Biomanufacturing Processes

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Eng. Life Sci. 2011, 11, No.

4, 443–451 443

Thorsten Peuker1 Technical Report


Alexandra Bogner2

1
Sartorius Stedim Biotech
Equipment design and facility layout
GmbH, Göttingen, for flexible biomanufacturing processes
Germany
2
Sartorius Stedim Systems In process development and manufacturing, the biopharmaceutical industry
GmbH, Melsungen, requires high flexibility in its production facilities. These suites must be capable of
Germany producing clinical material or even drug substances for the market. Several
products are manufactured in parallel or shortly after one another, which is
challenging especially for the equipment. Product titers are increasing due to
improved cell line performances, and personalized medicine will lead to tailor-
made drugs for a smaller group of patients. The resulting reduced upstream
volumes enhance the opportunities for complete single-use manufacturing trains.
These requirements must be considered already during the design phase of the
manufacturing facility. Engineering efforts have to be reduced as much as possible
in order to reduce cost and timelines. Generic platform concepts for the overall
process enable a shorter execution time as well as more efficient qualification
procedures. In this contribution, the focus is on production processes for
monoclonal antibodies (mAb) as a major product class of the biopharma-
ceutical industry. Two case studies introduce process platform concepts inte-
grating single-use equipment for an existing building and for a greenfield
facility.

Keywords: Bioprocesses / Facility design / Monoclonal antibodies / Process platform /


Single-use and hybrid equipment

Received: September 30, 2010; revised: February 2, 2011; accepted: February 24, 2011

DOI: 10.1002/elsc.201000174

1 Introduction emerging markets, and a continuously growing population will


primarily fuel the global mAb production demand.
Antibodies are one of the most efficient weapons of the Modern biopharmaceutical production has changed in the
immune system. With up to 1011 molecule modifications, the last few years, due to the changes in overall process perfor-
variety in antibody specificity is nearly endless, which is mances on the one hand and the availability of new materials
necessary for the ability to bind millions of various antigens. and components on the other hand. There is a trend toward
The interaction between a monoclonal antibody (mAb) and its the replacement of stainless-steel equipment and the usage of
antigen epitope is highly specific, excluding all other epitopes. single-use equipment since implementation of the latter is easy
This unique characteristic of mAb opened up the door for the and very often much more cost efficient. Single-use equipment
development of new diagnostic methods and therapeutics, is already established in several unit operations and will be
especially for cancer therapy [1, 2]. mAb generated revenues of accepted in others in the near future [5–7]. In addition, it is
approx. $35 billion in 2009, accounting for 28% of the global obvious that flexible multiproduct facilities have different
biotech market [3]. The market size of mAb will further requirements than large-scale manufacturing of bulk drug
expand and replace some of the small molecules in the top-ten substances, which are produced in stationary and hard-piped
list of bestselling products, as predicted by EvaluatePharma in stainless-steel equipment. Time-to-market and cost pressure
its World Preview 2016 report [4]. A full R&D pipeline, on the biopharmaceutical industry are the main issues to solve,
in order to be ahead of the growing competition on the
Correspondence: Dr. Thorsten Peuker (thorsten.peuker@sartorius-
market.
stedim.com), Sartorius Stedim Biotech GmbH, August-Spindler-Street For the development of new drug substances, a strict and
11, 37079 Göttingen, Germany. regulated clinical test procedure has to be followed. If a drug
Abbreviations: API, active pharmaceutical ingredient; CIP, cleaning in candidate was successful in toxicological studies and the
place; HVAC, heating, ventilating, and air conditioning; SIP, ster- preclinical and clinical phases I and II, the material for phase
ilization in place III must already originate from the same equipment that is

& 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim http://www.els-journal.com
444 T. Peuker and A. Bogner Eng. Life Sci. 2011, 11, No. 4, 443–451

