In the biopharmaceutical industry,

Similar documents
In the current competitive biopharmaceutical

Efficient operation of the HyPerforma 5:1 Single-Use Bioreactor at low working volume

Perfusion and Beyond The XCell TM ATF System

Single-Use Cultivation in a Classical Format:

Efficient operation of the HyPerforma 5:1 Single-Use Bioreactor at low working volume

Outline. Upstream Processing: Development & Optimization

Disposable technologies are

Application Note. Increased efficiency and product quality with the UniVessel Single Use bioreactor for CHO fed-batch cultures.

BIOSTAT B-DCU. Product Presentation. Page 1

Your Guide to Upstream Processing Solutions From Research to Production

Reducing Total Cost Of Ownership In Media Filtration. White Paper. by Mandar Dixit and Chris Sullivan

single-use fermentors

One-step seed culture expansion from one vial of high-density cell bank to 2000 L production bioreactor

Significant time savings with simplified cell culturing using ReadyToProcess WAVE 25

Thermo Scientific HyClone Single-Use Bioreactor Products and Capabilities. Discovery Development Production

While attending a conference

Scalability of the Mobius CellReady Single-use Bioreactor Systems

BIOSTAT RM - Rocking Motion Bioreactors

Welcome Biotech 101. Outline. Before the Floor. Manufacturing Process. Supporting Cast 4/22/2013

Improving Single Use Bioreactor Design and Process Development: New Research Towards Intensifying Seed- Train and Scale-Up Methods Using 5:1 Turn-Down

Harvesting Technology Guide for mab Processes. Accelerated process development through the identification of optimal platform solutions

Single-Use Simplicity

Bioprocessing Challenges: High-Titer Mammalian-Based Cell Systems. Elements influencing the way Biologics may be manufactured /supplied in the future

How single-use connections advance aseptic processing: Increased process flexibility and reliability, reduced costs

Innovative Bioreactor Systems for Enhancing Vero Cell growth

Driving Value through Innovation in Biotech Manufacturing. Agenda

Cultivation of sensitive cell lines - Improving bioreactor performance by dynamic membrane aeration

Cell Growth Performance in Single-Use Bags

MAKING REAL-TIME PROCESS ANALYTICAL TECHNOLOGY IN BIOMANUFACTURING A REALITY

BPI.Seminar Semina opelmg ropelmg.com com

BD Resurge Media Supplements Chemically Defined. Protein Free. Animal Free.

Trends in Perfusion Bioreactors: Will Perfusion Be the Next Revolution in Bioprocessing? June 15, 2011 BioProcess International

Single-Use Simplicity

Single-Use Simplicity

Shortening Timelines for Upstream Bioprocessing of Protein-based Therapeutics

9 Reasons To Consider A Single-Use Fermentor

Inspiring Advances in Bioprocessing. John Bonham-Carter March 14, 2017

Accelerate Bioprocess Development Using the Modular Automated Sampling Technology (MAST ) Platform

Hollow Fiber Bioreactors: Single-Use. Perfusion. Scalable. Continuous Manufacturing.

Single Use Capacitance Measurements of Viable Cell Volume

Optimizing the HyPerforma Single-Use Bioreactor for adherent cell culture on microcarriers

Optimizing the HyPerforma Single-Use Bioreactor for adherent cell culture on microcarriers

Scaling Down Bioreactor Process Development: Comparison of Microbioreactor and Bench Scale Solutions.

Presented at the ECI Conference on Integrated Continuous Biomanufacturing Castelldefels, Spain, October 21, 2013

Implementation of a Micro Bioreactor System for Platform Cell Culture Process Development at Cobra Biologics

Fast Trak Services: Long-term collaboration supports speed to market in single-use biomanufacturing

Kelly Thom Associate Principal Scientist Fujifilm Diosynth Biotechnologies. Your Biologics and Vaccines CDMO Partner of Choice.

BioPAT ViaMass Real-time, Inline Biomass Measurement for Cell Culture Processes in single-use. 03 May 2013 Page 1

BIOSTAT RM Culturing Convenience

Efficient and controlled expansion of IgG1 producing CHO-DG44 cells using the ActiCHO Media System and WAVE Bioreactor

Continuous Processing Progress in Manufacturing

High throughput cell culture technology as a route to improved process understanding. Seth Rodgers, CTO Bioprocessors Jan 29, 2007

ProCellics. In-Line & Real-Time Bioprocess Raman Analyzer

Powerful, Intelligent & Intuitive Mixing For Your cgmp Biomanufacturing

XDR Single-Use Bioreactors

Contents. 01 Introduction to Continuous Manufacturing: Technology Landscape and Trends. 02 A Brief History of Perfusion Biomanufacturing

Continuously Improving Bioprocesses: Biopharmaceutical Capabilities

BioBundles. Real bioreactors, really small.

