1
|
Mattson Cypert BK, Menard K, Chu G, McDevitt T, Verona RI, Rupnow B, Packman K. Mechanisms of Response and Resistance to PSMA×CD3 Bispecifics in CD34+ Humanized Mice. Mol Cancer Ther 2025; 24:740-752. [PMID: 40008870 DOI: 10.1158/1535-7163.mct-23-0779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/06/2024] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Prostate cancer is considered immunologically "cold," with low mutational burden, tumor-infiltrating immune cells, and PD-L1 levels, culminating in poor response to immune checkpoint therapies. CD3 bispecific redirection antibodies can elicit T cell-mediated cytotoxicity and hold promise for immune cell recruitment into prostate tumors. CD3 redirection antibodies in solid tumors are still in the early phases of clinical development, and it is not yet understood whether these potential therapies will achieve the high response rates observed in hematologic malignancies or result in durable T-cell responses. In this study, we demonstrated that treatment with a prostate-specific membrane antigen (PSMA)-targeted CD3 redirector resulted in efficacy against LnCaP.AR human prostate xenografts in CD34+ cord blood-humanized mice. Efficacy correlated with T-cell infiltration into tumors with an activated phenotype and also increased PD-L1 expression. Engineered overexpression of PD-L1 in LNCaP.AR tumors resulted in resistance to PSMA×CD3 bispecific antibody treatment, whereas sensitivity was restored in combination with anti-PD-1 antibody pembrolizumab. PSMA×CD3 and anti-PD-1 combination treatment resulted in complete tumor responses in approximately 20% of mice and elicited immune responses that delayed growth of rechallenged tumors. In a second prostate model, patient-derived LuCaP 86.2 xenografts, PSMA×CD3 monotherapy treatment resulted in complete responses in 25% of mice. When PSMA×CD3-treated responder mice were rechallenged with LuCaP 86.2 tumors, partial control of tumor regrowth was associated with the expansion of effector memory T cells. These studies show that PSMA×CD3 treatment elicits antitumor memory T-cell responses and that combination with PD-1 blockade can enhance these effects in tumors with immune-suppressive tumor microenvironments.
Collapse
Affiliation(s)
| | - Krista Menard
- Janssen Research and Development, Spring House, Pennsylvania
| | - Gerald Chu
- Janssen Research and Development, Spring House, Pennsylvania
| | | | - Raluca I Verona
- Janssen Research and Development, Spring House, Pennsylvania
| | - Brent Rupnow
- Janssen Research and Development, Spring House, Pennsylvania
| | | |
Collapse
|
2
|
Chen M, Zhou Y, Bao K, Chen S, Song G, Wang S. Multispecific Antibodies Targeting PD-1/PD-L1 in Cancer. BioDrugs 2025; 39:427-444. [PMID: 40106158 DOI: 10.1007/s40259-025-00712-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
The development of immune checkpoint inhibitors has revolutionized the treatment of patients with cancer. Targeting the programmed cell death protein 1 (PD-1)/programmed cell death 1 ligand 1(PD-L1) interaction using monoclonal antibodies has emerged as a prominent focus in tumor therapy with rapid advancements. However, the efficacy of anti-PD-1/PD-L1 treatment is hindered by primary or acquired resistance, limiting the effectiveness of single-drug approaches. Moreover, combining PD-1/PD-L1 with other immune drugs, targeted therapies, or chemotherapy significantly enhances response rates while exacerbating adverse reactions. Multispecific antibodies, capable of binding to different epitopes, offer improved antitumor efficacy while reducing drug-related side effects, serving as a promising therapeutic approach in cancer treatment. Several bispecific antibodies (bsAbs) targeting PD-1/PD-L1 have received regulatory approval, and many more are currently in clinical development. Additionally, tri-specific antibodies (TsAbs) and tetra-specific antibodies (TetraMabs) are under development. This review comprehensively explores the fundamental structure, preclinical principles, clinical trial progress, and challenges associated with bsAbs targeting PD-1/PD-L1.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Yuli Zhou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kaicheng Bao
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Siyu Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Guoqing Song
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| |
Collapse
|
3
|
Radić L, Offersgaard A, Kadavá T, Zon I, Capella-Pujol J, Mulder F, Koekkoek S, Spek V, Chumbe A, Bukh J, van Gils MJ, Sanders RW, Yin VC, Heck AJR, Gottwein JM, Sliepen K, Schinkel J. Bispecific antibodies against the hepatitis C virus E1E2 envelope glycoprotein. Proc Natl Acad Sci U S A 2025; 122:e2420402122. [PMID: 40193609 PMCID: PMC12012487 DOI: 10.1073/pnas.2420402122] [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: 10/05/2024] [Accepted: 02/19/2025] [Indexed: 04/09/2025] Open
Abstract
Hepatitis C virus (HCV) currently causes about one million infections and 240,000 deaths worldwide each year. To reach the goal set by the World Health Organization of global HCV elimination by 2030, it is critical to develop a prophylactic vaccine. Broadly neutralizing antibodies (bNAbs) target the E1E2 envelope glycoproteins on the viral surface, can neutralize a broad range of the highly diverse circulating HCV strains, and are essential tools to inform vaccine design. However, bNAbs targeting a single E1E2 epitope might be limited in neutralization breadth, which can be enhanced by using combinations of bNAbs that target different envelope epitopes. We have generated 60 immunoglobulin G (IgG)-like bispecific antibodies (bsAbs) that can simultaneously target two distinct epitopes on E1E2. We combine non- or partially overlapping E1E2 specificities into three types of bsAbs, each containing a different hinge length. The majority of bsAbs shows retained or increased potency and breadth against a diverse panel of HCV pseudoparticles and HCV produced in cell culture compared to monospecific and cocktail controls. Additionally, we demonstrate that changes in the hinge length of bsAbs can alter the binding stoichiometry to E1E2. These results provide insights into the binding modes and the role of avidity in bivalent targeting of diverse E1E2 epitopes.This study illustrates how potential cooperative effects of HCV bNAbs can be utilized by strategically designing bispecific constructs. These HCV bsAbs can guide vaccine development and unlock novel therapeutic and prophylactic strategies against HCV and other (flavi)viruses.
