搜档网
当前位置:搜档网 › Trends_in_Biotech-Single-Use

Trends_in_Biotech-Single-Use

Special Issue:Celebrating30years of biotechnology

Single-use disposable technologies for biopharmaceutical manufacturing

Abhinav A.Shukla1and Uwe Gottschalk2

1KBI Biopharma,Durham,NC,USA

2Sartorius-Stedim Biotech,Goettingen,Germany

The manufacture of protein biopharmaceuticals is con-ducted under current good manufacturing practice (cGMP)and involves multiple unit operations for up-stream production and downstream puri?cation.Until recently,production facilities relied on the use of rela-tively in?exible,hard-piped equipment including large stainless steel bioreactors and tanks to hold product intermediates and buffers.However,there is an increas-ing trend towards the adoption of single-use technolo-gies across the manufacturing process.Technical advances have now made an end-to-end single-use manufacturing facility possible,but several aspects of single-use technology require further improvement and are continually evolving.This article provides a perspec-tive on the current state-of-the-art in single-use technol-ogies and highlights trends that will improve performance and increase the market penetration of disposable manufacturing in the future.

The advent of single-use technologies

Many different factors have combined to encourage the current surge of interest in single-use or disposable tech-nologies for biopharmaceutical manufacturing(Table1). The production of all drugs is tightly controlled by cGMP guidelines to reduce or prevent bioburden,that is,contam-ination of the product stream with bacteria,viruses,and other potentially harmful adventitious agents[1].A key aspect of cGMP manufacturing plants is the cleaning, between runs,of vessels and other equipment that comes into contact with the product[2].This is a laborious and time-consuming requirement that means the manufactur-ing process must be taken off line,and the cleaning proce-dure must be extensively validated and documented[3]to demonstrate the elimination of bioburden and residual product,the latter to prevent cross-contamination in mul-tiproduct manufacturing plants.Single-use technologies were?rst introduced as a means to avoid cleaning and validation requirements while simultaneously reducing the risk of contamination,particularly in cell culture pro-cesses.Single-use technologies also reduce the need for utilities such as the steam used to sterilize product contact equipment by steaming-in-place(SIP)before each use.The environmental bene?ts of the reduced energy demand can be said to outweigh the increase in solid waste generated by the disposal of single-use devices[4].

The economic bene?ts of single-use equipment are not restricted to the faster campaign turnaround times(for the same and different products)once a production facility is operating.Indeed,the cost bene?ts can be experienced at the beginning of clinical development for a product because single-use equipment reduces the time required to get a facility up and running,and the apparatus tends to be less expensive than stainless steel counterparts.It has been estimated that designing a new production facility based on single-use systems can reduce capital costs by up to40% compared to a conventional hard-piped facility[5].Even where major components such as fermenters and buffer tanks are stainless steel,the introduction of single-use buffer tanks can enhance operational?exibility and stretch production capability by removing bottlenecks caused by vessel capacity and changeover times.The?ve most com-mon reasons cited for adopting disposable technologies are the elimination of cleaning requirements,the reduced risk of cross-contamination,the faster turnaround between campaigns,the increased convenience and?exibility of disposable technologies,and the reduced time for a new facility to become operational[6].

The economic bene?ts of disposable technologies are becoming more important because biopharmaceutical manufacturers are facing increasing pressure to reduce product costs while maintaining product quality.The costs of production received little attention in the early years of the industry because the pro?t margins on critical,life-saving biopharmaceuticals often exceeded98%.But as the pace of drug discovery and commercialization slows and fewer blockbuster drugs reach the market[7],competition in the sector is getting hotter,particularly in key thera-peutic areas.For example,biopharmaceuticals have sig-ni?cantly improved the standard of care in the rheumatoid arthritis market and now there are no fewer than seven such products indicated for this disease[7,8].Another key driver of costs is the rising market for generics or biosimi-lars,which are supported by most regulatory health au-thorities because they reduce the costs placed on national health infrastructures[9].Biosimilars are intended to be priced lower than the innovator product,thus creating a demand for more cost-effective production[10].The grow-ing share of emerging biopharmaceutical markets around the world has also squeezed manufacturing costs because of the demand to colocate manufacturing facilities with these markets and the trend towards legislation that requires locally-sourced analytical release testing(quality

Review

Corresponding author:Shukla,A.A.(ashukla@https://www.sodocs.net/doc/2f11442024.html,).