designated for market supply [8]. Figure 1 shows a typical 2 Design of generic process platforms
timeline of an active pharmaceutical ingredient (API)
development and the corresponding engineering phases A biopharmaceutical manufacturing concept is more than the
for facility and equipment. Since the engineering, construc- sum of its elements.
tion, and qualification phases for customized production The design of the purification process is influenced by the
plants sometimes take more than 3 years, these activities physicochemical properties of the product and the nature of
have to start almost at the same time as drug develop- the process- and product-related impurities [9]. Despite small
ment. As a consequence, companies have to decide for variations in the chemical nature of mAb, many companies
major investments at an early stage (preclinical) where no have defined platform purification processes [10–12]. Each
guarantee exists that the product will be successful. unit operation is made of standardized technological modules
Suppliers should support the manufacturing industry by (e.g. sensors, storage bags, mixing devices, filters, transfer sets,
predefined configurable unit operations or even generic plat- connectors, sampling devices, etc.). Major benefits of process
form processes. This will reduce time and engineering efforts platforms are savings in time and cost, process optimization,
during design and construction of such development and and security of supply for single-use components [8, 13, 14].
manufacturing plants and hence could enable postponement In order to be fast and efficient in the design and execution of
of the investment decision until a later phase without loss of projects, these technological modules should be well defined
time. and should not have to be changed or even newly developed.

Figure 1. Timelines of drug development and


corresponding engineering phases for facility
and equipment.

Figure 2. Process platforms available for


mAb production in clinical phases and
production.

& 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim http://www.els-journal.com
Eng. Life Sci. 2011, 11, No. 4, 443–451 Equipment design and facility layout 445

There are different process platforms available for mAb, 2.1 mAb manufacturing process
matching the varying product amounts in the different clinical
phases (Fig. 2). A generalized mAb platform process is Currently, the most efficient expression systems are Chinese
described in detail (Fig. 3) [10, 11]. hamster ovary (CHO) cells and mouse myeloma cell lines.
Main criteria in the selection of cell lines are the human

Figure 3. (A) Flow diagram for a mid-scale mAb upstream process with corresponding single-use equipment. (B) Flow diagram for a mid-
scale mAb downstream process with corresponding single-use equipment.

& 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim http://www.els-journal.com
446 T. Peuker and A. Bogner Eng. Life Sci. 2011, 11, No. 4, 443–451