Collapse
Affiliation(s)
- Laura Radić
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| | - Anna Offersgaard
- Copenhagen Hepatitis C Program, Department of Infectious Diseases, Copenhagen University Hospital–Hvidovre, Hvidovre2650, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N2200, Denmark
| | - Tereza Kadavá
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Ian Zon
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| | - Joan Capella-Pujol
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| | - Fabian Mulder
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| | - Sylvie Koekkoek
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| | - Vera Spek
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| | - Ana Chumbe
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| | - Jens Bukh
- Copenhagen Hepatitis C Program, Department of Infectious Diseases, Copenhagen University Hospital–Hvidovre, Hvidovre2650, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N2200, Denmark
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| | - Rogier W. Sanders
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY10065
| | - Victor C. Yin
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Judith M. Gottwein
- Copenhagen Hepatitis C Program, Department of Infectious Diseases, Copenhagen University Hospital–Hvidovre, Hvidovre2650, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N2200, Denmark
| | - Kwinten Sliepen
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| | - Janke Schinkel
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam1105 AZ, the Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam1105 AZ, the Netherlands
| |
Collapse
|
4
|
Wen J, Cui W, Yin X, Chen Y, Liu A, Wang Q, Meng X. Application and future prospects of bispecific antibodies in the treatment of non-small cell lung cancer. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0470. [PMID: 40192238 PMCID: PMC12032835 DOI: 10.20892/j.issn.2095-3941.2024.0470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/05/2025] [Indexed: 04/29/2025] Open
Abstract
As the leading cause of cancer-related deaths, lung cancer remains a noteworthy threat to human health. Although immunotherapies, such as immune checkpoint inhibitors (ICIs), have significantly increased the efficacy of lung cancer treatment, a significant percentage of patients are not sensitive to immunotherapies and patients who initially respond to treatment can quickly develop acquired drug resistance. Bispecific antibodies (bsAbs) bind two different antigens or epitopes simultaneously and have been shown to enhance antitumor efficacy with suitable safety profiles, thus attracting increasing attention as novel antitumor therapies. At present, in addition to the approved bsAb, amivantamab, three novel bsAbs (KN046, AK112, and SHR-1701) are being evaluated in phase 3 clinical trials and many bsAbs are being evaluated in phase 1/2 clinical trials for patients with non-small cell lung cancer (NSCLC). Herein we present the structure, classification, and mechanism of action underlying bsAbs in NSCLC and introduce related clinical trials. Finally, we discuss challenges, potential solutions, and future prospects in the context of cancer treatment with bsAbs.
Collapse
Affiliation(s)
- Junxu Wen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Wenxing Cui
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Yu Chen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Ailing Liu
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Qian Wang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
5
|
Liao C, Engelberts P, van Dijk M, Timmermans A, Martens JWM, Neubert E, Danen EH. CD3xHER2 bsAb-Mediated Activation of Resting T-cells at HER2 Positive Tumor Clusters Is Sufficient to Trigger Bystander Eradication of Distant HER2 Negative Clusters Through IFNγ and TNFα. Eur J Immunol 2025; 55:e202451589. [PMID: 40178291 PMCID: PMC11967296 DOI: 10.1002/eji.202451589] [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: 10/21/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
Bispecific antibodies (bsAbs) bridging CD3 on T-cells to tumor-associated antigens (TAA) on tumor cells can direct T-cell immunity to solid tumors. "Bystander killing", where T-cell targeting of TAA-positive tumor cells also leads to the eradication of TAA-negative cells, may overcome TAA heterogeneity. While bystander activity of activated, engineered T-cells has been shown to be robust and wide-reaching, spatiotemporal aspects of bsAb-mediated bystander activity are unresolved. Here, we developed a model where breast cancer tumoroids varying in HER2 expression were printed in to extracellular matrix (ECM) scaffolds. We generated (1) mixed tumors containing different ratios of HER2+ and HER2- tumor cells, and (2) HER2+ and HER2- tumoroids spaced at different distances within the ECM scaffold. Subsequently, tumors were exposed to peripheral blood-derived T-cells in the presence of CD3xHER2 bsAbs. We find that CD3xHER2 bsAb-mediated interaction of resting, nonactivated T-cells with HER2+ tumor cells is sufficient (1) to eliminate 50% HER2- cells in mixed tumor areas, and (2) to eradicate distant HER2- tumor areas. Such bystander killing involves paracrine IFNγ and TNFα activity but does not require T-cell accumulation in HER2- areas. These findings indicate that bystander eradication of TAA-negative cells can significantly contribute to bsAb therapy for solid tumors.
Collapse
Affiliation(s)
- Chen‐Yi Liao
- Leiden Academic Centre for Drug ResearchLeiden UniversityLeidenthe Netherlands
| | | | - Michiel van Dijk
- Leiden Academic Centre for Drug ResearchLeiden UniversityLeidenthe Netherlands
| | - Annemieke Timmermans
- Department of Medical OncologyErasmus MC Cancer InstituteErasmus University Medical CenterRotterdamthe Netherlands
| | - John W. M. Martens
- Department of Medical OncologyErasmus MC Cancer InstituteErasmus University Medical CenterRotterdamthe Netherlands
| | - Elsa Neubert
- Leiden Academic Centre for Drug ResearchLeiden UniversityLeidenthe Netherlands
| | - Erik H.J. Danen
- Leiden Academic Centre for Drug ResearchLeiden UniversityLeidenthe Netherlands
| |
Collapse
|
6
|
Sewnath CAN, Damelang T, Bentlage AEH, Ten Kroode L, Tuk CW, Visser R, Wuhrer M, Van Coillie J, Rispens T, van Egmond M, Vidarsson G. Enhancing activity of FcαRI-bispecific antibodies using glycoengineering. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf027. [PMID: 40156381 DOI: 10.1093/jimmun/vkaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/20/2025] [Indexed: 04/01/2025]
Abstract
Macrophages and natural killer (NK) cells can effectively kill tumor cells in the presence of anti-cancer IgG monoclonal antibodies (mAbs), but neutrophils are less effective. We previously showed that IgG1 bispecific antibodies (BsAb), which target the IgA Fc receptor (FcαRI, CD89) and a tumor associated antigen induce effective neutrophil recruitment and tumor cell killing in vivo. Here we investigated if the efficacy of an anti-EGFR (CetuximAb)/FcαRI-bispecific antibody could be further improved by implementing glycoengineering of the IgG-Fc, aimed at increasing FcγRIIIa/b binding and/or complement activity. Fc afucosylation was introduced to enhance antibody-dependent cellular cytotoxicity (ADCC) by FcγRIIIa on NK/macrophages, which can also reduce neutrophil-mediated ADCC through their GPI-linked FcγRIIIb. Fc galactylation was found to enhance antibody hexamerization and thereby complement dependent cytotoxicity (CDC). Low fucosylated BsAbs moderately increased NK cell-mediated tumor cell killing, but did not affect neutrophil-mediated tumor cell killing nor phagocytosis by macrophages. Glycoengineering of these EGFR-specific BsAb, which normally are devoid of CDC-activity, did not enable their complement activities. In conclusion, glycoengineered FcαRI BsAbs increased ADCC by NK cells but had little effect on neutrophil or macrophage mediated tumor killing.
Collapse
Affiliation(s)
- Céline A N Sewnath
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Cancer Immunology Program, Amsterdam, The Netherlands
| | - Timon Damelang
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Arthur E H Bentlage
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Luuk Ten Kroode
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Cancer Immunology Program, Amsterdam, The Netherlands
| | - Cornelis W Tuk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Cancer Immunology Program, Amsterdam, The Netherlands
| | - Remco Visser
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Manfred Wuhrer
- Department of Proteomics and Metabolomics, Leids Universitair Medisch Centrum (LUMC), LUMC, The Netherlands
| | - Julie Van Coillie
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Cancer Immunology Program, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gestur Vidarsson
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
- Department of Experimental Immunohematology and Landsteiner Laboratory, Sanquin Research, Amsterdam, Netherlands
| |
Collapse
|
7
|
Al-Jarrad A, Alouch SS, Chawla Y, Gonsalves WI. T-Cell Engagers In Multiple Myeloma: A Clinical Review. Blood Lymphat Cancer 2025; 15:1-10. [PMID: 40160334 PMCID: PMC11952057 DOI: 10.2147/blctt.s492116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
T-cell engagers (TCEs) are engineered to bind both the CD3 subunit on T-cells and specific antigens on tumor cells, triggering T-cell activation and tumor cell lysis. TCEs targeting B-cell maturation antigen (BCMA) and G-protein-coupled receptor, class C, group 5, member D (GPRC5D) have demonstrated significant clinical activity in heavily pretreated patients with relapsed/refractory multiple myeloma (RRMM). As a monotherapy, TCEs have had overall response rates (ORRs) of over 60%, with deep and durable hematological responses. Common toxicities include cytokine release syndrome (CRS), infections, and on-target off-tumor effects. Ongoing research looks to enhance the efficacy and tolerability of TCEs for the next generation of products to play an even bigger role in treating patients with MM.