0167-7799/$–see front matter?2012Elsevier Ltd.All rights reserved.https://www.sodocs.net/doc/2f11442024.html,/10.1016/j.tibtech.2012.10.004Trends in Biotechnology,March2013,Vol.31,No.3147

control testing).This favors pharmaceutical sellers that have a signi?cant manufacturing and economic presence in the local market,but these markets are typically smaller than the traditional US and EU markets thus creating a demand for smaller-scale commercial launch facilities that have a lower capital cost base if they use disposable equipment.A?nal consideration is the progressive in-crease in product titers,with many proteins now produced in cell culture with a titer exceeding5g/l.This means that smaller bioreactors are suf?cient for production scale. Whereas many commercial production facilities were installed with multiple10000–25000liter bioreactors, facilities may in the future be able to produce the same amounts of product with single-use2000–5000liter bio-reactors[11].

History of single-use technologies

Disposable tissue culture?asks and roller bottles have been used in the laboratory for many years,and some early biopharmaceutical products were manufactured using roll-er bottle cell cultures.With these exceptions,single-use technologies in large-scale biopharmaceutical production facilities have traditionally been con?ned to the early inoculum stages of the cell expansion process,which in-volved the use of shake?asks or T-?asks.The?rst single-use systems for large-scale production came about with the introduction of the WAVE bioreactor in1996[12].Dispo-sables are now widely used for inoculum expansion pro-cesses,but they have also been employed as the production bioreactor when smaller cell culture volumes are suf?cient [13].The?rst single-use stirred-tank bioreactor was launched by Hyclone in2004and had a working volume of250l.This system used a self-contained plastic bag that was placed in a stainless steel https://www.sodocs.net/doc/2f11442024.html,rger single-use stirred-tank bioreactor systems have been developed more recently(1000liters in2006,2000liters in2009)as well as various fermenters where mixing is achieved by rocking or orbital shaking[14–17].Box1provides further details about cell cultivation in single-use bioreactor systems including some of the challenges faced when making the transition from conventional systems.

Cell cultures are usually harvested by centrifugation and/or depth?ltration[18],and the convenience of dispos-able depth?lters means they are the most widely used

Table1.Factors driving the growth of single-use systems in biopharmaceutical manufacturing

Market factors Advantages Current limitations

Emphasis on production costs Flexible,multiproduct manufacturing facilities Biosimilars

Multiple,smaller manufacturing plants colocated with markets Increasing number of low-volume biopharmaceutical products Reduced capital costs for plant

construction and commissioning

Reduced risk for product cross-

contamination in a multiproduct facility

Rapid changeover

Lower utility costs due to reduced need for SIP

Reduced need for cleaning validation

Leachables and extractables

Prior investment in?xed equipment

Scales limited by current

2000liter cell culture bioreactor capacity

Limited number of vendors

High cost of disposables

Lack of universal standards for vendors

Solid waste disposal

Box1.Cell culture in single-use bioreactors

Cell culture was one of the first areas of bioprocessing to benefit from single-use systems,starting with the WAVE bioreactor which eliminated the need for cleaning or sterilization and thus reduced contamination rates significantly.A variety of other disposable bioreactors have been developed based on the‘cell culture in a bag’concept.

WAVE bioreactors

WAVE bioreactors feature a bag partially filled with cell culture medium and mounted on a tray that can be rocked to provide agitation and gas transfer.Oxygen enters the culture from the headspace above the culture medium.Parameters such as the rocking angle,rocking rate,and bag fill ratio can also influence the mass transfer rates in WAVE systems,which are now available at volumes of up to500liters and have become established as part of the seed expansion steps for most cell culture-based production operations. They are compatible with a wide variety of mammalian,insect,and plant cell lines[13].

Orbitally shaken bioreactors

Orbitally shaken single-use bioreactors[40]rely on the rotating motion of the culture vessel around a central axial shaft to provide mixing and oxygen transfer from the head space into the culture. Orbitally shaken bioreactors are now available up to a200liter working volume developed by Kuhner AG in collaboration with Excellgene and comarketed by Sartorius-Stedim.This is the only bioreactor system in which oxygen supply occurs by air alone and has been shown to be operable without any oxygen probes owing to the high gas transfer rates[41].

Pneumatically mixed bioreactors

Another addition to single-use cell culture systems has been the combination of single-use with airlift bioreactors in which mixing and oxygen are supplied via air bubbles without a mechanical agitator [42].Such systems are available from Cellexus(3–50liter volumes) and from PBS Biotech(3–500liter working volumes).

