Biotech J 2021 - Complete or Periodic Continuity in Continuous Manufacturing Platforms For Production of Monoclonal Antibodies

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Received: 14 October 2020 Revised: 21 March 2021 Accepted: 22 March 2021

DOI: 10.1002/biot.202000524

RESEARCH ARTICLE

Complete or periodic continuity in continuous manufacturing


platforms for production of monoclonal antibodies?

Nikhil Kateja Anamika Tiwari Garima Thakur Anurag S. Rathore

Department of Chemical Engineering, Indian


Institute of Technology Delhi, Hauz Khas, India Abstract
Background: Monoclonal antibodies (mAbs) currently dominate the biotherapeutic
Correspondence
Anurag S. Rathore, Department of Chemical market. This has resulted in significant efforts towards the development of a contin-
Engineering, Indian Institute of Technology,
uous integrated platform for the manufacturing of mAbs.
Hauz Khas, New Delhi 110016, India.
Email: asrathore@biotechcmz.com, Website: Main Methods and Major Results: In this study, a continuous mAb platform has been
www.biotechcmz.com
developed consisting of an Acoustic Wave Separator, a Cadence BioSMB PD system,
Nikhil Kateja, Anamika Tiwari, and Garima a customized coiled flow reactor, a modular single-pass TFF kit, an in-line diafiltra-
Thakur contributed equally to this work. tion module, and a continuous dead-end filtration skid. A three-step chromatographic
purification was performed in the platform consisting of Protein A capture chromatog-
raphy followed by an anion exchange membrane directly coupled to a cation exchange
chromatography. Two operational case studies have been executed on the platform,
namely complete continuous (“CC”) and periodic continuous (“PC”) modes of opera-
tion. The CC mode was designed to ensure that each unit operation had completely
continuous inflow and outflow by increasing the number of columns, filtration mod-
ules and tanks, while the PC mode operated in periodic pulses with scheduled flow and
hold steps. Both modes were designed to handle the same flow rate and titers from the
upstream bioreactor or fed-batch harvest tank, and were compared in terms of pro-
ductivity and operational complexity. Both modes offer viable options for continuous
processing of mAbs and result in achievement of target critical quality attribute pro-
files of the final drug product over 24 h of operation.
Conclusions and Implications: It was found that the CC mode was superior in terms of
specific productivity (20–50% higher) and consumable utilization (20% lower resin uti-
lization), while the PC mode was operationally simpler and had lower facility costs due
to significant reductions in the number of auxiliary equipment (pumps, columns, tanks,
and valves). The work successfully highlighted the pros and cons of both approaches,
and demonstrates that while several groups have amply shown the superiority of con-
tinuous processing over batch mode, there are intermediate variants which may be
optimal in a given situation.

KEYWORDS
continuous processing, integrated purification, monoclonal antibody

Abbreviations: mAb, monoclonal antibody; CEX, cation exchange chromatography; SEC, size exclusion chromatography; CV, column volume; CIP, cleaning in place; A280 , absorbance at 280 nm;
HCP, host cell protein

Biotechnol. J. 2021;16:2000524. www.biotechnology-journal.com © 2021 Wiley-VCH GmbH 1 of 14


https://doi.org/10.1002/biot.202000524
2 of 14 KATEJA ET AL.

1 INTRODUCTION

Development of end-to-end platforms for continuous manufacturing Recently, a continuous non-Protein A mAb manufacturing train from
of monoclonal antibodies (mAbs) is currently of significant interest to clarified fed-batch harvest to unformulated drug substance has been
both academic and industrial groups in the biotherapeutic industry.[1] demonstrated by integrating a precipitation-based capture step with
Continuous manufacturing has been shown to offer several advantages two polishing chromatography steps and viral inactivation using low pH
over batch manufacturing including reduced capital cost, enhanced hold.[6]
productivity, improved product consistency without batch-to-batch Researchers have also developed continuous platforms with vari-
variability, and the ability to handle high upstream process titers.[2–8] ous novel aspects of monitoring, modelling, and control to improve per-
The last decade has seen rapid growth in specialized technologies formance and consistency. An integrated continuous lab-scale process
designed to facilitate continuous mAb manufacturing. Innovations in consisting of perfusion cell culture with filter-based cell retention, a
upstream processing include perfusion cell culture systems for contin- customized two-column capture setup, a viral inactivation loop with
uous production of mAbs at high titer[9] and continuous clarification continuous flow, and two semi-continuous polishing steps using multi-
systems[10] such as continuous centrifugation,[11,12] alternating tan- column counter-current solvent gradient purification (MCSGP) on a
gential flow filtration,[13,14] and acoustic wave separation.[15,16] Down- Contichrom CUBE Combined system has been proposed.[28] Internal
stream innovations include versatile chromatography setups with in- recycle loops were implemented with model-based control to improve
built pumps, valves and sensors enabling integrated multi-column the performance of the bioreactor, maximize the yield of the cap-
operation, such as the Cadence BioSMB (Pall Life Sciences), Octave ture step, and optimize the purity-yield tradeoff in the polishing steps.
SMB (Tarpon Systems), ÄKTA PCC (GE Healthcare), Contichrom CUBE Another group developed a continuous platform from perfusion cell
(ChromaCon), and BioSC (Novasep).[17] For continuous viral inactiva- culture to formulated drug product and implemented at-line analytical
tion, customized flow reactors and hold tanks have been developed chromatography methods to monitor critical quality attributes by sam-
by various groups for achieving continuous low pH conditions.[18–22] pling the process material at various points to facilitate control deci-
For continuous formulation, innovative single-pass ultrafiltration and sions during continuous operation.[29] Researchers have also proposed
diafiltration modules have been developed which use novel flow paths a continuous train consisting of a perfusion bioreactor, twin-column
to achieve high concentration factors and have multiple inlets to capture step, viral inactivation in a customized hold tank, and single-
achieve 99.9% buffer exchange in a single pass of the process material column polishing step, along with a supervisory control and data acqui-
through the module.[23–25] sition system to digitally integrate unit operations along with analyzers
Integrating these technologies has been shown to result in a power- including biocapacitance probes and two at-line HPLCs.[30]
ful and highly versatile platform for continuous manufacturing of mAbs It is evident from the above mentioned literature that several
with continuous cell culture, clarification, capture, viral inactivation, robust mAb platforms have been successfully demonstrated at both
sterile filtration, polishing, concentration, and formulation into the final lab and pilot scale. Each platform uses different combinations of the
mAb drug product. A platform consisting of perfusion cell culture, alter- underlying enabling technologies, namely continuous clarification,
nating tangential flow filtration for clarification, capture chromatogra- multi-column capture and polishing chromatography, viral inactivation
phy on ÄKTA PCC, viral inactivation in a low pH hold tank, and polish- in scheduled batch-mode or continuous flow mode, and modular ultra-
ing chromatography on a custom-modified four-column ÄKTA PCC was filtration, to achieve continuous operation. Major innovations have
showcased as one of the first continuous trains running from cell cul- been with respect to novel chromatography setups,[17] customized
ture to unformulated drug substance.[26] Several platforms for contin- flow reactors,[21,31] integrated multi-polishing steps,[32] model-based
uous production of mAbs at pilot scale have also been demonstrated. A optimization,[33] and advanced supervisory control systems[30,34]
plant consisting of continuous perfusion followed by clarification, con- to implement continuous processes. Each platform has successfully
centration, and sterile filtration as the first half of downstream train, shown benefits over batch processing in terms of process productivity,
and then capture, viral inactivation, polishing, sterile filtration, formu- facility size and economic costs. However, it is also evident from the
lation, and bioburden reduction as the second half of the downstream wide variety of approaches that there are multiple ways of achieving
train has been proposed.[27] The two parts of the downstream train continuity, even when the base unit operations are identical. Some
were separated by a cooled storage step and the entire platform was of the case studies referenced above integrated continuous loading
built using only single-use equipment. Two Cadence BioSMB units were in capture chromatography with periodic viral inactivation,[26,28]
used for capture and polishing chromatography, respectively, and recir- polishing,[28] and ultrafiltration[35] steps by implementing scheduled
culation loops were used to achieve continuous viral inactivation and hold steps, while others chose to have uninterrupted flow between
formulation. Another pilot-scale integrated continuous process from the unit operations and increased the number of columns and mem-
perfusion cell culture to formulated drug substance using a BioSC branes to achieve this.[7] For a user intending to create a continuous
Multicolumn Chromatography system for capture chromatography, a platform, many questions would come to mind. Should we aim for
size exclusion chromatography column for continuous viral inactiva- complete continuity in each unit operation, or is periodic continuity
tion, and single-pass TFF modules for sterile filtration, anion exchange acceptable or even preferred? For example, when integrating capture
polishing, ultrafiltration, and diafiltration has also been proposed.[7] chromatography with continuous clarification, it is generally accepted
KATEJA ET AL. 3 of 14

