1
|
Mlakar V, Lesne L, Vossio S, Dupanloup I, Gloor Y, Moreau D, Ansari M. Microcavity-assisted cloning (MAC) of hard-to-clone HepG2 cell lines: cloning made easy. BMC Biotechnol 2024; 24:81. [PMID: 39407195 PMCID: PMC11481743 DOI: 10.1186/s12896-024-00911-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
Cloning is a key molecular biology procedure for obtaining a genetically homogenous population of organisms or cell lines. It requires the expansion of new cell populations starting from single genetically modified cells. Despite the technical progress, cloning of many cell lines remains difficult. Cloning often fails either due to the strenuous conditions associated with manipulating cells or because many cells don't tolerate a single-cell state. Here we describe a new cloning method utilizing low adhesion microcavity plates. This new technique, named microcavity-assisted cloning (MAC) was developed to clone difficult-to-clone HepG2 cells. The clones were produced following CRISPR/Cas9 knockout of the GSTA1 gene by a random distribution of 200, 400, and 800 cells into 550 microcavities of a 24-well low adhesion plate originally designed for the culture of spheroids. The knockout of GSTA1 was verified at the protein level using Western blotting. The advantages of the MAC method are its low cost, ease of the procedure, and the possibility of scaling up the throughput and automatization.
Collapse
Affiliation(s)
- Vid Mlakar
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| | - Laurence Lesne
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stefania Vossio
- University of Geneva, School of Chemistry and Biochemistry - Sciences II, ACCESS Geneva, Quai Ernest Ansermet 30, 1211, Geneva 4, Switzerland
| | - Isabelle Dupanloup
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Yvonne Gloor
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Dimitri Moreau
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Marc Ansari
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
| |
Collapse
|
2
|
Zeh N, Schmidt M, Schulz P, Fischer S. The new frontier in CHO cell line development: From random to targeted transgene integration technologies. Biotechnol Adv 2024; 75:108402. [PMID: 38950872 DOI: 10.1016/j.biotechadv.2024.108402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/03/2024]
Abstract
Cell line development represents a crucial step in the development process of a therapeutic glycoprotein. Chinese hamster ovary (CHO) cells are the most frequently employed mammalian host cell system for the industrial manufacturing of biologics. The predominant application of CHO cells for heterologous recombinant protein expression lies in the relative simplicity of stably introducing ectopic DNA into the CHO host cell genome. Since CHO cells were first used as expression host for the industrial production of biologics in the late 1980s, stable genomic transgene integration has been achieved almost exclusively by random integration. Since then, random transgene integration had become the gold standard for generating stable CHO production cell lines due to a lack of viable alternatives. However, it was eventually demonstrated that this approach poses significant challenges on the cell line development process such as an increased risk of inducing cell line instability. In recent years, significant discoveries of new and highly potent (semi)-targeted transgene integration systems have paved the way for a technological revolution in the cell line development sector. These advanced methodologies comprise the application of transposase-, recombinase- or Cas9 nuclease-mediated site-specific genomic integration techniques, which enable a scarless transfer of the transgene expression cassette into transcriptionally active loci within the host cell genome. This review summarizes recent advancements in the field of transgene integration technologies for CHO cell line development and compare them to the established random integration approach. Moreover, advantages and limitations of (semi)-targeted integration techniques are discussed, and benefits and opportunities for the biopharmaceutical industry are outlined.
Collapse
Affiliation(s)
- Nikolas Zeh
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH and Co.KG, Biberach an der Riss, Germany
| | - Moritz Schmidt
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH and Co.KG, Biberach an der Riss, Germany
| | - Patrick Schulz
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH and Co.KG, Biberach an der Riss, Germany
| | - Simon Fischer
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH and Co.KG, Biberach an der Riss, Germany.
| |
Collapse
|
3
|
Schardt JS, Sivaneri NS, Tessier PM. Monoclonal Antibody Generation Using Single B Cell Screening for Treating Infectious Diseases. BioDrugs 2024; 38:477-486. [PMID: 38954386 PMCID: PMC11645890 DOI: 10.1007/s40259-024-00667-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/04/2024]
Abstract
The screening of antigen-specific B cells has been pivotal for biotherapeutic development for over four decades. Conventional antibody discovery strategies, including hybridoma technology and single B cell screening, remain widely used based on their simplicity, accessibility, and proven track record. Technological advances and the urgent demand for infectious disease applications have shifted paradigms in single B cell screening, resulting in increased throughput and decreased time and labor, ultimately enabling the rapid identification of monoclonal antibodies with desired biological and biophysical properties. Herein, we provide an overview of conventional and emergent single B cell screening approaches and highlight their potential strengths and weaknesses. We also detail the impact of innovative technologies-including miniaturization, microfluidics, multiplexing, and deep sequencing-on the recent identification of broadly neutralizing antibodies for infectious disease applications. Overall, the coronavirus disease 2019 (COVID-19) pandemic has reinvigorated efforts to improve the efficiency of monoclonal antibody discovery, resulting in the broad application of innovative antibody discovery methodologies for treating a myriad of infectious diseases and pathological conditions.
Collapse
Affiliation(s)
- John S Schardt
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Neelan S Sivaneri
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Peter M Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
4
|
Masuda K, Kubota M, Nakazawa Y, Iwama C, Watanabe K, Ishikawa N, Tanabe Y, Kono S, Tanemura H, Takahashi S, Makino T, Okumura T, Horiuchi T, Nonaka K, Murakami S, Kamihira M, Omasa T. Establishment of a novel cell line, CHO-MK, derived from Chinese hamster ovary tissues for biologics manufacturing. J Biosci Bioeng 2024; 137:471-479. [PMID: 38472071 DOI: 10.1016/j.jbiosc.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/04/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024]
Abstract
Chinese hamster ovary (CHO) cells are widely used as a host for producing recombinant therapeutic proteins due to advantages such as human-like post-translational modification, correct protein folding, higher productivity, and a proven track record in biopharmaceutical development. Much effort has been made to improve the process of recombinant protein production, in terms of its yield and productivity, using conventional CHO cell lines. However, to the best of our knowledge, no attempts have been made to acquire new CHO cell lines from Chinese hamster ovary. In this study, we established and characterized a novel CHO cell line, named CHO-MK, derived from freshly isolated Chinese hamster ovary tissues. Some immortalized cell lines were established via sub-culture derived from primary culture, one of which was selected for further development toward a unique expression system design. After adapting serum-free and suspension culture conditions, the resulting cell line exhibited a considerably shorter doubling time (approximately 10 h) than conventional CHO cell lines (approximately 20 h). Model monoclonal antibody (IgG1)-producing cells were generated, and the IgG1 concentration of fed-batch culture reached approximately 5 g/L on day 8 in a 200-L bioreactor. The cell bank of CHO-MK cells was prepared as a new host and assessed for contamination by adventitious agents, with the results indicating that it was free from any such contaminants, including infectious viruses. Taking these findings together, this study showed the potential of CHO-MK cells with a shorter doubling time/process time and enhanced productivity in biologics manufacturing.
