Downstream Processing - Science topic

Fig. 2. Fourier transform infrared spectroscopy (FTIR) analysis of...

  • Recruit researchers
  • Join for free
  • Login Email Tip: Most researchers use their institutional email address as their ResearchGate login Password Forgot password? Keep me logged in Log in or Continue with Google Welcome back! Please log in. Email · Hint Tip: Most researchers use their institutional email address as their ResearchGate login Password Forgot password? Keep me logged in Log in or Continue with Google No account? Sign up

Information

  • Author Services

Initiatives

You are accessing a machine-readable page. In order to be human-readable, please install an RSS reader.

All articles published by MDPI are made immediately available worldwide under an open access license. No special permission is required to reuse all or part of the article published by MDPI, including figures and tables. For articles published under an open access Creative Common CC BY license, any part of the article may be reused without permission provided that the original article is clearly cited. For more information, please refer to https://www.mdpi.com/openaccess .

Feature papers represent the most advanced research with significant potential for high impact in the field. A Feature Paper should be a substantial original Article that involves several techniques or approaches, provides an outlook for future research directions and describes possible research applications.

Feature papers are submitted upon individual invitation or recommendation by the scientific editors and must receive positive feedback from the reviewers.

Editor’s Choice articles are based on recommendations by the scientific editors of MDPI journals from around the world. Editors select a small number of articles recently published in the journal that they believe will be particularly interesting to readers, or important in the respective research area. The aim is to provide a snapshot of some of the most exciting work published in the various research areas of the journal.

Original Submission Date Received: .

  • Active Journals
  • Find a Journal
  • Proceedings Series
  • For Authors
  • For Reviewers
  • For Editors
  • For Librarians
  • For Publishers
  • For Societies
  • For Conference Organizers
  • Open Access Policy
  • Institutional Open Access Program
  • Special Issues Guidelines
  • Editorial Process
  • Research and Publication Ethics
  • Article Processing Charges
  • Testimonials
  • Preprints.org
  • SciProfiles
  • Encyclopedia

ijms-logo

Article Menu

research papers on downstream processing

  • Subscribe SciFeed
  • Recommended Articles
  • PubMed/Medline
  • Google Scholar
  • on Google Scholar
  • Table of Contents

Find support for a specific problem in the support section of our website.

Please let us know what you think of our products and services.

Visit our dedicated information section to learn more about MDPI.

JSmol Viewer

Recent advances and future directions in downstream processing of therapeutic antibodies.

research papers on downstream processing

1. Introduction

2. developability assessment for lead antibody molecules, 3. adoption of single-use technologies, 4. continuous downstream processing, 5. mechanistic and statistical process modelling, 6. process analytical technologies, 7. summary and conclusions, conflicts of interest.

  • Vatsa, S. In Silico prediction of post-translational modifications in therapeutic antibodies. mAbs 2022 , 14 , 2023938. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Liu, H.; Ponniah, G.; Zhang, H.-M.; Nowak, C.; Neil, A.; Gonzalez-Lopez, N.; Patel, R.; Cheng, G.; Kita, A.Z.; Andrien, B. In vitro and in vivo modifications of recombinant and human IgG antibodies. mAbs 2014 , 6 , 1145–1154. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Kerwin, B.A.; Bennett, C.; Brodsky, Y.; Clark, R.; Floyd, J.A.; Gillespie, A.; Mayer, B.T.; McClure, M.; Siska, C.; Seaman, M.S.; et al. Framework mutations of the 10-1074 bnAb increase conformational stability, manufacturability, and stability while preserving full neutralization activity. J. Pharm. Sci. 2020 , 109 , 233–246. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Chi, B.; De Oliveira, G.; Gallagher, T.; Mitchell, L.; Knightley, L.; Gonzalez, C.C.; Russell, S.; Germaschewski, V.; Pearce, C.; Sellick, C.A. Pragmatic mAb lead molecule engineering from a developability perspective. Biotechnol. Bioeng. 2021 , 118 , 3733–3743. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Seeliger, D.; Schulz, P.; Litzenburger, T.; Spitz, J.; Hoerer, S.; Blech, M.; Enenkel, B.; Studts, J.; Garidel, P.; Karow, A.R. Boosting antibody developability through rational sequence optimization. mAbs 2015 , 7 , 505–515. [ Google Scholar ] [ CrossRef ]
  • Robotham, A.C.; Kelly, J.F. Detection and quantification of free sulfhydryls in monoclonal antibodies using maleimide labeling and mass spectrometry. mAbs 2019 , 11 , 757–766. [ Google Scholar ] [ CrossRef ]
  • Kim, N.A.; Kar, S.; Li, Z.; Das, T.K.; Carpenter, J.F. Mimicking low pH virus inactivation used in antibody manufacturing proceses: Effect of processing conditions and biophysical properties on antibody aggregation and particle formation. J. Pharm. Sci. 2021 , 110 , 3188–3199. [ Google Scholar ] [ CrossRef ]
  • Wãlchli, R.; Ressurreição, M.; Vogg, S.; Feidl, F.; Angelo, J.; Xu, X.; Ghose, S.; Li, Z.J.; Le Saoût, X.; Souquet, J.; et al. Understanding mAb aggregation during low pH viral inactivation and subsequent neutralization. Biotechnol. Bioeng. 2020 , 117 , 687–700. [ Google Scholar ] [ CrossRef ]
  • Saleh, D.; Hess, R.; Ahlers-Hesse, M.; Beckert, N.; Schönberger, M.; Rischawy, R.; Wang, G.; Bauer, J.; Blech, M.; Kluters, S.; et al. Modeling the impact of amino acid substitution in a monoclonal antibody on cation exchange chromatography. Biotechnol. Bioeng. 2021 , 118 , 2923–2933. [ Google Scholar ] [ CrossRef ]
  • Strop, P.; Ho, W.-H.; Boustany, L.M.; Abdiche, Y.N.; Lindquist, K.C.; Farias, S.E.; Rickert, M.; Appah, C.T.; Pascua, E.; Radcliffe, T.; et al. Generating bispecific human IgG1 and IgG2 antibodies from any antibody pair. J. Mol. Biol. 2012 , 420 , 204–219. [ Google Scholar ] [ CrossRef ]
  • Kittelmann, J.; Lang, K.M.H.; Ottens, M.; Hubbuch, J. Orientation of monoclonal antibodies in ion-exchange chromatography: A predictive quantitative structure-activity relationship modeling approach. J. Chromatogr. A 2017 , 1510 , 33–39. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Gudhka, R.B.; Bilodeau, C.L.; McCallum, S.A.; McCoy, M.A.; Roush, D.J.; Snyder, M.A.; Crammer, S.M. Identification of preferred multi-modal ligand-binding regions on IgG1 Fc using nuclear magnetic resonance and molecular dynamics simulations. Biotechnol. Bioeng. 2021 , 118 , 809–822. [ Google Scholar ] [ CrossRef ]
  • Lopes, A.G. Single-use in the biopharmaceutical industry: A review of current technology impact, challenges and limitation. Food Bioprod. Process. 2015 , 93 , 98–114. [ Google Scholar ] [ CrossRef ]
  • Carlbungco, C. Stainless Steel and Single-Use Systems: Observations from COVID-19. BioProcess International (Supply Chain Management: SUS in COVID-19). 2020. Available online: https://bioprocessintl.com/sponsored-content/stainless-steel-and-single-use-systems-supply-chain-management-observations-from-covid-19/ (accessed on 27 May 2022).
  • Barbaroux, M.; Horowski, B.; Mokuolu, S.; Petrich, M.; Whitford, W.; BPSA Sustainability Subcommittee and Flanagan, B. The green imperative. Part One: Life-cycle assessment and sustainability for single-use technologies in the biopharmaceutical industry. BioProcess Int. 2020 , 18 , 12–19. [ Google Scholar ]
  • Jacquemart, R.; Vandersluis, M.; Zhoa, M.; Sukhija, K.; Sidhu, N.; Stout, J. A single-use strategy to enable manufacturing of affordable biologics. Comput. Struct. Biotechnol. J. 2016 , 14 , 309–318. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Hummel, J.; Pagkaliwangan, M.; Gjoka, X.; Davidovits, T.; Stock, R.; Ransohoff, T.; Gantier, R.; Schofield, M. Modeling the downstream processing of monoclonal antibodies reveals cost advantages for continuous methods for a broad range of manufacturing scales. Biotechnol. J. 2019 , 14 , 1700665. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Bio-Process Systems Alliance (BPSA). X-ray Sterilization of Single-Use Bioprocess Equipment. Part 1. Industry Need 2021, Requirements and Risk Evaluation. Available online: www.bpsalliance.org (accessed on 20 April 2022).
  • Menzel, R.; Dorey, S.; Maier, T.; Pahl, I.; Hauk, A. X-ray sterilization of biopharmaceutical manufacturing equipment—Extractables profile of a film material and copolyester Tritan compared to gamma irradiation. Biotechnol. Prog. 2021 , 38 , e3214. [ Google Scholar ] [ CrossRef ]
  • Gaston, F.; Dupuy, N.; Marque, S.R.A.; Dorey, S. Evaluation of multilayer film stability by Raman spectroscopy after gamma-irradiation sterilization process. Vib. Spectrosc. 2018 , 96 , 52–59. [ Google Scholar ] [ CrossRef ]
  • Gaston, F.; Dupuy, N.; Marque, S.R.A.; Barbaroux, M.; Dorey, S. FTIR study of ageing of –irradiated biophamaceutical EVA based film. Polym. Degrad. Stab. 2016 , 129 , 19–25. [ Google Scholar ]
  • Madsen, B.; Britt, D.W.; Griffiths, F.; McKenna, E.; Ho, C.-H. Effect of sterilization techniques on the physiochemical properties of polysulfone hollow fibers. J. Appl. Polym. Sci. 2010 , 119 , 3429–3436. [ Google Scholar ] [ CrossRef ]
  • Gao, Y.; Allison, N. Extractables and leachables issues with the application of single-use technology in the biopharmaceutical industry. J. Chem. Technol. Biotechnol. 2015 , 91 , 289–295. [ Google Scholar ] [ CrossRef ]
  • Kavara, A.; Sokolowski, D.; Collins, M.; Schofield, M. Chapter 4—Recent advances in continuous downstream processing of antibodies and related products. In Approaches to the Purification, Analysis and Characterization of Antibody-Based Therapeutics ; Matte, A., Ed.; Elsevier: Amsterdam, The Netherlands, 2020; pp. 81–103. [ Google Scholar ]
  • Khanal, O.; Lenhoff, A.M. Developments and opportunities in continuous biopharmaceutical manufacturing. mAbs 2021 , 13 , e1903664. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Mahal, H.; Branton, H.; Farid, S.S. End-to-end continuous bioprocessing: Impact on facility design, cost of goods, and cost of development for monoclonal antibodies. Biotechnol. Bioeng. 2021 , 118 , 3468–3485. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kelly, W.; Scully, J.; Zhang, D.; Feng, G.; Lavengood, M.; Condon, J.; Knighton, J.; Bhatia, R. Understanding and modeling alternating tangential flow filtration for perfusion cell culture. Biotechnol. Prog. 2014 , 30 , 1291–1300. [ Google Scholar ] [ CrossRef ]
  • Madsen, E.; Kaiser, J.; Krühne, U.; Pinelo, M. Single pass tangential flow filtration: Critical operational variables, fouling, and main current applications. Sep. Purif. Technol. 2022 , 291 , 120949. [ Google Scholar ] [ CrossRef ]
  • Tan, R.; Hezel, F.; Franzreb, M. Continuous single pass diafiltration with alternating permeate flow direction for high efficiency buffer exchange. J. Memb. Sci. 2021 , 619 , 118695. [ Google Scholar ] [ CrossRef ]
  • Fedorenko, D.; Dutta, A.K.; Tan, J.; Walko, J.; Brower, M.; Pinto, N.D.S.; Zydney, A.L.; Shinkazh, O. Improved protein A resin for antibody capture in a continuous countercurrent tangential chromatography system. Biotechnol. Boeng. 2020 , 117 , 646–653. [ Google Scholar ] [ CrossRef ]
  • Amritar, V.; Adat, S.; Tejwani, V.; Rathore, A.; Bhambure, R. Engineering Staphylococcal Protein A for high-throughput affinity purification of monoclonal antibodies. Biotechnol. Adv. 2020 , 44 , 107632. [ Google Scholar ] [ CrossRef ]
  • Napoleone, A.; Lauén, I.; Linkgreim, T.; Dahllund, L.; Persson, H.; Andersson, O.; Olsson, A.; Hultqvist, G.; Frank, P.; Hall, M.; et al. Fed-batch production assessment of a tetravalent bispecific antibody: A case study on piggyBac stably transfected HEK293 cells. New Biotechnol. 2021 , 65 , 9–19. [ Google Scholar ] [ CrossRef ]
  • Davis, R.R.; Suber, F.; Heller, I.; Yang, B.; Martinez, J. Improving mAb capture productivity on batch and continuous processing using nanofiber PrismA adsorbents. J. Biotechnol. 2021 , 336 , 50–55. [ Google Scholar ] [ CrossRef ]
  • Nadar, S.; Shooter, G.; Somasundaram, B.; Shave, E.; Baker, K.; Lua, L.H.L. Intensified downstream processing of monoclonal antibodies using membrane technology. Biotechnol. J. 2021 , 16 , 2000309. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Dods, S.R.; Hardick, O.; Stevens, B.; Bracewell, D.G. Fabricating electrospun cellulose nanofiber adsorbents for ion-exchange chromatography. J. Chromatogr. A 2015 , 1376 , 74–83. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Arnold, L.; Lee, K.; Rucker-Pezzini, J.; Lee, J.H. Implementation of fully integrated continuous antibody processing: Effects on productivity and COGm. Biotechnol. J. 2019 , 14 , 1800061. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Gillespie, C.; Holstein, M.; Mullin, L.; Cotoni, K.; Tuccelli, R.; Caulmare, J.; Greenhalgh, P. Continuous in-line virus inactivation for next generation bioprocessing. Biotechnol. J. 2019 , 14 , 1700718. [ Google Scholar ] [ CrossRef ]
  • Martins, D.L.; Sencar, J.; Hammerschmidt, N.; Flicker, A.; Kindermann, J.; Kreil, T.R.; Jungbauer, A. Truly continuous low pH viral inactivation for biopharmaceutical process integration. Biotechnol. Bioeng. 2020 , 117 , 1406–1417. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kateja, N.; Nitika, N.; Fadnis, R.S.; Rathore, A.S. A novel reactor configuration for continuous virus inactivation. Biochem. Eng. J. 2021 , 167 , 107885. [ Google Scholar ] [ CrossRef ]
  • Pollard, J.; McDonald, P.; Hesslein, A. Lessons learned in building high-throughput process development capabilities. Eng. Life Sci. 2017 , 16 , 93–98. [ Google Scholar ] [ CrossRef ]
  • Kumar, V.; Bhalla, A.; Rathore, A.S. Design of experiments applications in bioprocessing: Concepts and approaches. Biotechnol. Prog. 2014 , 30 , 86–99. [ Google Scholar ] [ CrossRef ]
  • Kumar, V.; Lenhoff, A.M. Mechanistic modelling of preparative chromatography for biotherapeutics. Ann. Rev. Chem. Biomol. Eng. 2020 , 11 , 235–255. [ Google Scholar ] [ CrossRef ]
  • Babi, D.K.; Griesbach, J.; Hunt, S.; Insaidoo, F.; Roush, D.; Todd, R.; Staby, A.; Welsh, J.; Wittkopp, F. Opportunities and challenges for model utilization in the biopharmaceutical industry: Current versus future state. Curr. Opin. Chem. Eng. 2022 , 36 , 100813. [ Google Scholar ] [ CrossRef ]
  • Hanke, A.T.; Ottens, M. Purifying biopharmaceuticals: Knowledge-based chromatographic process development. Trends Biotechnol. 2014 , 22 , 210–220. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Benner, S.W.; Welsh, J.P.; Rauscher, M.A.; Pollard, J.M. Prediction of lab and manufacturing scale chromatography using min-columns and mechanistic modelling. J. Chromatogr. A 2019 , 1593 , 54–62. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Schmölder, J.; Kaspereit, M. A modular framework for the modelling and optimization of advanced chromatographic processes. Processes 2020 , 8 , 65. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Leweke, S.; von Lieres, E. Chromatography analysis and design toolkit (CADET). Comput. Chem. Eng. 2018 , 113 , 274–294. [ Google Scholar ] [ CrossRef ]
  • Aspen Chromatography™. Improving Process Design. Available online: https://www.aspentech.com/en/%20products/pages/aspen-chromatography (accessed on 7 June 2022).
  • Ypso-Ionic®. Available online: https://www.ypso-ionic.com. (accessed on 7 June 2022).
  • GoSilico™. Chromatography Modeling. Available online: https://www.cytivalifesciences.com/en/us/shop/chromatography/chromatography-modeling (accessed on 3 August 2022).
  • Rischawy, F.; Saleh, D.; Tobias, H.; Oelmeier, S.; Spitz, J.; Kluters, S. Good modelling practise for industrial chromatography: Mechanistic modelling of ion exchange chromatography of a bispecific antibody. Comput. Chem. Eng. 2019 , 130 , 106532. [ Google Scholar ] [ CrossRef ]
  • Briskot, T.; Stückler, F.; Wittkopp, F.; Williams, C.; Yang, J.; Konrad, S.; Doninger, K.; Griesbach, J.; Bennecke, M.; Hepbildikler, S.; et al. Prediction uncertainty assessment of chromatography models using Bayesian inference. J. Chromatogr. A 2019 , 1587 , 101–110. [ Google Scholar ] [ CrossRef ]
  • Shekhawat, L.K.; Kumar, V.; Rathore, A.S. Design of experiments applications in bioprocessing: Chromatography process development using split design of experiments. Biotechnol. Prog. 2018 , 35 , e2730. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Shekhawat, L.K.; Manvar, A.P.; Rathore, A.S. Enablers for QbD implementation: Mechanistic modeling for ion-exchange membrane chromatography. J. Memb. Sci. 2016 , 500 , 86–98. [ Google Scholar ] [ CrossRef ]
  • Sanchez-Reyes, G.; Graalfs, H.; Hafner, M.; Frech, C. Mechanistic modelling of ligand density variations on anion exchange chromatography. J. Sep. Sci. 2021 , 44 , 805–821. [ Google Scholar ] [ CrossRef ]
  • Gerzon, G.; Sheng, Y.; Kirkitadze, M. Process analytical technologies—Advances in bioprocess integration and future perspectives. J. Pharm. Biomed. Anal. 2021 , 207 , 114379. [ Google Scholar ] [ CrossRef ]
  • Maruthamuthu, M.K.; Rudge, S.R.; Ardekani, A.M.; Ladisch, M.R.; Verma, M.S. Process analytical technologies and data analytics for the manufacture of monoclonal antibodies. Trends Biotechnol. 2020 , 38 , 1169–1186. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • São Pedro, M.N.; Klijn, M.E.; Eppink, M.H.M.; Ottens, M. Process analytical technique (PAT) miniaturization for monoclonal antibody aggregate detection in continuous downstream processing. J. Chem. Technol. Biotechnol. 2021 . [ Google Scholar ] [ CrossRef ]
  • Wasalathanthri, D.P.; Rehmann, M.S.; Song, Y.; Gu, Y.; Mi, L.; Shao, C.; Chemmalil, L.; Lee, J.; Ghose, S.; Borys, M.C.; et al. Technology outlook for real-time quality attribute and process parameter monitoring in biopharmaceutical development—A review. Biotechnol. Bioeng. 2020 , 117 , 3182–3198. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Wasalathanthri, D.P.; Shah, R.; Ding, J.; Leone, A.; Li, Z.J. Process analytics 4.0. A paradigm shift in rapid analytics for biologics development. Biotechnol. Prog. 2021 , 37 , e3177. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Jiang, M.; Severson, K.A.; Love, J.C.; Madden, H.; Swann, P.; Zang, L.; Baatz, R.D. Opportunities and challenges of real-time release testing in biopharmaceutical manufacturing. Biotechnol. Bioeng. 2017 , 114 , 2445–2456. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Brestrich, N.; Rüdt, M.; Büchler, D.; Hubbuch, J. Selective protein quantification for preparative chromatography using variable pathelength UV/Vis spectroscopy and partial least squares regression. Chem. Engin. Sci. 2018 , 176 , 157–164. [ Google Scholar ] [ CrossRef ]
  • Ly, T.T.; Ruan, Y.; Du, B.; Jia, P.; Zhang, H. Fibre-optic surface plasmon resonance biosensor for monoclonal antibody titer quantification. Biosensors 2021 , 11 , 383. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Feidl, F.; Garbellini, S.; Luna, M.F.; Vogg, S.; Souquet, J.; Broly, H.; Morbidelli, M.; Butté, A. Combining mechanistic modeling and Raman spectroscopy for monitoring antibody chromatographic purification. Processes 2019 , 7 , 683. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Feidl, F.; Garbellini, S.; Vogg, S.; Sokolov, M.; Souquet, J.; Broly, H.; Butte, A.; Morbidelli, M. A new flow cell and chemometric protocol for implementing in-line Raman spectroscopy in chromatography. Biotechnol. Prog. 2019 , 35 , e2847. [ Google Scholar ] [ CrossRef ]
  • Rolinger, L.; Rüdt, M.; Hubbuch, J. Comparison of UV- and Raman-based monitoring of the Protein A load phase and evaluation of data fusion by PLS models and CNNs. Biotechnol. Bioeng. 2021 , 118 , 4255–4268. [ Google Scholar ] [ CrossRef ]
  • Yilmaz, D.; Mehdizadeh, H.; Navarro, D.; Shehzad, A.; O’Connor, M.; McCormick, P. Application of Raman spectroscopy in monoclonal antibody producing continuous systems for downstream process intensification. Biotechnol. Prog. 2019 , 36 , e2947. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Patel, B.A.; Gospodarek, A.; Larkin, M.; Kenrick, S.A.; Haverick, M.A.; Tugcu, N.; Brower, M.A.; Richardson, D.D. Multi-angle light scattering as a process analytical technology measuring real-time molecular weight for downstream process control. mAbs 2018 , 10 , 945–950. [ Google Scholar ] [ CrossRef ]
  • Tiwari, A.; Kateja, N.; Chanana, S.; Rathore, A.S. Use of HPLC as an enabler of process analytical technology in process chromatography. Anal. Chem. 2018 , 90 , 7824–7829. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Chemmalil, L.; Prabhakar, T.; Kuang, J.; West, J.; Tan, Z.; Ehamparanathan, V.; Song, Y.; Xu, J.; Ding, J.; Li, Z. Online/at-line measurement, analysis and control of product titre and critical product quality attributes (CQAs) during process development. Biotechnol. Bioeng. 2020 , 117 , 3757–3765. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Gillespie, C.; Wasalathanthri, D.P.; Ritz, D.B.; Zhou, G.; Davis, K.A.; Wucherpfennig, T.; Hazelwood, N. Systematic assessment of process analytical technologies for biologics. Biotechnol. Bioeng. 2021 , 119 , 423–434. [ Google Scholar ] [ CrossRef ]
  • West, J.M.; Feroz, H.; Xu, X.; Puri, N.; Holstein, M.; Ghose, S.; Ding, J.; Li, Z.J. Process analytical technology for on-line monitoring of quality attributes during single-use ultrafiltration/diafiltration. Biotechnol. Bioeng. 2021 , 118 , 2293–2300. [ Google Scholar ] [ CrossRef ]
  • Liu, Y.; Zhang, C.; Chen, J.; Fernandez, J.; Vellala, P.; Kulkarni, T.A.; Aguilar, I.; Ritz, D.; Lan, K.; Patel, P.; et al. A fully integrated online platform for real-time monitoring of multiple product quality attributes in biopharmaceutical processes for monoclonal antibody therapeutics. J. Pharm. Sci. 2022 , 111 , 358–367. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Rogers, R.S.; Abernathy, M.; Richardson, D.D.; Rouse, J.C.; Sperry, J.B.; Swann, P.; Wypych, J.; Yu, C.; Zhang, L.; Deshpande, R. A view on the importance of “multi attribute method” for measuring purity of biopharmaceuticals and improving overall control strategy. AAPS J. 2018 , 20 , 7. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Rogstad, S.; Yan, H.; Wang, X.; Powers, D.; Brorson, K.; Damdinsuren, B.; Lee, S. Multi-attribute for quality control of therapeutic proteins. Anal. Chem. 2019 , 91 , 14170–14177. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Jakes, C.; Füssl, F.; Zaborowska, I.; Bones, J. Rapid analysis of biotherapeutics using Protein A chromatography coupled to Orbitrap mass spectrometry. Anal. Chem. 2021 , 93 , 13505–13512. [ Google Scholar ] [ CrossRef ]

