International Journal of Pharmacy and Chemistry

| Peer-Reviewed |

Impact Factors in the Process Development for Therapeutic Antibodies

Received: 4 July 2018    Accepted: 26 July 2018    Published: 16 August 2018
Views:       Downloads:

Share This Article

Abstract

The research and development of therapeutic antibodies in the world is rapidly rising in recent years. It is a huge challenge to obtain a robust process and achieve the consistency of consistent product quality, because most of the antibody drugs are produced from mammalian cells of which the production processes are long and complex. To develop a robust and high yield production process for a biotherapeutic, one needs to have a general consideration of entire R&D project working flow. First of all, it is necessary to start with a good drug candidate, which not only meets the requirement for both the effectiveness and drug safety, but also suits for manufacture. Then, the expression system and host cell line used for the product expression need to be determined based on availability and regulatory requirements. Cell line development needs to combine with early upstream and downstream process developments as well as product quality assessment. Once a suitable stable cell line is selected, the Master Cell Bank (MCB) and the Working Cell Bank (WCB) are established according to the regulatory requirements, and tested for growth, production stability and cellular safety. Finally, process optimization and scale-up production are carried out to obtain a consistent production. Upstream process development is mainly to optimize a variety of process parameters, including physical parameters, chemical parameters, metabolite levels, and feed strategy and foam control; downstream process development focuses on product quality, recovery rate, and cost efficiency. For scale-up production, enterprises need to make decisions whether to use traditional stainless steel system or the single-use disposable system based on demands on product market potential and available technical supports. In this review, combined with the development of new technologies in recent years and the authors’ personal working experience, the factors that need to be considered in process development are discussed.

DOI 10.11648/j.ijpc.20180402.11
Published in International Journal of Pharmacy and Chemistry (Volume 4, Issue 2, March 2018)
Page(s) 16-22
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Therapeutic Antibody, Process Development, Quality Control

