1
|
Ko S, Jo M, Kyung M, Lee W, Ko WH, Na JH, Chun YS, Ko BJ, Jung ST. Engineering FcRn binding kinetics dramatically extends antibody serum half-life and enhances therapeutic potential. J Biol Eng 2025; 19:35. [PMID: 40251669 PMCID: PMC12007268 DOI: 10.1186/s13036-025-00506-y] [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: 03/18/2025] [Accepted: 04/10/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Optimizing the IgG Fc domain for neonatal Fc receptor (FcRn) binding is crucial for enhancing antibody pharmacokinetics. The prolonged serum half-life of IgG antibody is governed by its pH-dependent interaction with FcRn, enabling efficient binding at acidic endosomal pH, intracellular trafficking, and release at neutral serum pH. However, a critical yet previously unrecognized challenge in Fc engineering for extending the serum half-life of therapeutic antibodies is the intense competition with endogenous IgG for FcRn binding during intracellular trafficking, which limits FcRn-mediated transport and reduces the serum persistence of therapeutic antibodies. To address this, we developed an Fc variant that precisely modulates pH-dependent FcRn binding kinetics, accelerates FcRn association at acidic pH, and promotes rapid dissociation at neutral pH, thereby enhancing FcRn-driven intracellular transport, outcompeting endogenous IgG, and achieving unprecedented improvement in the serum half-life of therapeutic antibodies. RESULTS Using comprehensive site-directed saturation mutagenesis coupled with functional screening, we generated a diverse panel of Fc variants and identified two with distinct FcRn binding kinetics: YML (L309Y/Q311M/M428L), which exhibited superior FcRn association at acidic pH and accelerated dissociation at neutral pH, and EML (L309E/Q311M/M428L), which displayed attenuated binding kinetics. In human FcRn transgenic mice, YML extended the serum half-life of clinically used trastuzumab with a wild-type Fc by 6.1-fold, demonstrating a remarkable improvement over previously reported Fc-engineered variants, including PFc29 (Q311R/M428L) and DHS (L309D/Q311H/N434S), which represent the most effective Fc modifications for prolonging serum persistence to date. This in vivo validation underscores the pivotal role of FcRn kinetic tuning in overcoming endogenous IgG competition and maximizing FcRn-mediated antibody transport. Additionally, YML exhibited potent complement-dependent cytotoxicity (CDC) while maintaining favorable physicochemical properties. CONCLUSION This study presents a rational Fc engineering framework to optimize FcRn binding kinetics, addressing a previously unconsidered challenge-endogenous IgG competition during intracellular trafficking of therapeutic antibodies. The distinct kinetic behaviors of YML and EML highlight the critical necessity of precise control over pH-dependent association and dissociation rates in FcRn binding. YML represents a next-generation Fc platform, offering enhanced pharmacokinetics and improved effector functions, thus providing a powerful strategy for developing biologics with superior serum persistence and therapeutic efficacy.
Collapse
Affiliation(s)
- Sanghwan Ko
- Department of Biomedical Sciences, Graduate School, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Migyeong Jo
- Institute of Chemical Processes, Seoul National University, Gwanak-gu, Seoul, 08826, Republic of Korea
- Department of Chemical and Biological Engineering, College of Engineering, Seoul National University, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Munsu Kyung
- Department of Biomedical Sciences, Graduate School, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Wonju Lee
- Department of Biomedical Sciences, Graduate School, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Manufacturing Science & Technology Team, Manufacturing Science Group2, Samsung Bioepis, Incheon, Republic of Korea
| | - Woo Hyung Ko
- Department of Chemical and Biological Engineering, College of Engineering, Seoul National University, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Jung-Hyun Na
- School of Biopharmaceutical and Medical Science, Sungshin Women's University, Gangbuk-gu, Seoul, 01133, Republic of Korea
| | - Youn Seo Chun
- School of Biopharmaceutical and Medical Science, Sungshin Women's University, Gangbuk-gu, Seoul, 01133, Republic of Korea
| | - Byoung Joon Ko
- School of Biopharmaceutical and Medical Science, Sungshin Women's University, Gangbuk-gu, Seoul, 01133, Republic of Korea
| | - Sang Taek Jung
- Institute of Chemical Processes, Seoul National University, Gwanak-gu, Seoul, 08826, Republic of Korea.
- Department of Chemical and Biological Engineering, College of Engineering, Seoul National University, Gwanak-gu, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
2
|
Bernstein ZJ, Gierke TR, Dammen-Brower K, Tzeng SY, Zhu S, Chen SS, Wilson DS, Green JJ, Yarema KJ, Spangler JB. Production of site-specific antibody conjugates using metabolic glycoengineering and novel Fc glycovariants. J Biol Chem 2024; 300:108005. [PMID: 39551135 PMCID: PMC11697773 DOI: 10.1016/j.jbc.2024.108005] [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: 07/25/2024] [Revised: 10/23/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Molecular conjugation to antibodies has emerged as a growing strategy to combine the mechanistic activities of the attached molecule with the specificity of antibodies. A variety of technologies have been applied for molecular conjugation; however, these approaches face several limitations, including disruption of antibody structure, destabilization of the antibody, and/or heterogeneous conjugation patterns. Collectively, these challenges lead to reduced yield, purity, and function of conjugated antibodies. While glycoengineering strategies have largely been applied to study protein glycosylation and manipulate cellular metabolism, these approaches also harbor great potential to enhance the production and performance of protein therapeutics. Here, we devise a novel glycoengineering workflow for the development of site-specific antibody conjugates. This approach combines metabolic glycoengineering using azido-sugar analogs with newly installed N-linked glycosylation sites in the antibody constant domain to achieve specific conjugation to the antibody via the introduced N-glycans. Our technique allows facile and efficient manufacturing of well-defined antibody conjugates without the need for complex or destructive chemistries. Moreover, the introduction of conjugation sites in the antibody fragment crystallizable (Fc) domain renders this approach widely applicable and target agnostic. Our platform can accommodate up to three conjugation sites in tandem, and the extent of conjugation can be tuned through the use of different sugar analogs or production in different cell lines. We demonstrated that our platform is compatible with various use-cases, including fluorescent labeling, antibody-drug conjugation, and targeted gene delivery. Overall, this study introduces a versatile and effective yet strikingly simple approach to producing antibody conjugates for research, industrial, and medical applications.
Collapse
Affiliation(s)
- Zachary J Bernstein
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Taylor R Gierke
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kris Dammen-Brower
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Stanley Zhu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sabrina S Chen
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - D Scott Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kevin J Yarema
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jamie B Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Molecular Microbiology & Immunology, Johns Hopkins University School of Public Health, Baltimore, Maryland, USA.
| |
Collapse
|
3
|
Liu X, Song Y, Cheng P, Liang B, Xing D. Targeting HER2 in solid tumors: Unveiling the structure and novel epitopes. Cancer Treat Rev 2024; 130:102826. [PMID: 39270365 DOI: 10.1016/j.ctrv.2024.102826] [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: 05/14/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Human epidermal growth factor receptor-2 (HER2) is overexpressed in various solid tumor types, acting as an established therapeutic target. Over the last three decades, the fast-paced development of diverse HER2-targeted agents, notably marked by the introduction of the antibody-drug conjugate (ADC), yielding substantial improvements in survival rates. However, resistance to anti-HER2 treatments continues to pose formidable challenges. The complex structure and dynamic dimerization properties of HER2 create significant hurdles in the development of novel targeted therapeutics. In this review, we synthesize the latest insights into the structural intricacies of HER2 and present an unprecedented overview of the epitope characteristics of HER2-targeted antibodies and their derivatives. Furthermore, we delve into the correlation between anti-HER2 antibody binding epitopes and their respective functions, with a particular focus on their efficacy against resistant tumors. In addition, we highlight the potential of emerging anti-HER2 agents that target specific sites or non-overlapping epitopes, poised to transform the therapeutic landscape for HER2-positive tumors in the foreseeable future.