Collapse
Affiliation(s)
| | | | - Yogesh Chawla
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
8
|
Fournier L, Arras P, Pekar L, Kolmar H, Zielonka S, Toleikis L, Becker S. Enhancing NK cell-mediated tumor killing of B7-H6 + cells with bispecific antibodies targeting allosteric sites of NKp30. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200917. [PMID: 39811682 PMCID: PMC11730255 DOI: 10.1016/j.omton.2024.200917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/09/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025]
Abstract
In this work, we report the discovery and engineering of allosteric variable domains of the heavy chain (VHHs) derived from camelid immunization targeting NKp30, an activating receptor on natural killer (NK) cells. The aim was to enhance NK cell-mediated killing capacities by identifying VHHs that do not compete with the natural ligand of NKp30:B7-H6, thereby maximizing the recognition of B7-H6+ tumor cells. By relying on the DuoBody technology, bispecific therapeutic antibodies were engineered, creating a panel of bispecific antibodies against NKp30xEGFR (cetuximab moiety) or NKp30xHER2 (trastuzumab moiety), called natural killer cell engagers (NKCEs). These NKCEs were assessed for their killing capacities on B7-H6-expressing tumor cells. The results demonstrated an enhancement in NK killing capacities for both EGFR-expressing (HeLa) and HER2-expressing (SK-BR-3) cells, indicating the significance of the natural NKp30/B7-H6 axis in tumor recognition by the immune system. Notably, engineering NKCEs to allow natural recognition of B7-H6 was found to be more effective in promoting NKCE-mediated killing of B7-H6+ tumor cells via enhancement of cytokine release. This study highlights the potential of an enhanced-targeting approach, wherein tumor cell surface antigens are targeted while still enabling the natural recognition of the activating ligand (B7-H6) by the immune cells.
Collapse
Affiliation(s)
- Léxane Fournier
- Early Protein Supply and Characterization, Merck Healthcare KGaA, 64293 Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Paul Arras
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Lukas Pekar
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, 64283 Darmstadt, Germany
| | - Stefan Zielonka
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Lars Toleikis
- Early Protein Supply and Characterization, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Stefan Becker
- Early Protein Supply and Characterization, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| |
Collapse
|
9
|
Karbyshev MS, Kalashnikova IV, Dubrovskaya VV, Baskakova KO, Kuzmichev PK, Sandig V. Trends and challenges in bispecific antibody production. J Chromatogr A 2025; 1744:465722. [PMID: 39884073 DOI: 10.1016/j.chroma.2025.465722] [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: 10/31/2024] [Revised: 01/05/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Bispecific antibodies (bsAbs) represent a rapidly growing field of therapeutic agents. More bsAbs are being approved worldwide and are in various stages of clinical trials. However, the discovery and production of novel bsAbs presents significant challenges due to their complex structure. Thus, precise control of assembly and stability is required, given the many formats developed. This review examines recent trends in bsAb production, focusing on advancements in engineering platforms, production strategies, and challenges in large-scale manufacturing. Key developments include improvements in modular antibody design, novel expression systems, and optimization of bioprocessing techniques to enhance stability, yield, and efficacy. Additionally, the article explores the future potential of bsAbs as next-generation therapeutics, underscoring the growing impact of these innovations on expanding treatment options for patients with unmet medical needs.
Collapse
Affiliation(s)
- Mikhail S Karbyshev
- Department of Biotechnology, Moscow Polytechnic University (Moscow Polytech), Moscow, Russia; Department of Biochemistry and Molecular Biology, Pirogov Russian National Research Medical University, Moscow, Russia.
| | | | | | - Kristina O Baskakova
- Department of Biochemistry and Molecular Biology, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | | |
Collapse
|
10
|
Hushmandi K, Einollahi B, Lee EHC, Sakaizawa R, Glaviano A, Reiter RJ, Saadat SH, Farani MR, Huh YS, Aref AR, Salimimoghadam S, Kumar AP. Bispecific antibodies as powerful immunotherapeutic agents for urological cancers: Recent innovations based on preclinical and clinical evidence. Int J Biol Sci 2025; 21:1410-1435. [PMID: 39990653 PMCID: PMC11844292 DOI: 10.7150/ijbs.96155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/25/2024] [Indexed: 02/25/2025] Open
Abstract
Conventional immunotherapy has emerged as a key option for cancer treatment. However, its efficacy has been limited in urological cancers, especially prostate cancer, because of the immunosuppressive tumor microenvironment (TME), difficulty in drug delivery, aberrant immune response, and damage to normal cells. Bispecific antibodies (BsAbs) are engineered proteins with two different antigen-binding domains, designed using different technologies and in various formats. BsAb-based tumor immunotherapy has yielded optimistic results in preclinical and clinical investigations of many tumor types, including urological cancers. However, a series of challenges, including tumor heterogeneity, TME, Ab immunogenicity, adverse effects, serum half-life, low response rates, and drug resistance, hamper the application of BsAbs. In this review, we provide insights into the most common BsAb platforms with different mechanisms of action, which are under preclinical and clinical research, along with ways to overcome the challenges in BsAb administration for treating urological cancer.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reo Sakaizawa
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, Texas USA
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX Inc., Boston, MA, USA
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
Lacy ER, Nanjunda R, Klakamp SL, Kwok D, Nemeth JF, Powers GD, Caicedo HH, Jacobs SA. A novel label-free method to determine equilibrium dissociation constants of antibodies binding to cell surface proteins. Sci Rep 2025; 15:3352. [PMID: 39870691 PMCID: PMC11772844 DOI: 10.1038/s41598-024-82288-9] [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: 06/28/2024] [Accepted: 12/04/2024] [Indexed: 01/29/2025] Open
Abstract
Solution-based affinity assays are used for the selection and characterization of proteins that could be developed into therapeutic molecules. However, these assays have limitations for cell-surface proteins as in most cases their purification requires detergent solubilization and are unlikely to assume conformations in solution that resemble their native states in cell membranes. This report describes a novel electrochemiluminescence-based method, called MSD-CAT, for the affinity analysis of antibodies binding to cell-surface receptors. MSD-CAT was used to evaluate the binding of monoclonal antibodies, Fab fragments, and bispecific antibodies targeting the cell-surface receptor interleukin 3 receptor alpha (CD123) and the results were compared to data obtained using surface plasmon resonance (SPR). The data showed that MSD-CAT can be successfully applied to determine binding affinity on cells in a label free format and without the need for laborious solubilization procedures to generate recombinant antigen. In addition, this method has the potential for high-throughput application while enabling simultaneous determination of equilibrium dissociation constant (KD) and receptor density within the same experiment.
