From Process Development To Scale-up Facing The Challenges .

2y ago
11 Views
4 Downloads
4.21 MB
34 Pages
Last View : 25d ago
Last Download : 3m ago
Upload by : Rosa Marty
Transcription

eBookFrom Process Development to Scale-upFacing the Challengesin Vaccine UpstreamBioprocessingSponsored by

Continuous GrowthFibra-Cel disks—3-D growth matrix for perfusion and continuous processesSuspend your disbelief: The three-dimensional Fibra-Cel matrix entrapsanchorage dependent and suspension cells—for optimized growth conditionsand increased yields. Less susceptible to shear forces, clogging, and fouling Ideal for secreted product and vaccine production For use in autoclavable, sterilize-in-place or BioBLU Cel is a registered trademark owned by Imerys Minerals California, Inc., USA and licensed to Eppendorf, Inc., USA. Eppendorf , the Eppendorf Brand Design and BioBLU are registered trademarks of Eppendorf AG, Germany. All rights reserved, including graphics and images. Copyright 2018 by Eppendorf AG.

Driven by epidemic events and by governmental vaccination programs, there is a rising demand fordevelopment of new vaccines and the industry is growing at a double-digit rate. The vaccine industry is facingthe challenge of developing new products to serve so far unmet needs and fulfilling the demands on dose numbers,both in an economically viable way.Upstream bioprocessing is an important piece of the puzzle. High titer, robustness of the process, constant productquality, fast turn-around times, and scalability are some of the success factors. With a comprehensive portfolio ofscalable bioreactor and fermentor systems, software, single-use bioreactors, and worldwide service, Eppendorfstrives to support bioprocess engineers in tackling these challenges.With this ebook we would like to share expert views and case studies on some of the hot topics in vaccine bioprocessdevelopment. We hope the information on viral vector production, continuous bioprocessing, and process scale-upwill prove useful for your own development projects.

From process development to scale-upFacing the Challenges in Vaccine Upstream Bioprocessing5Interview:VaccineManufacturing9 ScalableProductionof AAV Vectors15 IntegratedContinuousManufacturingof Biologics23 rAAV Productionin SuspensionCAP -GT Cells inBioBLU 3c and 10cSingle-Use Vessels30 Vero Perfusion,Packed-BedVessels IntensifyVaccine ProductionCover:Avatar/Getty Images

INTERVIEWVaccine ManufacturingThe rising prevalence of several diseases and the emergence of several new ones are expectedto encourage the development of novel and effective drugs and vaccines, according to a marketresearch study by Transparency Market Research. In 2016, the global vaccines market was worth 28billion and is projected to reach a value of 48 billion by the end of 2025, notes the study. The marketis predicted to register a 6% CAGR between 2017 and 2025.GEN interviewed Mannan Khambati, of Bharat Serums & Vaccines, India, and George O. Lovrecz, PhD,adjunct professor of RMIT and Monash University, and Senior Principal Research scientist, CSIROProtein Production and Fermentation, Australia, to get a better sense of the manufacturing andeconomic issues involved in vaccine development.GEN: What is currently the most importantproduction platform for vaccines (eggs, cellculture plates, roller bottles, stirred-tankbioreactors)? Do you expect changes inthe upcoming years?Mr. Khambati: On the basis of the generaltrend and a cost/benefit analysis, a cell culture5 GENengnews.comprocess utilizing disposable stirred tank or aperfusion system fits perfectly into the scene.As the vaccine industry requires high volumethroughput, high yield cell culture along witha perfusion system should be the preferredchoice in the upcoming years. Disposable perfusion bioreactor systems should take over fromconventional systems to reduce process down-Mannan KhambatiGeorge O. Lovrecz, PhDADDITIONAL CONTENTVaccine development and production needflexible processes, optimized to balance costsand time-to-market. New technologies such assingle-use equipment and automation open upnew possibilities in upstream bioprocessing.Learn More