Stirred tank bioreactors

The majority of cell culture operations for large-scale biopharmaceu-tical production involve the use of stirred-tank bioreactors.The key development in this area was the concept of culturing cells in an integral plastic bag that could be mounted within a cylindrical frame to support the bag.Hyclone was the first market entrant with its Single Use Bioreactor(SUB)system,which used a top driven impeller for mixing and agitation.Xcellerex followed with the XDR disposable stirred tank bioreactors featuring magnetically-coupled bottom-driven agitators.A wide variety of small-volume and large-volume single-use stirred-tank bioreactors are now available(see Table2in main text). Early challenges with efficient mixing and aeration have been addressed and the disposable systems now appear to be equivalent to stainless steel stirred-tank systems for CHO cell cultures[20,43].

148

disposable devices in the industry,along with tubing and connectors(used in54%of all commercial processes[6]). Centrifugation has proved to be more dif?cult to convert into a single-use setup because of the complexity of the devices compared to?lters.However,two disposable cen-trifuges are now commercially available.KSep Systems has a technology based on revolving chambers that are ?tted with a single-use bag to enable closed system proces-sing,whereas Carr Centrifuges has launched the Unifuge system that is essentially a tubular bowl centrifuge lined with a bag.

Disposable systems have also been developed to handle downstream processing operations.The?rst single-use systems were in-process micro?lters for bioburden control between process steps[19].These?lters are typically housed in a plastic capsule that can be discarded after the process intermediate is?ltered.Membrane chromatog-raphy was?rst introduced as a process step in the late 1990s and constituted the?rst single-use technology that was intended to replace a conventional option(preparative chromatography on columns).Although membrane chro-matography has the lowest take-up of all disposable devices,currently featuring in19%of commercial process-es[6],it is also the most recent addition to the family of disposable concepts and has the strongest market growth, with a compound annual growth rate of nearly27%be-tween2006and2012[6].Column chromatography has also entered the single-use market,with the development of mixing and storage tanks that feature a plastic liner inside a stainless steel frame,and the GE Healthcare Akta Ready chromatography skid with a fully-disposable?uid?ow path so that the buffers and product do not come in contact with any?xed parts that require subsequent cleaning. Completely disposable chromatography columns have also been introduced and several companies offer pre-packed, pre-validated disposable columns in a range of sizes containing any resin chosen by the customer.Other dis-posable unit operations such as ultra?ltration/dia?ltration (UF/DF)and drug substance storage(liquid or frozen)have also been introduced into the marketplace.We list some examples of current single-use equipment and their ven-dors in Table2.The current state-of-the-art has enabled the setup of several manufacturing production trains that feature end-to-end disposable technologies[20–22].

An integrated single-use drug substance production process

Figure1shows a schematic representation of a typical biopharmaceutical manufacturing process with single-use technologies for all unit operations[23].This highlights the diverse range of unit operations that go into producing a biopharmaceutical product.The production process typi-cally begins by thawing a single vial of cells and increasing cell counts through the inoculum expansion process.The early inoculum process typically uses shake?asks or spin-ner?asks and then progressively larger volumes in WAVE or small stirred-tank bioreactors.A combination of WAVE and seed bioreactors is typically used in the seed train for mammalian cell culture processes.The seed bioreactor is used to inoculate the production bioreactor during which the cells shift from the growth phase to the product expression phase,resulting in the secretion of the product into the culture medium.Cell harvest and clari?cation removes whole cells and cell debris from the culture broth to provide clari?ed feed that can be loaded onto chroma-tography columns for downstream puri?cation.In large-scale processes,harvest and clari?cation typically involve centrifugation followed by depth?ltration and membrane ?ltration to remove cells and cell debris.For smaller bioreactors,depth?ltration may be used as the primary harvest and clari?cation step without centrifugation.