Click here to enlarge figure

MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

Matte, A. Recent Advances and Future Directions in Downstream Processing of Therapeutic Antibodies. Int. J. Mol. Sci. 2022 , 23 , 8663. https://doi.org/10.3390/ijms23158663

Matte A. Recent Advances and Future Directions in Downstream Processing of Therapeutic Antibodies. International Journal of Molecular Sciences . 2022; 23(15):8663. https://doi.org/10.3390/ijms23158663

Matte, Allan. 2022. "Recent Advances and Future Directions in Downstream Processing of Therapeutic Antibodies" International Journal of Molecular Sciences 23, no. 15: 8663. https://doi.org/10.3390/ijms23158663

Article Metrics

Article access statistics, further information, mdpi initiatives, follow mdpi.

MDPI

Subscribe to receive issue release notifications and newsletters from MDPI journals

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

Current research approaches in downstream processing of pharmaceutically relevant proteins

Affiliations.

  • 1 Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria; Bioseparation Engineering Group, School of Engineering and Design, Technical University of Munich, Garching, Germany. Electronic address: [email protected].
  • 2 Bioseparation Engineering Group, School of Engineering and Design, Technical University of Munich, Garching, Germany.
  • 3 Bioseparation Engineering Group, School of Engineering and Design, Technical University of Munich, Garching, Germany. Electronic address: [email protected].
  • PMID: 35930843
  • DOI: 10.1016/j.copbio.2022.102768

Biopharmaceuticals and their production are on the rise. They are needed to treat and to prevent multiple diseases. Therefore, an urgent need for process intensification in downstream processing (DSP) has been identified to produce biopharmaceuticals more efficiently. The DSP currently accounts for the majority of production costs of pharmaceutically relevant proteins. This short review gathers essential research over the past 3 years that addresses novel solutions to overcome this bottleneck. The overview includes promising studies in the fields of chromatography, aqueous two-phase systems, precipitation, crystallization, magnetic separation, and filtration for the purification of pharmaceutically relevant proteins.

Copyright © 2022 The Author(s). Published by Elsevier Ltd.. All rights reserved.

PubMed Disclaimer

Publication types

  • Search in MeSH

Related information

  • PubChem Compound (MeSH Keyword)

LinkOut - more resources

Full text sources.

  • ClinicalKey
  • Elsevier Science

Miscellaneous

  • NCI CPTAC Assay Portal

full text provider logo

  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

downstream processing Recently Published Documents

Total documents.

  • Latest Documents
  • Most Cited Documents
  • Contributed Authors
  • Related Sources
  • Related Keywords

Total In Vitro Biosynthesis of the Thioamitide Thioholgamide and Investigation of the Pathway

Thioholgamides are ribosomally synthesized and post-translationally modified peptides (RiPPs) with potent activity against cancerous cell lines and an unprecedented structure. Despite being one of the most structurally and chemically complex RiPPs, very few biosynthetic steps have been elucidated. Here, we report the complete in vitro reconstitution of the biosynthetic pathway. We demonstrate that thioamidation is the first step and acts as a gatekeeper for downstream processing. Thr dehydration follows thioamidation, and our studies reveal that both these modifications require the formation of protein complexes – ThoH/I and ThoC/D. Harnessing the power of AlphaFold we deduce that ThoD acts as a lyase and also propose putative catalytic residues. ThoF catalyzes the oxidative decarboxylation of the terminal Cys and the subsequent macrocyclization is facilitated by ThoE. This is followed by Ser dehydration, which is also carried out by ThoC/D. ThoG is responsible for histidine bis-N-methylation, which is a prerequisite for His β-hydroxylation – a modification carried out by ThoJ. The last step of the pathway is the removal of the leader peptide by ThoK to afford mature thioholgamide.

Explaining Valence Asymmetries in Value Learning: A Reinforcement Learning Account

To understand how acquired value impacts how we perceive and process stimuli, psychologists have developed the Value Learning Task (VLT; e.g., Raymond & O’Brien, 2009). The task consists of a series of trials in which participants attempt to maximize accumulated winnings as they make choices from a pair of presented images associated with probabilistic win, loss, or no-change outcomes. Despite the task having a symmetric outcome structure for win and loss pairs, people learn win associations better than loss associations (Lin, Cabrera-Haro, & Reuter-Lorenz, 2020). This asymmetry could lead to differences when the stimuli are probed in subsequent tasks, compromising inferences about how acquired value affects downstream processing. We investigate the nature of the asymmetry using a standard error-driven reinforcement learning model with a softmax choice rule. Despite having no special role for valence, the model yields the asymmetry observed in human behavior, whether the model parameters are set to maximize empirical fit, or task payoff. The asymmetry arises from an interaction between a neutral initial value estimate and a choice policy that exploits while exploring, leading to more poorly discriminated value estimates for loss stimuli. We also show how differences in estimated individual learning rates help to explain individual differences in the observed win-loss asymmetries, and how the final value estimates produced by the model provide a simple account of a post-learning explicit value categorization task.

Wood Ash Based Treatment of Anaerobic Digestate: State-of-the-Art and Possibilities

The problem of current agricultural practices is not limited to land management but also to the unsustainable consumption of essential nutrients for plants, such as phosphorus. This article focuses on the valorization of wood ash and anaerobic digestate for the preparation of a slow-release fertilizer. The underlying chemistry of the blend of these two materials is elucidated by analyzing the applications of the mixture. First, the feasibility of employing low doses (≤1 g total solids (TS) ash/g TS digestate) of wood ash is explained as a way to improve the composition of the feedstock of anaerobic digestion and enhance biogas production. Secondly, a detailed description concerning high doses of wood ash and their uses in the downstream processing of the anaerobic digestate to further enhance its stability is offered. Among all the physico-chemical phenomena involved, sorption processes are meticulously depicted, since they are responsible for nutrient recovery, dewatering, and self-hardening in preparing a granular fertilizer. Simple activation procedures (e.g., carbonization, carbonation, calcination, acidification, wash, milling, and sieving) are proposed to promote immobilization of the nutrients. Due to the limited information on the combined processing of wood ash and the anaerobic digestate, transformations of similar residues are additionally considered. Considering all the possible synergies in the anaerobic digestion and the downstream stages, a dose of ash of 5 g TS ash/g TS digestate is proposed for future experiments.