References
[1] Buss N, and Henderson S, “Monoclonal antibody therapeutics: history and future,” Current Opinion in Pharmacology. [J]. 2012, vol. 12, pp. 615–622.
[2] Dawn M Ecker, Susan Dana Jones, and Howard L Levine, “The therapeutic monoclonal antibody market,” mAbs 7:1, 9--14; 2015 BioProcess Technology Consultants, Inc.
[3] Shi H, Chen M, and Shi Y, “Challenges and prospects for monoclonal antibodies in China,” Journal of Commercial Biotechnology. [J]. 2013, vol. 19, pp. 48–54.
[4] Morrow Jr. KJ, “Advances in antibody manufacturing using mammalian cells,” Biotechnology Annual Review [J]. 2007, vol. 13, pp. 95–113.
[5] Nelson AL, Dhimolea E, and Reichert JM, “Development trends for human monoclonal antibody therapeutics,” Nature Reviews Drug Discovery [J]. 2010 vol. 9, no. 10, pp. 767–774.
[6] Kozlowski S, and Swann P, “Current and future issues in the manufacturing and development of monoclonal antibodies,” Advanced Drug Delivery Reviews [J]. 2006, vol. 58, no. 5-6, pp. 707-722.
[7] Jakobovits A, “Production of fully human antibodies by transgenic mice,” Current Opinion in Biotechnology [J]. 1995, vol. 6, no. 5, pp. 561–566.
[8] Chen Z, and Jiang J, “Monoclonal antibody—from murine to humanized,” Foreign Medical Science [J]. 2000, vol. 23, pp. 188–189.
[9] K. Knight, et al, “B lymphocyte development in the rabbit,” Int. Rev. Immunol. [J]. 1997, 15: 129–163.
[10] Iwamoto S, Nishimichi Y, Tateishi Y, Sato Y, Horiuchi H, Furusawa S, Sawamura T, and Matsuda H, “Generation and characterization of chicken monoclonal antibodies against human LOX-1,” [J]. MAbs 2009, Jul-Aug; 1(4): 357–363.
[11] Tiller T. “Single B cell antibody technologies,” N Biotechnol. [J]. 2011 Sep; 28(5):453-7.
[12] Liao C. “Early development of antibody therapeutics: key aspects of advancing an antibody drug from research to clinic,” Trends in Bio/Pharmaceutical Industry [J]. 2009, vol. 5, pp.18–23.
[13] Lowe D, Dudgeon K, Rouet R, Schofield P, Jermutus L, ChristD. “Aggregation, stability, and formulation of human antibody therapeutics; Advances in Protein Chemistry and Structural Biology” [J]. 2011, Volume 84, Pages 41–61.
[14] Shire SJ, Shahrokh Z, Liu Z, “Challenges in the development of high protein concentration formulations,” Current Trends in Monoclonal Antibody Development and Manufacturing. New York: Springer [J].2010, p131-147.
[15] Fan L, Kadura I, Krebs LE, Hatfield CC, Shaw MM, Frye CC, “Improving the efficiency of CHO cell line generation using glutamine synthetase gene knockout cells,” Biotechnol Bioeng. [J]. 2012, Apr; 109(4):1007-15.
[16] Kennard ML, Goosney DL, Monteith D, Zhang L, Moffat M, Fischer D, Mott JE, “The generation of stable, high MAb expressing CHO cell lines based on the artificial chromosome expression (ACE) technology,” Biotechnol. Bioeng [J]. 2009, 104: 540-553.
[17] Chen C, “Challenges and opportunities of monoclonal antibody manufacturing in China,” Trends in Bio/Pharmaceutical Industry [J]. 2009 vol. 5, pp. 28–33.
[18] Ju D, “Development of antibody drugs in China,” Trends in Bio/Pharmaceutical Industry [J]. 2009, vol. 5, pp. 34–39.
[19] Hartley J L, “Why proteins in mammalian cells?” [J]. Methods Mol Biol, 2012, 801:1-12.
[20] Backliwal G, Hildinger M, Chenuet S, Wulhfard S, De Jesus and Wurm F M, “Rational vector design and multi-pathway modulation of HEK 293E cells yield recombinant antibody titers exceeding 1 g/l by transient transfection under serum-free conditions,” Nucleic Acids Research, [J] 2008, 1–7
[21] Awotwe-Otoo D, Agarabi C, Wu GK, Casey E, Read E, Lute S, Brorson KA, Khan MA, Shah RB, “Quality by design: Impact of formulation variables and their interactions on quality attributes of a lyophilized monoclonal antibody,” Int J Pharmaceutics [J]. 2012, 438: 167-175.
[22] Higbee J and Tu H, “Biosimilars: challenges and promises cabs workshop on biosimilar therapeutics,” Trends in Bio/Pharmaceutical Industry, [J]. 2009, vol. 5, pp. 42–44.
[23] Beck A, and Reichert JM, “Approval of the first biosimilar antibodies in Europe: a major landmark for the biopharmaceutical industry,” mAbs [J]. 2013, vol. 5, no. 5, pp. 621–623.
[24] Schaub J, Clemens C, Schorn P, et al, “CHO gene expression profiling in biopharmaceutical process analysis and design,” [J]. 2010 Feb 1; 105(2):431-8.