CHAPTER 2 CULTURE TYPE

A Quality-by-Design Approach to Upstream Bioprocess Interrogation and Intensification

Thermo Scientific Metabolic Pathway Design Process Cell culture media and process optimization approaches for optimal biotherapeutic production

CHO-GSN PLATFORM STABLE CELL LINE GENERATION. NR v5

Fast Trak Services: Ongoing collaboration aims to accelerate biosimilar approval in China

Challenges in Scaling Up Newly Developed Microbial Manufacturing Processes

Bioreactors and Fermenters. Biometrix Corporation (800)

Advances in Single-Use Platforms for Commercial Manufacturing

During the past few years, use

croatia BIOSTAT B The Multi-Talent for Research and Development

Future biologics manufacturing

Summer Training Program In Industrial Biotechnology BiOZEEN, Bangalore

Cellca Technology Platform

Single-Use Products in BioManufacturing: Innovations Driving the Industry Bioprocess International SUPPLEMENT March 2011 By Eric Langer

miniperm The Bioreactor for the Production of Monoclonal Antibodies and Recombinant Proteins

Integrated Single-Use Bioreactor (S.U.B.) systems with controllers

Conversion of Bioreactors to Continuous Perfusion Using Hollow Fiber Cell Separators

Internal Presentation. BIOSTAT Cplus. The SIP Stainless Steel Fermenter Bioreactor for Your Lab

Enhancing Upstream and Downstream Process Methods Through Implementation of Single-Use Technologies. 06 February 2018

Challenges in the cgmp Manufacturing of hescs: Lessons Learned from Monoclonal Antibodies

Application of Disposable Technologies in Biopharmaceutical Manufacturing

Emerging and Enabling Technologies in Membrane Separations

GE Healthcare Cell Therapy Steve Hawthorn September 1st 2011

Intensification of a Perfusion Platform using Single-use XCell ATF Systems

F2 15/30 SIP PILOT SCALE BIOREACTORS

WELCOME TO THE FUTURE OF TRANSIENT EXPRESSION. Smart start guide for the ExpiCHO system

Accelerate Your Process Development With High-Throughput, Single-Use, Fully Automated Bioreactors. White Paper. by Mwai Ngibuini

Large-Scale Perfusion and Concentrated Fed-Batch Operation of BIOSTAT STR Single-Use Bioreactor

Cultivation of aggressive microbial cultures of Pichia and Bacillus using the HyPerforma 30 L and 300 L Single-Use Fermentors

Assessment of process performance and product quality in high-performing fedbatch

Critical Analytical Measurements for Bioreactor Optimization. controlling an organism s chemical environment leads to consistent and

Application of Single-Use Equipment for Buffer Storage and Distribution in Medium Size mab Production Facility

SCIENTIFIC. Process for Success. CHO ONE Media System. Expression Media for Fed-Batch Culture

Professor Wei-Shou Hu Spring 2007 ChEn 5751

Continuous Biomanufacturing: Relevant Experiences with Development, Hybrid Implementation, and Emerging Opportunities

F2 15/30 SIP PILOT SCALE BIOREACTORS

The Use of Disposable Technologies in Antibody Manufacturing Processes

The monoclonal antibody (MAb) market

Transcription:

S i n g l e - U s e TECHNOLOGIES Continuous Cell Culture Operation at 2,000-L Scale Michael Sherman, Vincent Lam, Melissa Carpio, Nick Hutchinson, and Christel Fenge In the biopharmaceutical industry, continuous manufacturing is often cited as a method for increasing the productivity of bioprocesses (1). Compared with batch processing, it has the potential to enable production of more product within a smaller facility footprint while improving product quality, particularly for sensitive and unstable molecules. Investigation into continuous methods is taking place for both upstream and downstream operations. For the full benefit of continuous processing to be realized, an argument has been made that cell culture, harvest, and product purification must be effectively integrated (2). That follows a trend within the industry to take an increasingly holistic view of bioprocesses and bioprocess platforms encompassing seed bioreactors through downstream processing to drug substance freezing and thawing. Cell culture operations play a vital role in determining overall process productivity. The industry has made great progress over the past couple decades in achieving higher product titers from fed-batch cell cultures. One consequence has been a move toward smaller bioreactors and replacement of older stainless steel equipment with single-use technologies. Some companies are choosing to develop perfusion processes. They are especially useful when a therapeutic product is labile and has a short halflife within a production bioreactor. Under perfusion control, protein can be constantly harvested from a culture vessel and purified before degradation occurs. The constant addition of nutrients and removal of toxic metabolites allows perfusion cultures to reach and sustain high cell densities over many weeks. Not only is that a highly productive condition, but it also allows cells to remain within a steady state and express consistent product. Advances in cell culture media and improvements in the robustness of microfilter cell-retention devices have contributed to the adoption of perfusion. Some disadvantages exist, however, including challenges in operating cell cultures for prolonged periods: e.g., preventing contamination and demonstrating consistency during validation from harvest and purification of multiple subbatches. Those concerns are particularly important during drug development for companies trying to rapidly reach their investigational new drug (IND) filing. So some companies are deciding to operate in perfusion mode, through which product is retained (sometimes called concentrated fed-batch mode) as fresh media is added and exhausted media removed using an ultrafiltration device. Very high cell densities can be generated and maintained that give exceptional productivity in a short time frame without the additional complexity of harvesting and purifying multiple sub-batches of product. We believe that the full potential of continuous upstream operations has yet to be realized. Applications of new technologies can help deliver upstream bioprocesses with even higher productivities at preclinical, clinical, and commercial manufacturing scales. With such high titers, even blockbuster drugs could be manufactured using 2,000-L singleuse bioreactors, and no additional scale-up step after process validation in phase 3 would be needed. That offers the advantage of omitting an additional scale-up step and eliminating the associated time and risk of further adapting a bioprocess. In addition, drug makers can exploit all the advantages of single-use manufacturing through perfusion operations. Concentrated Fed-Batch or Perfusion Operations: After inoculation of a bioreactor and an 22 BioProcess International 14(10)s November 2016 Supplement

Figure 1: Set-up of a concentrated perfusion or fed-batch set-up using the STR single-use bioreactor Overlay Sparger Balance Air CO 2 CO 2 Air O 2 N 2 Feed BioPAT Trace Exhaust cooler Bleed Bleed Figure 2: Comparing viable cell density (top) and cell viability (bottom) over time from a bioreactor with historical data generated from an existing single-use bioreactor for the MAb A process. Viable Cell Density ( 10 5 cells/ml) PO 2 Temperature ph BioPAT ViaMass cell density and viability Balance Refine ATF Filtrate Filtrate Viability (%) Figure 3: Comparing the percentage of packed cell volume data and packed-cell adjusted product titer over time from a with historical data generated from an existing single-use bioreactor for the MAb A process Packed Cell Volume %) Packed Cell Adjusted Titer (g/l) initial batch growth phase, removal of cell-free supernatant using a cell retention device begins at a constant harvest flow rate. Meanwhile, the culture is replenished with an equivalent volume of fresh medium. Control of its addition can be achieved through a feed that receives a signal from load cells or a platform balance that enables operators to maintain a defined bioreactor weight. Multiple approaches might be used, including non-automated daily feed rate adjustments based on nutrient levels. As cell density grows and nutrient consumption and metabolite formation increase, the exchange rate increases to provide a certain amount of fresh medium per cell per day or alternatively, to follow a predefined profile of medium exchange per day (3). On-line biomass measurement provides an automated option to control perfusion or cell bleed rate based on a cell-density signal. Using an additional concentrated feed can help operators control the glucose concentration in a bioreactor using feedback from at-line glucose and lactate measurements. The schematic in Figure 1 depicts a typical concentrated perfusion or fed-batch set-up. Key Considerations: Typical perfusion rates fall into the range of one to two bioreactor volumes per day. Applying a small cell-bleed stream enables establishment of a defined cell growth rate, and by that a high viability can be maintained that in turn mitigates clogging of the cell retention device (4). Depending on the pore size or cut-off of the cell retention membrane, a protein product will be either recovered in the cell-free harvest (microfiltration perfusion) or retained in the bioreactor (ultrafiltration perfusion). Most antibodies are relatively stable, so perfusion with accumulation of such products in a bioreactor offers a simple and straightforward approach to increase the space/ time yield of a given antibody-manufacturing facility. Microfiltration perfusion is the method of choice for recombinant proteins that are prone to degradation. And those that might show feedback inhibition should be removed from cell culture as fast as possible and transferred into a chilled harvest tank before subsequent purification. Using Sartorius stirred-tank, single-use bioreactors in combination with different sizes of cell retention devices, perfusion processes can be developed at the 2-L bench scale. For example, a company can use a Univessel single-use bioreactor in combination with a B or B-DCU controller and subsequently scale its process to 500 L, then 1,000 L in a STR system. At 1,000-L and 2,000-L scales, a retention device (e.g., ATF system filter modules from Repligen) connects through side ports of a single-use bioreactor bag with up to two 1-in. sterile connectors and operates in an external loop of the bioreactor. Alternatively, Spectrum Labs and other companies propose using single-use, hollow-fiber modules operated in tangential-flow mode for cell retention. Another cell 24 BioProcess International 14(10)s November 2016 Supplement