Production, downstream processing, and characterization of polyhydroxyalkanoates (PHAs) boosted by pyruvate supplement using mixed microbial culture (MMC) and organic wastewater

Enhancing downstream processing of algal-based biofuel using novel fused bacteria and green solvents.

The present study investigated the utilization of algal biomass to produce bio-oil and acetone, butanol, and ethanol (ABE) products. Novel Clostridia fusants (C. beijernickii + C. thermocellum-CbCt and C. acetobutylicum + C. thermocellocum-CaCt) were developed using protoplast fusion technique and subsequently subjected to UV radiation for strain enhancement. Resultant mutated fusants showed improvement in thermal stability and higher resistance to biobutanol toxicity. Algal biomass was initially subjected to various hydrolysis treatments prior to fermentation. Combination treatment of thermal, chemical, and enzymatic resulted in maximum sugar release of 27.78 g/L. Maximum biobutanol concentration from fermentation using CbCt resulted in 7.98 g/L. Fermentation using CaCt produced a concentration of 7.39 g/L. Oil extraction from virgin algae investigated a green, bio-based approach using terpenes with ultrasonication and a modified, Bligh and Dyer method, separately. Combination method, ultrasonication followed by the modified Bligh and Dyer, resulted in oil yield of 46.27% (dlimonene) and 39.85% (p-cymene). Oil extraction was also produced from an algae sample following fermentation. Combined extraction method using fermentation sample resulted in oil yield of 65.04%.

5 Bioconversion and downstream processing in the context of biorefinery: Principles and process examples

All-in-one superparamagnetic and sers-active niosomes for dual-targeted in vitro detection of breast cancer cells.

Abstract BackgroundIn vitro and in vivo biosensing through surface-enhanced Raman scattering often suffer from signal contamination diminishing both the limit of detection and quantification. However, overcoming the lack of specificity requires excessive nanoparticle concentrations, which may lead to adverse side effects if applied to patients. ResultsWe propose encapsulation of iron oxide (FexOy) and gold (Au) nanoparticles (NPs) into the bilayer structure of transferrin-modified niosomes. This approach enables achieving greatly enhanced and contamination-free SERS-signals in vitro as well as a dual-targeting functionality towards MCF-7 breast cancer cells. An in-depth characterization of FexOyNPs- and AuNPs-loaded niosomes (AuNPs/FexOyNPs/NIO) after magnetic downstream processing reveals defined hybrid niosome structures, which show a long-term SERS-signal stability in various media such as MCF-7 cell culture medium. In vitro 2D-SERS imaging unveil a successful incorporation of a non-toxic dose of hybrid NPs into MCF-7 cells, which leads to strong and almost contamination-free SERS-signals. The measured signal-to-noise ratio of the in vitro signal exceeds the values required by DIN 32645 for the successful validation of a detection method. ConclusionsThe hybrid niosomes can be considered a promising and efficient agent for the establishment and commercialization of a highly sensitive detection kit for monitoring cancerous tissue.

Commercial-scale Economic Comparison of Different Batch Modes for Upstream and Downstream Processing of Monoclonal Antibody

Nanovaccine delivery approaches and advanced delivery systems for the prevention of viral infections: from development to clinical application.

Viral infections causing pandemics and chronic diseases are the main culprits implicated in devastating global clinical and socioeconomic impacts, as clearly manifested during the current COVID-19 pandemic. Immunoprophylaxis via mass immunisation with vaccines has been shown to be an efficient strategy to control such viral infections, with the successful and recently accelerated development of different types of vaccines, thanks to the advanced biotechnological techniques involved in the upstream and downstream processing of these products. However, there is still much work to be done for the improvement of efficacy and safety when it comes to the choice of delivery systems, formulations, dosage form and route of administration, which are not only crucial for immunisation effectiveness, but also for vaccine stability, dose frequency, patient convenience and logistics for mass immunisation. In this review, we discuss the main vaccine delivery systems and associated challenges, as well as the recent success in developing nanomaterials-based and advanced delivery systems to tackle these challenges. Manufacturing and regulatory requirements for the development of these systems for successful clinical and marketing authorisation were also considered. Here, we comprehensively review nanovaccines from development to clinical application, which will be relevant to vaccine developers, regulators, and clinicians.

Export Citation Format

Share document.

Downstream Processing for Biopharmaceuticals Recovery

  • First Online: 10 January 2019

Cite this chapter

research papers on downstream processing

  • Anu Mehta 8  

Part of the book series: Environmental Chemistry for a Sustainable World ((ECSW,volume 26))

1173 Accesses

9 Citations

The invention of genetic engineering tools has given birth to a new type of pharmaceuticals known as biopharmaceuticals. These are the drug molecules that have therapeutic effects and are synthesised in biological cell systems. Drug like recombinant insulin is a prominent prototype example of biopharmaceutical which is commonly available in the market at cheap prices for diabetic patients, globally. The production of these therapeutic molecules differs from chemically synthesised low molecular weight drugs. Upstream and downstream processes altogether comprise the production process of biopharmaceuticals. The downstream processing costs 70% of the total production cost of a particular biopharmaceutical, largely contributed by expensive chromatographic techniques such as affinity, hydrophobic interaction, ion exchange and size exclusion. Although chromatography is a reliable and conventional approach to carry out single step purification of biopharmaceuticals, the columns are run in a series to increase the purification fold. This makes the process tedious, and problems like diffusional spreading and resolution are also observed with chromatography procedures. The concern is important as we aim to bring various biopharmaceuticals into market that can treat innumerable diseases at a cheap price.

The current chapter emphasises the process and technology related to the upstream process and the three chronological steps – initial recovery, purification and polishing – involved in downstream processing of biopharmaceuticals. The chapter encompasses the hurdles encountered in the downstream processing in particular with chromatography process that makes high-quality production of biopharmaceuticals an expensive affair thus making it difficult to reach the public. New technologies designed to offer faster and cheaper purification such as aqueous two-phase extraction system, and nano-magnetic-based antibodies separation system have been discussed further. Moreover, we have reviewed and emphasised the requirement of using combination of physical, mathematical, biological and computational approaches, which can help to design efficient production and purification systems for the ample, cheap and continuous market supply of this new category of drugs.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Subscribe and save.

  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
  • Available as EPUB and PDF
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

research papers on downstream processing

Scale-Up of Protein Purification: Downstream Processing Issues

research papers on downstream processing

Progress and Challenges in PEGylated Proteins Downstream Processing: A Review of the Last 8 Years

Recent developments in chromatographic purification of biopharmaceuticals.

Albertsson PA (1970) Partition of cell particles and macromolecules in polymer two-phase system. Adv Protein Chem 24:309–341. https://doi.org/10.1016/S0065-3233(08)60244-2

Article   CAS   Google Scholar  

Alford JR, Kendrick BS, Carpenter JF, Randolph TW (2008) High concentration formulations of recombinant human interleukin-1 receptor antagonist: II. aggregation kinetics. J Pharm Sci 97(8):3005–3021. https://doi.org/10.1002/jps.21205

Arakawa T, Tsumoto K, Nagase K, Ejima D (2007) The effects of arginine on protein binding and elution in hydrophobic interaction and ion-exchange chromatography. Protein Expr Purif 54(1):110–116. https://doi.org/10.1016/j.pep.2007.02.010

Aumann L, Morbidelli M (2007) A continuous multicolumn countercurrent solvent gradient purification (MCSGP) process. Biotechnol Bioeng 98(5):1043–1055. https://doi.org/10.1002/bit.21527

Azevedo AM, Aires-Barros MR (2011) New platforms for downstream processing of biopharmaceuticals. 1st Portuguese Meeting in Bioengineering, February 2011 Portuguese chapter of IEEE EMBS Instituto Superior Técnico, Technical University of Lisbon

Google Scholar  

Azevedo AM, Rosa PA, Ferreira IF, Aires-Barros MR (2007) Optimisation of aqueous two-phase extraction of human antibodies. J Biotechnol 132(2):209–217. https://doi.org/10.1016/j.jniotec.2007.04.002

Bailey JE, Ollis DF (1986) Biochemical engineering fundamentals, 2nd edn. Tata McGraw-Hill Education, Noida

Beck A (2011) Biosimilar, biobetter and next generation therapeutic antibodies. MAbs 3(2):107–110. https://doi.org/10.4161/mabs.3.2.14785

Article   Google Scholar  

Bennett AD, Rhind SK, Lowe PA, Hentschel CCG (1984) Eur Pat Appl 0131363

Bhambure R, Kumar K, Rathore AS (2011) High-throughput process development for biopharmaceutical drug substances. Trends Biotechnol 29(3):127–135. https://doi.org/10.1016/j.tibtech.2010.12.001

Biopharma International Editors (2012) Considerations for successful upstream process development. BioPharm Int 25(7)

BIOPHARMA: Biopharmaceutical Products in the U.S. and European Markets (2002) This is a list of new full FDA approvals for biopharmaceutical products. Available from: http://www.biopharma.com/approvals.html

Bloomingburg GF, Bauer JS, Carta G, Byers CH (1991) Continuous separation of proteins by annular chromatography. Ind Eng Chem Res 30(5):1061–1010. https://doi.org/10.1021/ie00053a031

Bonham-Carter J, Shevitz J (2011) A brief history of perfusion bio manufacturing. BioProcess Int. 9(9):28–30

Brouns TM, Elliott ML, Van Wie BJ (1990) U.S. Patent No. 4,939,087. U.S. Patent and Trademark Office, Washington, DC

Buchacher A, Iberer G (2006) Purification of intravenous immunoglobulin G from human plasma – aspects of yield and virus safety. Biotechnol J 1(2):148–163. https://doi.org/10.1002/biot.200500037

Casey C, Gallos T, Alekseev Y, Ayturk E, Pearl S (2011) Protein concentration with single-pass tangential flow filtration (SPTFF). J Membr Sci 384(1-2):82–88. https://doi.org/10.1016/j.memsci.2011.09.004

Chen J, Tetrault J, Ley A (2008) Comparison of standard and new generation hydrophobic interaction chromatography resins in the monoclonal antibody purification process. J Chromatogr A 1177(2):272–281. https://doi.org/10.1016/j.chroma.2007.07.083

Cheryan M (1986) Ultrafiltration handbook. Technomic, Lancaster

Chisti Y, Moo-Young M (1986) Disruption of microbial cells for intracellular products. Enzym Microb Technol 8:194–204. https://doi.org/10.1016/0141-0229(86)90087-6

Chon JH, Zarbis-Papastoitsis G (2011) Advances in the production and downstream processing of antibodies. New Biotechnol 28(5):458–463. https://doi.org/10.1016/j.nbt.2011.03.015

Courtney M, Buchwalder A, Tessier LH, Jaye M, Benavente A, Ballard A, Kohli V, Lathe R, Tolstoshev P, Lecocq JP (1984) High-level production of biologically active human alpha 1-antitrypsin in Escherichia coli. 1984. Proc Natl Acad Sci U S A 81(3):669–663. https://doi.org/10.1073/pnas.81.3.669

Crapisi A, Lante A, Pasini G, Spettoli P (1993) Enhanced microbial cell lysis by the use of lysozyme immobilised on different carriers. Process Biochem 28(1):17–21. https://doi.org/10.1016/0032-9592(94)80031-6

Crommelin DJ, Storm G, Verrijk R, de Leede L, Jiskoot W, Hennink WE (2003) Shifting paradigms: biopharmaceuticals versus low molecular weight drugs. Int J Pharm 266(1-2):3–16. https://doi.org/10.1016/S0378-5173(03)00376-4

Dean CR, Ward OP (1992) The use of EDTA or polymyxin with lysozyme for the recovery of intracellular products from Escherichia coli . Biotechnol Tech 6(2):133–138. https://doi.org/10.1007/BF02438819

del Val IJ, Kontoravdi C, Nagy JM (2010) Towards the implementation of quality by design to the production of therapeutic monoclonal antibodies with desired glycosylation patterns. Biotechnol Prog 26(6):1505–1527. https://doi.org/10.1002/btpr.470

Dunnill P, Lilly MD (1974) Purification of enzymes using magnetic bioaffinity materials. Biotechnol Bioeng 16:987–990. https://doi.org/10.1002/bit.260160710

Durocher Y, Butler M (2009) Expression systems for therapeutic glycoprotein production. Curr Opin Biotechnol 20(6):700–707. https://doi.org/10.1016/j.copbio.2009.10.008

Eiberle MK, Jungbauer A (2010) Technical refolding of proteins: do we have freedom to operate? Biotechnol J 5(6):547–559. https://doi.org/10.1002/biot.201000001

Erikson RA (1984) Disk stack centrifuges in biotechnology. In: American Institute of Chemical Engineers, National Meeting, American Institute of Chemical Engineers, p 9

Ferreira AM, Faustino VFM, Mondal D, Coutinho JAP, Freire MG (2016) Improving the extraction and purification of immunoglobulin G by the use of ionic liquids as adjuvants in aqueous biphasic systems. J Biotechnol 236:166–175. https://doi.org/10.1016/j.jbiotec.2016.08.015

Fish B, Williams R (2007) Avoiding pitfalls in scaling up biopharmaceutical production. Pharm Technol Eur 19(10):29. doi: not available

Frenzel A, Bergemann C, Kohl G, Reinard T (2003) Novel purification system for 6xHis-tagged proteins by magnetic affinity separation. J Chromatogr A 793:325–329. https://doi.org/10.1016/S1570-0232(03)00332-5

Frost, Sullivan (2004) Strategic analysis of downstream processing. In: Biopharmaceutical production

Gagnon P (2006) Polishing methods for monoclonal IgG purification. In: Shukla AA, Etzel MR, Gadam S (eds) Process scale bioseparations for the biopharmaceutical industry. Taylor & Francis, New York, pp 491–505

Chapter   Google Scholar  

Gagnon P (2012) Technology trends in antibody purification. J Chromatogr A 1221:57–70. https://doi.org/10.1016/j.chroma.2011.10.034

Gary W (2000) Biopharmaceutical benchmarks. Nat Biotechnol 18:832–833. https://doi.org/10.1038/nbt.3040

Gary W (2003) Biopharmaceutical and pharmaceutical biotechnology. In: Biopharmaceuticals: biochemistry and biotechnology, 2nd edn. Wiley, Chichester

Ghose S, Hubbard B, Cramer SM (2006) Evaluation and comparison of alternatives to Protein A chromatography: mimetic and hydrophobic charge induction chromatographic stationary phases. J Chromatogr A 1122(1-2):144–152. https://doi.org/10.1016/j.chroma.2006.04.083

Ghose S, Jin M, Liu J, Hickey J (2009) Integrated polishing steps for monoclonal antibody purification. In: Gottschalk U (ed) Process scale purification of antibodies. Wiley, New York, pp 145–141

Giovannini R, Freitag R (2001) Isolation of a recombinant antibody from cell culture supernatant: continuous annular versus batch and expanded-bed chromatography. Biotechnol Bioeng 73(6):522–529. https://doi.org/10.1002/bit.1087

Goeddel DV, Heyneker HL, Hozumi T, Arentzon R, Itakura K, Yansura DG, Ross MJ, Miozzari G, Crea R, Seeburg P (1979a) Direct expression in Escherichia coli of a DNA sequence coding for human growth hormone. Nature 281(5732):544–548. https://doi.org/10.1038/281544a0