Collapse
Affiliation(s)
- Eilyn R Lacy
- Johnson & Johnson, Therapeutics Discovery, Spring House, PA, USA.
| | - Rupesh Nanjunda
- Johnson & Johnson, Therapeutics Discovery, Spring House, PA, USA
- Johnson & Johnson, Therapeutics Development and Supply, Malvern, PA, USA
| | - Scott L Klakamp
- Johnson & Johnson, Therapeutics Discovery, Spring House, PA, USA
- Coherus BioSciences, Camarillo, CA, USA
| | - Deborah Kwok
- Johnson & Johnson, Therapeutics Discovery, Spring House, PA, USA
| | | | - Gordon D Powers
- Johnson & Johnson, Therapeutics Discovery, Spring House, PA, USA
| | - H Hugo Caicedo
- Johnson & Johnson, Therapeutics Discovery, Spring House, PA, USA
- Eli Lilly and Company, New York, NY, USA
| | - Steven A Jacobs
- Johnson & Johnson, Therapeutics Discovery, Spring House, PA, USA
| |
Collapse
|
12
|
Liu P, Gu C, Cao X, Zhang H, Wang Z, Yang Y, OuYang K, Zhen Y, Jia F, He X, Yu H, Deng S. Discovery of a common light chain bispecific antibody targeting PD-1 and PD-L1 by Hybridoma-to-Phage-to-Yeast (H2PtY) platform. Antib Ther 2025; 8:1-12. [PMID: 39839911 PMCID: PMC11744305 DOI: 10.1093/abt/tbae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 01/23/2025] Open
Abstract
Background Therapeutic antibody drugs targeting the PD-1 pathway are generally characterized by relatively low response rates and susceptibility to drug resistance during clinical application. Therefore, there is an urgent need for alternative therapeutic strategies to increase the immune response rate. Bispecific antibodies co-targeting PD-1 and PD-L1 may have greater potential to improve the efficacy of the immune checkpoint pathway. Method In this study, we developed a potent humanized common light chain (CLC) IgG shape bispecific antibody (bsAb), named JMB2005, based on Hybridoma-to-Phage-to-Yeast platform. The platform allowed us to discover CLC bsAb from traditional mice for any pair of given targets. Results JMB2005 exhibited favorable developability, good manufacturing property, and satisfactory efficacy, which could be given via subcutaneous injection at the concentration of 120 mg/mL. Mechanistically, JMB2005 could bridge tumor cells and T cells with both Fab arms and promote T-cells to function as direct tumor cell killers. It could also promote T cell activation by blocking the binding of PD-L1 to CD80. Furthermore, JMB2005 has exhibited a favorable half-life and has demonstrated promising anti-tumor therapeutic efficacy in vivo. Conclusion Consequently, the present study showed that the novel humanized CLC bsAb JMB2005 may represent a novel therapeutic agent of great clinical potential.
Collapse
Affiliation(s)
- Peipei Liu
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Chunyin Gu
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Xiaodan Cao
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Huawei Zhang
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Zongda Wang
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Yukun Yang
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - KeDong OuYang
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Yingying Zhen
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Fangfang Jia
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Xianqing He
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Haixiang Yu
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| | - Sujun Deng
- Biologics Innovation Institute, Shanghai Jemincare Pharmaceutical Co., Ltd., Lane 535, Huanqiao Road, Pudong New Area, Shanghai 201315, China
| |
Collapse
|
13
|
Mortaheb S, Pezeshki PS, Rezaei N. Bispecific therapeutics: a state-of-the-art review on the combination of immune checkpoint inhibition with costimulatory and non-checkpoint targeted therapy. Expert Opin Biol Ther 2024; 24:1335-1351. [PMID: 39503381 DOI: 10.1080/14712598.2024.2426636] [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: 07/18/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have revolutionized the field of cancer immunotherapy and have enhanced the survival of patients with malignant tumors. However, the overall efficacy of ICIs remains unsatisfactory and is faced with two major concerns of resistance development and occurrence of immune-related adverse events (irAEs). Bispecific antibodies (bsAbs) have emerged as promising strategies with unique mechanisms of action to achieve a better efficacy and safety than monoclonal antibodies (mAbs) or even their combination. BsAbs along with other bispecific platforms such as bispecific fusion proteins, nanobodies, and CAR-T cells may help to avoid development of resistance and reduce irAEs caused by on-target/off-tumor binding effects of mAbs. AREAS COVERED A literature search was performed using PubMed for English-language articles to provide a comprehensive overview of preclinical and clinical studies on bsAbs specified for both immune checkpoints and non-checkpoint molecules as a well-enhanced class of therapeutics. EXPERT OPINION Identifying suitable targets and selecting effective engineering platforms enhance the potential of bsAbs to address the challenges associated with conventional therapies such as ICIs, positioning them as a promising class of therapeutics in the landscape of cancer immunotherapy.
Collapse
Affiliation(s)
- Samin Mortaheb
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parmida Sadat Pezeshki
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Feng Y, Deyanat-Yazdi G, Newburn K, Potter S, Wortinger M, Ramirez M, Truhlar SME, Yachi PP. PD-1 antibody interactions with Fc gamma receptors enable PD-1 agonism to inhibit T cell activation - therapeutic implications for autoimmunity. J Autoimmun 2024; 149:103339. [PMID: 39608214 DOI: 10.1016/j.jaut.2024.103339] [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: 11/21/2023] [Revised: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 11/30/2024]
Abstract
PD-1 has emerged as a central inhibitory checkpoint receptor in maintaining immune homeostasis and as a target in cancer immunotherapies. However, targeting PD-1 for the treatment of autoimmune diseases has been more challenging. We recently showed in a phase 2a trial that PD-1 could be stimulated with the PD-1 agonist antibody peresolimab to treat rheumatoid arthritis. Here, we demonstrate that PD-1 antibodies can elicit agonism and inhibit T cell activation by co-localization of PD-1 with the T cell receptor via Fcγ receptor (FcγR) engagement. Three PD-1 agonist antibodies with different antigen binding domains, including the clinically validated PD-1 blocking antibody pembrolizumab, suppressed T cell activation to a similar degree; this finding suggests that a specific PD-1-binding epitope is not required for PD-1 agonism. We next explored whether antibody-mediated clustering was an important driver of inhibition of T cell activation; however, we found that a monovalent PD-1 antibody was not inferior to a conventional bivalent antibody in its ability to suppress T cell activation. Importantly, we found that affinity to PD-1 correlated positively with inhibition of T cell activation, with higher affinity antibodies exhibiting higher levels of inhibition. Using a series of human Fc mutants with altered affinities to various FcγRs, we dissected the contributions of FcγRs and found that multiple FcγRs rather than a single receptor contribute to agonist activity. Our work reveals an important role for FcγR binding in the activity of PD-1 antibodies, which has implications for optimizing both PD-1 agonist and antagonist antibodies.
