Sunteți pe pagina 1din 6

Strategies for Developing Design Spaces for Viral Clearance by Anion Exchange

Chromatography During Monoclonal Antibody Production


Daniel M. Strauss, Tony Cano, Nick Cai, Heather Delucchi, and Magdalena Plancarte
Process Research and Development, Genentech, Inc., One DNA Way, South San Francisco, CA 94080

Daniel Coleman
Non-Clinical Biostatistics, Genentech, Inc., One DNA Way, South San Francisco, CA 94080
Gregory S. Blank, Qi Chen, and Bin Yang
Process Research and Development, Genentech, Inc., One DNA Way, South San Francisco, CA 94080

DOI 10.1002/btpr.385
Published online January 29, 2010 in Wiley InterScience (www.interscience.wiley.com).

The quality-by-design (QbD) regulatory initiative promotes the development of process


design spaces describing the multidimensional effects and interactions of process varia-
bles on critical quality attributes of therapeutic products. However, because of the com-
plex nature of production processes, strategies must be devised to provide for design
space development with reasonable allocation of resources while maintaining highly de-
pendable results. Here, we discuss strategies for the determination of design spaces for
viral clearance by anion exchange chromatography (AEX) during purification of monoclo-
nal antibodies. We developed a risk assessment for AEX using a formalized method and
applying previous knowledge of the effects of certain variables and the mechanism of
action for virus removal by this process. We then use design-of-experiments (DOE) con-
cepts to perform a highly fractionated factorial experiment and show that varying many
process parameters simultaneously over wide ranges does not affect the ability of the
AEX process to remove endogenous retrovirus-like particles from CHO-cell derived feed-
stocks. Finally, we performed a full factorial design and observed that a high degree of
viral clearance was obtained for three different model viruses when the most significant
process parameters were varied over ranges relevant to typical manufacturing processes.
These experiments indicate the robust nature of viral clearance by the AEX process as
well as the design space where removal of viral impurities and contaminants can be
assured. In addition, the concepts and methodology presented here provides a general
approach for the development of design spaces to assure that quality of biotherapeutic
products is maintained. V C 2010 American Institute of Chemical Engineers Biotechnol.

Prog., 26: 750–755, 2010


Keywords: quality-by-design (QbD), design space, viral clearance, anion exchange
chromatography (AEX), design of experiments (DOE)

Introduction cess knowledge required for approval of a QbD filing


involves the formation of a design space, which is defined as
Because biotherapeutic products are complex and difficult ‘‘the multidimensional combination and interaction of input
to fully characterize, the process by which they are created variables and process parameters that have been demon-
is an important factor in defining the final drug product. As strated to provide assurance of quality.’’1 As long as changes
such, the characterization and validation of the processes are are made within the design space, the change can be dealt
critical parts of ensuring that products are consistently safe with by internal quality controls rather than requiring regula-
and effective for patients. Quality-by-design (QbD) is a regu- tory approval. A general guidance for the development of
latory initiative that encourages a thorough understanding of design spaces has been provided,1 but few examples of their
the process variables, which affect the quality of the product development in biotechnology settings have been
and how those parameters interact with one another.1 Once described.2–4 Additional case studies of design space devel-
approved, the QbD approach may then allow for more regu- opment will provide the industry with the concepts needed
latory flexibility in manufacturing process changes. The pro- to build systematic methods and take full advantage of the
QbD regulatory initiative.
The first step in the QbD process involves determining
Current address of Daniel M. Strauss: Bioproduct Research and
critical quality attributes (CQAs), which are those attributes
Development, Eli Lilly and Company, Indianapolis, IN 64221
Correspondence concerning this article should be addressed to of a product that characterize its quality, safety, and efficacy.
B. Yang at yang.bin@gene.com. Although many products may have distinct CQAs, viral