Collapse
Affiliation(s)
- Xinlin Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China
| | - Yunlong Song
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Panpan Cheng
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
4
|
Tong M, Liu P, Li C, Zhang Z, Sun W, Dong P, Fan N, Wang X, Liu J, Lv C, Cao Z, Wang Y. Interaction of Asn297-Linked Glycan Ligands with the Fc Fragment of the Immunoglobulin Class G1: A Molecular Dynamics Simulation Study. J Chem Inf Model 2024; 64:785-798. [PMID: 38262973 DOI: 10.1021/acs.jcim.3c01584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
The allosteric modulation of the homodimeric H10-03-6 protein to glycan ligands L1 and L2, and the STAB19 protein to glycan ligands L3 and L4, respectively, has been studied by molecular dynamics simulations and free energy calculations. The results revealed that the STAB19 protein has a significantly higher affinity for L3 (-11.38 ± 2.32 kcal/mol) than that for L4 (-5.51 ± 1.92 kcal/mol). However, the combination of the H10-03-6 protein with glycan L2 (1.23 ± 6.19 kcal/mol) is energetically unfavorable compared with that of L1 (-13.96 ± 0.35 kcal/mol). Further, the binding of glycan ligands L3 and L4 to STAB19 would result in the significant closure of the two CH2 domains of the STAB19 conformation with the decrease of the centroid distances between the two CH2 domains compared with the H10-03-6/L1/L2 complex. The CH2 domain closure of STAB19 relates directly to the formation of new hydrogen bonds and hydrophobic interactions between the residues Ser239, Val240, Asp265, Glu293, Asn297, Thr299, Ser337, Asp376, Thr393, Pro395, and Pro396 in STAB19 and glycan ligands L3 and L4, which suggests that these key residues would contribute to the specific regulation of STAB19 to L3 and L4. In addition, the distance analysis revealed that the EF loop in the H10-03-6/L1/L2 model presents a high flexibility and partial disorder compared with the stabilized STAB19/L3/L4 complex. These results will be helpful in understanding the specific regulation through the asymmetric structural characteristics in the CH2 and CH3 domains of the H10-03-6 and STAB19 proteins.
Collapse
Affiliation(s)
- Mingqiong Tong
- Shandong Engineering Research Center of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou 253023, China
| | - Peng Liu
- Faculty of Electrical Engineering, Universiti Teknologi Malaysia, UTM, Johor Bahru, Johor Darul Takzim 81310, Malaysia
- The Office of Academic Affairs, Dezhou University, Dezhou 253023, China
| | - Chaoqun Li
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemistry, Chemical Engineering and Materials, Handan University, Handan, Hebei 056005, China
| | - Zhongyu Zhang
- Shandong Engineering Research Center of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou 253023, China
| | - Wan Sun
- Shandong Engineering Research Center of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou 253023, China
| | - Pingxuan Dong
- Shandong Engineering Research Center of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou 253023, China
| | - Na Fan
- Shandong Engineering Research Center of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou 253023, China
| | - Xiaoyue Wang
- Shandong Engineering Research Center of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou 253023, China
| | - Jing Liu
- Shandong Engineering Research Center of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou 253023, China
| | - Chao Lv
- College of Chemistry and Chemical Engineering, Dezhou University, Dezhou 253023, China
| | - Zanxia Cao
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China
| | - Yan Wang
- College of Chemistry, Beijing Normal University, 19# Xinjiekouwai Street, Beijing 100875, China
| |
Collapse
|
5
|
Díaz-Fernández A, Ferapontov A, Vendelbo MH, Ferapontova EE. Electrochemical Cellulase-Linked ELASA for Rapid Liquid Biopsy Testing of Serum HER-2/ neu. ACS MEASUREMENT SCIENCE AU 2023; 3:226-235. [PMID: 37360033 PMCID: PMC10288612 DOI: 10.1021/acsmeasuresciau.2c00067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/05/2023] [Accepted: 04/05/2023] [Indexed: 06/28/2023]
Abstract
Non-invasive liquid biopsy assays for blood-circulating biomarkers of cancer allow both its early diagnosis and treatment monitoring. Here, we assessed serum levels of protein HER-2/neu, overexpressed in a number of aggressive cancers, by the cellulase-linked sandwich bioassay on magnetic beads. Instead of traditional antibodies we used inexpensive reporter and capture aptamer sequences, transforming the enzyme-linked immuno-sorbent assay (ELISA) into an enzyme-linked aptamer-sorbent assay (ELASA). The reporter aptamer was conjugated to cellulase, whose digestion of nitrocellulose film electrodes resulted in the electrochemical signal change. ELASA, optimized relative aptamer lengths (dimer vs monomer and trimer), and assay steps allowed 0.1 fM detection of HER-2/neu in the 10% human serum in 1.3 h. Urokinase plasminogen activator and thrombin as well as human serum albumin did not interfere, and liquid biopsy analysis of serum HER-2/neu was similarly robust but 4 times faster and 300 times cheaper than both electrochemical and optical ELISA. Simplicity and low cost of cellulase-linked ELASA makes it a perspective diagnostic tool for fast and accurate liquid biopsy detection of HER-2/neu and of other proteins for which aptamers are available.
Collapse
Affiliation(s)
- Ana Díaz-Fernández
- Interdisciplinary
Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Alexey Ferapontov
- Interdisciplinary
Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Mikkel Holm Vendelbo
- Department
of Nuclear Medicine & PET Centre, Aarhus
University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
- Department
of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Elena E. Ferapontova
- Interdisciplinary
Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| |
Collapse
|
6
|
Benedetti F, Stadlmayr G, Stadlbauer K, Rüker F, Wozniak-Knopp G. Selection of High-Affinity Heterodimeric Antigen-Binding Fc Fragments from a Large Yeast Display Library. Methods Mol Biol 2023; 2681:131-159. [PMID: 37405647 DOI: 10.1007/978-1-0716-3279-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Antigen-binding Fc (Fcab™) fragments, where a novel antigen binding site is introduced by the mutagenesis of the C-terminal loops of the CH3 domain, function as parts of bispecific IgG-like symmetrical antibodies when they replace their wild-type Fc. Their homodimeric structure typically leads to bivalent antigen binding. In particular, biological situations monovalent engagement, however, would be preferred, either for avoiding agonistic effects leading to safety issues, or the attractive option of combining a single chain (i.e., one half) of an Fcab fragment reactive with different antigens in one antibody. We present the strategies for construction and selection of yeast libraries displaying heterodimeric Fcab fragments and discuss the effects of altered thermostability of the basic Fc scaffold and novel library designs that lead to isolation of highly affine antigen binding clones.
Collapse
Affiliation(s)
- Filippo Benedetti
- Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Gerhard Stadlmayr
- Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Katharina Stadlbauer
- Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Florian Rüker
- Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Gordana Wozniak-Knopp
- Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria.
| |
Collapse
|
7
|
Mark JKK, Lim CSY, Nordin F, Tye GJ. Expression of mammalian proteins for diagnostics and therapeutics: a review. Mol Biol Rep 2022; 49:10593-10608. [PMID: 35674877 PMCID: PMC9175168 DOI: 10.1007/s11033-022-07651-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/25/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Antibodies have proven to be remarkably successful for biomedical applications. They play important roles in epidemiology and medicine from diagnostics of diseases to therapeutics, treating diseases from incessant chronic diseases such as rheumatology to pandemic outbreaks. With no end in sight for the demand for antibody products, optimizations and new techniques must be expanded to accommodate this. METHODS AND RESULTS This review discusses optimizations and techniques for antibody production through choice of discovery platforms, expression systems, cell culture mediums, and other strategies to increase expression yield. Each system has its own merits and demerits, and the strategy chosen is critical in addressing various biological aspects. CONCLUSIONS There is still insufficient evidence to validate the efficacy of some of these techniques, and further research is needed to consolidate these industrial production systems. There is no doubt that more strategies, systems, and pipelines will contribute to enhance biopharmaceutical production.
Collapse
Affiliation(s)
- Jacqueline Kar Kei Mark
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Minden, Malaysia
| | - Crystale Siew Ying Lim
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, No 1 Jalan Menara Gading, UCSI Heights, Taman Connaught, 56000, Kuala Lumpur, Cheras, Malaysia
| | - Fazlina Nordin
- Tissue Engineering Centre (TEC), Universiti Kebangsaan Malaysia Medical Centre (UKMMC), 56000, Kuala Lumpur, Cheras, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Minden, Malaysia.
| |
Collapse
|
8
|
Natale V, Stadlmayr G, Benedetti F, Stadlbauer K, Rüker F, Wozniak-Knopp G. Trispecific antibodies produced from mAb 2 pairs by controlled Fab-arm exchange. Biol Chem 2022; 403:509-523. [PMID: 35089662 DOI: 10.1515/hsz-2021-0376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022]
Abstract
Bispecific antibodies and antibody fragments are therapeutics of growing importance. They are clinically applied for effector cell engagement, enhanced targeting selectivity, addressing of multiple cellular pathways and active transfer of certain activities into difficult-to-reach compartments. These functionalities could profit from a third antigen specificity. In this work we have employed symmetrical bispecific parental antibodies of mAb2 format, which feature a novel antigen binding site in the CH3 domains, and engineered them with a minimal number of point mutations to guide the formation of a controlled Fab-arm exchanged trispecific antibody at a high yield after reduction and re-oxidation. Two model antibodies, one reactive with EGFR, Her2 and VEGF, and one with Fab-arms binding to Ang2 and VEGF and an Fc fragment binding to VEGF, were prepared and examined for heterodimeric status, stability, antigen binding properties and biological activity. Resulting molecules were of good biophysical characteristics and retained antigen reactivity and biological activity of the parental mAb2 constructs.