Collapse
MESH Headings
- Humans
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Receptors, IgG/metabolism
- Receptors, IgG/immunology
- Lymphocyte Activation/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/drug effects
- Autoimmunity
- Protein Binding
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Autoimmune Diseases/immunology
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/therapy
Collapse
Affiliation(s)
- Yiqing Feng
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN, USA
| | - Gordafaried Deyanat-Yazdi
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA
| | - Kristin Newburn
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN, USA
| | - Scott Potter
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA
| | - Mark Wortinger
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, USA
| | - Miriam Ramirez
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA
| | - Stephanie M E Truhlar
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA
| | - Pia P Yachi
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, CA, USA.
| |
Collapse
|
15
|
Doran D, Struble G, Moravcevic K, Wang P, Ji S. Development and validation of a robust RP-HPLC method to quantitate residual 2-mercaptoethylamine in drug product formulations containing amino acid additives. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1249:124379. [PMID: 39561467 DOI: 10.1016/j.jchromb.2024.124379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/23/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
Bispecific antibodies have a wide range of applications in cancer immunotherapy, some of which are manufactured by controlled Fab-arm exchange requiring the reductant 2-mercaptoethylamine (2-MEA). As a process impurity, monitoring the residual 2-MEA in bispecific antibody drug product process development is needed. A novel reversed phase-high performance liquid chromatography (RP-HPLC) method for measurement of residual 2-MEA that uses 7-fluorobenzofurazan-4-sulfonic acid ammonium salt (SBD-F) as a fluorescent-detection tag in drug product formulations containing high concentrations of arginine has been developed. Using a thiol tag for residual 2-MEA eliminates any potential interference from conventional tag binding to amine groups of the formulation arginine, and potentially resulting in overestimation of the amount of impurity in a given sample. The new method has been fully validated for specificity, linearity, accuracy, range, limit of quantitation, limit of detection, and robustness. This method therefore has potential to aid in detecting residual 2-MEA content for any process that utilizes 2-MEA for bispecific antibody manufacturing.
Collapse
Affiliation(s)
- Delaney Doran
- Therapeutics Development and Supply, Analytical Development - Janssen Research & Development, LLC, Malvern, PA, USA.
| | - Geoffrey Struble
- Therapeutics Development and Supply, Analytical Development - Janssen Research & Development, LLC, Malvern, PA, USA
| | - Katarina Moravcevic
- Therapeutics Development and Supply, Analytical Development - Janssen Research & Development, LLC, Malvern, PA, USA
| | - Pinger Wang
- Therapeutics Development and Supply, Analytical Development - Janssen Research & Development, LLC, Malvern, PA, USA
| | - Shelly Ji
- Therapeutics Development and Supply, Analytical Development - Janssen Research & Development, LLC, Malvern, PA, USA
| |
Collapse
|
16
|
van der Horst HJ, Mutis T. Enhancing Fc-mediated effector functions of monoclonal antibodies: The example of HexaBodies. Immunol Rev 2024; 328:456-465. [PMID: 39275983 PMCID: PMC11659923 DOI: 10.1111/imr.13394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
Since the approval of the CD20-targeting monoclonal antibody (mAb) rituximab for the treatment of lymphoma in 1997, mAb therapy has significantly transformed cancer treatment. With over 90 FDA-approved mAbs for the treatment of various hematological and solid cancers, modern cancer treatment relies heavily on these therapies. The overwhelming success of mAbs as cancer therapeutics is attributed to their broad applicability, high safety profile, and precise targeting of cancer-associated surface antigens. Furthermore, mAbs can induce various anti-tumor cytotoxic effector mechanisms including antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC), all of which are mediated via their fragment crystallizable (Fc) domain. Over the past decades, these effector mechanisms have been substantially improved through Fc domain engineering. In this review, we will outline the different approaches to enhance Fc effector functions via Fc engineering of mAbs, with a specific emphasis on the so-called "HexaBody" technology, which is designed to enhance the hexamerization of mAbs on the target cell surface, thereby inducing greater complement activation, CDC, and receptor clustering. The review will summarize the development, preclinical, and clinical testing of several HexaBodies designed for the treatment of B-cell malignancies, as well as the potential use of the HexaBody technology beyond Fc-mediated effector functions.
Collapse
Affiliation(s)
- Hilma J. van der Horst
- Department of HematologyCancer Center Amsterdam, Amsterdam UMC, VU Medical CenterAmsterdamThe Netherlands
- Present address:
Department of Fundamental Oncology, Ludwig Institute for Cancer ResearchUniversity of LausanneEpalingesSwitzerland
| | - Tuna Mutis
- Department of HematologyCancer Center Amsterdam, Amsterdam UMC, VU Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
17
|
Whitehead CA, Wines BD, Davies AM, McDonnell JM, Trist HM, Esparon SE, Hogarth PM. Stellabody: A novel hexamer-promoting mutation for improved IgG potency. Immunol Rev 2024; 328:438-455. [PMID: 39364646 PMCID: PMC11659935 DOI: 10.1111/imr.13400] [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] [Indexed: 10/05/2024]
Abstract
Advances in antibody engineering are being directed at the development of next generation immunotherapeutics with improved potency. Hexamerisation of IgG is a normal physiological aspect of IgG biology and recently described mutations that facilitate this process have a substantial impact upon monoclonal antibody behavior resulting in the elicitation of dramatically enhanced complement-dependent cytotoxicity, Fc receptor function, and enhanced antigen binding effects, such as targeted receptor agonism or microbe neutralization. Whereas the discovery of IgG hexamerisation enhancing mutations has largely focused on residues with exposure at the surface of the Fc-Fc and CH2-CH3 interfaces, our unique approach is the engineering of the mostly buried residue H429 in the CH3 domain. Selective substitution at position 429 forms the basis of Stellabody technology, where the choice of amino acid results in distinct hexamerisation outcomes. H429F results in monomeric IgG that hexamerises after target binding, so called "on-target" hexamerisation, while the H429Y mutant forms pH-sensitive hexamers in-solution prior to antigen binding. Moreover, Stellabody technologies are broadly applicable across the family of antibody-based biologic therapeutics, including conventional mAbs, bispecific mAbs, and Ig-like biologics such as Fc-fusions, with applications in diverse diseases.
Collapse
Affiliation(s)
- Clarissa A. Whitehead
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Bruce D. Wines
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Anna M. Davies
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's HouseLondonUK
| | - James M. McDonnell
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's HouseLondonUK
| | - Halina M. Trist
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
| | | | - P. Mark Hogarth
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
- Department of Clinical PathologyThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
18
|
Messaoudi S, Wai K, Marple A, Baniahmad SF, Wylie RG, Pelletier M, Craig M, Durocher Y, Greschner AA, Gauthier MA. Rapid Systematic Screening of Bispecific Antibody Surrogate Geometries for T-Cell Engagement Using DNA Nanotechnology. J Am Chem Soc 2024; 146:29824-29835. [PMID: 39412838 PMCID: PMC11529601 DOI: 10.1021/jacs.4c11648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/01/2024]
Abstract
Bispecific antibodies (bsAbs) are emerging immune-therapeutics, and many formats exist that differ considerably in structure. However, little systematic data exist about how the spatial organization of their components influences activity, requiring innovative approaches combining empirical and quantitative frameworks. This study presents a modular DNA nanotechnology platform to generate numerous bsAbs with surrogate geometries that span the structural features of the BiTE, IgG-like, and IgG-conjugate platforms to screen for T-cell engagement. Results highlight interesting structure-activity relationships regarding bsAb potency and selectivity and raise questions regarding the molecular phenomena underlying activity. To elucidate some effects, the platform was paired with a simple mathematical model. This work is thus one of the first to systematically investigate and reveal the importance of the spatial organization of bsAb components on activity and equally provides an accessible and convenient tool for rapidly mapping out such trends for other combinations of target epitopes.
