You are on page 1of 33

eBook Series

2015
Most Read
TopArticles
5
FORMULATION, FILL-FINISH
Evaluating Freeze-Thaw Processes in Biopharmaceutical Development

SINGLE-USE PRODUCTION
A Novel Seed-Train Process

MONOCLONAL ANTIBODIES
Hamster Phospholipase B-Like 2 (PLBL2)

BIOSIMILARS
Rapid Development and Scale-Up of Biosimilar Trastuzumab

QUALITY CONTROL
Fundamental Strategies for Viral Clearance
B i o P r o c e s s TECHNICAL

Evaluating Freeze–Thaw Processes


in Biopharmaceutical Development
Small-Scale Study Designs

Manasi Puri, Sorina Morar-Mitrica, George Crotts, and Douglas Nesta

R
egulations mandate that manufacture. Freezing provides
biopharmaceutical product f lexibility and cost savings by
quality be controlled enabling batch processing: Large
throughout manufacturing, volumes of an expensive biological
storage, transportation, and delivery drug substance can be frozen in
to patients (1). Operations often batches to allow the drug product
include freezing and thawing of a to be manufactured based on real-
bulk drug substance, dilution of time commercial or clinical
that purified substance to a target demands. Freezing a drug substance
concentration, filtration, filling into before drug product manufacture
a selected container–closure system, also reduces microbial growth risk
additional processing (e.g., and eliminates mechanical
lyophilization), inspection, (agitation) stresses during
packaging, storage, transport, and transportation.
delivery (2). Most important, this allows for a
Freezing is a common processing longer shelf life. Freezing decelerates component container–closure
step used to maintain stability and chemical degradation (because systems, such as vials or prefilled
quality of a drug substance during reaction kinetics depend directly on syringes with rubber stopper
development and production of temperature) and physical closures (2). Interactions with
biopharmaceutical products. It is degradation (immobilizing drug container surfaces (e.g., glass or
generally agreed upon that freezing substance in a frozen matrix limits polymers) and other constituents
drug substance maximizes protein–protein interactions) (3–13). (e.g., silicone oil) can significantly
productivity and reduces overall Furthermore, application of freeze– inf luence drug substance and drug
production costs by decoupling bulk thaw processes extends to storage of product stability. Such interactions
solution manufacture and storage process intermediates, allowing for may be further exacerbated with
steps from final product longer hold steps between mechanical stresses such as agitation
manufacturing operations. Although during transportation. Freezing can
freezing is considered to be a limit exposure of a protein to those
Product Focus:  Monoclonal
conservative approach, frozen interfaces and prevent shipping- and
antibodies
storage is one of the most efficient storage-related solution instabilities.
Process Focus:  Downstream and reliable methods to minimize Overall, freezing can be leveraged
processing, formulation, fill–finish protein interactions with container– to maximize protein stability
closures and extend the shelf life of throughout a supply chain. However,
Who Should Read:  Process/product biological products in solution-based additional controls may be needed to
development, manufacturing, and formulations (13). limit the number of freeze–thaw
analytical personnel Drug substance (typically stored cycles that occur at clinics,
in plastic bottles or single-use plastic pharmacies, or depot sites to that
Keywords:  Drug product, drug
bags) is exposed to various interfaces supported by available stability data.
substance, stability, freezing, thawing,
on contact with container–closure Doing so ensures that product
single-use technology
systems. Likewise, drug product quality is not compromised.
Level:  Intermediate often is filled into multiple-

BioProcess International 13(1) January 2015


Freeze –Thaw R elated to concentrated solutes — due to Passive freezing has been shown to
Process Challenges water crystallization — also can result in protein aggregation (18).
Freezing and thawing a result in a loss of a protein's It has been reported that active
biopharmaceutical product may thermodynamic stability, leading to control of freezing rates can reduce
change the chemical and physical unfolding events and eventually stresses that cause protein structural
properties of the product solution. causing aggregation. and functional losses. Freezing rates
That in turn can stress proteins, Fast freezing rates lead to smaller can be highly controlled at
may irreversibly denature complex ice-crystal formation, which predetermined levels; however, such
macromolecular structure, and exposes proteins to a large ice– an approach requires highly
could alter their stability (7, 13). liquid interface. Concentration and specialized and expensive
Because freeze–thaw process adsorption of proteins at the surface equipment (including freeze dryers
parameters have been identified to of ice crystals along the ice–liquid or commercially available controlled
play a significant role in product interface can result in their partial freeze–thaw systems). Actively
quality through alteration of a unfolding, increased aggregation, controlled thawing also involves
protein’s microenvironment, gaining and decreased biological activity (17, specialized equipment such as
product- and process-specific 18). One control strategy is the use programmable water baths and
knowledge is critical to using of formulation excipients that act as freeze dryers. In contrast, passive
freeze–thaw applications (11, 14, 15). stabilizers (e.g., surfactants and thawing can be achieved, for
Freezing and thawing can occur sugar cryoprotectants) to reduce the example, by placing a frozen
at varying rates, especially when level of protein denaturation container at room temperature or at
passively performed (at an induced upon exposure to ice-liquid 2–8 °C. Thus, the need to fully
uncontrolled rate). Those rates are interfaces (18). control freezing and thawing rates
known to affect protein stability. Of further concern is the fact should be evaluated carefully.
No standard and agreed definition that fast freezing can entrap air in Freeze–thaw studies are the most
of freezing or thawing rates has yet the ice. When released during direct approaches to evaluating the
been adopted across the industry thawing, the entrapped air can impact of freezing and thawing
(11, 14). Depending on the denature proteins as air–liquid rates and to defining process
equipment used, freezing rates may interfaces form (19, 20). conditions (14).
be slow (<1 °C/min), intermediate Correspondingly, slow thawing
(1–10 °C/min), or rapid (10–900 °C/ rates can result in ice S mall-S cale D esigns
min). recrystallization, and associated of Freeze –Thaw Studies
For example, fast freezing rates shear stress can damage proteins Early in development, material
are achieved by immersing samples (21). Ideally, rapid freezing and availability is usually limited. This
in subzero solvents or baths thawing are usually preferred for creates a need for reliable small-
consisting of alcohols or liquid protein stability (22). scale models for various freeze–thaw
nitrogen (80–900 °C/min). Other Concerns: Published processes that mimic, as closely as
Thawing is also performed at literature offers no consistent possible, the processes and
various rates. Like freezing, terminology or parameters to conditions that will occur at large
thawing can be slow (1–5 °C/min), describe and execute various rates of scale (2). Small-scale models must
or intermediate freezing and thawing. Freeze–thaw be designed or selected that are
(>5 °C/min). It is not always feasible studies in traditional stability predictive of potential large-scale
to monitor temperature profiles programs use passive freezing and freeze–thaw issues.
during freezing and thawing. thawing (19). Such procedures Figure 1 illustrates a systematic
Therefore, in design of a systematic emulate a freezing process in which approach to designing such studies
small-scale study, it is important to a drug substance or drug product is for evaluation of freezing and
incorporate an evaluation of simply placed on a shelf in a freezer thawing parameters as well as
different freezing and thawing rates storage chamber and permitted to protein stability following freeze–
(16, 17). cool and freeze at an uncontrolled thaw stress. In general, study design
Slow freezing rates can result in rate. Temperatures can vary among starts with selection of a
cryoconcentration, in which different commercial freezer units representative formulation and
proteins and excipients form used for storage, partly due to small-scale container–closure, the
concentration gradients near the differences in their size, cooling number of freeze–thaw cycles
freeze front and get excluded from capacity, and loads. Even within the needed for large-scale manufacture,
the ice–liquid interface (2). This same freezer chamber, the freezing freezing and thawing temperatures
can further lead to pH shifts and rate experienced by a protein and rates, and equipment and
phase separation among those solution could vary depending on protocols. Study design is followed
components, resulting in protein the freezer load and the container’s by execution of a protocol. Finally,
structural damage (2, 14). Exposure position within the chamber. long-term effects of freeze–thaw

January 2015 13(1) BioProcess International


stress are evaluated through a stability study evaluating be paid to selecting appropriate scaled-down container–
protein attributes under different storage conditions. closure systems or configurations.
Design of small-scale studies is based on the most For laboratory-scale studies, it is important to
representative freeze–thaw conditions to be used at a choose an appropriate fill volume with the
manufacturing site (8). Product stability depends on representative head space or surface-area-to-volume
selected formulation and container–closure materials. exposure. Scale-down is based on an attempt to
Thus, freeze–thaw evaluation first must include maintain the ratio of surface area to volume similar to
assessment of a product’s compatibility with the contact that at large scale. However, because of significantly
material of the container–closure system during different path lengths, cooling and heating rates
freezing and thawing. Second, special attention must encountered during freezing and thawing are generally
much slower at large scale. Thus, process times for
large-scale operations are longer than for small-scale
Figure 1:  Small-scale study design considerations for evaluating various
freezing and thawing process parameters
studies, particularly with passive freezing and thawing
processes. As a result, large-scale cryoconcentration
becomes an important factor governing product quality.
Drug Product/Substance It can occur at small-scale too but is magnified by bulk
freezing in which the extent of freeze concentration
can be directly correlated to solution volume (3).
Select representative small-scale container Moreover, it is not always feasible to monitor
(materials of construction and fill volume). temperature profiles during freezing and thawing. As
stated above, in designing a systematic small-scale
study, it is important to evaluate different freezing and
Select relevant number of freeze−thaw cycles thawing rates under both active-control and passive
aligned with production scale. conditions, then determine whether those rates
constitute a critical process parameter (CPP) for
product quality (14, 23).
Select representative freezing and thawing Our focus herein resides primarily on the impact of
temperatures and rates based on freezing and thawing rates on drug substance and drug
process knowledge. product stability in representative small-scale
container–closure systems. In addition, our small-scale
case studies were designed with an emphasis on
Select laboratory equipment to execute study. adopting a robust approach to understanding
interactions between formulations and process
conditions. Overall, these studies systematically and
comprehensively evaluate the impact of relevant
Execute study with appropriate stress protocol.
processing parameters (freezing and thawing rates),
formulation parameters (MAb concentration, pH,
excipient levels), and container–closure systems on
short- and long-term MAb storage stability. In these
four case studies, we assessed the impact of varying
Perform active Perform passive
(controlled rates) (uncontrolled rates) freezing and thawing rates on the stability of
freeze−thaw freeze−thaw • a low-concentration MAb-A drug product in glass
protocol. protocol. vials
• a high-concentration MAb-B drug product in glass
vials and drug substance in plastic bottles
• a MAb-C drug substance in single-use bags.
Here we show that understanding of a product’s
Test samples immediately after stress and/or robustness to freeze–thaw stress can be achieved using
place on long-term stability testing carefully designed scale-down models for freezing and
according to stability protocol.
thawing.

C ase Study 1
Perform analytical and biophysical testing. A Controlled Freeze–Thaw Study of MAb-A at a Low
Concentration (0.2 mg/mL): MAb-A was formulated as
a refrigerated liquid at low concentration (0.2 mg/mL)
for intravenous (IV) administration. Early clinical
Evaluate effects of freezing and thawing rates
on product quality.
in-use stability studies had revealed significant
adsorption onto IV bags and giving sets upon addition

BioProcess International 13(1) January 2015


Figure 2 (Case Study 1):  Low-
of the drug product to diluent in Fast Thaw:
concentration MAb-A solutions in
the bags due to the resulting low glass vials were subjected to two
Thermocirculating
Slow Freeze: From water bath set at 25 °C
dosing concentration (0.002 mg/ freeze–thaw cycles. Samples were
25 °C to −40 °C
mL). To overcome significant either fast-frozen in a dry ice and (freeze dryer)
protein loss to such binding, a acetone slurry (active fast freeze
protocol) or slow-frozen at a rate of
surfactant (polysorbate 80, PS80)
0.1 °C/min (active slow freeze
was introduced to the product protocol) using a laboratory freeze-
formulation at a concentration of dryer. Frozen samples were
0.1% (w/v). Although the addition subsequently thawed in a water bath
of PS80 to the formulation at ambient temperature (active fast
thaw protocol) or thawed at the rate Fast Freeze:
overcame the adsorption challenges 1/1 dry ice and
of 0.1 °C/min (active slow thaw
and provided protection from protocol) in a laboratory freeze dryer. acetone slurry Slow Thaw: From
−40 °C to 25 °C
mechanical stress, it also accelerated (freeze
Active Freezing Active
chemical degradation of the MAb, Rate Thawing Rate dryer)
in particular by oxidation. Fast freeze Fast thaw
Literature shows that degradation Slow freeze Slow thaw
products generated through the Fast freeze Slow thaw
oxidation-induced degradation Slow freeze Fast thaw
pathway lead to accelerated MAb
deamidation, fragmentation, and
aggregation (24). Similar behavior Figure 3 (Case Study 1):  Summary results of in solution observed by size-
was observed here. Therefore, %oxidation at methionine (Met) residues exclusion chromatography (SEC) or
oxidation of MAb-A needed to be following two actively controlled freeze–thaw ProteinSimple microf low imaging
cycles for MAb-A; refer to Figure 2 for study
controlled to maintain product setup and sample description.
(MFI) analyses, respectively. No
quality. significant changes were observed
10
This oxidation challenge was 9
in general appearance (GA), pH, or
% MAb A Oxidation

Met 254
identified shortly before 8 absorbance at 280 nm (A280)
at Met Residues

Met 430
Met 48
commencement of phase 3 clinical 7 analyses for all samples.
6
trials. A multicomponent control 5 This small-scale study design
strategy was devised to overcome 4 permitted rapid evaluation of the
the identified surfactant-induced 3 impact of extreme freezing and
2
chemical degradation and allow 1 thawing rates on MAb-A quality. It
on-time start of clinical trials and 0
Initial Fast Fast Slow Slow supported implementation of
submission of a licensing Fast Slow Fast Slow freezing and thawing processes
application. A key element of the Freeze−Thaw during manufacture and storage of
strategy entailed changing the this product in the selected
proposed long-term storage container–closure system. Study
conditions for this drug product slow-freeze, and slow-thaw results verified that control of two
from refrigerated to frozen, which protocols described. All solutions significant process parameters —
could sufficiently inhibit the were tested in glass vials (the drug freezing and thawing rates — would
observed chemical degradation. product container–closure) and not be required during handling of
However, before phase 3 supplies subjected to two freeze–thaw cycles. this product.
were manufactured, it was also As Figure 3 shows, MAb-A
imperative to assess the impact of samples subjected to two controlled C ase Study 2
process parameters (e.g., freeze– freeze–thaw cycles experienced no Evaluation of MAb-B Stability at 200
thaw rates) on drug product quality increase in oxidation over the initial mg/mL After Short-Term Exposure to
and stability. set of samples. Deamidation Subzero Temperatures Above Tg ’:
The experimental plan for a (previously noted as a common Protein concentration and container
small-scale, freeze–thaw study of degradation pathway for this MAb) type can significantly affect MAb
MAb-A consisted of a statistical also was not detected. Fluorescence behavior in a frozen state. So can
combination of two factors at two intensities did not change, storage temperature in relation to
levels each. Factor 1 was the type of indicating maintenance of the the glass transition temperature
stress (freeze or thaw), and factor 2 overall tertiary structure and (Tg′), potentially causing irreversible
was the stress rate (slow or fast). packing of the MAb when frozen damage such as loss of structure
Figure 2 shows the overall study and thawed at different rates. No through partial unfolding and
design, which consisted of four significant changes to the aggregate adsorption to a container through
combinations of stress and stress or fragment profile were observed, exposure of hydrophobic residues (7,
rates: the fast-freeze, fast-thaw, nor was any increase of particulates 11). Loss of structural integrity can

January 2015 13(1) BioProcess International


Table 1 (Case Study 2):  Summary of a small-scale study designed to expose MAb-B drug
substance and drug product in their representative container–closure systems to various subzero placebo control were filled into
temperatures under active control small-scale high-density
Container 72 Three Six One polyethylene (HDPE) bottles and
Freeze–Thaw Stress MAb-B Sample Type Hours Months Months Year glass vials that were representative
Perform one cycle Drug substance HDPE T T T NT of the proposed commercial
of controlled 200 mg/mL (100 mL)
freezing at 0 °C,
configuration. Active and placebo
Formulation buffer T T T NT
–5 °C, or –15 °C. solutions were cooled or frozen to
Drug product Glass Vial T T T T
Move to long-term
200 mg/mL (3 mL)
0 °C, –5 °C, and –15 °C by placing
2–8 °C storage.
Formulation buffer T T T T
filled containers in a laboratory
bath containing a heat-transfer
T = tested; NT = not tested
f luid. After about 72 hours of
exposure to the target temperature,
Figure 4 (Case Study 2):  Summary results of %monomer and %HMW (high–molecular-weight
samples were moved to long-term
species) by SEC for MAb-B drug product at 200 mg/mL; following one cycle of actively controlled
freeze cycle (72-hour storage at 0 °C, –5 °C, or –15 °C), the product was stored at 2–8 °C for ≤12 storage at 2–8 °C for up to 12
months. Refer to Table 1 for study setup and sample description. months.
100 Stability of the MAb-B solutions
after 72 hours at 0 °C was assessed immediately after
after 72 hours at −5 °C subzero temperature treatment and
after 72 hours at −15 °C again after long-term storage by
98
general appearance, testing pH and
A280, and using SEC, capillary
Monomer (%) MAb-B

isoelectric focusing (cIEF),


96 ProteinSimple MFI, reverse-phase
chromatography (RP-HPLC), and
100
surface plasmon resonance (SPR)
Monomer (%) MAb-B

94 98 potency analysis. Structural


96 integrity of the MAb was also
determined by biophysical testing:
94
f luorescence spectroscopy and
92 92 circular dichroism. Figure 4
90 summarizes SEC stability results.
0 3 6 9 12
Storage Time at 2−8 °C (months) MAb-B drug product at a
90 concentration of 200 mg/mL
0 3 6 9 12
Storage Time at 2−8 °C (months) exhibited no significant changes in
monomer concentration or
aggregate profile when stored for up
directly result in aggregate However, those restrictions added to 12 months (one year) at 2–8 °C
formation through increased significant supply-chain risk after 72-hour incubation at the
interaction of partially unfolded because the project progressed into three subzero temperatures.
molecules with each other or with later-phase clinical development. The results from the analytical
other species upon thawing. That For example, according to internal and biophysical testing showed that
phenomenon can be enhanced if the shipping temperature logs (data not MAb-B quality and stability in the
frozen storage temperature is near shown), temperature excursions selected container–closure system
or above Tg′ of the formulation (25). outside 2–8 °C were typically in the were not affected by short-term
Stability studies revealed that range of –1 °C to –11 °C and tended exposure to temperatures between
MAb-B failed acceptance criteria to be on the lower end of that –15 °C and 0 °C. These data were
for aggregates when frozen at range. Thus, we needed to justify a used to justify expansion of the
–20 °C (>Tg′) for a month. A more practical temperature excursion limits for this project,
significant increase in subvisible excursion range. which reduced the risk of stock-outs
particles also was observed. Not A small-scale freeze–thaw and clinical delays.
only were MAb-B drug substance procedure — together with a long-
and product both formulated at very term stability study — was C ase Study 3
high concentration (200 mg/mL), implemented to discharge shipment Drug Substance Robustness, Freeze–
but also they used a buffer system and storage excursion risks of short- Thaw, and Long-Term Stability in
with no cryoprotectant. term exposure to subzero Small-Scale System Configuration
Temperature excursions below 2 °C temperatures >Tg′. Table 1 displays for a 100-mg/mL MAb-C:
thus were strictly prohibited for the details of the study design. Commercial-scale production of
early phase clinical supplies. MAb-B and a corresponding drug substance is no longer limited