Goeddel DV, Kleid DG, Bolivar F, Heyneker HL, Yansura DG, Crea R, Hirose T, Kraszewski A, Itakura K, Riggs AD (1979b) Expression in Escherichia coli of chemically synthesized genes for human insulin. Proc Natl Acad Sci USA 76(1):106–110. doi: not available

Gözke G, Kirschhöfer F, Heissler S, Trutnau M, Brenner-Weiss G, Ondruschka J, Obst U, Posten C (2012) Filtration kinetics of chitosan separation by electrofiltration. Biotechnol J 7(2):262–274. https://doi.org/10.1002/biot.201000466

Gronemeyer P, Ditz R, Strube J (2014) Trends in upstream and downstream process development for antibody manufacturing. Bioengineering 1(4):188–212. https://doi.org/10.3390/bioengineering1040188

Gueorguieva L, Palani S, Rinas U, Jayaraman G, Seidel-Morgenstern A (2011) Recombinant protein purification using gradient assisted simulated moving bed hydrophobic interaction chromatography. Part II: process design and experimental validation. J Chromatogr A 1218(37):6402–6411. https://doi.org/10.1016/j.chroma.2011.07.008

Hanke AT, Ottens M (2014) Purifying biopharmaceuticals: knowledge-based chromatographic process development. Trends Biotechnol 32(4):210–220. https://doi.org/10.1016/j.tibtech.2014.02.001

Harrison STL (2011) Cell disruption. In: Comprehensive biotechnology, 2nd edn. Elsevier, Oxford, pp 619–639

Himeji D, Horiuchi T, Tsukamoto H, Hayashi K, Watanabe T, Harada M (2002) Characterization of caspase-8L: a novel isoform of caspase- 8 that behaves as an inhibitor of the caspase cascade. Blood 99:4070–4078. https://doi.org/10.1182/blood.V99.11.4070

Hodge G (2005) Media development for mammalian cell culture. Biopharm Int 18:54

Hofmann I, Schnolzer M, Kaufmann I, Franke WW (2002) Symplekin, a constitutive protein of karyo- and cytoplasmic particles involved in mRNA biogenesis in Xenopus laevis oocytes. Mol Biol Cell 13(5):1665–1676. https://doi.org/10.1091/mbc.01-12-0567

Hossler P, Khattak SF, Li ZJ (2009) Optimal and consistent protein glycosylation in mammalian cell culture. Glycobiology 19(9):936–949. https://doi.org/10.1093/glycob/cwp079

Huddleston J, Veide A, Köhler K, Flanagan J, Enfors SO, Lyddiatt A (1991) The molecular basis of partitioning in aqueous two-phase systems. Trends Biotechnol 9(11):381–388. https://doi.org/10.1016/0167-7799(91)90130-A

Huettmann H, Berkemeyer M, Buchinger W, Jungbauer A (2014) Preparative crystallization of a single chain antibody using an aqueous two-phase system. Biotechnol Bioeng 111(11):2192–2199. https://doi.org/10.1002/bit.25287

Iberer G, Schwinn H, Josic D, Jungbauer A, Buchacher A (2002) Continuous purification of a clotting factor IX concentrate and continuous regeneration by preparative annular chromatography. J Chromatogr 972(1):115–129. https://doi.org/10.1016/S0021-9673(02)01074-9

Itakura K, Hiroso T, Crea R, Riggs AD, Heyneker HL, Bolivar F, Boyer HW (1977) Expression in Escherichia coli of a chemically synthesized gene for the hormone somatostatin. Science 198(4321):1056–1063. https://doi.org/10.1126/science.412251

Ivory CF, Gilmartin M, Gobie WA, McDonald CA, Zollars RL (1995) A hybrid centrifuge rotor for continuous bioprocessing. Biotechnol Prog 11(1):21–32. https://doi.org/10.1021/bp00031a003

Jayapal KP, Wlaschin KF, Hu WS, Yap MGS (2007) Recombinant protein therapeutics from CHO cells – 20 years and counting. Chem Eng Prog 103(10):40–47. doi: not available

CAS   Google Scholar  

Jozala AF, Geraldes DC, Tundisi LL, Feitosa VA, Breyer CA, Cardoso SL, Mazzola PG, Oliveira-Nascimento L, Rangel-Yagui CO, Magalhães PO, Oliveira MA, Pessoa A Jr (2016) Biopharmaceuticals from microorganisms: from production to purification. Braz J Microbiol 47(1):51–63. https://doi.org/10.1016/j.bjm.2016.10.007

Jungbauer A (2013) Continuous downstream processing of biopharmaceuticals. Trends Biotechnol 31(8):479–492. https://doi.org/10.1016/j.tibtech.2013.05.011

Jungbauer A, Kaar W (2007) Current status of technical protein refolding. J Biotechnol 128(3):587–596. https://doi.org/10.1016/j.jbiotec.2006.12.004

Kato Y, Nakamura K, Kitamura T, Hasegawa M, Sasaki H (2004) Hydrophobic interaction chromatography at low salt concentration for the capture of monoclonal antibodies. J Chromatogr A 1036(1):45–50. https://doi.org/10.1016/j.chroma.2004.02.009

Kennedy RM (2005) Expanded-bed adsorption chromatography. Curr Protoc Protein Sci Jun; Chapter 8: Unit 8:8. https://doi.org/10.1002/0471140864.ps0808s40

Kramberger P, Urbas L, Štrancar A (2015) Downstream processing and chromatography based analytical methods for production of vaccines, gene therapy vectors, and bacteriophages. Hum Vaccin Immunother 11(4):1010–1021. https://doi.org/10.1080/21645515.2015.1009817

Kwon JS-II, Nayhouse M, Christofides PD, Orkoulas G (2014) Modeling and control of crystal shape in continuous protein crystallization. Chem Eng Sci 107:47–57. https://doi.org/10.1016/j.ces.2013.12.005

Lain B, Cacciuttolo MA, Zarbis-Papastoitsis G (2009) Development of a high-capacity Mab capture step based on cation-exchange chromatography. BioProcess Int 7(5):26–34

Lander R, Daniels C, Meacle F (2005) Efficient, scalable clarification of diverse bioprocess streams. Bioprocess Int 11:32–40

Langer ES (2011) Trends in perfusion bioreactors: will perfusion be the next revolution in bioprocessing? BioProcess Int. 9(10):18–22

Larsson PO (1994) Magnetically enhanced phase separation. Methods Enzymol 228:112–117. doi: not available

Lebreton B, Brown A, van Reis R (2008) Application of high-performance tangential flow filtration (HPTFF) to the purification of a human pharmaceutical antibody fragment expressed in Escherichia coli. Biotechnol Bioeng 100(5):964–974. https://doi.org/10.1002/bit.21842

Li F, Vijayasankaran N, Shen AY, Kiss R, Amanullah A (2010) Cell culture processes for monoclonal antibody production. MAbs 2(5):466–479. https://doi.org/10.4161/mabs.2.5.12720

Lin N, Mascarenhas J, Sealover NR, George HJ, Brooks J, Kayser KJ, Gau B, Yasa I, Azadi P, Archer-Hartmann S (2015) Chinese hamster ovary (CHO) host cell engineering to increase sialylation of recombinant therapeutic proteins by modulating sialyltransferase expression. Biotechnol Prog 31(2):334–346. https://doi.org/10.1002/btpr.2038

Liu HF, Ma J, Winter C, Bayer R (2010) Recovery and purification process development for monoclonal antibody production. MAbs 2(5):480–499. https://doi.org/10.4161/mabs.2.5.12645

Liu Z, Wickramasinghe SR, Qian X (2017) Membrane chromatography for protein purifications from ligand design to functionalization. Sep Sci Technol 52:299–319. https://doi.org/10.1080/01496395.2016.1223133

Lowe CR (2001) Combinatorial approaches to affinity chromatography. Curr Opin Chem Biol 5(3):248–256. https://doi.org/10.1016/S1367-5931(00)00199-X

Machold C, Schlegl R, Buchinger W, Jungbauer A (2005) Continuous matrix assisted refolding of a-lactalbumin by ion exchange chromatography with recycling of aggregates combined with ultradiafiltration. J Chromatogr A 1080(1):29–42. https://doi.org/10.1016/j.chroma.2005.03.018

Mahajan E, George A, Wolk B (2012) Improving affinity chromatography resin efficiency using semi-continuous chromatography. J Chromatogr A 1227:154–162. https://doi.org/10.1016/j.chroma.2011.12.106

Martinez Cristancho CA, David F, Franco-Lara E, Seidel-Morgenstern A (2013) Discontinuous and continuous purification of single-chain antibody fragments using immobilized metal ion affinity chromatography. J Biotechnol 163(2):233–242. https://doi.org/10.1016/j.jbiotec.2012.08.022

Middelberg A (1995) Process-scale disruption of microorganisms. Biotechnol Adv 13(3):491–551. https://doi.org/10.1016/0734-9750(95)02007-P

Mitragotri S, Burke PA, Langer R (2014) Overcoming the challenges in administering biopharmaceuticals: formulation and delivery strategies. Nat Rev Drug Discov 13(9):655–652. https://doi.org/10.1038/nrd4363

Mun S, Yi X, Kim JH, Wang NHL (2003) Optimal design of a size-exclusion tandem simulated moving bed for insulin purification. Ind Eng Chem Res 42(9):1977–1993. https://doi.org/10.1021/ie020680+

Ngantung FA, Miller PG, Brushett FR, Tang GL, Wang DI (2006) RNA interference of sialidase improves glycoprotein sialic acid content consistency. Biotechnol Bioeng 95(1):106–119. https://doi.org/10.1002/bit.20997

Nishiya Y, Hibi T, Oda JL (2002) A purification method of the diagnostic enzyme Bacillus uricase using magnetic beads and nonspecific protease. Protein Expr Purif 25:426–429. https://doi.org/10.1016/S1046-5928(02)00022-0

Odabasi M, Denizli A (2004) Cibacron Blue F3GA-attached magnetic poly(2-hydroxyethyl methacrylate) beads for human serum albumin adsorption. Polym Int 53:332–338. https://doi.org/10.1002/pi.1305

Ozdural AR et al (2007) A novel technology for virus vaccine purification: modeling and operation of continuous annular chromatography unit. In: AIChE annual meeting: 2007 Spring meeting and 3rd global congress on process safety. American Institute of Chemical Engineers, p 18

Parmar HC (2006) Biopharmaceuticals market overview In: Pharmatech.com . Pharmaceutical Technology Europe 18(3). https://economictimes.indiatimes.com/industry/healthcare/biotech/india-launches-flagship-program-to-boost-biopharma-production/articleshow/59390777.cms

Peeva L, da Silva Burgal J, Valtcheva I, Livingston AG (2014) Continuous purification of active pharmaceutical ingredients using multistage organic solvent nanofiltration membrane cascade. Chem Eng Sci 116:183–194. https://doi.org/10.1016/j.ces.2014.04.022

Pennica D, Hayflick JS, Bringman TS, Palladina MA, Goeddel DV (1985) Cloning and expression in Escherichia coli of the cDNA for murine tumor necrosis factor. Proc Natl Acad Sci U S A 82:6060–6064. doi:not available

Pirrung SM, van der Wielen LAM, van Beckhoven RFWC, van de Sandt EJAX, Eppink MHM, Ottens M (2017) Optimization of biopharmaceutical downstream processes supported by mechanistic models and artificial neural networks. Biotechnol Prog 33(3):696–707. https://doi.org/10.1002/btpr.2435

Rajendran A, Paredes G, Mazzotti M (2009) Simulated moving bed chromatography for the separation of enantiomers. J Chromatog A 1216(4):709–738. https://doi.org/10.1016/j.chroma.2008.10.075

Rathore AS, Agarwal H, Sharma AK, Pathak M, Muthukumar S (2015) Continuous processing for production of biopharmaceuticals. Prep Biochem Biotechnol 45(8):836–849. https://doi.org/10.1080/10826068.2014.985834

Rathore AS, Kumar D, Kateja N (2018) Recent developments in chromatographic purification of biopharmaceuticals. Biotechnol Lett 40(6):1–11. https://doi.org/10.1007/s10529-018-2552-1

Roque AC, Lowe CR, Taipa MA (2004) Antibodies and genetically engineered related molecules: production and purification. Biotechnol Prog 20(3):639–654. https://doi.org/10.1021/bp030070k

Ruanjaikaen K, Zydney AL (2011) Purification of singly-pegylated α-lactalbumin using charged ultrafiltration membranes. Biotechnol Bioeng 108:822–829. https://doi.org/10.1002/bit.22991

Saboya LV, Maillard MB, Lortal S (2003) Efficient mechanical disruption of Lactobacillus helveticus, Lactococcus lactis and Propionibacterium freudenreichii by a new high-pressure homogenizer and recovery of intracellular aminotransferase activity. J Ind Microbiol Biotechnol 30(1):1–5. https://doi.org/10.1007/s10295-002-0011-3

Safarik I, Safarikova M (1993) Batch isolation of hen egg white lysozyme with magnetic chitin. J Biochem Biophys Methods 27:327–330. https://doi.org/10.1016/0165-022X(93)90013-E

Saraswat M, Musante L, Ravidá A, Shortt B, Byrne B, Holthofer H (2013) Preparative purification of recombinant proteins: current status and future trends. Biomed Res Int 2013:312709. https://doi.org/10.1155/2013/312709

Schafer F, Romer U, Emmerlich M, Blumer J, Lubenow H, Steinert K (2002) Automated high-throughput purification of 6xHis-tagged proteins. J Biomol Tech 13:131–142. doi:not available

Schoemaker JM, Brasnett AH, Marston FA (1985) Examination of calf prochymosin accumulation in Escherichia coli: disulphide linkages are a structural component of prochymosin-containing inclusion bodies. EMBO J 4(3):775–780. doi:not available

Schoner RG, Ellis LF, Schoner BE (1985) Isolation and purification of protein granules from Escherichia coli cells overproducing bovine growth hormone. Bio Technol 3:151–154. https://doi.org/10.1038/nbt0285-151

Schuster M, Wasserbauer E, Ortner C, Graumann K, Jungbauer A, Hammerschmid F, Werner G (2000) Short cut of protein purification by integration of cell-disrupture and affinity extraction. Bioseparation 9(2):59–67. https://doi.org/10.1023/A:100813591

Scott JH, Schekman R (1980) Lyticase: endoglucanase and protease activities that act together in yeast cell lysis. J Bacteriol 142(2):414–423. doi:not available

Shao S, Gross V, Yan W, Guo T, Lazarev A, Abersold R (2015) Hands-free sample homogenisation and protein extraction from small tissue biopsy samples using pressure cycling technology and PCT micropestle (poster). In: US HUPO 2015 Conference, Tempe, AZ. http://www.pressurebiosciences.com/documents?task=document.viewdoc&id=64

Shinkai M, Kamihira M, Honda H, Kobayashi T (1992) Rapid purification of monoclonal antibody with functional magnetite particles. Kag Kog Ronbunshu 18:256–259. https://doi.org/10.1252/kakoronbunshu.18.256

Shukla AA, Hinckley P (2008) Host cell protein clearance during protein A chromatography: Development of an improved column wash step. Biotechnol Prog 24(5):1115–1121. https://doi.org/10.1002/btpr.50

Shukla AA, Thömmes J (2010) Recent advances in large-scale production of monoclonal antibodies and related proteins. Trends Biotechnol 28(5):253–261. https://doi.org/10.1016/j.tibtech.2010.02.001

Shukla AA, Hubbard B, Tressel T, Guhan S, Low D (2007) Downstream processing of monoclonal antibodies – application of platform approaches. J Chromatogr B Anal Technol Biomed Life Sci 848(1):28–39. https://doi.org/10.1016/jchromb.2006.09.026

