1
|
Yang Y, Dalvie NC, Brady JR, Naranjo CA, Lorgeree T, Rodriguez‐Aponte SA, Johnston RS, Tracey MK, Elenberger CM, Lee E, Tié M, Love KR, Love JC. Adaptation of Aglycosylated Monoclonal Antibodies for Improved Production in Komagataella phaffii. Biotechnol Bioeng 2025; 122:361-372. [PMID: 39543843 PMCID: PMC11718428 DOI: 10.1002/bit.28878] [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: 07/04/2024] [Revised: 10/17/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024]
Abstract
Monoclonal antibodies (mAbs) are a major class of biopharmaceuticals manufactured by well-established processes using Chinese Hamster Ovary (CHO) cells. Next-generation biomanufacturing using alternative hosts like Komagataella phaffii could improve the accessibility of these medicines, address broad societal goals for sustainability, and offer financial advantages for accelerated development of new products. Antibodies produced by K. phaffii, however, may manifest unique molecular quality attributes, like host-dependent, product-related variants, that could raise potential concerns for clinical use. We demonstrate here conservative modifications to the amino acid sequence of aglycosylated antibodies based on the human IgG1 isotype that minimize product-related variations when secreted by K. phaffii. A combination of 2-3 changes of amino acids reduced variations across six different aglycosylated versions of commercial mAbs. Expression of a modified sequence of NIST mAb in both K. phaffii and CHO cells showed comparable biophysical properties and molecular variations. These results suggest a path toward the production of high-quality mAbs that could be expressed interchangeably by either yeast or mammalian cells. Improving molecular designs of proteins to enable a range of manufacturing strategies for well-characterized biopharmaceuticals could accelerate global accessibility and innovations.
Collapse
Affiliation(s)
- Yuchen Yang
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Neil C. Dalvie
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Joseph R. Brady
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Christopher A. Naranjo
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Timothy Lorgeree
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Sergio A. Rodriguez‐Aponte
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Ryan S. Johnston
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Mary K. Tracey
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Carmen M. Elenberger
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | | | - Kerry R. Love
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - J. Christopher Love
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| |
Collapse
|
2
|
Krishna S, Jung ST, Lee EY. Escherichia coli and Pichia pastoris: microbial cell-factory platform for -full-length IgG production. Crit Rev Biotechnol 2025; 45:191-213. [PMID: 38797692 DOI: 10.1080/07388551.2024.2342969] [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: 12/13/2023] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 05/29/2024]
Abstract
Owing to the unmet demand, the pharmaceutical industry is investigating an alternative host to mammalian cells to produce antibodies for a variety of therapeutic and research applications. Regardless of some disadvantages, Escherichia coli and Pichia pastoris are the preferred microbial hosts for antibody production. Despite the fact that the production of full-length antibodies has been successfully demonstrated in E. coli, which has mostly been used to produce antibody fragments, such as: antigen-binding fragments (Fab), single-chain fragment variable (scFv), and nanobodies. In contrast, Pichia, a eukaryotic microbial host, is mostly used to produce glycosylated full-length antibodies, though hypermannosylated glycan is a major challenge. Advanced strategies, such as the introduction of human-like glycosylation in endotoxin-edited E. coli and cell-free system-based glycosylation, are making progress in creating human-like glycosylation profiles of antibodies in these microbes. This review begins by explaining the structural and functional requirements of antibodies and continues by describing and analyzing the potential of E. coli and P. pastoris as hosts for providing a favorable environment to create a fully functional antibody. In addition, authors compare these microbes on certain features and predict their future in antibody production. Briefly, this review analyzes, compares, and highlights E. coli and P. pastoris as potential hosts for antibody production.
Collapse
Affiliation(s)
- Shyam Krishna
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Sang Taek Jung
- BK21 Graduate Program, Department of Biomedical Sciences, Graduate School, Korea University, Seoul, Republic of Korea
| | - Eun Yeol Lee
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
3
|
Janezic EM, Doan A, Mai E, Bravo DD, Wang J, Kim HS, Spiess C, Bewley K, ElSohly A, Liang WC, Koerber JT, Richalet P, Vanhove M, Comps-Agrar L. A novel, label-free, pre-equilibrium assay to determine the association and dissociation rate constants of therapeutic antibodies on living cells. Br J Pharmacol 2024; 181:3836-3855. [PMID: 37783572 DOI: 10.1111/bph.16258] [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: 03/15/2023] [Revised: 09/19/2023] [Accepted: 09/23/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Monoclonal antibodies (Ab) represent the fastest growing drug class. Knowledge of the biophysical parameters (kon, koff and KD) that dictate Ab:receptor interaction is critical during the drug discovery process. However, with the increasing complexity of Ab formats and their targets, it became apparent that existing technologies present limitations and are not always suitable to determine these parameters. Therefore, novel affinity determination methods represent an unmet assay need. EXPERIMENTAL APPROACH We developed a pre-equilibrium kinetic exclusion assay using recent mathematical advances to determine the kon, koff and KD of monoclonal Ab:receptor interactions on living cells. The assay is amenable to all human IgG1 and rabbit Abs. KEY RESULTS Using our novel assay, we demonstrated for several monoclonal Ab:receptor pairs that the calculated kinetic rate constants were comparable with orthogonal methods that were lower throughput or more resource consuming. We ran simulations to predict the critical conditions to improve the performance of the assays. We further showed that this method could successfully be applied to both suspension and adherent cells. Finally, we demonstrated that kon and koff, but not KD, correlate with in vitro potency for a panel of monoclonal Abs. CONCLUSIONS AND IMPLICATIONS Our novel assay has the potential to systematically probe binding kinetics of monoclonal Abs to cells and can be incorporated in a screening cascade to identify new therapeutic candidates. Wide-spread adoption of pre-equilibrium assays using physiologically relevant systems will lead to a more holistic understanding of how Ab binding kinetics influence their potency.
Collapse
Affiliation(s)
| | | | - Elaine Mai
- Genentech, Inc, South San Francisco, California, USA
| | | | - Jianyong Wang
- Genentech, Inc, South San Francisco, California, USA
| | - Hok Seon Kim
- Genentech, Inc, South San Francisco, California, USA
| | | | | | - Adel ElSohly
- Genentech, Inc, South San Francisco, California, USA
| | | | | | | | | | | |
Collapse
|
4
|
de Moraes LMP, Marques HF, Reis VCB, Coelho CM, Leitão MDC, Galdino AS, Porto de Souza TP, Piva LC, Perez ALA, Trichez D, de Almeida JRM, De Marco JL, Torres FAG. Applications of the Methylotrophic Yeast Komagataella phaffii in the Context of Modern Biotechnology. J Fungi (Basel) 2024; 10:411. [PMID: 38921397 PMCID: PMC11205268 DOI: 10.3390/jof10060411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024] Open
Abstract
Komagataella phaffii (formerly Pichia pastoris) is a methylotrophic yeast widely used in laboratories around the world to produce recombinant proteins. Given its advantageous features, it has also gained much interest in the context of modern biotechnology. In this review, we present the utilization of K. phaffii as a platform to produce several products of economic interest such as biopharmaceuticals, renewable chemicals, fuels, biomaterials, and food/feed products. Finally, we present synthetic biology approaches currently used for strain engineering, aiming at the production of new bioproducts.
Collapse
Affiliation(s)
- Lidia Maria Pepe de Moraes
- Laboratory of Molecular Biology, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (L.M.P.d.M.); (H.F.M.); (L.C.P.); (A.L.A.P.); (J.L.D.M.)
| | - Henrique Fetzner Marques
- Laboratory of Molecular Biology, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (L.M.P.d.M.); (H.F.M.); (L.C.P.); (A.L.A.P.); (J.L.D.M.)
| | - Viviane Castelo Branco Reis
- Laboratory of Genetics and Biotechnology, Embresa Brasileira de Pesquisa Agropecuária (EMBRAPA) Agroenergy, Brasília 70770-901, DF, Brazil; (V.C.B.R.); (D.T.); (J.R.M.d.A.)
| | - Cintia Marques Coelho
- Laboratory of Synthetic Biology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (C.M.C.); (M.d.C.L.)
| | - Matheus de Castro Leitão
- Laboratory of Synthetic Biology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (C.M.C.); (M.d.C.L.)
| | - Alexsandro Sobreira Galdino
- Microbial Biotechnology Laboratory, Federal University of São João Del-Rei, Divinópolis 35501-296, MG, Brazil; (A.S.G.); (T.P.P.d.S.)
| | - Thais Paiva Porto de Souza
- Microbial Biotechnology Laboratory, Federal University of São João Del-Rei, Divinópolis 35501-296, MG, Brazil; (A.S.G.); (T.P.P.d.S.)
| | - Luiza Cesca Piva
- Laboratory of Molecular Biology, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (L.M.P.d.M.); (H.F.M.); (L.C.P.); (A.L.A.P.); (J.L.D.M.)
| | - Ana Laura Alfonso Perez
- Laboratory of Molecular Biology, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (L.M.P.d.M.); (H.F.M.); (L.C.P.); (A.L.A.P.); (J.L.D.M.)
| | - Débora Trichez
- Laboratory of Genetics and Biotechnology, Embresa Brasileira de Pesquisa Agropecuária (EMBRAPA) Agroenergy, Brasília 70770-901, DF, Brazil; (V.C.B.R.); (D.T.); (J.R.M.d.A.)
| | - João Ricardo Moreira de Almeida
- Laboratory of Genetics and Biotechnology, Embresa Brasileira de Pesquisa Agropecuária (EMBRAPA) Agroenergy, Brasília 70770-901, DF, Brazil; (V.C.B.R.); (D.T.); (J.R.M.d.A.)
| | - Janice Lisboa De Marco
- Laboratory of Molecular Biology, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (L.M.P.d.M.); (H.F.M.); (L.C.P.); (A.L.A.P.); (J.L.D.M.)
| | - Fernando Araripe Gonçalves Torres
- Laboratory of Molecular Biology, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (L.M.P.d.M.); (H.F.M.); (L.C.P.); (A.L.A.P.); (J.L.D.M.)
| |
Collapse
|
5
|
Kang M, Wang Z, Ge X. One-step production of fully biotinylated and glycosylated human Fc gamma receptors. Biotechnol Prog 2024; 40:e3392. [PMID: 37734055 PMCID: PMC10922510 DOI: 10.1002/btpr.3392] [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: 07/07/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023]
Abstract
Initiating and regulating humoral immunity, Fc gamma receptors (FcγRs) have been identified both as therapeutics and as drug targets, and thus production of biologically active FcγRs is highly demanded for biopharmaceutical development. Focusing on low-affinity FcγRs IIA (131H/R allotypes), IIB, and IIIA (176F/V), this study used human 293-F cells to achieve correct post-translational modifications (PTMs) including biotinylation, N-glycosylation, and disulfides. Approaches involving co-expression of FcγR-AviTag and Escherichia coli biotin ligase BirA, endoplasmic reticulum retention, stable and transient transfections, and optimization of transgene ratio were investigated. Protein electrophoresis under reducing and non-reducing conditions, enzymatic deglycosylation, streptavidin pull-down assays, and binding kinetic analysis collectively indicated that the produced FcγR ectodomains were fully biotinylated, N-glycosylated, had formed disulfide bond, and exhibited expected binding affinities toward IgG1 trastuzumab and its Fc mutants. A clear trade-off between production yield and PTM quality was also observed. Achieving multiple types of PTMs completely by one-step cell culture should have applications for the production of a variety of complex proteins of biomedical importance.