that the loading step should be continuous. However, should the actor and was continuously clarified with an Pall Cadence Acoustic
chromatographic method be designed so that elution is continuous, Wave Separator (AWS) followed by downstream processing steps of
or are periodic elutions also acceptable? Similar choices between Protein A chromatography, viral inactivation, sterile filtration, cation
continuous or scheduled periodic flows arise in other unit operations exchange (CEX) chromatography, single-pass ultrafiltration, and single-
as well, including viral inactivation, polishing chromatography, and pass diafiltration. The chromatography steps were carried out on a Pall
formulation. What are the impacts of either choice on process produc- Cadence BioSMB System. The continuous train was operated in two
tivity, facility fit, and operational ease? What considerations need to be modes, namely complete continuous (“CC”) and periodic continuous
taken into account to integrate completely continuous versus periodic (“PC”). Each mode was operated for 24 h and samples of the harvest
flows with neighboring unit operations? and final product were taken every 1 h and analyzed by offline ana-
In this work, we have leveraged our lab-scale mAb manufacturing lytical HPLC for quantification of mAb concentration, aggregate per-
train consisting of continuous clarification using acoustic wave sep- centage, and charge variant distribution. Figures 1A and 1B illustrate
aration, continuous capture, and polishing chromatography using a the process flow diagrams for the CC and PC modes respectively. The
Cadence BioSMB system, continuous viral inactivation using a coiled operating time of 24 h per mode was selected as it was sufficient to
flow inversion reactor,[36] and continuous formulation using single- demonstrate the key scheduling differences between the two modes
pass ultrafiltration and diafiltration modules, to explore these ques- for both the individual unit operations and the overall train includ-
tions in a targeted manner. The same platform has been run in two ing surge tanks. The operating time can be extended in each case as
modes, complete continuous (“CC”) and periodic continuous (“PC”). In per campaign requirements, as steady state is attained in both modes
CC mode, the inflows and outflows from each unit operation are com- in well under 24 h. The CC mode was designed to ensure that there
pletely continuous, with no scheduled hold time. This is achieved by was continuous inflow and outflow from all unit operations. Continu-
developing appropriate chromatography methods on the BioSMB with ous loading and elution were facilitated in all chromatography steps
sufficient columns for both continuous loading and continuous elu- by increasing the number of columns and sizing them appropriately,
tion, and by placing surge tanks to dampen concentration gradients and surge tanks were placed between the unit operations for damp-
between unit operations. In PC mode, the chromatography methods ening of concentration gradients. The PC mode was designed as an
are designed without the constraints of continuous loading and elu- operationally simpler alternative to the CC mode where the number
tion, and each unit operation is run with scheduled starts and stops in of columns and surge tanks was minimized, and chromatography elu-
sync with the rest of the continuous train. The harvest flowrate and tions were passed to the next unit operation in periodic pulses. Further
titer conditions are the same for each mode to allow for comparisons in explanations of the design of the CC and PC modes and comparison in
terms of process productivity, resin productivity, operational complex- terms of process scheduling, resin productivity, membrane productiv-
ity, and facility requirements including auxiliary equipment such as hold ity, overall process productivity, and train complexity are given in the
tanks, pumps and valves for each unit operation as well as the overall Discussion section.
train.

2.2.1 Continuous clarification


2 MATERIALS AND METHODS
A Pall Cadence Acoustic Wave Separator (AWS) was used for continu-
2.1 Materials ous clarification.[15] Four acoustic chambers were connected in series.
Feed flow rate from the fed batch harvest tank was set to 5 mL min-1
The mAb used in the study is an immunoglobulin G1 (IgG1) antibody for both CC and PC mode operation. The acoustic power was adjusted
with a molecular weight of around 150 kDa and a pI of 8.1. 20 L to facilitate >90% reduction of total starting cell density after the clar-
of clarified cell culture harvest containing the mAb at concentration ification. Prior to the runs, the chambers and tubing were cleaned with
5.0 g L-1 were obtained from a major Indian biopharmaceutical manu- 0.1 M NaOH and flushed with deionized water.
facturer. Analytical grade chemicals including glycine, tris base, acetic
acid (CH3 COOH), sodium chloride (NaCl), sodium dihydrogenphos-
phate (NaH2 PO4 ), disodium hydrogen phosphate (Na2HPO4 ), sodium 2.2.2 Continuous depth and sterile filtration
hydroxide (NaOH), hydrochloric acid (HCl), and histidine were pur-
chased from Merck, India. Buffer solutions were prepared using deion- A customized filtration skid was used for continuous dead-end
ized water. All the solutions were filtered through 0.2 µm filters. filtration.[37] A three-way solenoid valve was used to connect three
identical filters in parallel. The solenoid valves are shown as dark blue
diamonds in Figures 1A and 1B. In-line pressure sensors were used
2.2 Methods to monitor the feed pressure and trigger the valve to switch the flow
to a fresh filter whenever pressure exceeded 1 bar indicating filter
A continuous mAb production train was developed at lab scale. Cell fouling, thus ensuring uninterrupted continuous flow. One such skid
culture harvest was obtained in 10 L batches from a fed-batch biore- was fitted with three Supradisc HP PDE2 (0.2–3.5 um, 22 cm2 ) depth
4 of 14 KATEJA ET AL.

F I G U R E 1 Process flow diagrams showing the current platform for continuous mAb production adapted for (A) complete continuous (“CC”)
and (B) periodic continuous (“PC”) modes of operation

filters from Pall Life Sciences and placed after the AWS for continuous For continuous CEX chromatography, Poros HS resin (Applied
depth filtration. Another such skid was fitted with three Acrodisc 4652 Biosystems) was used with a salt gradient with pH 7, 20 mM sodium
(0.2 µm, 5.8 cm2 ) sterile filters from Pall Life Sciences and placed after phosphate buffer as equilibration buffer and pH 7, 20 mM sodium phos-
the pH neutralization step post viral inactivation for continuous sterile phate buffer containing 200 mM NaCl as elution buffer. Three columns
filtration. of 22.0 mL each were used in CC mode, while three columns of 27.0 mL
each were used in PC mode. Table S1 gives the details of the chro-
matography methods for both modes of operation, including column
2.2.3 Continuous chromatography specifications and duration, residence time and flow rate of the loading,
second-pass loading, wash, elution, cleaning, and equilibration steps.
For continuous Protein A chromatography, MabSelectSure resin (GE The column sizing and scheduling decisions are explained further in the
Healthcare) was used. The columns were equilibrated with 20 mM Discussion section. An anion exchange (“AEX”) membrane (Sartobind
phosphate and 150 mM NaCl buffer at pH 7.4 and then loaded with Q nano 1 mL, 4 mm bed height) was placed in-line prior to the CEX
filtered harvest. The unbound material was washed with equilibration columns such that the loading onto the CEX was performed through
buffer. Product was eluted using 100 mM glycine buffer at pH 3. Six the AEX membrane.
columns of 10.0 mL each were used in CC mode, while three columns All chromatography steps were performed on a single Pall Cadence
of 24 mL each were used in PC mode. The loading conditions of clar- BioSMB system using a customized Python-based control software.[38]
ified cell culture harvest from the AWS (flow rate 5.0 mL min-1 , con- The software layer controlled the flow rates of all seven pumps and
centration 5.0 g L-1 ) were kept constant in both modes of operation to the positions of all 240 valves in the BioSMB system in real time based
maintain comparability for productivity analyses. on chromatography scheduling methods coded into a Python-based
KATEJA ET AL. 5 of 14