glycosylation pattern, a stable expression system of the target product purity or yield. In the purification of antibodies, the
protein, and an as high as possible specific production rate molecular weight cut-off used for ultrafiltration/diafiltra-
[15]. The mAb titers usually reach 2–3 g/L. Production levels of tion (UF/DF) varies in the range of 30–500 kDa, depending
5 g/L have already been reported for highly optimized on whether the antibody shall be retained or not. Finally, a
mammalian cell culture systems, and titers of >10 g/L are microfiltration step under stringent conditions ensures
expected within the next decade [16]. This development sterility of the final product, as required [15]. Single-use
predicts lower upstream volumes in the future and benefits the equipment is available for all filtration steps, except for
usage of single-use bioreactors for the production scale, ultrafiltration/diafiltration, in large-scale production.
which is limited to 2000 L. Downstream processes of mAb
production were not able to keep pace with the rapidly
increasing titers in fermentation; thus, the production bottle- 2.2 Single-use equipment and hybrid solutions
neck has shifted from upstream to downstream. In particular,
the load capacities of chromatographic and membrane mate- So far, most of the necessary unit operations in mid-scale or
rials need to be improved [17]. The economy of a mAb large-scale production processes are made of reusable stainless-
product is intrinsically tied to the titer and the overall recovery steel equipment. This material has major advantages, such as
rate in the downstream process. Other significant benchmarks mechanical robustness and chemical and temperature resis-
in an mAb production process are protein quality and purity. tance; however, high capital investments and less flexibility are
The quality of any biopharmaceutical product is defined by the its drawbacks. For reusable equipment, well-established design
production process, which is basically determined by the criteria and experienced engineers are available to build
process equipment, the sequence of unit operations, and the production facilities almost everywhere around the world. But
operation parameters [15]. due to long lead items and extensive construction and quali-
The first step after cell culture is separation of the cells/ fication efforts, it takes more than 5 years to bring such a new
clarification of the harvest broth. Centrifugation is the most facility into operation.
common method for harvesting operations. Depth filters are During the last decade, single-use equipment has been
an adequate single-use alternative or are used in combination increasingly implemented [23, 24]. This is due to the fact that
with a centrifuge. Which approach turns out as the method new materials are available that are considered as save and
of choice depends on the final cell density in the culture reliable by the US and European Pharmacopeia (ICH Q7A:
broth, the turbidity, and the cell viability. A subsequent Protein Good Manufacturing Practice Guide for Active Pharmaceutical
A chromatography step serves as capturing step and as key Ingredients) [25]. In order to make maximum use of the
volume reduction step. The high purity that can be achieved benefits that disposables can bring to the manufacturing of
makes the concept of a platform process suitable for mAb. biopharmaceuticals, the challenge is to design and implement
Following polishing steps have to remove the remaining application-specific integrated process solutions that, with
amounts of host cell protein contaminants, DNA, leached respect to process engineering, automation and control, follow
Protein A, endotoxins, or other process-related impurities the same principles as conventional process designs. It is
[11, 18]. The main drawback of this step is the expensive important to accept that disposables have certain limitations,
Protein A chromatography medium at approx. 10 000 US$/L. but that they can be intelligently integrated in new and existing
A promising alternative for antibody capturing is cation process designs. How many disposables to use and where to
exchange chromatography [19, 20]. The low pH conditions of use them are issues that have to be evaluated from various
the Protein A pool benefit a subsequent low-pH viral inacti- aspects and carefully analyzed with respect to application and
vation step [19]. Two additional chromatography steps are customer-specific need. In addition, the possibility of a
typically employed in an mAb platform process, with the aim combination of reusable and single-use technologies (i.e.
of further purification and polishing. Common chromato- ‘‘hybrid technology’’) must be considered. Currently, dispo-
graphy methods are cation exchange, anion exchange, hydro- sables are mainly used during scale-up and clinical produc-
phobic interaction, hydroxyapatite, and size exclusion chro- tion. In commercial manufacturing, the use of disposables is
matography. Single-use membrane chromatography found its still mostly limited to tubings and filter cartridges. However,
place particularly in purification processes for the removal of with the expansion of disposable technologies into the area
impurities and contaminants [21]. In order to comply with the of commercial production, the need of integrated single-use
virus reduction corresponding to the FDA Q5A document, the process designs becomes absolutely obvious. Although in
mAb purification process includes a virus filtration step the past single-use products were recognized as useful
[11, 22] (www.fda.gov/downloads/RegulatoryInformation/ components in the manufacturing of biopharmaceuticals,
Guidances/UCM129101.pdf). Different filtration techniques nowadays there are more and more processes that are almost
are established in the mAb purification process. Microfiltration completely designed with single-use equipment. Even in
membranes are available with pore sizes between 0.01 and mid-scale processes (r2000 L upstream volume), several
10 mm. They are used between the main unit operations steps single-use package units and in-process fluid handling
to protect the chromatography resins or membranes or to systems are used, and their usage will certainly increase in
minimize bioburden and endotoxin levels during the purifi- the future. Currently, there are efforts to standardize single-
cation process. Ultrafiltration is used to concentrate the use process equipment in the same way as stainless-steel
product or for buffer exchange in order to condition the bioprocess equipment: DIN EN (German/European Indus-
load for the next column. Diafiltration enhances either try Standard), ASME BPE (American Society for Mechanical

& 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim http://www.els-journal.com
Eng. Life Sci. 2011, 11, No. 4, 443–451 Equipment design and facility layout 447

Engineering BioProcessing Equipment), and ISPE (Inter- errors. Industrial standards are available for the overall auto-
national Society for Pharmaceutical Engineering). mation system but also for almost every package unit. The
In general, there are a number of issues that have to be majority of automations tasks for this kind of plant are
solved before implementing single-use equipment. Process required for cleaning in place (CIP) and sterilization in place
compatibility, process efficacy, batch volume, filling and (SIP) of the reusable equipment.
discharge, degree of agitation (power input), operating pres- Automation solutions, ranking from dedicated local
sure and temperature, measurement of process values, material controllers to high-level system integration using programmable
handling/space requirements, and environmental, health, and logic controller/supervisory control and data acquisition (PLC/
safety aspects are of utmost importance [26]. SCADA) or distributed control systems (DCS), cannot be fully
applied for single-use parts yet. Common standards need to be
adjusted considering the different natures of both technologies.
2.3 Advantage of closed disposable systems By applying more and more single-use products and equipment,
CIP and SIP are no longer an issue. On the other hand, the unit
Biopharmaceutical production processes require sterile and closed operation approach discussed earlier will help to combine unit
containments in order to ensure process and user safety. Single- operations by using platform technology connection methods
use equipment can support this by predefined, configurable and well-established supervisory control and data acquisition
disposable solutions. Such single-use systems must fulfill the platforms. The digital control unit (DCU)-4 system of Sartorius
needs of the different unit operations. Reducing the complexity of Stedim Biotech is one example of applying well-established
predefined, configurable systems by maintaining flexibility automation systems to single-use and hybrid processes. It serves
through different configurations will be a key success factor mixing systems, cell cultivation systems, and filtration systems at
perceived by the end users. Several suppliers already launched all stages. All these unit operations can now be equipped with
such closed disposable solutions or systems (e.g. Sartorius Stedim either reusable or single-use components, relying on a well-
Biotech: FlexAct; Millipore: Mobius; GE: ReadyToProcess). Once established automation platform.
it is implemented in the process cycle, security of supply and fast Disposable sensors that are fully integrated into the
in-time delivery are the main constraints of the industry. respective single-use containment to control important
process parameters for each unit operation (e.g. dissolved
oxygen (DO), pH, conductivity, etc.). However, if users want
2.4 Automation and instrumentation to integrate classical sensors, this is also possible by applying
autoclavable connection systems.
Traditional facilities with stainless-steel equipment require a Considering that single-use equipment leads to more
high automation degree. There are only few procedures with interference of the personnel, integration into a plant-wide
human interaction, e.g. inoculation or sampling. This leads to manufacturing execution system (MES) is a prerequisite for a
high process safety and less losses of batches due to operational safe and documented production process.