Collapse
Affiliation(s)
- Kenji Masuda
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan; Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Michi Kubota
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Chitose Laboratory Corp., KSP EAST511, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Yuto Nakazawa
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Chigusa Iwama
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Chitose Laboratory Corp., KSP EAST511, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Kazuhiko Watanabe
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Naoto Ishikawa
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Yumiko Tanabe
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Satoru Kono
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Hiroki Tanemura
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Shinichi Takahashi
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Tomohiro Makino
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Takeshi Okumura
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan; Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Takayuki Horiuchi
- Chitose Laboratory Corp., KSP EAST511, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Koichi Nonaka
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan; Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Sei Murakami
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Masamichi Kamihira
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takeshi Omasa
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
5
|
Desmurget C, Frentzel J, Strembitska A, Sobkowiak K, Perilleux A, Souquet J, Borth N, Douet J. Combined approach of selective and accelerated cloning for microfluidic chip-based system increases clone specific productivity. Biotechnol J 2024; 19:e2300488. [PMID: 38803036 DOI: 10.1002/biot.202300488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
Improving current cell line development workflows can either focus on increasing the specific productivity of the cell lines or shortening timelines to reach the clinic as fast as possible. In this work, using the Beacon platform, we have combined two distinct protocols - early cloning with low-viability pools, and IgG membrane staining-, to concomitantly reach both objectives, and generate highly productive CHO clones in shorter timelines. Fast-sorting approaches using low-viability pools in combination with the Beacon platform have recently been reported to shorten CLD timelines. However, the low recovery led to a drastic reduction in the clone number obtained postcloning. Here, we report a combined approach of fast-sorting and fluorescent membrane staining. With this new protocol, the cells reach a correct recovery, allowing to fully exploit the Beacon screening capacities. In addition, by using a fluorescent staining recognizing the secreted IgG, we were able to enrich the fraction of highly secreting cells prior to cloning and we obtained significant increases in the cell's specific productivity. The combination of these two protocols has a synergistic effect, and as they help discarding the dead and nonproducing populations prior to cloning, they increase the throughput power of the Beacon platform and the detection of super productive clones.
Collapse
Affiliation(s)
- Caroline Desmurget
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Julie Frentzel
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Anastasiya Strembitska
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Katarzyna Sobkowiak
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Arnaud Perilleux
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Jonathan Souquet
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Julien Douet
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| |
Collapse
|
6
|
Abali F, Schasfoort R, Nijland S, Wittenberns J, Tibbe AGJ, den Hartog M, Boon L, Terstappen LWMM. A nanowell platform to identify, sort and expand high antibody-producing cells. Sci Rep 2024; 14:9457. [PMID: 38658627 PMCID: PMC11043069 DOI: 10.1038/s41598-024-60054-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 04/18/2024] [Indexed: 04/26/2024] Open
Abstract
Increased use of therapeutic monoclonal antibodies and the relatively high manufacturing costs fuel the need for more efficient production methods. Here we introduce a novel, fast, robust, and safe isolation platform for screening and isolating antibody-producing cell lines using a nanowell chip and an innovative single-cell isolation method. An anti-Her2 antibody producing CHO cell pool was used as a model. The platform; (1) Assures the single-cell origin of the production clone, (2) Detects the antibody production of individual cells and (3) Isolates and expands the individual cells based on their antibody production. Using the nanowell platform we demonstrated an 1.8-4.5 increase in anti-Her2 production by CHO cells that were screened and isolated with the nanowell platform compared to CHO cells that were not screened. This increase was also shown in Fed-Batch cultures where selected high production clones showed titers of 19-100 mg/L on harvest day, while the low producer cells did not show any detectable anti-Her2 IgG production. The screening of thousands of single cells is performed under sterile conditions and the individual cells were cultured in buffers and reagents without animal components. The time required from seeding a single cell and measuring the antibody production to fully expanded clones with increased Her-2 production was 4-6 weeks.
Collapse
Affiliation(s)
- Fikri Abali
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, University of Twente, PO Box 217, 7500AE, Enschede, The Netherlands
| | - Richard Schasfoort
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, University of Twente, PO Box 217, 7500AE, Enschede, The Netherlands
| | - Sanne Nijland
- VYCAP, Capitool 41, 7521PL, Enschede, The Netherlands
| | - Jelle Wittenberns
- Polpharma Biologics Utrecht B.V., Yalelaan 46, 3584 CM, Utrecht, The Netherlands
| | | | - Marcel den Hartog
- Polpharma Biologics Utrecht B.V., Yalelaan 46, 3584 CM, Utrecht, The Netherlands
| | - Louis Boon
- JJP Biologics, Bobrowiecka 6, 00-728, Warsaw, Poland
| | - Leon W M M Terstappen
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, University of Twente, PO Box 217, 7500AE, Enschede, The Netherlands.
- Department of General, Visceral and Pediatric Surgery, Heinrich-Heine University, University Hospital Düsseldorf, 40225, Düsseldorf, Germany.
| |
Collapse
|
7
|
Slagboom J, Lewis AH, Schouten WM, van Haperen R, Veltman M, Bittenbinder MA, Vonk FJ, Casewell NR, Grosveld F, Drabek D, Kool J. High throughput identification of human monoclonal antibodies and heavy-chain-only antibodies to treat snakebite. Toxicon X 2024; 21:100185. [PMID: 38425752 PMCID: PMC10901844 DOI: 10.1016/j.toxcx.2024.100185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/12/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Snakebite envenoming is a priority Neglected Tropical Disease that causes an estimated 81,000-135,000 fatalities each year. The development of a new generation of safer, affordable, and accessible antivenom therapies is urgently needed. With this goal in mind, rigorous characterisation of the specific toxins in snake venom is key to generating novel therapies for snakebite. Monoclonal antibodies directed against venom toxins are emerging as potentially strong candidates in the development of new snakebite diagnostics and treatment. Venoms comprise many different toxins of which several are responsible for their pathological effects. Due to the large variability of venoms within and between species, formulations of combinations of human antibodies are proposed as the next generation antivenoms. Here a high-throughput screening method employing antibody-based ligand fishing of venom toxins in 384 filter-well plate format has been developed to determine the antibody target/s The approach uses Protein G beads for antibody capture followed by exposure to a full venom or purified toxins to bind their respective ligand toxin(s). This is followed by a washing/centrifugation step to remove non-binding toxins and an in-well tryptic digest. Finally, peptides from each well are analysed by nanoLC-MS/MS and subsequent Mascot database searching to identify the bound toxin/s for each antibody under investigation. The approach was successfully validated to rapidly screen antibodies sourced from hybridomas, derived from venom-immunised mice expressing either regular human antibodies or heavy-chain-only human antibodies (HCAbs).