750 C 2010 American Institute of Chemical Engineers


V
Biotechnol. Prog., 2010, Vol. 26, No. 3 751

Table 1. LRV Values Obtained from Small-Scale Virus Reduction Experiments


mAb1-a* mAb1-b mAb2-a mAb2-b mAb3-a mAb3-b mAb4 mAb5 mAb6 mAb7
XMuLV 3.7†,‡ 6.0 3.5‡ 5.1 5.8 5.4 6.2 5.5 5.7 5.7
SV40 5.8 4.8 5.1 4.1 4.7 4.0 5.7 4.6 4.3 4.1
MMV 5.5 5.3 5.0 5.4 5.0 5.7 5.3 4.7 5.5 5.2
Product mAb8 mAb9 mAb10 mAb11 mAb12 mAb13 mAb14
XMuLV 5.7 5.4 5.9 5.4 6.1 5.3 5.9
* Letters represent multiple processes developed at different scales for the same mAb product.

‘‘’’ symbols indicate that no virus was detected in the product pool.

These values were obtained using infectivity assays, while all others were obtained using QPCR assays.

safety should be considered for all products, which are pro- Table 2. RVLP Design Space Parameters
duced in mammalian cell culture. These processes are known Parameters Low Center High
to produce endogenous retroviruses or retrovirus-like par-
Feedstock conductivity (mS/cm) 5.0 9.5 14.0
ticles (RVLPs),5–7 and they have the potential to be infected
Load density (mg/mL resin) 50 N/A* 200
by adventitious viruses during mAb production.8 Any viral Feedstock pH 7.0 7.8 8.5
impurities that are or could be present in the product pools Equil. buffer conductivity (mS/cm) 5.0 9.5 14.0
must be removed or inactivated by the product purification Feedstock impurity level No Varied HCCF
process.9–11 One unit operation that plays an important role spike spike
Buffer pH 7.0 7.8 8.5
in providing acceptable levels of the viral clearance CQA is Temperature ( C) 15 23 30
the anion exchange chromatography (AEX) step. AEX per- Flow rate (cm/h) 50 150 250
formed in product flow-through mode has been shown to be Equil. buffer tris-concentration (mM) 15 25 35
capable of providing a high level of removal of both nonen- Equilibration phase duration (CV) 3 5 7
veloped and enveloped viruses, including the endogenous * Both load density values were obtained for all runs.
RVLPs that are produced by CHO-cells.12–14 This process
has also been shown to be very robust. It has been tested (SevenMulti, Mettler Toledo, Columbus, OH), and either
over wide ranges of parameters and shown to consistently WFI or sodium acetate was added to achieve the desired
provide log reduction values greater than 4.15,16 In addition, conductivity. Selected loads were also spiked to 0.1% V/V
our group has validated this process for many different mAb with harvested cell culture fluid (HCCF) to introduce
products and processes (Table 1). In any case, no specific increased levels of host cell impurities. The feedstock was
product effect has been observed, and despite many differen- then filtered through a 0.45 lm PVDF filter (Millipore, Bill-
ces between those processes, the step has consistently pro- erica, MA), and the protein concentration was determined by
vided excellent LRV values. The robustness observed for optical density at 280 nm (OD280).
viral clearance by AEX makes it an excellent candidate for
A small-scale chromatography column (0.66-cm diameter,
the application of QbD concepts.
Bio-Chem Valve/OmniFit, Boonton, NJ) was packed with
In this work, we discuss strategies for developing design naive QSFF resin to 20-cm bed height. Chromatography runs
spaces for viral clearance and present examples of how we were performed with varying parameters as described in
have applied these concepts to AEX processes. We first de- Table 2. Column equilibration was first performed using tris-
velop a formalized risk assessment to cover multiple AEX acetate equilibration buffers with varied compositions and
processes by using previous knowledge of the effects of pa- for varied durations. The feedstock pool was then loaded
rameters on viral clearance as well as the mechanism of onto the column, and collection of the flow-through fraction
action of the process. We then present experimental meth- was started when the OD280 reached 0.1 unit above baseline.
ods, which utilize that risk assessment as well as DOE tech- Protein was loaded to a total of 200 g mAb/mL resin, chang-
niques, to develop an understanding of process variability on ing the collection vessel after the initial 50 g mAb/mL resin
viral clearance. Using a highly fractionated experimental was loaded and allowing for analysis of both load densities
design, we show that the AEX can provide complete re- from each chromatography run. The column was then
moval of the endogenous RVLPs present in CHO-cell washed with three column volumes (CV) of equilibration
derived feedstocks over wide ranges of many process param- buffer, followed by regeneration with four CV of 0.5 N
eters. Finally, we present a full factorial design space in NaOH and three CV of 0.1 N NaOH. Aliquots of the product
which very high LRV values are obtained for three different pools and remaining feedstock pools were collected and
model viruses for all combinations of parameters within stored at 80 C. Extraction of viral genomic material was
ranges typical of AEX manufacturing processes. performed either using the Virus Mini Kit v2.0 on a BioRo-
bot EZ1 (Qiagen, Valencia, CA) or the MagAttract Virus
Materials and Methods Mini Kit v1.3 on a BioRobot M48 (Qiagen, Valencia, CA).
In both cases, extractions were followed by treatment with
RVLP design space RNase-free DNaseI (Strategene, Santa Clara, CA) to remove
Stock monoclonal antibody intermediate protein A pools any contaminating CHO-cell genomic DNA, and QPCR
were obtained from the mAb15 commercial-scale manufac- assays were performed as described previously.17
turing operations. For each chromatography run, pool condi-
tions were adjusted to varying levels as indicated in Table 2.
The pool was first adjusted to the appropriate pH using tris- Model virus design space
base or HEPES acid. The conductivity of each pool was then Stock monoclonal antibody intermediate cation chroma-
measured using a temperature-adjusted conductivity meter tography pools were obtained from mAb1 commercial-scale
752 Biotechnol. Prog., 2010, Vol. 26, No. 3