Collapse
Affiliation(s)
- Sabrine Messaoudi
- Institut
National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| | - Kevin Wai
- Department
of Chemistry and Chemical Biology, McMaster
University, Hamilton, Ontario L8S 4M1, Canada
| | - April Marple
- Department
of Chemistry and Chemical Biology, McMaster
University, Hamilton, Ontario L8S 4M1, Canada
| | - Seyed Farzad Baniahmad
- Human
Health Therapeutics Research Centre, National
Research Council Canada, Montreal, Quebec H4P 2R2, Canada
| | - Ryan G. Wylie
- Department
of Chemistry and Chemical Biology, McMaster
University, Hamilton, Ontario L8S 4M1, Canada
| | - Marianne Pelletier
- Sainte-Justine
University Hospital Azrieli Research Centre, Montreal, Quebec H3T 1C5, Canada
- Département
de Mathématiques et de Statistique, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Morgan Craig
- Sainte-Justine
University Hospital Azrieli Research Centre, Montreal, Quebec H3T 1C5, Canada
- Département
de Mathématiques et de Statistique, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Yves Durocher
- Human
Health Therapeutics Research Centre, National
Research Council Canada, Montreal, Quebec H4P 2R2, Canada
| | - Andrea A. Greschner
- Institut
National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| | - Marc A. Gauthier
- Institut
National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| |
Collapse
|
19
|
Tornetta MA, Whitaker BP, Cantwell OM, Pisors ED, Han L, MacWilliams MP, Jiang H, Zhou F, Chiu ML. The process using a synthetic library that generates multiple diverse human single domain antibodies. Antib Ther 2024; 7:283-294. [PMID: 39381136 PMCID: PMC11456836 DOI: 10.1093/abt/tbae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 10/10/2024] Open
Abstract
Background Single domain antibodies (sdAbs) possess unique characteristics that make them highly effective for developing complex therapeutics. Methods Our process uses a fully synthetic phage display library to generate single domain antibodies that can bind to disease relevant antigen conformations. A human IGHV3 family scaffold makes up the phage display libraries, and these VHO libraries are applied to diverse phage biopannings against target antigens. After NGS processing, unique VHOs undergo automated cloning into expression constructs followed by transfections and purifications. Binding assays were used to determine VHO binding behaviors to the target proteins. Additional VHO interactions are measured against endogenous targets on cells by way of flow cytometry, cell internalization, and activation assays. Results We show that a fully synthetic phage display library can generate VHOs that bind to disease relevant antigen conformations. The diverse biopanning methods and processing of next-generation sequencing generated many VHO paratopes. These different VHO sequences can be expressed as Fc fusion proteins. Various screening assays resulted in VHOs representing different epitopes or activities. During the hit evaluation, we demonstrate how screening can identify distinct VHO activities that have been used to generate differentiated drug molecules in various bispecific and multispecific antibody formats. Conclusion We demonstrate how screening can identify distinct VHO activities that have been used to generate differentiated drug molecules in various bispecific and multispecific antibody formats.
Collapse
Affiliation(s)
- Mark A Tornetta
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Brian P Whitaker
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Olivia M Cantwell
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Eileen D Pisors
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Lu Han
- Biologics Discovery Department, Tavotek Biotherapeutics, Building C2, Suzhou Biomedical Industrial Park, Suzhou, Jiang Su 215000, China
| | - Maria P MacWilliams
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Hao Jiang
- Biologics Discovery Department, Tavotek Biotherapeutics, Building C2, Suzhou Biomedical Industrial Park, Suzhou, Jiang Su 215000, China
| | - Fulai Zhou
- Biologics Discovery Department, Tavotek Biotherapeutics, Building C2, Suzhou Biomedical Industrial Park, Suzhou, Jiang Su 215000, China
| | - Mark L Chiu
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
- Biologics Discovery Department, Tavotek Biotherapeutics, Building C2, Suzhou Biomedical Industrial Park, Suzhou, Jiang Su 215000, China
| |
Collapse
|
20
|
Xie Z, Protzer U. Activating adaptive immunity by bispecific, T-cell engager antibodies bridging infected and immune-effector cells is a promising novel therapy for chronic hepatitis B. Antiviral Res 2024; 229:105972. [PMID: 39084340 DOI: 10.1016/j.antiviral.2024.105972] [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: 01/30/2024] [Revised: 07/14/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Bispecific antibodies (bsAbs) are engineered immunoglobulins that combine two different antigen-binding sites in one molecule. BsAbs can be divided into two molecular formats: IgG-like and non-IgG-like antibodies. Structural elements of each format have implications for engaging the immune system. T cell engager antibodies (TCEs) are bsAbs designed to engage T cells with target cells. TCEs can be applied not only in cancer but also in infectious disease therapy to activate T-cell responses. In this review, we focus on current literature on the design and use of bsAbs as an innovative strategy to enhance adaptive antiviral immune responses. We summarized the novel T cell-related immunotherapies with a focus on TCEs, that are developed for the treatment of chronic hepatitis B. Chronic infection with the hepatitis B virus (HBV) had a death toll of 1.1 million humans in 2022, mainly due to liver cirrhosis and hepatocellular carcinoma developing in the more than 250 million humans chronically infected. A curative treatment approach for chronic hepatitis B is lacking. Combining antiviral therapy with immune therapies activating T-cell responses is regarded as the most promising therapeutic approach to curing HBV and preventing the sequelae of chronic infection. Attracting functionally intact T cells that are not HBV-specific and, therefore, have not yet been exposed to regulatory mechanisms and activating those at the target site in the liver is a very interesting therapeutic approach that could be achieved by TCEs. Thus, TCEs redirecting T cells toward HBV-positive cells represent a promising strategy for treating chronic hepatitis B and HBV-associated hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhe Xie
- Institute of Virology, School of Medicine and Health, Technical University of Munich / Helmholtz Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, School of Medicine and Health, Technical University of Munich / Helmholtz Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Sites, Germany.
| |
Collapse
|
21
|
Liao CY, Engelberts P, Ioan-Facsinay A, Klip JE, Schmidt T, Ruijtenbeek R, Danen EHJ. CD3-engaging bispecific antibodies trigger a paracrine regulated wave of T-cell recruitment for effective tumor killing. Commun Biol 2024; 7:983. [PMID: 39138287 PMCID: PMC11322607 DOI: 10.1038/s42003-024-06682-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
The mechanism of action of bispecific antibodies (bsAbs) directing T-cell immunity to solid tumors is incompletely understood. Here, we screened a series of CD3xHER2 bsAbs using extracellular matrix (ECM) embedded breast cancer tumoroid arrays exposed to healthy donor-derived T-cells. An initial phase of random T-cell movement throughout the ECM (day 1-2), was followed by a bsAb-dependent phase of active T-cell recruitment to tumoroids (day 2-4), and tumoroid killing (day 4-6). Low affinity HER2 or CD3 arms were compensated for by increasing bsAb concentrations. Instead, a bsAb binding a membrane proximal HER2 epitope supported tumor killing whereas a bsAb binding a membrane distal epitope did not, despite similar affinities and intra-tumoroid localization of the bsAbs, and efficacy in 2D co-cultures. Initial T-cell-tumor contact through effective bsAbs triggered a wave of subsequent T-cell recruitment. This critical surge of T-cell recruitment was explained by paracrine signaling and preceded a full-scale T-cell tumor attack.