Collapse
Affiliation(s)
- Veronica Natale
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Gerhard Stadlmayr
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Filippo Benedetti
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Katharina Stadlbauer
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Florian Rüker
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Gordana Wozniak-Knopp
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| |
Collapse
|
9
|
Capraz T, Kienzl NF, Laurent E, Perthold JW, Föderl-Höbenreich E, Grünwald-Gruber C, Maresch D, Monteil V, Niederhöfer J, Wirnsberger G, Mirazimi A, Zatloukal K, Mach L, Penninger JM, Oostenbrink C, Stadlmann J. Structure-guided glyco-engineering of ACE2 for improved potency as soluble SARS-CoV-2 decoy receptor. eLife 2021; 10:e73641. [PMID: 34927585 PMCID: PMC8730730 DOI: 10.7554/elife.73641] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
Infection and viral entry of SARS-CoV-2 crucially depends on the binding of its Spike protein to angiotensin converting enzyme 2 (ACE2) presented on host cells. Glycosylation of both proteins is critical for this interaction. Recombinant soluble human ACE2 can neutralize SARS-CoV-2 and is currently undergoing clinical tests for the treatment of COVID-19. We used 3D structural models and molecular dynamics simulations to define the ACE2 N-glycans that critically influence Spike-ACE2 complex formation. Engineering of ACE2 N-glycosylation by site-directed mutagenesis or glycosidase treatment resulted in enhanced binding affinities and improved virus neutralization without notable deleterious effects on the structural stability and catalytic activity of the protein. Importantly, simultaneous removal of all accessible N-glycans from recombinant soluble human ACE2 yields a superior SARS-CoV-2 decoy receptor with promise as effective treatment for COVID-19 patients.
Collapse
Affiliation(s)
- Tümay Capraz
- Institute for Molecular Modeling and Simulation, University of Natural Resources and Life Sciences (BOKU)ViennaAustria
| | - Nikolaus F Kienzl
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences (BOKU)ViennaAustria
| | - Elisabeth Laurent
- Institute of Molecular Biotechnology, Department of Biotechnology and Core Facility Biomolecular & Cellular Analysis, University of Natural Resources and Life Sciences (BOKU)ViennaAustria
| | - Jan W Perthold
- Institute for Molecular Modeling and Simulation, University of Natural Resources and Life Sciences (BOKU)ViennaAustria
| | | | - Clemens Grünwald-Gruber
- Institute of Biochemistry, Department of Chemistry, University of Natural Resources and Life SciencesViennaAustria
| | - Daniel Maresch
- Institute of Biochemistry, Department of Chemistry, University of Natural Resources and Life SciencesViennaAustria
| | - Vanessa Monteil
- Karolinska Institute, Department of Laboratory MedicineStockholmSweden
| | | | | | - Ali Mirazimi
- Karolinska Institute, Department of Laboratory MedicineStockholmSweden
- National Veterinary InstituteUppsalaSweden
| | - Kurt Zatloukal
- Diagnostic and Research Institute of Pathology, Medical University of GrazGrazAustria
| | - Lukas Mach
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences (BOKU)ViennaAustria
| | - Josef M Penninger
- IMBA - Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. BohrViennaAustria
- Department of Medical Genetics, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Chris Oostenbrink
- Institute for Molecular Modeling and Simulation, University of Natural Resources and Life Sciences (BOKU)ViennaAustria
| | - Johannes Stadlmann
- Institute of Biochemistry, Department of Chemistry, University of Natural Resources and Life SciencesViennaAustria
- IMBA - Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. BohrViennaAustria
| |
Collapse
|
10
|
Benedetti F, Stadlbauer K, Stadlmayr G, Rüker F, Wozniak-Knopp G. A Tetravalent Biparatopic Antibody Causes Strong HER2 Internalization and Inhibits Cellular Proliferation. Life (Basel) 2021; 11:life11111157. [PMID: 34833033 PMCID: PMC8624325 DOI: 10.3390/life11111157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
The overexpression of tyrosine kinase HER2 in numerous cancers, connected with fierce signaling and uncontrolled proliferation, makes it a suitable target for immunotherapy. The acquisition of resistance to currently used compounds and the multiplicity of signaling pathways involved prompted research into the discovery of novel binders as well as treatment options with multiple targeting and multispecific agents. Here we constructed an anti-HER2 tetravalent and biparatopic symmetrical IgG-like molecule by combining the Fab of pertuzumab with a HER2-specific Fcab (Fc fragment with antigen binding), which recognizes an epitope overlapping with trastuzumab. In the strongly HER2-positive cell line SK-BR-3, the molecule induced a rapid and efficient reduction in surface HER2 levels. A potent anti-proliferative effect, specific for the HER2-positive cell line, was observed in vitro, following the induction of apoptosis, and this could not be achieved with treatment with the mixture of pertuzumab and the parental Fcab. The inhibitory cytotoxic effect of our antibody as a single agent makes it a promising contribution to the armory of anti-cancer molecules directed against HER2-addicted cells.
Collapse
|
11
|
Soltermann F, Struwe WB, Kukura P. Label-free methods for optical in vitro characterization of protein-protein interactions. Phys Chem Chem Phys 2021; 23:16488-16500. [PMID: 34342317 PMCID: PMC8359934 DOI: 10.1039/d1cp01072g] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022]
Abstract
Protein-protein interactions are involved in the regulation and function of the majority of cellular processes. As a result, much effort has been aimed at the development of methodologies capable of quantifying protein-protein interactions, with label-free methods being of particular interest due to the associated simplified workflows and minimisation of label-induced perturbations. Here, we review recent advances in optical technologies providing label-free in vitro measurements of affinities and kinetics. We provide an overview and comparison of existing techniques and their principles, discussing advantages, limitations, and recent applications.
Collapse
Affiliation(s)
- Fabian Soltermann
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| | - Weston B. Struwe
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| | - Philipp Kukura
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| |
Collapse
|
12
|
Hoffmann D, Mereiter S, Jin Oh Y, Monteil V, Elder E, Zhu R, Canena D, Hain L, Laurent E, Grünwald-Gruber C, Klausberger M, Jonsson G, Kellner MJ, Novatchkova M, Ticevic M, Chabloz A, Wirnsberger G, Hagelkruys A, Altmann F, Mach L, Stadlmann J, Oostenbrink C, Mirazimi A, Hinterdorfer P, Penninger JM. Identification of lectin receptors for conserved SARS-CoV-2 glycosylation sites. EMBO J 2021; 40:e108375. [PMID: 34375000 PMCID: PMC8420505 DOI: 10.15252/embj.2021108375] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 12/23/2022] Open
Abstract
New SARS‐CoV‐2 variants are continuously emerging with critical implications for therapies or vaccinations. The 22 N‐glycan sites of Spike remain highly conserved among SARS‐CoV‐2 variants, opening an avenue for robust therapeutic intervention. Here we used a comprehensive library of mammalian carbohydrate‐binding proteins (lectins) to probe critical sugar residues on the full‐length trimeric Spike and the receptor binding domain (RBD) of SARS‐CoV‐2. Two lectins, Clec4g and CD209c, were identified to strongly bind to Spike. Clec4g and CD209c binding to Spike was dissected and visualized in real time and at single‐molecule resolution using atomic force microscopy. 3D modelling showed that both lectins can bind to a glycan within the RBD‐ACE2 interface and thus interferes with Spike binding to cell surfaces. Importantly, Clec4g and CD209c significantly reduced SARS‐CoV‐2 infections. These data report the first extensive map and 3D structural modelling of lectin‐Spike interactions and uncovers candidate receptors involved in Spike binding and SARS‐CoV‐2 infections. The capacity of CLEC4G and mCD209c lectins to block SARS‐CoV‐2 viral entry holds promise for pan‐variant therapeutic interventions.