manufacturing operations. For each chromatography run, This strategy led to increased parameter ranges and consider-
load pools were conditioned to varying levels as indicated in ation of many categorical parameters, such as buffer and salt
Table 4. The pool was first adjusted to the appropriate pH compositions, which are generally locked for a specific pro-
using 1.5 M tris-base. The conductivity of each pool was cess but are varied between different processes. We used a
then measured using a temperature-compensated conductivity variation of an FMEA risk assessment tool, taking into
meter (SevenMulti, Mettler Toledo, Columbus, OH), and ei- account the severity that variability in a parameter may have
ther WFI or 5 M NaCl was added to adjust the conductivity on viral clearance and the probability that the parameter
to the desired level. The feedstock was then filtered through would be variable. The effects of each factor were first ana-
a 0.22 lm PES filter, and the protein concentration was lyzed using previous data, which indicated their effects on
determined by optical density at 280 nm (OD280). viral clearance. For instance, our laboratory has previously
For each run, a small-scale chromatography column (0.66- determined that feedstock conductivity, pH, and salt compo-
cm diameter, Bio-Chem Valve/OmniFit) was packed with sition can have effects on LRV values, and they interact
naive QSFF resin to 20 cm bed height and equilibrated with with one another,15,16 whereas variables such as the buffer
eight CV equilibration buffer (25 mM tris, NaCl, pH and compositions and pooling criteria were not observed to
conductivity adjusted to match load pool). The feedstock impact viral clearance. In addition, while considering the
pool was then loaded onto the column, and collection of the effects of certain parameters, we also took into account the
flow-through fraction was started when the OD280 reached mechanism of viral clearance by this process. The binding
0.1 unit above baseline. The feedstocks were loaded at vary- interactions that allow removal of viruses from mAb prod-
ing flow rates and to varying load densities as indicated in ucts are primarily due to electrostatic forces.21 This knowl-
Table 4, and the columns were then washed with three CV edge emphasizes the impact that variability in factors, which
equilibration buffer. On completion of the chromatography are related to electrostatic forces, such as conductivity and
run, the protein concentration of the product pool was deter- pH, may have on viral clearance. In addition, the mechanism
mined by OD280, and aliquots of this pool as well as the suggests that impurities that compete for the electrostatic
remaining feedstock pool were collected and stored at binding sites, such as anion host cell proteins, may also
80 C. The samples were subject to treatment with DNaseI impact viral clearance. This observation increases the impact
(Applied Biosystems, Foster City, CA), followed by extrac- of parameters such as impurity level, as well as load density
tion of viral genomic material using the EZ1 Virus Mini Kit since it relates directly to total impurity load. For each pa-
v2.0 on a BioRobot EZ1 (Qiagen, Valencia, CA). QPCR rameter, we also gauged the probability that it would vary
assays used to quantify X-MuLV, SV40, and MMV viral between different processes by comparing the many AEX
particles were performed as previously described.13,18,19 processes previously developed in-house. The results of our
risk assessment indicate that feedstock conductivity and load
density are the parameters, which are most likely to impact
viral clearance by AEX, while feedstock pH, impurity levels,
Results and Discussion
and buffer conductivity also may have some impact (Figure
Risk assessment 1). This risk assessment provides a useful tool for comparing
As a multidimensional study of input variables, design the viral clearance ability of different processes or predicting
space experiments have the potential to be quite extensive. the effects of process changes. Furthermore, the strategy pre-
When performed in product flow-through mode during mAb sented for the formation of a risk assessment provides the
production, AEX is relatively straightforward compared with opportunity to apply previous knowledge and experience
other bind-and-elute chromatography steps.20 However, we with a process to guide the development of design space
have identified 18 sources of variability for this unit opera- experiments.
tion. Using two-level screening designs to test variables at
the high and low ends of a range, full factorial experiments
to investigate all combinations require at least 2n runs, where Fractionated design space for RVLP clearance
n is the number of parameters. For AEX, testing all combi- Another way to reduce the overall number of runs neces-
nations of the 18 variables identified would require over sary to explore a design space with many parameters is
260,000 runs, an unfeasible number, especially for viral through the use of DOE experiments. DOE techniques
clearance experiments, which, due to the high costs of virus ensure that the efficiency of an experiment matches the
stocks, are generally expensive to perform. Therefore, we desired outcome, maximizing reliable information gained
utilized two tools to reduce the number of runs required to while minimizing redundancy between runs. For initial
perform design space studies: development of a formalized design space experiments, two-level screening designs are
risk assessment and utilization of design-of-experiments well-suited to give information on which parameters have
(DOE) techniques. main effects on the desired outcome, and often give informa-
The goal of a risk assessment is to provide an unbiased tion as to the interactions of those variables. For a given
ranking of variables so that experiments can be devised that number of runs, DOE approaches range from highly fractio-
focus on the factors most likely to have an impact on pro- nated designs to nonfractionated full factorial designs.
cess performance or have significant interactions with other Highly fractionated designs can test many different parame-
factors. Because of the robust nature of viral clearance by ters, providing information on main effects of most parame-
AEX with regard to both products and process parameters, ters but only providing interactions between a few. Because
we focused on constructing a risk assessment based on the many parameters can be tested with these designs, they rely
ranges of variables from the AEX processes for many differ- less on the risk assessment to choose which parameters
ent mAb products. These processes are often optimized on a should be tested. However, because many parameters are
product-specific basis for yield and impurity removal, and adjusted for each run, the experiments can be logistically dif-
many parameters can vary significantly between products. ficult, and since the results contain more information than
Biotechnol. Prog., 2010, Vol. 26, No. 3 753