F I G U R E 2 Schematic showing BioSMB setup for (A) CC and (B) PC mode of operation and the ultrafiltration and diafiltration configuration
along with pumps and valves for (C) CC and (D) PC mode respectively

scheduler. Any desired method or combination of methods could be because of the extra surge tanks before and after the chromatography
coded into the Python scheduler and executed by the system, limited operations in CC mode, requiring pumping action to both supply and
only by the capabilities of the BioSMB system, namely the physical withdraw material from the surge tanks. Conversely, the PC mode does
limitations of not more than 16 columns and not more than 7 not have surge tanks between the chromatography operations, and so
feed/buffer streams. Real-time data collected from other on-line the AWS feed pump provides the driving force for Protein A loading,
or at-line process sensors could also be integrated with the control and the Protein A elution pump provides the driving force for CEX load-
layer to facilitate the implementation of advanced control logic such ing.
as loading and pooling decisions based on analytical tools like NIR The number of valves also differs due to the different number of
spectroscopy[38] or at-line HPLC,[39] as we have demonstrated in columns. The nature of the valve manifold with 240 valves and 16 col-
previous work. umn positions makes the BioSMB a versatile device in which we found
Figures 2A and 2B illustrate the operation of the BioSMB by the it possible to carry out both Protein A and CEX chromatography. How-
Python control layer for the CC and PC modes respectively. The ever, it is also possible to use two separate BioSMBs for each unit oper-
present case study is the first example in the literature of a single ation, as is conventional.[27,40] We have chosen to showcase the use
BioSMB being used to carry out both Protein A and CEX chromatogra- of a single BioSMB as it is an expensive equipment and it is of eco-
phy. The system was able to accommodate both chromatography steps nomic benefit as well as operational advantage to limit the number of
by working within the limitations of pumps and valves as follows. The such equipment in the continuous platform. Both PC and CC modes can
eight inlets of the BioSMB are used to supply Protein A load, Protein be run with either one or two BioSMBs, based on the user’s individual
A second-pass, CEX load, Protein A elution buffer, CEX elution buffer, preference. The design strategy and comparison of equipment, instru-
Protein A wash buffer, CEX wash buffer, and cleaning buffer (same for mentation and scheduling between the CC and PC modes is explored
Protein A and CEX) for both modes of operation. Both modes use an further in the Discussion section.
external gradient pump for providing the CEX elution gradient, and the
Python controller is used to sync the operation of the gradient pump
with the CEX elution schedule. Both modes use five of the six outlets 2.2.4 Continuous viral inactivation
of the BioSMB for Protein A elute, CEX elution pool, Protein A second-
pass, Protein A waste, and CEX waste respectively. The key differences Continuous viral inactivation was achieved using low pH hold in a coiled
between the two modes are the number of BioSMB pumps and valves flow inversion reactor (CFIR) developed in-house.[36,41,42,43] The setup
used. consisted of tubing with internal diameter 6.4 mm (Masterflex L/S 17
It can be seen from Figure 2A that the CC mode uses six of the tubing) wound around a central coiled axis. The length of the CFIR was
seven in-built BioSMB pumps, while the PC mode uses only four. This is adjusted to allow a total low pH hold time of 1 h at the process flow rate
6 of 14 KATEJA ET AL.

for both CC and PC mode operations. The pH of the Protein A elute was tively. Further comparison of the strategy design, process scheduling,
already 3.7, sufficient for viral inactivation. Therefore, pH adjustment and equipment requirements of the two modes is presented in the Dis-
at the entrance to the CFIR was not required. In CC mode operation, cussion section.
the pH was adjusted in the surge tank at the exit of the CFIR (Tank 3 in
Figure 1A) using a peristaltic pump to deliver a constant stream of neu-
tralization solution to the surge tank at a pre-determined flow rate to 2.3 Analysis methods
bring the pH up to 7. In PC mode operation, the flow through the CFIR
was driven by the Protein A elution pump of the BioSMB, as shown in 2.3.1 Analytical size exclusion chromatography
the scheduling chart in Figure 2B. The pH was adjusted in-line using a
Y-connector and a peristaltic pump which was triggered whenever the Superdex 200 column (1.0 mm × 300 mm) from Cytiva Lifesciences
BioSMB Protein A elution pump was triggered. 2 M Tris base was used was employed for size exclusion high performance liquid chromatogra-
as the pH neutralization solution in both modes of operation. phy (SEC-HPLC) for determination of % high molecular weight impu-
rities (HMWI%). Analysis was performed at 0.5 mL min-1 at 25◦ C
using 50 mM phosphate buffer of 300 mM NaCl, pH 6.8 as mobile
2.2.5 Continuous ultrafiltration and diafiltration phase for a runtime of 45 min. Samples, before injection, were filtered
using 0.2 µm, syringe filter. 10 µg was injected for each sample. UV
Single pass tangential flow filtration (SPTFF) was used for continuous absorbance at 280 nm was monitored.
ultrafiltration. A Cadence Single-Pass TFF Modular Kit (Pall Corpora-
tion, USA) was used. The modular setup was fitted with membranes of
regenerated cellulose with a molecular weight cut-off of 30 kDa. Each 2.3.2 Analytical affinity chromatography
membrane had an area of 93 cm2 . Process development was carried out
to construct the modular setups yielding the desired volumetric con- Analytical protein A affinity chromatography was performed for mAb
centration factors (VCFs) for the input flowrates and concentrations in quantification. A Bio-Monolith Protein A (4.95 × 5.2 mm) column from
both the CC and PC setups. The input conditions to the SPTFF mod- Agilent Technologies was used for the analysis. Analysis was performed
ule under CC mode operation were flow rate of 3.6 mL min-1 and con- at 0.5 mL min-1 at 25◦ C using equilibration buffer (50 mM sodium phos-
centration of 6.1 g L-1 . Similarly, the input conditions under PC mode phate, 150 mM sodium chloride at pH 7.5) and elution buffer (50 mM
operation were flow rate of 7.9 mL min-1 and concentration of 5.1 g L-1 . sodium phosphate, 150 mM NaCl, pH 2.5). For analysis, 5 µg of filtered
Further details on the process design which gave rise to these particu- sample was injected and UV absorbance was monitored at 280 nm.
lar flow rates in the two modes of operation are given in Section 3.1.3. Antibody concentration was calculated using a calibration curve (peak
The target final concentration in both cases was 20 g L-1 , corresponding area vs. protein amount) prepared form pure mAb sample of known
to VCFs of 3.3x and 4.0x in the CC and PC cases, respectively. Trans- concentration.
membrane pressure was fixed at around 1 bar in both cases using a
clamp on the retentate line and measuring the feed and retentate pres-
sures using in-line pressure sensors during process development stud- 2.3.3 Analysis of charge variants
ies. The target VCF was achieved by building a modular setup with 3
membranes (2 parallel + 1 in series) for the CC case and 6 membranes Levels of charge variants in the various samples were measured using
(3 parallel + 2 parallel + 1 in series) for the PC case. A Masterflex peri- analytical cation exchange high performance liquid chromatography
staltic pump was used to pump the material from the previous surge (CEX-HPLC). An Agilent Bio MAb NP5 (4.6 × 250 mm, Agilent technolo-
tank through the modular SPTFF in both cases. gies, India) column was connected to Agilent 1260 liquid chromatogra-
For diafiltration, a Pall Cadence In-Line Diafiltration (ILD) module phy system. Analysis was performed at 0.5 mL min-1 at 25◦ C using equi-
was used. The membranes were made of regenerated cellulose with libration buffer (15 mM sodium phosphate, pH 6.2) and elution buffer
a molecular weight cut-off of 30 kDa and area of 0.11 m2 . The same (150 mM sodium phosphate, pH 6.2). The method used for analysis of
unit was used in both CC and PC modes. The retentate stream from the charge variants has been published elsewhere.[44] UV absorbance at
modular SPTFF was connected to the feed port of the ILD module. A 280 nm was monitored.
six-headed peristaltic pump was used to pump the diafiltration buffer
(20 mM histidine, 5 mM acetate, pH 6) into the ILD. The flow rate of the
peristaltic pump was set to 7 times the retentate flowrate of the mod- 2.3.4 HCP and DNA measurement
ular SPTFF, ensuring diafiltration up to 7 diavolumes in a single pass
through the ILD. Prior to the runs, both the SPTFF and ILD were flushed Quantification of residual DNA and host cell proteins (HCP) in the
with deionized water and normalized water permeability test was con- purified mAb samples was performed using the Quant-iT PicoGreen
ducted. Figures 3A and 3B illustrate the operation of the ultrafiltration DNA kit (Invitrogen, UK) and CHO HCP ELISA kit (Cygnus Technolo-
and diafiltration steps for the CC and PC modes, respectively. Table gies, USA) respectively. All the reagents and buffers were prepared
S2 lists the operating parameters for the CC and PC modes, respec- according to manufacturer’s protocols. Detailed methods for both
KATEJA ET AL. 7 of 14