Facing the Challenges in Vaccine Upstream Bioprocessing Interview: Vaccine Manufacturingtime, optimize the utility requirement,and reduce equipment footprints, thus leadingto compact manufacturing suites and a lower riskof contamination or accidental exposure.GEN: How do you judge the importance ofcontinuous/perfusion bioprocess techniques invaccine production today? Do you expect it tobecome more important in the future?Dr. Lovrecz: Eggs and roller bottles are still themost important platforms, especially for traditional vaccines. This has to change and we allshould move to stirred (preferably single-use)tanks. This is made possible by the advances innovel vaccine generation methods (subunits, etc.)and high yield/high density cell cultures.Mr. Khambati: As the global demand for vaccinedoses increased, there was a need for a newmanufacturing method that could drasticallychange the way we handle high volumes today.The continuous/perfusion system has come as asavior to vaccine manufacturers as it can resolveboth issues of space and volume. With the adventof continuous manufacturing, small footprintequipment with high volume turnaround willbecome a reality in the near future.GEN: Which are the three main challengesin upstream bioprocessing for the productionof vaccines?Mr. Khambati: Low yield process, largevolume processing, and high downtime dueto maintenance, cleaning, and contamination.Dr. Lovrecz: Speed and flexibility (in the caseof a potential outbreak), and often sterility andoperator safety.6 GENengnews.comDr. Lovrecz: Perfusion systems are far moreimportant/central to traditional biologics (mAbs,cytokines, etc.). Yes, I expect it to be become moreimportant in the next five or more years.GEN: To increase scale we can scaleup (increasingthe working volume) or scaleout (increasing thenumber of bioreactors). Do you think one strategyis more promising than the other?The continuous/perfusion systemhas come as asavior to vaccinemanufacturersas it can resolveboth issues ofspace and volume.

Facing the Challenges in Vaccine Upstream Bioprocessing Interview: Vaccine ManufacturingMr. Khambati: In scaleup, usually manufacturingequipment like a bioreactor increases in size vertically while in scaleout the manufacturing suitesincrease horizontally. Due to constraintsin a build area it is always better to scale upthan scale out. In regard to the regulatory andoperational framework, it is better to have lessvariability in a process and hence scaleup wouldbe the preferred strategy over scaleout.Dr. Lovrecz: Scaleout is more important;however, continuous systems and potentiallyfurther genetic-engineering manipulations stilloffer an adequate solution, i.e., we might not havea need for larger single batch volumes after all.GEN: If you could hand in a wishlist, whichimprovements in upstream bioprocessing equipment for vaccine process developmentand production would you like to see?Mr. Khambati: There are few companies thatare manufacturing disposable perfusion systems.I would prefer disposable perfusion bioreactorsand continuous purification systems with prefilled7 GENengnews.comresin columns rather than traditional stainlesssteel or glass vessels being used in production,as well as in process development. Single-usepump heads, media preparation and storagevessels, sensors, genderless connections, etc.would also be a part of my wishlist.Dr. Lovrecz: Enhanced biological efficacy(so volumes could be kept low), single-usesystems for suspension cells, and improvedcell/cell debris removal.GEN: Do you expect certain cell lines or organismsto lose or gain importance for vaccine production?How important are microbial platforms?Mr. Khambati: Scientist are studying manyunique life forms to harness their potentialfor use for human health improvement. But microbial platforms are always going to bein demand for vaccine production due to theirunique advantages. The enormous databaseand ease of working with microbes makethem preferred choices for most vaccine manufacturing companies.Dr. Lovrecz: I believe that mammalian cell lineswill dominate for another 5–10 years but, potentially, insect lines and yeast systems may gainequal importance.GEN: For some vaccines, like seasonal flu vaccines,enormous numbers of doses are needed. Whichpossibilities for the improvement of biomanufacturing do you think will better fulfill thesedemands and avoid shortages?Mr. Khambati: With the rise in the number of casesof seasonal ailments like flu, the demand for itstreatment is always going to increase. The only waythe vaccine industry can fulfill this huge demandis by increasing its manufacturing capacities,improving titers, and harnessing the unlimitedpotential of continuous manufacturing methods.Dr. Lovrecz: Better antigenicity (novel platformssuch as “Molecular Clamp” or vaccinia virus–basedproduction) and greater yields (culturing conditions/cell lines) should be able to improve thevaccine biomanufacturing process. This mighttake 3–5 years before we will see significant results.