Figure 4. Facility layout for a mid-scale mAb, single-use mAb manufacturing process.

& 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim http://www.els-journal.com
448 T. Peuker and A. Bogner Eng. Life Sci. 2011, 11, No. 4, 443–451

Figure 5. (A–C) Layout for large-scale mAb manufacturing


building.

2.5 Facility requirements for implementation of investment economical, reduction of clean room demands is
single-use equipment essential. On the other hand, employing single-use equipment
requires more space for movement and handling. In this
The facility design must be according to the current Good article, we will introduce two typical scenarios as case studies.
Manufacturing Practice (cGMP). However, most importantly, In general, closed containments can be assumed as safe.
it should be process oriented and follow well-established However, since there are always interactions (e.g. sampling or
design principles. It has to consider material and personnel final processing), dedicated clean room environments have
flows and can be of modular design. In order to make the to be considered. At least ISO 8 air classification must be

& 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim http://www.els-journal.com
Eng. Life Sci. 2011, 11, No. 4, 443–451 Equipment design and facility layout 449

Figure 6. Flow diagram for a large-scale mAb production process.

provided if open-top mixing is used. If closed-bag systems are systems by Sartorius) or RAFT (rapid aseptic fluid transfer).
applied for raw material addition and liquid mixing (e.g. We have considered class D environments for cell cultivation
FlexAct BP), the air classification can be reduced to controlled (seed culture will be done in class C) and harvesting, and for
but nonclassified (CNC) area. Reduced heating, ventilating previrus downstream steps. Postvirus steps in downstream will
and air conditioning (HVAC) demands lead to cost savings for be carried out in a class C clean room (if required, the final
ongoing operational costs [26]. filling can be done under laminar flow in class B).
For the process design, it is important to realize that making
use of standardized solutions in particular for single-use
3 Case studies components (bags, transfer sets, filters, etc.) will become more
important in the future. The main issues to overcome are
3.1 Retrofitting of an existing facility for process security of supply and especially supply chain strategies for
development these consumables.

If an existing facility should be used for new processes,


retrofitting is required [13]. In this case study, we will focus on 3.2 Greenfield production facility for market supply
a new mAb process at a scale of 200 L batch volume and 5 g/L
product titer. The process was designed based on the generic Greenfield production facilities have other requirements
principles that are commonly accepted by the industry [14]. than existing facilities. Besides the process equipment, utility
Figure 3(A) and (B) shows the process flow diagram of a demands, HVAC requirements, and even construction work
complete single-use manufacturing train in upstream and have to be considered, which leads to longer project timelines.
downstream as well as the corresponding equipment. The In this contribution, we will focus only on the process
complete process can be arranged on one floor, with separate equipment for the production of mAb and the architecture of a
flows for personnel and material (Fig. 4). Buffer and media new building.
preparation will be done in a CNC environment. Intermediate A manufacturing building for larger-scale production
storage can be realized in a cold room until the material is should contain five stories in order to distribute all necessary
required at the point of use. Palletankss will be stored outside infrastructures. Figure 5 shows the layout for a greenfield
the process rooms in order to reduce clean room space. facility. On the first two floors (levels 1 and 0), administration
Transfer inside the process rooms is done by aseptic transfer and technical areas are located. The next two floors (levels 2
systems, e.g. SART SystemTM (single-use aseptic transfer and 3) are dedicated to process equipment, and the last floor