Collapse
Affiliation(s)
- Minhyo Kang
- Department of Chemical and Environmental Engineering, University of California Riverside, CA, USA
- Present address: Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC Canada
| | - Zening Wang
- Department of Chemical and Environmental Engineering, University of California Riverside, CA, USA
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, USA
| | - Xin Ge
- Department of Chemical and Environmental Engineering, University of California Riverside, CA, USA
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, USA
| |
Collapse
|
6
|
Fan X, Yuan Z, Zhao Y, Xiong W, Hsiao HC, Pare R, Ding J, Almosa A, Sun K, Zhang S, Jordan RE, Lee CS, An Z, Zhang N. Impairment of IgG Fc functions promotes tumor progression and suppresses NK cell antitumor actions. Commun Biol 2022; 5:960. [PMID: 36104515 PMCID: PMC9474879 DOI: 10.1038/s42003-022-03931-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 09/01/2022] [Indexed: 11/15/2022] Open
Abstract
Natural killer (NK) cells mediate antibody dependent cytotoxic killing of cancer cells via cross-linking FcγR on NK cells with IgG-Fc. Studies have shown that the single-hinge cleaved IgGs (scIgGs) have dysfunctional Fc and failed engagement with FcγRs on immune cells. However, little is known about how scIgGs impact on antitumor immunity in the tumor microenvironment. In this study, we revealed a significant association of tumor scIgGs with tumor progression and poor outcomes of breast cancer patients (n = 547). Using multiple mouse tumor models, we demonstrated that tumor scIgGs reduced NK cell cytotoxic activities and resulted in aggressive tumor progression. We further showed that an anti-hinge specific monoclonal antibody (AHA) rescued the dysfunctional Fc in scIgGs by providing a functional Fc and restored NK cell cytotoxic activity. These findings point to a novel immunotherapeutic strategy to enhance Fc engagement with FcγRs for activation of anticancer immunity.
Collapse
Affiliation(s)
- Xuejun Fan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, 77030, USA
| | - Zihao Yuan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, 77030, USA
| | - Yueshui Zhao
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, 77030, USA
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, 77030, USA
| | - Hao-Ching Hsiao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, 77030, USA
| | - Rahmawati Pare
- School of Medicine, Western Sydney University, Department of Anatomical Pathology, Liverpool Hospital, Cancer Pathology Laboratory, Ingham Institute for Applied Medical Research, Liverpool BC, NSW, 1871, Australia
- Medicine & Health Sciences, University Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia
| | - Jianmin Ding
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Ahmad Almosa
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, 77030, USA
| | - Songlin Zhang
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Robert E Jordan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, 77030, USA
| | - Cheok Song Lee
- School of Medicine, Western Sydney University, Department of Anatomical Pathology, Liverpool Hospital, Cancer Pathology Laboratory, Ingham Institute for Applied Medical Research, Liverpool BC, NSW, 1871, Australia
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, 77030, USA.
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Wang T, Liu L, Voglmeir J. mAbs N-glycosylation: Implications for biotechnology and analytics. Carbohydr Res 2022; 514:108541. [DOI: 10.1016/j.carres.2022.108541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/29/2022]
|
8
|
Rinnofner C, Felber M, Pichler H. Strains and Molecular Tools for Recombinant Protein Production in Pichia pastoris. Methods Mol Biol 2022; 2513:79-112. [PMID: 35781201 DOI: 10.1007/978-1-0716-2399-2_6] [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] [Indexed: 06/15/2023]
Abstract
Within the last two decades, the methylotrophic yeast Pichia pastoris (Komagataella phaffii) has become an important alternative to E. coli or mammalian cell lines for the production of recombinant proteins. Easy handling, strong promoters, and high cell density cultivations as well as the capability of posttranslational modifications are some of the major benefits of this yeast. The high secretion capacity and low level of endogenously secreted proteins further promoted the rapid development of a versatile Pichia pastoris toolbox. This chapter reviews common and new "Pichia tools" and their specific features. Special focus is given to expression strains, such as different methanol utilization, protease-deficient or glycoengineered strains, combined with application highlights. Different promoters and signal sequences are also discussed.
Collapse
Affiliation(s)
- Claudia Rinnofner
- Austrian Centre of Industrial Biotechnology (ACIB), Graz, Austria.
- Bisy GmbH, Hofstaetten/Raab, Austria.
| | - Michael Felber
- Austrian Centre of Industrial Biotechnology (ACIB), Graz, Austria
| | - Harald Pichler
- Austrian Centre of Industrial Biotechnology (ACIB), Graz, Austria
- Institute of Molecular Biotechnology, Graz University of Technology, Graz, Austria
| |
Collapse
|
9
|
Yanakieva D, Pekar L, Evers A, Fleischer M, Keller S, Mueller-Pompalla D, Toleikis L, Kolmar H, Zielonka S, Krah S. Beyond bispecificity: Controlled Fab arm exchange for the generation of antibodies with multiple specificities. MAbs 2022; 14:2018960. [PMID: 35014603 PMCID: PMC8757479 DOI: 10.1080/19420862.2021.2018960] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/13/2021] [Indexed: 01/07/2023] Open
Abstract
Controlled Fab arm exchange (cFAE) has proven to be a generic and versatile technology for the efficient generation of IgG-like bispecific antibodies (DuoBodies or DBs), with several in clinical development and one product, amivantamab, approved by the Food and Drug Administration. In this study, we expand the cFAE-toolbox by incorporating VHH-modules at the C-termini of DB-IgGs, termed DB-VHHs. This approach enables the combinatorial generation of tri- and tetraspecific molecules with flexible valencies in a straightforward fashion. Using cFAE, a variety of multispecific molecules was produced and assessed for manufacturability and physicochemical characteristics. In addition, we were able to generate DB-VHHs that efficiently triggered natural killer cell mediated lysis of tumor cells, demonstrating the utility of this format for potential therapeutic applications.
Collapse
Affiliation(s)
- Desislava Yanakieva
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Lukas Pekar
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Andreas Evers
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Markus Fleischer
- Protein and Cell Sciences, Merck Healthcare KGaA, Darmstadt, Germany
| | - Stephan Keller
- Protein and Cell Sciences, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Lars Toleikis
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Simon Krah
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
10
|
On the Use of Surface Plasmon Resonance Biosensing to Understand IgG-FcγR Interactions. Int J Mol Sci 2021; 22:ijms22126616. [PMID: 34205578 PMCID: PMC8235063 DOI: 10.3390/ijms22126616] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 01/01/2023] Open
Abstract
Surface plasmon resonance (SPR)-based optical biosensors offer real-time and label-free analysis of protein interactions, which has extensively contributed to the discovery and development of therapeutic monoclonal antibodies (mAbs). As the biopharmaceutical market for these biologics and their biosimilars is rapidly growing, the role of SPR biosensors in drug discovery and quality assessment is becoming increasingly prominent. One of the critical quality attributes of mAbs is the N-glycosylation of their Fc region. Other than providing stability to the antibody, the Fc N-glycosylation influences immunoglobulin G (IgG) interactions with the Fcγ receptors (FcγRs), modulating the immune response. Over the past two decades, several studies have relied on SPR-based assays to characterize the influence of N-glycosylation upon the IgG-FcγR interactions. While these studies have unveiled key information, many conclusions are still debated in the literature. These discrepancies can be, in part, attributed to the design of the reported SPR-based assays as well as the methodology applied to SPR data analysis. In fact, the SPR biosensor best practices have evolved over the years, and several biases have been pointed out in the development of experimental SPR protocols. In parallel, newly developed algorithms and data analysis methods now allow taking into consideration complex biomolecular kinetics. In this review, we detail the use of different SPR biosensing approaches for characterizing the IgG-FcγR interactions, highlighting their merit and inherent experimental complexity. Furthermore, we review the latest SPR-derived conclusions on the influence of the N-glycosylation upon the IgG-FcγR interactions and underline the differences and similarities across the literature. Finally, we explore new avenues taking advantage of novel computational analysis of SPR results as well as the latest strategies to control the glycoprofile of mAbs during production, which could lead to a better understanding and modelling of the IgG-FcγRs interactions.
Collapse
|
11
|
Afucosylated IgG Targets FcγRIV for Enhanced Tumor Therapy in Mice. Cancers (Basel) 2021; 13:cancers13102372. [PMID: 34069226 PMCID: PMC8156657 DOI: 10.3390/cancers13102372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Cancer treatments are increasingly based on therapeutic antibodies to clear tumors. While in vivo mouse models are useful to predict effectiveness of human antibodies it is not completely clear how useful these models are to test antibodies engineered with enhanced effector functions designed for humans. One of the changes considered for many new antibody-based drugs is the removal of fucose (resulting in afucosylated IgG) which enhances IgG-Fc receptor (FcγR) mediated effector functions in humans through FcγRIIIa. Here we show that afucosylated human IgG1 also have enhanced effector functions against peritoneal metastasis of melanoma cells in mice through the evolutionary related mouse FcγRIV. This shows that afucosylated human IgG is functionally recognized across species and shows that mouse tumor models can be used to assess the therapeutic potential of afucosylated IgG1. Abstract Promising strategies for maximizing IgG effector functions rely on the introduction of natural and non-immunogenic modifications. The Fc domain of IgG antibodies contains an N-linked oligosaccharide at position 297. Human IgG antibodies lacking the core fucose in this glycan have enhanced binding to human (FcγR) IIIa/b, resulting in enhanced antibody dependent cell cytotoxicity and phagocytosis through these receptors. However, it is not yet clear if glycan-enhancing modifications of human IgG translate into more effective treatment in mouse models. We generated humanized hIgG1-TA99 antibodies with and without core-fucose. C57Bl/6 mice that were injected intraperitoneally with B16F10-gp75 mouse melanoma developed significantly less metastasis outgrowth after treatment with afucosylated hIgG1-TA99 compared to mice treated with wildtype hhIgG1-TA99. Afucosylated human IgG1 showed stronger interaction with the murine FcγRIV, the mouse orthologue of human FcγRIIIa, indicating that this glycan change is functionally conserved between the species. In agreement with this, no significant differences were observed in tumor outgrowth in FcγRIV-/- mice treated with human hIgG1-TA99 with or without the core fucose. These results confirm the potential of using afucosylated therapeutic IgG to increase their efficacy. Moreover, we show that afucosylated human IgG1 antibodies act across species, supporting that mouse models can be suitable to test afucosylated antibodies.
