Bioreactor productivity and media cost comparison for different intensified cell culture processes

Process intensification in biomanufacturing has attracted a great deal of interest in recent years. Manufacturing platform improvements leading to higher cell density and bioreactor productivity have been pursued. Here we evaluated a variety of intensified mammalian cell culture processes for producing monoclonal antibodies. Cell culture operational modes including fed‐batch (normal seeding density or high seeding density with N‐1 perfusion), perfusion, and concentrated fed‐batch (CFB) were assessed using the same media set with the same Chinese Hamster Ovary (CHO) cell line. Limited media modification was done to quickly fit the media set to different operational modes. Perfusion and CFB processes were developed using an alternating tangential flow filtration device. Independent of the operational modes, comparable cell specific productivity (fed‐batch: 29.4 pg/cell/day; fed‐batch with N‐1 perfusion: 32.0 pg/cell/day; perfusion: 31.0 pg/cell/day; CFB: 20.1 – 45.1 pg/cell/day) was reached with similar media conditions. Continuous media exchange enabled much higher bioreactor productivity in the perfusion (up to 2.29 g/L/day) and CFB processes (up to 2.04 g/L/day), compared with that in the fed‐batch processes (ranging from 0.39 to 0.49 g/L/day), largely due to the higher cell density maintained. Furthermore, media cost per gram of antibody produced from perfusion was found to be highly comparable with that from fed‐batch; and the media cost for CFB was the highest due to the short batch duration. Our experimental data supports the argument that media cost for perfusion process could be even lower than that in a fed‐batch process, as long as sufficient bioreactor productivity is achieved. © 2016 American Institute of Chemical Engineers Biotechnol. Prog., 33:867–878, 2017

[1]  Gerhard Schembecker,et al.  Cost evaluation of antibody production processes in different operation modes , 2016 .

[2]  H. Eisen,et al.  The secretion of antibody by isolated lymph node cells. , 1961, The Journal of biological chemistry.

[3]  Paul W. Sauer,et al.  A high-yielding, generic fed-batch cell culture process for production of recombinant antibodies. , 2000, Biotechnology and bioengineering.

[4]  S. Chamow,et al.  Therapeutic antibody expression technology. , 2001, Current opinion in biotechnology.

[5]  Brian Kelley,et al.  Very Large Scale Monoclonal Antibody Purification: The Case for Conventional Unit Operations , 2007, Biotechnology progress.

[6]  Marco Jenzsch,et al.  Optimizing capacity utilization by large scale 3000 L perfusion in seed train bioreactors , 2013, Biotechnology progress.

[7]  Mohamed Al-Rubeai,et al.  Animal Cell Culture , 2015, Cell Engineering.

[8]  Suzanne S Farid,et al.  Process economics of industrial monoclonal antibody manufacture. , 2007, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[9]  Yao-ming Huang,et al.  Perfusion seed cultures improve biopharmaceutical fed‐batch production capacity and product quality , 2014, Biotechnology progress.

[10]  Ye Zhang,et al.  Very High Density of CHO Cells in Perfusion by ATF or TFF in WAVE Bioreactor™. Part I. Effect of the Cell Density on the Process , 2013, Biotechnology progress.

[11]  H. Dosch,et al.  Limiting dilution analysis of the B cell compartment in human bone marrow , 1986, European journal of immunology.

[12]  Brian Kelley,et al.  Industrialization of mAb production technology: The bioprocessing industry at a crossroads , 2009, mAbs.

[13]  Florian M Wurm,et al.  Manufacturing recombinant proteins in kg-ton quantities using animal cells in bioreactors. , 2011, European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V.

[14]  Jochen Strube,et al.  Challenges in biotechnology production—generic processes and process optimization for monoclonal antibodies , 2005 .

[15]  Rashmi Kshirsagar,et al.  Concentrated fed-batch cell culture increases manufacturing capacity without additional volumetric capacity. , 2016, Journal of biotechnology.

[16]  Ningning Ma,et al.  A single nutrient feed supports both chemically defined NS0 and CHO fed‐batch processes: Improved productivity and lactate metabolism , 2009, Biotechnology progress.

[17]  Konstantin B Konstantinov,et al.  The future of industrial bioprocessing: batch or continuous? , 2015, Biotechnology and bioengineering.