BioProcess International 13(1) January 2015


Figure 5 (Case Study 3):  Summary results of Table 2 (Case Study 3):  Summary of a small-scale, passive freeze–thaw study for MAb-C using
%monomer by SEC for MAb-C drug substance disposable 5-mL EVA bags filled at 40% volume with samples representative of formulation robustness
after five cycles of passive freeze–thaw cycles Freeze–Thaw MAb-C MAb-C
from ≤–60 °C to 2–8 °C, followed by six-month Stress Sample Conditions Concentration pH
storage at 2–8 °C; refer to Table 2 for study Perform five a Low MAb concentration, low pH 80 mg/mL 5.8
setup and sample description. cycles of passive b Low MAb concentration, high pH 80 mg/mL 6.2
freezing and
thawing from c High MAb concentration, low pH 120 mg/mL 5.8
100
≤–60 °C to 2–8°C d High MAb concentration, high pH 120 mg/mL 6.2
Monomer (%) MAb-C

98 e Center point (target formulation) 100 mg/mL 6.0

96
Figure 6 (Case Study 4):  A small-scale, controlled freeze–thaw study using single-use 30-mL
Sample A
94 Sample B Celsius bags for MAb-C; solutions were aliquoted into each bag at approximately 66% fill volume.
Sample C The 120-mg/mL samples were subjected to three freeze–thaw cycles in which the rates of freezing
Sample D
92 Sample E and thawing were actively controlled (table below). Samples were either frozen in liquid nitrogen
(active fast freeze protocol, graph below) and then thawed in a water bath at ambient temperature
90
−1 0 1 2 3 4 5 6 7 (active fast thaw protocol), or frozen from 25 °C to −70 °C at a controlled rate of 0.1 °C/min (active
Storage Time (months) slow freeze protocol) and thawed from −70 °C to 25 °C at a controlled rate of 0.1 °C/min (active slow
thaw protocol) by using Celsius S3 system (photo below). After three freeze–thaw cycles, the bags
to rigid geometric containers such were placed at 2–8 °C and 25 °C for long-term storage up to one, three, and six months.
as bottles and carboys. Single-use Sample Set Freezing Rate Thawing Rate
bag systems made of f lexible f Controlled Fast Controlled Fast
polymeric materials offer an g Controlled Slow Controlled Slow
alternative. One such example is the h Controlled Fast Controlled Slow
Celsius FFT (f lexible freeze–thaw) i Controlled Slow Controlled Fast
bag system from Sartorius Stedim j Uncontrolled Uncontrolled
Biotech. The FFT bag uses Celsius
film, of which the product-contact (graph)  Controlled slow freeze-thaw temperature profile
component is ethylene vinyl acetate at 0.1 °C/min using the Celsius® S3 System
(EVA). 30
(photo)  A 30-mL
20
The Celsius bag was identified as
10
Celsius bag
Temperature (°C)

the preferred container for phase 3


0
containing
MAb-C BDS at
and commercial supply of MAb-C −10
120 mg/mL
−20
drug substance formulated at 100
−30 after being
mg/mL. Upon information review
−40 subjected to
and consultation with the vendor, −50
controlled
−60
fast-freezing
5-mL Flexboy bags from Sartorius −70
protocol using
Stedim Biotech were determined to
0 50 100
Time (hours)
150
liquid nitrogen
represent a suitable scale-down
model of commercial sizes in the
Celsius bag system. 2–8 °C storage chamber (passive month storage at 2–8 °C regardless
The goal of this study was to thawing protocol), each at about of sample type.
assess robustness of the drug- 12 hours duration. Results from this study confirm
substance formulation to freeze– MAb-C stability was assessed long-term stability of the MAb-C
thaw stress and subsequent storage after bags were subjected to five product within specified
at various conditions in the 5-mL passive freeze–thaw cycles from formulation robustness ranges
bags. Table 2 lists the two ≤–60 °C to 2–8 °C and after storage following five passive freeze–thaw
formulation robustness factors at ≤–60 °C, –40 °C, –20 °C, cycles. Consequently, no active
incorporated into this study design: 2–8 °C, and 25 °C for one, three, freeze–thaw control was
MAb concentration (±20% target) and six months. Samples were implemented at the drug-substance
and pH range (±0.2 units from analyzed for general appearance, manufacturing site. Instead, filled
target). Solutions of MAb-C were pH, and freezing-point osmolality, Celsius bags were frozen by placing
aliquoted into each bag at about and by microf luidic chip them in a standard ≤–60 °C freezer,
40% fill-volume capacity. Samples electrophoresis (Caliper Life and the substance was thawed at
were subjected to five freeze–thaw Sciences), SEC, cIEF, and SPR. 2–8 °C in a cold room.
cycles in which the rates of freezing Figure 5 displays SEC %monomer
and thawing were passively results, showing no significant C ase Study 4
controlled. Samples were frozen by change in the purity profile for Active and Passive Freeze–Thaw of
placing them horizontally in MAb-C drug substance after five MAb-C at 120 mg/mL in 30-mL
≤–60 °C storage chambers (passive passive freeze–thaw cycles from Celsius Bags: Combining actively
freezing protocol) and thawed in a ≤–60 °C to 2–8 °C followed by six- controlled freeze–thaw technology

January 2015 13(1) BioProcess International


Table 3 (Case Study 4):  SEC and cIEF results for MAb-C drug substance subjected to three cycles
of active freezing and thawing from –70 °C to 25 °C and stored up to six months at 2–8 °C; refer to lengths make freezing and thawing
Figure 6 for study setup and sample description. rates extremely difficult to match.
One approach we adopted at small
Size-Exclusion Chromatography Capillary Isoelectric Focusing
MAb-C scale was to show that freezing and
Sample Monomer HMW LMW Main Acidic Basic thawing rates do not significantly
Initial 97.5% 1.3% 1.2% 57.6% 34.8% 7.6% inf luence product quality in the
f 96.9% 1.9% 1.3% 57.1% 35.2% 7.7% selected formulations. Neither
g 96.9% 1.9% 1.3% 57.2% 35.4% 7.5%
freezing nor thawing rates (both
significant process parameters)
h 96.7% 2.0% 1.3% 56.2% 35.3% 8.5%
needed to be controlled during
i 96.8% 1.9% 1.3% 56.7% 35.9% 7.5% product handling.
j 96.8% 1.9% 1.3% 57.6% 35.0% 7.6% In all four case studies, freeze–
thaw cycles tested at small scale
mimicked the product-specific
with single-use bags can greatly samples stored at 2–8 °C for six system and process for potential
reduce overall cost of goods and months. application at large-scale
enhance operational f lexibility (26). The results of this study — production. For active control of
The Celsius S3 system from together with those described in freezing and thawing rates, we used
Sartorius Stedim Biotech is case study 3 above — indicate that laboratory approaches such as a
designed to reproduce active control active control of freezing and freeze dryer, liquid nitrogen, dry-
of large-scale freezing and thawing thawing rates during manufacture ice acetone slurry, and a water bath
conditions in small-scale EVA bags was not necessary for MAb-C. (in addition to specialized
(of 30 mL and 100 mL total Although such results may not be technologies such as the Celsius S3
volume) (8, 26). This study was observed for all MAbs in freeze–thaw system). These studies
designed to assess whether an development, they were not exemplified how small-scale freeze–
actively controlled freezing and unexpected here because the thaw approaches can be
thawing process using the Celsius formulation was specifically implemented in the
S3 system would provide an designed to withstand stresses that biopharmaceutical industry to
advantage over passive freezing and can be induced by freezing and confirm container–closure selection,
thawing for MAb-C. Resulting thawing. verify formulation robustness,
data were used to determine support temperature excursion
whether to purchase a controlled D emonstrating D ue D iligence limits, set process parameters, and
freeze–thaw system for the Freeze–thaw processes are routinely (most important) justify the need
manufacturing site. used to improve storage and for either active or passive freeze–
Figure 6 summarizes the study shipping stability in production and thaw procedures and associated
design and procedure. Single-use storage of biopharmaceutical equipment before expensive
30-mL Celsius bags were used. solutions. However, freezing and/or manufacturing campaigns begin.
Samples were subjected to three thawing processes can adversely Although our intent in these studies
freeze–thaw cycles with different affect protein product stability and was not to evaluate concentration
rates of freezing and thawing quality. For this reason, relevant effects, their results illustrate the
achieved using Celsius S3 system, a studies are needed to assess success of such designs across a
liquid nitrogen bath, and a water formulation robustness to freeze– wide range of concentrations
bath. GA, pH, freezing-point thaw stress and container–closure (0.2 mg/mL MAb-A in case study 1
osmolality, microf luidic chip compatibility with as well as to and 200 mg/mL MAb-B in case
electrophoresis, SEC, cIEF, SPR, define freezing and thawing study 2).
and PS80 testing were used to test parameters for large-scale Understanding the product
MAb-C stability under stresses of production. robustness to freeze–thaw stress can
extreme freeze–thaw rates followed Designing freeze–thaw studies at be achieved using carefully designed
by long-term storage (at 2–8 °C and conditions representative of stresses scale-down models for freezing and
25 °C for up to six months) in this expected at large-scale production thawing. Also, we propose that
small-scale system configuration. is a significant challenge requiring systematic small-scale designs
As Table 3 shows, no significant consideration of multiple factors similar to those discussed here
changes were observed in either the such as container–closure material could be adopted as predictive,
size (measured using SEC) or and size, fill volume, and freezing robust approaches and applied
charge (measured using cIEF) and thawing rates. Although small- across the biopharmaceutical
profiles of MAb-C after three and large-scale contact materials industry to assess the impact of
freeze–thaw cycles. A similar can be matched with relative ease, freeze–thaw process on product
observation was made for cycled the significantly different path quality and to support

BioProcess International 13(1) January 2015


manufacturing, storage, and Freeze-Concentration. BioPharm Int. 15(5) Reichert JM. Antibody-Based
2002: 22–34. Therapeutics to Watch in 2011. MAbs 3(1)
handling decisions.
14 Bhatnagar BS, Bogner RH, Pikal MJ. 2011: 76–99.
Protein Stability During Freezing: Karow AR, Bahrenburg S, Garidel P.
R eferences Separation of Stresses and Mechanisms of Buffer Capacity of Biologics — from Buffer
1 Ammann C. Stability Studies Needed
Prog. 29(2) 2013: 480–492. •
Protein Stabilization. Pharm. Dev. Technol. Salts to Buffering By Antibodies. Biotechnol.
to Define the Handling and Transport 12(5) 2007: 505–523.
Conditions of Sensitive Pharmaceutical or
15 Singh SK, et al. Large-Scale Freezing
Biotechnological Products. A APS Pharm.
of Biologics: A Practitioner’s Review, Part 2:
Sci. Tech. 12(4) 2011: 1264–1275.
Practical Advice. BioProcess Int. 7(10) 2009: Corresponding author Manasi Puri is a
2 Rathore N, Rajan RS. Current 34–42. scientist; Sorina Morar-Mitrica is a
Perspectives on Stability of Protein Drug
16 Kelley B. Industrialization of MAb senior scientific investigator; George
Products During Formulation, Fill and
Finish Operations. Biotechnol. Prog. 24(3)
Production Technology: The Bioprocessing Crotts is manager; and Douglas Nesta is
Industry at a Crossroads. MAbs 1(5) 2009: director of biopharmaceutical product
2008: 504–514.
440–449. sciences at GlaxoSmithKline, 709
3 Kolhe P, et al. Large-Scale Freezing
17 Chang BS, Kendrick BS, Carpenter Swedeland Road, King of Prussia, PA
of Biologics: Understanding Protein and
JF. Surface-Induced Denaturation of Proteins 19406; 1-610-270-5900; manasi.5.puri@
Solute Concentration Changes in a
During Freezing and Its Inhibition By gsk.com.
Cryovessel — Part 2. BioPharm Int. 23(7)
Surfactants. J. Pharm. Sci. 85(12) 1996: 1325–
2010: 40–49.
1330.
4 Kolhe P, et al. Large-Scale Freezing
18 Strambini GB, Gabellieri E. Proteins For electronic or printed reprints, contact
of Biologics: Understanding Protein and
in Frozen Solutions: Evidence of Ice-Induced Rhonda Brown of Foster Printing Service,
Solute Concentration Changes in a
Partial Unfolding. Biophysical. J. 70(2I) 1996: rhondab@fosterprinting.com, 1-866-879-9144
Cryovessel — Part 1. BioPharm Int. 23(6)
971–976. x194. Download personal-use–only PDFs
2010: 53–60.
19 Lashmar UT, Vanderburgh M, Little online at www.bioprocessintl.com.
5 Kolhe P, Amend E, Singh SK.
SJ. Bulk Freeze–Thawing of Macromolecules
Impact of Freezing on pH of Buffered
Effects of Cryoconcentration on Their
Solutions and Consequences for Monoclonal
Formulation and Stability. BioProcess Int. 5(6)
Antibody Aggregation. Biotechnol. Prog.
2007, 44–54.
26(3) 2010: 727–733.
20 Miller R, et al. Dynamics of Protein
6 Kolhe P, Badkar A. Protein and
and Mixed Protein/Surfactant Adsorption
Solute Distribution in Drug Substance
Layers at the Water–Fluid Interface. Adv.
Containers During Frozen Storage and
Colloid Interface Sci. 86(1) 2000: 39–82.
Post-Thawing: A Tool to Understand and
Define Freezing-Thawing Parameters in 21 Cao E, et al. Effect of Freezing and
Biotechnology Process Development. Thawing Rates on Denaturation of Proteins
Biotechnol. Prog. 27(2) 2011: 494–504. in Aqueous Solutions. Biotechnol. Bioeng.
82(6) 2003: 684–690.
7 Kueltzo LA, et al. Effects of Solution
Conditions, Processing Parameters, and 22 Yan L, Zhu Z. Development of
Container Materials on Aggregation of a Antibody-Based Therapeutics for Oncology
Monoclonal Antibody During Freeze– Indications. Drug Dev. Res. 67(9) 2006: 699–
Thawing. J. Pharm. Sci. 97(5) 2008: 1801– 728.
1812. 23 Heller MC, Carpenter JF, Randolph
8 Le Saout X, et al. Safe Freeze–Thaw TW. Protein Formulation and Lyophilization
of Protein Drug Products: A QbD Cycle Design: Prevention of Damage Due to
Approach. BioPharm Int. 23(12) 2010: Freeze-Concentration Induced Phase
28–43. Separation. Biotechnol. Bioeng. 63(2) 1999:
166–174.
9 Miller MA, et al. Frozen-State
Storage Stability of a Monoclonal Antibody: 24 Luo Q , et al. Chemical Modifications
Aggregation Is Impacted By Freezing Rate in Therapeutic Protein Aggregates Generated
and Solute Distribution. J. Pharm. Sci. Under Different Stress Conditions. J. Biol.
102(4) 2013: 1194–1208. Chem. 286(28) 2011: 25134–25144.
10 Rodrigues MA, et al. Effect of 25 Singh SK, et al. Best Practices for the
Freezing Rate and Dendritic Ice Formation Formulation and Manufacturing of Biotech
on Concentration Profiles of Proteins Drug Products. BioPharm Int. 22(6) 2009:
Frozen in Cylindrical Vessels. J. Pharm. Sci. 32–48.
100(4) 2011: 1316–1329. 26 Voute N et al. Disposable Technology
11 Singh SK, et al. Large-Scale Freezing for Controlled Freeze–thaw of
of Biologics. BioProcess Int. 7(9) 2009: Biopharmaceuticals at Manufacturing Scale.
32–44. BioProcess Int. 2(9) 2004: S40–S43.
12 Singh SK, et al. Frozen State Storage
Instability of a Monoclonal Antibody:
Aggregation As a Consequence of Trehalose Further R eading
Crystallization and Protein Unfolding. Elvin JC, Couston RG, van der Walle
Pharm. Res. 28(4) 2011: 873–885. CF. Therapeutic Antibodies: Market
Considerations, Disease Targets and
13 Webb SD, et al. Freezing Bulk-Scale
Bioprocessing. Int. J. Pharm. 440(1) 2013:
Biopharmaceuticals Using Common
83–98.
Techniques — and the Magnitude of

January 2015 13(1) BioProcess International


S i n g l e -U s e PRODUCTION

A Novel Seed-Train Process


Using High-Density Cell Banking, a Disposable Bioreactor,
and Perfusion Technologies

by Benjamin Wright, Mike Bruninghaus, Mike Vrabel, Jason Walther, Neha Shah,
Seul-A Bae, Timothy Johnson, Jin Yin, Weichang Zhou, and Konstantin Konstantinov