Facing the Challenges in Vaccine Upstream Bioprocessing Interview: Vaccine ManufacturingGEN: Economic considerations are an importantfactor in vaccine development. Which strategiesfor cost reduction can you envision to motivatethe development of new vaccines to so-far overlooked diseases?Mr. Khambati: Strategies for making vaccinemanufacturing cost effective and viable havealways been a big challenge for companiesengaged in this business. The applicationof manufacturing strategies like continousprocessing, low footprint plant with low utilitydemands, better process control, and reduced failures can substantially improve the bottom lines.A number of manufacturers are also exploring thepossibility of developing mixture vaccines (e.g.,pentavalent or hexavalent) as therapies againstmultiple life-threatening diseases. Thus, reducingthe number of dosing events and improving themanufacturing logistics are important economicconsiderations.8 GENengnews.comDr. Lovrecz: Single-use flexible bioreactorsusing suspension cell lines, better immunogenicity, improved cell lines, higher final densityand, perhaps, novel platforms should enable thedevelopment of new vaccines especially for thedeveloping world and for overlooked diseases.The Coalition for Epidemic Preparedness Innovations (CEPI) and similar initiatives are reassuring. n

TUTORIALScalable Productionof AAV VectorsRange of Methods Used in Generatingand Purifying AAV VectorsThe Vectors derived from adeno-associatedvirus (AAV) provide promising gene delivery vehicles that can be used effectively inlarge-scale productions for preclinical targetidentification/validation studies, or used inlarge animal models and clinical trials of humangene therapy.9 GENengnews.comWhy is AAV one of the most promising viralgene transfer vectors? Notably, recombinantadeno-associated virus (rAAV) vectors comein different serotypes (AAV 1–9), each withdifferent tissue tropisms. rAAV provides a highrate of gene transfer efficiency, long-term geneexpression, and natural replication deficiency.It is nonpathogenic and does not have thecapability of altering biological properties uponintegration of the host cell.ADDITIONAL CONTENTScale-down models are important to optimizea bioprocess, troubleshoot, and implementchanges. Researchers at Généthon describescale-down model development for a viralvector production process in Sf9 cells.Read application note4X-image/Getty ImagesEva Szarek, Ph.D., andJeffrey Hung, Ph.D.

Facing the Challenges in Vaccine Upstream Bioprocessing Scalable Production of AAV VectorsHowever, achieving preclinical efficacy testing,especially in large animal models and toxicologystudies, requires vector quantities that simplycannot be produced in a laboratory setting or inmost research-grade vector core facilities. Currentmethods for transfection require use of adherentHEK 293 cell cultures, expanded by preparingmultiple culture plates. Ideally, a single large-scalesuspension culture would be a replacement formultiple culture plates.In this tutorial, we examine some of the currentlyavailable schemes used in generating rAAV fromsuspension cultures, and describe what it takes toachieve scalable rAAV production.Scalable ProductionTwo basic systems for growing cells in cultureexist: monolayers on an artificial substrate(i.e., adherent culture) and free-floating inculture medium (i.e., suspension culture).rAAV vector production uses a triple transfectionmethod performed in adherent HEK 293 cells,which is the most common and reliable method10 GENengnews.com(Lock et al., 2010), albeit resource intensive.Due to its scalability and cost, rAAV cell suspensions are more desirable.To simplify scalability and dramatically decreaseoperational costs and capital investments, useof bioreactors provides process simplification,from pre-culture to final product. Two examplesare the iCELLis from Pall Life Sciences, designedfor adherent cell culture applications, and theWAVE Bioreactor from GE Healthcare LifeSciences, ready for batch culture, fed-batchculture, perfusion culture, and cultivation ofadherent cells.Both are designed for convenient handlingand control of cell cultures up to 25 L. Bothenable rapid and scalable rAAV production.Recently, Grieger et al. (2016) showed suspensionHEK293 cell lines generated greater than 1 105vector genome-containing particles (vg)/cellor greater than 1 1014 vg/L of cell culture whenharvested 48 hours post-transfection, a protocoldeveloped and used to successfully manufactureGMP Phase I clinical AAV vectors.Large-scale productions require consistent andreproducible. AAV produced for clinical usesmust be thoroughly analyzed to identify themain purity, potency, safety, and stability factorsdescribed below.PurityEmpty capsids typically take 50–95% of the totalAAV particles generated in cell culture, dependingon specific serotypes and protocols used. Emptycapsids may solicit deleterious immune responseagainst AAV (Zaiss and Muruve, 2005). It is desirable they be minimized during production andremoved during purification.AAV empty capsids are composed of an AAVcapsid shell identical to that of the desiredproduct, but lacking a nucleic acid moleculepackaged within. Gradient ultracentrifugationusing iodixanol is effective in separating emptycapsids. Assessment and measurement can bedone by either electron microscopy or A260/A280spectrometry. It may be difficult to distinguishAAV capsids containing small fragments of DNA