After harvest and clari?cation,downstream puri?cation aims to produce a pure protein suitable for clinical dosing into humans.This is generally achieved through a combi-nation of column chromatography and/or membrane chro-matography,buffer exchange using UF/DF,and viral ?ltration/inactivation steps.Several chromatography steps are often involved using orthogonal separation prin-ciples to achieve high resolution and peak separation, traditionally involving a mixture of capture chromatogra-phy(where the product binds to the resin allowing con-taminants to be washed through)and?ow-through chromatography,which is used for polishing,that is,the product?ows through and contaminants are retained on the https://www.sodocs.net/doc/2f11442024.html,rge,reusable steel chromatography columns have been the key enabling technology at the heart of most bioprocess separations because disposable solutions have not been available at a comparable scale and the cost of resins means that regeneration and reuse has been re-quired to make processes economically viable.As discussed above,the trend more recently has been to use product-dedicated?xed columns and fully disposable columns where this proves cost-effective(Table2).Whereas large columns remain the best solution for most bind-and-elute steps,disposable membrane cassettes are becoming more popular for?ow-through chromatography steps.Mem-brane chromatography involves the use of synthetic porous membranes containing the same functional groups as packed resins,which is advantageous for polishing steps because there is much less mass transfer resistance and therefore more ef?cient hydrodynamic behavior,which means the membranes can be operated at higher?ow rates with a lower overall buffer consumption and a much shorter processing cycle[24].Disposable membranes are therefore becoming established as a platform for the re-moval of host cell DNA by anion exchange,which can be performed with a membrane bed height of4mm at?ow rates of more than600cm/h[25,26].The availability of disposables in a variety of sizes and functionalities also facilitates scaling up,particularly given that parameters such as frontal surface area,bed volume,?ow rate,and static binding capacity scale in a linear fashion,whereas the normalized dynamic capacity remains constant at10% or complete breakthrough.

Throughout the process,several ancillary unit opera-tions are used for buffer and media batching in mixing vessels,the storage/hold of process intermediates between steps,and membrane?ltration to augment bioburden control.The entire process must be connected to allow the transfer of process intermediates from one unit opera-tion to the next.Appropriate sensors and detectors must also be integrated to ensure that the production process

149

remains within speci?ed tolerances.Finally,the puri?ed drug substance must be transferred to appropriate con-tainers for storage and shipping.In a single-use facility,all of the main and ancillary unit operations,the connections between them,and the monitoring devices need to be developed with disposable product contact surfaces.This can become a challenge when developing an end-to-end single-use facility because although several vendors offer

Table2.Single-use bioprocessing technologies:current scales and vendors a

Unit operation Examples of single-use technology on the market

Bag cultures20–300liter Wave TM Bioreactors(GE Healthcare)

<300liter Biostat Cultibag(Sartorius Stedim)

<500liter BioWave(Sartorius Stedim)

<25liter Appli?ex(Applicon)

<160liter Tsunami Bioreactor(Tsunami Bio)

Production bioreactors Single Use Bioreactor(SUB)<2000liter(Hyclone/ThermoFisher Scienti?c)

XDR Disposable Stirred Tank Reactor<2000liter(Xcellerex)

Biostat Cultibag STR Plus<200liter(Sartorius Stedim Biotech)

Bench-top bioreactors

(laboratory scale,

but could also be used for seed train)CelligenBLU Single Use Bioreactor(New Brunswick)<14liter CELLtainer Single Use Microbial Bioreactor(Lonza)<15liter Mobius Cellready(Applikon and Millipore)<2.4liter

XDR-10(Xcellerex)4.5–10liter

Centrifugation kSep1400and6000(for1–6000liters)(kSep Systems)

Unifuge(Carr Centritech)for up to1000liters

Depth?ltration POD(Millipore)

Stax(Pall)

Zeta Plus(Cuno)

Sartoclear P(Sartorius-Stedim)

Chromatography columns ReadyToProcess(GE)

Opus(Repligen)

GoPure(Life Technologies)

Uno monolith(BioRad)

CIM1monolithic columns(BIA Separations)

Membrane chromatography Mustang(Pall)

Sartobind(Sartorius)

Chromasorb(Millipore)

Chromatography skids Akta Ready(GE)

In-process micro?ltration ReadyToProcess HF(GE)

KrosFlo(Spectrum)

UF/DF membranes Pellicon(Millipore)

Omega(Pall)

(Sartorius)

UF/DF skids Sci?ex(Scilog)

Mobius FlexReady(Millipore)

Allegro(Pall)

Cadence single pass(Pall)

Viral?ltration Planova15N and20N(Asahi)

Viresolve Vpro(Millipore)

Virosart CPV(Sartorius)

DV20(Pall)

Mixing(bag mixing with rotating stirrer)6–2000liter Levmixer TM(ATMI Life Sciences)

6–2000liter Magnetic Mixer(ATMI Life Sciences)

100–1000liter Mobius TM(Millipore)

100–1000liter XDM Quad TM(Xcellerex)

50–1000liter Flexel13D LevMix(Sartorius-Stedim Biotech)

50–2000liter Single Use Mixer(Hyclone/ThermoFisher Scienti?c)