F I G U R E 3 Scheduling charts for the operation of the mAb platform in (A) CC mode and (B) PC mode including details of the chromatography
and ultrafiltration sub-steps. Text in red gives details of the time (“t”) of each step in min and the flow rate (“u”) of each step in mL/min

these analyses have been laid down in our previous publications and conditions of feed flowrate and titer from a fed-batch harvest. The pro-
the same were followed.[6,42] cess flow diagrams for the two modes have been presented in Figure 1
and the detailed process scheduling charts are illustrated in Figure 3.
The CC mode was designed to ensure that both inflow and out-
3 RESULTS AND DISCUSSION flow from each unit operation was completely continuous (Figure 3A).
Thus, the AWS was continuously supplied with material from the fed-
3.1 Leveraging the mAb production platform to batch harvest tank at a rate of 5 mL min-1 , and the outflow was con-
achieve varying levels of continuity tinuously loaded onto the BioSMB for Protein A capture chromatog-
raphy. The capture method was designed to ensure that at least one
The aim of the present work was to use the same mAb platform con- column from the multi-column setup was always undergoing elution.
sisting of an Acoustic Wave Separator (AWS), a 16-column BioSMB PD Full elutions were collected in a surge tank with constant flow into the
system, a coiled flow inversion reactor (CFIR), a modular single-pass CFIR for viral inactivation. The CFIR outflow was therefore also con-
ultrafiltration (SPTFF) device, and an in-line diafiltration (ILD) module tinuous and led to a surge tank where the pH was neutralized, prior
to demonstrate two variants of continuous operation, namely complete to continuous loading onto the same BioSMB for CEX chromatography
continuous (“CC”) and periodic continuous (“PC”), and compare the in bind-and-elute mode (Figure 2A) via a flow-through AEX membrane.
resulting processes in terms of process productivity, consumable pro- Once again, the polishing method was designed to ensure that at least
ductivity, operational complexity, and facility fit, under near-identical one column in the multi-column setup was always undergoing loading
8 of 14 KATEJA ET AL.

and another elution. Volumetric pooling was performed on the elutions and second-pass loading in sequence, with each step of 12 min length
and the pools were collected in a surge tank. From this tank, the mate- (Figure 3A). Given these constraints of loading time, flow rate and titer,
rial was continuously pumped through an SPTFF+ILD setup for for- the Protein A columns were sized at 10 mL volume (12.7 cm height,
mulation followed by storage of the final drug product. Since the flow 1 cm diameter) in CC mode, resulting in a loading residence time of 2
through the SPTFF+ILD was required to be uninterrupted, a parallel min and resin binding capacity of 30 g L-1 .
set of modules was required to which the process flow stream could be The PC mode was designed to minimize operational complexity.
switched during periodic membrane cleaning operations every 8–12 h One of the simplest modes of continuous Protein A chromatography
(Figure 2C). All the tanks in CC mode were maintained at five times the is the three-column periodic counter-current (“PCC”) method.[45] In
elution volume of the preceding step to dampen concentration gradi- this mode, the loading step determines the cycling rate of the columns.
ents in the elution peaks. The durations of the non-loading steps (wash, elution, cleaning, and
In the PC mode of operation, the loading onto Protein A chro- equilibration) are set so that their combined duration is equivalent
matography was performed continuously by designing a multi-column to that of the loading step. This ensures a cyclical operation where
method where one column was always in the loading phase. This was one column is always in the loading phase and another is in second-
because continuous loading onto capture chromatography is an essen- pass while the third undergoes all the remaining steps. Matching the
tial requirement of continuous platforms so that they are capable of elution, wash, cleaning, and equilibration steps with those in the CC
handling continuous upstream perfusion processes. All the subsequent mode resulting in a loading time of 48 min, and columns were sized
unit operations were controlled by the Protein A elutions, without any at 24 mL volume (12 cm height, 1.6 cm diameter) to match the height
additional constraints of continuous inflow or outflow. Therefore, the as closely as possible with the columns in the CC mode. This resulted
overall process flow through the continuous platform was in a pulsed in a loading residence time of 4.8 min and resin binding capacity of
manner, driven by the schedule of the elutions from the Protein A 50 g L-1 . In both PC and CC mode, Protein A columns were oper-
columns (Figure 3B). Protein A elution was directly loaded onto the ated at 70–80% breakthrough, as is optimal for continuous Protein A
CFIR and subsequently onto the CEX columns after in-line pH adjust- chromatography.[6,45,46] It is evident that there was a large difference
ment without any intermediate hold tanks followed by membrane AEX in Protein A resin binding capacity between the CC mode (30 g L-1 ) and
in flow through mode. Each elution was essentially a pulse of material, the PC mode (50 g L-1 ). This is because both trains were designed to
with a 1:1 relationship between the Protein A and CEX columns, and handle the same product flow from upstream in g/min, so as to maintain
no mixing between pulses. The SPTFF+ILD setup was run as shown comparability of the overall train by having the same g per min for the
in Figure 2D, with cleaning operations scheduled in the pause time entire process. Therefore, the necessity of having continuous elution
between CEX elution pulses. A tank was required between the CEX to fulfil the requirements of complete continuity led to faster cycling
elution pool and the SPTFF+ILD setup to prevent concentration gra- times in Protein A, imposed a constraint on the system to have smaller
dients in ultrafiltration feed stream. The tank was also operated in a columns and therefore less residence time in CC mode, impacting the
pulsed manner, with one CEX elution pool being completely processed binding capacity. In PC mode, the loading step was much longer as it
by the SPTFF+ILD modules prior to the next elution being collected in was no longer required to have continuous elution, and the residence
the tank, as shown in Figure 3B. time was lengthened, leading to higher binding capacity.
The BioSMB setups used to implement the six-column capture step
in CC mode and three-column capture step in PC mode are shown in
3.1.1 Design of continuous capture Figure 2, and the scheduling for both modes are detailed in Figure 3. It
chromatography in CC and PC modes should be highlighted that the fixed constraints of load flow rate, load
titer, elution column volumes, and elution residence time result in sig-
One of the major benefits of continuous capture chromatography is nificantly different capture step design in both modes. Adding the addi-
the ability to achieve higher binding capacities by placing a second- tional constraint of continuous elutions to the CC mode led to doubling
pass column in series after the main loading column to capture break- of the number of columns, and the necessity of matching the durations
through material and enable loading up to 70–90% breakthrough, com- of the loading and elution steps.[6] This resulted in a shorter loading
pared to batch-mode where there is only one column and loading is step with reduced residence time and binding capacity. The average
done only up to 5–10% breakthrough to prevent loss of product.[6,45,46] concentration of the elute in the CC mode was 5.7 g L-1 , significantly
This principle of periodic counter-current chromatography (“PCC”) lower than the value of 9.5 g L-1 in the PC mode. However, the total
was applied to both CC and PC modes of operation. The loading flow resin utilized was lower in the CC mode by around 17%.
rate and titer was fixed at 5.0 mL min-1 and 5.0 g L-1 , and elution
was fixed at 5 column volumes and 2.4 min residence time in both
modes (Table S1). This resulted in an elution of 12 min duration in both 3.1.2 Design of continuous polishing
modes. In CC mode, the duration of the loading and elution steps were chromatography in CC and PC modes
matched in order to facilitate both continuous loading and elution. This
led to the development of a six-column method where each of the six The flow rate and concentration of the CEX load was determined by
columns cycled through loading, wash, elution, cleaning, equilibration, those of the elution of the capture step, which had flow rates of 4.2 and
KATEJA ET AL. 9 of 14