A
typical cell culture process Figure 1:  Comparing a conventional with an improved seed-train process for inoculation of a
begins with thawing of a 500-L perfusion bioreactor; the new process uses high-density (HD) cell banking, single-use
cryopreserved cell-bank vial, technology, and high-density perfusion at the n – 1 stage to allow for high-density inoculation in
the production bioreactor. HD cell banking and disposables improve operational success by
followed by successive reducing seed-train complexity and contamination potential. HD perfusion at the n – 1 stage allows
expansions into larger culture vessels for HD inoculation of the production bioreactor, reducing the time to steady-state cell density by
such as shake flasks, spinners, Wave 4–5 days and increasing productivity by 10% for a 50-day run.
bags, and stirred bioreactors (1). When
culture volume and cell density meet Conventional Seed Train
predetermined criteria, the culture is
transferred to a production bioreactor
Standard WCB
in which cells continue to grow and 1 mL @ 2 × 107 cells/mL
express product. This approach
presents several challenges.
Shake flasks or spinners used in the
initial stages require manual 50-L SS batch 500-L SS batch
125-mL to 10-L glass spinners (n − 1) + ATF production
manipulations inside a laminar flow
hood, making them vulnerable to Improved Seed Train
contamination and operator error.
Also, because a conventional HD WCB
cryopreserved cell-bank vial contains 4.5 mL @ 10 × 107 cells/mL
low cell numbers, the seed-train
process is time consuming. It may take
even longer and become more complex
when production bioreactors are very 50-L SUB + ATF 500-L SUB + ATF
large because additional large-volume 2-L and 20-L Wave bioreactors
(n − 1) production
culture vessels are required to support
scale up to a production bioreactor.
Finally, production bioreactors are 450 million viable cells/vial to directly facility flexibility, time savings in
usually inoculated at a density <0.5 × inoculate a 20-L Wave bag (2). That setup, cleaning, and sterilization, and
106 viable cells/mL, so a necessary study revealed that HD cell-bank use reduced capital investment (4–6).
growth phase of 5–10 days offers little could eliminate several intermediate Disposable Wave bioreactors from GE
practical value because cell shake-flask expansion steps and reduce Healthcare Life Sciences are routinely
concentrations (and thus initial product process times by up to nine days. used in seed-train expansion processes
yield) during that period will be low. Heidemann et al. combined HD cell because they decrease contamination
Previous efforts have shown that banks with a single seed bioreactor risk (typically requiring no
high-density (HD) cell banking can be capable of operating at several working manipulations in laminar-flow hoods).
an effective means to reduce the volumes to reduce seed-train expansion Also, because such bioreactors are
number of seed-train steps required duration by 60–70% (3). disposable, no resources are needed for
and also improve operational success in Single-use technology offers assembly, cleaning, or sterilization (7).
seed-train processes. Tao et al. reported numerous advantages over stainless Industry interest in perfusion cell-
using HD cell banks containing steel systems, including enhanced culture processes has been increasing

M arch 2015 13(3)s BioProcess International


Figure 2:  Viable cell density (VCD) profiles for a complete three-stage Figure 3:  Viable cell density (VCD) as a function of capacitance for n – 1
seed train bioreactor run
120 120
Viable Cell Density (106 cells/mL)

Viable Cell Density (106 cells/mL)


100 100

80 80

60 60

40 40

20 20
Actual
Predicted
0 0
0 2 4 6 8 10 12 14 16 18 20 22 24 26 0 50 100 150 200
Culture Time (days) Capacitance (pF)

(8). Several companies have allowing for direct inoculation into a a 20-L Wave CellBag bioreactor at a
successfully used perfusion bioreactors 2-L Wave bioreactor. Replacing 7.5-L working volume. A model 20/50
in manufacturing commercial spinners (125 mL to 10 L in the figure) EHTD Wave bioreactor system (GE
therapeutics (9). Perfusion processes with Wave bioreactors (2 L and 20 L) Healthcare Life Sciences) served as the
can reach high cell densities in significantly reduces the number of rocking platform for both the 2-L and
relatively small bioreactor volumes (10) required manipulations in a laminar 20-L bags, maintaining cultures at a
and remove unstable product from flow hood. That improves operational temperature of 37 °C, a 16-rpm
cultures more quickly (11) than batch success by allowing operation under a rocking speed, and a 7° rocking angle.
and fed-batch processes. Because of “closed” system. Use of ATF perfusion A 20% O2 and 5% CO2 gas mixture
those and other advantages, perfusion technology at the n – 1 stage (50-L flowed into the head space at a rate of
is often considered in the context of bioreactor with ATF perfusion device 0.25 slpm.
production bioreactors rather than as in the figure) also enables cell densities Seed-Train n – 1 Perfusion Culture:
an element of seed expansion. to be ≥50 × 106 viable cells/mL. That We used a 15-L glass bioreactor
Whitaker (12) and Heidemann (3) allows for an inoculation density of 5 × (Broadley-James Corporation)
briefly mentioned using perfusion in 106 viable cells/mL in the 500-L equipped with an ATF4 perfusion
seed expansion, but they did not discuss production bioreactor rather than 0.5 × device (Refine Technology) to mimic
its potential to improve terminal seed- 106 viable cells/mL in the conventional the n – 1 seed-train stage (50-L
train stages, to reduce bioreactor size or cell culture process. Higher seeding bioreactor in Figure 1). A 20-µm
steps, or to reduce the growth-phase density helps reduce the 500-L sintered sparger added oxygen to
duration for production bioreactors. production bioreactor growth phase by control dissolved oxygen (DO) at 40%,
Pohlscheidt (13) used a 3,000-L n – 1 about five days, saving manufacturing and a 1-mm drilled hole sparger added
perfusion bioreactor with an inclined plant time for a 500-L bioreactor nitrogren to control the dissolved CO2
cell settler to achieve n – 1 cell densities operating for 50 days. level <120-mmHg pressure. As
up to 15.8 × 106 viable cells/mL, needed, we added 0.5 M sodium
decreasing cultivation times by 20% in Materials and Methods carbonate (Na 2CO3) to the culture for
a fed-batch production process. Cell Line and Media: All experiments maintaining pH >6.85 and a 10%
However, higher cell densities are hard used a commercially available FoamAway antifoam solution (Life
to obtain with inclined settler devices chemically defined medium (Life Technologies) to control the foam level.
because of perfusion rate limitations Technologies) supplemented with We seeded a 15-L bioreactor at 10-L
and long residence times for cells in 4 mM glutamine (Sigma-Aldrich). The working volume with 0.5 × 106 viable
suboptimal conditions (10). Chinese hamster ovary (CHO) cell line cells/mL and operated the culture in
Here we describe a novel seed-train is proprietary to Genzyme and produces batch mode until day 2, when perfusion
process using HD cell banking, a recombinant human enzyme. began. Initially, an online capacitance
disposable Wave bioreactors, and Batch Seed-Train Culture in 2-L and sensor (Aber Instruments) and a DeltaV
Refine Technology’s Alternating 20-L Bags: We thawed an HD cell- programmable logic controller
Tangential Flow (ATF) perfusion cell bank vial (10 × 107 viable cells/mL, (Emerson Process Management)
culture technologies. Figure 1 compares 4.5 mL/vial) into a 2-L Wave CellBag controlled the cell-specific perfusion
an example of this process for a 500-L bioreactor (GE Healthcare Life rate (CSPR) at 0.2 nL/cell/day. We
production perfusion bioreactor with a Sciences) at a 1-L working volume. capped the perfusion rate at four
conventional seed-train process. Using When viable cell density (VCD) in the bioreactor working volumes per day
the HD cell bank approach eliminates 2-L Wave bag reached 3.0 × 106 viable (RV/day) after the VCD reached 20 ×
two intermediate spinner steps by cells/mL, we expanded the culture into 106 viable cells/mL.

BioProcess International 13(3)s M arch 2015


50-mL Spin Tube Batch Refeed Model for Inoculation 9, which is the density required to inoculate a 500-L bioreactor
Density Evaluation: We removed aliquots of the culture at 5 × 106 viable cells/mL (from a 50-L n – 1 bioreactor).
from the n – 1 seed-train bioreactor when its cell density Initiated on day 2, n – 1 bioreactor perfusion was
reached 25 × 106, 50 × 106, and 100 × 106 viable cells/mL controlled to maintain a CSPR of 0.2 nL/cell/day by an
and subsequently inoculated those samples into 50-mL spin online capacitance probe to automatically increase the feed
tubes (TPP Techno Plastic Products) at three different rate as cell density rose. Success of such a perfusion-rate
inoculation densities in triplicate: 0.5 × 106, 2.5 × 106, and control strategy depends on the capacitance probe’s ability to
5.0 × 106 viable cells/mL in a working volume of 10 mL. accurately estimate VCD. Here, we used the strategy only
Refeeds were performed once daily because continuous until day 7 because the perfusion rate was capped at
perfusion could not be performed at this small scale. 4 RV/day, but the probe was able to accurately estimate VCD
Starting on day 1 we refed the cultures by removing the throughout the run to day 12, when 110 × 106 viable cells/mL
spin tubes from their incubator, spinning cells down at was reached (Figure 3). Using control strategy during the cell
1,100 rpm for five minutes, removing the supernatant, and
then adding fresh media to resuspend the cells. That refeed Figure 4:  Viable cell density (VCD, solid line) and viability (dashed line) for
strategy is designed to provide the same CSPR across spin tubes inoculated at (a) 0.5 × 106 viable cells/mL, (b) 2.5 × 106 viable
cells/mL, and (c) 5.0 × 106 viable cells/mL inoculation cell density; n – 1
different inoculation density conditions. A Multitron II
stage final cell density is 25 × 106 (blue trends), 50 × 106 viable cells/mL
shaking incubator (HT Infors) maintained all spin tubes at (green trends), 100 × 106 cells/mL (red trends), respectively. Shaded areas
a temperature of 37 °C with a rocking rate of 160 rpm, a represent ±1 standard deviation (n = 3).
rocking angle of 45°, relative humidity of 80%, and CO2
120 100
concentration of 5%.

Viable Cell Density (106 cells/mL)


Production Bioreactor: To mimic the 500-L production 100 A 80
bioreactor in Figure 1, we operated 15-L bioreactors
(Broadley-James Corporation) with 10-L working volumes 80

Viability (%)
in perfusion mode using an ATF4 perfusion device (Refine 60
Technology) with 0.2-µm polyethersulfone filters. A 60
20-µm sintered sparger added oxygen to control DO at 40
40
40%, and a 1-mm drilled hole sparger added nitrogen to
maintain dissolved CO2 level <120-mmHg pressure. We 20 20
maintained pH >6.85 by adding 0.5 M Na 2CO3 and
controlled foam levels by adding 10% FoamAway antifoam 0 0
solution through a peristaltic pump. 0 1 2 3 4 5 6 7 8
One set of production bioreactors was inoculated at a low Culture Time (days)
cell density (0.5 × 106 viable cells/mL) from a seed vessel 120 100
Viable Cell Density (106 cells/mL)

operated in batch mode. Perfusion started on day 1 at


100
0.5 RV/day and increased daily by 0.5 RV/day until reaching B 80
a 2 RV/day perfusion rate. Another set of production reactors 80

Viability (%)
was inoculated at a high cell density (5.0 × 106 viable 60
cells/mL) from an n – 1 15-L perfusion seed vessel when its 60
density reached 50 × 106 viable cells/mL. Because of that 40
high inoculation density, perfusion started at 2 RV/day 40
immediately after inoculation. For all production bioreactors,
20 20
an online capacitance sensor and bleed pump controlled the
VCD at 40 × 106 viable cells/mL. All production bioreactors 0
operated for 50 days. 0
0 1 2 3 4 5
Analytical Methods: We determined VCD using a Culture Time (days)
ViCell XR cell viability analyzer (Beckman Coulter), 120 100
Viable Cell Density (106 cells/mL)

measured pH and pCO2 off-line using a RAPIDLab blood


gas analyzer (Siemens), and quantified protein productivity 100
C 80
with a proprietary photometric enzymatic activity assay.
80
Viability (%)

60
Results and Discussion 60
HD Cell Culture Using ATF Perfusion in the n – 1 Bioreactor:
40
Figure 2 shows the VCD growth profile of our seed-train 40
process including the two Wave bioreactor stages and the
n – 1 stage. The 2-L and 20-L Wave stages lasted eight and 20 20
five days, respectively. The n – 1 bioreactor operated for
0
12 days, reaching a final VCD of 110 × 106 viable cells/mL 0 1 2 3 4 5
0
and >98% cell viability. VCD reached 50 × 106 cells/mL on day Culture Time (days)

M arch 2015 13(3)s BioProcess International


Figure 5:  Viable cell density (VCD) profiles for 10-L production bioreactors Figure 6:  Cumulative product titer as a function of integral viable cell
inoculated at 0.5 × 106 viable cells/mL from an n – 1 bioreactor at 2 concentration for 10-L production bioreactors inoculated at
.5 × 106 viable cells/mL (n = 2) compared with 10-L production bioreactors 0.5 × 106 viable cells/mL from an n – 1 bioreactor at
inoculated at 5.0 × 106 viable cells/mL from an n – 1 bioreactor at 2.5 × 106 viable cells/mL (red points) compared with 10-L production
50 × 106 viable cells/mL (n = 2); dashed line represents the target viable cell bioreactors inoculated at 5.0 × 106 viable cells/mL from an n – 1 bioreactor
density for steady-state operation, and shaded areas represent the range. at 50 × 106 viable cells/mL (green points); error bars represent the range.
3,000,000

Cumulative Product Acitivity (U/L)


70 .
Viable Cell Density (106 cells/mL)

Low-density n − 1 + inoculation
60 2,500,000 High-density n − 1 + inoculation
(n = 2)
50 2,000,000
40
1,500,000
30
1,000,000
20 (n = 2)
Low-density n − 1 + inoculation 500,000
10 High-density n − 1 + inoculation
Target steady-state cell density
0 0
0 10 20 30 40 50 0 500 1,000 1,500 2,000
Culture Time (days) IVCC (106 cells/day/mL)

growth phase allows much more cultures only up to 20 × 106 viable phase duration by four to five days,
accurate control of CSPR, which can be cells/mL to match their CSPR to that increasing productivity by 10% for a
advantageous. Previous reports have of the perfusion bioreactor model. We 50-day run. Additionally, we have
shown that growth and productivity observed no growth differences among shown the n – 1 bioreactor could reach
can be affected by a CSPR difference of the n – 1 density conditions for all 100 × 106 viable cells/mL, theoretically
0.05–0.10 nL/cell/day (14). Our strategy three inoculation densities tested. But inoculating a 500-L bioreactor at 10 ×
also reduces the risk of incorrect or the 100 × 106 viable cells/mL condition 106 viable cells/mL, which would
missed set-point changes because had a noticeably lower starting viability reduce the growth phase duration by
perfusion rate is automatically increased at all inoculation densities than at the an additional five to six days.
as the culture grows. In a stepwise other n – 1 densities. Alternatively, our seed-train process
approach, by contrast, operators must To study the effects of n – 1 density can inoculate a production bioreactor
adjust the rate manually (15). and inoculation density on production operated in either batch or fed-batch
Evaluating the Effects on Cell Growth bioreactor cell growth and productivity, mode. For a 21-day fed-batch cell
of n – 1 Split Density and Production we inoculated bioreactors at 5.0 × culture process using a 500-L bioreactor,
Bioreactor Inoculation Density: Using a 106 viable cells/mL from an n – 1 an inoculation density of 5.0 × 106 viable
small-scale spin tube model, we bioreactor at 50 × 106 viable cells/mL cells/mL could reduce the production
determined how n – 1 cell density and compared those with bioreactors bioreactor duration by 25%. Overall,
affected cell growth in the production inoculated at 0.5 × 106 viable cells/mL that could allow for an additional five to
bioreactor. We removed culture samples from an n – 1 bioreactor at 2.5 × six batches per year, increasing
from an n – 1 bioreactor when cell 106 viable cells/mL. Figure 5 illustrates manufacturing productivity by 25–30%.
densities were 25 × 106 viable cells/mL, comparable cell growth for both In most cases, batch and fed-batch
50 × 106 viable cells/mL, and 100 × conditions. Cell viability remained processes use production bioreactors that
106 viable cells/mL, respectively. Then >90% for both inoculation conditions are much larger than those used for
we split those into three aliquots diluted throughout the 50-day run. Figure 6 perfusion processes, which requires
to 0.5 × 106, 2.5 × 106, or 5 × 106 viable plots cumulative productivity against several seed-train stages in large-scale
cells/mL with fresh media for integrated VCD and indicates a similar stainless steel bioreactors. Figure 7
inoculation in triplicate into spin tubes. specific production rate between compares a conventional seed-train
Daily refeeds began on day 1 at rates conditions. Most notably, the steady- process used to inoculate a 10,000-L
adjusted based on the inoculation state cell density of 40 × bioreactor with another process using a
density of the spin tubes. That allowed 106 viable cells/mL (Figure 5) and the 50-L n – 1 bioreactor using an ATF
each inoculation density condition to required titer for purification were perfusion device. The 50-L n – 1
experience the same CSPR at reached four to five days sooner for the bioreactor at 50–100 × 106 viable
corresponding cell densities. highest inoculation density condition. cells/mL can inoculate a 10,000-L
Figure 4 shows cell count and Economic and Operational bioreactor at 0.25-0.50 × 106 viable
viability profiles plotted to compare the Advantages: Our seed-train process cells/mL, thus eliminating two
n – 1 density conditions at each has both economic and operational intermediate seed-train stages. The
inoculation density. Because the advantages. For example, inoculating a benefit here is an increase in facility
maximum perfusion rate in the spin- 500-L perfusion bioreactor at 5.0 × capacity use because no space and
tube model is 1 RV/day, we grew these 106 viable cells/mL reduces the growth utilities are required for 250-L n – 2 and