Collapse
Affiliation(s)
- Julien Slagboom
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
| | - Abigail H. Lewis
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
| | - Wietse M. Schouten
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
| | - Rien van Haperen
- Department of Cell Biology and Genetics, Faculty of Medicine, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
- Harbour BioMed, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
| | - Mieke Veltman
- Department of Cell Biology and Genetics, Faculty of Medicine, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
- Harbour BioMed, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
| | - Mátyás A. Bittenbinder
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
- Naturalis Biodiversity Center, 2333 CR, Leiden, the Netherlands
| | - Freek J. Vonk
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
- Naturalis Biodiversity Center, 2333 CR, Leiden, the Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Frank Grosveld
- Department of Cell Biology and Genetics, Faculty of Medicine, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
- Harbour BioMed, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
| | - Dubravka Drabek
- Department of Cell Biology and Genetics, Faculty of Medicine, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
- Harbour BioMed, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
| | - Jeroen Kool
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
| |
Collapse
|
8
|
Colina AS, Shah V, Shah RK, Kozlik T, Dash RK, Terhune S, Zamora AE. Current advances in experimental and computational approaches to enhance CAR T cell manufacturing protocols and improve clinical efficacy. FRONTIERS IN MOLECULAR MEDICINE 2024; 4:1310002. [PMID: 39086435 PMCID: PMC11285593 DOI: 10.3389/fmmed.2024.1310002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/08/2024] [Indexed: 08/02/2024]
Abstract
Since the FDA's approval of chimeric antigen receptor (CAR) T cells in 2017, significant improvements have been made in the design of chimeric antigen receptor constructs and in the manufacturing of CAR T cell therapies resulting in increased in vivo CAR T cell persistence and improved clinical outcome in certain hematological malignancies. Despite the remarkable clinical response seen in some patients, challenges remain in achieving durable long-term tumor-free survival, reducing therapy associated malignancies and toxicities, and expanding on the types of cancers that can be treated with this therapeutic modality. Careful analysis of the biological factors demarcating efficacious from suboptimal CAR T cell responses will be of paramount importance to address these shortcomings. With the ever-expanding toolbox of experimental approaches, single-cell technologies, and computational resources, there is renowned interest in discovering new ways to streamline the development and validation of new CAR T cell products. Better and more accurate prognostic and predictive models can be developed to help guide and inform clinical decision making by incorporating these approaches into translational and clinical workflows. In this review, we provide a brief overview of recent advancements in CAR T cell manufacturing and describe the strategies used to selectively expand specific phenotypic subsets. Additionally, we review experimental approaches to assess CAR T cell functionality and summarize current in silico methods which have the potential to improve CAR T cell manufacturing and predict clinical outcomes.
Collapse
Affiliation(s)
- Alfredo S. Colina
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Viren Shah
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, United States
| | - Ravi K. Shah
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tanya Kozlik
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ranjan K. Dash
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, United States
| | - Scott Terhune
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, United States
| | - Anthony E. Zamora
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
9
|
Grob A, Enrico Bena C, Di Blasi R, Pessina D, Sood M, Yunyue Z, Bosia C, Isalan M, Ceroni F. Mammalian cell growth characterisation by a non-invasive plate reader assay. Nat Commun 2024; 15:57. [PMID: 38167870 PMCID: PMC10761699 DOI: 10.1038/s41467-023-44396-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Automated and non-invasive mammalian cell analysis is currently lagging behind due to a lack of methods suitable for a variety of cell lines and applications. Here, we report the development of a high throughput non-invasive method for tracking mammalian cell growth and performance based on plate reader measurements. We show the method to be suitable for both suspension and adhesion cell lines, and we demonstrate it can be adopted when cells are grown under different environmental conditions. We establish that the method is suitable to inform on effective drug treatments to be used depending on the cell line considered, and that it can support characterisation of engineered mammalian cells over time. This work provides the scientific community with an innovative approach to mammalian cell screening, also contributing to the current efforts towards high throughput and automated mammalian cell engineering.
Collapse
Affiliation(s)
- Alice Grob
- Department of Chemical Engineering, Imperial College London, London, UK
- Imperial College Centre for Synthetic Biology, Imperial College London, London, UK
| | - Chiara Enrico Bena
- Italian Institute for Genomic Medicine, Torino, Italy
- Université Paris-Saclay (INRAE), AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Roberto Di Blasi
- Department of Chemical Engineering, Imperial College London, London, UK
- Imperial College Centre for Synthetic Biology, Imperial College London, London, UK
| | - Daniele Pessina
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Matthew Sood
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Zhou Yunyue
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Carla Bosia
- Italian Institute for Genomic Medicine, Torino, Italy.
- Department of Applied Science and Technology, Politecnico di Torino, Torino, Italy.
| | - Mark Isalan
- Imperial College Centre for Synthetic Biology, Imperial College London, London, UK.
- Department of Life Sciences, Imperial College London, London, United Kingdom.
| | - Francesca Ceroni
- Department of Chemical Engineering, Imperial College London, London, UK.
- Imperial College Centre for Synthetic Biology, Imperial College London, London, UK.
| |
Collapse
|
10
|
Park JH, Heo NY, Lee HM, Lee EJ, Park S, Lee GM, Kim YG. Streamlined in vitro screening system of synthetic signal peptides in Chinese hamster ovary cells for therapeutic protein production. J Biotechnol 2023; 375:12-16. [PMID: 37634828 DOI: 10.1016/j.jbiotec.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Increasing the screening efficiency and maintaining the N-terminal cleavage pattern are key factors in the development of an in vitro synthetic signal peptide screening system for high therapeutic protein production in Chinese hamster ovary (CHO) cells. This study improved the in vitro screening system of synthetic signal peptides in CHO cells for therapeutic protein production by modifying the expression vector. Incorporating a leaky stop codon with IgG transmembrane and cytoplasmic domains into the expression vector improved the proportion of high producers in establishing stable CHO cell pools. The selected signal peptides from stable CHO cell pools that were generated using degenerate codon-based oligonucleotides with a conserved polar carboxy-terminal domain in the native signal peptide showed similar N-terminal cleavage patterns to the native one. In addition, replacing native signal peptide with selected synthetic signal peptides did not influence the sialylated N-linked glycan formation and biological activity of therapeutic Fc-fusion glycoprotein in CHO cells. Thus, an in vitro synthetic signal peptide screening system can be used for therapeutic Fc-fusion glycoprotein production in CHO cells with an enhanced specific protein productivity while maintaining the N-terminal cleavage pattern similar to the native one.
Collapse
Affiliation(s)
- Jong-Ho Park
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea; Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Na-Yeong Heo
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hoon-Min Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Eun-Ji Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Soomin Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea.