10.0 mL min-1 , and average concentrations of 5.7 and 9.5 g L-1 in the affect the residence time and permeate flux respectively.[35] Since
CC and PC modes, respectively. The elution conditions were fixed at 15 both the feed flowrate and the feed concentration in the CC and PC
column volumes and 3 min residence time in both modes, with pooling modes were constrained by the CEX elution, it was necessary to design
of 7.5 column volumes. This resulted in an elution time of 45 min for the SPTFF module configuration such that the output concentration
both modes. Once again, the constraints of continuous loading and elu- target of 20 g L-1 was achieved, corresponding to VCFs of 3.3x and
tion in CC mode led to significant differences in the polishing method 4.0x in the CC and PC cases, respectively. The module configurations
compared to that of the PC mode. Three columns with equal duration were scouted experimentally and it was determined that a 2 parallel
of loading and elution steps were sufficient to achieve the desired con- + 1 series configuration was appropriate for the CC case, while a
tinuity in both steps, as shown in Figure 3A. Under the constraints of 3+2+1 configuration was appropriate for the PC case to achieve the
45 min loading time, 4.3 mL min-1 load flowrate and 5.7 g L-1 load con- required concentration target.
centration, the CEX columns in CC mode were sized at 21.6 mL volume Smooth operation of the ultrafiltration and diafiltration step in
(10.8 cm height, 1.6 cm diameter) with binding capacity of 51 g L-1 and the continuous train requires not only proper membrane sizing and
a loading residence time of 5 min. module configuration, but also setups for periodic buffer flush and
For PC mode, the CEX method was designed so that the elution from cleaning of the membrane modules. For the CC mode, both ultra-
the first capture column would be completely loaded on to the first CEX filtration and diafiltration were carried out continuously with no
column, and so on for each of the three columns. The CEX loadings were pause time. Therefore, a parallel SPTFF+ILD setup was required to
therefore in the form of periodic pulses synced to the Protein A elu- facilitate cleaning without disruption of the flow stream, as shown in
tions. Since the duration of the Protein A elution in PC mode was 12 Figure 2C. For the PC mode, the flow through the membrane modules
min, this was the required loading time. Under the constraints of 12 min was periodic. Therefore, cleaning could be scheduled during periods
loading time, 10.2 mL min-1 load flow rate and 9.5 g L-1 load concentra- of non-operation without requiring a parallel membrane setup, as
tion, the CEX columns in PC mode were sized at 27 mL volume (13.3 cm shown in Figure 2D. It can be seen that the CC mode requires a total
height, 1.6 cm diameter) with binding capacity of 43 g L-1 and loading of six valves to enable process material to be directed through one
residence time of 2.7 min. The elution, wash, cleaning and equilibration set of SPTFF+ILD membranes, while the other undergoes buffer flush
conditions were the same as those in PC mode. and cleaning. Conversely, the PC mode requires only two valves as
The BioSMB setups used to implement the three-column setups in continuous flow is not a constraint, allowing processing, buffer flush
CC and PC modes have been shown in Figure 2, and the scheduling for and cleaning to be scheduled sequentially. The CC mode also requires
both modes are detailed in Figure 3. The sizing and number of columns an extra pump as both material processing and flush/cleaning are
was determined by the capture step in both cases, though the addi- carried out simultaneously. Both modes have a total of four tanks for
tional constraint of continuous elution in CC mode led to some process the process material, flush buffer, cleaning buffer, and formulation
design differences. The average concentration of the elute in the CC buffer, respectively.
mode was 6.1 g L-1 , slightly higher than the value of 5.1 g L-1 in the
PC mode. The total resin utilized was lower in the CC mode by around
20%. Also, there was a non-productive time of around 42 min for each 3.1.4 Comparing start up and shut down times in
column in PC mode, as shown by the white space in the scheduling CC and PC modes
chart in Figure 3B. This was due to the necessity of waiting for Pro-
tein A elutions in order to load the CEX columns due to the pulsed The presence of surge tanks also impacted the start-up and shut
approach. down times of the two modes. All the surge tanks in CC mode were
maintained at 5x the elution volume of the preceding step, except
the first tank which was maintained at 5x the loading volume of the
3.1.3 Design of continuous ultrafiltration and Protein A step. Protein A loading was started after ST01 had filled to
diafiltration in CC and PC modes 300 mL, taking 1 h. Then, loading onto the CFIR was started after five
Protein A elutions were collected in ST02, also taking 1 h. The CEX was
The input conditions to the SPTFF module were determined by the started after five CFIR outputs had collected in ST03, taking another
elution output of the CEX step. Under CC mode operation, the feed 1 h. Finally, the ILC and ILD were started after five CEX elutions were
flow rate to the SPTFF was 3.6 mL min-1 and concentration was 6.1 g collected, taking an additional 4 h. Therefore, the total time for the
L-1 . Similarly, the input conditions under PC mode operation were CC mode to reach steady state was 7 h. Conversely, the lack of surge
flow rate of 7.9 mL min-1 and concentration of 5.1 g L-1 . In single-pass tanks in PC mode allowed it to reach steady state as soon as the first
ultrafiltration, the target concentration must be achieved in a single cycle was completed. The first Protein A elution passed through the
pass through the module. This is done by increasing the membrane CFIR and onto the CEX columns immediately, and the subsequent CEX
length by having several smaller membranes in various series and elution immediately passed through the ILC and ILD. The total start
parallel configurations. Moreover, the flow rate through the module up time was therefore equivalent to the time of Protein A loading,
and the concentration of the input feed material critically affect the which is just 48 min, after which there was no difference between
volumetric concentration factor (VCF) achieved in a single pass, as they start-up operation and steady-state operation due to the periodic
10 of 14 KATEJA ET AL.

scheduling. The logic for shut-down time can be similarly constructed, ally shown significant productivity gains of 20–50%.[6,45,46] Figure 4C
and shut-down is much faster for PC mode compared to CC mode. shows the results of a similar productivity comparison between the CC
Though PC mode is much faster to start-up and shut-down, there and PC modes of operation of our platform. The total process produc-
are few options for at-line process monitoring as there are no surge tivity was calculated from the retentate stream of the ILD module. In
tanks where the product is collected without gradients and easily CC mode, the ILD retentate stream has a constant flow rate 1.1 mL
accessed by immersion probes, though in-line options for monitoring min-1 and concentration of 19.9 g L-1 , while in PC mode the flow rate
are possible in both CC and PC modes. There is also limited scope to and concentration are 20.1 g /L-1 and 2.0 mL min-1 with periodic flow
pause individual unit operations and carry out repairs without stop- (25.5 min on and 22.5 min off), as shown in the scheduling charts in Fig-
ping the whole train, due to the lack of tanks for holding intermediate ure 3. This resulted in the same total productivity of 21.8 g mAb pro-
material. duced per minute in both modes. This is expected as both platforms
are operating at steady-state and are processing the same amount of
upstream material, namely 5.1 g L-1 at 5.0 mL min-1 flowrate equivalent
3.1.5 Product quality results in CC and PC modes to 25.5 g mAb per minute incoming from the upstream fed-batch tank
of operation with 95% yield in the capture step and 90% yield in the polishing step.
However, CC mode had higher specific productivity for Protein A
The continuous platform was successfully demonstrated for both CC resin, CEX resin, and SPTFF membrane area. The faster cycle times and
and PC modes over 24 h runs, with the final drug product consistently smaller column volumes in CC mode, designed to facilitate both contin-
achieving the target critical quality attribute profile of 20 g L-1 mAb uous loading and continuous elution, allowed higher productivity per
concentration, 15% acidic species content and <0.2% aggregate unit resin volume in the chromatography steps. The specific produc-
content, as shown in Figure 4A which summarizes the offline analytics tivity for Protein A and CEX were around 20% and 29% higher in CC
carried out every hour during the continuous operation. Further, mode compared to PC mode, respectively, in units of mg elute/min/mL
HCP concentration of <10 ppm and DNA concentration of <10 ppb resin. In the case of specific productivity of SPTFF, the continuous flow
were obtained in the pooled drug product for both CC and PC mode, in CC mode facilitated lower flowrates compared to the PC mode,
compared to the initial harvest which had HCP content of 92,811 where the operation in the form of periodic pulses and pauses led to
± 4919 ppm and HCDNA content of 978 ± 64 ppm. Representative higher flowrates during the periods of operation to compensate for the
elution chromatograms for the capture and polishing steps are shown periods of hold. As explained in Section 2.2.5, the differences in feed
in Figure 4B. Finally, it may be required to have an additional step for flowrate and feed concentration necessitated different SPTFF module
viral filtration at the end of the process. A similar skid as described configuration to achieve the same target concentration of 20 g L-1 in
in Section 2.2.2 for continuous dead-end depth and sterile filtration both modes. This led to a total productivity difference of 52% in the CC
can be implemented for continuous viral filtration. There would be mode compared to PC mode in units of mg retentate/min/cm2 STPFF
no significant difference between the CC and PC modes as it is a membrane area.
simple flow-through membrane separation with minimal flow rate It is clear that the complete continuous mode of operation offers
constraints. Alternatively, batch-mode viral filtration of the pooled superior specific productivity due to the lower consumable require-
drug product at the end of either the CC or PC mode continuous train ments of resin and membrane area, and that both modes have the same
could be done at the end of the continuous campaign or infrequently overall process productivity governed by the upstream material flow.
throughout. Any combination approaches, such as capture chromatography in CC
mode followed by polishing in PC mode, would result in reduction in
specific productivity, while the total productivity remains unchanged.
3.2 Considerations for choosing between However, it is evident from the process flow diagrams in Figure 1 and
complete and periodic continuity the scheduling charts in Figure 3 that the PC mode is operationally sim-
pler than the CC mode. These differences are summarized in Table 1. It
It is clear that both CC and PC are viable modes of operation for the is evident from the facility related parameters in Table 1 that the opera-
continuous mAb platform. Both were able to handle the same upstream tional simplicity of the PC mode results in economic advantages on the
flow rate and titer and achieve the target critical quality attribute pro- facility front. Fewer vessels, columns, SPTFF modules, and pumps are
file in the final drug product. The basic methods of the unit operations required, decreasing the capital investment cost. Another point to note
were the same in both modes, though there were differences in the col- is that while the lab-scale version of the BioSMB can accommodate up
umn sizing, membrane sizing, number of columns, number of filtration to 16 columns, the pilot-scale version of the equipment can only accom-
modules, and number of auxiliary equipment including tanks, pumps modate up to eight. Since the CC mode of operation has a total of nine
and valves between the two processes. One of the useful metrics of columns, implementing it at pilot scale would necessitate an additional
comparison between different processes is productivity—both overall BioSMB system at a significant capital cost. This would not be required
process productivity and specific productivity of different unit opera- for the PC mode which has only six columns in total.
tions. Multiple researchers have compared these parameters between In summary, increasing level of continuity results in increasing
batch and continuous processes, and continuous processes have gener- specific productivity and consequentially lower cost of manufacturing.
KATEJA ET AL. 11 of 14