Collapse
|
12
|
Brady JR, Love JC. Alternative hosts as the missing link for equitable therapeutic protein production. Nat Biotechnol 2021; 39:404-407. [PMID: 33782611 DOI: 10.1038/s41587-021-00884-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Joseph R Brady
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
13
|
Brooks SM, Alper HS. Applications, challenges, and needs for employing synthetic biology beyond the lab. Nat Commun 2021; 12:1390. [PMID: 33654085 PMCID: PMC7925609 DOI: 10.1038/s41467-021-21740-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Synthetic biology holds great promise for addressing global needs. However, most current developments are not immediately translatable to 'outside-the-lab' scenarios that differ from controlled laboratory settings. Challenges include enabling long-term storage stability as well as operating in resource-limited and off-the-grid scenarios using autonomous function. Here we analyze recent advances in developing synthetic biological platforms for outside-the-lab scenarios with a focus on three major application spaces: bioproduction, biosensing, and closed-loop therapeutic and probiotic delivery. Across the Perspective, we highlight recent advances, areas for further development, possibilities for future applications, and the needs for innovation at the interface of other disciplines.
Collapse
Affiliation(s)
- Sierra M Brooks
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hal S Alper
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
14
|
Cambay F, Raymond C, Brochu D, Gilbert M, Tu TM, Cantin C, Lenferink A, Grail M, Henry O, De Crescenzo G, Durocher Y. Impact of IgG1 N-glycosylation on their interaction with Fc gamma receptors. CURRENT RESEARCH IN IMMUNOLOGY 2020; 1:23-37. [PMID: 35493857 PMCID: PMC9040152 DOI: 10.1016/j.crimmu.2020.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 11/08/2022] Open
Abstract
The effector functions of the IgGs are modulated by the N-glycosylation of their Fc region. Particularly, the absence of core fucosylation is known to increase the affinity of IgG1s for the Fcγ receptor IIIa expressed by immune cells, in turn translating in an improvement in the antibody-dependent cellular cytotoxicity. However, the impact of galactosylation and sialylation is still debated in the literature. In this study, we have investigated the influence of high and low levels of core fucosylation, terminal galactosylation and terminal α2,6-sialylation of the Fc N-glycans of trastuzumab on its affinity for the FcγRIIIa. A large panel of antibody glycoforms (i.e., highly α2,6-sialylated or galactosylated IgG1s, with high or low levels of core fucosylation) were generated and characterized, while their interactions with the FcγRs were analysed by a robust surface plasmon resonance-based assay as well as in a cell-based reporter bioassay. Overall, IgG1 glycoforms with reduced fucosylation display a stronger affinity for the FcγRIIIa. In addition, fucosylation, and the presence of terminal galactose and sialic acids are shown to increase the affinity for the FcγRIIIa as compared to the agalactosylated forms. These observations perfectly translate in the response observed in our reporter bioassay. Rapid production in CHO cells of IgGs bearing defined and relevant N-glycans IgG1 N-glycosylation influence upon FcγRs binding studied in a robust SPR assay Excellent correlation between the EC50 from a cell-based assay and the affinities
Collapse
|
15
|
Huynh HH, Morita N, Sakamoto T, Katayama T, Miyakawa T, Tanokura M, Chiba Y, Shinkura R, Maruyama JI. Functional production of human antibody by the filamentous fungus Aspergillus oryzae. Fungal Biol Biotechnol 2020; 7:7. [PMID: 32514366 PMCID: PMC7257131 DOI: 10.1186/s40694-020-00098-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Monoclonal antibodies (mAbs) as biopharmaceuticals take a pivotal role in the current therapeutic applications. Generally mammalian cell lines, such as those derived from Chinese hamster ovaries (CHO), are used to produce the recombinant antibody. However, there are still concerns about the high cost and the risk of pathogenic contamination when using mammalian cells. Aspergillus oryzae, a filamentous fungus recognized as a GRAS (Generally Regarded As Safe) organism, has an ability to secrete a large amount of proteins into the culture supernatant, and thus the fungus has been used as one of the cost-effective microbial hosts for heterologous protein production. Pursuing this strategy the human anti-TNFα antibody adalimumab, one of the world's best-selling antibodies for the treatment of immune-mediated inflammatory diseases including rheumatoid arthritis, was chosen to produce the full length of mAbs by A. oryzae. Generally, N-glycosylation of the antibody affects immune effector functions such as antibody-dependent cell-mediated cytotoxicity (ADCC) via binding to the Fc receptor (FcγR) on immune cells. The CRISPR/Cas9 system was used to first delete the Aooch1 gene encoding a key enzyme for the hyper-mannosylation process in fungi to investigate the binding ability of antibody with FcγRIIIa. RESULTS Adalimumab was expressed in A. oryzae by the fusion protein system with α-amylase AmyB. The full-length adalimumab consisting of two heavy and two light chains was successfully produced in the culture supernatants. Among the producing strains, the highest amount of antibody was obtained from the ten-protease deletion strain (39.7 mg/L). Two-step purifications by Protein A and size-exclusion chromatography were applied to obtain the high purity sample for further analysis. The antigen-binding and TNFα neutralizing activities of the adalimumab produced by A. oryzae were comparable with those of a commercial product Humira®. No apparent binding with the FcγRIIIa was detected with the recombinant adalimumab even by altering the N-glycan structure using the Aooch1 deletion strain, which suggests only a little additional activity of immune effector functions. CONCLUSION These results demonstrated an alternative low-cost platform for human antibody production by using A. oryzae, possibly offering a reasonable expenditure for patient's welfare.
Collapse
Affiliation(s)
- Hung Hiep Huynh
- Department of Biotechnology, The University of Tokyo, Tokyo, Japan
| | - Naoki Morita
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Toshihiro Sakamoto
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Takuya Katayama
- Department of Biotechnology, The University of Tokyo, Tokyo, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan
| | - Takuya Miyakawa
- Department of Applied Biological Chemistry, The University of Tokyo, Tokyo, Japan
| | - Masaru Tanokura
- Department of Applied Biological Chemistry, The University of Tokyo, Tokyo, Japan
| | - Yasunori Chiba
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki Japan
| | - Reiko Shinkura
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan
- Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Jun-ichi Maruyama
- Department of Biotechnology, The University of Tokyo, Tokyo, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
16
|
ELISA assay employing epitope-specific monoclonal antibodies to quantify circulating HER2 with potential application in monitoring cancer patients undergoing therapy with trastuzumab. Sci Rep 2020; 10:3016. [PMID: 32080226 PMCID: PMC7033231 DOI: 10.1038/s41598-020-59630-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/02/2020] [Indexed: 11/08/2022] Open
Abstract
Circulating HER2 extracellular domain (HER2 ECD) levels were proposed as a surrogate for HER2 tissue expression to monitor breast cancer patients for early relapse or responses to standard or HER2-targeted therapies, such as the monoclonal antibody (mAb) trastuzumab. Currently, available commercial ELISA assays for HER2 ECD rely on antibodies recognizing undisclosed or unknown epitopes. In this work, two ELISA assays employing MGR2 and MGR3 epitope-specific mAbs for HER2 ECD were developed and validated, showing good assay precision and linearity of the dose-response signal within the dynamic range of 0.19-12.50 ng mL-1 and detection limits of 0.76 and 0.75 ng mL-1 for the MGR2 and MGR3 assays, respectively. The developed assay showed a good agreement with two widely used commercial kits for HER2 ECD quantification in serum samples from breast cancer patients. A complete characterization of mAb-HER2 ECD interaction was performed by means of surface plasmon resonance using trastuzumab as control for both epitope mapping and kinetics analysis. The epitopes recognized by the two mAbs showed no overlap with trastuzumab, which was confirmed by trastuzumab interference analysis in serum samples. The method showed to be a practical approach to determine HER2 ECD with a high degree of sensitivity, reliability and recovery in samples containing mAbs-based therapies.
Collapse
|
17
|
Dalvie NC, Leal J, Whittaker CA, Yang Y, Brady JR, Love KR, Love JC. Host-Informed Expression of CRISPR Guide RNA for Genomic Engineering in Komagataella phaffii. ACS Synth Biol 2020; 9:26-35. [PMID: 31825599 PMCID: PMC7814401 DOI: 10.1021/acssynbio.9b00372] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There is growing interest in the use of nonmodel microorganisms as hosts for biopharmaceutical manufacturing. These hosts require genomic engineering to meet clinically relevant product qualities and titers, but the adaptation of tools for editing genomes, such as CRISPR-Cas9, has been slow for poorly characterized hosts. Specifically, a lack of biochemical characterization of RNA polymerase III transcription has hindered reliable expression of guide RNAs in new hosts. Here, we present a sequencing-based strategy for the design of host-specific cassettes for modular, reliable, expression of guide RNAs. Using this strategy, we achieved up to 95% gene editing efficiency in the methylotrophic yeast Komagataella phaffii. We applied this approach for the rapid, multiplexed engineering of a complex phenotype, achieving humanized product glycosylation in two sequential steps of engineering. Reliable extension of simple gene editing tools to nonmodel manufacturing hosts will enable rapid engineering of manufacturing strains tuned for specific product profiles and potentially decrease the costs and timelines for process development.
Collapse
Affiliation(s)
- Neil C. Dalvie
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 01239, United States
| | - Justin Leal
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 01239, United States
| | - Charles A. Whittaker
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 01239, United States
| | - Yuchen Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 01239, United States
| | - Joseph R. Brady
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 01239, United States
| | - Kerry R. Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 01239, United States
| | - J. Christopher Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 01239, United States
| |
Collapse
|
18
|
Antibody glycosylation: impact on antibody drug characteristics and quality control. Appl Microbiol Biotechnol 2020; 104:1905-1914. [DOI: 10.1007/s00253-020-10368-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/02/2020] [Accepted: 01/09/2020] [Indexed: 12/21/2022]
|
19
|
Bozovičar K, Bratkovič T. Evolving a Peptide: Library Platforms and Diversification Strategies. Int J Mol Sci 2019; 21:E215. [PMID: 31892275 PMCID: PMC6981544 DOI: 10.3390/ijms21010215] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/22/2019] [Accepted: 12/25/2019] [Indexed: 12/22/2022] Open
Abstract
Peptides are widely used in pharmaceutical industry as active pharmaceutical ingredients, versatile tools in drug discovery, and for drug delivery. They find themselves at the crossroads of small molecules and proteins, possessing favorable tissue penetration and the capability to engage into specific and high-affinity interactions with endogenous receptors. One of the commonly employed approaches in peptide discovery and design is to screen combinatorial libraries, comprising a myriad of peptide variants of either chemical or biological origin. In this review, we focus mainly on recombinant peptide libraries, discussing different platforms for their display or expression, and various diversification strategies for library design. We take a look at well-established technologies as well as new developments and future directions.