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea.
| |
Collapse
|
11
|
Hu S, Ye J, Shi S, Yang C, Jin K, Hu C, Wang D, Ma H. Large-Area Electronics-Enabled High-Resolution Digital Microfluidics for Parallel Single-Cell Manipulation. Anal Chem 2023; 95:6905-6914. [PMID: 37071892 DOI: 10.1021/acs.analchem.3c00150] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Large-area electronics as switching elements are an ideal option for electrode-array-based digital microfluidics. With support of highly scalable thin-film semiconductor technology, high-resolution digital droplets (diameter around 100 μm) containing single-cell samples can be manipulated freely on a two-dimensional plane with programmable addressing logic. In addition, single-cell generation and manipulation as foundations for single-cell research demand ease of operation, multifunctionality, and accurate tools. In this work, we reported an active-matrix digital microfluidic platform for single-cell generation and manipulation. The active device contained 26,368 electrodes that could be independently addressed to perform parallel and simultaneous droplet generation and achieved single-cell manipulation. We demonstrate a high-resolution digital droplet generation with a droplet volume limit of 500 pL and show the continuous and stable movement of droplet-contained cells for over 1 h. Furthermore, the success rate of single droplet formation was higher than 98%, generating tens of single cells within 10 s. In addition, a pristine single-cell generation rate of 29% was achieved without further selection procedures, and the droplets containing single cells could then be tested for on-chip cell culturing. After 20 h of culturing, about 12.5% of the single cells showed cell proliferation.
Collapse
Affiliation(s)
- Siyi Hu
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
| | - Jingmin Ye
- Guangdong ACXEL Micro & Nano Tech Co., Ltd, Foshan, Guangdong Province 528000, P. R. China
| | - Subao Shi
- Guangdong ACXEL Micro & Nano Tech Co., Ltd, Foshan, Guangdong Province 528000, P. R. China
| | - Chao Yang
- Guangdong ACXEL Micro & Nano Tech Co., Ltd, Foshan, Guangdong Province 528000, P. R. China
| | - Kai Jin
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
| | - Chenxuan Hu
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
| | - Dongping Wang
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
| | - Hanbin Ma
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
- Guangdong ACXEL Micro & Nano Tech Co., Ltd, Foshan, Guangdong Province 528000, P. R. China
| |
Collapse
|
12
|
Hou Y, Wang H, Fu R, Wang X, Yu J, Zhang S, Huang Q, Sun Y, Fukuda T. A review on microrobots driven by optical and magnetic fields. LAB ON A CHIP 2023; 23:848-868. [PMID: 36629004 DOI: 10.1039/d2lc00573e] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Due to their small sizes, microrobots are advantageous for accessing hard-to-reach spaces for delivery and measurement. However, their small sizes also bring challenges in on-board powering, thus usually requiring actuation by external energy. Microrobots actuated by external energy have been applied to the fields of physics, biology, medical science, and engineering. Among these actuation sources, light and magnetic fields show advantages in high precision and high biocompatibility. This paper reviews the recent advances in the design, actuation, and applications of microrobots driven by light and magnetic fields. For light-driven microrobots, we summarized the uses of optical tweezers, optoelectronic tweezers, and heat-mediated optical manipulation techniques. For magnetically driven microrobots, we summarized the uses of torque-driven microrobots, force-driven microrobots, and shape-deformable microrobots. Then, we compared the two types of field-driven microrobots and reviewed their advantages and disadvantages. The paper concludes with an outlook for the joint use of optical and magnetic field actuation in microrobots.
Collapse
Affiliation(s)
- Yaozhen Hou
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, 100081, China.
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
| | - Huaping Wang
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, 100081, China.
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| | - Rongxin Fu
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Xian Wang
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada
| | - Jiangfan Yu
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
| | - Shuailong Zhang
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, 100081, China.
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| | - Qiang Huang
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, 100081, China.
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Toshio Fukuda
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
13
|
Breukers J, Ven K, Struyfs C, Ampofo L, Rutten I, Imbrechts M, Pollet F, Van Lent J, Kerstens W, Noppen S, Schols D, De Munter P, Thibaut HJ, Vanhoorelbeke K, Spasic D, Declerck P, Cammue BPA, Geukens N, Thevissen K, Lammertyn J. FLUIDOT: A Modular Microfluidic Platform for Single-Cell Study and Retrieval, with Applications in Drug Tolerance Screening and Antibody Mining. SMALL METHODS 2023; 7:e2201477. [PMID: 36642827 DOI: 10.1002/smtd.202201477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/21/2022] [Indexed: 06/17/2023]
Abstract
Advancements in lab-on-a-chip technologies have revolutionized the single-cell analysis field. However, an accessible platform for in-depth screening and specific retrieval of single cells, which moreover enables studying diverse cell types and performing various downstream analyses, is still lacking. As a solution, FLUIDOT is introduced, a versatile microfluidic platform incorporating customizable microwells, optical tweezers and an interchangeable cell-retrieval system. Thanks to its smart microfluidic design, FLUIDOT is straightforward to fabricate and operate, rendering the technology widely accessible. The performance of FLUIDOT is validated and its versatility is subsequently demonstrated in two applications. First, drug tolerance in yeast cells is studied, resulting in the discovery of two treatment-tolerant populations. Second, B cells from convalescent COVID-19 patients are screened, leading to the discovery of highly affine, in vitro neutralizing monoclonal antibodies against SARS-CoV-2. Owing to its performance, flexibility, and accessibility, it is foreseen that FLUIDOT will enable phenotypic and genotypic analysis of diverse cell samples and thus elucidate unexplored biological questions.