Collapse
Affiliation(s)
- David Hoffmann
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Stefan Mereiter
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Yoo Jin Oh
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Vanessa Monteil
- Department of Laboratory Medicine, Unit of Clinical Microbiology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | | | - Rong Zhu
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Daniel Canena
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Lisa Hain
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Elisabeth Laurent
- Department of Biotechnology and BOKU Core Facility Biomolecular & Cellular Analysis, University of Natural Resources and Life Sciences, Vienna, Austria
| | | | - Miriam Klausberger
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Gustav Jonsson
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Max J Kellner
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Maria Novatchkova
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Melita Ticevic
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Antoine Chabloz
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | | | - Astrid Hagelkruys
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Friedrich Altmann
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Lukas Mach
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Johannes Stadlmann
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.,Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Chris Oostenbrink
- Department for Material Sciences and Process Engineering, Institute for Molecular Modeling and Simulation, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Ali Mirazimi
- Department of Laboratory Medicine, Unit of Clinical Microbiology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden.,National Veterinary Institute, Uppsala, Sweden
| | | | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
13
|
Jäger S, Wagner TR, Rasche N, Kolmar H, Hecht S, Schröter C. Generation and Biological Evaluation of Fc Antigen Binding Fragment-Drug Conjugates as a Novel Antibody-Based Format for Targeted Drug Delivery. Bioconjug Chem 2021; 32:1699-1710. [PMID: 34185508 DOI: 10.1021/acs.bioconjchem.1c00240] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Fragment crystallizable (Fc) antigen binding fragments (Fcabs) represent a novel antibody format comprising a homodimeric Fc region with an engineered antigen binding site. In contrast to their full-length antibody offspring, Fcabs combine Fc-domain-mediated and antigen binding functions at only one-third of the size. Their reduced size is accompanied by elevated tissue penetration capabilities, which is an attractive feature for the treatment of solid tumors. In the present study, we explored for the first time Fcabs as a novel scaffold for antibody-drug conjugates (ADCs). As model, various HER2-targeting Fcab variants coupled to a pH-sensitive dye were used in internalization experiments. A selective binding on HER2-expressing tumor cells and receptor-mediated endocytosis could be confirmed for selected variants, indicating that these Fcabs meet the basic prerequisite for an ADC approach. Subsequently, Fcabs were site-specifically coupled to cytotoxic monomethyl auristatin E yielding homogeneous conjugates. The conjugates retained HER2 and FcRn binding behavior of the parent Fcabs, showed a selective in vitro cell killing and conjugation site-dependent serum stability. Moreover, Fcab conjugates showed elevated penetration in a spheroid model, compared to their full-length antibody and Trastuzumab counterparts. Altogether, the presented results emphasize the potential of Fcabs as a novel scaffold for targeted drug delivery in solid cancers and pave the way for future in vivo translation.
Collapse
Affiliation(s)
- Sebastian Jäger
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany.,Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Str. 4, 64287 Darmstadt, Germany
| | - Tim R Wagner
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Str. 4, 64287 Darmstadt, Germany
| | - Nicolas Rasche
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Str. 4, 64287 Darmstadt, Germany
| | - Stefan Hecht
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Christian Schröter
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| |
Collapse
|
14
|
Laurent E, Sieber A, Salzer B, Wachernig A, Seigner J, Lehner M, Geyeregger R, Kratzer B, Jäger U, Kunert R, Pickl WF, Traxlmayr MW. Directed Evolution of Stabilized Monomeric CD19 for Monovalent CAR Interaction Studies and Monitoring of CAR-T Cell Patients. ACS Synth Biol 2021; 10:1184-1198. [PMID: 33843201 PMCID: PMC8155657 DOI: 10.1021/acssynbio.1c00010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CD19 is among the most relevant targets in cancer immunotherapy. However, its extracellular domain (ECD) is prone to aggregation and misfolding, representing a major obstacle for the development and analysis of CD19-targeted therapeutics. Here, we engineered stabilized CD19-ECD (termed SuperFolder) variants, which also showed improved expression rates and, in contrast to the wild type protein, they could be efficiently purified in their monomeric forms. Despite being considerably more stable, these engineered mutants largely preserved the wild type sequence (>98.8%). We demonstrate that the variant SF05 enabled the determination of the monovalent affinity between CD19 and a clinically approved FMC63-based CAR, as well as monitoring and phenotypic characterization of CD19-directed CAR-T cells in the blood of lymphoma patients. We anticipate that the SuperFolder mutants generated in this study will be highly valuable tools for a range of applications in basic immunology and CD19-targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Elisabeth Laurent
- Department of Biotechnology and BOKU Core Facility Biomolecular and Cellular Analysis, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Anna Sieber
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Benjamin Salzer
- St. Anna Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Anna Wachernig
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Jacqueline Seigner
- Department of Chemistry, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Manfred Lehner
- St. Anna Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - René Geyeregger
- St. Anna Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, 1090 Vienna, Austria
| | - Ulrich Jäger
- Department of Internal Medicine, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, 1090 Vienna, Austria
| | - Michael W. Traxlmayr
- Department of Chemistry, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
15
|
Klausberger M, Duerkop M, Haslacher H, Wozniak-Knopp G, Cserjan-Puschmann M, Perkmann T, Lingg N, Aguilar PP, Laurent E, De Vos J, Hofner M, Holzer B, Stadler M, Manhart G, Vierlinger K, Egger M, Milchram L, Gludovacz E, Marx N, Köppl C, Tauer C, Beck J, Maresch D, Grünwald-Gruber C, Strobl F, Satzer P, Stadlmayr G, Vavra U, Huber J, Wahrmann M, Eskandary F, Breyer MK, Sieghart D, Quehenberger P, Leitner G, Strassl R, Egger AE, Irsara C, Griesmacher A, Hoermann G, Weiss G, Bellmann-Weiler R, Loeffler-Ragg J, Borth N, Strasser R, Jungbauer A, Hahn R, Mairhofer J, Hartmann B, Binder NB, Striedner G, Mach L, Weinhäusel A, Dieplinger B, Grebien F, Gerner W, Binder CJ, Grabherr R. A comprehensive antigen production and characterisation study for easy-to-implement, specific and quantitative SARS-CoV-2 serotests. EBioMedicine 2021; 67:103348. [PMID: 33906067 PMCID: PMC8099623 DOI: 10.1016/j.ebiom.2021.103348] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 04/02/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Antibody tests are essential tools to investigate humoral immunity following SARS-CoV-2 infection or vaccination. While first-generation antibody tests have primarily provided qualitative results, accurate seroprevalence studies and tracking of antibody levels over time require highly specific, sensitive and quantitative test setups. METHODS We have developed two quantitative, easy-to-implement SARS-CoV-2 antibody tests, based on the spike receptor binding domain and the nucleocapsid protein. Comprehensive evaluation of antigens from several biotechnological platforms enabled the identification of superior antigen designs for reliable serodiagnostic. Cut-off modelling based on unprecedented large and heterogeneous multicentric validation cohorts allowed us to define optimal thresholds for the tests' broad applications in different aspects of clinical use, such as seroprevalence studies and convalescent plasma donor qualification. FINDINGS Both developed serotests individually performed similarly-well as fully-automated CE-marked test systems. Our described sensitivity-improved orthogonal test approach assures highest specificity (99.8%); thereby enabling robust serodiagnosis in low-prevalence settings with simple test formats. The inclusion of a calibrator permits accurate quantitative monitoring of antibody concentrations in samples collected at different time points during the acute and convalescent phase of COVID-19 and disclosed antibody level thresholds that correlate well with robust neutralization of authentic SARS-CoV-2 virus. INTERPRETATION We demonstrate that antigen source and purity strongly impact serotest performance. Comprehensive biotechnology-assisted selection of antigens and in-depth characterisation of the assays allowed us to overcome limitations of simple ELISA-based antibody test formats based on chromometric reporters, to yield comparable assay performance as fully-automated platforms. FUNDING WWTF, Project No. COV20-016; BOKU, LBI/LBG.