[25] Agarabi C. D., Schiel J. E, Lute S. C, et al, “Bioreactor Process Parameter Screening Utilizing a Plackett–BurmanDesign for a Model Monoclonal Antibody,” Journal of Pharmaceutical Sciences [J]. 2015104:1919–1928, 2015.
[26] Kishishita S, Nishikawa T, Shinoda Y, Nagashima H and Aoyag H, “Effect of temperature shift on levels of acidic charge variants in IgG monoclonal antibodies in Chinese hamster ovary cell culture Journal of Bioscience and Bioengineering,” [J] 2015, VOL. 119 No. 6, 700-705.
[27] Ishii Y, Murakami J, Sasaki K, Tsukahara M, and Wakamatsu K, “Efficient folding/assembly in Chinese hamster ovary cells is critical for high quality (low aggregate content) of secreted trastuzumab as well as for high production: Stepwise multivariate regression analyses,” Journal of Bioscience and Bioengineering [J] 2014, VOL. 118 No. 2, 223-230.
[28] Davami F, Eghbalpour F, Nematollahi L, Barkhordari F, Mahboudi F, “Effects of Peptone Supplementation in Different Culture Media on Growth, Metabolic Pathway and Productivity of CHO DG44 Cells; a New Insight into Amino Acid Profiles,” Iran Biomed J. [J]. 2015, 19(4):194-205.
[29] Almo SC, Love JD, “Better and faster: improvements and optimization for mammalian recombinant protein production,” [J]. 2014, 26: 39-43.
[30] Ozturk SS, “Equipment for large-scale mammalian cell culture,” Adv Biochem Engng-Biotech [J]. 2014, 139: 69-92.
[31] Eibl R, Löffelholz C, Eibl D, “Disposable bioreactors for inoculum production and protein expression,” Methods Mol Biol. [J]. 2014, 1104:265-84.
[32] Hammond M, Nunn H, Rogers G, et al, “Identification of a leachable compound detrimental to cell growth in single-use bioprocess containers,” PDA J Pharm Sci Technol. [J]. 2013, Mar-Apr; 67(2):123-34.
[33] Li F, Vijayasankaran N, Shen A, Kiss R and Amanullah A, “Cell culture processes for monoclonal antibody production,” mAbs [J] 2010, 2:5, 466-477.
[34] Angarita M, Müller-Späth T, Baur D, Lievrouw R, Lissens G, Morbidelli M, “Twincolumn Capture SMB: A novel cyclic process for protein A affinity chromatography,” J Chromatogr [J]. 2015, A 1389: 85-95.
[35] Bae JE, Jeong EK, Lee JI, Lee JI, Kim IS, Kum JS, “Evaluation of viral inactivation efficacy of a continuous flow ultraviolet-C reactor (UVivatec),” Kor J Microbiol Biotechnol [J]. 2009, 4: 377-382.
[36] Weaver J, Husson SM, Murphy L, Wickramasinghe SR, “Anion exchange membrane adsorbers for flow-through polishing steps: Part II. Virus, host cell protein, DNA clearance, and antibody recovery,” Biotechnol Bioeng [J]. 2013, 110: 500-510.
[37] Zheng K, Bantog C, and Bayer R, “The impact of glycosylation on monoclonal antibody conformation and stability,” MAbs, [J]. 2011, Nov-Dec; 3(6): 568–576.
[38] Pacis E, Yu M, Autsen J, Bayer R, Li F, “Effects of cell culture conditions on antibody Nlinked glycosylation - What affects high mannose 5 glycoform,” Biotechnol Bioeng [J]. 2011, 108:2348-2358.
[39] Joshi V, Shivach T, Kumar V, Yadav N, Rathore A, “Avoiding antibody aggregation during processing: Establishing hold times” Biotechnol J [J]. 2014, 9: 1195-1205.
[40] Chames P, Baty D, “Bispecific antibodies for cancer therapy: The light at the end of the tunnel?” mAbs [J]. 2009, 1:6, 539-547.
[41] Schaefera I, Regulaa JT, Bähnera M, et al, “Immunoglobulin domain crossover as a generic approach for the production of bispecific IgG antibodies,” [J]. Proc Natl Acad Sci USA, 2011, 108(27): 11187-11192.
[42] Asher M. "Maturing antibody–drug conjugate pipeline hits 30," Nature Reviews Drug Discovery [J]. 2013, 12 (5): 329–32.
[43] Shinkazh O, Kanani D, Barth M, Long M, Hussain D, Zydney AL, “Countercurrent tangential chromatography for large-scale protein purification,” Biotechnol Bioeng [J]. 2011, 108:582-591, 2011.
[44] Jungbauer A, “Continuous downstream processing of biopharmaceuticals,” Trends Biotechnol [J]. 2013, 31: 479-492, 2013.
[45] Gagnon P, “Monoclonal antibody purification with hydroxyapatite,” New Biotechnology, [J]. 2009, vol. 25, no. 5, pp. 287–293.
[46] Hammerschmidt N, Tscheliessnig A, Sommer R, Helk B, Jungbauer A, “Economics of recombinant antibody production processes at various scales: Industry-standard compared to continuous precipitation,” Biotechnol [J]. 2014, J 9: 766-775, 2014.
Cite This Article
  • APA Style