Collapse
Affiliation(s)
- Jolien Breukers
- Department of Biosystems, Biosensors group, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
- LISCO, KU Leuven Institute for Single Cell Omics, ON4 Herestraat 49, Leuven, 3000, Belgium
| | - Karen Ven
- Department of Biosystems, Biosensors group, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
- LISCO, KU Leuven Institute for Single Cell Omics, ON4 Herestraat 49, Leuven, 3000, Belgium
- MabMine: KU Leuven Single B Cell Mining Platform, KU Leuven, ON2 Herestraat 49, 3000, Leuven, Belgium
| | - Caroline Struyfs
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, Leuven, 3001, Belgium
| | - Louanne Ampofo
- Department of Biosystems, Biosensors group, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
- MabMine: KU Leuven Single B Cell Mining Platform, KU Leuven, ON2 Herestraat 49, 3000, Leuven, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, ON 2 Herestraat 49, Leuven, 3000, Belgium
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, ON2 Herestraat 49, Leuven, 3000, Belgium
| | - Iene Rutten
- Department of Biosystems, Biosensors group, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
- LISCO, KU Leuven Institute for Single Cell Omics, ON4 Herestraat 49, Leuven, 3000, Belgium
| | - Maya Imbrechts
- MabMine: KU Leuven Single B Cell Mining Platform, KU Leuven, ON2 Herestraat 49, 3000, Leuven, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, ON 2 Herestraat 49, Leuven, 3000, Belgium
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, ON2 Herestraat 49, Leuven, 3000, Belgium
| | - Francesca Pollet
- Department of Biosystems, Biosensors group, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
| | - Julie Van Lent
- Department of Biosystems, Biosensors group, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
| | - Winnie Kerstens
- Translational Platform Virology and Chemotherapy, Rega Institute, KU Leuven, Rega - Herestraat 49, Leuven, 3000, Belgium
| | - Sam Noppen
- Laboratory of Virology and Chemotherapy, Rega Institute, KU Leuven, Rega - Herestraat 49, Leuven, 3000, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Rega Institute, KU Leuven, Rega - Herestraat 49, Leuven, 3000, Belgium
| | - Paul De Munter
- Department of Internal Medicine, University Hospitals Leuven, UZ Herestraat 49, Leuven, 3000, Belgium
- Laboratory for Clinical Infectious and Inflammatory Disorders, KU Leuven, UZ Herestraat 49, Leuven, 3000, Belgium
| | - Hendrik Jan Thibaut
- Translational Platform Virology and Chemotherapy, Rega Institute, KU Leuven, Rega - Herestraat 49, Leuven, 3000, Belgium
| | - Karen Vanhoorelbeke
- MabMine: KU Leuven Single B Cell Mining Platform, KU Leuven, ON2 Herestraat 49, 3000, Leuven, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, ON 2 Herestraat 49, Leuven, 3000, Belgium
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Etienne Sabbelaan 53, Kortrijk, 8500, Belgium
| | - Dragana Spasic
- Department of Biosystems, Biosensors group, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
| | - Paul Declerck
- MabMine: KU Leuven Single B Cell Mining Platform, KU Leuven, ON2 Herestraat 49, 3000, Leuven, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, ON 2 Herestraat 49, Leuven, 3000, Belgium
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, ON2 Herestraat 49, Leuven, 3000, Belgium
| | - Bruno P A Cammue
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, Leuven, 3001, Belgium
| | - Nick Geukens
- MabMine: KU Leuven Single B Cell Mining Platform, KU Leuven, ON2 Herestraat 49, 3000, Leuven, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, ON 2 Herestraat 49, Leuven, 3000, Belgium
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, ON2 Herestraat 49, Leuven, 3000, Belgium
| | - Karin Thevissen
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, Leuven, 3001, Belgium
| | - Jeroen Lammertyn
- Department of Biosystems, Biosensors group, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
- LISCO, KU Leuven Institute for Single Cell Omics, ON4 Herestraat 49, Leuven, 3000, Belgium
- MabMine: KU Leuven Single B Cell Mining Platform, KU Leuven, ON2 Herestraat 49, 3000, Leuven, Belgium
- LIMNI, KU Leuven Institute for Micro- and Nanoscale Integration, Celestijnenlaan 200F, Leuven, 3001, Belgium
| |
Collapse
|
14
|
Zhu D, Wang Z, Xu Y, Lin J, Qiu M, Liu J, Li X. Novel application of anti-human Fc nanobody for screening high-producing CHO cells for monoclonal antibody. Eng Life Sci 2022; 22:608-618. [PMID: 36247827 PMCID: PMC9550735 DOI: 10.1002/elsc.202200028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/08/2022] Open
Abstract
Animal-derived anti-IgG secondary antibodies are currently employed to stain and screen of human monoclonal antibody(mAb)-producing cells, but using animal-derived antibodies may raise the concerns of high cost, complicated operations and biological safety issues in biopharmaceutical manufacturing. Nanobodies(VHHs) are attractive forms of antibodies for their straightforward engineering and expression in both eukaryotic and prokaryotic systems. Using phage-displayed immune llama VHH library, we identified new anti-Fc VHHs that could bind to human Fc with high affinity. In GFP fusion format, the anti-Fc VHH-GFP generated dramatically stronger FACS signals than AF488 conjugated anti-IgG antibodies when used for staining mAb-producing CHO cells. Furthermore, preparative sorting of CHO cells based on anti-Fc VHH-GFP staining resulted in the enrichment of cell lines capable of synthesizing mAb at high productivity. This safe and cost-efficient anti-Fc VHH-GFP may optimize the process of generating highly productive cell lines for therapeutic mAb production compared to conventional animal-derived fluorescent antibodies.
Collapse
Affiliation(s)
- Di Zhu
- ChengduMedical CollegeSichuan ProvinceChina
| | - Zheng Wang
- Shanghai Bao Pharmaceuticals Co.Ltd.ShanghaiChina
| | - Yunxia Xu
- Shanghai Bao Pharmaceuticals Co.Ltd.ShanghaiChina
| | - Jing Lin
- ABLINK Biotech Co.Ltd.ChengduChina
| | - Mei Qiu
- ABLINK Biotech Co.Ltd.ChengduChina
| | - Jianghai Liu
- ChengduMedical CollegeSichuan ProvinceChina
- ABLINK Biotech Co.Ltd.ChengduChina
| | - Xinlei Li
- ChengduMedical CollegeSichuan ProvinceChina
| |
Collapse
|
15
|
Miwa H, Dimatteo R, de Rutte J, Ghosh R, Di Carlo D. Single-cell sorting based on secreted products for functionally defined cell therapies. MICROSYSTEMS & NANOENGINEERING 2022; 8:84. [PMID: 35874174 PMCID: PMC9303846 DOI: 10.1038/s41378-022-00422-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 05/13/2023]
Abstract
Cell therapies have emerged as a promising new class of "living" therapeutics over the last decade and have been particularly successful for treating hematological malignancies. Increasingly, cellular therapeutics are being developed with the aim of treating almost any disease, from solid tumors and autoimmune disorders to fibrosis, neurodegenerative disorders and even aging itself. However, their therapeutic potential has remained limited due to the fundamental differences in how molecular and cellular therapies function. While the structure of a molecular therapeutic is directly linked to biological function, cells with the same genetic blueprint can have vastly different functional properties (e.g., secretion, proliferation, cell killing, migration). Although there exists a vast array of analytical and preparative separation approaches for molecules, the functional differences among cells are exacerbated by a lack of functional potency-based sorting approaches. In this context, we describe the need for next-generation single-cell profiling microtechnologies that allow the direct evaluation and sorting of single cells based on functional properties, with a focus on secreted molecules, which are critical for the in vivo efficacy of current cell therapies. We first define three critical processes for single-cell secretion-based profiling technology: (1) partitioning individual cells into uniform compartments; (2) accumulating secretions and labeling via reporter molecules; and (3) measuring the signal associated with the reporter and, if sorting, triggering a sorting event based on these reporter signals. We summarize recent academic and commercial technologies for functional single-cell analysis in addition to sorting and industrial applications of these technologies. These approaches fall into three categories: microchamber, microfluidic droplet, and lab-on-a-particle technologies. Finally, we outline a number of unmet needs in terms of the discovery, design and manufacturing of cellular therapeutics and how the next generation of single-cell functional screening technologies could allow the realization of robust cellular therapeutics for all patients.