Collapse
Affiliation(s)
- Miriam Klausberger
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Mark Duerkop
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; Novasign GmbH Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Gordana Wozniak-Knopp
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; CD Laboratory for innovative Immunotherapeutics, Vienna, Austria
| | - Monika Cserjan-Puschmann
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; ACIB-Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Nico Lingg
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; ACIB-Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Patricia Pereira Aguilar
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; ACIB-Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Elisabeth Laurent
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; BOKU Core Facility Biomolecular & Cellular Analysis, University of Natural Resources and Life Sciences (BOKU),Vienna, Austria
| | - Jelle De Vos
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; Department of Chemical Engineering, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Manuela Hofner
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - Barbara Holzer
- Austrian Agency for Health and Food Safety (AGES), Department for Animal Health, Moedling, Austria
| | - Maria Stadler
- Institute of Immunology, University of Veterinary Medicine, Vienna, Austria
| | - Gabriele Manhart
- Institute for Medical Biochemistry, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | - Klemens Vierlinger
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - Margot Egger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz and Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Lisa Milchram
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - Elisabeth Gludovacz
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Nicolas Marx
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Christoph Köppl
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; ACIB-Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Christopher Tauer
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Jürgen Beck
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Daniel Maresch
- BOKU Core Facility Mass Spectrometry, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Clemens Grünwald-Gruber
- BOKU Core Facility Mass Spectrometry, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria; Department of Chemistry, University of Natural Resources and Life Sciences (BOKU) Vienna, Austria
| | | | - Peter Satzer
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; ACIB-Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Gerhard Stadlmayr
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; CD Laboratory for innovative Immunotherapeutics, Vienna, Austria
| | - Ulrike Vavra
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences (BOKU) Vienna, Austria
| | - Jasmin Huber
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - Markus Wahrmann
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Austria
| | - Farsad Eskandary
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Austria
| | - Marie-Kathrin Breyer
- Department of Respiratory and Critical Care Medicine and Ludwig Boltzmann Institute for Lung Health, Otto Wagner Hospital, Vienna, Austria
| | - Daniela Sieghart
- Division of Rheumatology, Department of Medicine III, Medical University of Vienna, Austria
| | - Peter Quehenberger
- Department of Laboratory Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Gerda Leitner
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Austria
| | - Robert Strassl
- Department of Laboratory Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Alexander E Egger
- Central Institute for Medical and Chemical Laboratory Diagnosis, Innsbruck University Hospital, Innsbruck, Austria
| | - Christian Irsara
- Central Institute for Medical and Chemical Laboratory Diagnosis, Innsbruck University Hospital, Innsbruck, Austria
| | - Andrea Griesmacher
- Central Institute for Medical and Chemical Laboratory Diagnosis, Innsbruck University Hospital, Innsbruck, Austria
| | - Gregor Hoermann
- Central Institute for Medical and Chemical Laboratory Diagnosis, Innsbruck University Hospital, Innsbruck, Austria; MLL Munich Leukemia Laboratory, Munich, Germany
| | - Günter Weiss
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| | - Rosa Bellmann-Weiler
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| | - Judith Loeffler-Ragg
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Richard Strasser
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences (BOKU) Vienna, Austria
| | - Alois Jungbauer
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; ACIB-Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Rainer Hahn
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; ACIB-Austrian Centre of Industrial Biotechnology, Graz, Austria
| | | | - Boris Hartmann
- Austrian Agency for Health and Food Safety (AGES), Department for Animal Health, Moedling, Austria
| | - Nikolaus B Binder
- Technoclone Herstellung von Diagnostika und Arzneimitteln GmbH, Vienna, Austria
| | - Gerald Striedner
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria; Novasign GmbH Vienna, Austria; ACIB-Austrian Centre of Industrial Biotechnology, Graz, Austria; enGenes Biotech GmbH, Vienna, Austria
| | - Lukas Mach
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences (BOKU) Vienna, Austria
| | - Andreas Weinhäusel
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - Benjamin Dieplinger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz and Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria.
| | - Wilhelm Gerner
- Institute of Immunology, University of Veterinary Medicine, Vienna, Austria; Christian Doppler Laboratory for an Optimized Prediction of Vaccination Success in Pigs, University of Veterinary Medicine, Vienna, Austria; Present address: The Pirbright Institute, Pirbright, United Kingdom
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Reingard Grabherr
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
16
|
Ubbiali D, Orlando M, Kovačič M, Iacobucci C, Semrau MS, Bajc G, Fortuna S, Ilc G, Medagli B, Oloketuyi S, Storici P, Sinz A, Grandori R, de Marco A. An anti-HER2 nanobody binds to its antigen HER2 via two independent paratopes. Int J Biol Macromol 2021; 182:502-511. [PMID: 33848543 DOI: 10.1016/j.ijbiomac.2021.04.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/10/2021] [Accepted: 04/05/2021] [Indexed: 01/02/2023]
Abstract
High-resolution structural data of complexes between antibodies and membrane receptors still represent a demanding task. In this study, we used complementary sets of experimental data to obtain a structural model of the complex formed by the human epidermal growth factor receptor 2 (HER2) and its specific nanobody A10. First we identified by NMR the residues that bind or rearrange as a consequence of the complex formation. In parallel, the complex was cross-linked, digested and the resulting peptides were characterized by mass-spectrometry to define maximal distance restraints between HER2 and A10 amino acids in their complex. These independent datasets guided a docking process, refined by molecular dynamics simulations, to develop a model of the complex and estimate per-residue free-energy contributions. Such a model explains the experimental data and identifies a second, non-canonical paratope, located in the region opposite to the conventional nanobody paratope, formed by the hypervariable loop regions LH1 and LH3. Both paratopes contributed substantially to the overall affinity by binding to independent HER2 epitopes. Nanobody mutants with substitution of key interaction residues, as indicated by the model, possess significantly lower affinity for HER2. This is the first described case of a "natural" biparatopic nanobody, directly selected by in-vitro panning.
Collapse
Affiliation(s)
- Daniele Ubbiali
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Marco Orlando
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy; Department of Biotechnology and Life Sciences, University of Insubria, Via J. H. Dunant 3, 21100 Varese, Italy
| | - Matic Kovačič
- Slovenian NMR Center, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Claudio Iacobucci
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Marta S Semrau
- Structural Biology Lab, Elettra Sincrotrone Trieste S.C.p.A., 34149, Basovizza, Trieste, Italy; CIBIO, Centre for Integrative Biology, University of Trento, via Sommarive 9, Povo 38123, Italy
| | - Gregor Bajc
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Sara Fortuna
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
| | - Gregor Ilc
- Slovenian NMR Center, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Barbara Medagli
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
| | - Sandra Oloketuyi
- Lab of Environmental and Life Sciences, University of Nova Gorica, Vipavska cesta 13, 5000 Rožna Dolina, Nova Gorica, Slovenia
| | - Paola Storici
- Structural Biology Lab, Elettra Sincrotrone Trieste S.C.p.A., 34149, Basovizza, Trieste, Italy
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Rita Grandori
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy
| | - Ario de Marco
- Lab of Environmental and Life Sciences, University of Nova Gorica, Vipavska cesta 13, 5000 Rožna Dolina, Nova Gorica, Slovenia.
| |
Collapse
|
17
|
Pinet L, Wang YH, Deville C, Lescop E, Guerlesquin F, Badache A, Bontems F, Morellet N, Durand D, Assrir N, van Heijenoort C. Structural and dynamic characterization of the C-terminal tail of ErbB2: Disordered but not random. Biophys J 2021; 120:1869-1882. [PMID: 33741354 DOI: 10.1016/j.bpj.2021.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 01/23/2023] Open
Abstract
ErbB2 (or HER2) is a receptor tyrosine kinase overexpressed in some breast cancers and associated with poor prognosis. Treatments targeting the receptor extracellular and kinase domains have greatly improved disease outcome in the last 20 years. In parallel, the structures of these domains have been described, enabling better mechanistic understanding of the receptor function and targeted inhibition. However, the ErbB2 disordered C-terminal cytoplasmic tail (CtErbB2) remains very poorly characterized in terms of structure, dynamics, and detailed functional mechanism. Yet, it is where signal transduction is triggered via phosphorylation of tyrosine residues and carried out via interaction with adaptor proteins. Here, we report the first description, to our knowledge, of the ErbB2 disordered tail at atomic resolution using NMR, complemented by small-angle x-ray scattering. We show that although no part of CtErbB2 has any fully populated secondary or tertiary structure, it contains several transient α-helices and numerous transient polyproline II helices, populated up to 20 and 40%, respectively, and low but significant compaction. The presence of some structural elements suggests, along the lines of the results obtained for EGFR (ErbB1), that they may have a functional role in ErbB2's autoregulation processes. In addition, the transient formation of polyproline II helices is compliant with previously suggested interactions with SH3 domains. All in all, our in-depth structural study opens perspectives in the mechanistic understanding of ErbB2.