Facing the Challenges in Vaccine Upstream Bioprocessing Scalable Production of AAV Vectorsnot readily distinguished from completely emptycapsids using density centrifugation or electronmicroscopy.dual-labeled hybridizable probe for detectionand quantification of amplified DNA junctionsequence.Helper virus-dependent replication-competentAAV (rcAAV), also referred to as “wild-type” or“pseudo-wild-type” AAV, is an AAV capsid particlecontaining AAV rep and cap flanked by ITR. Thistype of AAV (rcAAV) is able to replicate in the presence of a helper virus.rcAAV DNA sequence titer is calculated by directcomparison to the fluorescent signal generatedfrom known plasmid dilution bearing the sameDNA sequence. A positive signal indicates an intactleft IRT-rep gene junction has been detected andamplified, representing the maximum possiblercAAV contamination level present in the rAAVvector sample being analyzed. It does not indicate whether the DNA sequence is infectious orcapable of helper-virus assisted replication.Though wild-type AAV is unable to replicateautonomously and requires co-infection withhelper viruses, such as adenovirus, the expression of AAV rep or cap from rcAAV present in anAAV vector increases the risk of immunotoxicity invector-transduced tissues. Replication competentrcAAV is a rare ( 10-8) and yet deleterious event.To assess rcAAV generation, target DNA sequencespans left AAV2 ITR D-Sequence and AAV2 repsequence. An intact left AAV ITR-rep gene junctionis a requisite feature for AAV replication to occurin vivo in the presence of a helper virus. The assayemploys sequence-specific PCR primers and a11 GENengnews.comPotencyHigh potency of AAV vectors is achieved by carefully selecting and isolating full capsids. Physicaland functional titers can be measured to assessthe actual potency of AAV production. Physicaltiter measures the encapsidated AAV vectorgenome, a key mediator and indicator of therapeutic effect. Measurement of vector genomes byquantitative real-time PCR is the closest physicalindicator of rAAV vectors. Functional titer isestablished by measuring transgene proteinexpression in a dose-dependent manner,following transduction into appropriate cell lines.SafetySafety concerns comprise infectious agentsused to generate AAV vectors. Mechanismsto inactivate infectious viruses include heatinactivation of adenoviruses and detergentinactivation of enveloped viruses. A completelist of product release tests can include adventitious virus tests of porcine, canine, and bovineviruses (Table).Payload IncreaseDeveloping viral vector comes with key featuresscientists strive for, including large payloadcapacity. With rAAV, the limited packagingcapacity precludes the design of vectors forthe treatment of diseases associated with largergenes; AAV has a packaging capacity of up to4.5 kb for packaging foreign DNA.

Facing the Challenges in Vaccine Upstream Bioprocessing Scalable Production of AAV VectorsTable. Method of removal and measurement of AAV vector production impuritiesAAV Vector ProductionImpurityMethod of RemovalMethod of MeasurementResidual host cell DNA/RNA(nuclease-sensitive)Nuclease treatment(Benzonase)qPCR using amplicons to generic host cell genome(e.g., 18SRNA gene); qPCR using amplicons forsequences of specific concern (e.g., AdE1); qPCRusing amplicons for non-vector genome sequencesResidual host cell proteinUltracentrifugation; ionexchange chromatographyELISA using polyclonal antibodies detecting representative proteinsResidual plasmid DNA(nuclease-sensitive)Ultracentrifugation;ion exchangechromatographyqPCR using amplicons for helper virus sequences;infectious titer of helper viruses; ELISA or Westernblotting for helper virus proteinsResidual helper viruses(nucleic acids and proteins)Nuclease treatment(Benzonase)Various, depending on componentAAV empty capsidsUltracentrifugation;ion exchangechromatographyElectron microscopy; spectrophotometryEncapsidated hostcell nucleic acids(nuclease-resistant)qPCR using amplicons to generic host cell genomesequencesTable continued on page 1512 GENengnews.comFigure 1. Elements in optimization.Before optimization, the trans-splicingefficiency is about 1–5% when comparedto GFP expression from single vector.After optimization, the efficiency reachesabout 25–50%.