Collapse
Affiliation(s)
- Hiromi Miwa
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
| | - Robert Dimatteo
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
| | - Joseph de Rutte
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
- Partillion Bioscience, Los Angeles, CA 90095 USA
| | - Rajesh Ghosh
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
- Department of Mechanical and Aerospace Engineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
16
|
Yang W, Zhang J, Xiao Y, Li W, Wang T. Screening Strategies for High-Yield Chinese Hamster Ovary Cell Clones. Front Bioeng Biotechnol 2022; 10:858478. [PMID: 35782513 PMCID: PMC9247297 DOI: 10.3389/fbioe.2022.858478] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/23/2022] [Indexed: 12/20/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are by far the most commonly used mammalian expression system for recombinant expression of therapeutic proteins in the pharmaceutical industry. The development of high-yield stable cell lines requires processes of transfection, selection, screening and adaptation, among which the screening process requires tremendous time and determines the level of forming highly productive monoclonal cell lines. Therefore, how to achieve productive cell lines is a major question prior to industrial manufacturing. Cell line development (CLD) is one of the most critical steps in the production of recombinant therapeutic proteins. Generation of high-yield cell clones is mainly based on the time-consuming, laborious process of selection and screening. With the increase in recombinant therapeutic proteins expressed by CHO cells, CLD has become a major bottleneck in obtaining cell lines for manufacturing. The basic principles for CLD include preliminary screening for high-yield cell pool, single-cell isolation and improvement of productivity, clonality and stability. With the development of modern analysis and testing technologies, various screening methods have been used for CLD to enhance the selection efficiency of high-yield clonal cells. This review provides a comprehensive overview on preliminary screening methods for high-yield cell pool based on drug selective pressure. Moreover, we focus on high throughput methods for isolating high-yield cell clones and increasing the productivity and stability, as well as new screening strategies used for the biopharmaceutical industry.
Collapse
Affiliation(s)
- Wenwen Yang
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang, China
| | - Junhe Zhang
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang, China
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Tianyun Wang, ; Junhe Zhang,
| | - Yunxi Xiao
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
| | - Wenqing Li
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| | - Tianyun Wang
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang, China
- *Correspondence: Tianyun Wang, ; Junhe Zhang,
| |
Collapse
|
17
|
Hannart H, Berger A, Aeberli L, Forchelet D, Uffer N, Muller G, Barrandon Y, Renaud P, Bonzon D. Traceable impedance-based single-cell pipetting, from a research set-up to a robust and fast automated robot: DispenCell-S1. SLAS Technol 2022; 27:121-129. [DOI: 10.1016/j.slast.2021.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
18
|
Zhang S, Xu B, Elsayed M, Nan F, Liang W, Valley JK, Liu L, Huang Q, Wu MC, Wheeler AR. Optoelectronic tweezers: a versatile toolbox for nano-/micro-manipulation. Chem Soc Rev 2022; 51:9203-9242. [DOI: 10.1039/d2cs00359g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This review covers the fundamentals, recent progress and state-of-the-art applications of optoelectronic tweezers technology, and demonstrates that optoelectronic tweezers technology is a versatile and powerful toolbox for nano-/micro-manipulation.
Collapse
Affiliation(s)
- Shuailong Zhang
- School of Mechatronical Engineering, Beijing Institute of Technology, Room 711, Building No 6, Science and Technology Park, 5 Zhongguancun South St, Haidian District, Beijing, 100081, China
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| | - Bingrui Xu
- School of Mechatronical Engineering, Beijing Institute of Technology, Room 711, Building No 6, Science and Technology Park, 5 Zhongguancun South St, Haidian District, Beijing, 100081, China
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
| | - Mohamed Elsayed
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Fan Nan
- Institute of Nanophotonics, Jinan University, Guangzhou 511443, China
| | - Wenfeng Liang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang, 110168, China
| | - Justin K. Valley
- Berkeley Lights, Inc, 5858 Horton Street #320, Emeryville, CA 94608, USA
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
| | - Qiang Huang
- School of Mechatronical Engineering, Beijing Institute of Technology, Room 711, Building No 6, Science and Technology Park, 5 Zhongguancun South St, Haidian District, Beijing, 100081, China
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| | - Ming C. Wu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720, USA
| | - Aaron R. Wheeler
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| |
Collapse
|
19
|
Tihanyi B, Nyitray L. Recent advances in CHO cell line development for recombinant protein production. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 38:25-34. [PMID: 34895638 DOI: 10.1016/j.ddtec.2021.02.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/02/2021] [Accepted: 02/23/2021] [Indexed: 12/20/2022]
Abstract
Recombinant proteins used in biomedical research, diagnostics and different therapies are mostly produced in Chinese hamster ovary cells in the pharmaceutical industry. These biotherapeutics, monoclonal antibodies in particular, have shown remarkable market growth in the past few decades. The increasing demand for high amounts of biologics requires continuous optimization and improvement of production technologies. Research aims at discovering better means and methods for reaching higher volumetric capacity, while maintaining stable product quality. An increasing number of complex novel protein therapeutics, such as viral antigens, vaccines, bi- and tri-specific monoclonal antibodies, are currently entering industrial production pipelines. These biomolecules are, in many cases, difficult to express and require tailored product-specific solutions to improve their transient or stable production. All these requirements boost the development of more efficient expression optimization systems and high-throughput screening platforms to facilitate the design of product-specific cell line engineering and production strategies. In this minireview, we provide an overview on recent advances in CHO cell line development, targeted genome manipulation techniques, selection systems and screening methods currently used in recombinant protein production.
Collapse
Affiliation(s)
- Borbála Tihanyi
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter stny 1/C, 1117 Budapest, Hungary
| | - László Nyitray
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter stny 1/C, 1117 Budapest, Hungary.
| |
Collapse
|
20
|
Pybus LP, Kalsi D, Matthews JT, Hawke E, Barber N, Richer R, Young A, Saunders FL. Coupling picodroplet microfluidics with plate imaging for the rapid creation of biomanufacturing suitable cell lines with high probability and improved multi-step assurance of monoclonality. Biotechnol J 2021; 17:e2100357. [PMID: 34633760 DOI: 10.1002/biot.202100357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND There is an expectation from regulatory agencies that cell lines used in the commercial production of biopharmaceuticals are derived from a single cell progenitor. Traditional methods of single cell cloning include the use of the limiting dilution cloning method which often requires multiple rounds of low cell density cell plating and either microscopic evaluation that wells contain single cells and/or the calculation of a statistically derived probability of monoclonality. METHODS AND RESULTS We have combined the single cell screening, deposition and picodroplet imaging ability of Sphere Fluidics' Cyto-Mine technology with the plate imaging capability of the Solentim Cell Metric to create a novel workflow for the generation of high producing clonal cell lines with both high probability and assurance of monoclonality. The efficiency of three key stages of the process (single cell picodroplet encapsulation, single picodroplet dispensation and single cell settling in the focal plane of the plate imager) was determined and a probability calculation was derived using the Wilson Score Interval method. The combined probability that a single cell is encapsulated into a picodroplet, is deposited into the correct well of a 96-well plate and that a cell settles into the focal plane of the plate imager yields a combined > 99% probability of monoclonality. Furthermore, visual verification of a single cell progenitor is obtained at multiple steps throughout the cloning workflow. CONCLUSION This novel methodology for the rapid creation of high quality clonal cell lines for biomanufacturing purposes has many advantages over more traditional approaches including improved assurance of single cell derivation, integrated imaging capability, assay flexibility, equipment utilization time and in-process cell line segregation.