Collapse
Affiliation(s)
| | - Tomaž Bratkovič
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, SI-1000 Ljubljana, Slovenia;
| |
Collapse
|
20
|
Tackling Cancer with Yeast-Based Technologies. Trends Biotechnol 2019; 37:592-603. [DOI: 10.1016/j.tibtech.2018.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/24/2018] [Accepted: 11/30/2018] [Indexed: 12/19/2022]
|
21
|
Zhang Y, Fan C, Zhang L, Ma X. Glycosylation-dependent antitumor therapeutic monoclonal antibodies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 163:471-485. [PMID: 31030759 DOI: 10.1016/bs.pmbts.2019.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The therapeutic market for monoclonal antibodies (MAbs) has grown exponentially since 2000. It is expected that the world-wide market for MAbs could reach $125 billion in 2020. For cancer treatment alone, more than 30 MAbs have been approved by the US Food and Drug Administration since 1997. Unlike structure-defined small molecule-based anti-cancer drugs, the expensive MAb is a mixture of heterogeneously glycosylated proteins. All MAbs typically have a single N-glycosylation site on each of the Fc region. The clinical efficacy of the MAbs depends on the N-glycan structures. Loss of N-glycosylation on the MAbs leads to the loss of the ability to activate complement, to bind to Fc receptors, and to induce antibody-dependent cellular cytotoxicity (ADCC). Moreover, antigen-antibody complexes produced from N-glycan-deficient MAbs are failed to be eliminated rapidly from the blood circulation. Even in certain cases, the N-glycan heterogeneity does not significantly influence pharmacokinetics or half-life of MAbs, reduced terminal galactosylation decreases complement-dependent cytotoxicity, the absence of core fucosylation enhances ADCC due to the increased affinities for the FcγRIIIа receptor, and high sialylation levels reduce ADCC activity and impact inflammatory responses. Furthermore, only mammalian cell lines that make human-like N-glycan structures can be used for MAbs production since certain mammalian cell lines can produce non-human glycan epitopes such as galactose-α-1,3-galactose and N-glycolylneuraminic acid (NGNA), which can trigger unwanted immune response. Therefore, mastering the knowledge of N-glycan structures and glycobiology is the key to produce and provide patients with reliable MAbs with consistent glycosylation profile and expected clinical efficacy.
Collapse
Affiliation(s)
- Yiran Zhang
- Systems Biology and Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, China; Department of Orthopedics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chun Fan
- Department of Stomatology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lijuan Zhang
- Systems Biology and Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xuexiao Ma
- Department of Orthopedics, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
22
|
Khan AH, Noordin R. Strategies for humanizing glycosylation pathways and producing recombinant glycoproteins in microbial expression systems. Biotechnol Prog 2018; 35:e2752. [DOI: 10.1002/btpr.2752] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 10/26/2018] [Accepted: 11/16/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Amjad Hayat Khan
- Inst. for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia 11800 Penang Malaysia
| | - Rahmah Noordin
- Inst. for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia 11800 Penang Malaysia
| |
Collapse
|
23
|
The yeast stands alone: the future of protein biologic production. Curr Opin Biotechnol 2018; 53:50-58. [DOI: 10.1016/j.copbio.2017.12.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/13/2022]
|
24
|
Zhang S, Mukherjee S, Fan X, Salameh A, Mujoo K, Huang Z, Li L, To'a Salazar G, Zhang N, An Z. Novel association of DJ-1 with HER3 potentiates HER3 activation and signaling in cancer. Oncotarget 2018; 7:65758-65769. [PMID: 27582551 PMCID: PMC5323190 DOI: 10.18632/oncotarget.11613] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/15/2016] [Indexed: 01/01/2023] Open
Abstract
HER3/ErbB3 has emerged as a new therapeutic target for cancer. Currently, more than a dozen anti-HER3 antibodies are in clinical trials for treatment of various cancers. However, limited understanding of the complex HER3 signaling in cancer and lack of established biomarkers have made it challenging to stratify cancer patients who can benefit from HER3 targeted therapies. In this study, we identified DJ-1/PARK7 (Parkinson Protein 7) as a novel interaction partner of HER3 and demonstrated the potential of DJ-1 as a biomarker for anti-HER3 cancer therapy. DJ-1 association with HER3 protects HER3 from ubiquitination and degradation through the proteasomal pathway in breast cancer cells. However, neuregulin 1 (NRG-1) mediated HER3 activation results in a reduced association of DJ-1 with HER3. DJ-1 shRNA knockdown in cancer cells resulted in decreased levels of HER3 and its downstream signaling through the PI3K/AKT and Ras/Raf/ERK pathways. DJ-1 shRNA knockdown cancer cells significantly reduced cell proliferation and migration in vitro and tumor growth in vivo. Conversely, overexpression of DJ-1 increased HER3 levels and promoted cancer cell proliferation in vitro and tumor growth in vivo. Notably, cancer cells with high DJ-1 expression showed more sensitivity than DJ-1 knockdown cells to anti-HER3 antibody inhibition. In addition, there was a significant co-expression of HER3 and DJ-1 in tumor tissues of breast cancer patients. Taken together, these results suggest that high DJ-1 expression in breast cancer cells predicts elevated HER3 signaling and may therefore serve as a biomarker for HER3 targeted antibody cancer therapies.
Collapse
Affiliation(s)
- Shu Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Current address: Clinical Research Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Seema Mukherjee
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xuejun Fan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ahmad Salameh
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kalpana Mujoo
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Current address: Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas, USA
| | - Zhao Huang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Current address: Stemcentrx, Inc., South San Francisco, California, USA
| | - Leike Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Georgina To'a Salazar
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
25
|
Cymer F, Beck H, Rohde A, Reusch D. Therapeutic monoclonal antibody N-glycosylation – Structure, function and therapeutic potential. Biologicals 2018; 52:1-11. [DOI: 10.1016/j.biologicals.2017.11.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/13/2017] [Accepted: 11/14/2017] [Indexed: 12/25/2022] Open
|
26
|
Egashira Y, Nagatoishi S, Kiyoshi M, Ishii-Watabe A, Tsumoto K. Characterization of glycoengineered anti-HER2 monoclonal antibodies produced by using a silkworm–baculovirus expression system. J Biochem 2018; 163:481-488. [DOI: 10.1093/jb/mvy021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 12/23/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yuriko Egashira
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
- Protein Development Center, Sysmex Corporation, Sayama, Saitama 350-1332, Japan
| | - Satoru Nagatoishi
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
- Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Masato Kiyoshi
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
- Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
27
|
Glycoengineering of antibody (Herceptin) through yeast expression and in vitro enzymatic glycosylation. Proc Natl Acad Sci U S A 2018; 115:720-725. [PMID: 29311294 DOI: 10.1073/pnas.1718172115] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Monoclonal antibodies (mAbs) have been developed as therapeutics, especially for the treatment of cancer, inflammation, and infectious diseases. Because the glycosylation of mAbs in the Fc region influences their interaction with effector cells that kill antibody-targeted cells, and the current method of antibody production is relatively expensive, efforts have been directed toward the development of alternative expressing systems capable of large-scale production of mAbs with desirable glycoforms. In this study, we demonstrate that the mAb trastuzumab expressed in glycoengineered P. pastoris can be remodeled through deglycosylation by endoglycosidases identified from the Carbohydrate Active Enzymes database and through transglycosylation using glycans with a stable leaving group to generate a homogeneous antibody designed to optimize the effector functions. The 10 newly identified recombinant bacterial endoglycosidases are complementary to existing endoglycosidases (EndoA, EndoH, EndoS), two of which can even accept sialylated tri- and tetraantennary glycans as substrates.
Collapse
|
28
|
Engineering of Yeast Glycoprotein Expression. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 175:93-135. [DOI: 10.1007/10_2018_69] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Abstract
The methylotrophic yeast Pichia pastoris has become an increasingly popular host for recombinant protein expression in recent times. MRL pioneered a glycoengineered humanized P. pastoris expression system that could produce glycoproteins with glycosylation profiles similar to mammalian systems. Therapeutic glycoproteins produced by the humanized P. pastoris platform have shown comparable folding, stability, and in vitro and in vivo efficacies in preclinical models to their counterparts produced from the CHO cells. P. pastoris offers a cost and time efficient alternative platform for therapeutic protein production. This chapter describes a protocol for using P. pastoris to produce full-length monoclonal antibodies. It covers a broad spectrum of antibody expression technologies in P. pastoris, including expression vector construction, yeast transformation, high-throughput strain selection, fermentation, and antibody purification.
Collapse
Affiliation(s)
- Adam Nylen
- Biologics Discovery, MRL, 33 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Ming-Tang Chen
- Biologics Discovery, MRL, 33 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| |
Collapse
|
30
|
Purcell O, Opdensteinen P, Chen W, Lowenhaupt K, Brown A, Hermann M, Cao J, Tenhaef N, Kallweit E, Kastilan R, Sinskey AJ, Perez-Pinera P, Buyel JF, Lu TK. Production of Functional Anti-Ebola Antibodies in Pichia pastoris. ACS Synth Biol 2017; 6:2183-2190. [PMID: 28786662 DOI: 10.1021/acssynbio.7b00234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The 2013-2016 Ebola outbreak highlighted the limited treatment options and lack of rapid response strategies for emerging pathogen outbreaks. Here, we propose an efficient development cycle using glycoengineered Pichia pastoris to produce monoclonal antibody cocktails against pathogens. To enable rapid genetic engineering of P. pastoris, we introduced a genomic landing pad for reliable recombinase-mediated DNA integration. We then created strains expressing each of the three monoclonal antibodies that comprise the ZMapp cocktail, and demonstrated that the secreted antibodies bind to the Ebola virus glycoprotein by immunofluorescence assay. We anticipate that this approach could accelerate the production of therapeutics against future pathogen outbreaks.