Table 3. LRV Values from Fractional Factorial Design Space of


RVLP Removal
Load Density (g/L resin)
Run 50 200
1.* 3.7 3.7
2.* 3.5 3.5
3 3.3 3.3
4 3.3 3.3
5 3.4 3.4
6 3.4 3.4
7 3.3 3.3
8 2.9 2.9
9 3.2 3.2
10 3.2 3.2
11 3.2 3.2
12 3.2 3.2
13 3.2 3.2
14 3.2 3.2
15 3.0 3.0
16 3.0 3.0
17 3.0 3.0
18 3.0 3.0
19.* 3.4 3.4
20.* 3.5 3.5
* Center point runs.

Table 4. Model Virus Design Space Parameters


Parameters Low Center High
Conductivity* (mS/cm) 5.5 7.3 9.0
Load density (mg/mL resin) 10 80 150
pH* 7.5 8.0 8.5
Figure 1. Risk assessment for viral clearance by AEX per- Flow rate† (cm/h) 43 115 186
formed in product flow-through mode. * Conductivity and pH of feedstock and buffer were matched.

Process variables for a QSFF step performed in mAb flow- Accounts for contact time variation from production variations in
through mode are listed in the order of highest risk priority to both flow rate and bed height.
lowest risk priority with bars indicating the magnitude of the
risk priority numbers associated with each factor.
stocks and column temperature runs performed in pairs,
allowing two runs per day. In addition, we collected multiple
the familiar one-factor-at-a-time experiments, some research- flow-through pools for each run allowing for LRV values to
ers and regulatory personal may require the assistance of a be calculated for different load densities. This design strat-
statistician to help in analysis and interpretation of the data, egy allowed for 10 of the 18 AEX variables to be varied
particularly when using designs with complex aliasing struc- (Table 2), while two others, feedstock and buffer salt compo-
tures. Full factorial designs, on the other hand, provide both sitions, were held at worse case conditions for viral clear-
main effects and high-level interactions for all parameters. ance.16 We observed that virus titers in all of the product
For a given number of runs, fewer parameters can be tested pools were below the limit of quantification (LOQ) for the
with these designs, so although they rely heavily on the risk QPCR assay, indicating that all 20 runs achieved complete
assessment in choosing the most important factors, they are or near-complete removal of RVLPs for all load densities
logistically easier to carry out. In addition, since all combi- tested. LRV values for the runs, which were calculated from
nations of all the included parameters are tested, it is easy to the input titers and QPCR LOQ values (Table 3), varied
grasp the full meaning of the resulting data. Our experimen- from 2.9 to 3.7, with variations primarily stemming from
tal strategy included an iterative approach, including a highly the QPCR assays of the feedstocks. Since complete removal
fractionated design to test many of the AEX parameters, re- was observed for all of the runs, the data cannot be used to
evaluation of the risk assessment based on the outcome of determine the effects of the varied chromatography parame-
that data, and then a full factorial design to test the most sig- ters. Re-evaluation of the risk assessment based on these
nificant parameters. data did not result in changes to the previous order of
We first investigated the effects of many AEX variables parameters. However, the data does indicate that variation of
using a highly fractionated two-level screening design. To even the most important AEX variables does not have a
minimize the cost associated with this preliminary experi- measurable impact on its ability to remove RVLPs. Further-
ment, we chose to test removal of in-process RVLPs using a more, the robust removal of RVLPs over these broad ranges
protein A pool as a feedstock. This methodology allows for of AEX parameters establishes a minimal design space
the determination of LRV values by the AEX process with- where consistent RVLP removal can be achieved by this
out the need for expensive virus spiking, although the maxi- process.
mum LRV values which can be obtained are limited by the
starting RVLP titers. We used a fractional factorial design to
test nine different parameters over large ranges. The design Full factorial design space for model virus clearance
consisted of 16 runs plus four center point runs, and the runs The design space from the first experiment provides strong
were grouped to form a split plot design with similar feed- evidence that viruses can be removed over wide ranges of
754 Biotechnol. Prog., 2010, Vol. 26, No. 3