Collapse
Affiliation(s)
- Leon P Pybus
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Devika Kalsi
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Joe T Matthews
- School of Mathematics, Statistics and Physics, Newcastle University, Newcastle upon Tyne, UK
| | - Ellie Hawke
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK.,Current Address: Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Nicholas Barber
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Rachel Richer
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Alison Young
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Fay L Saunders
- Mammalian Cell Culture Process Development, FUJIFILM Diosynth Biotechnologies, Billingham, UK
| |
Collapse
|
21
|
Tejwani V, Chaudhari M, Rai T, Sharfstein ST. High-throughput and automation advances for accelerating single-cell cloning, monoclonality and early phase clone screening steps in mammalian cell line development for biologics production. Biotechnol Prog 2021; 37:e3208. [PMID: 34478248 DOI: 10.1002/btpr.3208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
Mammalian cell line development is a multistep process wherein timelines for developing clonal cells to be used as manufacturing cell lines for biologics production can commonly extend to 9 months when no automation or modern molecular technologies are involved in the workflow. Steps in the cell line development workflow involving single-cell cloning, monoclonality assurance, productivity and stability screening are labor, time and resource intensive when performed manually. Introduction of automation and miniaturization in these steps has reduced the required manual labor, shortened timelines from months to weeks, and decreased the resources needed to develop manufacturing cell lines. This review summarizes the advances, benefits, comparisons and shortcomings of different automation platforms available in the market for rapid isolation of desired clonal cell lines for biologics production.
Collapse
Affiliation(s)
- Vijay Tejwani
- Biotechnology R&D, Clone Development Team, Lupin Limited, Pune, India
| | - Minal Chaudhari
- Biotechnology R&D, Clone Development Team, Lupin Limited, Pune, India
| | - Toyaj Rai
- Biotechnology R&D, Clone Development Team, Lupin Limited, Pune, India
| | - Susan T Sharfstein
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York, USA
| |
Collapse
|
22
|
São Pedro MN, Silva TC, Patil R, Ottens M. White paper on high-throughput process development for integrated continuous biomanufacturing. Biotechnol Bioeng 2021; 118:3275-3286. [PMID: 33749840 PMCID: PMC8451798 DOI: 10.1002/bit.27757] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/15/2021] [Accepted: 03/12/2021] [Indexed: 12/25/2022]
Abstract
Continuous manufacturing is an indicator of a maturing industry, as can be seen by the example of the petrochemical industry. Patent expiry promotes a price competition between manufacturing companies, and more efficient and cheaper processes are needed to achieve lower production costs. Over the last decade, continuous biomanufacturing has had significant breakthroughs, with regulatory agencies encouraging the industry to implement this processing mode. Process development is resource and time consuming and, although it is increasingly becoming less expensive and faster through high-throughput process development (HTPD) implementation, reliable HTPD technology for integrated and continuous biomanufacturing is still lacking and is considered to be an emerging field. Therefore, this paper aims to illustrate the major gaps in HTPD and to discuss the major needs and possible solutions to achieve an end-to-end Integrated Continuous Biomanufacturing, as discussed in the context of the 2019 Integrated Continuous Biomanufacturing conference. The current HTPD state-of-the-art for several unit operations is discussed, as well as the emerging technologies which will expedite a shift to continuous biomanufacturing.
Collapse
Affiliation(s)
| | - Tiago C. Silva
- Department of BiotechnologyDelft University of TechnologyDelftThe Netherlands
| | - Rohan Patil
- Global CMC DevelopmentSanofiFraminghamMassachusettsUSA
| | - Marcel Ottens
- Department of BiotechnologyDelft University of TechnologyDelftThe Netherlands
| |
Collapse
|
23
|
Diep J, Le H, Le K, Zasadzinska E, Tat J, Yam P, Zastrow R, Gomez N, Stevens J. Microfluidic chip-based single-cell cloning to accelerate biologic production timelines. Biotechnol Prog 2021; 37:e3192. [PMID: 34323013 PMCID: PMC9285370 DOI: 10.1002/btpr.3192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/06/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022]
Abstract
Cell line development (CLD) represents a critical, yet time‐consuming, step in the biomanufacturing process as significant resources are devoted to the scale‐up and screening of several hundreds to thousands of single‐cell clones. Typically, transfected pools are fully recovered from selection and characterized for growth, productivity, and product quality to identify the best pools suitable for single‐cell cloning (SCC) using limiting dilution or fluorescence‐activated cell sorting (FACS). Here we report the application of the Berkeley Lights Beacon Instrument (BLI) in an early SCC process to accelerate the CLD timeline. Transfected pools were single‐cell cloned when viabilities reached greater than 85% or during selection when viabilities were less than 30%. Clones isolated from these accelerated processes exhibited comparable growth, productivity, and product quality to those derived from a standard CLD process and fit into an existing manufacturing platform. With these approaches, up to a 30% reduction in the overall CLD timeline was achieved. Furthermore, early process‐derived clones demonstrated equivalent long‐term stability compared with standard process‐derived clones over 50 population doubling levels (PDLs). Taken together, the data supported early SCC on the BLI as an attractive approach to reducing the standard CLD timeline while still identifying clones with acceptable manufacturability.
Collapse
Affiliation(s)
- Jonathan Diep
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Huong Le
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Kim Le
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Ewelina Zasadzinska
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Jasmine Tat
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Pheng Yam
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Ryan Zastrow
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Natalia Gomez
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Jennitte Stevens
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| |
Collapse
|
24
|
Zhang W, Li Q, Jia F, Hu Z, Wei Z. A Microfluidic Chip for Screening and Sequencing of Monoclonal Antibody at a Single-Cell Level. Anal Chem 2021; 93:10099-10105. [PMID: 34264632 DOI: 10.1021/acs.analchem.1c00918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The pairing of heavy and light chains of an antibody decides the specificity of monoclonal antibodies (mAbs). Acquisition of the genes encoding variable regions of paired heavy and light chains (VH:VL) is crucial, but it is a labor- and cost-intensive process in traditional methods. The emerging microfluidic chips have brought us to a portal of directly acquiring natively paired VH:VL genes by sequencing single target cells. This study presents a novel method in which all processing steps for acquiring natively paired VH:VL genes from single cells are finished in a single microfluidic chip, not multiple discrete devices. The microfluidic chip performs single-cell trapping/in situ fluorescence examination of antibody specificity/cell lysis/gene amplification all at a single-cell level. By a proof-of-concept validation of efficiently acquiring paired VH:VL genes of anti-CD45 mAbs from single hybridoma cells, the microfluidic chip has been proved capable of trapping/screening/lysing single antibody-secreting cells and performing an on-chip reverse transcription-polymerase chain reaction. The presented method has realized remarkably improved cell loss/human labor/time cost, and more importantly, determinacy of native VH:VL gene pairing, which is one of the most decisive factors of effectiveness for antibody discovery.