Collapse
Affiliation(s)
- Oliver Purcell
- Synthetic
Biology Center, Department of Electrical Engineering and Computer
Science, Department of Biological Engineering, 500 Technology Square, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Patrick Opdensteinen
- Department
of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraβe 6, 52074 Aachen, Germany
| | - William Chen
- Synthetic
Biology Center, Department of Electrical Engineering and Computer
Science, Department of Biological Engineering, 500 Technology Square, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Ky Lowenhaupt
- Synthetic
Biology Center, Department of Electrical Engineering and Computer
Science, Department of Biological Engineering, 500 Technology Square, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Alexander Brown
- Department
of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Mario Hermann
- Synthetic
Biology Center, Department of Electrical Engineering and Computer
Science, Department of Biological Engineering, 500 Technology Square, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jicong Cao
- Synthetic
Biology Center, Department of Electrical Engineering and Computer
Science, Department of Biological Engineering, 500 Technology Square, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Niklas Tenhaef
- Department
of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Eric Kallweit
- Department
of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Robin Kastilan
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraβe 6, 52074 Aachen, Germany
| | - Anthony J. Sinskey
- Department
of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Pablo Perez-Pinera
- Department
of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Johannes F. Buyel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraβe 6, 52074 Aachen, Germany
- Institute
for Molecular Biotechnology, RWTH Aachen University, Worringerweg
1, 52074 Aachen, Germany
| | - Timothy K. Lu
- Synthetic
Biology Center, Department of Electrical Engineering and Computer
Science, Department of Biological Engineering, 500 Technology Square, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
31
|
The state-of-play and future of antibody therapeutics. Adv Drug Deliv Rev 2017; 122:2-19. [PMID: 27916504 DOI: 10.1016/j.addr.2016.11.004] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/26/2016] [Accepted: 11/28/2016] [Indexed: 12/22/2022]
Abstract
It has been over four decades since the development of monoclonal antibodies (mAbs) using a hybridoma cell line was first reported. Since then more than thirty therapeutic antibodies have been marketed, mostly as oncology, autoimmune and inflammatory therapeutics. While antibodies are very efficient, their cost-effectiveness has always been discussed owing to their high costs, accumulating to more than one billion dollars from preclinical development through to market approval. Because of this, therapeutic antibodies are inaccessible to some patients in both developed and developing countries. The growing interest in biosimilar antibodies as affordable versions of therapeutic antibodies may provide alternative treatment options as well potentially decreasing costs. As certain markets begin to capitalize on this opportunity, regulatory authorities continue to refine the requirements for demonstrating quality, efficacy and safety of biosimilar compared to originator products. In addition to biosimilars, innovations in antibody engineering are providing the opportunity to design biobetter antibodies with improved properties to maximize efficacy. Enhancing effector function, antibody drug conjugates (ADC) or targeting multiple disease pathways via multi-specific antibodies are being explored. The manufacturing process of antibodies is also moving forward with advancements relating to host cell production and purification processes. Studies into the physical and chemical degradation pathways of antibodies are contributing to the design of more stable proteins guided by computational tools. Moreover, the delivery and pharmacokinetics of antibody-based therapeutics are improving as optimized formulations are pursued through the implementation of recent innovations in the field.
Collapse
|
32
|
Royle KE, Polizzi K. A streamlined cloning workflow minimising the time-to-strain pipeline for Pichia pastoris. Sci Rep 2017; 7:15817. [PMID: 29150665 PMCID: PMC5693959 DOI: 10.1038/s41598-017-16172-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/08/2017] [Indexed: 02/06/2023] Open
Abstract
Although recent advances in E. coli self-assembly have greatly simplified cloning, these have not yet been harnessed for the high-throughput generation of expression strains in the early research and discovery phases of biopharmaceutical production. Here, we have refined the technique and incorporated it into a streamlined workflow for the generation of Pichia pastoris expression strains, reducing the timeline by a third and removing the reliance on DNA editing enzymes, which often require troubleshooting and increase costs. We have validated the workflow by cloning 24 human proteins of biopharmaceutical value, either as direct therapeutics or as research targets, which span a continuous range in size and GC content. This includes demonstrating the applicability of the workflow to three-part assemblies for a monoclonal antibody and its single-chain antibody fragments derivatives. This workflow should enable future research into recombinant protein production by P. pastoris and a synthetic biology approach to this industrial host.
Collapse
Affiliation(s)
- Kate E Royle
- Department of Life Sciences, Imperial College London, London, UK.,Centre for Synthetic Biology and Innovation, Imperial College London, London, UK
| | - Karen Polizzi
- Department of Life Sciences, Imperial College London, London, UK. .,Centre for Synthetic Biology and Innovation, Imperial College London, London, UK.
| |
Collapse
|
33
|
Qiao C, Lang X, Luo L, Geng S, Lv M, Geng J, Li X, Feng J, Shen B, Li Y. Multi-parametric analysis reveals enhanced G2-phase arrest of an optimized anti-HER2 antibody to inhibit breast cancer. Biotechnol Lett 2017; 39:1309-1323. [PMID: 28560579 DOI: 10.1007/s10529-017-2364-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/25/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To find a "me-better" antibody by epitope-specific antibody optimization and multi-parametric analysis. RESULTS Using epitope-specific library based on the commercial drug, Pertuzumab/2C4, we screened a novel human anti-HER2 antibody, MIL5, which has slightly higher affinity than the drug. MIL5 and 2C4 share the same epitope to bind HER2; however, MIL5 bound to HER2 His235-His245 more tightly than 2C4, which could be the main reason of its enhanced affinity. In vivo experiments also showed MIL5 had stronger anti-cancer activity than 2C4; however, the classical flow cytometry assays to detect cell apoptosis or cycling did not show convincing evidence of the advantages of MIL5. Thus we introduced the multi-parameter in-cell analysis method to evaluate the superiority of MIL5 to 2C4 in arresting cancer cells in G2-phase to inhibit cell growth and/or proliferation. CONCLUSION Multi-parametric method confirmed stronger arrest of G2 by MIL5 to show better anti-cancer function both in vitro and in vivo than 2C4.
Collapse
Affiliation(s)
- Chunxia Qiao
- Laboratory of Molecular Immunology, Institute of Basic Medical Sciences, P. O. Box 130 (3), Taiping Road #27 Haidian, Beijing, 100850, China
| | - Xiaoling Lang
- Economic-Technological Development Area, Beijing Mabworks Biotech Company Ltd., Beijing, 101111, China
| | - Longlong Luo
- Laboratory of Molecular Immunology, Institute of Basic Medical Sciences, P. O. Box 130 (3), Taiping Road #27 Haidian, Beijing, 100850, China
| | - Shusheng Geng
- Economic-Technological Development Area, Beijing Mabworks Biotech Company Ltd., Beijing, 101111, China
| | - Ming Lv
- Laboratory of Molecular Immunology, Institute of Basic Medical Sciences, P. O. Box 130 (3), Taiping Road #27 Haidian, Beijing, 100850, China
| | - Jing Geng
- Laboratory of Molecular Immunology, Institute of Basic Medical Sciences, P. O. Box 130 (3), Taiping Road #27 Haidian, Beijing, 100850, China
| | - Xinying Li
- Laboratory of Molecular Immunology, Institute of Basic Medical Sciences, P. O. Box 130 (3), Taiping Road #27 Haidian, Beijing, 100850, China
| | - Jiannan Feng
- Laboratory of Molecular Immunology, Institute of Basic Medical Sciences, P. O. Box 130 (3), Taiping Road #27 Haidian, Beijing, 100850, China
| | - Beifen Shen
- Laboratory of Molecular Immunology, Institute of Basic Medical Sciences, P. O. Box 130 (3), Taiping Road #27 Haidian, Beijing, 100850, China
| | - Yan Li
- Laboratory of Molecular Immunology, Institute of Basic Medical Sciences, P. O. Box 130 (3), Taiping Road #27 Haidian, Beijing, 100850, China. .,Economic-Technological Development Area, Beijing Mabworks Biotech Company Ltd., Beijing, 101111, China.
| |
Collapse
|
34
|
A shear-enhanced CNT-assembly nanosensor platform for ultra-sensitive and selective protein detection. Biosens Bioelectron 2017; 97:143-149. [PMID: 28587929 DOI: 10.1016/j.bios.2017.05.053] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/15/2017] [Accepted: 05/29/2017] [Indexed: 01/04/2023]
Abstract
Detection and quantification of low-concentration proteins in heterogeneous media are generally plagued by two distinct obstacles: lack of sensitivity due to high dissociation equilibrium constant KD and non-specificity due to an abundance of non-targets with similar KD. Herein, we report a nanoscale protein-sensing platform with a non-equilibrium on-off switch that employs dielectrophoretic and hydrodynamic shear forces to overcome these thermodynamic limitations with irreversible kinetics. The detection sensitivity is achieved with complete association of the antibody-antigen-antibody (Ab-Ag-Ab) complex by precisely and rapidly assembling carbon nanotubes (CNT) across two parallel electrodes via sequential DC electrophoresis and AC dielectrophoresis (DEP), and with single-CNT electron tunneling conductance. The high selectivity is achieved with a critical hydrodynamic shear rate between the activated dissociation shear rates of target and non-target linkers of the aligned CNTs. We are able to reach detection limits of 100 attomolar (aM) and 10 femtomolar (fM) in pure samples for two ELISA assays with low and high dissociation constant: biotin/streptavidin (10 fM) and HER2/HER2 antibody (0.44 ± 0.07nM), respectively. For both models, irreversible capture and shearing allow us to tune the dynamic range up to 5 decades by increasing the CNT numbers. We also demonstrate in spiked serum sample high selectivity towards target HER2 proteins against non-target HER2 isoform of a similar KD. The detection limit for HER2 in serum is lower than 100fM.
Collapse
|
35
|
Suzuki T, Baba S, Ono M, Nonaka K, Ichikawa K, Yabuta M, Ito R, Chiba Y. Efficient antibody production in the methylotrophic yeast Ogataea minuta by overexpression of chaperones. J Biosci Bioeng 2017; 124:156-163. [PMID: 28356218 DOI: 10.1016/j.jbiosc.2017.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 01/20/2023]
Abstract
A production system for a therapeutic monoclonal antibody was developed using the methylotrophic yeast Ogataea minuta IFO10746. The genetically engineered O. minuta secreted a detectable amount of anti-TRAIL receptor antibody into the culture supernatant, and the secreted antibody was purified by multiple column chromatography steps. In the purification process, both fully and partially assembled antibodies were detected and isolated. The fully assembled antibody from O. minuta showed almost the same biological activity as that derived from mammalian cells despite the distinct glycosylation profile, whereas the partially assembled antibody showed no cytotoxic activity. To increase the production of active antibody in O. minuta, we overexpressed selected chaperone proteins (included protein disulfide isomerase (OmPDI1), thiol oxidase (OmERO1), and immunoglobulin heavy chain binding protein (OmKAR2)) known to assist in the proper folding (in the endoplasmic reticulum) of proteins destined for secretion. Each of these chaperones enhanced antibody secretion, and together these three factors yielded 16-fold higher antibody accumulation while increasing the ratio of the fully assembled antibody compared to that from the parental strain. Supplementation of a rhodanine-3-acetic acid derivative (R3AD_1c), an inhibitor of O-mannosylation, further increased the secretion of the correctly assembled antibody. These results indicated that the co-overexpression of chaperones is an effective way to produce the correctly assembled antibody in O. minuta.