Collapse
Affiliation(s)
- Louise Pinet
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Gif-sur-Yvette, France; Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Ying-Hui Wang
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Gif-sur-Yvette, France; SGS Taiwan LTD, New Taipei City, Taiwan
| | - Célia Deville
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Gif-sur-Yvette, France; IGBMC, University of Strasbourg, CNRS UMR, Illkirch, France
| | - Ewen Lescop
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Françoise Guerlesquin
- LISM, Institut de Microbiologie de la Méditerranée, CNRS and Aix-Marseille University, Marseille, France
| | - Ali Badache
- Centre de Recherche en Cancérologie de Marseille, Aix-Marseille Univ, INSERM, Institut Paoli-Calmettes, CNRS, Marseille, France
| | - François Bontems
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Gif-sur-Yvette, France; Department of Virology, Institut Pasteur, CNRS UMR 3569, Paris, France
| | - Nelly Morellet
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Dominique Durand
- I2BC, Université Paris-Saclay, CNRS UMR 9198, Gif-sur-Yvette, France
| | - Nadine Assrir
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Carine van Heijenoort
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Gif-sur-Yvette, France.
| |
Collapse
|
18
|
Benedetti F, Stracke F, Stadlmayr G, Stadlbauer K, Rüker F, Wozniak-Knopp G. Bispecific antibodies with Fab-arms featuring exchanged antigen-binding constant domains. Biochem Biophys Rep 2021; 26:100959. [PMID: 33718630 PMCID: PMC7920882 DOI: 10.1016/j.bbrep.2021.100959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/27/2022] Open
Abstract
Monoclonal antibodies can acquire the property of engagement of a second antigen via fusion methods or modification of their CDR loops, but also by modification of their constant domains, such as in the mAb2 format where a set of mutated amino acid residues in the CH3 domains enables a high-affinity specific interaction with the second antigen. We tested the possibility of introducing multiple binding sites for the second antigen by replacing the Fab CH1/CL domain pair with a pair of antigen-binding CH3 domains in a model scaffold with trastuzumab variable domains and VEGF-binding CH3 domains. Such bispecific molecules were produced in a “Fab-like” format and in a full-length antibody format. Novel constructs were of expected molecular composition using mass spectrometry. They were expressed at a high level in standard laboratory conditions, purified as monomers with Protein A and gel filtration and were of high thermostability. Their high-affinity binding to both target antigens was retained. Finally, the Her2/VEGF binding domain-exchanged bispecific antibody was able to mediate a potentiated surface Her2-internalization effect on the Her2-overexpressing cell line SK-BR-3 due to improved level of cross-linking with the endogenously secreted cytokine. To conclude, bispecific antibodies with Fabs featuring exchanged antigen-binding CH3 domains offer an alternative solution in positioning and valency of antigen binding sites. Fab constant domains can be efficiently exchanged for antigen-binding CH3 domains. Such mutagenesis results in bispecific antibodies with correct chain pairing. Domain-exchanged bispecific Fab- and IgG-like formats are of favorable biophysical properties. Resulting bispecific antibodies show high-affinity binding to both target antigens.
Collapse
Key Words
- Ab, antibody
- BLI, biolayer interferometry
- BSA, bovine serum albumin
- Bispecific antibody
- CDR, complementarity determining region
- DSC, differential scanning calorimetry
- Domain-exchanged antibody
- EC50, half-maximal effective concentration
- FBS, fetal bovine serum
- FITC, fluorescein isothiocyanate
- Fab constant domain exchange
- Fab, fragment antigen binding
- Fc, fragment crystallizable
- Fcab, Fc with antigen binding properties
- HPLC-SEC, high pressure liquid chromatography-size exclusion chromatography
- Her2 internalization
- IgG, immunoglobulin G
- LC-ESI-MS, liquid chromatography-electrospray ionization-mass spectrometry
- PBS, phosphate buffered saline
- PE, phycoerythrin
- PEI, polyethylenimine
- PNGase F, Peptide:N-glycosidase F
- RMSD, root mean square deviation
- TRA, trastuzumab
- Tm, melting temperature
- VEGF, vascular endothelial growth factor
- “Knobs-into-holes” heterodimerization
Collapse
Affiliation(s)
- Filippo Benedetti
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Florian Stracke
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Gerhard Stadlmayr
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Katharina Stadlbauer
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Florian Rüker
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Gordana Wozniak-Knopp
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| |
Collapse
|
19
|
Song Y, Qian Y, Huang Z, Khattak SF, Li ZJ. Computational insights into O-glycosylation in a CTLA4 Fc-fusion protein linker and its impact on protein quality attributes. Comput Struct Biotechnol J 2020; 18:3925-3935. [PMID: 33335689 PMCID: PMC7734232 DOI: 10.1016/j.csbj.2020.11.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
The hinge region of immunoglobulin G1 (IgG1) is used as a common linker for Fc-fusion therapeutic proteins. With the advances of high-resolution mass spectrometry and sample treatment strategies, unexpected O-linked glycosylation has been observed in the linker. However, the molecular mechanism involved in this unusual posttranslational modification is unknown. In this study, we applied site-direct mutagenesis, mass spectrometry, analytical chromatography, and computational modeling to investigate O-glycosylation processes in a clinically used CTLA4 Fc-fusion protein and its impacts on protein quality attributes. Surprisingly, O-glycans could be formed at new sites when an initial O-glycosylation site was eliminated, and continued to occur until all potential O-glycosylation sites were nulled. Site-preference of O-glycosylation initiation was attributed to the complex formation between the linker peptide and glycan transferase whereas the O-glycosylating efficiency and the linker flexibility were correlated using molecular modeling and simulations. As predicted, O-glycan-free CTLA4 Fc-fusion proteins were more homogenous for sialylation, and interestingly less prone to protein aggregation. Attenuating protein aggregation was a desirable effect, and could be related to the reduced presence of linker O-glycans that hindered inter-chain disulfide bond reformation. Findings from this study shed light on new therapeutic protein design and development.
Collapse
Affiliation(s)
- Yuanli Song
- Biologics Development, Bristol Myers Squibb Company, 38 Jackson Road, Devens, MA 01434, USA
| | - Yueming Qian
- Biologics Development, Bristol Myers Squibb Company, 38 Jackson Road, Devens, MA 01434, USA
| | - Zhe Huang
- Biologics Development, Bristol Myers Squibb Company, 38 Jackson Road, Devens, MA 01434, USA
| | - Sarwat F Khattak
- Biologics Development, Bristol Myers Squibb Company, 38 Jackson Road, Devens, MA 01434, USA
| | - Zheng Jian Li
- Biologics Development, Bristol Myers Squibb Company, 38 Jackson Road, Devens, MA 01434, USA
| |
Collapse
|
20
|
Shibata-Seki T, Nagaoka M, Goto M, Kobatake E, Akaike T. Direct visualization of the extracellular binding structure of E-cadherins in liquid. Sci Rep 2020; 10:17044. [PMID: 33046720 PMCID: PMC7552386 DOI: 10.1038/s41598-020-72517-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/19/2020] [Indexed: 12/16/2022] Open
Abstract
E-cadherin is a key Ca-dependent cell adhesion molecule, which is expressed on many cell surfaces and involved in cell morphogenesis, embryonic development, EMT, etc. The fusion protein E-cad-Fc consists of the extracellular domain of E-cadherin and the IgG Fc domain. On plates coated with this chimeric protein, ES/iPS cells are cultivated particularly well and induced to differentiate. The cells adhere to the plate via E-cad-Fc in the presence of Ca2+ and detach by a chelating agent. For the purpose of clarifying the structures of E-cad-Fc in the presence and absence of Ca2+, we analyzed the molecular structure of E-cad-Fc by AFM in liquid. Our AFM observations revealed a rod-like structure of the entire extracellular domain of E-cad-Fc in the presence of Ca2+ as well as trans-binding of E-cad-Fc with adjacent molecules, which may be the first, direct confirmation of trans-dimerization of E-cadherin. The observed structures were in good agreement with an X-ray crystallographic model. Furthermore, we succeeded in visualizing the changes in the rod-like structure of the EC domains with and without calcium. The biomatrix surface plays an important role in cell culture, so the analysis of its structure and function may help promote cell engineering based on cell recognition.