Collapse
Affiliation(s)
- Chen-Yi Liao
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | | | | | - Janna Eleonora Klip
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Thomas Schmidt
- Leiden Institute of Physics, Leiden University, Leiden, the Netherlands
| | | | - Erik H J Danen
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
22
|
Ramos KE, Okba NMA, Tan J, Bandawane P, Meade PS, Loganathan M, Francis B, Shulenin S, Holtsberg FW, Aman MJ, McMahon M, Krammer F, Lai JR. Broadly protective bispecific antibodies that simultaneously target influenza virus hemagglutinin and neuraminidase. mBio 2024; 15:e0108524. [PMID: 38899870 PMCID: PMC11253627 DOI: 10.1128/mbio.01085-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 06/21/2024] Open
Abstract
Monoclonal antibodies (mAbs) are an attractive therapeutic platform for the prevention and treatment of influenza virus infection. There are two major glycoproteins on the influenza virion surface: hemagglutinin (HA), which is responsible for viral attachment and entry, and neuraminidase (NA), which mediates viral egress by enzymatically cleaving sialic acid to release budding particles from the host cell surface. Broadly neutralizing antibodies (bNAbs) that target the conserved HA central stalk region, such as CR9114, can inhibit both viral entry and egress. More recently, broadly binding mAbs that engage and inhibit the NA active site, such as 1G01, have been described to prevent viral egress. Here, we engineered bispecific antibodies (bsAbs) that combine the variable domains of CR9114 and 1G01 into a single molecule and evaluated if simultaneous targeting of two different glycoproteins improved antiviral properties in vitro and in vivo. Several CR9114/1G01 bsAbs were generated with various configurations of the two sets of the variable domains ("bsAb formats"). We found that combinations employing the addition of a single-chain variable fragment in the hinge region of an IgG scaffold had the best properties in terms of expression, stability, and binding. Further characterization of selected bsAbs showed potent neutralizing and egress-inhibiting activity. One such bsAb ("hSC_CR9114_1G01") provided higher levels of prophylactic protection from mortality and morbidity upon challenge with H1N1 than either of the parental mAbs at low dosing (1 mg/kg). These results highlight the potential use of bsAbs that simultaneously target HA and NA as new influenza immunotherapeutics. IMPORTANCE Infection by the influenza virus remains a global health burden. The approaches utilized here to augment the activity of broadly protective influenza virus antibodies may lead to a new class of immunotherapies with enhanced activity.
Collapse
MESH Headings
- Neuraminidase/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Viral/immunology
- Animals
- Humans
- Mice
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Monoclonal/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice, Inbred BALB C
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/drug effects
Collapse
Affiliation(s)
- Kevin E. Ramos
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Nisreen M. A. Okba
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jessica Tan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Pooja Bandawane
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Philip S. Meade
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Madhumathi Loganathan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Benjamin Francis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - M. Javad Aman
- Integrated BioTherapeutics, Inc., Rockville, Maryland, USA
| | - Meagan McMahon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Jonathan R. Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
23
|
Chernyi N, Gavrilova D, Saruhanyan M, Oloruntimehin ES, Karabelsky A, Bezsonov E, Malogolovkin A. Recent Advances in Gene Therapy for Hemophilia: Projecting the Perspectives. Biomolecules 2024; 14:854. [PMID: 39062568 PMCID: PMC11274510 DOI: 10.3390/biom14070854] [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: 05/27/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
One of the well-known X-linked genetic disorders is hemophilia, which could be hemophilia A as a result of a mutation in the F8 (factor VIII) gene or hemophilia B as a result of a mutation in the F9 (factor IX) gene, leading to insufficient levels of the proteins essential for blood coagulation cascade. In patients with severe hemophilia, factor VIII or factor IX activities in the blood plasma are considerably low, estimated to be less than 1%. This is responsible for spontaneous or post-traumatic bleeding episodes, or both, leading to disease complications and death. Current treatment of hemophilia relies on the prevention of bleeding, which consists of expensive lifelong replacement infusion therapy of blood plasma clotting factors, their recombinant versions, or therapy with recombinant monoclonal antibodies. Recently emerged gene therapy approaches may be a potential game changer that could reshape the therapeutic outcomes of hemophilia A or B using a one-off vector in vivo delivery and aim to achieve long-term endogenous expression of factor VIII or IX. This review examines both traditional approaches to the treatment of hemophilia and modern methods, primarily focusing on gene therapy, to update knowledge in this area. Recent technological advances and gene therapeutics in the pipeline are critically reviewed and summarized. We consider gene therapy to be the most promising method as it may overcome the problems associated with more traditional treatments, such as the need for constant and expensive infusions and the presence of an immune response to the antibody drugs used to treat hemophilia.
Collapse
Affiliation(s)
- Nikita Chernyi
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia; (N.C.); (M.S.); (E.S.O.)
| | - Darina Gavrilova
- Department of Biology and General Genetics, First Moscow State Medical University (Sechenov University), Moscow 105043, Russia;
| | - Mane Saruhanyan
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia; (N.C.); (M.S.); (E.S.O.)
| | - Ezekiel S. Oloruntimehin
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia; (N.C.); (M.S.); (E.S.O.)
| | - Alexander Karabelsky
- Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354530, Russia;
| | - Evgeny Bezsonov
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia; (N.C.); (M.S.); (E.S.O.)
- Department of Biology and General Genetics, First Moscow State Medical University (Sechenov University), Moscow 105043, Russia;
| | - Alexander Malogolovkin
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia; (N.C.); (M.S.); (E.S.O.)
- Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354530, Russia;
| |
Collapse
|
24
|
Goebeler ME, Stuhler G, Bargou R. Bispecific and multispecific antibodies in oncology: opportunities and challenges. Nat Rev Clin Oncol 2024; 21:539-560. [PMID: 38822215 DOI: 10.1038/s41571-024-00905-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/02/2024]
Abstract
Research into bispecific antibodies, which are designed to simultaneously bind two antigens or epitopes, has advanced enormously over the past two decades. Owing to advances in protein engineering technologies and considerable preclinical research efforts, bispecific antibodies are constantly being developed and optimized to improve their efficacy and to mitigate toxicity. To date, >200 of these agents, the majority of which are bispecific immune cell engagers, are in either preclinical or clinical evaluation. In this Review, we discuss the role of bispecific antibodies in patients with cancer, including history and development, as well as innovative targeting strategies, clinical applications, and adverse events. We also discuss novel alternative bispecific antibody constructs, such as those targeting two antigens expressed by tumour cells or cells located in the tumour microenvironment. Finally, we consider future research directions in this rapidly evolving field, including innovative antibody engineering strategies, which might enable more effective delivery, overcome resistance, and thus optimize clinical outcomes.