Collapse
Affiliation(s)
- Weikai Zhang
- Department of Biomedical Engineering, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Qin Li
- Department of Biomedical Engineering, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Fei Jia
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Zhiyuan Hu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.,School of Nanoscience and Technology, Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, P. R. China.,Center for Neuroscience Research, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108 Fujian Province, China.,School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, P. R. China
| | - Zewen Wei
- Department of Biomedical Engineering, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
25
|
Weinguny M, Klanert G, Eisenhut P, Lee I, Timp W, Borth N. Subcloning induces changes in the DNA-methylation pattern of outgrowing Chinese hamster ovary cell colonies. Biotechnol J 2021; 16:e2000350. [PMID: 33484505 DOI: 10.1002/biot.202000350] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 01/17/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
Chinese hamster ovary (CHO) cells are the most extensively used mammalian production system for biologics intended for use in humans. A critical step in the establishment of production cell lines is single cell cloning, with the objective of achieving high productivity and product quality. Despite general use, knowledge of the effects of this process is limited. Importantly, single cell cloned cells display a wide array of observed phenotypes, which so far was attributed to the instability and variability of the CHO genome. In this study we present data indicating that the emergence of diverse phenotypes during single cell cloning is associated with changes in DNA methylation patterns and transcriptomes that occur during the subcloning process. The DNA methylation pattern of each analyzed subclone, randomly picked from all outgrowing clones of the experiment, had unique changes preferentially found in regulatory regions of the genome such as enhancers, and de-enriched in actively transcribed sequences (not including the respective promoters), indicating that these changes resulted in adaptations of the relative gene expression pattern. The transcriptome of each subclone also had a significant number of individual changes. These results indicate that epigenetic regulation is a hidden, but important player in cell line development with a major role in the establishment of high performing clones with improved characteristics for bioprocessing.
Collapse
Affiliation(s)
- Marcus Weinguny
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria.,Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| | - Gerald Klanert
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Peter Eisenhut
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria.,Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| | - Isac Lee
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, USA
| | - Nicole Borth
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria.,Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| |
Collapse
|
26
|
Zhang W, Li R, Jia F, Hu Z, Li Q, Wei Z. A microfluidic chip for screening high-producing hybridomas at single cell level. LAB ON A CHIP 2020; 20:4043-4051. [PMID: 33005908 DOI: 10.1039/d0lc00847h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Hybridomas are a commonly used, or even the only option, for laboratory study and pilot production of monoclonal antibodies (mAbs), which are crucial for both targeted therapy and biomedical study. A long-term culture of hybridomas will inevitably induce a heterogenization of the whole hybridoma population, resulting in a continuous growth of non-producing hybridomas. To overcome the limits of existing methods of screening heterogeneous hybridomas, in which the whole multi-round screening process is performed in multi-well plates or other discrete modules, this study presents a novel method in which all processing steps of a multi-round hybridoma screening are finished in a single microfluidic chip. This microfluidic chip comprehensively performs hybridoma trapping/proliferating/transferring and fluorescent identification of protein-antibody binding at single cell level. By performing a two-round screening of anti-CD45 mAb secreting hybridomas, the novel microfluidic chip was proved capable of screening several single high-producing hybridomas with minimum cell loss/human labor/time cost, and more importantly, enhanced accuracy and definite monoclonality, which is one of the most important properties of mAb production.
Collapse
Affiliation(s)
- Weikai Zhang
- Department of Biomedical Engineering, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | | | | | | | | | | |
Collapse
|
27
|
Choi JR. Advances in single cell technologies in immunology. Biotechniques 2020; 69:226-236. [PMID: 32777935 DOI: 10.2144/btn-2020-0047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/06/2020] [Indexed: 11/23/2022] Open
Abstract
The immune system is composed of heterogeneous populations of immune cells that regulate physiological processes and protect organisms against diseases. Single cell technologies have been used to assess immune cell responses at the single cell level, which are crucial for identifying the causes of diseases and elucidating underlying biological mechanisms to facilitate medical therapy. In the present review we first discuss the most recent advances in the development of single cell technologies to investigate cell signaling, cell-cell interactions and cell migration. Each technology's advantages and limitations and its applications in immunology are subsequently reviewed. The latest progress toward commercialization, the remaining challenges and future perspectives for single cell technologies in immunology are also briefly discussed.
Collapse
Affiliation(s)
- Jane Ru Choi
- Centre for Blood Research, Life Sciences Centre, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
- Department of Mechanical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
28
|
Weinguny M, Klanert G, Eisenhut P, Jonsson A, Ivansson D, Lövgren A, Borth N. Directed evolution approach to enhance efficiency and speed of outgrowth during single cell subcloning of Chinese Hamster Ovary cells. Comput Struct Biotechnol J 2020; 18:1320-1329. [PMID: 32612755 PMCID: PMC7306589 DOI: 10.1016/j.csbj.2020.05.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/16/2020] [Accepted: 05/17/2020] [Indexed: 01/08/2023] Open
Abstract
Chinese Hamster Ovary (CHO) cells are the working horse of the pharmaceutical industry. To obtain high producing cell clones and to satisfy regulatory requirements single cell cloning is a necessary step in cell line development. However, it is also a tedious, labor intensive and expensive process. Here we show an easy way to enhance subclonability using subcloning by single cell sorting itself as the selection pressure, resulting in improved subcloning performance of three different host cell lines. These improvements in subclonability also lead to an enhanced cellular growth behavior during standard batch culture. RNA-seq was performed to shed light on the underlying mechanisms, showing that there is little overlap in differentially expressed genes or associated pathways between the cell lines, each finding their individual strategy for optimization. However, in all three cell lines pathways associated with the extracellular matrix were found to be enriched, indicating that cells struggle predominantly with their microenvironment and possibly lack of cell-to-cell contact. The observed small overlap may hint that there are multiple ways for a cell line to achieve a certain phenotype due to numerous genetic and subsequently metabolic redundancies.
Collapse
Key Words
- CHO
- CHO cells
- CHO, Chinese hamster ovary
- Cell line development
- Cell sorting
- Chinese Hamster Ovary Cells
- CoI, clusters of interest
- DE, directed evolved
- Directed Evolution
- ECM, extracellular matrix
- ES, enrichment score
- FACS
- FACS, fluorescent-activated cell sorting
- Fluorescent-activated cell sorting
- GSEA, gene set analysis
- Growth enhancement
- Growth improvement
- LDC, limiting dilution cloning
- NES, negative enrichment score
- PC, principal component
- PCA, principal component analysis
- POI, product of interest
- RNA Sequencing
- RNA-Seq
- RNA-Seq, RNA sequencing
- SCC, single cell cloning
- Single Cell Cloning
- Single Cell Subcloning
- Subcloning
- lfcSE, logfoldstandard error
Collapse
Affiliation(s)
- Marcus Weinguny
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| | - Gerald Klanert
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Peter Eisenhut
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| | | | | | | | - Nicole Borth
- ACIB Gmbh, Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
- Corresponding author at: Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|