Collapse
Affiliation(s)
- Teiko Shibata-Seki
- Biomaterials Center for Regenerative Medical Engineering, Foundation for Advancement of International Science, 24-16 Kasuga, 3-chome, Tsukuba, Ibaraki, 305-0821, Japan
| | - Masato Nagaoka
- Biomaterials Center for Regenerative Medical Engineering, Foundation for Advancement of International Science, 24-16 Kasuga, 3-chome, Tsukuba, Ibaraki, 305-0821, Japan
| | - Mitsuaki Goto
- Biomaterials Center for Regenerative Medical Engineering, Foundation for Advancement of International Science, 24-16 Kasuga, 3-chome, Tsukuba, Ibaraki, 305-0821, Japan.
| | - Eiry Kobatake
- School of Life Science and Technology, Tokyo Institute of Technology, G1-13, 4259, Nagatsuta, Midori-ku, Yokohama, Kanagawa, 226-8502, Japan
| | - Toshihiro Akaike
- Biomaterials Center for Regenerative Medical Engineering, Foundation for Advancement of International Science, 24-16 Kasuga, 3-chome, Tsukuba, Ibaraki, 305-0821, Japan
| |
Collapse
|
21
|
Reimann L, Schwäble AN, Fricke AL, Mühlhäuser WWD, Leber Y, Lohanadan K, Puchinger MG, Schäuble S, Faessler E, Wiese H, Reichenbach C, Knapp B, Peikert CD, Drepper F, Hahn U, Kreutz C, van der Ven PFM, Radziwill G, Djinović-Carugo K, Fürst DO, Warscheid B. Phosphoproteomics identifies dual-site phosphorylation in an extended basophilic motif regulating FILIP1-mediated degradation of filamin-C. Commun Biol 2020; 3:253. [PMID: 32444788 PMCID: PMC7244511 DOI: 10.1038/s42003-020-0982-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/01/2020] [Indexed: 01/10/2023] Open
Abstract
The PI3K/Akt pathway promotes skeletal muscle growth and myogenic differentiation. Although its importance in skeletal muscle biology is well documented, many of its substrates remain to be identified. We here studied PI3K/Akt signaling in contracting skeletal muscle cells by quantitative phosphoproteomics. We identified the extended basophilic phosphosite motif RxRxxp[S/T]xxp[S/T] in various proteins including filamin-C (FLNc). Importantly, this extended motif, located in a unique insert in Ig-like domain 20 of FLNc, is doubly phosphorylated. The protein kinases responsible for this dual-site phosphorylation are Akt and PKCα. Proximity proteomics and interaction analysis identified filamin A-interacting protein 1 (FILIP1) as direct FLNc binding partner. FILIP1 binding induces filamin degradation, thereby negatively regulating its function. Here, dual-site phosphorylation of FLNc not only reduces FILIP1 binding, providing a mechanism to shield FLNc from FILIP1-mediated degradation, but also enables fast dynamics of FLNc necessary for its function as signaling adaptor in cross-striated muscle cells.
Collapse
Affiliation(s)
- Lena Reimann
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Anja N Schwäble
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Anna L Fricke
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Wignand W D Mühlhäuser
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Yvonne Leber
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121, Bonn, Germany
| | - Keerthika Lohanadan
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121, Bonn, Germany
| | - Martin G Puchinger
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, A-1030, Vienna, Austria
| | - Sascha Schäuble
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Erik Faessler
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Heike Wiese
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Institute of Pharmacology and Toxicology, University of Ulm, 89081, Ulm, Germany
| | - Christa Reichenbach
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Bettina Knapp
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Christian D Peikert
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Bioinformatics Research & Development, BioNTech SE, 55131, Mainz, Germany
| | - Friedel Drepper
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Udo Hahn
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Clemens Kreutz
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, 79104, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121, Bonn, Germany
| | - Gerald Radziwill
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Kristina Djinović-Carugo
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, A-1030, Vienna, Austria
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121, Bonn, Germany
| | - Bettina Warscheid
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
22
|
Chromatographic assay to probe the binding energy and mechanisms of homologous proteins to surface-bound ligands. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1136:121927. [DOI: 10.1016/j.jchromb.2019.121927] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/08/2019] [Accepted: 12/03/2019] [Indexed: 01/01/2023]
|
23
|
Göritzer K, Turupcu A, Maresch D, Novak J, Altmann F, Oostenbrink C, Obinger C, Strasser R. Distinct Fcα receptor N-glycans modulate the binding affinity to immunoglobulin A (IgA) antibodies. J Biol Chem 2019; 294:13995-14008. [PMID: 31362986 PMCID: PMC6755811 DOI: 10.1074/jbc.ra119.009954] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/23/2019] [Indexed: 01/10/2023] Open
Abstract
Human immunoglobulin A (IgA) is the most prevalent antibody class at mucosal sites with an important role in mucosal defense. Little is known about the impact of N-glycan modifications of IgA1 and IgA2 on binding to the Fcα receptor (FcαRI), which is also heavily glycosylated at its extracellular domain. Here, we transiently expressed human epidermal growth factor receptor 2 (HER2)-binding monomeric IgA1, IgA2m(1), and IgA2m(2) variants in Nicotiana benthamiana ΔXT/FT plants lacking the enzymes responsible for generating nonhuman N-glycan structures. By coinfiltrating IgA with the respective glycan-modifying enzymes, we generated IgA carrying distinct homogenous N-glycans. We demonstrate that distinctly different N-glycan profiles did not influence antigen binding or the overall structure and integrity of the IgA antibodies but did affect their thermal stability. Using size-exclusion chromatography, differential scanning and isothermal titration calorimetry, surface plasmon resonance spectroscopy, and molecular modeling, we probed distinct IgA1 and IgA2 glycoforms for binding to four different FcαRI glycoforms and investigated the thermodynamics and kinetics of complex formation. Our results suggest that different N-glycans on the receptor significantly contribute to binding affinities for its cognate ligand. We also noted that full-length IgA and FcαRI form a mixture of 1:1 and 1:2 complexes tending toward a 1:1 stoichiometry due to different IgA tailpiece conformations that make it less likely that both binding sites are simultaneously occupied. In conclusion, N-glycans of human IgA do not affect its structure and integrity but its thermal stability, and FcαRI N-glycans significantly modulate binding affinity to IgA.
Collapse
Affiliation(s)
- Kathrin Göritzer
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Aysegül Turupcu
- Department of Material Sciences and Process Engineering, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Daniel Maresch
- Department of Chemistry, Division of Biochemistry, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Jan Novak
- Department of Microbiology, University of Alabama, Birmingham, Alabama 35294
| | - Friedrich Altmann
- Department of Chemistry, Division of Biochemistry, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Chris Oostenbrink
- Department of Material Sciences and Process Engineering, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Christian Obinger
- Department of Chemistry, Division of Biochemistry, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Richard Strasser
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| |
Collapse
|
24
|
Yeast Surface Display and Cell Sorting of Antigen-Binding Fc Fragments. Methods Mol Biol 2019. [PMID: 30737746 DOI: 10.1007/978-1-4939-9024-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Since the introduction of the yeast display platform, this method has increasingly gained popularity for the discovery and affinity maturation of antibodies and other protein scaffolds intended for antigen recognition. Yeast display is particularly well suited for the selection of antigen-binding Fc fragments (Fcabs) as it allows rapid combinatorial library construction via gap repair-driven homologous recombination and an efficient display of a glycosylated Fc able to interact with Fcγ receptors. Apart from expression-related normalization, isolation of properly folded Fcabs can be guided efficiently by simultaneous staining with ligands such as protein A, FcγRI, or the conformation-sensitive anti-FigCH2 antibody, whose binding is critically dependent on the integrity of the Fc structure. The particular properties of the Fcab scaffold, such as its homodimeric state which can promote binding to multiple antigen molecules, require modifications of traditional affinity maturation strategies. Preferred to equilibrium selections are kinetically driven antigen selections, designed to specifically influence the binding off-rate, which in many cases augments the desired biological effect. A simple design of a yeast-displayed heterodimeric Fc fragment is described and can be used as a general guideline for affinity selection of Fcabs with an asymmetric binding site. Overall, this chapter underlines the importance of the versatile yeast display technique for the optimization of the novel Fcab scaffold for antigen recognition.
Collapse
|
25
|
Everett KL, Kraman M, Wollerton FPG, Zimarino C, Kmiecik K, Gaspar M, Pechouckova S, Allen NL, Doody JF, Tuna M. Generation of Fcabs targeting human and murine LAG-3 as building blocks for novel bispecific antibody therapeutics. Methods 2018; 154:60-69. [PMID: 30208333 DOI: 10.1016/j.ymeth.2018.09.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/31/2018] [Accepted: 09/04/2018] [Indexed: 01/07/2023] Open
Abstract
The immunoglobulin superfamily protein lymphocyte-activation gene 3 (LAG-3) participates in immune suppression and has been identified as a suitable target for cancer therapies. In order to generate bispecific antibodies targeting LAG-3, Fcabs (Fc-region with antigen binding) targeting human and murine LAG-3 were generated from phage libraries. These Fcabs bind to LAG-3, inhibiting its interaction with MHC class II, and induce IL-2 production in a T cell assay. Bispecific antibodies, known as mAb2, were produced by replacing the Fc region of a monoclonal antibody with Fcab sequences in the CH3 domain. mAb2 containing anti-LAG-3 Fcabs have mAb-like biophysical characteristics and retain LAG-3 binding and functional activity. mAb2 can thus be generated using multiple Fabs to investigate bispecific parings and develop novel therapeutics.