F I G U R E 4 (A) Critical quality attributes of mAb concentration, % acidic species and % aggregate content measured in the fed-batch harvest
and final drug product for the two modes of operation, (B) Representative elution chromatogram for the Protein A and cation exchange
chromatography in the two modes of operation along with analytical SEC and CEX-HPLC profile for the final drug product, (C) Comparison of total
and specific productivities for the two modes of operation
12 of 14 KATEJA ET AL.

TA B L E 1 Comparison of the two modes of operation in terms of 4 CONCLUSIONS


facility and consumables requirements

Facility Related Parameters A mAb platform has been developed consisting of continuous clari-

Parameter CC Mode PC Mode fication, depth filtration, capture chromatography, viral inactivation,
polishing chromatography, and formulation. A series of enabling
No. of tanks 4 1
technologies have been used as the building blocks of the platform,
No. of protein A columns 6 3
including an Acoustic Wave Separator, a Cadence BioSMB PD system,
No. of CEX columns 3 3
a customized coiled flow reactor, a modular single-pass TFF kit, an
No. of SPTFF modules 2 1 in-line diafiltration module, and a continuous dead-end filtration
No. of ILD modules 2 1 skid. Two operational cases were executed on the platform, namely
No. BioSMB pumps used (out of 7) 6 4 complete continuous (“CC”) and periodic continuous (“PC”) operation.
No. of BioSMB valves used (out of 240) 75 48 The CC mode was designed to ensure that each unit operation had
No. of auxiliary pumps 6 4 completely continuous inflow and outflow by increasing the number

No. of auxiliary valves 8 4


of columns, filtration modules and tanks, which is what is typically
expected from the term “continuous processing”. Alternatively, the PC
Consumables
mode operated in periodic pulses in line with the intrinsic periodicity of
Volume of protein A resin x 1.2x
chromatography unit operations, where loading and elution are always
Volume of CEX resin x 1.2x sequential. Both modes are viable options for continuous processing
Volume of AEX membrane x x of mAbs and allow the target critical quality attribute profiles of the
Membrane area per SPTFF module x 2x final drug product to be achieved over 24 h of operation.
The two cases were designed to highlight the variety of approaches
in which continuity can achieved in mAb processing, even when the
underlying platform is identical. Both modes were designed to han-
However, so does the complexity of hardware and operations and dle the same flow rate and titers from the upstream bioreactor or
as a result there may be situations where an intermediate level of fed-batch harvest tank, and were compared in terms of total process
continuity may be more suitable. For example, in CC mode, the Protein productivity, specific productivity of the unit operations in terms of
A resin undergoes a larger number of cycles compared to the PC mode. resin and membrane utilization, and operational complexity in terms
In the current case studies, the Protein A cycle time is 12 min in CC of advanced scheduling requirements and number of auxiliary equip-
mode and 48 min in PC mode, resulting in a 4x faster rate of cycling. ment including tanks, valves and pumps. It was found that the CC mode
This is a potential benefit in the case of manufacturing for clinical was superior in terms of specific productivity and consumable utiliza-
trials, where a single manufacturing campaign is planned and it is desir- tion, while the PC mode was operationally simpler and had lower facil-
able to fully exhaust consumables at the end of the campaign.[47,48] ity costs due to significant reductions in the number of auxiliary equip-
Another potential benefit of CC mode is the large number of tanks ment. The work successfully demonstrates that while superiority of
which provide opportunities for in-process monitoring in surge tanks continuous processing over batch mode is evident, varying extent of
where the product is collected without in-process gradients and easily continuity can be achieved. It is of benefit to both industrial and aca-
accessible by immersion probes or conveniently accessible for at-line demic researchers to be aware of the pros and cons of different types
HPLC analysis, in line with US FDA guidelines on Quality by Design in of continuity when designing processes on their respective continuous
biopharmaceutical manufacturing[49,50] by monitoring critical quality platforms using similar enabling technologies.
attributes and critical process parameters throughout the continu-
ous train. On the other hand, the PC mode offers the advantage by ACKNOWLEDGMENTS
preventing mixing of process streams in any of the tanks. The pulsed Authors would like to acknowledge funding support from Depart-
approach in PC mode allows for easy definition of a “batch” as a single ment of Biotechnology, Ministry of Science and Technology (num-
Protein A elution pulse passes through the subsequent unit operations ber BT/COE/34/SP15097/2015) and Ministry of Human Resources
of viral inactivation, polishing chromatography, sterile filtration and and Development (Uchchatar Avishkar Yojana/MHRD 21–105/2015-
formulation without ever being mixed with other pulses. This 1:1 TS.II/TC). Nikhil Kateja would like to thank Council of Scientific
matching between capture and elution columns mitigates the effect of and Industrial Research (CSIR), New Delhi (India) for financial
column failure or in-process errors such as wrong elution gradient, and assistance in the form of Research Associate Fellowship (File no.
allays many of the concerns felt by the batch-processing community 09/086(1363)/2019-EMR-1). Anamika Tiwari and Garima Thakur
in terms of the risks associated with pooling drug substance with would like to acknowledge support from TCS Research Scholar Pro-
different product quality profiles in continuous processes.[51] The final gram (RSP) cycles 14 and 15, respectively.
decision of going for complete, periodic or hybrid continuity in a mAb
platform process should be taken after due consideration of these CONFLICT OF INTEREST
factors. The authors declare no financial or commercial conflict of interest.
KATEJA ET AL. 13 of 14