Facing the Challenges in Vaccine Upstream Bioprocessing Scalable Production of AAV VectorsTable(con’t). Method of removal and measurement of AAV vector production impuritiesAAV Vector ProductionImpurityMethod of RemovalMethod of MeasurementqPCR using amplicons to specific sequencesof concern (e.g., E1A)Encapsidated helpercomponent DNA(nuclease-resistant)replication-competent AAVnoninfectious AAV particlesqPCR using amplicons for helper backbonesequencesOther, including aggregated,degraded, and oxidized AAVvectorsAd-dependent amplification; Ad-dependentinfectivity in susceptible cells; various, includingsize-exclusion chromatography, dynamic lightscattering, electrophoresisUtilizing the split vector system, which exploitshead-to-tail concatamerization formation, hasbeen developed to circumvent AAV small packaging capacity. Two main approaches includetrans-splicing and homologous recombinationmethods; both depend upon recombinationbetween two vector genomes, with each genomeencoding approximately half the transgene,within the same cell to achieve gene expression.ViGene has developed a trans-splicing systemthat can expand the payload to 8 kb.13 GENengnews.comWe setup a screen for a more efficient transsplicing system. Here, the GFP reporter was splitand cloned into two AAV vectors. Based on GFPexpression, after co-transfection in HEK-293 cells,we scored expression efficiency of the combination of different trans-splicing elements andincluded the selection of a splicing donor andacceptor sequence, and the annealing sequence inthe intron. Our best vector generated about 70%expression compared to single vector (Figure 1).

Facing the Challenges in Vaccine Upstream Bioprocessing Scalable Production of AAV VectorsWe generated and purified both vectors in AAV9trans-splicing and efficiency and expressionlevels were tested in vitro and in vivo. Following72-hours transduction in HEK-293 cells by AAV9virus (Figure 2), GFP expression was detectablein approximately 70% of cells, about 50% whencompared to single vector GFP expression.Similar efficiency and expression levels wereobserved in RGC neurons, following two-weekintraocular injection.A deeper understanding of the molecular basisfor inherited and acquired diseases continuesto drive the broader adoption of AAV as thevector of choice for treating many diseases.Numerous Phase I and II trials utilizing AAVhave been performed for various inheritedand acquired diseases.A continuation to greatly increase AAV vectoryield, improve AAV potency and purity, andincrease payload size will further make AAVa bigger player in gene therapy. n14 GENengnews.comFigure 2. Trans-splicing AAV vector expression of GFP in vitro and in vivo.Eva Szarek, Ph.D., is scientific marketingand sales managerJeffrey Hung, Ph.D. (jhung@vigenebio.com),is chief commercial officer and GM of cGMPbusiness at ViGene Biosciences.

ROUNDUPIntegrated ContinuousManufacturing of Biologics:Trends in the FieldBiosimilar Manufacturers, Take Note:This Stream Is for YouADDITIONAL CONTENTRandi Hernandez15 GENengnews.comDuring upstream bioprocess developmentbioprocess engineers need to decide between abatch, fed-batch or continuous/perfusion process.Analyzing the benefits of their respective usage atbench scale allows confident process decisions.Watch Webinarreptile8488/Getty ImagesAlthough estimates vary slightly, many industry experts predict continuous manufacturing will, at the least, cut the cost of manufacturing biologics in half. Thus, it will be anattractive option for drug makers looking to trimmanufacturing budgets. But not all biologicswould be feasible candidates for a truly integrated processing stream, and a truly continuous linemay require a larger initial capital investment.To learn more about which companies andproducts will likely incorporate continuousmanufacturing first—and to understand which“hot-button” questions regarding implementation still require attention and clarification—GENspoke to pioneers in the field of continuousbiomanufacturing, including MassimoMorbidelli, Ph.D., professor of chemical reactionengineering at the Institute for Chemical andBioengineering at ETH Zürich; Andrew Zydney,