Collapse
Affiliation(s)
- Katy L Everett
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom.
| | - Matthew Kraman
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Francisca P G Wollerton
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Carlo Zimarino
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Katarzyna Kmiecik
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Miguel Gaspar
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Sarka Pechouckova
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Natalie L Allen
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Jacqueline F Doody
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Mihriban Tuna
- F-star Biotechnology Ltd, Eddeva B920, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| |
Collapse
|
26
|
Chakravarty S, Ung AR, Moore B, Shore J, Alshamrani M. A Comprehensive Analysis of Anion-Quadrupole Interactions in Protein Structures. Biochemistry 2018; 57:1852-1867. [PMID: 29482321 PMCID: PMC6051350 DOI: 10.1021/acs.biochem.7b01006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The edgewise interactions of anions with phenylalanine (Phe) aromatic rings in proteins, known as anion-quadrupole interactions, have been well studied. However, the anion-quadrupole interactions of the tyrosine (Tyr) and tryptophan (Trp) rings have been less well studied, probably because these have been considered weaker than interactions of anions hydrogen bonded to Trp/Tyr side chains. Distinguishing such hydrogen bonding interactions, we comprehensively surveyed the edgewise interactions of certain anions (aspartate, glutamate, and phosphate) with Trp, Tyr, and Phe rings in high-resolution, nonredundant protein single chains and interfaces (protein-protein, DNA/RNA-protein, and membrane-protein). Trp/Tyr anion-quadrupole interactions are common, with Trp showing the highest propensity and average interaction energy for this type of interaction. The energy of an anion-quadrupole interaction (-15.0 to 0.0 kcal/mol, based on quantum mechanical calculations) depends not only on the interaction geometry but also on the ring atom. The phosphate anions at DNA/RNA-protein interfaces interact with aromatic residues with energies comparable to that of aspartate/glutamate anion-quadrupole interactions. At DNA-protein interfaces, the frequency of aromatic ring participation in anion-quadrupole interactions is comparable to that of positive charge participation in salt bridges, suggesting an underappreciated role for anion-quadrupole interactions at DNA-protein (or membrane-protein) interfaces. Although less frequent than salt bridges in single-chain proteins, we observed highly conserved anion-quadrupole interactions in the structures of remote homologues, and evolutionary covariance-based residue contact score predictions suggest that conserved anion-quadrupole interacting pairs, like salt bridges, contribute to polypeptide folding, stability, and recognition.
Collapse
Affiliation(s)
- Suvobrata Chakravarty
- Chemistry & Biochemistry, South Dakota State University, Brookings, SD, USA, 57007
- BioSNTR, Brookings, SD, USA, 57007
| | - Adron R. Ung
- Chemistry & Biochemistry, South Dakota State University, Brookings, SD, USA, 57007
| | - Brian Moore
- University Networking and Research Computing, South Dakota State University, Brookings, SD, USA, 57007
| | - Jay Shore
- Chemistry & Biochemistry, South Dakota State University, Brookings, SD, USA, 57007
| | - Mona Alshamrani
- Chemistry & Biochemistry, South Dakota State University, Brookings, SD, USA, 57007
| |
Collapse
|
27
|
Lobner E, Humm AS, Mlynek G, Kubinger K, Kitzmüller M, Traxlmayr MW, Djinović-Carugo K, Obinger C. Two-faced Fcab prevents polymerization with VEGF and reveals thermodynamics and the 2.15 Å crystal structure of the complex. MAbs 2017; 9:1088-1104. [PMID: 28816592 PMCID: PMC5627596 DOI: 10.1080/19420862.2017.1364825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Fcabs (Fc domain with antigen-binding sites) are promising novel therapeutics. By engineering of the C-terminal loops of the CH3 domains, 2 antigen binding sites can be inserted in close proximity. To elucidate the binding mode(s) between homodimeric Fcabs and small homodimeric antigens, the interaction between the Fcabs 448 and CT6 (having the AB, CD and EF loops and the C-termini engineered) with homodimeric VEGF was investigated. The crystal structures of these Fcabs, which form polymers with the antigen VEGF in solution, were determined. However, construction of heterodimeric Fcabs (JanusFcabs: one chain Fc-wt, one chain VEGF-binding) results in formation of distinct JanusFcab–VEGF complexes (2:1), which allowed elucidation of the crystal structure of the JanusCT6–VEGF complex at 2.15 Å resolution. VEGF binding to Janus448 and JanusCT6 is shown to be entropically unfavorable, but enthalpically favorable. Structure-function relationships are discussed with respect to Fcab design and engineering strategies.
Collapse
Affiliation(s)
- Elisabeth Lobner
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| | - Anne-Sophie Humm
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,c Department for Structural and Computational Biology , Max F. Perutz Laboratories, University of Vienna , Dr. Bohr-Gasse 9, Vienna , Austria
| | - Georg Mlynek
- c Department for Structural and Computational Biology , Max F. Perutz Laboratories, University of Vienna , Dr. Bohr-Gasse 9, Vienna , Austria
| | - Konstantin Kubinger
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| | - Michael Kitzmüller
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| | - Michael W Traxlmayr
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| | - Kristina Djinović-Carugo
- c Department for Structural and Computational Biology , Max F. Perutz Laboratories, University of Vienna , Dr. Bohr-Gasse 9, Vienna , Austria.,d Department of Biochemistry, Faculty of Chemistry and Chemical Technology , University of Ljubljana , Večna pot 113, Ljubljana , Slovenia
| | - Christian Obinger
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| |
Collapse
|
28
|
Vega JF, Ramos J, Cruz VL, Vicente-Alique E, Sánchez-Sánchez E, Sánchez-Fernández A, Wang Y, Hu P, Cortés J, Martínez-Salazar J. Molecular and hydrodynamic properties of human epidermal growth factor receptor HER2 extracellular domain and its homodimer: Experiments and multi-scale simulations. Biochim Biophys Acta Gen Subj 2017. [PMID: 28642126 DOI: 10.1016/j.bbagen.2017.06.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND In a broad range of human carcinomas gene amplification leads to HER2 overexpression, which has been proposed to cause spontaneous dimerization and activation in the absence of ligand. This makes HER2 attractive as a therapeutic target. However, the HER2 homodimerization mechanism remains unexplored. It has been suggested that the "back-to-back" homodimer does not form in solution. Notwithstanding, very recently the crystal structure of the HER2 extracellular domain homodimer formed with a "back-to-head" interaction has been resolved. We intend to explore the existence of such interactions. METHODS A combination of experiments, molecular dynamics and hydrodynamic modeling were used to monitor the transport properties of HER2 in solution. RESULTS & CONCLUSIONS We have detected the HER2 extracellular domain homodimer in solution. The results show a high degree of molecular flexibility, which ultimately leads to quite higher values of the intrinsic viscosity and lower values of diffusion coefficient than those corresponding to globular proteins. This flexibility obeys to the open conformation of the receptor and to the large fluctuations of the different domains. We also report that for obtaining the correct hydrodynamic constants from the modeling one must consider the glycosylation of the systems. GENERAL SIGNIFICANCE Conformational features of epidermal growth factor receptors regulate their hydrodynamic properties and control their activity. It is essential to understand the dynamics of these systems and the role of the specific domains involved. To find biophysical correlations between dynamics and macroscopic transport properties is of general interest for researches working in this area. This article is part of a Special Issue entitled "Biochemistry of Synthetic Biology - Recent Developments" Guest Editor: Dr. Ilka Heinemann and Dr. Patrick O'Donoghue.
Collapse
Affiliation(s)
- J F Vega
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/ Serrano 113 bis, 28006 Madrid, Spain.
| | - J Ramos
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/ Serrano 113 bis, 28006 Madrid, Spain
| | - V L Cruz
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/ Serrano 113 bis, 28006 Madrid, Spain
| | - E Vicente-Alique
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/ Serrano 113 bis, 28006 Madrid, Spain
| | - E Sánchez-Sánchez
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/ Serrano 113 bis, 28006 Madrid, Spain
| | - A Sánchez-Fernández
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/ Serrano 113 bis, 28006 Madrid, Spain
| | - Y Wang
- Sino Biological, Inc., Beijing, People's Republic of China
| | - P Hu
- Sino Biological, Inc., Beijing, People's Republic of China
| | - J Cortés
- Ramon y Cajal University Hospital, Ctra. de Colmenar Viejo, km 9,100, 28034 Madrid, Spain; Vall D'Hebron Institute of Oncology (VHIO), Paseo Vall Hebron 119-129, 08035 Barcelona, Spain
| | - J Martínez-Salazar
- BIOPHYM, Department of Macromolecular Physics, Instituto de Estructura de la Materia, IEM-CSIC, C/ Serrano 113 bis, 28006 Madrid, Spain
| |
Collapse
|