Collapse
Affiliation(s)
- Takeshi Suzuki
- Daiichi Sankyo Co., Ltd., Biologics Technology Research Laboratories, 2716-1 Kurakake, Akaiwa, Chiyoda, Gunma 370-0503, Japan
| | - Satoshi Baba
- Daiichi Sankyo Co., Ltd., Biologics Technology Research Laboratories, 2716-1 Kurakake, Akaiwa, Chiyoda, Gunma 370-0503, Japan
| | - Minako Ono
- Daiichi Sankyo Chemical Pharma Co., Ltd., 389-4 Ohtsurugi, Shimokawa, Izumi, Iwaki, Fukushima 971-8183, Japan
| | - Koichi Nonaka
- Daiichi Sankyo Co., Ltd., Biologics Technology Research Laboratories, 2716-1 Kurakake, Akaiwa, Chiyoda, Gunma 370-0503, Japan
| | - Kimihisa Ichikawa
- Daiichi Sankyo Co., Ltd., Biologics Technology Research Laboratories, 2716-1 Kurakake, Akaiwa, Chiyoda, Gunma 370-0503, Japan
| | - Masayuki Yabuta
- Daiichi Sankyo Co., Ltd., Biologics Technology Research Laboratories, 2716-1 Kurakake, Akaiwa, Chiyoda, Gunma 370-0503, Japan
| | - Rie Ito
- Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Yasunori Chiba
- Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.
| |
Collapse
|
36
|
Calow J, Behrens AJ, Mader S, Bockau U, Struwe WB, Harvey DJ, Cormann KU, Nowaczyk MM, Loser K, Schinor D, Hartmann MWW, Crispin M. Antibody production using a ciliate generates unusual antibody glycoforms displaying enhanced cell-killing activity. MAbs 2016; 8:1498-1511. [PMID: 27594301 PMCID: PMC5098438 DOI: 10.1080/19420862.2016.1228504] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Antibody glycosylation is a key parameter in the optimization of antibody therapeutics. Here, we describe the production of the anti-cancer monoclonal antibody rituximab in the unicellular ciliate, Tetrahymena thermophila. The resulting antibody demonstrated enhanced antibody-dependent cell-mediated cytotoxicity, which we attribute to unusual N-linked glycosylation. Detailed chromatographic and mass spectrometric analysis revealed afucosylated, oligomannose-type glycans, which, as a whole, displayed isomeric structures that deviate from the typical human counterparts, but whose branches were equivalent to fragments of metabolic intermediates observed in human glycoproteins. From the analysis of deposited crystal structures, we predict that the ciliate glycans adopt protein-carbohydrate interactions with the Fc domain that closely mimic those of native complex-type glycans. In addition, terminal glucose structures were identified that match biosynthetic precursors of human glycosylation. Our results suggest that ciliate-based expression systems offer a route to large-scale production of monoclonal antibodies exhibiting glycosylation that imparts enhanced cell killing activity.
Collapse
Affiliation(s)
| | - Anna-Janina Behrens
- b Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford , Oxford , UK
| | | | | | - Weston B Struwe
- b Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford , Oxford , UK
| | - David J Harvey
- b Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford , Oxford , UK
| | - Kai U Cormann
- c Plant Biochemistry, Ruhr University Bochum , Bochum , Germany
| | - Marc M Nowaczyk
- c Plant Biochemistry, Ruhr University Bochum , Bochum , Germany
| | - Karin Loser
- d Department of Dermatology , University of Münster , Münster , Germany
| | - Daniel Schinor
- e Wessling GmbH, Pharmaanalytik Münster , Münster , Germany
| | | | - Max Crispin
- b Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford , Oxford , UK
| |
Collapse
|
37
|
Kim HJ, Kim A, Miyata K, Kataoka K. Recent progress in development of siRNA delivery vehicles for cancer therapy. Adv Drug Deliv Rev 2016; 104:61-77. [PMID: 27352638 DOI: 10.1016/j.addr.2016.06.011] [Citation(s) in RCA: 329] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 05/21/2016] [Accepted: 06/16/2016] [Indexed: 12/13/2022]
Abstract
Recent progress in RNA biology has broadened the scope of therapeutic targets of RNA drugs for cancer therapy. However, RNA drugs, typically small interfering RNAs (siRNAs), are rapidly degraded by RNases and filtrated in the kidney, thereby requiring a delivery vehicle for efficient transport to the target cells. To date, various delivery formulations have been developed from cationic lipids, polymers, and/or inorganic nanoparticles for systemic delivery of siRNA to solid tumors. This review describes the current status of clinical trials related to siRNA-based cancer therapy, as well as the remaining issues that need to be overcome to establish a successful therapy. It, then introduces various promising design strategies of delivery vehicles for stable and targeted siRNA delivery, including the prospects for future design.
Collapse
|
38
|
Perez-Pinera P, Han N, Cleto S, Cao J, Purcell O, Shah KA, Lee K, Ram R, Lu TK. Synthetic biology and microbioreactor platforms for programmable production of biologics at the point-of-care. Nat Commun 2016; 7:12211. [PMID: 27470089 PMCID: PMC4974573 DOI: 10.1038/ncomms12211] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 06/07/2016] [Indexed: 11/10/2022] Open
Abstract
Current biopharmaceutical manufacturing systems are not compatible with portable or distributed production of biologics, as they typically require the development of single biologic-producing cell lines followed by their cultivation at very large scales. Therefore, it remains challenging to treat patients in short time frames, especially in remote locations with limited infrastructure. To overcome these barriers, we developed a platform using genetically engineered Pichia pastoris strains designed to secrete multiple proteins on programmable cues in an integrated, benchtop, millilitre-scale microfluidic device. We use this platform for rapid and switchable production of two biologics from a single yeast strain as specified by the operator. Our results demonstrate selectable and near-single-dose production of these biologics in <24 h with limited infrastructure requirements. We envision that combining this system with analytical, purification and polishing technologies could lead to a small-scale, portable and fully integrated personal biomanufacturing platform that could advance disease treatment at point-of-care.
Collapse
Affiliation(s)
- Pablo Perez-Pinera
- Synthetic Biology Group, Department of Biological Engineering and Electrical Engineering &Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Ningren Han
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Sara Cleto
- Synthetic Biology Group, Department of Biological Engineering and Electrical Engineering &Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Jicong Cao
- Synthetic Biology Group, Department of Biological Engineering and Electrical Engineering &Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Oliver Purcell
- Synthetic Biology Group, Department of Biological Engineering and Electrical Engineering &Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Kartik A Shah
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Kevin Lee
- Pharyx Inc., Woburn, Massachusetts 01801, USA
| | - Rajeev Ram
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Timothy K Lu
- Synthetic Biology Group, Department of Biological Engineering and Electrical Engineering &Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02412, USA
| |
Collapse
|
39
|
Wronska MA, O'Connor IB, Tilbury MA, Srivastava A, Wall JG. Adding Functions to Biomaterial Surfaces through Protein Incorporation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5485-5508. [PMID: 27164952 DOI: 10.1002/adma.201504310] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/16/2016] [Indexed: 06/05/2023]
Abstract
The concept of biomaterials has evolved from one of inert mechanical supports with a long-term, biologically inactive role in the body into complex matrices that exhibit selective cell binding, promote proliferation and matrix production, and may ultimately become replaced by newly generated tissues in vivo. Functionalization of material surfaces with biomolecules is critical to their ability to evade immunorecognition, interact productively with surrounding tissues and extracellular matrix, and avoid bacterial colonization. Antibody molecules and their derived fragments are commonly immobilized on materials to mediate coating with specific cell types in fields such as stent endothelialization and drug delivery. The incorporation of growth factors into biomaterials has found application in promoting and accelerating bone formation in osteogenerative and related applications. Peptides and extracellular matrix proteins can impart biomolecule- and cell-specificities to materials while antimicrobial peptides have found roles in preventing biofilm formation on devices and implants. In this progress report, we detail developments in the use of diverse proteins and peptides to modify the surfaces of hard biomaterials in vivo and in vitro. Chemical approaches to immobilizing active biomolecules are presented, as well as platform technologies for isolation or generation of natural or synthetic molecules suitable for biomaterial functionalization.
Collapse
Affiliation(s)
- Małgorzata A Wronska
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Iain B O'Connor
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Maura A Tilbury
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Akshay Srivastava
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - J Gerard Wall
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| |
Collapse
|
40
|
Bull-Hansen B, Berstad MB, Berg K, Cao Y, Skarpen E, Fremstedal AS, Rosenblum MG, Peng Q, Weyergang A. Photochemical activation of MH3-B1/rGel: a HER2-targeted treatment approach for ovarian cancer. Oncotarget 2016; 6:12436-51. [PMID: 26002552 PMCID: PMC4494949 DOI: 10.18632/oncotarget.3814] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 03/11/2015] [Indexed: 11/28/2022] Open
Abstract
HER2-targeted therapy has been shown to have limited efficacy in ovarian cancer despite frequent overexpression of this receptor. Photochemical internalization (PCI) is a modality for cytosolic drug delivery, currently undergoing clinical evaluation. In the present project we studied the application of PCI in combination with the HER2-targeted recombinant fusion toxin, MH3-B1/rGel, for the treatment of ovarian cancer. The SKOV-3 cell line, resistant to trastuzumab- and MH3-B1/rGel- monotherapy, was shown to respond strongly to PCI of MH3-B1/rGel to a similar extent as observed for the treatment-sensitive SK-BR-3 breast cancer cells. Extensive hydrolytic degradation of MH3-B1/rGel in acidic endocytic vesicles was indicated as the mechanism of MH3-B1/rGel resistance in SKOV-3 cells. This was shown by the positive Pearson's correlation coefficient between Alexa488-labeled MH3-B1/rGel and Lysotracker in SKOV-3 cells in contrast to the negative Pearson's correlation coefficient in SK-BR-3 cells. The application of PCI to induce the release of MH3-B1/rGel was also demonstrated to be effective on SKOV-3 xenografts. Application of PCI with MH3-B1/rGel was further found highly effective in the HER2 expressing HOC-7 and NuTu-19 ovarian cancer cell lines. The presented results warrant future development of PCI in combination with MH3-B1/rGel as a novel therapeutic approach in preclinical models of ovarian cancer.