[18]  M. Care,et al.  In Vitro Generation of Long-lived Human Plasma Cells , 2012, The Journal of Immunology.

[19]  B. Kelley,et al.  An improved manufacturing process for Xyntha/ReFacto AF , 2010, Haemophilia : the official journal of the World Federation of Hemophilia.

[20]  Gerhard Schembecker,et al.  Developing the biofacility of the future based on continuous processing and single-use technology. , 2015, Journal of biotechnology.

[21]  C. Hoy,et al.  Multiple cell culture factors can affect the glycosylation of Asn-184 in CHO-produced tissue-type plasminogen activator. , 2000, Biotechnology and bioengineering.

[22]  Suzanne S Farid,et al.  Fed‐batch and perfusion culture processes: Economic, environmental, and operational feasibility under uncertainty , 2013, Biotechnology and bioengineering.

[23]  Paola Lettieri,et al.  Life‐cycle and cost of goods assessment of fed‐batch and perfusion‐based manufacturing processes for mAbs , 2016, Biotechnology progress.

[24]  R. Werner,et al.  Economic aspects of commercial manufacture of biopharmaceuticals. , 2004, Journal of biotechnology.

[25]  Charles L. Cooney,et al.  White Paper on Continuous Bioprocessing , 2014 .

[26]  Suzanne S Farid,et al.  Established bioprocesses for producing antibodies as a basis for future planning. , 2006, Advances in biochemical engineering/biotechnology.

[27]  Sen Xu,et al.  High-density mammalian cell cultures in stirred-tank bioreactor without external pH control. , 2016, Journal of biotechnology.

[28]  M. Fussenegger,et al.  Molecular engineering of exocytic vesicle traffic enhances the productivity of Chinese hamster ovary cells , 2009, Biotechnology and bioengineering.

[29]  Konstantin B Konstantinov,et al.  White paper on continuous bioprocessing. May 20-21, 2014 Continuous Manufacturing Symposium. , 2015, Journal of pharmaceutical sciences.

[30]  Sen Xu,et al.  Improving lactate metabolism in an intensified CHO culture process: productivity and product quality considerations , 2016, Bioprocess and Biosystems Engineering.

[31]  T. Bibila,et al.  In Pursuit of the Optimal Fed‐Batch Process for Monoclonal Antibody Production , 1995, Biotechnology progress.

[32]  Jason Walther,et al.  Delivering Steady-State Product Quality with An Intensified And Integrated Perfusion Cell Culture Process , 2015 .

[33]  Daniel Cummings,et al.  Integrated continuous production of recombinant therapeutic proteins , 2012, Biotechnology and bioengineering.

[34]  Nimish Dalal,et al.  Cell culture process operations for recombinant protein production. , 2014, Advances in biochemical engineering/biotechnology.

[35]  Yashas Rajendra,et al.  Generation of stable Chinese hamster ovary pools yielding antibody titers of up to 7.6 g/L using the piggyBac transposon system , 2016, Biotechnology progress.

[36]  R. Ahmed,et al.  Humoral immunity due to long-lived plasma cells. , 1998, Immunity.

[37]  Mohamed Al-Rubeai,et al.  Bioreactor Systems for Producing Antibody from Mammalian Cells , 2011 .

[38]  Huong Nguyen,et al.  A new large‐scale manufacturing platform for complex biopharmaceuticals , 2012, Biotechnology and bioengineering.

[39]  Sevim Duvar,et al.  Developing an upstream process for a monoclonal antibody including medium optimization , 2013, BMC Proceedings.

[40]  Jason Walther,et al.  The business impact of an integrated continuous biomanufacturing platform for recombinant protein production. , 2015, Journal of biotechnology.

[41]  Michele M. Myers,et al.  Addressing Changes Associated with Technology Transfer: A Case Study , 2010 .

[42]  Alex Xenopoulos,et al.  A new, integrated, continuous purification process template for monoclonal antibodies: Process modeling and cost of goods studies. , 2015, Journal of biotechnology.

[43]  Thomas Ryll,et al.  Maximizing productivity of CHO cell‐based fed‐batch culture using chemically defined media conditions and typical manufacturing equipment , 2010, Biotechnology progress.

[44]  Marcella Yu,et al.  Effects of cell culture conditions on antibody N‐linked glycosylation—what affects high mannose 5 glycoform , 2011, Biotechnology and bioengineering.