Figure 4: Viable cell density (VCD) and viability (%) over time were comparable with historical data from the existing single-use bioreactor for the MAb short B process. VCD ( 10 5 cells/ml) Viabilty (%) STR STR retention option is to use a continuous centrifuge such as a ksep single-use centrifuge from Sartorius or a Carr Centritech Cell system from PneumaticScaleAngelus. The advantage of continuous centrifugation is that it eliminates filter clogging and allows dead cells to be selectively separated and removed depending on the chosen centrifugal force. For all those options, it is critical that the external loop be as short as possible to keep from exposing cell cultures to uncontrolled conditions, most critically those related to temperature and dissolved oxygen (DO). Single-Use Bioreactor Configurations Suitable for Intensified Cell Cultures: The key to successful perfusion operations is an efficient aeration system that provides k L a values above 10 15 h 1 to supply cultures with sufficient oxygen. During an intensified culture run, large amounts of carbon dioxide are formed and must be removed to prevent inhibitory effects on productivity or possible negative effects on product quality. With STR single-use bioreactors, that is achieved with a combination sparger that microsparges compressed air or pure oxygen through defined 150-µm holes while providing a stripping gas flow through 0.8-mm holes simultaneously. Figure 5: Difference in aeration performance between a bioreactor and an existing single-use bioreactor for the short MAb B process Total Gas Flow (slpm) Pressure (psig) 180 160 140 120 100 80 60 40 20 0 0.50 0.45 0.40 0.35 0.30 0.25 0.20 0.15 0.10 0.05 0.00 Run #4 Run #1 Run #1 Run #2 This single-use sparger design emulates a successful aeration strategy that has been used for many years in conventional stainless steel bioreactors. A problem that should not be underestimated is excessive aerosol formation in exhaust gas due to high gas-flow rates and protein contents in concentrated cell cultures. A specifically developed single-use exhaust condenser design based on the well-known principle of plate heat exchangers can reduce the risk of blocked filters and dramatically increase process reliability. Additional safety interlocks in Sartorius bioreactor-control software prevent overflow in the event of a clogged cell retention device. Furthermore, all feed s and gas flows are interrupted if pressure in a STR bioreactor exceeds its maximum defined operating pressure. Here, we explore the use of this 2,000-L single-use bioreactor for production of two different monoclonal antibodies (MAbs) by continuous culture. Methods A large US biotech company evaluated the performance of the new system against that of existing single-use and stainless steel bioreactors in two perfusion processes. Both processes Run #3 Run #4 Run #2 Run #3 used a common cell-retention device with Chinese hamster ovary (CHO) cell lines to produce two different MAbs. With a 2,000-L maximum working volume, the 2000 is the largest model in the STR range of single-use bioreactors from Sartorius Stedim Biotech. Low-shear agitation with three-blade segment impellers provides homogeneous mixing. A microsparger designed not to shed particles has defined 150-µm holes to allow oxygen transfer rates of 40 h 1. A classical stirred-tank design enables application of wellknown and established scale-up criteria such as power-input per unit volume or tip speed. Geometric similarity from the ambr 250 microbioreactor (with its working volume of just 250 ml) across the Sartorius range of single-use bioreactors to this 2,000-L unit allows for seamless scale-up (5). Bags for STR bioreactors are made of the new S80 multilayer polyethylene film, recently introduced by Sartorius for the Flexsafe range of bioprocess containers. Biomanufacturers can use the same film for storage, and it will be available in the future also for mixing, shipping, and freeze thaw applications. That will help companies