Mixing(bag mixing with rocking)20–1000liter Wave TM(GE Healthcare)

30–5000liter HyNetics(HyNetics Corp)

5–2000liter SALTUS(Meissner)

Connections/tubing C-?ex weldable tubing

Bioquate DAC(Bioquate)

Ready-mate DAC(GE)

KleenPak(Pall)

Quick-connect,MPC,Saniquik,AseptiQuik,sanitary TC(Colder)

Bulk drug substance cold storage CX5-14HDPE Labtainer(Hyclone)

Bioeaze PE(SAFC)

Flexel(Sartorius)

Bulk drug substance freeze-thaw CelsiusPak(Sartorius)

Platinum UltraPak(Lonza)

PETG or polycarbonate bottles(Nalgene)

a Although every effort has been made to provide up-to-date examples of single-use products currently on the market,the authors cannot be held accountable for any omissions or errors.

150

off-the-shelf bags,?uid path designs,and con?gurations with reasonable lead times,the use of too many different platforms creates a requirement to build,sterilize,and stock a large number of different tubing and connector sets in order to bridge the gap between systems.To avoid this issue,several vendors now offer custom connector sets, although it is important to ensure the size of the connectors is appropriate to maintain the intended?ow rate.Depend-ing on requirements,a connection can be made open,for example with Luer Lok?ttings(Value Plastics),sanitary tri-clamps,or quick-connects(Colder Products),or closed if an aseptic connection is required,for example,for bulk ?lling.In the latter case,it is appropriate to use Kleenpak sterile connectors(Pall),ReadyMate DAC(GE Health-care),Opta SFT(Sartorius Stedim),Lynx ST(Millipore), or Pure-Fit SC connectors(Holland Applied Technologies). Tube welding can also be used to make sterile connections, and several tube welders are available commercially from companies such as GE Healthcare(Sterile Tube Fuser), Sartorius(BioWelder),and Terumo.The tubing can be selected from several different vendors,for example,C-Flex(Cole Parmer),PharmaPure(Saint Gobain),Advan-ta?ex heat sealable tubing(AdvantaPure),and SaniPure 60(Saint Gobain)heat sealable tubing(Table2).

End-to-end drug substance manufacturing using single-use systems are now becoming more widely established. Indeed,the bene?t of single-use technologies are fully realized when they are used throughout the production process.This concept is now being extended to mobile manufacturing facilities that can be set up and operated within a very short time period.Vendors are now increas-ingly cognizant of this market and are starting to offer integrated solutions for biopharmaceutical manufacturers, which makes it easier to source and combine various unit operations together.

Current challenges facing single-use technologies

Key challenges facing the developers and end-users of single-use bioprocessing technologies include their limited scale,the restricted diversity of options,the lack of stan-dardization,and some remaining performance issues that can be addressed by further research and development[6].

Most disposable technologies have been developed for a maximum upstream production scale of2000liters cell culture.Table2provides examples of the single-use com-ponents available for different unit operations and the scales that are currently offered.It seems clear that the scales are limited compared to conventional technologies but it should be emphasized that these restrictions are not technological but are based on demand,thus larger sys-tems will probably be introduced as the demand for them grows.The adoption of single-use technologies has been growing year on year since2004and disposables are pre-dicted to reach a20%share of the bioprocess technologies market within the next3–5years.Already,up to90%of manufacturers and contract research organizations use disposable?lters and tubing at some stage of the develop-ment pipeline,whereas77%use disposable bioreactors and 58%use disposable membrane adsorbers[6].

The diversity of options is another current limitation of disposable technologies.This is less apparent for laboratory equipment but increasingly prevalent as we move towards process scale manufacturing and it correlates with the number of vendors offering particular categories of equip-ment.For example,many different vendors offer disposable ?lters and tubing,a handful offer disposable bioreactors, and two supply disposable https://www.sodocs.net/doc/2f11442024.html,petition be-tween vendors often drives innovation so the diversity of disposable devices is bound to increase as more vendors enter the market.This may be stimulated by the interest in single-use technology now shown by larger suppliers,a key example being the recent acquisition of Xcellerex Systems by GE Healthcare.

Another challenge that affects all disruptive technolo-gies is the absence of standardization and regulation of the quality of materials used.One of the key reasons cited by manufacturers for not taking up disposable technologies is the lack of a validation process to determine the nature, quantity and risk associated with leachables and extrac-tables from the disposable plastics,which could potentially contaminate product intermediates.