BioProcess International 13(3)s M arch 2015


Figure 7:  Comparing a conventional seed-train process to the future seed-train process for 8 Langer ES. Trends in Perfusion
inoculation of a 10,000-L batch or fed-batch bioreactor; the future seed-train process uses high- Bioreactors. BioProcess Int. 9(10) 2011: 18–22.
density cell banking, disposables, and high-density perfusion at the n – 1 stage to eliminate two 9 Chu L, Robinson DK. Industrial
large-volume seed-train stages. Choices for Protein Production By Large-
Scale Cell Culture. Curr. Opin. Biotechnol.
12, 2001: 180–187.
Conventional Seed Train
10 Voisard D, et al. Potential of Cell
Retention Techniques for Large-Scale High-
Density Perfusion Culture of Suspended
Standard WCB
Mammalian Cells. Biotechnol. Bioeng. 82(7) 2003:
1 mL @ 2 × 107 cells/mL
751–765.
11 Kadouri A, Spier RE. Some Myths and
Message Concerning the Batch and Continuous
10,000-L SS batch Culture of Animal Cells. Cytotechnol. 24, 1997:
50-L SS batch 250-L batch 2,500-L batch
125-mL to 10-L (n − 1) production 89–98.
(n − 3) (n − 2)
glass spinners 12 Whitaker SC, Francis R, Siegel RC.
Validation of a Continuously Perfused Cell
Improved Seed Train Culture Process for Production of Monoclonal
HD WCB Antibody. ACS Symp Ser. 698, 1998: 28–43.
4.5 mL @ 10 × 107 cells/mL 13 Pohlscheidt M, et al. Optimizing Capacity
Utilization By Large Scale 3,000-L Perfusion in
Seed Train Bioreactors. Biotechnol. Progr. 29(1)
2013: 222–229.
14 Dowd JE, et al. Optimization and Control
2-L and 20-L Wave bioreactors 50-L SUB + ATF 500-L SUB + ATF of Perfusion Cultures Using a Viable Cell Probe
(n − 1) production
and Cell Specific Perfusion Rates. Cytotechnol.
42(1) 2003: 35–45.
15 Kurkela R, Fraune E, Vihko P. Pilot-Scale
2,500-L n – 1 bioreactors. Additional in several ways: An inoculation
Production of Murine Monoclonal Antibodies in
large savings could be realized in density of 10 × 10 6 viable cells/mL Agitated, Ceramic-Matrix or Hollow-Fiber Cell
up-front capital expenditures. could be obtained in a 500-L Culture Systems. BioTechniques 15, 1993: 674–683.
bioreactor. For batch or fed-batch
A Simplified Seed Train processes that require larger Further Reading
We have successfully demonstrated a production bioreactors (e.g., 10,000- Abu-Absi SF, et al. Defining Process Design
novel seed-train cell culture process L), one or two stages can be Space for Monoclonal Antibody Cell Culture.
involving HD cell banking, single-use eliminated from the expansion Biotechnol. Bioeng. 106(6) 2010: 894–905.
technology, and perfusion at the n – 1 process by using a single 50-L Konstantinov KB, et al. The ‘‘Push-to-Low’’
Approach for Optimization of High-Density
bioreactor stage. This new procedure perfusion bioreactor at the n – 1
Perfusion Cultures of Animal Cells. Adv. Biochem.
reduces the seed-train process stage. Eng. Biotechnol. 101, 2006: 75–98.
complexity by decreasing the number Woodside SM, Bowen BD, Piret JM.
of small-scale expansion stages and R eferences Mammalian Cell Retention Devices for Stirred
1 Li F, et al. Current Therapeutic
eliminating the need for labor-intensive
175. •
Perfusion Bioreactors. Cytotechnol. 28(1) 1998: 163–
Antibody Production and Process
operations such as cleaning, assembly, Optimization. Bioprocessing J. September–
and sterilization. Manufacturing October 2005: 1–8.
operations can rely on the same general 2 Tao Y, et al. Development and Corresponding author Benjamin Wright is a
seed-train process with a 50-L SUB at Implementation of a Perfusion-Based High process engineer, Neha Shah is a staff scientist
the n – 1 stage and a 500-L SUB at the Cell Density Cell Banking Process. I, Jin Yin is associate director, Mike Vrabel is a
Biotechnol. Progr. 27(3) 2011: 824–829. process engineer, Jason Walther is a staff
production bioreactor stage.
3 Heidemann R, et al. A New Seed- scientist II, Seul-A Bae is a process engineer,
Use of ATF perfusion at the n – 1 Train Expansion Method for Recombinant and Konstantin Konstantinov is vice
stage can help cultures achieve viable Mammalian Cell Lines. Cytotechnol. 38(1–3) president, all in late-stage process
cell densities ≤100 × 10 6 viable cells/ 2002: 99–108. development; Mike Bruninghaus is a
mL in 12 days without 4 Smelko JP, et al. Performance of High manufacturing specialist supervisor in
compromising culture growth Intensity Fed-Batch Mammalian Cell upstream purification operations; and
characteristics after further Cultures in Disposable Bioreactor Systems. Timothy Johnson is director of special projects
Biotechnol. Progr. 27(5) 2011: 1358–1364.
expansion steps. Based on our work, and strategic support at Genzyme, a Sanofi
5 Papavasileiou V, Siletti C, Petrides D.
a 500-L production bioreactor can Systematic Evaluation of Single-Use Systems
Company, 200 Staples Drive, Framingham, MA
be inoculated at a cell density of 01702; 1-508-271-2080; benjamin.wright@
Using Process Simulation Tools. BioPharm
5 × 10 6 viable cells/mL to reduce the Int. 21, March–April 2011: S16–S29.
genzyme.com. Weichang Zhou is vice
time to steady-state cell density by president of biologics process development at
6 Monge M, Sinclair A. Disposables
WuXi AppTec.
four to five days and provide a 10% Cost Contributions: A Sensitivity Analysis.
increase in the overall productivity BioPharm Int. 22, April 2009: 24–29.
of a 50-day run. This n – 1 7 Singh V. Disposable Bioreactor for
Cell Culture Using Wave Induced Agitation.
perfusion technology can be adapted Cytotechnol. 30, 1999: 149–158.

M arch 2015 13(3)s BioProcess International


B i o P r o c e s s TECHNICAL

Hamster Phospholipase
B-Like 2 (PLBL2)
A Host-Cell Protein Impurity in Therapeutic Monoclonal
Antibodies Derived from Chinese Hamster Ovary Cells

Martin Vanderlaan, Wendy Sandoval, Peter Liu, Julie Nishihara, George Tsui, Margaret Lin,
Feny Gunawan, Sara Parker, Robert Ming Wong, Justin Low, Xiangdan Wang, Jihong Yang,
Karthik Veeravalli, Patrick McKay, Chris Yu, Lori O’Connell, Benjamin Tran, Rajesh Vij, Chris
Fong, Kathleen Francissen, Judith Zhu-Shimoni, Valerie Quarmby, and Denise Krawitz

A
ll recombinant protein number of anti-HCP antibodies in the
biotherapeutics must be tested for immunoassay reagents will be directed
the presence of residual host-cell against each particular protein impurity.
protein (HCP) impurities (1–3). Moreover, the assay results are
The most common analytical method for immunologically weighted: Highly
doing so is a polyclonal sandwich immunogenic HCPs will be detected
immunoassay. Polyclonal anti-HCP more readily than non- or weakly
antibodies are selected to recognize the immunogenic HCPs.
broadest population of HCPs possible. During biopharmaceutical production,
The immunogen and analytical standard a single HCP can copurify with a
are produced from a blank-run product through the downstream
fermentation that mimics the production process. By contrast, the HCP
run but lacks the specific biotherapeutic immunogen and assay standard contain
protein. Because of the large number of the thousands of host proteins that could PLBL2 models from PSI, the online protein
impurities present in harvested cell- be final-product impurities. Antibodies model portal WWW.PROTEINMODELPORTAL.ORG
culture fluid (HCCF) that might used in such assays often are affinity
copurify with the protein of interest, an purified against the same diverse mixture it. Likewise, if a copurifying impurity is
HCP immunoassay must detect a broad of proteins as those used in the highly immunogenic, the assay could
spectrum of proteins. Thus, a limited immunogen. So the relevance of the overquantify its value. For these reasons,
antibody population and assay standard orthogonal analytical methods often are
to a particular product-copurifying used to ensure product purity (3).
Product Focus:  Monoclonal
impurity might not be optimal. That As noted in the earliest publications
antibodies (CHO derived)
disconnect makes enzyme-linked on this topic (4), one property of HCP
Process Focus:  Production and immunosorbent assays (ELISAs) only ELISAs is that a particular test sample
downstream processing semiquantitative for HCPs. may not dilute in parallel with the
Typically, a single HCP numerical analytical HCP standard used. In that
Who Should Read:  Process/ value is reported for such assay results, paper, nonlinear sample dilution was
product development, analytical, and representing the ratio of HCPs (ng) to ascribed to “antigen excess”: a single or
manufacturing personnel product (mg). That value can reflect a limited number of copurifying HCP
single protein or a collection of several impurities saturating available antibodies
Keywords:  HCP ELISAs, copurifying
such impurities. Furthermore, if a to that particular protein. As a sample is
host impurities, chromatography,
potentially significant copurifying diluted, the measured dilution-corrected
in-process pools, assay nonlinearity
impurity is nonimmunogenic (or weakly impurity level rises. As the authors note,
Level:  Advanced immunogenic), then the assay could miss for cases in which dilution-corrected

BioProcess International 13(4) A pril 2015


values eventually plateau, the HCP node cells (6). For further development, directly to the filters and mixed with a
antigen has been diluted sufficiently to we selected a pair of MAbs that were vortex mixer before sample recovery. The
fall within the linear range of the assay suitable for generating a sandwich entire retentate was transferred to a
for that particular HCP, so the plateau ELISA. Transient transfection cultures single lane on an SDS polyacrylamide
value is reported as an estimate of that of CHO cells produced antibodies with gel electrophoresis (SDS-PAGE) gel.
impurity level. When no plateau is cloned mouse immunoglobulin (IgG) After resolving that lane by
reached, the value from the most dilute genes from the hybridomas, and we electrophoresis, we stained the results
sample is reported, which might be only affinity purified those antibodies using either with SYPRO Ruby fluorescent
a minimum estimate of the HCP level. protein A chromatography. We used one protein dye or SimplyBlue Safe stain
In theory, if different products show member of the antibody pair as a coating (both from Life Technologies) and rinsed
nonlinear dilution behaviors in an HCP antibody and biotinylated the other for with water before imaging.
ELISA, each product might have its own use as a detection antibody. As described elsewhere (10), we
unique copurifying HCP antigen in For the PLBL2 polyclonal antibody- excised gel bands covering either the
excess. However, here we report that the based assay, we immunized rabbits with entire 30–70 kDa region or discrete,
same HCP — phospholipase B-like 2 the CHO-derived PLBL2. The visible bands within that range, then
(PLBL2) — copurifies with multiple resulting antibodies were affinity washed those in 25 mM ammonium
Chinese hamster ovary (CHO)-produced purified over a PLBL2 column made by bicarbonate (Sigma) containing 5%
antibody therapeutics. In all cases, this coupling PLBL2 to activated glycerol- acetonitrile (Burdick and Jackson),
impurity manifests as a nonlinear CPG. We either used those purified followed by 25 mM ammonium
dilution in our CHO protein (CHOP) antibodies to coat microplates or bicarbonate in 100 μL of 50:50
ELISA. conjugated them to horseradish acetonitrile:water for 20 minutes before
peroxidase (HRP) for use as detection 100% acetonitrile. The in-gel trypsin
Materials and Methods antibodies in the sandwich ELISA. digestion (0.2 μg, Promega) in 25 mM
In-Process Pool Samples: All in-process Using methods that are well- ammonium bicarbonate at pH 8 was
pool samples came from therapeutic described in literature (7), we formatted a incubated overnight at 37 °C, then
humanized monoclonal antibodies platform CHOP assay as a sandwich arrested by addition of 5 μL of 1% formic
(MAbs) currently or formerly in ELISA. It’s based on direct conjugation acid. We extracted peptides from the gel
development at Genentech (a member of of HRP to the detection antibody, with slices using 0.1% formic acid containing
the Roche group). We named our detection using tetramethylbenzidine 5% acetonitrile (50 μL) and finally with
samples accordingly: MAb 1 served as a (TMB, formulated as SureBlue Reserve 50 μL of pure acetonitrile. Extractions
negative control, having no detectable from KPL) as a substrate. For this assay, were pooled, dried completely, and
CHOP as measured by multiple the biotinylated second antibody was reconstituted in a volume of 10 μL with
analytical methods; MAb 2 had extreme detected by streptavidin-HRP followed 0.1% formic acid.
dilution-nonlinearity in the platform by washing and TMB substrate. We injected half the total sample
CHOP ELISA and was investigated Purification and Identification of volumes (5 µL) with an autosampler onto
further to identify the cause of that PLBL2 As an Impurity in MAb Products: a 75-μm × 150-mm Waters column at a
nonlinearity (5). Other MAbs had We purified PLBL2 from final MAb 2 1-μL/min flow rate using a Waters
varying levels of nonlinear CHOP product by first loading a 1-mg sample of nanoAcquity ultraperformance liquid
dilution. product in PBS with 0.05% Tween 20 chromatography (UPLC) system. It
Development of Antibodies and mobile phase onto a 1.6- to 2.0-mL applied a gradient from 98% Solvent A
ELISAs for CHOP and PLBL2: For the CHT-1 ceramic hydroxyapatite column (water + 0.1% formic acid) to 80%
CHOP assay, we concentrated HCCF from Bio-Rad Laboratories (8, 9). Solvent B (acetonitrile + 0.08% formic
from a culture of nontransfected CHO Continued washing with PBS-Tween acid) over a 40-minute period. We
cells by ultrafiltration, then diafiltered it preceded a step elution with 0.5 M analyzed the samples on-line using
against phosphate-buffered saline (PBS). sodium phosphate to remove the nanospray ionization into a hybrid LTQ-
We used the resulting mixture of CHO antibody from the column. We collected Orbitrap XL mass spectrometer
proteins as an immunogen to generate fractions throughout the chromatography (Thermo Scientific). Data were collected
anti-CHOP antibodies and an analytical process and assayed them with the in data-dependent mode with the parent
standard for our assay. Antisera from CHOP ELISA, pooling CHOP-positive ion analyzed by Fourier-transform mass
immunized goats were purified over an fractions for further analysis. spectrometry (FTMS), and we selected
affinity column made by immobilizing Because the pooled fractions had low the eight most abundant ions for
the immunogen on activated glycerol- concentrations of a potentially sticky fragmentation and analysis with the
controlled pore glass (glyceryl-CPG) protein, we took care to minimize losses LTQ instrument. Using the Mascot
from EMD Millipore. on surfaces and filters. Pools were search algorithm (Matrix Sciences), we
For the PLBL2 MAb-based assay, we concentrated to 20 μL using Amicon analyzed tandem mass spectrometric
immunized mice with CHO-derived spin filters prewet with sodium-dodecyle data against the Uniprot database
PLBL2 (see below) and made sulfate (SDS). Then we added Laemmli including peptide sequences from all
hybridomas from their spleen and lymph sample buffer containing 2% SDS mammals.

A pril 2015 13(4) BioProcess International


Gene Sequence Primers enzymatic cleavage of three rhuMAbs in
PBS at an enzyme:antibody ratio of
pRK-F 5′-GGTGACACTATAGAATAACATCCACTTTGCC-3′
1:200. We incubated the mixture
pRK-R 5′-GTGAAATTTGTGATGCTATTGCTTTATTTGTAACC-3′ overnight at ambient temperature before
PLBL2-mid-F 5′-CGCATCATCAAGAAGTACCAGCTGCAG-3′ affinity purification across MAbSelect
PLBL2-mid-R 5′-GCACGTACTTCCACAGGGCTGG-3′ Sure protein A and Capto-L protein L
resins (both from GE Healthcare) to
Cloning, Expression, and Purification formulated on a Superdex 200 size- remove IdeS and undigested MAbs
of Hamster PLBL2: Technicians at Blue exclusion chromatography (SEC) column before beginning our SPR studies. Here,
Heron Biotechnology synthesized and from GE Healthcare. We collected CHO PLBL2 was immobilized onto
cloned the hamster PLBL2 gene fractions from that column and analyzed two of the four flow cells (FC2 and FC4)
(G3I6T1_CRIGR) with a C-terminal them with SDS-PAGE, high-pressure of a Series S CM5 sensor chip using a
hexahistidine-tag into a Hoffmann-La liquid chromatography (HPLC), and standard amine coupling and blocking
Roche proprietary vector. The sequence standard Western blotting techniques procedure recommended by the
of the PLBL2 gene was confirmed using (using antibodies to detect CHOPs and manufacturer and ~4,000-RU
the following primers: pRK-F, pRK-R, phospholipase B2) to determine purity. immobilization levels. We used the two
PLBL2-mid-F, and PLBL2-mid-R (see Surface Plasmon Resonance (SPR): To remaining FCs (FC1 and FC3) as in-line
the “Gene Sequence Primers” box above). evaluate binding interactions between references for FC2 and FC4 and treated
Using standard methods, we CHO PLBL2 and intact antibodies or them similarly (without immobilized
transformed One Shot TOP10 fragments, we used SPR technology on a proteins) before blocking. We diluted
chemically competent Escherichia coli cells Biacore T200 instrument from GE intact antibodies and fragments to 1 μM
(Life Technologies) with the pRK5sm– Healthcare. Using two different formats in HBS-EP+ buffer and ran them over
PBLB2 plasmid using standard methods, ensured that molecules for comparison FCs coated with immobilized CHO
then selected transformants on Luria were kept in the mobile phase as analytes PLBL2 for five minutes at a 5-μL/min
broth (LB) agar plates containing 50 µg/ instead of being immobilized as ligands flow rate. In-line reference-corrected
mL carbenicillin antibiotic. From an onto sensor chips. All SPR experiments responses were reported five seconds
overnight culture of TOP10 cells, we were carried out at 25 °C and corrected before the end of each sample injection.
purified the plasmids using a QIAprep against signals from the in-line reference And we regenerated the sensor chip by
spin maxiprep kit (Qiagen). Transient cell and buffer-blank. injecting 10 mM Glycine (pH 2.0) over
transfections at the 35-L scale were With the first format, we evaluated the immobilized CHO PLBL2 at a
expressed PLBL2 as described elsewhere interactions of PLBL2 with various 30-μL/min flow rate for 10 seconds.
(11). rhuMAbs. Here, anti-hu IgG Fc (GE CHOP ELISA Blocking with Anti-PLBL2
We ultrafiltered HCCF tenfold by Healthcare) was covalently immobilized Antibodies: To determine whether
tangential-flow filtration (TFF) using to all four flow cells (FCs) of a Series S PLBL2 was the only CHOP impurity
10-kDa molecular weight cutoff CM5 sensor chip (GE Healthcare) to a causing nonlinear MAb 2 sample
(MWCO) membranes. The ultrafiltered level of ~6,500 response units (RU). We dilution, we assayed in-process pool
HCCF was diafiltered against 10 then used immobilized anti-hu IgG Fc to samples potentially containing both
volumes of a PBS-NaCl buffer. Then we capture rhuMAbs diluted into HBS-EP+ PLBL2 and other CHOP in our PLBL2
applied the resulting material to a buffer — 10 mM 4-(2-hydroxyethyl)-1- ELISA and in our CHOP ELISA.
Ni-NTA immobilized metal-affinity piperazineethanesulfonic acid, 150 mM Samples were assayed either neat or after
chromatography column (Qiagen) and sodium chloride, 3 mM preincubation with rabbit polyclonal anti-
eluted with an increasing imidazole ethylenediaminetetraacetic acid, 0.05% PLBL2.. Sample dilutions were assayed
gradient. After conditioning the polysorbate 20, pH 7.4 — for FC2, FC3, by CHOP ELISA either neat (undiluted)
Ni-NTA pool with a buffer containing and FC4 to achieve an immobilization or after preincubation with rabbit
sodium sulfate, we applied it to an Octyl level of ~900 RU. FC1 served as an polyclonal anti-PLBL2. In particular, we
Sepharose hydrophobic-interaction in-line reference cell. With CHO noted the impact of anti-PLBL2
chromatography (HIC) column from GE PLBL2 diluted to 1.0 µM in HBS-EP+ antibodies on the CHOP ELISA and
Healthcare. The HIC column was eluted buffer subsequently injected into all four whether the sample diluted nonlinearly.
with a decreasing sodium sulfate FCs at a 50-µL/min flow rate for 60
gradient. seconds, dissociation proceeded for three Results
We rechromatographed the resulting minutes. At the end of each association/ We used a platform CHOP ELISA to
pool on a Ni-NTA column step-eluted dissociation cycle, we regenerated the detect CHOP impurities in
with a high concentration of imidazole, sensor chip by injecting 3 M magnesium biopharmaceuticals produced by a
then concentrated the resulting pool chloride into all four FCs for 30 seconds. relatively harmonized upstream cell
using centrifugal filtration units equipped Using the second format, we culture process. Using a common assay
with 10-kDa MWCO membranes compared PLBL2 binding interactions for multiple products makes it possible to
(EMD Millipore). The concentrated with full-length, F(ab′)2, and Fc identify atypical results in specific clinical
Ni-NTA rechromatographed pool was fragments generated from Genovis IdeS candidates. In one such case, we observed