Collapse
Affiliation(s)
- Bente Bull-Hansen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Maria B Berstad
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kristian Berg
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Yu Cao
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA.,Current address: The Scripps Research Institute, Department of Chemistry, La Jolla, CA, USA
| | - Ellen Skarpen
- Department of Biochemistry, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ane Sofie Fremstedal
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Michael G Rosenblum
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - Qian Peng
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Anette Weyergang
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
41
|
Enhanced truncated-t-PA (CT-b) expression in high-cell-density fed-batch cultures of Pichia pastoris through optimization of a mixed feeding strategy by response surface methodology. Bioprocess Biosyst Eng 2016; 39:565-73. [PMID: 26758714 DOI: 10.1007/s00449-016-1538-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 01/06/2016] [Indexed: 12/27/2022]
Abstract
Recently, Pichia pastoris has been the focal point of interest as an expression system for production of many recombinant proteins. The study and optimization of feeding strategy are of major importance to achieve maximum volumetric productivity in fed-batch cultivations. Among different feeding strategies used in P. pastoris fed-batch cultures, those trying to maintain a constant specific growth rate have usually resulted in superior productivities. The objective of the present study was to investigate and optimize the co-feeding of glycerol and methanol to attain maximum expression of t-PA in P. pastoris fed-batch cultures with constant specific growth rate. The experiments were designed by response surface methodology, considering the specific feeding rates of methanol and glycerol as independent variables. In each experiment, glycerol and methanol were fed according to a predetermined equation to maintain a constant specific growth rate. It was found that with glycerol feeding for higher specific growth rates, the inhibitory properties of glycerol are more pronounced, while the best expression level was achieved when the ratio of µ set glycerol to that of methanol was around 1.67. In all specific growth rates tested, almost a similar ratio of the specific glycerol feeding rate to that of methanol led to the maximum protein production and activity. The statistical model predicted the optimal operating conditions for µ set glycerol and that of methanol to be 0.05 and 0.03 h(-1), respectively. Applying the optimum strategy, maximum of 52 g/L biomass, 300 mg/L t-PA and 340,000 IU/mL enzyme activity were obtained.
Collapse
|
42
|
Challenges to production of antibodies in bacteria and yeast. J Biosci Bioeng 2015; 120:483-90. [DOI: 10.1016/j.jbiosc.2015.03.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/12/2015] [Accepted: 03/12/2015] [Indexed: 12/21/2022]
|
43
|
Rouwendal GJ, van der Lee MM, Meyer S, Reiding KR, Schouten J, de Roo G, Egging DF, Leusen JH, Boross P, Wuhrer M, Verheijden GF, Dokter WH, Timmers M, Ubink R. A comparison of anti-HER2 IgA and IgG1 in vivo efficacy is facilitated by high N-glycan sialylation of the IgA. MAbs 2015; 8:74-86. [PMID: 26440530 DOI: 10.1080/19420862.2015.1102812] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Monomeric IgA has been proposed as an alternative antibody format for cancer therapy. Here, we present our studies on the production, purification and functional evaluation of anti-HER2 IgA antibodies as anti-cancer agents in comparison to the anti-HER2 IgG1 trastuzumab. MALDI-TOF MS analysis showed profound differences in glycosylation traits across the IgA isotypes and cell lines used for production, including sialylation and linkage thereof, fucosylation (both core and antennary) and the abundance of high-mannose type species. Increases in sialylation proved to positively correlate with in vivo plasma half-lives. The polymerization propensity of anti-HER2 IgA2m2 could be suppressed by an 18-aa deletion of the heavy chain tailpiece - coinciding with the loss of high-mannose type N-glycan species - as well as by 2 cysteine to serine mutations at positions 320 and 480. The HER2 F(ab')2-mediated anti-proliferative effect of the IgA2m1 and IgA2m2 subtypes was similar to IgG1, whereas the IgA1 isotype displayed considerably lower potency and efficacy. The Fc-mediated induction of antibody-dependent cell-mediated cytotoxicity (ADCC) using human whole blood ADCC assays did not demonstrate such clear differences between the IgA isotypes. However, the potency of the anti-HER2 IgA antibodies in these ADCC assays was found to be significantly lower than that of trastuzumab. In vivo anti-tumor activity of the anti-HER2 IgA antibodies was compared to that of trastuzumab in a BT-474 breast cancer xenograft model. Multiple dosing and sialylation of the IgA antibodies compensated for the short in vivo half-life of native IgA antibodies in mice compared to a single dose of IgG1. In the case of the IgA2m2 antibody, the resulting high plasma exposure levels were sufficient to cause clear tumor stasis comparable to that observed for trastuzumab at much lower plasma exposure levels.
Collapse
Affiliation(s)
| | | | - Saskia Meyer
- b Laboratory for Translational Immunology; University Medical Center Utrecht ; Utrecht ; The Netherlands
| | - Karli R Reiding
- c Center for Proteomics and Metabolomics; Leiden University Medical Center ; Leiden ; The Netherlands
| | - Jan Schouten
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - Guy de Roo
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - David F Egging
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - Jeanette Hw Leusen
- b Laboratory for Translational Immunology; University Medical Center Utrecht ; Utrecht ; The Netherlands
| | - Peter Boross
- b Laboratory for Translational Immunology; University Medical Center Utrecht ; Utrecht ; The Netherlands
| | - Manfred Wuhrer
- c Center for Proteomics and Metabolomics; Leiden University Medical Center ; Leiden ; The Netherlands.,d Division of BioAnalytical Chemistry; VU University Amsterdam ; Amsterdam , The Netherlands
| | | | - Wim H Dokter
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - Marco Timmers
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - Ruud Ubink
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| |
Collapse
|
44
|
Zhang N, Deng H, Fan X, Gonzalez A, Zhang S, Brezski RJ, Choi BK, Rycyzyn M, Strohl W, Jordan R, An Z. Dysfunctional Antibodies in the Tumor Microenvironment Associate with Impaired Anticancer Immunity. Clin Cancer Res 2015. [PMID: 26224871 DOI: 10.1158/1078-0432.ccr-15-1057] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Studies have demonstrated that cancer-associated matrix metalloproteinases (MMP) can generate single peptide bond cleavages in the hinge region of immunoglobulin G1 (IgG1). This study investigated the cleavage of endogenous IgGs by MMPs in the tumor microenvironment and the consequences of the IgG hinge cleavage for humoral immunity. EXPERIMENTAL DESIGN We investigated the occurrence of single peptide bond cleaved IgGs (scIgG) in tumor tissues and plasma samples collected from a cohort of breast cancer patients (n = 60). Samples from healthy people (n = 20) were used as the control. Antibody hinge cleavage was detected by multiple assays, including IHC, ELISA, and flow cytometry. A correlation analysis was conducted between scIgG levels and patient clinical parameters. RESULTS Levels of scIgGs in tumors were significantly higher than in normal tissues. In addition, scIgG levels in tumors were enriched compared with that in the plasma of the same patients. The appearance of scIgGs in tumor tissues was associated with altered host IgG content and decreased IgG1. Increased tumor scIgGs were found to be positively correlated with adverse clinical factors, such as elevated tumor-associated macrophages, increased expression of MMP9 and other MMPs, and local metastasis to axillary lymph nodes. CONCLUSIONS The study contributes to mounting evidence for the presence of hinge-cleaved antibodies with reduced Fc immune effector function in the tumor microenvironment. The results highlight a link between tumor scIgGs and poor patient outcomes, and reveal a component of compromised humoral immunity within tumors that could point to new immunotherapeutic strategies to rescue host immunity.
Collapse
Affiliation(s)
- Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas.
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Xuejun Fan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Anneliese Gonzalez
- Division of Oncology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Songlin Zhang
- Clinical Pathology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Randall J Brezski
- Biologics Research, Biotechnology Center of Excellence, Janssen R&D, LLC, Spring House, Pennsylvania
| | - Byung-Kwon Choi
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Michael Rycyzyn
- Biologics Research, Biotechnology Center of Excellence, Janssen R&D, LLC, Spring House, Pennsylvania
| | - William Strohl
- Biologics Research, Biotechnology Center of Excellence, Janssen R&D, LLC, Spring House, Pennsylvania
| | - Robert Jordan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas. Biologics Research, Biotechnology Center of Excellence, Janssen R&D, LLC, Spring House, Pennsylvania
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
45
|
Kurogochi M, Mori M, Osumi K, Tojino M, Sugawara SI, Takashima S, Hirose Y, Tsukimura W, Mizuno M, Amano J, Matsuda A, Tomita M, Takayanagi A, Shoda SI, Shirai T. Glycoengineered Monoclonal Antibodies with Homogeneous Glycan (M3, G0, G2, and A2) Using a Chemoenzymatic Approach Have Different Affinities for FcγRIIIa and Variable Antibody-Dependent Cellular Cytotoxicity Activities. PLoS One 2015. [PMID: 26200113 PMCID: PMC4511734 DOI: 10.1371/journal.pone.0132848] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Many therapeutic antibodies have been developed, and IgG antibodies have been extensively generated in various cell expression systems. IgG antibodies contain N-glycans at the constant region of the heavy chain (Fc domain), and their N-glycosylation patterns differ during various processes or among cell expression systems. The Fc N-glycan can modulate the effector functions of IgG antibodies, such as antibody-dependent cellular cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC). To control Fc N-glycans, we performed a rearrangement of Fc N-glycans from a heterogeneous N-glycosylation pattern to homogeneous N-glycans using chemoenzymatic approaches with two types of endo-β-N-acetyl glucosaminidases (ENG'ases), one that works as a hydrolase to cleave all heterogeneous N-glycans, another that is used as a glycosynthase to generate homogeneous N-glycans. As starting materials, we used an anti-Her2 antibody produced in transgenic silkworm cocoon, which consists of non-fucosylated pauci-mannose type (Man2-3GlcNAc2), high-mannose type (Man4-9GlcNAc2), and complex type (Man3GlcNAc3-4) N-glycans. As a result of the cleavage of several ENG'ases (endoS, endoM, endoD, endoH, and endoLL), the heterogeneous glycans on antibodies were fully transformed into homogeneous-GlcNAc by a combination of endoS, endoD, and endoLL. Next, the desired N-glycans (M3; Man3GlcNAc1, G0; GlcNAc2Man3GlcNAc1, G2; Gal2GlcNAc2Man3GlcNAc1, A2; NeuAc2Gal2GlcNAc2Man3GlcNAc1) were transferred from the corresponding oxazolines to the GlcNAc residue on the intact anti-Her2 antibody with an ENG'ase mutant (endoS-D233Q), and the glycoengineered anti-Her2 antibody was obtained. The binding assay of anti-Her2 antibody with homogenous N-glycans with FcγRIIIa-V158 showed that the glycoform influenced the affinity for FcγRIIIa-V158. In addition, the ADCC assay for the glycoengineered anti-Her2 antibody (mAb-M3, mAb-G0, mAb-G2, and mAb-A2) was performed using SKBR-3 and BT-474 as target cells, and revealed that the glycoform influenced ADCC activity.