Siew WE, Livingston AG, Ates C, Merschaert A (2013) Continuous solute fractionation with membrane cascades – a high productivity alternative to diafiltration. Sep Purif Technol 102:1–14. https://doi.org/10.1016/j.seppur.2012.09.017

Strohl WR (2015) Fusion proteins for half-life extension of biologics as a strategy to make biobetters. BioDrugs 29(4):215–239. https://doi.org/10.1007/s40259-015-0133-6

Szoka PR, Schreiber AB, Chan H, Murthy J (1986) A general method for retrieving the components of a genetically engineered fusion protein. DNA 5(1):11–20. https://doi.org/10.1089/dna.1986.5.11

Tarrant RD, Velez-Suberbie ML, Tait AS, Smales CM, Bracewell DG (2012) Host cell protein adsorption characteristics during protein A chromatography. Biotechnol Prog 28(4):1037–1044. https://doi.org/10.1002/btpr.1581

Top drugs by sales revenue in 2015: Who sold the biggest blockbuster drugs? (2016) The PharmaCompass Newsletter. http://www.pharmacompass.com/radiocompass-blog/top-drugs-by-sales-revenue-in-2015-whosold-thebiggest-blockbuster-drugs

Tsumoto K, Ejima D, Nagase K, Arakawa T (2007) Arginine improves protein elution in hydrophobic interaction chromatography. The cases of human interleukin-6 and activin-A. J Chromatogr A 1154(1–2):81–86. https://doi.org/10.1016/j.chroma.2007.02.061

van Reis R, Zydney A (2007) Bioprocess membrane technology. J Membr Sci 297:16–50. https://doi.org/10.1016/j.memsci.2007.02.045

van Reis R, Gadam S, Frautschy LN, Orlando S, Goodrich EM, Saksena S, Kuriyel R, Simpson CM, Pearl S, Zydney AL (1997) High performance tangential flow filtration. Biotechnol Bioeng 56(1):71–82. https://doi.org/10.1002/(SICI)1097-0290(19971005)56:1<71::AID-BIT8>3.0.CO;2-S

Varma SK (2009) An overview of biopharmaceutical industry in India. Pharmabiz Chronicle Specials. http://saffron.pharmabiz.com/article/detnews.asp?articleid=53092&sectionid=50

Wang X, Rivière I (2016) Clinical manufacturing of CAR T cells: foundation of a promising therapy. Mol Ther Oncolyt 3:16015. https://doi.org/10.1038/mto.2016.15

Warikoo V, Godawat R, Brower K, Jain S, Cummings D, Simons E, Johnson T, Walther J, Yu M, Wright B, McLarty J, Karey KP, Hwang C, Zhou W, Riske F, Konstantinov K (2012) Integrated continuous production of recombinant therapeutic proteins. Biotechnol Bioeng 109(12):3018–3029. https://doi.org/10.1002/bit.24584

Watt JG (1970) Automatically controlled continuous recovery of plasma protein fractions for clinical use. A preliminary report. Vox Sang 18(1):42–61. https://doi.org/10.1111/j.1423-0410.1970.tb01428x

Wellhoefer M, Sprinzl W, Hahn R, Jungbauer A (2013) Continuous processing of recombinant proteins: integration of inclusion body solubilization and refolding using simulated moving bed size exclusion chromatography with buffer recycling. J Chromatogr A 1319:107–117. https://doi.org/10.1016/j.chroma.2013.10.039

Wikstrom P, Flygare S, Grondalen A, Larsson PO (1987) Magnetic aqueous two-phase separation: a new technique to increase rate of phase-separation, using dextran-ferrofluid or larger iron oxide particles. Anal Biochem 1(2):331–339. https://doi.org/10.1016/0003-2697(87)90173-4s

Wilson AW, Neumann PJ (2012) The cost-effectiveness of biopharmaceuticals: a look at the evidence. MAbs 4(2):281–288. https://doi.org/10.4161/mabs.4.2.18812

Winkler ME, Blaber M, Bennett GL, Holmes W, Vehar GA (1985) Purification and characterization of recombinant urokinase from Escherichia coli . Bio Technol 3:990–1000. https://doi.org/10.1038/nbt1185-990

Yamane-Ohnuki N, Kinoshita S, Inoue-Urakubo M, Kusunoki M, Iida S, Nakano R, Wakitani M, Niwa R, Sakurada M, Uchida K, Shitara K, Satoh M (2004) Establishment of FUT8 knockout Chinese Hamster Ovary cells: An ideal host cell line for producing completely defucosylated antibodies with enhanced antibody dependent cellular cytotoxicity. Biotechnol Bioeng 87(5):614–622. https://doi.org/10.1002/bit.20151

Zang Y, Kammerer B, Eisenkolb M, Lohr K, Kiefer H (2011) Towards protein crystallization as a process step in downstream processing of therapeutic antibodies: screening and optimization at microbatch scale. PLoS One 6(9):1–8. https://doi.org/10.1371/journal.pone.0025282

Zhou JX, Tressel T (2006) Basic concepts in Q membrane chromatography for large-scale antibody production. Biotechnol Prog 22(2):341–349. https://doi.org/10.1021/bp050425v

Zhu J (2012) Mammalian cell protein expression for biopharmaceutical production. Biotechnol Adv 30(5):1158–1170. https://doi.org/10.1016/j.biotechadv.2011.08.022

Zydney AL (2015) Continuous downstream processing for high value biological products: a review. Biotechnol Bioeng 113(3):465–475. https://doi.org/10.1002/bit.25695

Zydney AL, van Reis R (2001) High performance tangential flow filtration. In: Wang WK (ed) Membrane separations in biotechnology, 2nd edn. Marcel Dekker, New York, pp 277–298

Download references

Acknowledgements

The author is highly thankful to the editor and reviewers for suggesting valuable suggestions.

Author information

Authors and affiliations.

Department of Biochemistry, Kurukshetra University, Kurukshetra, Haryana, India

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Anu Mehta .

Editor information

Editors and affiliations.

Indian Institute of Integrative Medicine, CSIR, Jammu, India

Divya Arora

Guru Angad Dev Veterinary and Animal Science University, Ludhiana, Punjab, India

Chetan Sharma

Sundeep Jaglan

Aix Marseille University, CNRS, IRD, INRA, Coll France, CEREGE, Aix en Provence, France

Eric Lichtfouse

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this chapter

Mehta, A. (2019). Downstream Processing for Biopharmaceuticals Recovery. In: Arora, D., Sharma, C., Jaglan, S., Lichtfouse, E. (eds) Pharmaceuticals from Microbes. Environmental Chemistry for a Sustainable World, vol 26. Springer, Cham. https://doi.org/10.1007/978-3-030-01881-8_6

Download citation

DOI : https://doi.org/10.1007/978-3-030-01881-8_6

Published : 10 January 2019

Publisher Name : Springer, Cham

Print ISBN : 978-3-030-01880-1

Online ISBN : 978-3-030-01881-8

eBook Packages : Earth and Environmental Science Earth and Environmental Science (R0)

Share this chapter

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Publish with us

Policies and ethics

  • Find a journal
  • Track your research
  • Architecture and Design
  • Asian and Pacific Studies
  • Business and Economics
  • Classical and Ancient Near Eastern Studies
  • Computer Sciences
  • Cultural Studies
  • Engineering
  • General Interest
  • Geosciences
  • Industrial Chemistry
  • Islamic and Middle Eastern Studies
  • Jewish Studies
  • Library and Information Science, Book Studies
  • Life Sciences
  • Linguistics and Semiotics
  • Literary Studies
  • Materials Sciences
  • Mathematics
  • Social Sciences
  • Sports and Recreation
  • Theology and Religion
  • Publish your article
  • The role of authors
  • Promoting your article
  • Abstracting & indexing
  • Publishing Ethics
  • Why publish with De Gruyter
  • How to publish with De Gruyter
  • Our book series
  • Our subject areas
  • Your digital product at De Gruyter
  • Contribute to our reference works
  • Product information
  • Tools & resources
  • Product Information
  • Promotional Materials
  • Orders and Inquiries
  • FAQ for Library Suppliers and Book Sellers
  • Repository Policy
  • Free access policy
  • Open Access agreements
  • Database portals
  • For Authors
  • Customer service
  • People + Culture
  • Journal Management
  • How to join us
  • Working at De Gruyter
  • Mission & Vision
  • De Gruyter Foundation
  • De Gruyter Ebound
  • Our Responsibility
  • Partner publishers

research papers on downstream processing

Your purchase has been completed. Your documents are now available to view.

book: Downstream Processing in Biotechnology

Downstream Processing in Biotechnology

  • Edited by: Venko N. Beschkov and Dragomir Yankov
  • X / Twitter

Please login or register with De Gruyter to order this product.

  • Language: English
  • Publisher: De Gruyter
  • Copyright year: 2021
  • Audience: Biochemical Engineers, Biochemists, Organic Chemists, Microbiologists.
  • Front matter: 11
  • Main content: 166
  • Illustrations: 100
  • Coloured Illustrations: 100
  • Keywords: Bioprocess Engineering ; Biotechnology ; Product Reprocessing ; Separation Processes
  • Published: June 21, 2021
  • ISBN: 9783110574111
  • ISBN: 9783110573954

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • v.7(1); 2024 Jan
  • PMC10791043

Logo of abt

Challenges and solutions for the downstream purification of therapeutic proteins

GenScript ProBio Biotechnology Co., Ltd, Nanjing, Jiangsu 21100, P.R. China

Associated Data

The authors confirm that the data supporting the findings of this study are available within the article.

The innovation in recombinant protein technology has brought forth a host of challenges related to the purification of these therapeutic proteins. This article delves into the intricate landscape of developing purification processes for artificially designed therapeutic proteins. The key hurdles include controlling protein reduction, protein capture, ensuring stability, eliminating aggregates, removing host cell proteins and optimizing protein recovery. In this review, we outline the purification strategies in order to obtain products of high purity, highlighting the corresponding solutions to circumvent the unique challenges presented by recombinant therapeutic proteins, and exemplify the practical applications by case studies. Finally, a perspective towards future purification process development is provided.

Statement of Significance: The diversity of protein structures has led to an increased complexity of product- and process-related impurities. The article summarizes the primary challenges encountered during the downstream processing of proteins, and presents effective solutions and case studies to tackle each major challenge.

INTRODUCTION

Therapeutic proteins played a pivotal role in combating numerous diseases over the recent decades. Subsequently, the landscape of recombinant products has expanded exponentially, encompassing a diverse array of biologics, including blood products, hormones, cytokines and monoclonal antibodies (mAbs) [ 1 ]. With the advancement of genetic engineering technology, research in protein drugs achieved significant breakthroughs. The application of technologies such as fusion proteins, peptides and engineered antibodies diversified the construction of protein drugs, allowing for customization based on disease characteristics and patient needs [ 2 ]. Progress in protein expression systems has enabled the synthesis of virtually any desired protein or peptide [ 3 ]. Notably, this technological advancement has empowered the creation of unprecedented molecules that diverge from natural proteins, exemplified by fusion proteins and bispecific antibodies. These novel categories of therapeutic proteins exhibit unique functionalities that hold immense potential in enhancing drug efficacy.

In protein cell culture, there are impurities that can potentially trigger adverse reactions or allergic responses. Immune responses to these impurities can result in the loss of tolerance to their endogenous counterparts leading to serious adverse events [ 4 ]. The ICH Q6B guideline sets out the principles for consideration in setting product quality specifications (appearance, identity, purity and impurities, potency and quantity). Simultaneously, while navigating market price regulations and competition, pharmaceutical companies must address the vital challenges of cost-effective production and achieving high product recovery rate to ensure sustainable profitability.

Extracting high-purity target products from complex mixtures is an extremely challenging task. The diversity of protein structures has led to an increased complexity of product- and process-related impurities. Relying solely on lessons derived from mAbs has proven somewhat inadequate for the purification of complex recombinant proteins. For example, bispecific antibodies (BsAbs) are an emerging class of biotherapeutics with design diversity, each with a unique combination of antigen-binding domains that bind two different antigenic epitopes [ 5 ]. The multi-target design allows the simultaneous introduction of BsAb-specific byproducts, such as mispaired products, undesired fragments and higher levels of aggregates. The presence of these impurities that closely resemble the target antibody makes separation difficult, so it is usually necessary to develop additional purification strategies to obtain high-purity products. Antibody fusion proteins combine the advantages of antibody targeting and long half-life [ 6 ]. However, they suffer from chain mismatches, product cleavage and aggregates. Acidic instability can give rise to protein fragmentation or aggregation during affinity elution, thereby compromising protein quality. Acidic proteins with isoelectric points similar to host cell proteins (HCPs) make the separation of HCPs difficult. Recombinant proteins designed without tags lack specific affinity domains, thereby presenting significant obstacles in the removal of HCPs and the capture of the target product. In the face of these challenges, optimizing individual unit operations and the overall purification process becomes particularly important.

This review outlines the existing challenges encountered in the purification of intricate therapeutic proteins, including multispecific antibodies, fusion proteins and recombinant proteins. The main challenges in protein purification processes include protein reduction, protein capture, maintaining protein stability, removal of aggregates and HCPs and achieving high recovery [ 7–9 ]. The corresponding solutions that have been proposed to circumvent the unique challenges are presented. These approaches hold the potential to expedite the advancement of novel products in research and development, shorten the time required for commercialization and reduce the production cost significantly.

SOLUTIONS FOR CHALLENGING PROTEIN PURIFICATION

Protein purification is a complex process involving multiple steps and diverse principles. The first unit operation is the removal of cells and cell debris from the culture broth. This essential step is referred to as “clarification” and can be achieved through physical methods, such as filtration. The purification process typically involves three primary steps: capture (isolation, concentration, volume reduction), intermediate purification (removal of bulk impurities or main protein contaminants) and polishing (removal of trace impurities, closely related contaminants and protein aggregates). These purification stages commonly utilize various chromatography techniques due to their excellent resolving capabilities. Finally, the product formulation involves obtaining the drug product through buffer exchange, and additives are added to enhance the stability and shelf life of the protein product.

This section summarizes the primary challenges encountered during the downstream processing of proteins, and presents effective solutions and case studies to tackle each major challenge.

Protein reduction

Proteins contain multiple disulfide bonds, and the correct pairing of disulfide bonds is crucial for protein structure and activity. Ideally, disulfide bonds are properly paired before protein secretion to the extracellular environment, but in the complex oxidative-reductive environment of cell culture, disulfide bond breakage will occur easily [ 10 ]. In living organisms, the oxidation and reduction of disulfide bonds formed through the exchange of thiols and disulfides are catalyzed by thioredoxin (Trx) [ 11 ]. Release of sufficient intracellular enzymes, proteins and cofactors into the cell culture medium due to cell lysis can cause disulfide bond reduction [ 12 ]. The development of high-expression cell culture processes has resulted in an increasing occurrence of reduction of protein inter-chain disulfide bonds due to higher reductase release. Several studies have been conducted to understand disulfide bond reduction, yielding a range of recommendations to prevent its occurrence during the manufacturing process [ 10 , 13 ].