BioProcess International 13(4) A pril 2015


an atypical, profoundly nonlinear dilution Collected fractions were assayed in the Figure 1:  Nonlinear dilution of in-process pool
samples for a MAb product in clinical
of samples late in purification. Figure 1 CHOP ELISA (Figure 2a’s trace with development; twofold dilution series were
shows the CHOP ratios (ng CHOP/mg diamond marks), with immunoreactive plotted according to CHOP ratios (ng CHOP/mg
product) of twofold serially diluted material (eluted early in the product) for samples taken from the initial
samples from the initial HCCF and chromatogram) well-separated from the harvested cell culture fluid (HCCF) and several
several intermediate purification pools. product. A small amount of such material intermediate downstream purification pools.
Pools early in the purification process dilute in
Those early in the downstream process coeluted with the product and was not parallel to the assay standard (giving dilution-
dilute in parallel with the assay standard pooled with other immunopositive independent CHOP ratio values); later pools
(giving dilution-independent CHOP fractions for subsequent study. show increasing CHOP ratio values as samples
ratio values); later pools show increasing As described above, the CHOP- are diluted. The only processing between the
values as samples are diluted. positive fractions (fractions 8–20) were third step and final pools is an ultrafiltration/
diafiltration (UF/DF) concentration and buffer
We calculated the coefficient of pooled, concentrated, and analyzed by exchange step.
variation (CV) for assays at each of those SDS-PAGE. Sypro Ruby-stained bands
10 6
sample dilutions. If the CV was ≤20%, were excised, digested with trypsin, and HCCF

we concluded that differences across the resulting gel-extracted peptides were 105

CHOP Ratio (ng/mg)


dilutions are within acceptable levels of sequenced by mass spectrometry. After 104
variability for ELISA sample replicates, comparing the peptide sequences with First step pool
and the sample was reported as databases of product, CHO proteins, and 103
Second step pool
exhibiting no dilution nonlinearity. If the other mammalian proteins, we identified 102 Third step pool

CV is >20% and inspection shows an phospholipase B-like 2 (PLBL2) as the


101
increasing CHOP level with increasing main impurity.
Final pool
sample dilution, then a sample was We observed several dye-stained 0
0.00001 0.0001 0.10 10.0
reported to have dilution-dependent bands (Figure 2b). Although some minor Product Concentration (mg/mL)
behavior. As illustrated in Figure 1, the bands were product-related, we identified
Black diamonds = HCCF X = third step pool
CV of dilutions for later pools was 155%, the three major bands as having black squares = first step pool * = final pool
well above the cut-off value. homology to hamster PLBL2. Literature open squares = second step pool

One explanation for nonlinear sample on PLBL2 (12, 13) and information from
dilution is that a single CHOP is present its UniProt entry (Q8NHP8) describes ≥2 ng/lane and shows staining of both
in excess of available anti-CHOP the protein as a mannose-6-phosphate the full-length protein and the smaller
antibodies (3, 4), so it exceeds the glycosylated lysosomal molecule that is clipped fragment. The CHOP ELISA
ELISA’s antibody binding capacity. synthesized as a 66-kDa preproenzyme. standard detected four bands. No anti-
Under such an interpretation, sample Under mildly acidic conditions, this PLBL2 reactive bands were detected in a
dilution to the assay limit gives the most enzyme is clipped into two chains that MAb 1 sample, which served as a
reasonable estimate of the CHOP ratio. remain strongly associated in native negative control throughout the study
For the Figure 1 preparation, we conditions; however, in the presence of (ELISA PLBL2 ratio in MAb 1 is below
estimated the CHOP ratio in the final SDS and when heated, those chains the assay’s limit of detection, 0.3 ng/mg).
product to be ~300 ng/mg. separate into N- and C-terminal However, the same two bands detected
Because that sample contained an fragments. That explains the three bands in the PLBL2 standard were detected in
abnormally high level of a potential on the gel: those two clipped fragments two MAb 2 manufacturing runs: PLBL2
CHOP impurity in the final purification and the full-length protein. ratios of 120 ng/mg for run 2, and 300
pool, we investigated it further using an Having identified our putative ng/mg for run 3. Run 3 corresponds to
approach described elsewhere (2). This CHOP impurity, we had the PLBL2 that shown in Figure 1, which had
involves capturing the MAb on a gene cloned for transient expression in pronounced nonlinear dilution in the
chromatographic support, where it is CHO cells, then purified the results and total CHOP ELISA.
tightly bound, and then collecting flow- used them to raise both mouse We revised the MAb 2
through and washes in an effort to isolate monoclonal and rabbit polyclonal manufacturing process to reduce PLBL2
the dissociated impurity from the antibodies. We developed PLBL2- levels and observed no anti-PLBL2
product. The approach is based on the specific ELISAs using those reagents. reactive bands in final-product samples
assumption that the reason for impurity Both ELISAs gave comparable results from the resulting updated process.
copurification is an interaction between it across multiple products and intermediate Corresponding ELISA data for those
and the MAb product, which must be pools. We used the MAb-based ELISA lots showed PLBL2 levels <1 ng/mg. We
disrupted for impurity dissociation. to assess PLBL2 in all our MAb have found that purification processes
For that reason, we captured our portfolio biotherapeutics, including both can be modified successfully to reduce
product on a 1-mL CHT column that commercial products and those in clinical PLBL2 levels to <1 ng/mg.
was washed with PBS-Tween (Figure 2). development. Our survey of Genentech commercial
The UV chromatogram trace shows Figure 3 shows an immunoblot using biologics showed that PLBL2 is
product elution late in the chromatogram the rabbit polyclonal antisera. In the gel, undetectable in Herceptin, Avastin,
at >150 mM phosphate concentrations. the PLBL2 standard is visible down to Perjeta, Rituxan, and Xolair products.

A pril 2015 13(4) BioProcess International


Figure 2a:  Chromatographic separation of CHOP impurities from the product; (a) UV trace shows Figure 2b:  Chromatographic separation of
product elution late in the chromatogram at concentrations of phosphate >150 mM. Collected CHOP impurities from the product; CHOP-
fractions were assayed in the CHOP ELISA (diamonds) and show immunopositive material eluted positive fractions 8–20 were pooled,
early in the chromatogram (well-separated from product). A small amount of immunoreactive concentrated, and analyzed on a sodium-
material coeluted with product and was not included in subsequent studies. dodecyl sulfate polyacrylamide gel
500 electrophoresis (SDS-PAGE) gel.

400 CHOP ELISA of Fractions


CHOP (ng/mL)

300 UV Trace of IgG Product


200 250
C2 C1
100
150 A1
0 A2
0 5 10 15 20 25 30 35 40 A3 IgG
100
−100
Fraction
75 A4
A5
A6
Table 1:  SYPRO Ruby stained bands were excised and digested with trypsin; the resulting peptides 50
were analyzed by mass spectrometry. The coverage plot maps identified peptides of PLBL2 to the
highest database hit. Coverage: 195/585 (33%)
37 A7 PLBL2

MA A PMD R S P D G R A V R A L R L A L A L A S L T E V L L N C P A G A L P T QG P G R R R QN L A8
25 A9
D P P V S R V R S V L L D A A S GQ L R L V D G I H P Y A V AWA N L T N A I R E T GWA Y L D L G
20
IgG
A10
T N G S Y N D S L Q A Y A A G V V E A S V S E E L I Y M H WM N T M V N Y C G P F E Y E V G Y C E K
L K S F L E I N L E WM Q R E M E L S Q D S P Y W H Q V R L T L L Q L K G L E D S Y E G R L T F P T 15
G R F T I K P L G F L L L Q I A G D L E D L E Q A L N K T S T K L S L G S G S C S A I I K L L P G A 10
R D L L V A H N TWN S Y QNM L R I I K K Y Q L Q F R QG P Q E A Y P L I A G N N L V F S S Y P G
T I F S G DD F Y I L G S G L V T L E T T I G N K N P A LWK Y V Q P QG C V L EW I R N I V A N R
L A L D G A T W A D I F K Q F N S G T Y N N QWM I V D Y K A F I P N G P S P G S R V L T I L E Q I
lane 1 = reference material
lane 2 = ELISA positive fractions
P GMV V V A D K T E D L Y K T T YWA S Y N I P F F E I V F N A S G L QD L V A Q Y G DWF S Y T
K N P R A Q I F Q R DQ S L V E DMN S MV R L I R Y NN F L HD P L S L C E A C I P K P N A E N A
I S A R S D L N P A N G S Y P F Q A L Y Q R P H G G I D V K V T S F S L A K RM S M L A A S G P TW
D Q L P P F QW S L S P F R S M L HMG Q P D L W T F S P I S V PWD with antibodies), PLBL2 binding was
completely blocked in the PLBL2
Minimum PSMs: 0 1 2 5 Modified
ELISA. In the CHOP ELISA, samples
diluted linearly with a low CHOP value
However, we did find it as an impurity in samples. Additionally, although PLBL2 (<1 ng/mg). That result showed that all
several products that are still in clinical is suspected to have enzymatic activity, the previously observed nonlinearity
development. Nonlinear dilution no associated substrates have been could be explained by PLBL2 in the
behavior of samples in the CHOP published (14). If the protein is sample.
ELISA often evidenced PLBL2 as the enzymatically active, then its substrate Using the PLBL2-specific ELISA,
CHOP present in antigen excess. In remains unidentified. we assessed PLBL2 levels in HCCF
some cases, the CHOP ELISA showed We spiked purified PLBL2 into the from a number of production cell cultures
low CHOP ratio values in the final CHOP ELISA (Figure 4). At low and blank-run cultures. Levels varied
product (<2 ng/mg). But PLBL2 was concentrations (<10 ng/mL), the CHOP widely among production runs (Figure
present at similar levels based on assay accurately quantifies PLBL2 spikes. 5). For many MAbs, we measured several
PLBL2-specific ELISA results, Increasing concentration of PLBL2 HCCF samples for different production
suggesting that PLBL2 could be the decreases spike recovery consistent with cultures, and the bar graphs show their
predominant CHOP species even in saturation of the anti-PLBL2 antibodies minimum (solid bar) and maximum
products with low total CHOP. We in the overall CHOP ELISA. The most (hashed bar) values. With a median
implemented purification improvements accurate PLBL2 estimate (using only the PLBL2 level of 2.5 μg/mL, values
to reduce PLBL2 levels in all clinical CHOP ELISA) occurs when a sample is ranged 0.76–7.7 μg/mL. If expressed
products for which it was detected. diluted to near an assay’s quantitation relative to the total CHOP content of the
Reduction of PLBL2 from MAb 2 pools limit. That supports the “antigen-excess” HCCF, PLBL2 content ranged 0.16%–
(Figure 3, Runs 5–8) illustrates the hypothesis as the cause of sample dilution 1.2% of the CHOP, with a median value
success of those improvement efforts. nonlinearity in the CHOP ELISA. of 0.37%. Levels range over about an
We also evaluated fluorogenic To further establish a connection order of magnitude across cultures.
synthetic phospholipase substrates (Life between nonlinear dilution of samples Previous studies on MAb
Technologies) to determine to determine and PLBL2 as the causative impurity, we biotherapeutics have shown that the
whether the enzyme was active but found incubated in-process pools with a tenfold biggest influence on CHOP level in
no enzymatic activity with those molar excess of rabbit anti-PLBL2 before protein A pools is the antibody rather
substrates for either our purified PLBL2 ELISA testing. With that pretreatment than nonspecific binding to the
or MAb biotherapeutic in-process (designed to coat PLBL2 in the samples chromatography media (15). So

BioProcess International 13(4) A pril 2015


Figure 3:  Immunoblot using rabbit polyclonal antisera to PLBL2; (left to right) the lanes correspond Figure 4:  PLBL2 assayed in the platform
to MW standards, a PLBL2 standard at three different protein concentrations, the CHOP standard, enzyme-linked immunosorbent assay (ELISA)
MAb 1, and six lots of MAb 2. The first two of those six represent MAb 2 prepared before the for CHOP; at low levels, PLBL2 recovery is near
protocol was optimized, and the last four came after changes made to reduce PLBL2 levels. 100%. As PLBL2 levels increase, the assay
response does not similarly increase, which is
consistent with the interpretation that only a
limited number of antibodies is available to
250 bind PLBL2. Here, “antigen excess” is expected
150 to lead to nonlinear dilution in samples
100 containing >10 ng/mL of PLBL2.
75 1.5

Optical Density
50 1.0
37
0.5

25 0.0
20 1 10 100
Protein Concentration (ng/mL)
W

2- PL 2
PL 2
B2

rd

Ab b 1

M 2 n3
M 2R n2
M 2R n5
M 2R n6
2 n7
8
-n LB
ng B

n
M

da

Ab Ru
Ab Ru
Ab u
Ab u
Ab u
Ru
A
10 g P

M
an

the first paper that described an ELISA


g

M 2
-n

St

for HCPs— in that case for E. coli


20

OP

proteins in recombinant human growth


CH

hormone (4).
Although the nonlinearity observed in
potentially small differences in antibody PLBL2. In some cases, PLBL2 detection our CHOP ELISA for multiple different
sequence might greatly influence CHOP was low or nonexistent with those products could have resulted from
interactions with the antibodies. In an antibody preparations (data not shown). multiple, different CHOP species, our
effort to understand why CHO-derived Thus, some CHOP ELISAs might be results show that a single CHOP had
PLBL2 copurifies with only some insensitive to this particular impurity.If independently copurified with multiple
MAbs, we conducted two more studies. the ELISA reagents are insensitive to antibodies. Several teams have put forth
First, we used SPR to look for PLBL2, then samples shown to have low the “hitchhiker impurities” idea that
PLBL2–antibody (or antibody fragment) CHOP values might yet contain high major HCP impurities in final
interactions (Figure 6). This levels of this impurity. therapeutic products are there because
demonstrated interactions between they bind to the product (15–20). Sisodiya
immobilized MAbs and PLBL2 and vice Discussion et al. found that antibody–CHOP
versa. However, measuring such PLBL2 may be a relatively common and interactions contributed significantly to
interactions required micromolar levels potentially overlooked HCP impurity in carrying CHOP along in the recovery
of PLBL2 in the mobile phase. The therapeutic MAbs produced by CHO process and observed significant
binding was readily reversible, with a cultures. In our case, HCP ELISA differences between antibodies in their
return to baseline in minutes. These dilution nonlinearity was not an assay interactions with CHOP (15).
observations suggest that the binding artifact; it was a consequence of antigen Those observations have been
interaction is relatively weak, and only excess and reflected the need to dilute confirmed with PLBL2 as a specific
high MAb concentrations during samples “into the range of the assay” so CHOP. It appears to copurify based on
purification processes allow these there would be sufficient antibody to an association with the Fab′2 portion of
impurities to be carried along. capture and detect the HCP impurity. It particular antibodies. Also, the
Further SPR studies showed that is important, therefore, to test samples for probability that PLBL2 will copurify
PLBL2 was interacting with the F(ab′)2 HCP impurities at multiple dilutions. with a given antibody may depend on the
portion of the MAb rather than its Fc Nonlinear responses should be particular clone selected for production.
region (Figure 7). MAb 1 served as the investigated because they may indicate CHO cells are heterogeneous in PLBL2
negative control, and MAb 2 was the the presence of a single or limited expression levels, as illustrated by the
positive antibody. These results show that number of HCP species that can copurify production cultures whose HCCF
most of the SPR signal is derived from with product and might be present in PLBL2 levels differ by over tenfold.
the F(ab′)2 fragment. MAb 6 had a very large amounts. Similarly, PLBL2 levels in blank runs
weakly positive SPR signal to both the Identification of sample dilution vary, so anti-CHOP antibody
intact molecule and its F(ab′)2 fragment. nonlinearity thus is an important signal preparations produced in response to
Finally, we assessed whether other that an HCP may be present in antigen those blank-run immunogens also could
CHOP ELISA antibody preparations — excess. Our description of the antigen- differ in their ability to detect PLBL2.
both those from commercial kits and excess phenomenon and associated Initially, we evaluated a commercial
developed in-house — were responsive to nonlinearity is not new. It was detailed in ELISA test kit for hamster PLBL2 from

A pril 2015 13(4) BioProcess International


Figure 5:  Levels of PLBL2 in HCCF samples displayed as μg/mL (left) and percentage of the total CHOP (right) from 13 different MAb products; for cases in
which multiple independent production cultures were measured for the same MAb, the solid bar shows the lowest observed value, and the stippled bar
shows the highest observed value. Wide variation among cultures in PLBL2 levels is shown by absolute μg/mL and as relative percentage of the total CHOP.
9 1.4
8 1.2
PLBL2 in HCCF (µg/mL)

PLBL2 as % of CHOP
1.0
6
5 0.8
4 0.6
3
0.4
2
1 0.2

0 0.0
1 2 3 4 5 6 7 8 9 10 11 12 13 1 2 3 4 5 6 7 8 9 10 11 12 13
Monoclonal Antibody ID Number Monoclonal Antibody ID Number