Table 5. LRV Values from Full Factorial Design Space of Model mammalian-cell derived mAb products. Through the system-
Virus Removal atic development of a risk assessment and two carefully
Model Virus designed experiments, we have shown that the AEX process
Run X-MuLV SV40 MMV can remove the endogenous RVLP impurities over ranges of
1.* 5.9 5.4 5.7
parameters much wider than those typically found in produc-
2 5.7 4.9 5.4 tion processes, and we have provided ranges of process vari-
3 5.8 5.4 5.5 ables where AEX can also be assured of providing complete
4 6.1 5.5 5.7 or near-complete removal of potential adventitious virus
5 5.7 5.5 5.7 contaminants.
6 5.8 5.1 5.5
7 5.7 5.3 5.5
8 6.0 5.6 5.8
9 5.8 5.3 5.6 Literature Cited
10 6.0 5.7 5.7 1. ICH (International Conference on Harmonization). Pharmaceuti-
11 5.6 4.9 5.3
cal Development Revision 1 Q8 (R1). Available at: http://
12 6.0 5.9 5.8
13 5.6 5.2 5.5
www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatory-
14 6.1 5.6 5.8 Information/Guidances/ucm073507.pdf; 2009.
15 5.6 4.9 5.3 2. Harms J, Xiangyang W, Kim T, Yang X, Rathore AS. Defining
16 5.7 5.1 5.4 process design space for biotech products: case study of Pichia
17 5.9 5.7 5.7 pastoris fermentation. Biotechnol Prog. 2008;24:655–662.
18.* 5.8 5.5 6.0 3. Li F, Hashimura Y, Pendleton R, Harms J, Collins E, Lee B. A
systematic approach for scale-down model development and
* Center point runs.
characterization of commercial cell culture processes. Biotech-
nol Prog. 2006;22:696–703.
4. Nail SL, Searles JA. Elements of quality by design in develop-
ment and scale-up of freeze-dried parenterals. BioPharm Int.
many variables and that none of the variables appear to com- 2008.
pletely disrupt viral clearance within those ranges. However, 5. Anderson KP, Low MA, Lie YS, Keller GA, Dinowitz M. En-
the limited LRV values obtained by that methodology are dogenous origin of defective retrovirus like particles from a
not optimal to determine the full potential of the process to recombinant Chinese hamster ovary cell line. Virology. 1991;
remove viruses or the effects that variables may have on 181:305–311.
6. Brorson K, de Wit C, Hamilton E, Mustafa M, Swann PG, Kiss
higher levels of clearance, and the parameter ranges tested R, Taticek R, Polastri G, Stein KE, Xu Y. Impact of cell culture
are wider than those found in actual manufacturing proc- process changes on endogenous retrovirus expression. Biotech-
esses. We therefore performed a second experiment using a nol Bioeng. 2002;80:257–267.
full factorial design to determine removal of spiked model 7. Lieber MM, Benveniste RE, Livingston DM, Todaro GJ. Mam-
viruses over parameter ranges more representative of those malian cells in culture frequently release type C viruses. Sci-
used in actual manufacturing settings (Table 4). The model ence. 1973;182:56–59.
viruses used for this experiment, X-MuLV, SV40, and 8. Garnick RL. Experience with viral contamination in cell culture.
Dev Biol Stand. 1996;88:49–56.
MMV, represent a broad range of virus classes including 9. CBER(Center for Biologics Evaluation and Research, Federal