Inhibiting the activity of reductase is an effective approach to address the antibody reduction issue. Our own practice demonstrated different resolution strategies for protein reduction ( Fig. 1 ). Figure 1(A) illustrates a mAb molecule with severe reduction issues. During the clarification stage, it was observed that the antibody experienced significant molecular fragmentation after being stored at low temperatures for up to 24 hours, resulting in a decrease in the target molecule’s CE-SDS-NR purity from 97.2 to 57.7%. The reduction of antibody was effectively suppressed by adding 0.5 mM CuSO 4 to the harvested samples. Copper ions can directly inhibit the activity of Trx, thereby suppressing enzyme-mediated molecular reduction [ 14 ]. Another BsAb molecule exhibits the same reduction issue, which becomes more severe with increasing incubation time ( Fig. 1B ). After 7 hours of incubation at room temperature, the CE-SDS-NR purity of the target antibody decreased from the initial 85.4 to 56.7%. Different strategies were employed to inhibit the reduction reaction, and the results demonstrated that continuous ventilation, addition of copper sulfate and cysteamine effectively suppressed molecular breakage. Air sparging is the preferred method due to its convenience in operation and absence of additives. Continuous air sparging can increase the dissolved oxygen in the liquid, reduce the generation of NADPH and maintain the electron source of thioredoxin in a deficient state, thereby inhibiting antibody redox reactions [ 15 , 16 ].

An external file that holds a picture, illustration, etc.
Object name is tbad028f1.jpg

CE-SDS-NR results of samples under different conditions. (A) mAb molecule (pI = 7.2, Mw = 144 kDa). (B) BsAb molecule (pI = 7.8, Mw = 175 kDa).

In addition, inhibiting or slowing down antibody reduction can also be achieved by adjusting pH and temperature to reduce thioredoxin catalytic reactivity, as well as implementing rapid purification to reduce catalytic reaction time [ 10 , 17 ]. Based on the mechanism of this system, Table 1 summarizes common strategies to address antibody reduction issue in downstream purification.

Solutions for antibody reduction in downstream purification

MechanismSolution strategies
Inhibit Trx and hexokinase activity1. Induce EDTA to inhibit hexokinase activity during cell harvest
2. Induce Cu , Zn to inhibit Trx activity during cell harvest
Reduce reductant and enzyme amount1. Induce oxidants such as O , H O and cysteine as competitors
2. Control depth filtration pressure to prevent enzyme release
Slow down enzyme reaction rateReduce pH and temperature before protein capture
Shorten enzyme reaction timeReduce sample storage time before protein capture

Protein capture

Protein capture is a recurring topic, and researchers have designed different tags on proteins to achieve effective separation [ 18 ]. While the capture of mAbs has been well-established, the capture methods for complex fusion proteins, tag-free recombinant proteins, and vaccines can vary significantly, depending on the unique properties of each protein.

Antibody fusion proteins

Affinity chromatography (AC) is widely used in the capture of mAbs, which relies on the specific binding between the target antibody with AC resin. As the complexity of molecular design increases, various antibody fragment-based fusion proteins have been developed, such as Fc-fusion proteins, ScFv-fusion proteins and VHH-fusion proteins.

As shown in Figure 2 , antibodies are composed of different structural domains, and different domains can be designed and produced as recombinant fusion proteins with therapeutic activity. In antibody process development, Protein A, Protein G and Protein L AC have been developed for the capture of antibodies at different sites [ 7 ]. Fc-fusion proteins are the most common design structure and can typically be captured using Protein A and Protein G chromatography. Protein A capture is made possible through the interaction between Protein A and the Fc region of the target molecule as well as the heavy chain variable domain (VH) region of the HC for targets belonging to the VH3 gene family [ 19 ]. Protein G recognizes the Fab and Fc antibody regions in a manner similar to Protein A but with different binding specificities. The protein designed based on ScFv is another form of composition, and these forms are achieved through different combinations of chains and linkers. Protein L and Protein A can specifically recognize the VL and VH regions of ScFv for selective capture.

An external file that holds a picture, illustration, etc.
Object name is tbad028f2.jpg

Schematic representation of IgG and fusion proteins classified based on their affinity binding domain.

Selection of the optimal affinity resin is the primary task of protein capture. Theoretically, Protein A, Protein G and Protein L can all be used for capture of Fab fragment, they showed vastly different binding capability in applications. Different ligand structures, ligand densities and resin matrix led to significant difference in protein capture capability among same ligand types. While the principle can offer guidance for the selection of capture resins during process development, it is important to note that various resins may demonstrate distinct specificities for different molecules. Our own practice showed that Protein L resin demonstrated a superior capture ability for a Fab fragment, and Protein L resin from different sources displayed distinct capture capabilities. ( Supplementary Fig. S1 ).

Besides the selection of resin, the choice of buffer system is also crucial for protein capture, especially for unstable fusion proteins. AC typically employs acidic elution to separate the target protein from the resin, under which condition the protein may become unstable. In addition, for acidic fusion proteins, the adjustment of pH across its isoelectric point (pI) may lead to protein precipitation or aggregation. Thus, for fusion proteins that are sensitive to acidic conditions, it is recommended to select a resin with lower affinity and reduce the buffering capacity of the elution buffer to increase the elution pH. Our own practice showed that the effect of affinity resin, elution buffer concentration and pH on the capture of an acidic fusion protein ( Fig. 3 ). This protein precipitates when exposed to acidic conditions. Affinity resin#2 has been specifically designed to enable the elution of protein at high pH and avoid protein precipitation under acidic conditions. Additionally, the affinity resin#2 exhibits lower affinity to protein, allowing for high recovery during high pH elution. Reducing the buffering capacity of the elution buffer can further increase the pH of the protein eluate. After optimization, the pH of protein eluate remains above its pI throughout the entire capture process, thus avoiding post elution pH adjustment and product loss through aggregation.

An external file that holds a picture, illustration, etc.
Object name is tbad028f3.jpg

Effect of affinity resin and elution conditions on the eluate pH and recovery of acidic fusion protein (pI = 5.4, Mw = 111 kDa).

Tag free recombinant protein

Due to the lack of affinity domains, tag-free recombinant proteins can only rely on non-specific capture, typically based on the protein’s physicochemical properties such as charge, hydrophobicity and molecular weight. Figure 4 recommends the purification strategy for tag-free fusion proteins. Ion exchange chromatography (IEX) typically provides high resolution under mild conditions with high binding capacity, and is, therefore, recommended for capturing recombinant proteins. Excessive adsorption of pigments on anion exchange chromatography (AEX) resin causes a decline in product purity and makes it challenging to regenerate the resin, consequently reducing its operational lifespan. Hence, cation exchange chromatography (CEX) is used as the preferred ion exchange capture method. Additionally, the sample pH should differ by at least 0.5 unit from the isoelectric point of the target molecule to ensure product stability. The pH and conductivity need to be fine-tuned to maximize the binding capacity and separation. Hydrophobic interaction chromatography (HIC) resins have been employed in protein purification steps and offer major advantages such as high adsorption capacity, high selectivity, mild elution conditions, and low cost [ 20 ]. By selecting a resin with suitable hydrophobic strength and optimizing the high-salt loading condition, better separation effects can be achieved. Furthermore, HIC elution is performed under low-salt conditions, making it relatively friendly for subsequent processes.

An external file that holds a picture, illustration, etc.
Object name is tbad028f4.jpg

The purification process of tag-free recombinant protein.

Our own practice demonstrated that the capture strategy of an acidic recombinant protein with a pI of 5.9 ( Fig. 5 ). The protein solubility was influenced by pH and conductivity, with precipitation occurring at low pH and low conductivity. On the other hand, to enable capture by CEX, pH should be kept below the protein’s pI and conductivity should be low to maintain sufficient electrostatic interaction. Therefore, the loading condition needs to be carefully adjusted to enable protein capture while maintaining protein stability. From Figure 5(B) , pH had a greater impact on recovery than conductivity within the test range. Based on these results, the loading conditions were set at pH 5.5 and conductivity of 11 mS/cm to achieve optimal capture capability.

An external file that holds a picture, illustration, etc.
Object name is tbad028f5.jpg

Counter plot of solution conductivity and pH versus recovery. (A) Recovery from protein precipitation at different pH and conductivity. (B) The CEX recovery at different loading pH and conductivity.

Unstable protein

Ensuring protein stability throughout the process is the primary consideration of protein purification, as most proteins are susceptible to changes in temperature, pH, or salt concentration [ 11 ]. Due to the complexity of artificially designed non-natural proteins, they are prone to issues such as precipitation and reduced quality during conventional purification process. The following summarizes the factors causing protein instability and the strategies to address this problem during protein purification process.

Buffer system and conditions

Inappropriate pH condition is the most common factor leading to protein instability, which results in rapid protein aggregation and fragmentation [ 21 ]. The elution buffer for AC is typically acidic, and the prolonged exposure to this condition leads to a sustained decline in protein activity. Therefore, upon elution, the pH should be adjusted back to neutral as soon as possible to prevent long-term activity reduction. In addition, the buffer system and protein concentration are also crucial for stability. For loosely folded proteins, inappropriate solution conditions can lead to partial exposure of hydrophobic regions, thereby accelerating the formation of aggregates.

Stabilizers

During the virus inactivation process, organic solvents have been employed as a solution to address the instability issue caused by low pH. However, the addition of inappropriate organic reagents can induce higher degree of aggregation formed through non-covalent binding. Addition of PS80 alone with proper concentration and treatment time can achieve the desired viral inactivation [ 22 ]. Surfactants not only have the ability to inactivate viruses but also helps to maintain protein stability. During the process development, significant precipitation occurred during the affinity step for proteins with strong hydrophobicity. Considering the relatively extended molecular structure and the presence of exposed hydrophobic regions in the molecule, a surfactant was introduced in the process buffer to stabilize the protein structure and enhance the product recovery significantly.

Other factors

Heat and shear forces can cause irreversible protein aggregation, while freezing and dehydration may result in conformational changes in some intricate protein structure and affect their activities. Although excipients are typically added to stabilize the drug substance (DS) stability during freeze-thaw, adding extra excipients in the process intermediate is not desirable during the purification process. Therefore, shortening the purification process duration and minimizing the sample freeze–thaw is the most effective and convenient approach.

Table 2 summarizes the factors causing protein structural changes during the purification process and their corresponding resolution strategies.

The factors affecting protein stability during the purification process and the corresponding resolution strategies

Influence factorsStageStrategies
Molecular structureAllAdd stabilizers such as PS80
For proteins that are sensitive to metal ions, ensure the purity of the reagents and optimize the amount added
pHACShorten the storage time in low pH condition
Choose high pH elution affinity resin or optimize elution buffer system
VINUse S/D inactivation or other inactivation reagents
Ionic conditionsHICScreen highly hydrophobic resin to reduce the conductivity required during sample loading
Try flow-through mode to avoid high conductivity loading
AllMaintain optimal salt concentration for salt solubilized proteins
Buffer systemAllChoose the optimal buffer system
FreezingIntermediatesShorten the processing time to avoid freezing
Add cryoprotectants without affecting the process
DSAdd cryoprotectants
TemperatureAllAvoid high-temperature conditions, thaw at low temperature
Shear forceUFDFUse low shear equipment, reduce operation pressure and add protectants

Aggregate removal

Protein aggregation is one of the most challenging aspects in protein purification. Part of the aggregates originate from the protein formation during cell culture processes due to covalent or non-covalent interaction. The late-stage aggregation is largely dependent on buffer conditions, including pH, ionic strength, buffer system and additives [ 23 , 24 ]. Different modes of chromatography are utilized to remove aggregates, depending on the variations in the physical and chemical properties between the target protein and the aggregates.

Affinity-based purification

Both monomers and aggregates can be captured by affinity resins. The differences in affinity can yield distinct separation effects on aggregates, as Protein A can simultaneously bind to the Fc and Fab regions of the antibody, and the binding affinity depends on the design of the resin and antibody [ 25 ]. As shown in Figure 6 , the affinity between Protein A and antibody depends on the structure of both. Native-Protein A has weak interaction with the Fab region as well as strong binding with the Fc region. In contrast, recombinant Protein A lacks interaction with Fab. Studies found that each monomer can bind to only one Protein A ligand, while each dimer can bind to two ligands [ 26 ]. Therefore, aggregates of one mAb are more strongly retained compared with the mAb monomer. On the other hand, during aggregate formation, the Fc regions might be buried inside or partially lost [ 25 ]. For this type of aggregates, the interaction with recombinant Protein A becomes weaker than the interaction between mAb monomer and protein A. Frequently, incorporating suitable wash steps before elution can effectively eliminate these aggregates at an early stage.

An external file that holds a picture, illustration, etc.
Object name is tbad028f6.jpg

Protein A-mAb interaction. The polymer can simultaneously bind multiple ligands of the resin, while the mAb can only bind a single ligand. The binding site and binding capacity of aggregate and affinity resin depend on protein A and aggregate types.

Additives are commonly used to separate aggregates and target proteins. The addition of calcium chloride/polyethylene glycol (PEG) or sodium chloride/PEG combinations to the wash and elution buffers can effectively enhance the separation of aggregates and target antibodies [ 27 ]. The Hofmeister series of salts can improve resin selectivity by adjusting the hydrophobic interactions between antibody species and Protein A ligands. The use of magnesium chloride and calcium chloride as elution additives has resulted in excellent separation between bispecific products and Fc-Fc homodimers [ 28 ].

Charge-based purification

Aggregation gives rise to protein surface coverage, leading to disparities in surface charge when compared to individual monomers. Although aggregates are usually more tightly bound to the CEX resin than monomeric proteins, the separation between the two is generally difficult, especially when the aggregates are dimers [ 29 ]. Besides screening different types of cationic resin, the separation of monomers and aggregates can be improved by adding excipients to the mobile phase [ 30 ]. The utilization of a pH-conductivity hybrid gradient elution method has demonstrated its efficacy in eliminating aggregates from samples, if a consistent, linear and reproducible pH profile is provided [ 31 ].

Hydrophobicity-based purification

HIC presents notable benefits in aggregate removal, owing to the increased surface hydrophobicity exhibited by aggregates in comparison to monomers [ 32 ]. The efficacy of separation through HIC can be influenced by protein hydrophobicity, salt type and concentration, and hydrophobicity of the resin. By reducing the salt concentration or using resins with stronger hydrophobicity, the target protein can flow through while the aggregates and other impurities are retained, thus avoiding protein instability caused by high conductivity [ 33 ]. Additives with different polarities can alter the hydrophobic interactions among proteins, impurities and the resin, thereby improving resolution and achieving better separation.

Multimodal-based purification

Some proteins and aggregates cannot be separated effectively by using chromatography with a single mechanism. Multimodal chromatography allows for separation based on charge, hydrophobicity, hydrogen bonding, and other factors in a single step, and therefore provides better selectivity. Ceramic hydroxyapatite (CHT™) is one type of multimodal resin with a dual exchange mechanism based on phosphate groups for cation exchange and calcium ion metal chelation. Experimental results have demonstrated the excellent efficacy of CHT™ resin in removing aggregates, DNA, endotoxins, and other impurities [ 34 ]. Therefore, in the development of BsAbs, fusion proteins, and recombinant proteins, CHT™ resin can be considered for the removal of challenging impurities.

The formation mechanism of aggregates is complex, which leads to heterogeneous structural characteristics. Therefore, the removal of aggregates requires thorough optimization efforts. Table 3 summarizes the methods and optimization strategies for removing protein aggregates during the purification process.