Cusio Bio in China. However, the kit did with recombinant therapeutics. We have hamster PLBL2.
not react with our purified PLBL2 observed no cases in which only the One primary concern about HCP
standard, making it unsuitable for use in N-terminal or C-terminal peptides were impurities is their potential to be
establishing product impurity levels. We present, nor any case in which one or the immunogenic. As foreign proteins, they
are unaware of any other commercially other fragment was in excess. might elicit an immune response in
available kits or reagents for detection of Phospholipases exist widely in nature exposed patients (23, 24). Hamster
hamster PLBL2, so we developed our and can be found in bee, spider, and PLBL2 has ~80% amino acid sequence
own immunoassays. snake venoms as well as in bacteria. identity with its human ortholog, and
PLBL2 was originally described in a However, nonmammalian species have many of their differences are surface-
study that screened for proteins distinct genes and share low sequence exposed residues. Given the multiple
containing mannose-6-phosphate. In homology with mammalian PLBL2. In amino-acid differences between human
both published literature and our own particular, although a phospholipase has and hamster PLBL2, it is reasonable to
experience, the two PLBL2 constituent been found to be a virulence factor in assume that PLBL2 would be
peptides remain closely associated after fungi, and infected humans have immunogenic in humans. So for patient
acid hydrolysis. Denaturing conditions generated antibodies to this enzyme (21, safety, PLBL2 levels should be reduced
are required to dissociate those 22), that molecule is unrelated to PLBL2. as much as practically possible in
fragments. Consistency between our two The natural substrate for PLBL2 is as yet recombinant biotherapeutics (25).
PLBL2-specific ELISAs (one based on a unreported, but it may be an arylamidase We have no reason to think that our
pair of mouse MAbs and one based on (14). Synthetic substrates used to measure observations are unique to Genentech
rabbit polyclonal antibodies) supports the bacterial phospholipases were not CHO cells or the Genentech production
idea that the whole protein copurifies hydrolyzed by our purified recombinant process. It is possible that PLBL2 is a

Figure 6:  Representative surface plasmon resonance (SPR) sensograms showing reversible binding between 1 μM PLBL2 and immobilized MAb
biotherapeutics; (left) MAb 1, with which PLBL2 tends to copurify, shows positive binding. Multiple traces in the MAb 1 panel show measurement
reproducibility with independent MAb 1 preparations (lots), each of which was made with the new process and is free of copurifying PLBL2. But the
positive interaction of PLBL2 and the antibody remains a property of the antibody. (right) MAb 1, with which PLBL2 does not copurify, does not respond
in the sensogram, showing that this antibody does not exhibit the PLBL2 binding properties of MAb 2. (* sample start)
20 20

17 17
MAb 1
14 14
MAb 2
Response Units

Response Units

11 11

8 8
Injection Injection
5 5 start stop

2 2

−1 −1
−10 10 30 50 70 90 110 130 150 −10 10 30 50 70 90 110 130 150
Time (seconds*) Time (seconds*)

BioProcess International 13(4) A pril 2015


Figure 7:  Average SPR binding levels for 1 μM of antibody and antibody
frequent impurity in CHO-derived MAb biotherapeutics across fragments to immobilized CHO PLBL2 — the first three bars show the
the industry. It is also possible that widely used commercial binding response values of intact MAbs; the next three bars show
CHOP ELISAs do not detect this impurity, or they could binding response values of the F(ab’)2 fragments of those MAbs; the final
significantly underreport its presence. three bars show binding response values of their Fc portions. Reference
flow-cell subtracted binding response values of samples to immobilized
We have instituted process changes to reduce PLBL2 levels
CHO PLBL2 were reported as the response five seconds before the end of
in all our MAb products. As illustrated herein for one product, each injection. Data are reported as average values, and error bars
it is possible to reduce PLBL2 to levels near or <1 ng/mg. We represent standard deviations of four experimental replicates.
reduced patient risk with a combination of a sensitive assay for
PLBL2 and implementation of PLBL2-reducing process 1,800
changes. 1,600
1,400
References
1,200
1 Tscheliessnig AL, et al. Host Cell Protein Analysis in Therapeutic

Response Units
Protein Bioprocessing: Methods and Applications. Biotechnol. J. 8(6) 2013: 1,000
655–670.
800

1,459
2 Wang X, Hunter AK, Mozier NM. Host Cell Proteins in Biologics
Development: Identification, Quantitation, and Risk Assessment. Biotechnol. 600
Bioeng. 103(3) 2009: 446–458. 400

766
3 Zhu-Shimoni J, et al. Host Cell Protein Testing By ELISAs and the
200 85
Use of Orthogonal Methods. Biotechnol. Bioeng. 111(12) 2014: 2367–2379. −3
31 −2 9 −1 −1 −4
4 Anicetti VR, et al. Immunoassay for the Detection of E. coli Proteins 0
in Recombinant DNA Derived Human Growth Hormone. 2 1 6 2 1 6 2 1 6 Buffer
−200
J. Immunol. Meth. 91(2) 1986: 213–224. Monoclonal Antibody
5 Corren, J, et al. Lebrikizumab Treatment in Adults with Asthma.
New England J. Med. 365(12) 2011: 1088–1098. 1992: 319–326.
6 McCullogh KC, Spier RE. Monoclonal Antibodies in Biotechnology. 20 Zhang Q , et al. Comprehensive Tracking of Host Cell Proteins
Cambridge University Press: New York, NY, 2009. During Monoclonal Antibody Purifications Using Mass Spectrometry. MAbs
7 Wild D. The Immunoassay Handbook: Theory and Applications of Ligand 6(3) 2014:659–670.
Binding, ELISA, and Related Techniques, Fourth Edition. Elsevier Science: 21 Chen SC, et al. Purification and Characterization of Secretory
Oxford, UK, 2013. Phospholipase B, Lysophospholipase, and Lysophospholipase/Transacylase
8 Gagnon P, et al. A Ceramic Hydroxyapatite-Based Purification from a Virulent Strain of the Pathogenic Fungus Cryptococcus neoformans.
Platform: Simultaneous Removal of Leached Protein A, Aggregates, DNA, Biochem. J. 347(2) 2000: 431–439.
and Endotoxins from MAbs. BioProcess Int. 4(2) 2006: 50–60. 22 Santangelo RT, et al. Detection of Antibodies to Phospholipase B in
9 Stanker LH, Vanderlaan M, Juarez-Salinas H. One-Step Purification Patients Infected with Cryptococcus Neoformans By Enzyme-Linked
of Mouse Monoclonal Antibodies from Ascites Fluid By Hydroxylapatite Immunosorbent Assay (ELISA). Med. Mycol. 43(4) 2005: 335–341.
Chromatography. J. Immunol. Meth. 76(1) 1985: 157–169. 23 Bailey-Kellogg C, et al. CHOPPI: A Web Tool for the Analysis of
10 Champion KM, et al. Comparison of the Escherichia coli Proteomes for Immunogenicity Risk from Host Cell Proteins in CHO-Based Protein
Recombinant Human Growth Hormone Producing and Nonproducing Production. Biotechnol. Bioeng. 111(11) 2014: 2170–2182.
Fermentations. Proteomics 3(7) 2003: 1365–1373. 24 Rosenberg AS, Worobec A. Immunogenicity Concerns of Therapeutic
11 Wong AW, Baginski TK, Reilly DE. Enhancement of DNA Uptake Protein Products, Part 2: Considering Host-Specific and Product-Specific
in FUT8-Deleted CHO Cells for Transient Production of Afucosylated Factors Impacting Immunogenicity. BioPharm Int. December 2004.
Antibodies. Biotechnol. Bioeng. 106(5) 2010: 751–763. 25 Koren E, et al. Recommendations on Risk-Based Strategies for

Products. J. Immunol. Meth. 333(1–2) 2008: 1–9. •


12 Deuschl F, et al. Molecular Characterization of the Hypothetical 66.3- Detection and Characterization of Antibodies Against Biotechnology
kDa Protein in Mouse: Lysosomal Targeting, Glycosylation, Processing, and
Tissue Distribution. FEBS Lett. 580(24) 2006: 5747–5752.
13 Lakomek K, et l. Initial Insight into the Function of the Lysosomal
66.3 Kda Protein from Mouse By Means of X-Ray Crystallography. BMC Corresponding author Martin Vanderlaan is in analytical operations at
Struct. Biol. 9, 2009: 56. Genentech (a Member of the Roche Group), 1 DNA Way, South San
14 Repo H, et al. Is the Bovine Lysosomal Phospholipase B-Like Protein Francisco, CA 94080; 1-650-225-4439, fax 1-650-225-5695; vdl@gene.
an Amidase? Proteins 82(2) 2014: 300–311. com. Julie Nishihara, George Tsui, Margaret Lin, Feny Gunawan,
15 Sisodiya VN, et al. Studying Host Cell Protein Interactions with Sara Parker, Robert Ming Wong, Karthik Veeravalli, Patrick
Monoclonal Antibodies Using High-Throughput Protein A Chromatography. McKay, Chris Yu, Lori O’Connell, Benjamin Tran, Chris Fong, Judith
Biotechnol. J. 7(10) 2012: 1233–1241. Zhu-Shimoni, and Denise Krawitz are in technical development;
16 Hunter AK, et al. Separation of Product Associating E. coli Host Cell Wendy Sandoval, Peter Liu, and Rajesh Vij are in research; Justin
Proteins OppA and DppA from Recombinant Apolipoprotein A-I(Milano) in Low, Xiangdan Wang, Jihong Yang, and Valerie Quarmby are in
an Industrial HIC Unit Operation. Biotechnol. Progr. 25(2) 2009: 446–453. development sciences; and Kathleen Francissen is in CMC regulatory
17 Levy NE, et al. Identification and Characterization of Host Cell — all at Genentech, as well.
Protein Product-Associated Impurities in Monoclonal Antibody
Bioprocessing. Biotechnol. Bioeng. 111(5) 2014: 904–912.
18 Nogal B, Chhiba K, Emery JC. Select Host Cell Proteins Coelute For electronic or printed reprints, contact Rhonda Brown of Foster
with Monoclonal Antibodies in Protein A Chromatography. Biotechnol. Progr. Printing Service, rhondab@fosterprinting.com, 1-866-879-9144 x194.
28(2) 2012: 454–458. Download personal-use–only PDFs online at www.bioprocessintl.
19 Wilson MR, Easterbrook-Smith SB. Clusterin Binds By a Multivalent com.
Mechanism to the Fc and Fab Regions of IgG. Biochim. Biophys. Acta. 1159(3)

A pril 2015 13(4) BioProcess International


B i o P r o c e s s TECHNICAL

Rapid Development and Scale-Up


of Biosimilar Trastuzumab
A Case Study of Integrated Cell Line and Process Development

Kumar Dhanasekharan, Claudia Berdugo-Davis, Xiaoming Liu, Carl Richey,


Zaneer Segu, David Calabrese, Valerie LeFourn, Pierre-Alain Girod, and Victor Vinci

C
ompared with that for new Figure 1a:  Cell-line development process used herein, with clone selection in 96-well and
drugs, biosimilar expansion to 6-well plates performed manually.
development faces Limiting dilution
Screening 1 Fed-batch
significantly condensed Vector
construction
Transfection of clones,
Retransfection
of lead clones
and screening 2
of clones, then
evaluation in
Cryo-
preservation
timelines from cell line to first-in- then expansion
expansion
shake flasks

human (FIH) trials. A biosimilar


development program needs to
accelerate quickly toward preclinical Limiting ELISA and ELISA and ELISA, SDS-PAGE,
and phase 1 studies; phase 2 studies dilution, expansion to expansion to and expansion
96-well plating 24-well plates 6-well plates to shake flask
typically are not required because
dose response and other patient-
treatment concepts are already
Figure 1b:  Analysis of isolated clones in
established by the original, European Medicines Agency 96-well plates
comparator medicine. Phase 3 (EMA), the US Food and Drug
140
studies typically are limited to Administration (FDA), and the Screening 1 Screening 2
fewer patients, which ultimately World Health Organization 120
shortens overall timelines and costs. (WHO) (1–3). This often requires 100
Titer (mg/L)

The key challenge remains: multiple iterations during early 80


demonstrating comparability and phase development compared to 60
high similarity based on in-depth that for an originator product (4). 40
analytics as outlined in a number of An integrated approach to cell line 20
guidance documents from the development and early stage 0
screening and process development 1 21 41 61 81
is therefore imperative for making Ranked Clones
Product Focus:  Biosimilars
appropriate decisions in pursuing
(monoclonal antibodies)
the right candidate for preclinical protein. Although recombinant
Process Focus:  Production and and clinical manufacturing. Here, technologies are extremely powerful
Downstream Processing we present such an approach for a tools for expression of
biosimilar trastuzumab, from cell- biotherapeutics, manufacturing
Who Should Read:  Process and line through early phase success depends on identifying cell
product development, manufacturing, development with manufacturing- lines that stably produce high
and analytical personnel ready titers and clinically relevant protein titers. A critical part of
product quality attributes. biopharmaceutical development is
Keywords:  Genetic engineering, DoE,
Our platform begins with choosing the right cells for meeting
cell line and process optimization,
development of a high-producing the objective of confirming cell
scale-up, and process control
Chinese hamster ovary (CHO) cell productivities, cell-line stability,
Level:  Intermediate line expressing the desired target and final-product quality. The most

BioProcess International 13(4) A pril 2015


Figure 2:  Cell culture process design for 2-L and 20-L studies
common gene expression systems
are based on dihydrofolate reductase
(DHFR) and glutamine synthetase
(GS). P2
P0 P1
However, identifying suitable
cell lines is traditionally a time-
25 20 L 20 L
consuming, labor-intensive process mL
100
3L
mL 10 L
because their productivity and
stability can vary enormously.
Several thousands of clones must be 2L 2L
screened to find those with both
the highest yield and the best
Figure 3:  Downstream process design for 2-L and 20-L studies
product quality traits. To improve
cell-line selection and reduce Protein A with Low-pH
CEX with Polishing AEX Viral
timelines, we developed a stable cell MAbSelect viral
POROS XS with POROS HQ filtration
UF/DF
Sure inactivation
line using the SUREtechnology
Platform system from Selexis SA. It
is based on DNA-based elements
(Selexis Genetic Elements, SGEs) operations, with comparable process commercial chemically defined
that control the dynamic and product attributes. Every step (CD) product from Irvine
organization of chromatin within in this process was supported by Scientific. To keep the environment
all mammalian cells. They allow for detailed analyses: e.g., N-glycan of the cells unchanged throughout
unwinding a transgene’s genomic analysis to demonstrate the whole procedure, we used the
environment — a prerequisite for comparability and quality, and size- same medium as a basal medium for
efficient gene expression. The SGE exclusion chromatography (SEC) transfection, single-cell cloning,
approach has been shown to prevent and capillary isoelectric focusing and production. We incorporated
transgene silencing and provide (cIEF) to confirm quality based on genes encoding the Hc and Lc and
high transcription rates for all size variants and charge variants. selection markers into SUREtech
copies of the transgene, thus Finally, we have developed a vectors containing SGEs to
enabling higher and more stable process ready for transfer to maintain transcription at the
expression of recombinant proteins. manufacturing using chemically maximum level.
Subsequently, we performed defined medium under an Figure 1a is a schematic of the
design-of-experiment (DoE) studies aggressive timeline, with a product development process we used to
using shake f lasks to identify base titer in the range of 3–4 g/L, while establish the cell line for use in this
media and feed media strategies, meeting product quality attributes study. DNA was delivered by
confirming the results at 2-L scale. comparable to those of the electroporation to improve
We further optimized the 2-L innovator molecule. transfection efficiencies. We
process to meet desired product applied an antibiotic selection to
titers and quality. A three-column M aterials and M ethods create the stable pools, picking
downstream process was developed We adapted a current good about 100 clones for manual
to yield high monomer content. We manufacturing practice (CGMP) expansion from 96-well to 24- and
successfully scaled up our process to compliant CHO-K1 ATCC six-well plates. Based on
a pilot-scale setup for both derivative host cell line to grow in immunoglobulin G (IgG) titers, we
upstream and downstream BalanCD Growth A media, a transferred selected clones to shake

Figure 4:  Media screening with temperature shift (left); media screening with no temperature shift (right)
3.0
Viable Cell Density (107 cells/mL)

100 3.0 100


Viable Cell Density (107 cells/mL)

90 90
2.5 2.5
80 Flask 6 80
2.0 70 Flask 3 70
Viability (%)

2.0
Viability (%)

60 60
1.5 Flask 10 50 1.5 50
40 40
Flask 11 Flask 4
1.0 30 1.0
Flask 12 Flask 8 Flask 1 Flask 7
30
Flask 13 Flask 14 20 Flask 2 20
0.5 0.5
10 Flask 5 10
Flask 9
0.0 0 0.0 0
0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 0 2 4 6 8 10 12 14 16 18 20 22 24 26 28
Time (days) Time (days)