both enveloped and nonenveloped, and RNA and DNA con- Drug Administration). Points to consider in the manufacture and
taining viruses. To limit the number of runs while maximiz- testing of monoclonal antibody products for human use. Avail-
ing the number of variables tested, some parameters were able at: http://www.fda.gov/downloads/BiologicsBloodVaccines/
varied together. For instance, the conductivity of the feed- GuidanceComplianceRegulatoryInformation/OtherRecommenda-
stock and equilibration buffer were matched for each run, as tionsforManufacturers/UCM153182.pdf; 1997.
were the pH of the feedstock and equilibration buffer, a 10. EMEA(European Medicines Agency). Guideline on virus safety
evaluation of biotechnological investigational medicinal prod-
methodology previously used to test these parameters.15 In ucts. Available at: http://www.emea.europa.eu/pdfs/human/bwp/
addition, bed heights and flow rates were combined into con- 39849805enfin.pdf; 2008.
tact times, which were then included experimentally as vari- 11. ICH(International Conference on Harmonization). Viral safety
ation in flow rate. In this way, seven different AEX evaluation of biotechnology products derived from cell lines of
parameters were combined into four factors in the full facto- human or animal origin Q5A. Available at: http://www.fda.gov/
rial design, which, with the addition of two center point downloads/Drugs/GuidanceComplianceRegulatoryInformation/
Guidances/ucm073454.pdf; 1999.
runs, were studied in an 18-run experiment. For all of those
12. Norling L, Lute S, Emery R, Khuu W, Voisard M, Xu Y, Chen
runs, we observed complete or near-complete removal of all Q, Blank G, Brorson K. Impact of multiple re-use of anion-
three model viruses to levels below the LOQ’s for their re- exchange chromatography media on virus removal. J Chroma-
spective assays (Table 5). Calculation of LRV’s using the togr A. 2005;1069:79–89.
input titers and assay LOQ’s resulted in LRV’s of 5.6 to 13. Shi L, Chen Q, Norling LA, Lau AS, Krejci S, Xu Y. Real time
6.1 for X-MuLV, 4.9 to 5.9 for SV40, and 5.3 to 6.0 quantitative PCR as a method to evaluate xenotropic murine
for MMV, with variations again due to the QPCR assay. leukemia virus removal during pharmaceutical protein purifica-
tion. Biotechnol Bioeng. 2004;87:884–896.
These data indicate that this AEX process is highly robust
14. Strauss DM, Lute S, Brorson K, Blank GS, Chen Q, Yang B.
over the design space and that there are no practically signif- Removal of endogenous retrovirus-like particles from CHO-cell
icant effects or interactions of any of the parameters tested derived products using Q sepharose fast flow chromatography.
over relevant ranges. Biotechnol Prog. 2009;25:1194–1197.
15. Curtis S, Lee K, Blank GS, Brorson K, Xu Y. Generic/matrix
evaluation of SV40 clearance by anion exchange chromatogra-
Conclusions phy in flow-through mode. Biotechnol Bioeng. 2003;84:179–
186.
We have developed strategies to create design spaces for 16. Strauss DM, Gorrell J, Plancarte M, Blank GS, Chen Q, Yang
the removal of viruses during downstream purification of B. Anion exchange chromatography provides a robust,
Biotechnol. Prog., 2010, Vol. 26, No. 3 755