& 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim http://www.els-journal.com
450 T. Peuker and A. Bogner Eng. Life Sci. 2011, 11, No. 4, 443–451

(level 4) is used for HVAC systems. A technical shaft in the as media or buffer preparation, as it is already reality in the
center of the building provides space for piping and distri- food industry. This will also lead to lower costs and overall
bution systems for utilities, media, and energy. It will also improved processes.
support all infrastructural needs that should be not placed in a
clean room.
The process equipment follows, as for the mid-scale case, The authors have declared no conflict of interest.
well-established unit operations [14]. Hybrid solutions are
combinations of single-use and stainless-steel equipment. The
process design was done based on the following assumptions: 5 References
A scale-up factor of 1:5 was considered for the bioreactors; in
total, it was planned to execute 21 batches per year with an [1] Michl, D., Heinemann, A., Medizinische Biotechnologie in
overall product yield of 4.4 kg mAb per batch. In an annual Deutschland 2009. Boston Consulting Group Report 2009,
operating time of 330 days, we could realize a minimum cycle 18–38.
time of 8.2 days, which would result in a maximum of 37 [2] Elgert, K. D., Immunology: Understanding the Immune
batches per year. The flow diagram for the complete process is System, 2nd Edn, Wiley-Blackwell, Hoboken 2009,
shown in Fig. 6. The buffer and media preparation is located pp. 139–178.
on the second level in order to make maximum use of gravi- [3] Monoclonal Antibodies in Cancer Therapy: World Market
metric flow. The distribution of buffer and media will be done Prospects 2010–2025, visiongain, London 2010, pp. 28–38.
in two different ways. Bags will be used as much as possible, [4] World Preview 2016, EvaluatePharma, 2010.
but there is also still a need for hard-piping. Especially, initial [5] Brecht, R., Disposable bioreactors: maturation into
media load for the production bioreactors (media filling via pharmaceutical glycoprotein manufacturing. in: Scheper, T.,
media filter into bioreactor) can only be realized in a certain Eibl, R., Eibl, D. (Eds.), Disposable Bioreactors, Springer,
time frame with this approach. The upstream process and the Berlin 2009, pp. 1–31.
downstream process are arranged on the first level of the [6] Gottschalk, U., Disposables in downstream processing. in:
building. Both streams are separated by the technical shaft. Scheper, T., Eibl, R., Eibl, D. (Eds.), Disposable Bioreactors,
For feeding purposes in fed-batch processes, Palletankss Springer, Berlin 2009, pp. 171–184.
with 500-L 3-D bags are considered. Feeding from the media [7] Eibl, R., Eibl, D., Application of disposable bag bioreactors
bag into the bioreactor for fed-batch operation is conveyed in tissue engineering and for the production of
by a peristaltic pump. The required media filter and valve therapeutic agents. Adv. Biochem. Eng. Biotechnol. 2009, 112,
groups are shared with the bioreactor. In order to clean and 183–207.
steam the pipework, SIP and CIP/rinse steps are carried out [8] Werner, R. G., Economic aspects of commercial manu-
in between operations. For this kind of hybrid process facture of biopharmaceuticals. J. Biotechnol. 2004, 113,
design, it is important to allow for enough space for the 171–182.
movement of Palletankss with 3-D bags (Flexel) and to [9] Humphreys, D. P., Bowering, L., Production of antibody Fab’
consider handling of filled 2-D bags (Flexboys). fragments in E. coli. in: An, Z. (Ed.), Therapeutic Monoclonal
Antibodies: From Bench to Clinic, Wiley, Hoboken 2009,
pp. 589–622.
4 Concluding remarks [10] Kelley, B., Blank, G., Lee, A., Downstream processing of
monoclonal antibodies: current practices and future oppor-
Single-use equipment has changed the process design in tunities. in: Gottschalk, U. (Ed.), Process Scale Purification of
biopharmaceutical production in the last years. The resulting Antibodies, Wiley, Hoboken 2009, pp. 1–24.
new demands have to be taken into account during the design [11] Shukla, A. A., Hubbard, B., Tressel, T., Guhan, S., Low, D.,
of processes and even whole facilities. Platform processes and Downstream processing of monoclonal antibodies – appli-
facility layouts should support producers in postponing their cation of platform approaches. J. Chromatogr. B 2007,
investment decision until their drug candidate has shown first 848, 28–39.
positive results in clinical trials. The benefit of these platform [12] Hagel, L., Jagschies, G., Sofer, G., Handbook of Process
processes is to significantly reduce the upfront engineering and Chromatography – Development, Manufacturing, Validation
design work. and Economics, 2nd Edn, Academic Press, London 2008,
The developments in single-use technologies will speed up pp. 1–21.
in the next years. In the future, they will increasingly penetrate [13] Ravisé, A., Cameau, E., De Abreu, G., Pralong, A., Hybrid
entire biopharmaceutical manufacturing processes. Conse- and disposable facilities for manufacturing of biopharma-
quently, security of supply of these product contact assemblies ceuticals: pros and cons. Adv. Biochem. Eng. Biotechnol. 2009,
is one of the major issues. Worldwide supply chain concepts, 115, 185–219.
with manufacturing sites all over the world, are a prerequisite [14] Kelley, B., Industrialization of mAb production technology –
to fulfill the highest level of security of supply for single-use the bioprocessing industry at a crossroads. mAbs 2009, 1,
systems and components. 443–452.
Reducing human interactions is one of the major require- [15] Strube, J., Sommerfeld, S., Lohrmann, M., Process develop-
ments for the next years. By applying more automation, in ment and optimization for biotechnology production –
future there may be even fully automated unit operations such monoclonal antibodies, in: Subramanian, G. (Ed.), Biose-