Figures 6: Viable cell density (VCD) and viability (%) over time were comparable with historical data from the stainless steel bioreactor for the long MAb B process. Figure 7: Difference in aeration performance between a bioreactor and a stainless steel bioreactor for the long MAb B process 250 VCD ( 10 5 cells/ml) Total Gas Flow (slpm) 200 150 100 50 Viabilty (%) reduce the number of materials their products will come into contact with from beginning to end of their manufacturing processes. In the context of cell culture, S80 film delivers reproducible cell growth comparable to that of borosilicate glass (6, 7). It is physically robust as measured by tensile strength and other test methods and adheres to the ASTM D4169-09 guideline for shipping applications (8). Flexsafe bags are manufactured with a holistic strategy that takes consistent quality, assurance of supply, change control, and business continuity into consideration (9). This bioreactor (including consumables) has been verified at the 1,000-L scale using a CHO fed-batch MAb production process (10). To benchmark the system and demonstrate its functional equivalence with an existing single-use bioreactor, the company used a continuous cell culture process for production of MAb A. We compared data from four runs with historical data averaging eight previous runs in the existing bioreactor. To test the new system s capabilities, we also compared its performance with that of the existing single-use and a stainless steel bioreactor for production of MAb B. This is a more demanding process than what was used to produce MAb A because it generates higher cell densities that demand more oxygen. 0 Such conditions can be challenging for single-use bioreactors, which are typically not designed to handle exhaust gases from such intense processes. Results MAb A Bioreactor Functional Equivalence Benchmarking: Figure 2 compares viable cell density (VCD) and cell viability (%) data from a system with historical data generated from an existing single-use bioreactor. Averaged data from the former falls within one standard deviation of the mean generated from those historical data. Figure 3(top) compares the packed-cell volume data (%) from both bioreactors. Results show a high degree of comparability between the two systems. Figure 3(bottom) compares their packed-cell adjusted product titers, and once again the two systems are comparable. From these results, we can conclude that the new system is functionally equivalent to the 2,000-L single-use bioreactor already in use. Here, we observed that less oxygen was needed to maintain the DO set point of the culture for the new system compared with historical data, which suggests a better oxygen transfer capability (not shown). MAb B Testing STR with a More Challenging Process: Two different MAb B production processes were run in a system. One process takes 25% longer than the other to run. For the shorter process, VCD and percent viability were comparable with those of historical data from the existing single-use bioreactor (Figure 4). Packed-cell adjusted product titer data also were comparable (not shown). However, a significant difference was observed between the two 2,000-L single-use bioreactors during this shorter process: Total gas flow and bag pressure typically were lower in the new system than in the other single-use bioreactor. Apparently, oxygen transfer within the former is particularly efficient. Its lower gas flow rates minimize shear damage from bubble bursting and lowers the risk of exhaust-filter blocking due to foaming. That risk is mitigated further by the new bioreactor s large headspace. Figure 5 illustrates the difference in aeration performance between the two single-use bioreactors. These findings raised the possibility that MAb B could be manufactured in a system with the longer process. Historically that was possible only in a stainless steel bioreactor because of limitations in oxygen transfer with single-use bioreactors that had been previously assessed from other suppliers. A second set of culture runs compared the system with a longer stainless steel bioreactor reference process. Figures 6 and 7 show that comparable performance was achieved. The lower total gas flow rates provided by the single-use bioreactor were sufficient to support cell growth and product Supplement November 2016 14(10)s BioProcess International 27