Facing the Challenges in Vaccine Upstream Bioprocessing Integrated Continuous Manufacturing of Biologics: Trends in the FieldPh.D., distinguished professor of chemical engineering, The Pennsylvania State University;Michelle Najera, Ph.D., downstream development scientist, CMC Biologics; Gerard Gach, chiefmarketing officer, LEWA-Nikkiso America; DanaPentia, Ph.D., senior application scientist, andJohn Bonham-Carter, director of upstream salesand business development at Repligen; GerbenZijlstra, platform marketing manager, continuousbiomanufacturing, Sartorius Stedim Biotech; andKarol Lacki, Ph.D., vice president of technologydevelopment at Avitide.Common conclusions from the experts were thatthe use of surge tanks in continuous lines hasboth benefits and drawbacks (see the Bioprocessing Perspectives column on page 22 in theSeptember 15 issue of GEN); enzyme-replacementproducts and monoclonal antibodies (mAbs) willlikely be the first product candidate types selectedfor integrated continuous manufacture (morespecifically, mAb-based biosimilars); and manufacturers are less likely to switch legacy productsfrom existing batch processes to continuousoperation. The high prices of affinity resins are16 GENengnews.comnot expected to decline significantly in the nearfuture, and continuous production could result inresin cost savings—but the experts explain thereare many benefits of continuous production ofbiologics besides those related to cost.Most of the products that would likely be madein integrated continuous lines will be new products, and will also primarily be labile biologics orthose which have uncertain demand. Continuousoperation, says Bonham-Carter, will allow engineers to react to fluctuations in product demandand give companies the option to build late (orbuild out) significantly, if required. Dr. Morbidelliasserts that peptides, fusion proteins, scaffoldswith mAbs, and other products “containing sensitive antennary glycostructures that are needed forbiological activity” would also be types of therapies that could be made in continuous flows.To tell which pharma manufacturers are likely tointegrate end-to-end continuous manufactureinto production lines first, investors and industryinsiders should follow company patent filingsand peer-review articles authored by companyrepresentatives. As Dr. Zydney points out, manu-facturers that are already “clearly interested” infully continuous biomanufacturing lines includeGenzyme, Merck, Bayer, and Sanofi, amongothers. Also doing work in this space: NovoNordisk, Novartis, Amgen, Shire, Pfizer, WuXi,and BiosanaPharma. In addition, some contractmanufacturing organizations (CMOs) are in thecontinuous biomanufacturing field (such as CMCBiologics), and these organizations are poised tohelp biopharma clients lower the cost to clinic byway of fully continuous manufacturing strategies.Regardless of who is or is not investigating continuous, “What is definitely a right development trend isthat vendors do support the change and offer technologies that enable continuous operations,” says Dr.Lacki of Avitide, a company that makes affinity resins.“At the end of the day, it will be up to the end user todecide how a process needs to be operated, but thechoice will be made on a thorough assessment ofcommercially available technologies.”GEN: What types of biologic medications arethe best/most feasible candidates for integratedcontinuous manufacture?

Facing the Challenges in Vaccine Upstream Bioprocessing Integrated Continuous Manufacturing of Biologics: Trends in the FieldDr. Najera: Any product with an expensivechromatography resin is a suitable candidate forcontinuous chromatography, especially for earlyphase clinical products, where resin cost can becost prohibitive. Continuous capture is also attractive for high-titer processes, since several smallvolumes can be used instead of a single largecolumn. In fact, facility fit limitations are commonfor high-titer processes ( 5 g/L), and continuouschromatography can help manufacturers avoidthe capital costs associated with procurement oflarge stainless-steel columns ( 80 cm diameter)and associated equipment. Finally, the concept ofintegrated continuous manufacture, (i.e., a fullycontinuous process), would be best suited for awell-established late-phase process with a stablemarket demand where cost-savings are realizedthrough basic efficiencies related to batch versuscontinuous processing.Dr. Zydney: This is a difficult question toanswer—it very much depends upon one’sperspective. In some ways, the ‘best’ candidates17 GENengnews.comIntegrated ContinuousManufacturing of BiologicsKarl Rix, Ph.D.,Head of Business Unit Bioprocess, EppendorfGEN: What types of biologic medications are the best/most feasible candidates for integrated continuousmanufacture?Dr. Rix: Besides for the production of less stable proteinswhich in a perfusion process are constantly harvestedfrom the culture, perfusion processes could be especiallyadvantagous to respond to fluctuations in the market.Scale can be varied by modulating the process time or bychanging the number of parallel production lines. This canbe easier than traditional scale-up of a fed-batch process.GEN: The manufacturers of Prezista (Janssen), asmall-molecule drug, got FDA approval to change itsprocesses to a continuous method. To your knowledge,are any biologics manufacturers looking into a similarmanufacturing change? Do you think manufacturers aremore likely to address the end-to-end manufacture of atotally new product, rather than for an existing product?Dr. Rix: Though there have been examples, it is still quiteunlikely that a manufacturer would change an approvedbatch or fed-batch manufacturing process to continousoperation. Usually, the decision for a process modewill be made in product development. In that phase,influencing variables, including product titer, costs formedia, labor, and equipment, and product quality, can besystematically assessed.GEN: To date, why do you think so few biologicmanufacturers have explored the use of an end-to-endcontinuous line?Dr. Rix: In my view, one issue ist that processdevelopment as well as process monitoring strategiesfor continuous processes are less well establishedthan for traditional fed-batch processes. This includesamong others cell line and media development, onlinemonitoring technologies, and scale-down modeldevelopment. This might increase process developmenttime and risks. And in my opinion, regulatoryuncertainties are still an issue.