AUTHOR CONTRIBUTIONS 12. Richardson, A., & Walker, J. (2018). Continuous solids-discharging cen-
Nikhil Kateja: conceptualization; investigation; methodology; writing- trifugation: A solution to the challenges of clarifying high-cell-density
mammalian-cell cultures. BioProcess International.
original draft. Garima Thakur: conceptualization; investigation;
13. Karst, D. J., Serra, E., Villiger, T. K., Soos, M., & Morbidelli, M. (2016).
methodology; writing-original draft. Anamika Tiwari: conceptualiza- Characterization and comparison of ATF and TFF in stirred biore-
tion; investigation; methodology; writing-original draft. Anurag S. actors for continuous mammalian cell culture processes. Biochemical
Rathore: Conceptualization; funding acquisition; project administra- Engineering Journal, 110, 17–26. https://doi.org/10.1016/j.bej.2016.
02.003.
tion; supervision; writing-review and editing.
14. Hadpe, S. R., Sharma, A. K., Mohite, V. V., & Rathore, A. S. (2017). ATF
for cell culture harvest clarification: Mechanistic modelling and com-
DATA AVAILABILITY STATEMENT parison with TFF. Journal of Chemical Technology & Biotechnology, 92(4),
Data available on request from the authors. The data that support the 732–740. https://doi.org/10.1002/jctb.5165.
15. Hong, J. S., Azer, N., Agarabi, C., & Fratz-Berilla, E. J. (2020). Primary
findings of this study are available from the corresponding author upon
clarification of CHO harvested cell culture fluid using an acoustic sep-
reasonable request. arator. JoVE (Journal of Visualized Experiments), (159), e61161. https:
//doi.org/10.3791/61161.
ORCID 16. Collins, M., & Levison, P. (2016). Development of high performance
Anurag S. Rathore https://orcid.org/0000-0002-5913-4244 integrated and disposable clarification solution for continuous Biopro-
cessing. Bioprocess International, 14, 30–33.
17. Steinebach, F., Müller-Späth, T., & Morbidelli, M. (2016). Continu-
REFERENCES ous counter-current chromatography for capture and polishing steps
1. Rathore, A. S., Agarwal, H., Sharma, A. K., Pathak, M., & Muthukumar, S. in biopharmaceutical production. Biotechnology Journal, 11(9), 1126–
(2015). Continuous processing for production of biopharmaceuticals. 1141. https://doi.org/10.1002/biot.201500354.
Preparative Biochemistry and Biotechnology, 45(8), 836–849. https://doi. 18. David, L., Maiser, B., Lobedann, M., Schwan, P., Lasse, M., Ruppach, H., &
org/10.1080/10826068.2014.985834. Schembecker, G. (2019). Virus study for continuous low pH viral inac-
2. Yang, O., Prabhu, S., & Ierapetritou, M. (2019). Comparison between tivation inside a coiled flow inverter. Biotechnology and Bioengineering,
batch and continuous monoclonal antibody production and economic 116(4), 857–869. https://doi.org/10.1002/bit.26872.
analysis. Industrial & Engineering Chemistry Research, 58(15), 5851– 19. Gillespie, C., Holstein, M., Mullin, L., Cotoni, K., Tuccelli, R., Caulmare,
5863. https://doi.org/10.1021/acs.iecr.8b04717.s001. J., & Greenhalgh, P. (2019). Continuous in-line virus inactivation for
3. Zydney, A. L. (2016). Continuous downstream processing for high next generation bioprocessing. Biotechnology Journal, 14(2), 1700718.
value biological products: A review. Biotechnology and Bioengineering, https://doi.org/10.1002/biot.201700718.
113(3), 465–475. https://doi.org/10.1002/bit.25695. 20. Klutz, S., Lobedann, M., Bramsiepe, C., & Schembecker, G. (2016). Con-
4. Jungbauer, A. (2013). Continuous downstream processing of biophar- tinuous viral inactivation at low pH value in antibody manufacturing.
maceuticals. Trends in Biotechnology, 31(8), 479–492. https://doi.org/ Chemical Engineering and Processing: Process Intensification, 102, 88–
10.1016/j.tibtech.2013.05.011. 101. https://doi.org/10.1016/j.cep.2016.01.002.
5. Konstantinov, K. B., & Cooney, C. L. (2015). White paper on continu- 21. Parker, S. A., Amarikwa, L., Vehar, K., Orozco, R., Godfrey, S., Coffman,
ous bioprocessing May 20–21 2014 continuous manufacturing sym- J., Shamlou, P., & Bardliving, C. L. (2018). Design of a novel continuous
posium. Journal of Pharmaceutical Sciences, 104(3), 813–820. https:// flow reactor for low pH viral inactivation. Biotechnology and Bioengi-
doi.org/10.1002/jps.24268. neering, 115(3), 606–616. https://doi.org/10.1002/bit.26497.
6. Kateja, N., Kumar, D., Sethi, S., & Rathore, A. S. (2018). Non-protein 22. Martins, D. L., Sencar, J., Hammerschmidt, N., Flicker, A., Kindermann,
A purification platform for continuous processing of monoclonal anti- J., Kreil, T. R., & Jungbauer, A. (2020). Truly continuous low pH viral
body therapeutics. Journal of Chromatography A, 1579, 60–72. https: inactivation for biopharmaceutical process integration. Biotechnology
//doi.org/10.1016/j.chroma.2018.10.031. and Bioengineering, 117(5), 1406–1417. https://doi.org/10.1002/bit.
7. Arnold, L., Lee, K., Rucker-Pezzini, J., & Lee, J. H. (2019). Implemen- 27292.
tation of fully integrated continuous antibody processing: Effects on 23. Jabra, M. G., Yehl, C. J., & Zydney, A. L. (2019). Multistage continuous
productivity and COGm. Biotechnology Journal, 14(2), 1800061. https: countercurrent diafiltration for formulation of monoclonal antibod-
//doi.org/10.1002/biot.201800061. ies. Biotechnology Progress, 35(4), e2810. https://doi.org/10.1002/btpr.
8. Hummel, J., Pagkaliwangan, M., Gjoka, X., Davidovits, T., Stock, R., Ran- 2810.
sohoff, T., Gantier, R., & Schofield, M. (2019). Modeling the down- 24. Yehl, C. J., & Zydney, A. L. (2020). Single-use, single-pass tangential flow
stream processing of monoclonal antibodies reveals cost advantages filtration using low-cost hollow fiber modules. Journal of Membrane Sci-
for continuous methods for a broad range of manufacturing scales. ence, 595, 117517. https://doi.org/10.1016/j.memsci.2019.117517.
Biotechnology Journal, 14(2), 1700665. https://doi.org/10.1002/biot. 25. Rucker-Pezzini, J., Arnold, L., Hill-Byrne, K., Sharp, T., Avazhanskiy, M.,
201700665. & Forespring, C. (2018). Single pass diafiltration integrated into a fully
9. Gupta, S. K. (2017). Upstream continuous process development. Con- continuous mAb purification process. Biotechnology and Bioengineering,
tinuous Biomanufacturing: Innovative Technologies and Methods. 115(8), 1949–1957. https://doi.org/10.1002/bit.26708.
10. Somasundaram, B., Pleitt, K., Shave, E., Baker, K., & Lua, L. H. 26. Godawat, R., Konstantinov, K., Rohani, M., & Warikoo, V. (2015). End-
(2018). Progression of continuous downstream processing of mon- to-end integrated fully continuous production of recombinant mono-
oclonal antibodies: Current trends and challenges. Biotechnology clonal antibodies. Journal of Biotechnology, 213, 13–19. https://doi.org/
and Bioengineering, 115(12), 2893–2907. https://doi.org/10.1002/bit. 10.1016/j.smim.2009.11.002.
26812. 27. Klutz, S., Magnus, J., Lobedann, M., Schwan, P., Maiser, B., Niklas, J.,
11. Mehta, S. (2014). Automated single-use centrifugation solution for Temming, M., & Schembecker, G. (2015). Developing the biofacility of
diverse biomanufacturing process. In G. Subramanian (Ed.), Continuous the future based on continuous processing and single-use technol-
Processing in Pharmaceutical Manufacturing (pp. 385–399). Wiley-VCH ogy. Journal of Biotechnology, 213, 120–130. https://doi.org/10.1016/
Verlag GmbH & Co. KGaA. j.jbiotec.2015.06.388.
14 of 14 KATEJA ET AL.