Cell culture is the most sensitive aspect of a biomanu-facturing process in terms of operability in a single-use system.The plastic bags used for cell culture have the potential to either bind media components[27,28]or to contribute leachables that could adversely impact cell growth.The greatest challenge appears to occur when cultivating cholesterol-dependent NS0cell lines,because the lipid components of the media are depleted by binding to the plastic bioreactor surfaces.However,this issue has been addressed by the development of a novel cholesterol supplementation strategy for these cells[29].In our expe-rience,most CHO cell lines do not have any issues in being operated in single-use bioreactor systems.

The issue of organic compounds leaching from plastic surfaces is often cited as a concern for the use of single-use technologies because the leachables could interfere with cell growth and activity.Leachables are chemicals that migrate from the product contact surface into the process ?uid(e.g.,buffer,water,or process intermediate)under normal exposure conditions,whereas extractables are che-micals that can be removed from the product contact surfaces using appropriate solvents under extreme expo-sure conditions to facilitate their identi?cation and quan-titation.The extractable pro?les released from bioreactor bags under extreme temperature and solvent conditions are typically characterized by vendors.Leachables are typically a subset of the extractables and are expected to be released at very low levels under normal conditions.For example,certain bags used to formulate and weigh out media powders for feeds could impact cell culture due to the presence of anti-static agents.The evaluation of cell culture in single-use bioreactors is a prudent step when setting up a new cell culture platform for the?rst time.A key area for future advancement is to arrive at a common standard set of conditions or analytical techniques to quantify extractables across different vendors and product types.The Bioprocess Systems Alliance(BPSA)is current-ly working towards a set of standards but more work needs to be done to meet the requirements of end-users[30].

The advent of fully integrated bioprocessing using sin-gle-use systems creates the need to source multiple parts

152

and systems from a variety of vendors.In addition to integrating systems from different vendors into a single process,supply of parts and disposables has to be carefully mapped and synchronized.From a supply chain perspec-tive,it is often suitable to treat these as long lead items and conduct facility?t exercises prior to process transfer to plan for these items.

Future directions for single-use technologies

The coming decade is likely to see the increased acceptance of single-use technologies as a standard component of the biopharmaceutical manufacturing process.This has al-ready occurred with disposable tubing and?lters,and to a certain extent buffer and media storage bags,where market growth has slowed to less than10%.Other types of disposable equipment are still becoming established and bene?tting from innovative developments to create more diverse end-user choices.Mixing systems,bioreactors,and membrane adsorbers are in this category and the growth rate in each sector is20–30%[6].There is less activity in other equipment categories and market options are limit-ed,for example,centrifuges and UF/DF skids.Therefore, although single-use technologies are?nding signi?cant mainstream applications in bioprocess operations with many new production trains already starting to employ these systems,further research is needed to increase the diversity available for the unit operations,which have yet to fully embrace the disposables revolution.

Although scalability is one of the current challenges for vendors of disposable technologies,future commercial pro-duction will likely be carried out at the5000liter scale because of the increasing titers of cell cultures.Therefore, we anticipate that the next frontier for single-use bioreac-tors will be an expansion to the5000liter production scale, making single-use technologies a viable replacement for stainless steel in a wider segment of the production space. This increase in scale will drive an increase in throughput and/or scale for other single-use technologies,particularly centrifugation,depth?ltration,UF/DF,and the?ow rates achievable using disposable skids.

In the future it will also be necessary to develop sensor and monitoring technologies that are compatible with sin-gle-use facilities,in order to facilitate the integration of process analytical technologies(PAT).The PAT initiative was introduced by the US FDA to improve the online monitoring of manufacturing processes and facilitate con-trol and(if necessary)correction during a campaign rather than testing the product against speci?cations after manu-facture[31,32].PAT was aligned with the quality by design (QbD)initiative,which aimed to prevent production errors as far as possible by building quality control into process development[33,34].Examples of recent PAT applications include the use of Raman spectroscopy to examine the lot-to-lot variability of cell culture media[35],the use of chro-matographic pro?les to predict chromatography bed stabil-ity and decay in binding capacity[36],and the use of liquid chromatography/mass spectroscopy(LC/MS)to evaluate glycan heterogeneity in monoclonal antibodies[37].These analytical technologies are not exclusive to the single-use market,but the trend towards single-use production sys-tems highlights the need to align the development of sensing technologies with innovations in processing systems.There-fore,single-use sensor technologies are likely to be another key area that sees signi?cant development in the next decade.There is an urgent need for non-invasive sensor technologies that can monitor the health and performance of cell cultures[38].These can include novel chemicals that are added to the cultures to indicate changes in cell metabolism as well as wave-based technologies that can monitor pro-cesses by taking external measurements through a site window.The development of these techniques is certainly the next step towards achieving the goal of PAT for cell culture technologies.