Strategies for aggregate removal using different types of chromatography

Type of chromatographyModeOptimization strategy
ACB-E1. Affinity resin screening
2. Wash buffer selection
3. Elution buffer selection
4. Additive selection in the elution buffer, such as polyols, amino acids and high-valent metal salts.
5. Elution pH optimization
IEXF-TIncreasing pH to remove aggregates based on weak binding mode.
B-E1. Investigation of sample pH and conductivity to modulate protein binding.
2. Elution methods selection, exploring the resolution differences between salt elution and pH elution.
3. Combination of elution pH and conductivity as optimization factors.
4. Additive selection in the elution buffer, such as polyols and amino acids.
HICB-E1. Selection of resin, where strong hydrophobic resins can be used for lower conductivity loading, and weak hydrophobic resins for higher conductivity loading.
2. Additive selection in the elution buffer, such as polyols and amino acids
F-T1. Selection of resin depending on hydrophobicity.
2. Sample pH and conductivity optimization.

Note: B-E, bind-and-elute; F-T, flow-through.

HCP removal

HCPs are heterogeneous mixtures of proteins secreted by living cells and intracellular proteins released upon cell death and lysis, with the majority of HCP impurities being acidic proteins. Over 6000 HCPs derived from CHO cells have been identified through proteomics and other methods [ 35 ]. In downstream processes, protein purification typically involves the use of orthogonal chromatographic separation methods to achieve HCP removal. Nevertheless, achieving higher titers of such proteins during cell cultivation proves difficult, which consequently leads to higher HCP contents for tag-free recombinant proteins (> 10 000 ppm) and BsAb or fusion proteins (a few thousand ppm) as compared with typical mAb (< 2000 ppm) after capture.

Depth filtration

Depth filtration (DF) is used for solid–liquid separation and can also be employed for HCP removal utilizing electrostatic interaction between the charged binder and proteins. Therefore, two separation strategies are employed: first, controlling the conditions to promote HCP precipitation; and second, removing soluble HCPs through electrostatic interaction with the depth filter material. The pI of the majority of CHO cell-derived HCPs falls within the pH range of 4.5–7.5, and they become less soluble during the neutralization process, leading to aggregation and precipitation. In mAb development, controlling the pH after low pH inactivation allows HCPs to precipitate from solution, enabling their removal during intermediate depth filtration [ 36 ]. An optimal process with pH adjustment can be designed to achieve selective precipitation of HCPs while minimizing product loss.

Affinity chromatography

AC is an ideal tool for protein capture, as the target protein can be strongly bound to the resin in a specific manner. However, some HCPs are co-eluted with the products due to non-specific interaction with the Protein A resin or non-specific binding with the target proteins [ 37 ]. A common approach is to use wash buffer with high salt concentration or low pH (5–5.5) to remove a part of the HCP prior to elution, but these conditions often do not completely disrupt the binding between HCPs and the target protein. Recent studies have found that using high pH and high conductivity wash buffer solutions can remove HCPs more effectively, as the electrostatic interactions between the mAb and most HCPs are repulsive under alkaline conditions (pH > 8) [ 36 ]. Since HCPs can bind to proteins through various mechanisms, including electrostatic interactions, hydrophobic interactions, and hydrogen bonding, reagents that disrupt these interactions can be used to effectively remove HCPs. To date, wash buffer solutions containing different additives have been tested for HCP removal [ 38–40 ]. Histidine hydrochloride has shown good HCP removal results, as it can form hydrogen bonds with proteins, disrupting hydrophobic and electrostatic interactions [ 40 ].

Ion exchange chromatography

The separation of HCPs is attributed to charge–charge interactions. Most HCPs belong to acidic proteins, so AEX in flow-through mode is effective for HCP removal in the case of basic target proteins. In the flow-through mode, weak partitioning chromatography (WPC) has been proposed for the purification of mAbs using a relatively high pH to enhance HCP removal [ 41 ]. For the bind-and-elute mode, the pI differences between HCPs and target proteins can be exploited to remove parts of HCPs prior to protein elution, thus minimizing co-elution of HCPs with the target protein. Additionally, amino acids have been used in various chromatographic separations to enhance the clearance of HCP during mAb purification processes [ 42 , 43 ]. Our own practice showed the use of CEX in combination with additional wash steps and amino acid additives in the elution buffer to achieve effective HCP removal for a recombinant protein with a pI of 5.9 ( Fig. 7 ). The increase of wash buffer strength induces a partial loss of the target protein, but it proves effective in removing weakly bound HCPs. The addition of arginine in the elution buffer further reduces HCP content. The presence of arginine stabilizes the pH after protein elution, preventing the elution of more strongly bound HCPs. Therefore, by adding a wash step and optimizing the elution buffer, the HCP removal capability can be significantly enhanced.

An external file that holds a picture, illustration, etc.
Object name is tbad028f7.jpg

The CEX chromatograms and quality results for different wash and elution conditions for a tag-free recombinant protein. (A) No wash, elution buffer: Tris-HAc. (B) Wash + elution buffer: Tris-HAc. (C) Wash + elution buffer: arginine. (D) The results for different wash and elution conditions.

Hydrophobic interaction chromatography

Amino acids, which are the building blocks of proteins, differ in both charge and polarity, resulting in variations in the hydrophobicity between the target protein and HCP. HIC in both bind-and-elute mode and low salt flow-through mode have demonstrated great HCP removal capabilities [ 44 ]. The selectivity can be improved by controlling the type and concentration of salt, temperature, pH and ligands used in the HIC process. Figure S2 summarizes the effects of HIC in HCP removal for our selected platform projects. Both the bind-and-elute mode and the flow-through mode can achieve good HCP removal, with an average removal rate of over 90%.

Multimodal chromatography

Multimodal resins possess dual properties of ionic exchange and hydrophobicity, the combination of multiple interactions may provide better selectivity. Capto™ Adhere resin, harnessing both anion exchange and hydrophobic interaction functionalities, could achieve a clearance capability of up to 2–3 LRV to reduce HCP levels below 10 ppm [ 45 ]. Capto™ MMC resin, with both cation exchange and hydrophobic properties, is typically used in bind-and-elute mode and has demonstrated good HCP and impurity removal capabilities under optimal process conditions [ 46 ]. CHT™ resin, operating in flow-through mode, has also exhibited excellent competence in HCP removal [ 47 ]. However, achieving enhanced separation through multimodal chromatography requires comprehensive Design of Experiments study to explore the potential interaction of different factors.

Among all impurities related to the process, HCPs are usually of particular concern because they can affect the safety, efficacy and stability of the product. Recently, the development of high density and perfusion cell culture has introduced more challenges in the purification of increased level of HCPs and other impurities. Figure 8 recommends effective strategies for HCP removal at different stages of downstream processing. The purification should be optimized towards maximizing the overall removal capability of the process.

An external file that holds a picture, illustration, etc.
Object name is tbad028f8.jpg

Effective strategies for HCP removal at different stages of downstream processing.

Protein recovery

For complex proteins, the presence of a significant level of aggregates and impurities such as HCPs leads to lower yields. Therefore, maximizing recovery while maintaining product quality becomes a pivotal challenge for protein purification process development.

DF is used for the initial clarification of cell culture broth to protect chromatography columns. Selecting appropriate DF filter is key to reduce the protein loss due to adsorption. Uneven charges on a portion of the protein surface result in electrostatic interactions with the charged membrane, causing protein adsorption. Hence, investigation of the DF membrane with different materials improves the protein recovery rate and reduces loss from protein adsorption. Besides, partial adsorption can be mitigated by increasing the conductivity of the wash buffer. Nevertheless, special attention should be given to the adjustment of wash buffer salt concentration, as excessively high conductance is potentially unsuitable for the subsequent separation phase.

In AC, strong binding of protein on the resin may make the elution of protein difficult, leading to low protein recovery. Previous studies have shown that the presence of high ionic strength may promote hydrophobic interactions between protein and Protein A, therefore requires lower pH for elution [ 48 ]. Additionally, fusion proteins generally exhibit weaker stability compared with mAbs, especially under low pH conditions, which pose challenges for elution at lower pH. By using arginine buffer or other additives, elution can be achieved at high pH while ensuring high recovery rate. HIC is often associated with low yield due to unfavorable solvent conditions, or denaturation of target protein on the hydrophobic surface. Various methods have been explored to improve the recovery rate of HIC. In HIC, bind-and-elute mode frequently led to overly strong protein binding, posing challenges in protein elution. Arginine and hexylene glycol can weaken the interactions between proteins and the resin matrix, thereby improving the recovery rate and purity of various proteins [ 49 ].

While striving for high purity by addressing issues such as the removal of aggregates and HCPs, part of the protein product is inevitably lost. Based on the data from our platform, protein loss primarily occurs during depth filtration and various chromatography steps. For depth filtration, selection of appropriate membranes and optimization of wash buffers are the main measures to address the protein loss. For chromatography, the addition of excipients in the product intermediate and process buffer is an effective strategy to improve the recovery by modulating the binding affinity between proteins and resins.

FUTURE PERSPECTIVES AND CONCLUSION

The development of therapeutic protein drugs is a multifaceted systemic engineering endeavor that involves diverse disciplines. In comparison to the substantial advancements seen in upstream processing areas such as molecular design and cell line engineering, there have been relatively fewer reports on the development of protein downstream purification methods. As we have discussed in this review, the diversity of BsAbs, fusion proteins, and recombinant proteins presents a significant challenge in developing effective and economical purification processes.

Chromatography techniques play a pivotal role in protein purification; however, the development of resins lags behind the demand arising from antibody technology advancement. Affinity resins and multimodal resins represent the most promising chromatographic media for addressing challenges in protein purification. Both types of resins exhibit stronger selectivity for impurities, enabling better differentiation between different proteins and process-related impurities. In contrast, progress in the development of filtration steps has been slow-paced, making ideal impurity removal based on filtration processes difficult to achieve. Nevertheless, the combination of resins and filtration processes in product design may offer a promising direction for breakthroughs in consumables. Such products would merge the efficiency of filtration processes with the selectivity of chromatographic processes, potentially enhancing the efficiency and economy of purification significantly.

In addition to anticipating suppliers to provide improved purification consumables, efficient experimental design is another pursuit in the development of purification processes. The design of protein purification processes is heavily based on empirical knowledge and a large number of trial-and-error experiments, often resulting in suboptimal overall processes and low utilization of raw materials. Addressing this issue primarily involves reducing the number of experiments and increasing experimental efficiency. Rational experimental design is the basis for reducing the number of experiments, requiring sufficient understanding of the target protein. Computational biology represents a future trend that leverages computer models and machine-learning algorithms to predict the properties of target proteins, identify correlations and optimize purification process conditions, thereby reduces trial-and-error experiments. This has the potential to fundamentally enhance the accuracy and efficiency of purification process development. On the other hand, the introduction of high-throughput screening technology has greatly improved the efficiency of protein purification. High-throughput screening accelerates the evaluation, optimization and selection of purification processes by parallel processing a large number of samples. The combination of computer-aided design and high-throughput screening is an excellent option for future purification process development, which is worth looking forward to.

In summary, this overview demonstrates that challenges still exist in seeking to improve the development of bioprocess purification, while also highlights the significant progress that has been made. Through the development of rational processes, we can address the majority of challenges to meet regulatory requirements. With the continuous advancement of technology, the exploration of more efficient, accurate and environmentally friendly protein purification methods will continue to drive the development of this field.

Supplementary Material

Supplementary_data_tbad028, acknowledgements.

The authors thank Weijuan Shen, Qianqian Zhu, Zhaoxin Liu, Shuyu Zhou, Weiwei Gu and Peilei Liu for helping generate data shown in the case studies in this article.

Contributor Information

Shuo Tang, GenScript ProBio Biotechnology Co., Ltd, Nanjing, Jiangsu 21100, P.R. China.

Jiaoli Tao, GenScript ProBio Biotechnology Co., Ltd, Nanjing, Jiangsu 21100, P.R. China.

Ying Li, GenScript ProBio Biotechnology Co., Ltd, Nanjing, Jiangsu 21100, P.R. China.

This work was funded by GenScript ProBio Biotechnology Co., Ltd.

CONFLICT OF INTEREST STATEMENT

Shuo Tang, Jiaoli Tao and Ying Li are current employees of GenScript ProBio Biotechnology Co., Ltd.

AUTHOR CONTRIBUTIONS

Shuo Tang (Data curation-lead, Formal analysis-lead, Investigation-equal, Project administration-equal, Writing-original draft-lead, Writing-review and editing-equal), Jiaoli Tao (Data curation-supporting, Investigation-supporting, Writing-original draft-supporting) and Ying Li (Conceptualization-lead, Investigation-lead, Methodology-lead, Project administration-equal, Supervision-lead, Writing-review and editing-lead).

DATA AVAILABILITY

Ethics and consent statement.

No patient consent is required.

ANIMAL RESEARCH STATEMENT

Not applicable.

Academia.edu no longer supports Internet Explorer.

To browse Academia.edu and the wider internet faster and more securely, please take a few seconds to  upgrade your browser .

Enter the email address you signed up with and we'll email you a reset link.

  • We're Hiring!
  • Help Center

paper cover thumbnail

Downstream Processing

Profile image of Pablo Moreira

Related Papers

Biotechnology and Bioengineering

Balaji Somasundaram

research papers on downstream processing

Biotechnology Progress

Nigel Titchener-hooker

International Journal for Research in Applied Science & Engineering Technology (IJRASET)

IJRASET Publication

The creation of therapeutic monoclonal antibodies has increased recently. mAbs have grown significantly in relevance, and these compounds are now more crucial than ever in the global biotechnological drug industry. For the treatment of various diseases, a lot of biotechnology companies are investing in the manufacture of monoclonal antibodies. The two main processes in the synthesis of these antibodies are bioreactor production and purification. This article goes into great detail on the current purification techniques utilized for these compounds. The fundamental monoclonal antibody recovery and purification procedures, including cell harvesting, Protein A affinity chromatography, and further polishing steps, are all enumerated.

International Journal of Pharmaceutics

Christopher Maycock

Shritama Ray

Monoclonal antibody (mAb) therapy is a form of immunotherapy that uses mAbs to bind mono-specifically to certain cells or proteins. This may then stimulate the patient&#39;s immune system to attack those cells. MAbs are currently used to treat medical conditions such as cancer, diabetes, arthritis, psoriasis, and Crohn’s Disease, but have the potential to treat countless diseases and disorders. In 2015, the mAb market was valued at $85.4 billion, and is expected to reach $138.6 billion by 2024.1 In manufacturing, mAbs are typically produced in suspension in a series of fed-batch bioreactors using genetically engineered cells originally obtained from Chinese Hamster Ovaries (CHO).2 In this proposal, two upstream bioreactor designs were analyzed for economic comparison given an annual production goal of 100 kg of mAb, with the first design culminating in a 20,000 L volume at low mAb titer and the second design culminating with a 2,000 L volume at high mAb titer. Following upstream mAb...

Patricia Rowicki

The emergence of monoclonal antibody (mAb) therapies has created a need for faster and more efficient bioprocess development strategies in order to meet timeline and material demands. In this work, a high‐throughput process development (HTPD) strategy implementing several high‐throughput chromatography purification techniques is described. Namely, batch incubations are used to scout feasible operating conditions, miniature columns are then used to determine separation of impurities, and, finally, a limited number of lab scale columns are tested to confirm the conditions identified using high‐throughput techniques and to provide a path toward large scale processing. This multistep approach builds upon previous HTPD work by combining, in a unique sequential fashion, the flexibility and throughput of batch incubations with the increased separation characteristics for the packed bed format of miniature columns. Additionally, in order to assess the applicability of using miniature column...