28. Klutz, S., Magnus, J., Lobedann, M., Schwan, P., Maiser, B., Niklas, 42. Kateja, N., Agarwal, H., Saraswat, A., Bhat, M., & Rathore, A. S. (2016).
J., Temming, M., & Schembecker, G. (2017). Design and operation Continuous precipitation of process related impurities from clarified
of a continuous integrated monoclonal antibody production pro- cell culture supernatant using a novel coiled flow inversion reactor
cess. Biotechnology Progress, 33(5), 120–130. https://doi.org/10.1016/ (CFIR). Biotechnology Journal, 11(10), 1320–1331. https://doi.org/10.
j.jbiotec.2015.06.388. 1002/biot.201600271.
29. Patel, B. A., Pinto, N. D. S., Gospodarek, A., Kilgore, B., Goswami, K., 43. Kateja, N., Nitika, N., Fadnis, R. S., & Rathore, A. S. (2021). A novel
Napoli, W. N., Desai, J., Heo, J. H., Panzera, D., Pollard, D., Richard- reactor configuration for continuous virus inactivation. Biochemical
son, D., Brower, M., & Richardson, D. D. (2017). On-line ion exchange Engineering Journal, 167, 107885. https://doi.org/10.1016/j.bej.2020.
liquid chromatography as a process analytical technology for mon- 107885.
oclonal antibody characterization in continuous bioprocessing. Ana- 44. Joshi, V., Kumar, V., & Rathore, A. S. (2015). Rapid analysis of charge
lytical Chemistry, 89(21), 11357–11365. https://doi.org/10.1021/acs. variants of monoclonal antibodies using non-linear salt gradient in
analchem.7b02228. cation-exchange high performance liquid chromatography. Journal of
30. Feidl, F., Vogg, S., Wolf, M., Podobnik, M., Ruggeri, C., Ulmer, N., Wälchli, Chromatography A, 1406, 175–185. https://doi.org/10.1016/j.chroma.
R., Souquet, J., Broly, H., Butté, A., & Morbidelli, M. (2020). Process- 2015.06.015.
wide control and automation of an integrated continuous manufactur- 45. Godawat, R., Brower, K., Jain, S., Konstantinov, K., Riske, F., & Warikoo,
ing platform for antibodies. Biotechnology and Bioengineering, 117(5), V. (2012). Periodic counter-current chromatography–design and oper-
1367–1380. https://doi.org/10.1002/bit.27296. ational considerations for integrated and continuous purification of
31. Rathore, A. S., & Kateja, N. (2016). A coiled flow inversion reactor proteins. Biotechnology Journal, 7(12), 1496–1508. https://doi.org/10.
enables continuous processing. BioPharm International, 29(12), 32–35. 1002/biot.201200068.
32. Kateja, N., Kumar, D., Godara, A., Kumar, V., & Rathore, A. S. (2017). 46. Warikoo, V., Godawat, R., Brower, K., Jain, S., Cummings, D., Simons,
Integrated chromatographic platform for simultaneous separation of E., Johnson, T., Walther, J., Yu, M., Wright, B., Mclarty, J., Karey, K.
charge variants and aggregates from monoclonal antibody therapeu- P., Hwang, C., Zhou, W., Riske, F., & Konstantinov, K. (2012). Inte-
tic products. Biotechnology Journal, 12(11), 1700133. https://doi.org/ grated continuous production of recombinant therapeutic proteins.
10.1002/biot.201700133. Biotechnology and Bioengineering, 109(12), 3018–3029. https://doi.org/
33. Shi, C., Gao, Z. Y., Zhang, Q. L., Yao, S. J., Slater, N. K., & Lin, D. Q. 10.1002/bit.24584.
(2020). Model-based process development of continuous chromatog- 47. Pollock, J., Bolton, G., Coffman, J., Ho, S. V., Bracewell, D. G., & Farid, S. S.
raphy for antibody capture: A case study with twin-column system. (2013). Optimising the design and operation of semi-continuous affin-
Journal of Chromatography A, 1619, 460936. https://doi.org/10.1016/ ity chromatography for clinical and commercial manufacture. Journal
j.chroma.2020.460936. of Chromatography A, 1284, 17–27. https://doi.org/10.1016/j.chroma.
34. Steinebach, F., Angarita, M., Karst, D. J., Müller-Späth, T., & Morbidelli, 2013.01.082.
M. (2016). Model based adaptive control of a continuous capture pro- 48. Xenopoulos, A. (2015). A new, integrated, continuous purification pro-
cess for monoclonal antibodies production. Journal of Chromatography cess template for monoclonal antibodies: process modeling and cost of
A, 1444, 50–56. https://doi.org/10.1016/j.chroma.2016.03.014. goods studies. Journal of Biotechnology, 213, 42–53. https://doi.org/10.
35. Thakur, G., Thori, S., & Rathore, A. S. (2020). Implementing PAT for 1016/j.jbiotec.2015.04.020.
single-pass tangential flow ultrafiltration for continuous manufac- 49. Rathore, A. S., & Winkle, H. (2009). Quality by design for biopharma-
turing of monoclonal antibodies. Journal of Membrane Science, 613, ceuticals. Nature Biotechnology, 27(1), 26–34. https://doi.org/10.1038/
118492. https://doi.org/10.1016/j.memsci.2020.118492. nbt0109-26.
36. Rathore, A. S., Nigam, K. D. P., Pathak, M., Agarwal, H., Sharma, A. K., 50. Rathore, A. S. (2009). Roadmap for implementation of quality by design
Kateja, N., & Hebbi, V. (2016). A coiled flow inverter reactor for con- (QbD) for biotechnology products. Trends in Biotechnology, 27(9), 546–
tinuous refolding of denatured recombinant proteins and other mixing 553. https://doi.org/10.1016/j.tibtech.2009.06.006.
operations. International Application No. PCT/IN2016/000022, Publi- 51. Coffman, J., Lin, H., Wang, S., Godfrey, S., Orozco, R., Yildirim, S., Salm,
cation No. WO/2016/116947. J., Hiller, G., Gagnon, M., Farner, R., Kottmeier, B., Sullivan, D. (2017).
37. Thakur, G., Hebbi, V., Parida, S., & Rathore, A. S. (2020). Automation Balancing continuous, integrated, and batch processing in Proc. of the
of dead end filtration: An enabler for continuous processing of bio- 2017 Integrated Continuous Biomanufacturing III (Eds: Farid, S., Goudar,
therapeutics. Frontiers in Bioengineering and Biotechnology, 8, 758. https: C., Alves, P., Warikoo, V.), ECI Symposium Series. https://dc.engconfintl.
//doi.org/10.3389/fbioe.2020.00758. org/biomanufact_iii/71/.
38. Thakur, G., Hebbi, V., & Rathore, A. S. (2020). An NIR-based PAT
approach for real-time control of loading in Protein A chromatogra-
phy in continuous manufacturing of monoclonal antibodies. Biotechnol-
ogy and Bioengineering, 117(3), 673–686. https://doi.org/10.1002/bit. SUPPORTING INFORMATION
27236. Additional supporting information may be found online in the Support-
39. Tiwari, A., Kateja, N., Chanana, S., & Rathore, A. S. (2018). Use of ing Information section at the end of the article.
HPLC as an enabler of process analytical technology in process chro-
matography. Analytical Chemistry, 90(13), 7824–7829. https://doi.org/
10.1021/acs.analchem.8b00897.
40. Ötes, O., Bernhardt, C., Brandt, K., Flato, H., Klingler, O., Landrock,
How to cite this article: Kateja, N., Tiwari, A., Thakur, G., &
K., Lohr, V., Stähler, R., & Capito, F. (2020). Moving to CoPACaPAnA:
Implementation of a continuous protein A capture process for anti- Rathore, A. S. (2021). Complete or periodic continuity in
body applications within an end-to-end single-use GMP manufactur- continuous manufacturing platforms for production of
ing downstream process. Biotechnology Reports, 26, e00465. monoclonal antibodies?. Biotechnol J, 16:e2000524.
41. Sharma, A. K., Agarwal, H., Pathak, M., Nigam, K. D., & Rathore, A. S.
https://doi.org/10.1002/biot.202000524
(2016). Continuous refolding of a biotech therapeutic in a novel Coiled
Flow Inverter Reactor. Chemical Engineering Science, 140, 153–160.
https://doi.org/10.1016/j.ces.2015.10.009.

You might also like