A pril 2015 13(4) BioProcess International


Table 1:  Typical analytical control strategy for early stage development (Char. = characterization) Analytical Abbreviations
Analytical Control Strategy
CD: Circular dichroism
Critical Quality Attribute Analytical Method In-Process Release Stability Char.
CE: Capillary electrophoresis
IgG Protein A HPLC × ×
Protein concentration UV-A280 × × × CE-SDS: Sodium dodecyl sulfate
Protein identification LC-MS peptide × ×
capillary electrophoresis
mapping cIEF: Capillary isoelectric focusing
Physicochemical LC-MS peptide × × DSC: Differential scanning calorimetry
characterization mapping
Biophysical analysis Far-UV CD × ×
ELISA: Enzyme-linked immunosorbent
assay
DSC thermal × ×
analysis FL-HPLC: High-performance liquid
Differential × × chromatography with fluorescence
fluorimetry detection
Functional analysis Binding ELISA × GC: Gas chromatography
Cell-based assays ×
HPLC: High-performance liquid
Residual DNA PCR × chromatography
Residual protein A Protein A ELISA × ×
iCE: Immunoaffinity capillary
Residual host-cell protein (HCP) HCP ELISA × ×
electrophoresis
Bioburden Plate-count × ×
methods ICP-MS: Inductively coupled plasma
Endotoxin LAL assay × × mass spectrometry
Charge variants (deamidation, IEX-HPLC × × × × IEX-HPLC: Ion-exchange high-
N-terminal pyroglutamates, cIEF or iCE × × × performance liquid chromatography
sialylation)
IgG: Immunoglobulin G
Size variants (aggregation, SEC-HPLC × × × ×
dimers, fragmentation) SDS-PAGE × × × ×
LAL: Limulus amebocyte lysate
CE-SDS × × × × LC-MS: Liquid chromatography–mass
Process impurities (Triton RP-HPLC × × spectrometry
X-100, residual solvents, GC × × PCR: Polymerase chain reaction
phosphenes, formulation
components) ICP-MS × × RP-HPLC: Reverse-phase high-
Glycan profile, intact protein FL-HPLC, LC-MS × × performance liquid chromatography
mass SDS-PAGE: Sodium dodecyl sulfate
Nonglycosylated product CE-SDS × × polyacrylamide gel electrophoresis
SEC-HPLC: Size-exclusion high-
f lasks, then further examined the Strategy Development: To evaluate a performance liquid chromatography
best performers during a 14-day set of commercially available cell UV-A280: 280-nm absorbance
fed-batch process. culture media with different feed ultraviolet spectroscopy
We accomplished those multiple options, we performed a basal media
steps of expansion without any screening study. To evaluate the
antibiotic selection, carefully media types for both basal and feed and confirm process parameters
characterizing and selecting clones media, total feed amount, feed such as temperature shift, pH, and
for attributes that would be duration, and temperature shift, we glucose levels, we ran two sets of
compatible with our production performed 14 shake-f lask studies four 2-L scale bioreactors,
requirements. Finally, we banked with 100-mL working volume (WV). evaluating two different
and tested 10 qualified clones for And we subsequently inoculated an temperature shift conditions (34 °C
cell identity and for endogenous and additional 12 shake f lasks to further and 32 °C). Glucose was fed up to 4
adventitious microbial contaminants optimize feeding strategy. g/L when it fell below 3 g/L; pH
(bacteria, fungi, and mycoplasma). To monitor those shake f lasks for was controlled using a sodium
To analyze their expression cell growth and viability, we used a bicarbonate–carbon dioxide system.
constructs at the nucleic-acid level Vi-Cell counter from Beckman The system controlled dissolved
(genetic stability), we assessed the Coulter; to monitor for metabolites, oxygen (DO) at a set point of 40%
constructs by quantitative we used a BioProfile Analyzer with a fixed air-f low rate and O2 on
polymerase chain reaction (qPCR). instrument from Nova Biomedical. demand. We defined our feed
Finally, we selected a single clone We took titer samples on day 3 and strategy, total feed amount, and
for further process development. then daily after day 5, finally feed duration based on results from
harvesting the f lasks when the our shake-f lask studies.
Process Development cultures reached ~60% viability. For the seed train, we thawed
and Scale-Up Cell Culture Process and passaged cells about every three
Basal Media Screening and Feed Development: To further evaluate days. For the last seed-train passage

BioProcess International 13(4) A pril 2015


Figure 5:  Effect of feeding timing and frequency on titer Figure 7:  Cell culture process scale-up; (a) viable cell density and viability,
(b) titer, (c) glucose profile, and (d) lactate profile
4.0
Flask 10
3.5 Flask 8
Flask 9 A 100
3.0 90
Flask 5 2.5

Viable Cell Density (107 cells/mL)


Flask 13 20-L (1), 1% feed D3−D18
2.5
Titer (g/L)

Flask 3 80
Flask 11 20-L (2), 1% feed D3−D18
2.0 Flask 4 2.0 70
Flask 2

Viability (%)
Flask 7
1.5 Flask 6 60
Flask 1 1.5
1.0 50
2-L golden batch
0.5 40
1.0
0.0 30
0 2 4 6 8 10 12 14 16 18 20 22 24
0.5 20
Time (days) 20-L (2), 1% feed D3−D12, 2% feed D13−18
10
20-L (1), 1% feed D3−D12, 2% feed D13−18
0.0 0
Figure 6:  Comparing the effects of temperature shift (TS) to 32 °C and 34 °C 0 2 4 6 8 10 12 14 16 18 20
from 37 °C on viable cell density, cell viability, and expression titer Time (days)
3.0 100 3.0
20-L (1), 1% feed D3−D18 B
90
Viable Cell Density (107 cells/mL)

20-L (2), 2% feed D3−D12, 1% feed D13−D18


2.5 2.5
20-L (1), 2% feed D3−D12, 1% feed D13−D18
80
20-L (2), 1% feed D3−D18
70 2.0
2.0

Titer (g/L)
Viability (%)
2-L TS 34 °C (1)
60
1.5
1.5 50
2-L TS 32 °C (1) 2-L TS 32 °C (2) 40 1.0
1.0
30
2-L TS 34 °C (2) 2-L golden batch
0.5
0.5 20
10
2-L Baseline 0.0
0.0 0 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
0 2 4 6 8 10 12 14 16 18 20 22 24 26 Time (Days)
Time (days) 9
C
8 20-L (1), 1% feed D3−D18
(n – 1) before bioreactor inoculation, we expanded the 20-L (2), 1% feed D3−D18
7
cells in a Wave bioreactor from GE Healthcare 20-L (2), 2% feed D3−D12, 1% feed D13−D18
20-L (1), 2% feed D3−D12, 1% feed D13−D18
equipped with temperature control. Figure 2 shows this 6
Glucose (g/L)

2-L golden batch


process. 5
We sampled the culture daily to measure cell
4
density, viability, and metabolite levels. After day 3, we
sampled daily for analytical purposes (including titer 3
and quality attributes). Bioreactors were harvested 2
when cultures reached ~60% viability. Using a sterile
1
process, we collected the cell culture contents and
centrifuged them before filtering the supernatant 0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
through a 0.2-µm Pall Acropak 500 filter. Filtrate was Time (Days)
collected and stored in a –80 °C freezer for downstream 6
processing. 2-L golden batch
D
Cell Culture Process Scale-Up to Pilot Scale: To 5
demonstrate scalability, we scaled the 2-L process up to
20 L. We performed several 20-L bioreactor runs using 4
Lactate (g/L)

a Sartorius-Stedim BB30 stainless-steel bioreactor. The


system maintained scale-independent parameters (pH, 3
pCO2 , feed strategy, temperature shift, and so on) as 20-L (1), 2% feed D3−D12, 1% feed D13−D18
defined in the 2-L bioreactor cell cultures. Scale- 2
dependent parameters were scaled appropriately. We 20-L (1), 1% feed D3−D18
designed the air f low rate to a constant volume per 1
20-L (1), 1% feed D3−D18
volume per minute (vvm) that translated to 0.15 20-L (2), 2% feed D3−D12, 1% feed D13−D18
standard liters per minute (SLPM) with a ring sparger. 0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
Agitation rate scale-up was based on constant power Time (Days)

A pril 2015 13(4) BioProcess International


per unit volume of f luid (P/V), which translated to 100 eluting the protein with 50 mM sodium acetate at pH
rpm. 3.4.
Downstream Development and Scale-Up: We After a viral inactivation step, we titrated the pool
developed and scaled up a three-column purification to pH 5.0. Next, cation exchange (CEX)
process to meet the requirements for our 20-L scale chromatography in bind–elute mode used POROS XS
bioreactor run. The downstream process starts with media from Life Technologies, with equilibration using
harvest clarification using a POD depth filtration 50 mM sodium acetate followed by a 4-CV wash at pH
system from EMD Millipore, followed by column 5.0. We used a gradient elution with sodium chloride
operations on a GE Healthcare ÄKTA Pilot system. from 75 to 270 mM over 20 CV. Then a polishing
The protein A capture step used MabSelect SuRe resin anion-exchange (AEX) chromatography used POROS
from GE Healthcare with phosphate-buffered saline HQ resin (also from Life Technologies) in f low-
(PBS) equilibration followed by loading and washing through mode, with a 3-CV chase at pH 7.8–8.0.
with PBS. After a secondary wash with PBS and 1 M Finally, we processed the eluate through a Virosart
sodium chloride, we ran a final wash with PBS before CPV virus filter followed by an ultrafiltration/

Figure 8:  Cell culture process optimized scale-up; (a) VCD and viability, (b) titer, (c) glucose profile, and (d) lactate profile

100 4.0
Viable Cell Density (×107 cells/mL)

A 90
B
2.5 3.5
20-L (2), 1% D3−18 2-L optimized: bolus feed
80
2-L golden batch 3.0
2.0 70 Viability (%) 2-L optimized: slow feed
20-L optimized: bolus feed
60 2.5
1.5 50 Titer (g/L) 2.0
20L (2), 1% D3−18
40
1.0 1.5
2-L optimized: bolus feed 30 20L-optimized: slow feed
2-L optimized: slow feed 1.0
5.0 20
20-L optimized, slow feed
20-L optimized, bolus feed 10 0.5
2-L golden batch
0.0 0 0.0
0 2 4 6 8 10 12 14 16 18 20 22 0 2 4 6 8 10 12 14 16 18 20 22 24
Time (Days) Time (Days)
8.0 6
20L-optimized, slow feed
2L-optimized: bolus feed C D
7.0
20L-optimized, bolus feed 5
20L-02 1% D3-18 2-L golden batch
6.0 2L-optimized: slow feed
4
Glucose (g/L)

Lactate (g/L)

2L golden batch
5.0 20-L (2) 1% feed D3−18
20-L optimized: slow feed
4.0 3
3.0
2
2-L optimized: bolus feed 20-L optimized: bolus feed
2.0
1 2L optimized: slow feed
1.0
0.0 0
0 2 4 6 8 10 12 14 16 18 20 22 0 2 4 6 8 10 12 14 16 18 20 22
Time (Days) Time (Days)

Figure 9:  Purity measured by size-exclusion high-performance liquid chromatography (SEC-HPLC) and charge variants measured by capillary isoelectric
focusing (cIEF) from Cook Pharmica’s 20-L run compared with the innovator molecule
140 140
Main Peak
120 Monomer 120
RS-Innovator
100
Wavelength (AU)

100
Wavelength (AU)

2-L Run

80 80
60 Acidic
60 Peaks
40 40 Basic
20 Peaks
20 Innovator
0 0
−20 20-L Run
−20
8 9 10 11 12 13 14 15 16 17 18
Time (minutes) 8 9 10 11 12 13 14 15 16 17 18
Time (minutes)

BioProcess International 13(4) A pril 2015


Figure 10:  Comparing N-glycans released from the innovator product (black) and Cook Pharmica’s
diafiltration (UF/DF) operation in 20-L run (blue)
an ÄKTA Crossf low system from
GE Healthcare. 25.0
Analytical Development: Table 1

G0F
22.5
shows a typical analytical strategy
20.0
during early development. For the

Measurement? (units?)
current proof-of-concept study, we 17.5
considered a subset of those 15.0

G1Fa
methods as presented below. 12.5
10.0
R esults

G0F-GlcNAc
G0-GlcNAc
We made research cell banks 7.5

G1Fb

G2f+2SA
G2f+SA
(RCBs) from the single clone we

G2f
5.0

G0

M5

G1

G2
had selected. Figure 4 shows our 20-L Run
2.5
initial shake-f lask media screening Innovator
0.0
results. Those results helped us 8 9 10 11 12 13 14 15 16 17 18
identify basal media (Media A) and Time (minutes)
feed media (Feed B) with a Fucose Mannose Galactose N-acetyl-D-glucosamine Sialic acid

temperature shift (TS) to provide


the best performance in terms of
Figure 11:  Relative glycoform content from two different 20-L bioreactor runs; afucosylated and
viable cell density (VCD) and fucosylated glycoforms are comparable for both innovator and 20-L bioreactor runs.
viability over the duration of the
88.79 89.91 89.73
runs. Innovator
Our feed-strategy optimization 77.8 75.88
20 L (1)
results (Figure 5) indicate a 20-L (2)
nonlinear relationship between total 54.52
feed percentage and titer. Higher
total feed percentages seem to be 34.86
beneficial up to 16–20%; beyond
that point, however, additional feed 14.52 15.93
did not increase titer significantly. 5.65
8.33 7.27 7.22
Based on statistical analysis using 1.57 1.76

JMP software from SAS Institute, G0 G1 G2 Fucosylated Afucosylated


daily feeds from days 3 to 18 gave
the best titer results.
A temperature-shift study using pH >6.6 results in cell cultures with whereas viability of the 2-L process
2-L bioreactors showed that the 34 lower peak VCD but extended declines slower, so titer continues to
°C condition helped cultures reach longevity of viability >60%. To go beyond 2.5 g/L.
higher VCDs, but that viability achieve higher cell densities while We hypothesized that increased
declines more sharply and earlier ensuring extended longevity, we set mixing speed (P/V ) and slower
than it does at 32 °C. Results are the pH dead band to 6.8–7.2 and additions of feed media and glucose
consistent between duplicates, based our TS criteria on reaching a might be required to reduce
although a shift in growth rate was cell density of 1 × 10 6 cells/mL. variability in control of pH, feed,
observed in one run (Figure 6). And to minimize base consumption and glucose control. To test our
Based on protein A HPLC titer for pH control, we increased the hypothesis, we tried slow and bolus
measurements, cell culture post-TS pH dead band to 6.6–7.2. feed media and glucose additions
performance was slightly higher for Cultures controlled at lower coupled with increased agitation
a TS to 32 °C than it was for a TS glucose levels (around 1 g/L) had rate. As part of scale-up
to 34 °C. lower VCDs than those topped at 4 confirmation, we ran additional 2-L
We studied the combined effects g/L under the same pH conditions. cultures in parallel with the same
of pH dead-band and glucose So we decided to maintain the increased agitation to correspond to
control to establish a baseline 2-L glucose levels at 4 g/L. a constant P/V and similar feed
process that we labeled as the Process Scale-Up Results: As addition schemes.
“golden batch.” Maintaining pH Figure 7 shows, metabolic profiles As the data show in Figure 8, in
above 6.8 led to higher cell growth for the process at 20-L scale all cases the peak cell density was
rates and higher peak VCDs, but compare favorably with those at 2-L higher than it had been in previous
viability dropped to 60% or less scale, although 20-L expression runs, and titer in one of the 20-L
sooner. By contrast, maintaining titers f latten out at about 2 g/L runs reached 3.35 g/L. Although

A pril 2015 13(4) BioProcess International


the second 20-L run continued on a galactosylated glycoforms appear to Scientific, and Patient Safety Perspectives.


J. Natl. Compr. Canc. Netw. 9, 2011: S1–S22.
similar track, a viability drop led us be present in the biosimilar, and G0
to harvest the reactor early in that appears to be relatively more
case. One key noticeable feature in abundant than it is in the innovator.
these runs is the dramatic inf luence Further iterative developments are Corresponding author Kumar
on lactate profile of the controlled- ongoing to improve comparability Dhanasekharan (Kumar.
feeding strategy in later stages of and similarity through the use of Dhanasekharan@cookpharmica.com) is
culture, in which the metabolism different culture media director of process development;
shifted to lactate consumption. supplements. Additional Claudia Berdugo-Davis is a senior
scientist, and Xiaoming Liu is an
We evaluated product quality characterization of the innovator
associate scientist in cell culture; Carl
using size-exclusion high- molecule is also required to help us
Richey and Zaneer Segu are senior
performance liquid chromatography understand lot-to-lot variability of scientists in biologics process
(SEC-HPLC) for size variants, our biosimilar MAb. development and innovation; and Victor
capillary isoelectric focusing (cIEF) Vinci is chief scientific officer and vice
for charge variants, and N-glycan A S uccessful Platform president at Cook Pharmica, 1300 South
analysis for glycosylation profiles. We have demonstrated an Patterson Drive, Bloomington, IN 47402.
Although purity was 99.8% after integrated cell-line development David Calabrese is director of cell line
three column steps, at the end of effort with process development and services, Valerie LeFourn is head of cell
the UF/DF step an overall purity of scale-up as a successful model for culture, and Pierre-Alain Girod is chief
99.1% monomer and host-cell expediting early phase development scientific officer at Selexis SA, 18 Chemin
des Aulx, 1228 Plan-les-Ouates,
protein (HCP) <0.05 ng/mg were for biosimilar antibodies. Our
Switzerland.
reached with an overall product approach did not compromise on
yield of 77%. the scientifically systematic
As measured by SEC-HPLC, the DoE-based approach, but it took a For electronic or printed reprints, contact
biosimilar purity is comparable with targeted set of experimental studies Rhonda Brown of Foster Printing Service,
that of the innovator molecule. to meet the requirements of product rhondab@fosterprinting.com, 1-866-879-9144
Additional product attributes — quality as well as process scale-up x194. Download personal-use–only PDFs
online at www.bioprocessintl.com.
such as the cIEF charge-variant needs. This approach was
profiles — are also comparable successfully demonstrated using a
(Figure 9). biosimilar Trastuzumab as a model
Because our monoclonal antibody molecule, with scale-up from 2-L
(MAb) molecule is a biosimilar bench scale through a 20-L pilot
product, its N-glycan profile is an scale process that will be scalable to
important critical quality attribute larger GMP operations for clinical
(CQA) for the design space production.
according to the quality by design
(QbD) paradigm. Figure 10 shows R eferences
the results of our comparison of 1 CBER/CDER. Guidance for Industry:
Scientific Considerations in Demonstrating
N-glycans released from the
Biosimilarity to a Reference Product. US Food
innovator product (black) and Cook and Drug Administration: Rockville, MD,
Pharmica’s 20-L run product. February 2012; www.fda.gov/downloads/
Clearly, the glycoforms present in drugs/
the innovator’s molecule are also guidancecomplianceregulatoryinformation/
guidances/UCM291128.pdf.
present in the material we
2 EMEA/CHMP/BMWP/42832/2005.
produced. That glycosylation profile
Guideline on Similar Biological Medicinal
is reproducible and consistent across Products Containing Biotechnology-Derived
multiple runs. Proteins As Active Substance: Non-clinical and
Relative afucosylation content in Clinical Issues. European Medicines Agency:
both products is also comparable London, UK, 2006; www.ema.europa.eu/
docs/en_GB/document_library/Scientific_
(Figure 11). This is another
guideline/2009/09/WC500003920.pdf.
important CQA for a MAb such as
3 Guidelines on Evaluation of Similar
trastuzumab, which has bioactivity Biotherapeutic Products. World Health
based on its binding affinity toward Organization: Geneva, Switzerland,
Fcγ receptors on effector cells and October 2009; www.who.int/biologicals/
clearance of target cells through publications/trs/areas/biological_
therapeutics/BS2110Dft_guidelines_Final_
antibody-dependent cell-mediated
HK_IK_29July_09.pdf.
cytotoxicity (ADCC) activity.
4 Zelenetz AD, et al. NCCN
However, fewer end-capped Biosimilars White Paper: Regulatory,

