performance. The aggregate clearance was and batch modes of operation. However, the amount of sorbent
50% for both batch and continuous operation. Notably, the volume required to operate the continuous process is considerably reduced.
For the Protein A sorbent, continuous operation results in a 94%
X. Gjoka et al. / Journal of Biotechnology 242 (2017) 11–18 17

Fig. 7. Chronograms showing the 3 chromatography processes, (a) Protein A, (b) AEX and (c) MMCEX. Column positions are shown by the radial “spokes” (Column 1, C1,
through Column 8, C8). The outer annular ring shows the material the column is receiving and the inner annular ring shows where the flow out of the column is directed.
The columns proceed around the cycle in clockwise direction.

reduction of resin volume. For the polishing steps, total AEX mem- analysis in combination with several in-line process analytical tech-
brane volume would be reduced by 74%, while the MMCEX resin nologies. This means that in the near future, real-time release of
volume would be reduced by 96%. This is a significant reduction drug product will be possible. This will reduce the need for large
in the amount of resin required and could result in consider- inventories of a given drug substance and ultimately lower the cost
able cost savings. Buffer consumption would also be reduced by of that drug to the patient.
44% by transferring from batch to integrated continuous process-
ing. The buffer consumption reduction comes from operating with
increased capacity, combining unit operations and intensification References
of the process through both changing the order of unit operations
Brower, M., Hou, Y., Pollard, D., 2014. Monoclonal Antibody Continuous Processing
and reducing the number of column volumes required for some of Enabled by Single Use. Continuous Processing in Pharmaceutical
the process steps. The dramatic reduction in buffer consumption Manufacturing.
might be a reason to convert facilities that are limited by buffer Castilho, L.R., 2014. Continuous Animal Cell Perfusion Processes: the First Step
Toward Integrated Continuous Biomanufacturing. Continuous Processing in
production to switch to continuous processing. These results show Pharmaceutical Manufacturing.
a compelling case for continuous production. However, an over- Ecker, D.M., Jones, S.D., Levine, H.L., 2015. The Therapeutic Monoclonal Antibody
all cost benefit analysis between batch and continuous for various Market. In MAbs, Vol. 7. Taylor & Francis, pp. 9–14 (January, No. 1).
Fromison, J., 2009. Disposables in clinical manufacturing. Am. Pharm. Rev. 12,
scales of production will be addressed in a subsequent publication. 20–27.
Girard, V., Hilbold, N.J., Ng, C.K., Pegon, L., Chahim, W., Rousset, F., Monchois, V.,
2015. Large-scale monoclonal antibody purification by continuous
4. Conclusion chromatography, from process design to scale-up. J. Biotechnol. 213, 65–73.
Gjoka, X., Rogler, K., Martino, R.A., Gantier, R., Schofield, M., 2015. A
straightforward methodology for designing continuous monoclonal antibody
Four mAb purification unit operations, Protein A, low pH viral
capture multi-column chromatography processes. J. Chrom. A 1416, 38–46.
inactivation, AEX, and MMCEX were optimized for batch pro- Godawat, R., Brower, K., Jain, S., Konstantinov, K., Riske, F., Warikoo, V., 2012.
cessing and converted into continuous unit operations using two Periodic counter-current chromatography–design and operational
considerations for integrated and continuous purification of proteins.
Cadence BioSMB PD continuous purification systems. The study
Biotechnol. J. 109 (12), 1496–1508.
demonstrates that significant operational efficiency and quality Gottschalk, U., 2013. Biomanufacturing: time for change? Pharm. Bioprocess. 1 (1),
advantages can be realized by moving from single column batch 7–9.
processing to multi-column processing on a Cadence BioSMB PD Hernandez, R., 2015. Continuous manufacturing: a changing processing paradigm.
Biopharm. Int. 28 (April (4)), Retrieved from: http://www.
system. The study also demonstrates that further operational effi- biopharminternational.com/continuous-manufacturing-changing-processing-
ciency is made possible by connecting and operating multiple unit paradigm.
operations together and those continuous processes can be oper- Hodge, G., 2004. Disposable components enable a new approach to
biopharmaceutical manufacturing. Biopharm. Int. 17, 38–49.
ated at steady state in order to produce a homogeneous, high purity Horowitz, A., 2010. Is the reign of batch processing coming to an end? Bioprocess
drug substance from start to finish. Online, Retrieved from: http://www.bioprocessonline.com/doc/is-the-reign-
In this study, processing continuously enabled a significant of-batch-processing-coming-to-0001.
Kaltenbrunner, O., Diaz, L., Hu, X., Shearer, M., 2016. Continuous bind-and-elute
reduction in consumable needs (volume of chromatography media, protein A capture chromatography: optimization under process scale column
buffer volume). With such a drastic reduction, it follows that con- constraints and comparison to batch operation. Biotechnol. Progr. 32 (4),
tinuous processing will have a proportionate impact on facility 938–948.
Kamarck, M.E., 2006. Building biomanufacturing capacity −the chapter.
footprint leading to a reduction in both operating and capital
Kelley, B., 2009. Industrialization of mAb Production Technology: the
expenditure. Reduction in capital expenditure could prove valuable Bioprocessing Industry at a Crossroads. In MAbs, Vol. 1. Taylor & Francis, pp.
when advancing drugs through clinical trials because only a fraction 443–452 (September, No. 5).
Konstantinov, K.B., Cooney, C.L., 2015. White paper on continuous bioprocessing.
of drugs make it through the FDA. Reducing the capital expendi-
may 20–21, 2014 continuous manufacturing symposium. J. Pharm. Sci. 104 (3),
ture for each potential treatment during clinical trials reduces the 813–820.
cost associated with the high risks of failure during clinical trials. Lee, S.L., O’Connor, T.F., Yang, X., Cruz, C.N., Chatterjee, S., Madurawe, R.D.,
Furthermore, this type of process intensification, where multiple Woodcock, J., 2015. Modernizing pharmaceutical manufacturing: from batch
to continuous production. J. Pharm. Innov. 10 (3), 191–199.
columns are cycled far more frequently than in batch processing, Mahajan, E., George, A., Wolk, B., 2012. Improving affinity chromatography resin
paves the way for dynamic process control using multi-variate data efficiency using semi-continuous chromatography. J. Chrom. A 1227, 154–162.
18 X. Gjoka et al. / Journal of Biotechnology 242 (2017) 11–18