Collapse
Affiliation(s)
- Masaki Kurogochi
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Masako Mori
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Kenji Osumi
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Mami Tojino
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Shu-ichi Sugawara
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Shou Takashima
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Yuriko Hirose
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Wataru Tsukimura
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Mamoru Mizuno
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Junko Amano
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Akio Matsuda
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Masahiro Tomita
- Immuno-Biological Laboratories Co., Ltd., 1091-1 Naka, Fujioka-shi, Gunma, Japan
| | - Atsushi Takayanagi
- Department of Molecular Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Shin-Ichiro Shoda
- Graduate School of Engineering, Tohoku University, Aoba-ku, Sendai, Japan
| | - Takashi Shirai
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
46
|
Gomathinayagam S, Laface D, Houston-Cummings NR, Mangadu R, Moore R, Shandil I, Sharkey N, Li H, Stadheim TA, Zha D. In vivo anti-tumor efficacy of afucosylated anti-CS1 monoclonal antibody produced in glycoengineered Pichia pastoris. J Biotechnol 2015; 208:13-21. [PMID: 26015261 DOI: 10.1016/j.jbiotec.2015.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/21/2015] [Accepted: 05/13/2015] [Indexed: 11/15/2022]
Abstract
Monoclonal antibody (mAb) therapy has been successfully used for the treatment of B-cell lymphomas and is currently extended for the treatment of multiple myeloma (MM). New developments in MM therapeutics have achieved significant survival gains in patients but the disease still remains incurable. Elotuzumab (HuLuc63), an anti-CS1 monoclonal IgG1 antibody, is believed to induce anti-tumor activity and MM cytotoxicity through antibody dependent cellular cytotoxicity (ADCC) and inhibition of MM cell adhesion to bone marrow stromal cells (BMSCs). Modulations of the Fc glycan composition at the N297 site by selective mutations or afucosylation have been explored as strategies to develop bio-better therapeutics with enhanced ADCC activity. Afucosylated therapeutic antibodies with enhanced ADCC activity have been reported to possess greater efficacy in tumor growth inhibition at lower doses when compared to fucosylated therapeutic antibodies. The N-linked glycosylation pathway in Pichia pastoris has been engineered to produce human-like N-linked glycosylation with uniform afucosylated complex type glycans. The purpose of this study was to compare afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris with fucosylated anti-CS1 mAb expressed in mammalian HEK293 cells through in vitro ADCC and in vivo tumor inhibition models. Our results indicate that Fc glycosylation is critical for in vivo efficacy and afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris shows a better in vivo efficacy in tumor regression when compared to fucosylated anti-CS1 mAb expressed in HEK293 cells. Glycoengineered Pichia pastoris could provide an alternative platform for generating homogeneous afucosylated recombinant antibodies where Fc mediated immune effector function is important for efficacy.
Collapse
Affiliation(s)
- Sujatha Gomathinayagam
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Drake Laface
- Biologics Discovery, Palo Alto, Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, United States
| | - Nga Rewa Houston-Cummings
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Ruban Mangadu
- Biologics Discovery, Palo Alto, Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, United States
| | - Renee Moore
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Ishaan Shandil
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Nathan Sharkey
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Huijuan Li
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Terrance A Stadheim
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Dongxing Zha
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States.
| |
Collapse
|
47
|
Shi Y, Fan X, Deng H, Brezski RJ, Rycyzyn M, Jordan RE, Strohl WR, Zou Q, Zhang N, An Z. Trastuzumab triggers phagocytic killing of high HER2 cancer cells in vitro and in vivo by interaction with Fcγ receptors on macrophages. THE JOURNAL OF IMMUNOLOGY 2015; 194:4379-86. [PMID: 25795760 DOI: 10.4049/jimmunol.1402891] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/19/2015] [Indexed: 11/19/2022]
Abstract
Trastuzumab has been used for the treatment of HER2-overexpressing breast cancer for more than a decade, but the mechanisms of action for the therapy are still being actively investigated. Ab-dependent cell-mediated cytotoxicity mediated by NK cells is well recognized as one of the key mechanisms of action for trastuzumab, but trastuzumab-mediated Ab-dependent cellular phagocytosis (ADCP) has not been established. In this study, we demonstrate that macrophages, by way of phagocytic engulfment, can mediate ADCP and cancer cell killing in the presence of trastuzumab. Increased infiltration of macrophages in the tumor tissue was associated with enhanced efficacy of trastuzumab whereas depletion of macrophages resulted in reduced antitumor efficacy in mouse xenograft tumor models. Among the four mouse FcγRs, FcγRIV exhibits the strongest binding affinity to trastuzumab. Knockdown of FcγRIV in mouse macrophages reduced cancer cell killing and ADCP activity triggered by trastuzumab. Consistently, an upregulation of FcγRIV expression by IFN-γ triggered an increased ADCP activity by trastuzumab. In an analogous fashion, IFN-γ priming of human macrophages increased the expression of FcγRIII, the ortholog of murine FcγRIV, and increased trastuzumab-mediated cancer cell killing. Thus, in two independent systems, the results indicated that activation of macrophages in combination with trastuzumab can serve as a therapeutic strategy for treating high HER2 breast cancer by boosting ADCP killing of cancer cells.
Collapse
Affiliation(s)
- Yun Shi
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030; National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, People's Republic of China; and
| | - Xuejun Fan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Randall J Brezski
- Biologics Research, Janssen Research & Development, LLC, Spring House, PA 19002
| | - Michael Rycyzyn
- Biologics Research, Janssen Research & Development, LLC, Spring House, PA 19002
| | - Robert E Jordan
- Biologics Research, Janssen Research & Development, LLC, Spring House, PA 19002
| | - William R Strohl
- Biologics Research, Janssen Research & Development, LLC, Spring House, PA 19002
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, People's Republic of China; and
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030;
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030;
| |
Collapse
|
48
|
Laukens B, De Visscher C, Callewaert N. Engineering yeast for producing human glycoproteins: where are we now? Future Microbiol 2015; 10:21-34. [PMID: 25598335 PMCID: PMC7617146 DOI: 10.2217/fmb.14.104] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Yeast has advanced as an alternative for mammalian cell culture for the production of recombinant therapeutic glycoproteins. Engineered yeast strains not only allow to mimic the human N-glycosylation pathway but also specific types of human O-glycosylation. This is of great value for therapeutic protein production and indispensable to determine the structure-function relationships of glycans on recombinant proteins. However, as the technology matures, some limitations have come up that may hamper biomedical applications and must be considered to exploit the full potential of the unprecedented glycan homogeneity obtained on relevant biopharmaceuticals. In this special report, we focus on the recent developments in N- and O-glycosylation engineering in yeasts of industrial importance, to produce recombinant therapeutics with customized glycans.
Collapse
Affiliation(s)
- Bram Laukens
- Unit for Medical Biotechnology, Inflammation Research Centre (IRC), VIB-UGent, Technologiepark 927, B-9052 Ghent-Zwijnaarde, Belgium
- Department of Biochemistry & Microbiology, Laboratory for Protein Biochemistry & Biomolecular Engineering, Ghent University, K.L.-Ledeganckstraat 35, B-9000 Ghent, Belgium
| | - Charlotte De Visscher
- Unit for Medical Biotechnology, Inflammation Research Centre (IRC), VIB-UGent, Technologiepark 927, B-9052 Ghent-Zwijnaarde, Belgium
- Department of Biochemistry & Microbiology, Laboratory for Protein Biochemistry & Biomolecular Engineering, Ghent University, K.L.-Ledeganckstraat 35, B-9000 Ghent, Belgium
| | - Nico Callewaert
- Unit for Medical Biotechnology, Inflammation Research Centre (IRC), VIB-UGent, Technologiepark 927, B-9052 Ghent-Zwijnaarde, Belgium
- Department of Biochemistry & Microbiology, Laboratory for Protein Biochemistry & Biomolecular Engineering, Ghent University, K.L.-Ledeganckstraat 35, B-9000 Ghent, Belgium
- Department of Medical Protein Research, VIB-UGent, Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| |
Collapse
|
49
|
Fan X, Brezski RJ, Deng H, Dhupkar PM, Shi Y, Gonzalez A, Zhang S, Rycyzyn M, Strohl WR, Jordan RE, Zhang N, An Z. A Novel Therapeutic Strategy to Rescue the Immune Effector Function of Proteolytically Inactivated Cancer Therapeutic Antibodies. Mol Cancer Ther 2014; 14:681-91. [DOI: 10.1158/1535-7163.mct-14-0715] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/11/2014] [Indexed: 11/16/2022]
|
50
|
Inactivation of a GAL4-like transcription factor improves cell fitness and product yield in glycoengineered Pichia pastoris strains. Appl Environ Microbiol 2014; 81:260-71. [PMID: 25344235 DOI: 10.1128/aem.02619-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
With a completely reengineered and humanized glycosylation pathway, glycoengineered Pichia pastoris has emerged as a promising production host for the manufacture of therapeutic glycoproteins. However, the extensive genetic modifications have also negatively affected the overall fitness levels of the glycoengineered host cells. To make glycoengineered Pichia strains more compatible with a scalable industrial fermentation process, we sought to identify genetic solutions to broadly improve cell robustness during fermentation. In this study, we report that mutations within the Pichia pastoris ATT1 (PpATT1) gene (a homolog of the Saccharomyces cerevisiae GAL4 [ScGAL4] transcriptional activator) dramatically increased the cellular fitness levels of glycoengineered Pichia strains. We demonstrate that deletion of the PpATT1 gene enabled glycoengineered Pichia strains to improve their thermal tolerance levels, reduce their cell lysis defects, and greatly improve fermentation robustness. The extension of the duration of fermentation enabled the PpATT1-modified glycoengineered Pichia strains to increase their product yields significantly without any sacrifice in product quality. Because the ATT1 gene could be deleted from any Pichia strains, including empty hosts and protein-expressing production strains alike, we suggest that the findings described in this study are broadly applicable to any Pichia strains used for the production of therapeutic proteins, including monoclonal antibodies, Fc fusions, peptides, hormones, and growth factors.
Collapse
|