& 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim http://www.els-journal.com
Eng. Life Sci. 2011, 11, No. 4, 443–451 Equipment design and facility layout 451

paration and Bioprocessing, Wiley-VCH, Weinheim 2007, pp. [21] Curling, J., The development of antibody purification tech-
65–99. nologies. in: Gottschalk, U. (Ed.), Process Scale Purification of
[16] Hagel, L., Jagschies, G., Sofer, G., Handbook of Process Antibodies, Wiley, Hoboken 2009, pp. 25–52.
Chromatography – Development, Manufacturing, Validation [22] FDA Q5A Guidance Document: viral safety evaluation of
and Economics, 2nd Edn, Academic Press, London 2008, biotechnology products derived from cell lines of human or
pp. 23–39. animal origin, Federal Register, 63 (185), 1998, p. 51074.
[17] Werner, R. G., The development and production of [23] 6th Annual Report and Survey of Biopharmaceutical
biopharmaceuticals: Technological and economic success Manufacturing Capacity and Production – A Study of
factors. BioProcess Int. 2005, 3, 6–15. Biotherapeutic Developers and Contract Manufacturing
[18] Hagel, L., Jagschies, G., Sofer, G., Handbook of Process Organizations, Bioplan Associates Inc., 2009.
Chromatography – Development, Manufacturing, Validation [24] Rao, G., Moreira, A., Brorson, K., Disposable bioprocessing:
and Economics, 2nd Edn, Academic Press, London 2008, The future has arrived. Biotechnol. Bioeng. 2009, 102,
pp. 41–80. 348–356.
[19] Lain, B., Cacciuttolo, M. A., Zarbis-Papastoitsis, G., Devel- [25] Goldstein, A., Perrone, P., Method for implementing dispo-
opment of a high-capacity mAb capture step based on sables into a bioprocess facility. ISPE Knowledge Brief 2010.
cation-exchange chromatography. BioProcess Int. 2009, 3, [26] Martin, L., Bartow, M., Schaidle, B., Tuttle, G., Sandstrom,
26–34. C., Managing large volume, single-use disposables in
[20] Morrow, K. J., Frontiers in bioprocessing: 2007. in: Langer, biopharmaceutical manufacturing. in: Langer, E. S. (Ed.),
E. S. (Ed.), Advances in Large-Scale Biopharmaceutical Advances in Large-Scale Biopharmaceutical Manufacturing
Manufacturing and Scale-Up Production, 2nd Edn., ASM and Scale-Up Production, ASM Press, Washington DC 2007,
Press, Washington DC 2007, pp. 1–34. pp. 737–766.

& 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim http://www.els-journal.com

You might also like