Palmer, E., 2015. Vertex, J&J, GSK, Novartis all working on continuous Strohl, William R., Strohl, Lila M., 2012. Therapeutic Antibody Engineering: Current
manufacturing facilities. FiercePharma, Retrieved from: http://www. and Future Advances Driving the Strongest Growth Area in the Pharmaceutical
fiercepharma.com/supply-chain/vertex-j-j-gsk-novartis-all-working-on- Industry. Elsevier, pp. 443.
continuous-manufacturing-facilities. Toueille, M., Uzel, A., Depoisier, J.F., Gantier, R., 2011. Designing new monoclonal
Pezzini, J., Joucla, G., Gantier, R., Toueille, M., Lomenech, A.M., Le Sénéchal, C., antibody purification processes using mixed-mode chromatographysorbents. J.
Garbay, B., Santarelli, X., Cabanne, C., 2011. Antibody capture by mixed-mode Chromatogr. B 879, 836–843.
chromatography: a comprehensive study from determination of optimal Vogel, J.H., Nguyen, H., Giovannini, R., Ignowski, J., Garger, S., Salgotra, A., Tom, J.,
purification conditions to identification of contaminating host cell proteins. J. 2012. A new large-scale manufacturing platform for complex
Chromatogr. A 1218, 8197–8208. biopharmaceuticals. Biotechnol. Bioeng. 9, 3049–3058.
Pollock, J., Bolton, G., Coffman, J., Ho, S.V., Bracewell, D.G., Farid, S.S., 2013. Warikoo, V., Godawat, R., Brower, K., Jain, S., Cummings, D., Simons, E., Johnson, T.,
Optimising the design and operation of semi-continuous affinity Walther, J., Yu, M., Wright, B., McLarty, J., Karey, K.P., Hwang, C., Zhou, W.,
chromatography for clinical and commercial manufacture. J. Chrom. A 1284, Riske, F., Konstantinov, K., 2012. Integrated and fully continuous production of
17–27. recombinant therapeutic proteins. Biotechnol. Bioeng. 109, 3018–3029.
Reichert, J.M., 2016. Antibodies to Watch in 2016. In MAbs, Vol. 8. Taylor & Francis,
pp. 197–204 (February, No. 2).

You might also like