Another key area for the expansion of single-use tech-nologies is microbial fermentation.Traditionally the chal-lenge here has been to provide adequate mass transfer required for fermentation and also to cope with the heat generated by microbial cultures[39].Current single-use microbial fermenters are therefore limited to the50liter scale,and the absence of higher-volume fermenters for the production of biopharmaceuticals on the clinical scale is an area of critical unmet need.

Concluding remarks

Single-use technologies began as an innovative alternative to?xed equipment for the production of biopharmaceutical proteins,but they have since become established as a strong competitive technology for many parts of the pro-duction and processing chain,outcompeting conventional hard-piped stainless steel components in terms of econo-my,convenience,and quality.The increasing pressure on capital and operating costs,the risk of product cross-con-tamination,and the cost of cleaning validation all conspire to push manufacturers away from?xed equipment and to embrace the signi?cant advantages of disposable processes for an expanding network of cGMP manufacturing facili-ties.The growth of single-use manufacturing technologies are anticipated to reduce biopharmaceutical manufactur-ing costs thus aiding the launch of biosimilars,facilitating clinical entry for a wider range of innovative products and an expansion of biomanufacturing activities closer to mar-kets for products manufactured in these facilities.For these reasons,we feel that these technologies have the potential to signi?cantly alter the biopharmaceutical land-scape in the years to come.

Several recent innovations have facilitated the imple-mentation of integrated manufacturing facilities based en-tirely on single-use technologies.Nevertheless,further innovation is required to increase the number of suppliers, the diversity of platforms,the capacity of disposable bio-reactors,standardization of vendor support packages,and the integration of biosensor technologies for non-invasive process control.We anticipate that all these areas will expand signi?cantly in the years to come,bringing dispos-able technologies to the forefront of biopharmaceutical manufacturing in both clinical and commercial manufactur-ing settings.

References

1International Conference for Harmonization(2000)Q7:Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients, ICH

2FDA(2004)Equipment Cleaning and Maintenance,FDA

153

3FDA(2006)Guide to Inspections Validation of Cleaning Processes,FDA 4Gottschalk,U.(2010)Disposables in downstream processing.In Disposable Bioreactors(Eibl,R.and Eibl,D.,eds),In Advances in Biochemical Engineering/Biotechnology(Vol.115),pp.171–183, Springer

5Pais-Chanfrau,J.et al.(2009)The impact of disposables on project economics in a new antibody plant:a case study.BioPharm.Int.22,62–74

6Langer, E.S.(2012)9th Annual Report and Survey of Biopharmaceutical Manufacturing Capacity and Production.A Study of Biotherapeutic Developers and Contract Manufacturing Organizations,BioPlan Associates

7Walsh,G.(2010)Biopharmaceutical benchmarks2010.Nat.

Biotechnol.28,917–924

8Olsen,N.and Stein,M.(2004)New drugs for rheumatoid arthritis.N.

Engl.J.Med.350,2167–2179

9Rathore, A.(2009)Follow-on protein products:scienti?c issues, development and challenges.Trends Biotechnol.27,698–705

10Gottschalk,U.et al.(2012)The need for innovation in biomanufacturing.Nat.Biotechnol.30,489–492

11Kelley, B.(2009)Industrialization of mAb production technology.

Monocl.Antibod.1,443–452

12Singh,V.(1999)Disposable bioreactor for cell culture using wave induced agitation.Cytotechnology30,149–158

13Haldankar,R.et al.(2006)Serum-free suspension large-scale transient transfection of CHO cells in WAVE bioreactors.Mol.Biotechnol.34, 191–199

14De Jesusa,M.J.et al.(2004)TubeSpin satellites:a fast track approach for process development with animal cells using shaking technology.

Biochem.Eng.J.17,217–223

15Baldi,L.et al.(2005)Transient gene expression in suspension HEK-293cells:application to large-scale protein production.Biotechnol.

Prog.21,148–153

16Stettler,M.et al.(2006)New disposable tubes for rapid and precise biomass assessment for suspension cultures of mammalian cells.