Facing the Challenges in Vaccine Upstream Bioprocessing Integrated Continuous Manufacturing of Biologics: Trends in the Fieldfor integrated continuous manufacturing areproducts for which there are particular challengesin using batch operations. For example, a highlylabile product that degrades over time wouldbenefit dramatically from the use of a continuousproce

Fibra-Cel is a registered trademark owned by Imerys Minerals California, Inc., USA and licensed to Eppendorf, Inc., USA. Eppendorf, the Eppendorf Brand Design and BioBLU .

Related Documents:

CCC-466/SCALE 3 in 1985 CCC-725/SCALE 5 in 2004 CCC-545/SCALE 4.0 in 1990 CCC-732/SCALE 5.1 in 2006 SCALE 4.1 in 1992 CCC-750/SCALE 6.0 in 2009 SCALE 4.2 in 1994 CCC-785/SCALE 6.1 in 2011 SCALE 4.3 in 1995 CCC-834/SCALE 6.2 in 2016 The SCALE team is thankful for 40 years of sustaining support from NRC

Svstem Amounts of AaCl Treated Location Scale ratio Lab Scale B en&-Scale 28.64 grams 860 grams B-241 B-161 1 30 Pilot-Plant 12500 grams MWMF 435 Table 2 indicates that scale up ratios 30 from lab-scale to bench scale and 14.5 from bench scale to MWMW pilot scale. A successful operation of the bench scale unit would provide important design .

work/products (Beading, Candles, Carving, Food Products, Soap, Weaving, etc.) ⃝I understand that if my work contains Indigenous visual representation that it is a reflection of the Indigenous culture of my native region. ⃝To the best of my knowledge, my work/products fall within Craft Council standards and expectations with respect to

Scale Review - Review the E - flat scale. Friday 5/29/2020. Scale Review - Review the c minor scale. Sight Reading. Monday 6/1/2020. History - Read 20th Century Packet - Complete listenings and quiz. Scale Review - Practice the B - flat Major scale. Tuesday 6/2/2020. Scale Review - Practice the g melodic minor scale. Wednes

Remember, this is just an abridged form of the major scale. It's not a 'separate', distinct scale. It's just the major scale, in a simpler form. Can you see that this has just a few notes less? Minor Scale Minor Pentatonic Scale Remember, this is just an abridged form of the minor scale. It's not a 'separate', distinct scale.

So if you start playing the major scale pattern on the 7th fret you will be playing the B Major Scale. If you start playing the minor scale pattern on the 10th fret you will be playing the D Minor Scale. If you start playing the minor pentatonic scale on the 3rd fret you will be playing the G Minor Pentatonic Scale. Have fun! !! !

The scale has an Auto Shut Off feature which automatically turns off the scale after three minutes of inactivity. To turn the scale on Press on the front of the scale. When the scale is first turned on, wait a few seconds for the scale to stabilize before weighing items. The di

the scale factor of the drawing? a. drawing length —— actual length 5 cm — 10 mm 1 cm — 2 mm The scale is 1 cm:2 mm. b. Write the scale with the same units. Use the fact that 1 cm 10 mm. scale factor 1 cm — 2 mm 10 mm — — 2 mm 5 1 The scale factor is 5:1. 3. A model has a scale of 1