predictable process to ensure viral safety of biotechnology prod- 20. Fahrner RL, Knudsen HL, Basey CD, Galan W, Feuerhelm D,
ucts. Biotechnol Bioeng. 2009;102:168–175. Vanderlaan M, Blank GS. Industrial purification of pharmaceuti-
17. De Wit C, Fautz C, Xu Y. Real-time quantitative PCR for retro- cal antibodies: development, operation, and validation of chro-
virus-like particle quantification in CHO cell culture. Biologi- matography processes. Biotechnol Genet Eng Rev. 2001;18:301–
cals. 2000;28:137–148. 327.
18. Shi L, Ho J, Norling LA, Roy M, Xu Y. A real time quantita- 21. Strauss DM, Lute S, Tebaykina Z, Frey DD, Ho C, Blank GS,
tive PCR-based method for the detection and quantification of Brorson K, Chen Q, Yang B. Understanding the mechanism of
simian virus 40. Biologicals. 1999;27:241–252. virus removal by Q sepharose fast flow chromatography during
19. Zhan D, Roy MR, Valera C, Cardenas J, Vennari JC, Chen JW, the purification of CHO-cell derived biotherapeutics. Biotechnol
Liu S. Detection of minute virus of mice using real time quanti- Bioeng. 2009;104:371–380.
tative PCR in assessment of virus clearance during the purifica-
tion of Mammalian cell substrate derived biotherapeutics.
Biologicals. 2002;30:259–270. Manuscript received Nov 13, 2009.

S-ar putea să vă placă și