Ana Azevedo

The commercial potential of monoclonal antibodies (mAbs) has been continuously increasing during the last years alongside with the number of approved mAb-based drugs and clinical trials. Despite their effectiveness and safety, the general access to this class of biopharmaceuticals is barred by high selling prices. Downstream processing is now considered the bottleneck in the manufacturing of mAbs. Therefore, the design of novel and economic operations and their implementation in the current technology platforms constitutes a pressing need. This review provides an insight into the current state-of-the-art in mAbs purification, focusing on multimodal chromatography as one of the viable options to upgrade the established purification train.

jonathan Coffman

Muthu Kumar

Acta Scientific Medical Sciences

Burak Erkal

Loading Preview

Sorry, preview is currently unavailable. You can download the paper by clicking the button above.

RELATED PAPERS

Journal of Membrane Science

Eva Rosenberg , S. Hepbildikler , Wolfgang Kuhne

Journal of Clinical Medicine

sanjeevani ghone

Gregory Zarbis-Papastoitsis , marco cacciuttolo

xionghui li

Journal of Pharmaceutical and Biomedical Analysis

Evelin Farsang

American Journal of Biochemistry and Biotechnology

Ali Demirci

Biotechnology Journal

Chia-Yun Sun

Alex Chatel

Journal of biotechnology

Boris Napadensky , Andrew Tustian

Darrin Tisdale

Chemical Engineering Science: X

Ruud van Beckhoven

IRJET Journal

abiy berhanu

Separations

Amarande Murisier

Journal of Chromatography a

Daniel Bracewell

Journal of Chromatography A

Maria Chiara Zorzoli

Biologicals

Anja ter Avest

Biotechnology and applied biochemistry

Martin Vanderlaan

Kerstin Thurow

  •   We're Hiring!
  •   Help Center
  • Find new research papers in:
  • Health Sciences
  • Earth Sciences
  • Cognitive Science
  • Mathematics
  • Computer Science
  • Academia ©2024

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 13 September 2024

The Wild West of spike-in normalization

  • Lauren A. Patel   ORCID: orcid.org/0000-0003-3495-5887 1 , 2 , 3 ,
  • Yuwei Cao   ORCID: orcid.org/0009-0000-4119-4399 3 , 4 ,
  • Eric M. Mendenhall 5 ,
  • Christopher Benner 2 &
  • Alon Goren   ORCID: orcid.org/0000-0001-5669-9357 3  

Nature Biotechnology volume  42 ,  pages 1343–1349 ( 2024 ) Cite this article

Metrics details

  • Chromatin analysis
  • Chromatin immunoprecipitation

The proper use of spike-in normalization in ChIP-seq improves sensitivity for detecting genome-wide changes between conditions, but improper use is common, calling some biological conclusions into question. A survey of public datasets generates guidelines for implementation of spike-in normalization for future ChIP-seq experiments.

This is a preview of subscription content, access via your institution

Access options

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

24,99 € / 30 days

cancel any time

Subscribe to this journal

Receive 12 print issues and online access

195,33 € per year

only 16,28 € per issue

Buy this article

  • Purchase on SpringerLink
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

research papers on downstream processing

Data availability

Data generated in this study (Fig. 1b , Fig. 4 , and Supplementary Figs. 2–6 and 8–14 ) are at GSE273915 . Public data used to generate Fig. 1a are from GSE60104 . UCSC Genome Browser sessions are available for Fig. 1b , Fig. 4 human (target) data and Fig. 4 yeast (spike-in) data .

Code availability

Code is available on a Github repository.

Orlando, D. A. et al. Cell Rep. 9 , 1163–1170 (2014).

Article   CAS   PubMed   Google Scholar  

Bonhoure, N. et al. Genome Res. 24 , 1157–1168 (2014).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Meers, M. P., Bryson, T. D., Henikoff, J. G. & Henikoff, S. eLife 8 , e46314 (2019).

Article   PubMed   PubMed Central   Google Scholar  

Chen, K. et al. Mol. Cell. Biol. 36 , 662–667 (2015).

Article   PubMed   Google Scholar  

Jiang, L. et al. Genome Res. 21 , 1543–1551 (2011).

Skene, P. J. & Henikoff, S. eLife 6 , e21856 (2017).

Wooten, M., Takushi, B., Ahmad, K. & Henikoff, S. Sci. Adv. 9 , eadg3257 (2023).

Terekhanova, N. V. et al. Nature 623 , 432–441 (2023).

Javasky, E. et al. Genome Res. 28 , 1455–1466 (2018).

Guertin, M. J., Cullen, A. E., Markowetz, F. & Holding, A. N. Nucleic Acids Res. 46 , e75 (2018).

Egan, B. et al. PLoS One 11 , e0166438 (2016).

Ma, Z. et al. eLife 7 , e35368 (2018).

Wang, Z. et al. Nat. Genet. 54 , 295–305 (2022).

Yano, S. et al. Nat. Commun. 13 , 4440 (2022).

Nakato, R. et al. Nat. Commun. 14 , 5647 (2023).

Greulich, F., Wierer, M., Mechtidou, A., Gonzalez-Garcia, O. & Uhlenhaut, N. H. Cell Rep. 34 , 108742 (2021).

Okabe, A. et al. Nat. Genet. 52 , 919–930 (2020).

Kent, W. J. et al. Genome Res. 12 , 996–1006 (2002).

Landt, S. G. et al. Genome Res. 22 , 1813–1831 (2012).

Cherry, J. M. et al. Nucleic Acids Res. 26 , 73–79 (1998).

Love, M. I., Huber, W. & Anders, S. Genome Biol. 15 , 550 (2014).

Grzybowski, A. T., Chen, Z. & Ruthenburg, A. J. Mol. Cell 58 , 886–899 (2015).

Vale-Silva, L. A., Markowitz, T. E. & Hochwagen, A. BMC Genom. 20 , 54 (2019).

Article   Google Scholar  

Download references

Acknowledgements

Research reported in this publication was supported in part by US National Institutes of Health/National Institute of Mental Health grant R01MH127077 (A.G. and C.B.) and National Science Foundation grant 2003358 (A.G.). We thank I. Simon for providing conceptual guidance and feedback on the manuscript, R. Wachs for helping with illustrations and the Pillus lab at UCSD for providing S. cerevisiae cells and guidance on yeast culture and ChIP-seq.

Author information

Authors and affiliations.

Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA

Lauren A. Patel

Department of Medicine, Division of Endocrinology & Metabolism, University of California San Diego, La Jolla, CA, USA

Lauren A. Patel & Christopher Benner

Department of Medicine, Division of Genomics & Precision Medicine, University of California San Diego, La Jolla, CA, USA

Lauren A. Patel, Yuwei Cao & Alon Goren

Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, USA

HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA

Eric M. Mendenhall

You can also search for this author in PubMed   Google Scholar

Corresponding authors

Correspondence to Christopher Benner or Alon Goren .

Ethics declarations

Competing interests.

L.A.P., Y.C., C.B. and A.G. are inventors on related patent applications.

Peer review

Peer review information.

Nature Biotechnology thanks the anonymous reviewers for their contribution to the peer review of this work.

Supplementary information

Supplementary information.

Descriptions of Supplementary Tables 1–5, Supplementary Figs. 1–15, Table 1 and Methods.

Supplementary Table

Supplementary Tables 2–5.

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Patel, L.A., Cao, Y., Mendenhall, E.M. et al. The Wild West of spike-in normalization. Nat Biotechnol 42 , 1343–1349 (2024). https://doi.org/10.1038/s41587-024-02377-y

Download citation

Published : 13 September 2024

Issue Date : September 2024

DOI : https://doi.org/10.1038/s41587-024-02377-y

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

research papers on downstream processing

COMMENTS

  1. Current research approaches in downstream processing of

    Owing to a rapidly increasing market demand for therapeutic proteins, for example, monoclonal antibodies (mAbs), the industry is facing the challenge of efficiently manufacturing large-product amounts [1].After recent process-intensification improvements in the upstream processing (USP), the current bottleneck of production processes shifts to in the purification of protein products in the ...

  2. 15518 PDFs

    Explore the latest full-text research PDFs, articles, conference papers, preprints and more on DOWNSTREAM PROCESSING. Find methods information, sources, references or conduct a literature review ...

  3. Downstream Processing

    Downstream process: toward cost/energy effectiveness. Ramesh Kumar, ... Alak Kumar Ghosh, in Handbook of Biofuels, 2022. Abstract. Downstream processing (DSP) involves multistaged unit operations after upstream processes that improve the quality of the final product in terms of concentration and purity. One of the most important objectives of the DSP, in addition to maximizing product recovery ...

  4. Recent Advances and Future Directions in Downstream Processing of

    A variety of downstream processing technological advances have led to a paradigm shift in how therapeutic antibodies are developed and manufactured. A key driver of change has been the increased adoption of single-use technologies for process development and manufacturing. ... Feature papers represent the most advanced research with significant ...

  5. Status and future developments for downstream processing of biological

    Downstream processing plays an important role in the supply of pharmaceuticals and, in particular, biopharmaceuticals, where a large proportion of the production costs are attributable to the recovery and purification processes (Straathof, 2011) although this may have shifted to upstream processes in the case of cell and gene therapy ...

  6. Research review paper Downstream processing technologies in the

    Unit operations and their assembly to downstream processing (DSP) routes for carbohydrate products are the focus of the current study (Hatti-Kaul, 2010; Illanes et al., 2016; Moreno et al., 2014a). Functional carbohydrates can be classified structurally into monosaccharides, disaccharides, oligosaccharides and polysaccharides.

  7. Downstream Processing; Applications and Recent Updates

    Development of efficient and economical downstream processing strategies has been applied in industrial manufacture of enzymes because of their important role not only in food and agricultural industries but also used in textile, paper and pharmaceutical industries, scientific research, etc (Roque et al., 2004).

  8. Current research approaches in downstream processing of

    Therefore, an urgent need for process intensification in downstream processing (DSP) has been identified to produce biopharmaceuticals more efficiently. The DSP currently accounts for the majority of production costs of pharmaceutically relevant proteins. This short review gathers essential research over the past 3 years that addresses novel ...

  9. Downstream Processing Technologies/Capturing and Final Purification

    Downstream processing (DSP) is engaged in the separation and refinement of mixtures of components. In its simplest definition, DSP encompasses a tool box of separation techniques designed to achieve mass transfer phenomena, converting mixtures of substances into subsets of mixtures or fractions [].At its onset, industrial DSP of proteins considered many of the traditional chemical unit ...

  10. Downstream process development strategies for effective bioprocesses

    The platform process most applied in biopharmaceutical downstream processing (DSP) is the systematic purification of mAbs and Fc fusion proteins. Shukla et al. 41 described the antibody platform process in several consecutive steps, which are consistent in process structure, but single unit operations are exchangeable. After cell harvest and ...

  11. downstream processing Latest Research Papers

    Downstream Processing . Leader Peptide . Oxidative Decarboxylation . Cancerous Cell . In Vitro Reconstitution . Modified Peptides . In Vitro Biosynthesis. Thioholgamides are ribosomally synthesized and post-translationally modified peptides (RiPPs) with potent activity against cancerous cell lines and an unprecedented structure.

  12. Downstream Processing for Biopharmaceuticals Recovery

    However research and development of the concerned firms is working to get advanced ways to improve the downstream processing of biopharmaceuticals. The upcoming future trends are in silico-based methods: algorithmic (super structure optimisation, model-based hybrid approach) and non-algorithmic (heuristic approach based, experimental and ...

  13. Recent Advances and Future Directions in Downstream Processing of

    A variety of downstream processing technological advances have led to a paradigm shift in how therapeutic antibodies are developed and manufactured. A key driver of change has been the increased adoption of single-use technologies for process development and manufacturing. ... This is an active area for further research and development. 5 ...

  14. Intensified Downstream Processing of Monoclonal Antibodies Using

    Moreover, downstream processing steps now require the capacity to process 15-100 kg of mAb per batch, despite being traditionally designed to process 5-10 kg of mAb. The challenge in processing such large quantities of products is that the resin-based chromatography operations are limited by scale-up to a maximum column diameter of 2.0 to 2 ...

  15. Downstream Processing in Biotechnology

    The current book gives an excellent insight into downstream processing technology and explains how to establish a successful strategy for an efficient recovery, isolation and purification of biosynthetic products. In addition to the overview of purification steps and unit operations, the authors provide practical information on capital and operating costs related to downstream processing.

  16. Downstream Processing Research Papers

    Perspectives for an improved downstream processing of biologically produced diols, especially 1,3-propanediol are discussed based on our own experience and recent work. It is argued that separation technologies such as aqueous two-phase extraction with short chain alcohols, pervaporation, reverse osmosis, and in situ extractive or pervaporative ...

  17. Downstream Processing

    Downstream processing refers to the series of unit operations used to isolate, purify, and concentrate the product. Downstream processing often determines the economic feasibility of the process. The first operation is cell separation, which can be done by cross-flow microfiltration. When a microfilter or ultrafilter is combined in a semi ...

  18. Downstream Processing of Biotechnology Products

    This chapter provides an overview of the chemical and biophysical properties of proteins and their main contaminants such as DNA and endotoxins. It discusses commonly used expression systems and the general structure of downstream processes needed to achieve the desired product purity. The chapter focuses on the production of recombinant ...

  19. Challenges and solutions for the downstream purification of therapeutic

    The article summarizes the primary challenges encountered during the downstream processing of proteins, and presents effective solutions and case studies to tackle each major challenge. ... With the advancement of genetic engineering technology, research in protein drugs achieved significant breakthroughs. The application of technologies such ...

  20. (PDF) Overview of Upstream and Downstream Processing of

    Academia.edu is a platform for academics to share research papers. Overview of Upstream and Downstream Processing of Biopharmaceuticals ... fed-batch, continuous, perfusion • Downstream processing - Philosophy - Chromatography - Examples • Conclusions 2 What is a bioprocess? • Application of natural or genetically manipulated ...

  21. (PDF) Downstream Processing

    This solvent-detergent 442 Chapter 11 - Downstream Processing f (S/D) treatment, originated at the New York Blood Center in the 1980s, is the most widespread method of viral inactivation in plasma fractionation processes and is used in the production of some recombinant proteins.

  22. Research review paper Current and novel approaches to downstream

    The low dry weight content of microalgae cultures (typically around 1 g L −1) makes harvesting and concentration an expensive unit operation of any biorefinery.Harvesting and dewatering are the main downstream processes required for any applications where the whole microalgae biomass can be used directly as the final product, such as for animal and fish feed or food supplements (e.g ...

  23. The Wild West of spike-in normalization

    The proper use of spike-in normalization in ChIP-seq improves sensitivity for detecting genome-wide changes between conditions, but improper use is common, calling some biological conclusions into ...

  24. Full article: Histogel-based techniques for embedding organoids in

    Organoid generation from intestinal resections. Segments of human colonic tissue were supplied by Donor Network West. Donor Network West received IRB approval of research, appropriate informed consent of all subjects contributing biological materials, and all other authorizations, consents, or permissions as necessary for the transfer and use of the biological materials for research at Genentech.

  25. Downstream processing of proteins: Recent advances

    Research review paper. Downstream processing of proteins: Recent advances. Author links open overlay panel John R. Ogez a, James C. Hodgdon a, Marc P. Beal a, Stuart E ... DOWNSTREAM PROCESSING OF PROTEINS 471 CELL DISINTEGRATION Mechanical disintegration is the most common form of cell disruption used in industry today, although treatment with ...

  26. Us Strategic Metals Announces Three-way Strategic Partnership for

    Today's announcement adds a processing LOI with USSM that builds upon this, with a three-way strategic partnership (the "Agreement") exploring collaboration on domestic US processing solutions ...