BioProcess International 13(4) A pril 2015


FOCUS  ON... QUALITY

Fundamental Strategies
for Viral Clearance
Part 1: Exploring the Regulatory Implications

by Kathryn Martin Remington

O
ver the past several decades, biologics) are expected to be submitted
biologics such as monoclonal for approval. In addition, biobetters,
antibodies (MAbs) and (considered improved versions of
recombinant proteins have innovator biologics) are gaining
provided therapeutic benefits and efficacy popularity.
for the treatment of human disease. Pharmaceutical companies developing
Completion of the human genome biosimilars and biobetters must confirm
project (launched in 1990) produced a that the efficacy, safety, and purity of
draft of the genome in 2001. A full their products are similar to those of
sequence was published on the 50th original innovator-approved drugs (2). In
anniversary (2003) of the initial this process, viral safety is of upmost
publication of Watson and Crick’s papers importance.
on the double-helical structure of DNA Because production of all
(1). That large volume of genetic biopharmaceutical products carries an
H1N1 influenza virus
information has been translated into (WWW.VISUALSCIENCE.RU.COM) inherent risk of potentially introducing
usable scientific knowledge, helping adventitious viruses, viral safety is an
researchers in their hunt for better posttranslational modifications, they have essential consideration in the
therapeutic approaches and establishing become the preferred host for production development and manufacture of these
the functional relationships between of complex protein therapeutics. products. Viruses possessing diverse
genes and clinical disorders. Ongoing It has been more than a decade after characteristics can potentially
research has led to new understandings, the completion of the human genome contaminate therapeutic agents, so
which have paved the way for many new sequence and more than 30 years after different methods of virus inactivation
designs of recombinant biologics. regulatory approval of the first genetically and removal are required to guarantee a
Today, most biopharmaceutical engineered drug. On 28 October 1982, safe drug supply.
products are proteins expressed in the FDA approved the first recombinant Although viral safety is a focal point
recombinant hosts. Microbial systems are medicine produced using genetic for regulatory authorities, the potential
attractive platforms because of their low engineering: recombinant human insulin, for transmission of diseases —
cost, high productivity, and rapid Humulin, developed by Eli Lilly and specifically viral — through any biologic
implementation. And bacterial expression Company and Genentech. The drug was that uses a mammalian-derived
systems present no concerns about expressed in bacteria. Today, such component in its manufacturing process
adventitious mammalian viruses. biologics represent a major portion of the is a real risk.
However, many biopharmaceutical pharmaceutical market, with most Current state of the art precludes
molecules are simply too big or too approved in the United States and claiming absolute absence of viral
complex to be made by bacteria. For Europe as glycosylated proteins produced presence in biologics. The practical
example, properly folded proteins by mammalian cell culture. approach applied by most regulatory
(including MAbs with appropriate Patents are expiring soon for more bodies is evaluating safety case by case.
glycosylation) require posttranscriptional than 20 first-generation blockbuster Such a strategy considers current, real,
metabolic machinery that is available biologics. As a result, an increasing and perceived virus contamination
only in mammalian cells. And because number of biosimilars (which can be concerns on the basis of source materials
such cells can add relevant human-like similar but not identical to innovator used for manufacturing each product as

January 2015 13(1) BioProcess International


well as availability of adequate Vulnerability
technologies for detection and clearance Sourcing of raw materials is one of the
of viral agents. The quality, safety, and greatest concerns for regulators and
Through international collaborations, efficacy of biologics biopharmaceutical manufacturers. Most
regulatory guidelines stress a holistic depend on careful incidents of viral contamination result
approach that includes appropriate CONTROL of process from using poorly characterized
sourcing of materials; demonstrating the materials. This risk is intensified by the
capability of a manufacturing process for inputs and operating sourcing of raw materials from multiple
viral clearance; and testing in process, conditions. vendors. And because large volumes of
among others. Critical use of an effective process media and gases are used to
viral safety program involves careful produce biologics, viral contamination is
selection and testing of cell lines and requires a high level of reproducibility in not necessarily a rare occurrence.
original raw materials for adventitious commercial development and Evaluation of raw materials has
viruses. In addition, in-process testing manufacture to guarantee high-quality, become a critical component of any risk
can identify contaminants such as consistent biological products. The level of mitigation strategy. Such a strategy can
endogenous retroviral particles, which are complexity is generally influenced by a require evaluating source materials
typically associated with mouse and host of factors, including individual cell beyond what can generally be expected or
hamster cells. Viral testing is characteristics, growing environment, and is reported about the source. For example,
complemented by viral clearance studies, nutrients provided during manufacturing many processes now limit or entirely
which demonstrate the capacity of (5–8). move away from animal‑derived
manufacturing steps to inactivate or Whereas synthetic small-molecule materials for producing recombinant cell
remove potential viral contaminants. pharmaceuticals rely on a chemical lines or media formulations. But there is
analysis of components to assess purity, still a risk that drug products have been
A Market in Transition the quality, safety, and efficacy of complex exposed to animal-derived materials at
Over the past decades, the development biologics depend on careful control of one or multiple points in the supply
and production of medicinal products for process inputs and operating conditions. chain. Furthermore, it is not always
human use has undergone a major shift. Processing involves many manufacturing possible to move away from animal-
Among factors affecting the global stages and parameters, as well as stringent derived materials, which presents a risk of
market for pharmaceutical manufacturing control mechanisms and process control introducing virus contamination into a
is a paradigm shift in the need for (9). manufacturing system.
medicinal products. Demands and Although biopharmaceutical Rigorous evaluation and mitigation
patient needs are shifting from production may be challenging, are required to eliminate potential risks
blockbuster drugs to specialized, even guaranteeing virological safety is even of contamination. Some manufacturers
personalized medicine (3). Because many more complex. Aided by advanced use virus inactivation methods such as
innovator drugs are going off patent in detection, donor selection, raw materials UV-C inactivation, high-temperature
the next few years, the industry is moving screening, virus removal, and inactivation, short-time (HTST) treatment, or
toward developing biosimilars and biomanufacturers have greatly increased removal (virus reduction filtration)
biobetters. the safety profile of their products. Such procedures for culture media or media
A September 2013 report published therapies include tissue-, blood- and components. The viral reduction
by IMS Health suggests that the global plasma-derived products, MAbs, potential of such risk‑mitigation
market for biologics is expected to grow recombinant proteins, vaccines, and procedures can be evaluated in a viral
from US$169 billion in 2012 to $221 animal products carrying an inherent risk clearance study. Similarly, the viral
billion in 2017 (4). The authors of that of transmitting viruses based on the reduction potential of downstream
report expect that biologics will continue source material, manufacturing processes, manufacturing process steps (designed to
to outpace the growth of overall and routes of administration. complement rigorous upstream screening
pharmaceutical spending and expect it to But despite stringent efforts on the and mitigation steps) also can be
be driven by increased demand for MAb part of manufacturers, companies evaluated in a viral clearance study.
innovator drugs and recombinant human continue to face threats of new and Lessons learned from past incidents
insulin. The authors also predict that by emerging viruses. Unfortunately, show the importance of thoroughly
2016, the market for biosimilars will be endogenous and adventitious viruses can understanding the complexity of (and
worth about $5 billion, with expected escape detection. That might be the result possible risks in) the supply chain.
exponential growth afterward. of limited assay sensitivity or limitations Regulatory authorities can use such
in detection methods, which require understanding — as well as the
Complexity increased efforts of viral clearance. As knowledge of the extent to (and the
The manufacturing process of biologics is part of such work, viral clearance studies nature in which) biologics include
challenging and complex because such are designed to help demonstrate virus animal-derived components — to assess
products are derived from plants, reduction, achieved by a number of steps how much viral clearance is required.
animals, or mammalian cells or in the manufacturing process in case
supplemented with reagents derived from process intermediates are contaminated. Mammalian Cells
living systems or cell lines. The process With its intrinsic complexity, the
production of recombinant their ability to remove or inactivate viral Risk Management
biopharmaceuticals poses a series of contaminants (11–14). A risk-based approach to viral safety is
unique manufacturing challenges. Viral The European Medicines Agency consistent with the approach that is now
safety is particularly challenging when (EMA), US Food and Drug used to ensure quality in
mammalian cells are used. Although Administration (FDA), and other biopharmaceuticals and is outlined in
iatrogenic incidents have occurred in the authorities have adopted guidelines that several guidance documents. Such
past decade as a result of biologics focus on testing and evaluating viral documents include ICH Q8, which
contamination, incomplete inactivation of safety of biotechnology products derived includes process analytical technology
viruses, or contamination of excipients in from characterized cell lines of human or and quality by design; ICH Q9, which
a nonrecombinant product, only a limited animal origin. Those guidelines provide a includes quality risk management; and
number of known and reported events general framework for testing production ICH Q10, which presents guidelines on
have taken place. (To date there has been cell banks and bulk harvests for viral pharmaceutical quality system (12–14).
no transmission of an infectious virus to a contaminants and designing viral Use of a risk-based approach to viral
patient from a recombinant biotech clearance evaluation studies. The EMA safety is consistent with the viral safety
product. However, such transmissions also has adopted guidelines concerning “tripod.” Selection and characterization
have occurred with plasma-derived clinical trials and submissions based on of source materials will define potential
products, and some have resulted in an investigational medicinal product contaminants that may be present. New
disease. Furthermore, although dossier that includes viral safety technologies such as massively parallel
contamination events for products evaluation (15). sequencing are powerful tools for
derived from mammalian cells have been detecting and identifying contaminants
reported, they were detected before the Investigational Products that may be present, especially for novel
drugs were administered.) The EMA has provided a regulatory or complex source materials. The
In 2010, Kerr and Nims reported that guideline governing virus safety for resulting information can provide a
during a specific 10-year period, biologics investigational medicinal biotechnology framework for more directed routine
derived from a number of mammalian products (15). This document, which testing.
cell culture processes were evaluated became effective in 2009, applies to Testing provides key information
using in vitro virus screening assays. clinical trial material derived from well- about the design of a viral clearance
Reported results showed that only characterized cell lines and harmonizes study. Understanding the potential
reovirus type 2 and Cache Valley virus (a the regulatory expectations throughout contaminants will guide selection of a
bunyavirus contaminant) were detected the European Union. This guideline virus panel for study so that models of
in biologics manufactured in Chinese acknowledges that, for clinical trial potential contaminants are included.
hamster ovary (CHO) cells. Other materials, the manufacturing process and Testing data also will direct the level of
reports showed that the murine full product characterization is still in clearance that is expected from
parvovirus mouse minute virus and development. It offers reduced viral safety manufacturing processes. For example,
contamination with vesivirus 2117 had testing and viral clearance study scope retroviral-like particles are quantitated in
also been detected in biologics compared with requirements for bulk harvests from CHO cell lines. Viral
manufactured using CHO cells (10). marketing authorization provided in clearance for a process should be high
ICH Q5A. enough that the risk of that contaminant
Myth and Solution The viral safety of products intended in a final dose of product is less than one
As a result of stringent regulatory for clinical trials is a priority. However, in a million.
oversight, biologics manufacturers are the EMA guideline acknowledges that For many years, the focus of viral
required to show that their products are working cell banks may not yet be clearance has been on downstream
safe for human use. And although established, and it lifts the requirement manufacturing. Although downstream
biopharmaceutical scientists and for testing cells at the limit of in vitro viral clearance steps are still essential,
regulatory authorities understand that age. For viral clearance studies, a reduced many manufacturers are looking at
absolute freedom of viral contamination virus panel can be used (a retrovirus and upstream viral clearance steps. Including
is impossible, much can be done to a parvovirus). Robustness studies are not a viral inactivation or removal step for cell
ensure viral safety. This is reflected in the required for products at this stage of culture media or media components is
regulatory landscape. development, again acknowledging that another way to mitigate the risk of viral
The International Conference on process parameter ranges may not be contamination. Steps such as UV-C
Harmonisation of Technical established yet. If a product used for a inactivation, HTST, and even virus
Requirements for Registration of clinical trial has been manufactured reduction filtration can provide yet
Pharmaceuticals for Human Use (ICH) using relatively new chromatography another layer of viral safety for a
is a unique collaboration of regulatory resins, then clearance studies on aged manufacturing process.
authorities and the pharmaceutical resins and resin sanitization studies are
industries of Europe, Japan, and the not expected.These guidelines were Facility Design
United States. The organization has developed with input from industry and One often-underestimated approach to
developed guidelines requiring other regulatory agencies that worked viral safety is the design of a
manufacturers of biologics to demonstrate together to assure viral safety.
January 2015 13(1) BioProcess International
manufacturing facility. To comply with follow regulatory requirements to ensure Danbury, CT, September 2013.
regulatory directions, it is important to adequate virus removal. During the past 5 Vyas VV, et al. Clinical Manufacturing of
Recombinant Human Interleukin 15: Production
consider where viral clearance takes place decade, viral clearance studies have Cell Line Development and Protein Expression in
in the production and manufacturing received increased regulatory scrutiny and E. coli with Stop Codon Optimization. Biotechnol.
process and which steps provide effective become important factors to consider in Prog. 28(2) 2013: 497–507.
viral inactivation or removal. Well- the design of manufacturing processes. 6 Hofland P. An Introduction to Recombinant
designed facilities ensure adequate Viral clearance in downstream Biologics and Immunotherapy. TVCC, Almere, The
segregation of process intermediates that purification can involve a number of Netherlands, January 2003.
have been through an effective viral methods to inactivate or remove viruses 7 Schellekens H. Biosimilar Therapeutics —
What Do We Need to Consider? NDT Plus 2(sup
reduction step from those intermediates from a final therapeutic product. Because 1) 2009: i27–i36.
that have not. Also, dedicated viral contamination incidents also can occur in 8 Zhang J. Mammalian Cell Culture for
clearance must be planned in the bioreactors (adding associated regulatory Biopharmaceutical Production. Manual of
manufacturing process to alleviate safety implications and costs associated with Industrial Microbiology and Biotechnology, Third
concerns. decontamination), some manufacturers Edition. Baltz RH, Davies JE, Demain AL, Eds.
As demonstrated by viral incidences have chosen to also include upstream American Society of Microbiology: Washington,
DC, 2010; 157–178.
with vesivirus 2117, the possible barriers in their processes.
9 Lee JF, Litten JB, Grampp G.
introduction of undetected viruses from Biomanufacturing processes are varied Comparability and Biosimilarity: Considerations
unknown sources is a great risk. Once a and can include a number of upstream for the Healthcare Provider. Curr. Med. Res. Opin.
viral contaminant enters the and downstream steps. Virus-removing 28(6) 2012: 1053–1058.
manufacturing process, the only effective (nano-) filters (designed for the removal 10 Kerr A, Nims R. Adventitious Viruses
control involves robust viral inactivation of almost all virus types through a size- Detected in Biopharmaceutical Bulk Harvest
or removal steps. exclusion mechanism and Samples Over a 10-Year Period. PDA J. Pharm. Sci.
Technol. 64(5) 2012: 481–485.
chromatographic resins in column or
11 ICH 5A: Viral Safety Evaluation of
Lessons Learned membrane configurations) are typically Biotechnology Products Derived from Cell Lines of
The most important lessons to be learned included downstream. Upstream steps Human or Animal Origin. The International
from regulatory guidelines —and a may include UV-C inactivation and Conference on Harmonisation of Technical
general principle that can be applied to HTST. Exposure to low pH or solvent/ Requirements for Registration of Pharmaceuticals
the manufacture of all detergent treatment steps provides for Human Use: Geneva, Switzerland, October
1997.
biopharmaceuticals — is that viral inactivation of enveloped viruses
12 ICH Q8: Pharmaceutical Development. The
clearance studies must be reviewed case downstream. International Conference on Harmonisation of
by case. Each study involves product- In addition to viral clearance, a typical Technical Requirements for Registration of
specific risk factors. If a modular manufacturing process also contains steps Pharmaceuticals for Human Use: Geneva,
approach to viral clearance is used for to purify a biopharmaceutical product. Switzerland, August 2009.
similar products and processes, you must Athough such steps are not necessarily 13 ICH Q9: Quality Risk Management. The
demonstrate the absence of product- designed to provide virus reduction, they International Conference on Harmonisation of
Technical Requirements for Registration of
specific influences on viral clearance. may have the potential to do so. Pharmaceuticals for Human Use: Geneva,
That may involve verification of Chromatography is a good example of a Switzerland, November 2005.
processing parameters, analytical testing purification operation that can help 14 ICH Q10: Pharmaceutical Quality System.
of process intermediates, and/or some achieve viral clearance. However, because The International Conference on Harmonisation
evaluation of viral clearance. Testing a number of operating parameters can of Technical Requirements for Registration of
source materials will help you identify influence viral reduction, Pharmaceuticals for Human Use: Geneva,
Switzerland, June 2008.
potential contaminants and the level of chromatography is generally less robust
15 Guideline on Virus Safety Evaluation of
virus reduction to be achieved in than viral reduction by other methods Biotechnological Investigational Medicinal Products.
manufacturing. such as inactivation.
UK, July 2008. •
Evaluation of Medicines for Human Use: London,
Past incidents have shown that the In the conclusion of this two-part
correct approach ultimately helps prevent series, I discuss effective planning of viral
costly mistakes. Such mistakes can delay clearance studies as well as the
regulatory approval of new biologics and implementation of viral clearance Kathryn Martin Remington, PhD, is a
principal scientist, development services,
halt current production and strategies.
clearance, at BioReliance; kathy.remington@
manufacturing of already approved bioreliance.com.
drugs. References
1 Collins FS, et al. A Vision for the Future
of Genomics Research. Nature 422, 2003: 835–
Importance of Viral Clearance For reprints, contact Rhonda Brown of Foster
847.
Effective viral clearance is required to Printing Service, rhondab@fosterprinting.com,
2 Islam R. Bioanalytical Challenges of 1-866-879-9144 x194. Download personal-
guarantee the availability of a safe supply Biosimilars. Bioanalysis 6(3) 2014: 349–356. use–only PDFs at www.bioprocessintl.com.
of drug products for human use. Because 3 Weiler A. A New Dawn for Western
adventitious viruses present in original CMOs. Pharm. Technol. 1 February 2014: S14–S17.
materials can contaminate biologics 4 The Global Use of Medicines: Outlook Through
during processing, manufacturers must 2017. IMS Institute for Healthcare Informatics:
BioProcess International 13(1) January 2015

You might also like