    Xiaopei Cui, Xiangyang Zhu. (2018). Impact Factors in the Process Development for Therapeutic Antibodies. International Journal of Pharmacy and Chemistry, 4(2), 16-22. https://doi.org/10.11648/j.ijpc.20180402.11

    Copy | Download

    ACS Style

    Xiaopei Cui; Xiangyang Zhu. Impact Factors in the Process Development for Therapeutic Antibodies. Int. J. Pharm. Chem. 2018, 4(2), 16-22. doi: 10.11648/j.ijpc.20180402.11

    Copy | Download

    AMA Style

    Xiaopei Cui, Xiangyang Zhu. Impact Factors in the Process Development for Therapeutic Antibodies. Int J Pharm Chem. 2018;4(2):16-22. doi: 10.11648/j.ijpc.20180402.11

    Copy | Download

  • @article{10.11648/j.ijpc.20180402.11,
      author = {Xiaopei Cui and Xiangyang Zhu},
      title = {Impact Factors in the Process Development for Therapeutic Antibodies},
      journal = {International Journal of Pharmacy and Chemistry},
      volume = {4},
      number = {2},
      pages = {16-22},
      doi = {10.11648/j.ijpc.20180402.11},
      url = {https://doi.org/10.11648/j.ijpc.20180402.11},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.ijpc.20180402.11},
      abstract = {The research and development of therapeutic antibodies in the world is rapidly rising in recent years. It is a huge challenge to obtain a robust process and achieve the consistency of consistent product quality, because most of the antibody drugs are produced from mammalian cells of which the production processes are long and complex. To develop a robust and high yield production process for a biotherapeutic, one needs to have a general consideration of entire R&D project working flow. First of all, it is necessary to start with a good drug candidate, which not only meets the requirement for both the effectiveness and drug safety, but also suits for manufacture. Then, the expression system and host cell line used for the product expression need to be determined based on availability and regulatory requirements. Cell line development needs to combine with early upstream and downstream process developments as well as product quality assessment. Once a suitable stable cell line is selected, the Master Cell Bank (MCB) and the Working Cell Bank (WCB) are established according to the regulatory requirements, and tested for growth, production stability and cellular safety. Finally, process optimization and scale-up production are carried out to obtain a consistent production. Upstream process development is mainly to optimize a variety of process parameters, including physical parameters, chemical parameters, metabolite levels, and feed strategy and foam control; downstream process development focuses on product quality, recovery rate, and cost efficiency. For scale-up production, enterprises need to make decisions whether to use traditional stainless steel system or the single-use disposable system based on demands on product market potential and available technical supports. In this review, combined with the development of new technologies in recent years and the authors’ personal working experience, the factors that need to be considered in process development are discussed.},
     year = {2018}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Impact Factors in the Process Development for Therapeutic Antibodies
    AU  - Xiaopei Cui
    AU  - Xiangyang Zhu
    Y1  - 2018/08/16
    PY  - 2018
    N1  - https://doi.org/10.11648/j.ijpc.20180402.11
    DO  - 10.11648/j.ijpc.20180402.11
    T2  - International Journal of Pharmacy and Chemistry
    JF  - International Journal of Pharmacy and Chemistry
    JO  - International Journal of Pharmacy and Chemistry
    SP  - 16
    EP  - 22
    PB  - Science Publishing Group
    SN  - 2575-5749
    UR  - https://doi.org/10.11648/j.ijpc.20180402.11
    AB  - The research and development of therapeutic antibodies in the world is rapidly rising in recent years. It is a huge challenge to obtain a robust process and achieve the consistency of consistent product quality, because most of the antibody drugs are produced from mammalian cells of which the production processes are long and complex. To develop a robust and high yield production process for a biotherapeutic, one needs to have a general consideration of entire R&D project working flow. First of all, it is necessary to start with a good drug candidate, which not only meets the requirement for both the effectiveness and drug safety, but also suits for manufacture. Then, the expression system and host cell line used for the product expression need to be determined based on availability and regulatory requirements. Cell line development needs to combine with early upstream and downstream process developments as well as product quality assessment. Once a suitable stable cell line is selected, the Master Cell Bank (MCB) and the Working Cell Bank (WCB) are established according to the regulatory requirements, and tested for growth, production stability and cellular safety. Finally, process optimization and scale-up production are carried out to obtain a consistent production. Upstream process development is mainly to optimize a variety of process parameters, including physical parameters, chemical parameters, metabolite levels, and feed strategy and foam control; downstream process development focuses on product quality, recovery rate, and cost efficiency. For scale-up production, enterprises need to make decisions whether to use traditional stainless steel system or the single-use disposable system based on demands on product market potential and available technical supports. In this review, combined with the development of new technologies in recent years and the authors’ personal working experience, the factors that need to be considered in process development are discussed.
    VL  - 4
    IS  - 2
    ER  - 

    Copy | Download

Author Information
  • Department of Process Development, Huabo Biopharma, Shanghai, China

  • Department of Process Development, Huabo Biopharma, Shanghai, China

  • Sections