formation, demonstrating the system s superior oxygen transfer. Packed-cell adjusted product titer data also were comparable (not shown). Figure 6 also shows performance of the single-use system at two different agitation rates. Clearly the oxygen transfer rate need not be a limiting parameter with that bioreactor, thereby opening the door to longer processes, higher cell densities. and even greater productivity. As Good As or Better Than We have demonstrated that the system provides equivalent performance to that of an existing single-use bioreactor for continuous production of MAb A. However, because of its superior oxygen transfer rates, less gassing is needed. That reduces the risk of exhaust-filter blockage from excessive foaming and pressure build-up in the bioreactor. In a more demanding process for a second antibody, that superior oxygen transfer efficiency allowed a culture to run for a longer time than was achieved previously with single-use bioreactors. Process performance of the new system was comparable to that of a stainless steel reference process. So with a system, even higher cell densities and more MAb product can be generated from a single continuous run while retaining all the benefits of single-use processing. Newly emerging technologies should enhance cell culture process development, scale-up, and process control significantly. Once these are applied in combination with highperforming, single-use bioreactors, then extremely high cell densities will become a reality. Control of critical process parameters requires advanced process control software and appropriate process analyzers. Singleuse, precalibrated, and presterilized sensors are available for measurement of ph and DO. Oxygen and carbon dioxide sensors can be used in bioreactor exhaust lines both to monitor and provide additional insight into cell culture metabolism. Measurement of nutrients and metabolites such as glucose and lactate Newly emerging technologies should enhance cell culture process development, scale-up, and process control significantly. Applied in combination with high-performing, single-use bioreactors, they will make VERY HIGH cell densities possible. can be used during continuous cell cultures to provide information about nutrient supplies and the general metabolic state of a culture. That information can help companies determine the most appropriate nutrient feeding rates using fully automated control loops. Automated viable-biomass measurements (e.g., those provided by BioPAT ViaMass sensors from Sartorius Stedim Biotech) can be completely integrated into single-use bioreactors to provide operatorindependent information on viable biomass. That reduces the need for manual sampling through direct in-line measurements. By incorporating such sensors into a bioreactor control system, automatic monitoring and control of viable biomass will be feasible for continuous bioprocesses. Bioreactor and cell culture technology continue to evolve, allowing ever higher cell densities to be achieved and maintained. Whether biopharmaceutical companies choose to work with fed-batch processes or develop continuous cell culture methods will depend on their individual circumstances. However technological innovations will allow them to enhance their speed to clinical testing and develop robust high-titer processes that operate within predefined design spaces. References 1 Croughan M, Konstantinov KB, Cooney CL. The Future of Industrial Bioprocessing: Batch or Continuous? Biotechnol. Bioeng. 112(4) 2015: 648 651. 2 Warikoo V, et al. Integrated Continuous Production of Recombinant Therapeutic Proteins. Biotechnol. Bioeng. 109(12) 2012: 3018 3029. 3 Adams T, et al. Increasing Efficiency in Protein Supply and Cell Production By Combining Single-Use Bioreactor Technology and Perfusion. BioPharm Int. May 2011: S4 S11. 4 Fenge C, et al. Evaluation of a Spin Filter During Perfusion Culture of Recombinant CHO Cells. Animal Cell Technology: Development, Processes, and Products. Butterworth-Heinemann: Oxford, UK, 1992; 365 370. 5 De Wilde D, et al. Superior Scalability of Single-Use Bioreactors. BioProcess Int. 12(8) 2014: S14 S19. 6 Hammond M, et al. A Cytotoxic Leachable Compound from Single-Use Bioprocess Equipment That Causes Poor Cell Growth Performance. Biotechnol. Prog. 30, 2014: 332 337. 7 Fenge C, et al. Consistently Superior Cell Growth Achieved with New Polyethylene Film Formulation. BioProcess Int. 12(8) 2014: S34 S37. 8 Vachette E, et al. Robust and Convenient Single-Use Processing: The Superior Strength and Flexibility of Flexsafe Bags. BioProcess Int. 12(8) 2014: S38 S42. 9 Cappia JM, et al. Enhanced Assurance of Supply for Single-Use Bags. BioProcess Int. 12(8) 2014: S14 S19. 10 Reglin R, et al. Verification of New Flexsafe STR Single-Use Bioreactor Bags Using a CHO Fed-Batch Monoclonal Antibody Production Process at 1,000-L Scale. BioProcess Int. 12(8) 2014: S53 S57. Mikal Sherman was a applications specialist; Vincent Lam and Melissa Carpio are applications specialists; Nick Hutchinson is a technical writer; and corresponding author Christel Fenge, PhD, is vice president of marketing fermentation technologies, all in fermentation technologies at Sartorius Stedim Biotech GmbH, August-Spindler-Straße 11, DE-37079 Goettingen, Germany; christel.fenge@ sartorius.com. For reprints, contact Rhonda Brown of Foster Printing Service, rhondab@fosterprinting.com, 1-866-879-9144 x194. 28 BioProcess International 14(10)s November 2016 Supplement