Biotechnol.Bioeng.95,1228–1233

17Muller,N.et al.(2007)Scalable transient gene expression in Chinese hamster ovary cells in instrumented and noninstrumented cultivation systems.Biotechnol.Lett.29,703–711

18Shukla,A.and Kandula,J.(2008)Harvest and recovery of monoclonal antibodies from large-scale mammalian cell culture.BioPharm.Int.21, 34–45

19Van Reis,R.and Zydney, A.(2001)Membrane separations in biotechnology.Curr.Opin.Biotechnol.12,208–211

20Shukla, A.et al.(2012)Vertical Integration of disposables in biopharmaceutical drug substance manufacturing.Bioprocess Int.

10,34–47

21Fuller,M.and Pora,H.(2008)Introducing disposable systems into biomanufacturing–a CMO case study.Bioprocess Int.6,30–36

22Wilson,J.(2006)A fully disposable monoclonal antibody manufacturing train.Bioprocess Int.4,34–36

23Shukla,A.and Thommes,J.(2010)Advances in large-scale production of monoclonal antibodies and related proteins.Trends Biotechnol.28, 253–26124Walter,J.K.(1998)Strategies and considerations for advanced economy in downstream processing of biopharmaceutical proteins.

In Bioseparation and Bioprocessing.Processing,Quality and Characterization,Economics,Safety and Hygiene(Subramanian,G., ed.),pp.447–460,Wiley VCH

25Gottschalk,U.(2005)New and unknown challenges facing biomanufacturing.BioPharm.Int.18,24–28

26Gottschalk,U.(2006)The Renaissance of protein puri?cation.

BioPharm.Int.19,8–9

27Altaras,G.et al.(2007)Quantitation of interaction of lipids with polymer surfaces in cell culture.Biotechnol.Bioeng.96,999–1007

28Okonkowski,J.et al.(2007)Cholesterol delivery to NS0cells: challenges and solutions in disposable linear low-density polyethylene-based bioreactors.J.Biosci.Bioeng.103,50–59

29Tao,Y.et al.(2012)Novel cholesterol feeding strategy enables cultivation of NS0cells in low-density polyethylene-based bioreactors.Biotechnol.Lett.34,1453–1458

30Smith-McCollum,B.and Rosin,L.(2006)The Bioprocess Systems Alliance.Bioprocess Int.4,6–8

31FDA(2004)Guidance for Industry:PAT–a Framework for Innovative Pharmaceutical Development,Manufacturing and Quality Assurance, FDA

32Hinz, D.C.(2006)Process analytical technologies in the pharmaceutical industry:the FDA’s PAT initiative.Anal.Bioanal.

Chem.384,1036–1042

33Yu,L.X.(2008)Pharmaceutical quality by design:product and process development,understanding,and control.Pharm.Res.25,781–791 34FDA(2003)Final Report on Pharmaceutical cGMPs for the21st Century–a Risk-based Approach,FDA

35De Beer,T.et al.(2011)Near infrared and Raman spectroscopy for the in-process monitoring of pharmaceutical production processes.Int.J.

Pharm.417,32–47

36Hou,Y.et al.(2011)Improved Process Analytical Technology(PAT)for Protein A chromatography using predictive PCA tools.Biotechnol.

Bioeng.108,59–68

37Kuribayashi,R.et al.(2012)Rapid evaluation for heterogeneities in monoclonal antibodies by liquid chromatography/mass spectrometry with a column-switching system.J.Pharm.Biomed.Anal.67–68,1–9 38Eibl, D.et al.(2011)Single-use equipment in biopharmaceutical manufacture:a brief introduction.In Single-use Technology in Biopharmaceutical Manufacture(Eibl,R.and Eibl,D.,eds),pp.1–11,John Wiley&Sons

39Oosterhuis,N.and van den Berg,H.(2011)How multi-purpose is a single-use bioreactor?Biopharm.Int.24,51–56

40Muller,N.et al.(2004)Orbital shaker technology for the cultivation of mammalian cells in suspension.Biotechnol.Bioeng.89,400–406

41Tissot,S.et al.(2011)Ef?cient and reproducible mammalian cell bioprocesses without probes and controllers?N.Biotechnol.28,382–390

42Lee,B.et al.(2011)Characterization of novel pneumatic mixing for single-use bioreactor application.BMC Proc.5,1–2

43Smelko,J.et al.(2011)Performance of high intensity mammalian fed batch cell cultures in disposable bioreactors.Biotechnol.Prog.27, 1358–1364

154

相关主题