Collapse
Affiliation(s)
- Maria-Elisabeth Goebeler
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany.
- National Center for Tumour Diseases, NCT WERA, University Hospital Würzburg, Würzburg, Germany.
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
| | - Gernot Stuhler
- National Center for Tumour Diseases, NCT WERA, University Hospital Würzburg, Würzburg, Germany
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Ralf Bargou
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
- National Center for Tumour Diseases, NCT WERA, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
25
|
Cech P, Skórka K, Dziki L, Giannopoulos K. T-Cell Engagers-The Structure and Functional Principle and Application in Hematological Malignancies. Cancers (Basel) 2024; 16:1580. [PMID: 38672662 PMCID: PMC11048836 DOI: 10.3390/cancers16081580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Recent advancements in cancer immunotherapy have made directing the cellular immune response onto cancer cells a promising strategy for the treatment of hematological malignancies. The introduction of monoclonal antibody-based (mAbs) targeted therapy has significantly improved the prognosis for hematological patients. Facing the issues of mAb-based therapies, a novel bispecific antibody (BsAb) format was developed. T-cell engagers (TCEs) are BsAbs, which simultaneously target tumor-associated antigens on tumor cells and CD3 molecules present on T-cells. This mechanism allows for the direct activation of T-cells and their anti-tumor features, ultimately resulting in the lysis of tumor cells. In 2014, the FDA approved blinatumomab, a TCE directed to CD3 and CD19 for treatment of acute lymphoblastic leukemia. Since then, numerous TCEs have been developed, allowing for treating different hematological malignancies such as acute myeloid leukemia, multiple myeloma, and non-Hodgkin lymphoma and Hodgkin lymphoma. As of November 2023, seven clinically approved TCE therapies are on the market. TCE-based therapies still have their limitations; however, improving the properties of TCEs, as well as combining TCE-based therapies with other forms of treatment, give hope to find the cures for currently terminal diseases. In this paper, we summarized the technical basis of the TCE technology, its application in hematology, and its current issues and prospects.
Collapse
Affiliation(s)
| | - Katarzyna Skórka
- Department of Experimental Hematooncology, Medical University of Lublin, 20-093 Lublin, Poland; (P.C.); (L.D.); (K.G.)
| | | | | |
Collapse
|
26
|
Zhou F, Ben Y, Jiang H, Tan S, Mu G, Zha Z, Dong S, Huang S, Zhou Y, Jin Y, Chiu ML. A Novel Dual-Fc Bispecific Antibody with Enhanced Fc Effector Function. Biochemistry 2024; 63:958-968. [PMID: 38426700 PMCID: PMC11025548 DOI: 10.1021/acs.biochem.3c00481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
Bispecific antibodies (BsAbs) are undergoing continued development for applications in oncology and autoimmune diseases. While increasing activity by having more than one targeting arm, most BsAb engineering employs single Fc engagement as monoclonal antibodies. Here, we designed a novel immunoglobulin gamma-1 (IgG1)-derived dual-Fc BsAb containing two Fc regions and two distinct asymmetric antigen binding arms comprising a Fab arm and another VHH domain. In conjunction with the knob-into-hole technology, dual-Fc BsAbs could be produced with a high yield and good stability. We explore how Fc engineering effects on dual-Fc constructs could boost the desired therapeutic efficacy. This new format enabled simultaneous bispecific binding to corresponding antigens. Furthermore, compared to the one-Fc control molecules, dual-Fc BsAbs were shown to increase the avidity-based binding to FcγRs to result in higher ADCC and ADCP activities by potent avidity via binding to two antigens and Fc receptors. Overall, this novel BsAb format with enhanced effector functionalities provides a new option for antibody-based immunotherapy.
Collapse
Affiliation(s)
- Fulai Zhou
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Yinyin Ben
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Hao Jiang
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Siwen Tan
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Guangmao Mu
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Zhengxia Zha
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Shuting Dong
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Sheng Huang
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Yijun Zhou
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Ying Jin
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
| | - Mark L. Chiu
- Research
& Development Department, Tavotek Biotherapeutics, Suzhou 215000, China
- Research
& Development, Tavotek Biotherapeutics, Spring House, Pennsylvania 19102, United States
| |
Collapse
|
27
|
Poskute R, Sankaran PK, Sewell L, Lepore G, Shrubsall R, Dewis L, Watanabe Y, Wong V, Pascual Fernandez L, Mishra R, Holt A, Sou S, Harris C, Moreno Rodriguez C, Cankorur-Cetinkaya A, Smith J, Lonska N, Powell A, Cui T, Cheeks M, Lindo V. Identification and quantification of chain-pairing variants or mispaired species of asymmetric monovalent bispecific IgG1 monoclonal antibody format using reverse-phase polyphenyl chromatography coupled electrospray ionization mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1237:124085. [PMID: 38513430 DOI: 10.1016/j.jchromb.2024.124085] [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: 01/10/2024] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 03/23/2024]
Abstract
Developing a knob-into-hole asymmetric bispecific IgG1 monoclonal antibody (mAb) poses manufacturing challenges due to the expression of chain pairing variants, also called mispaired species, in the desired product. The incorrect pairing of light and heavy chains could result in heterogeneous mispaired species of homodimers, heterodimers, light chain swapping, and low molecular weight species (LMWS). Standard chromatography, capillary electrophoretic, or spectroscopic methods poorly resolve these from the main variants. Here, we report a highly sensitive reverse-phase polyphenyl ultra-high-performance liquid chromatography (RP-UHPLC) method to accurately measure mispaired species of Duet mAb format, an asymmetric IgG1 bispecific mAb, for both process development and quality control analytical tests. Coupled with electrospray ionization mass spectrometry (ESI-MS), it enabled direct online characterization of mispaired species. This single direct assay detected diverse mispaired IgG-like species and LMWS. The method resolved eight disulfide bonds dissociated LMWS and three mispaired LMWS. It also resolved three different types of IgG-like mispaired species, including two homodimers and one heterodimer. The characterization and quantification simultaneously enabled the cell line selection that produces a lesser heterogeneity and lower levels of mispaired species with the desired correctly paired product. The biological activity assessment of samples with increased levels of these species quantified by the method exhibited a linear decline in potency with increasing levels of mispaired species in the desired product. We also demonstrated the utility of the technique for testing in-process intermediate materials to determine and assess downstream purification process capability in removing diverse mispaired IgG-like species and LMWS to a certain level during the downstream purification process. Our investigation demonstrates that adopting this method was vital in developing asymmetric bispecific mAb from the initial stage of cell line development to manufacturing process development. Therefore, this tool could be used in the control strategy to monitor and control mispaired species during manufacturing, thus improving the quality control of the final product.
Collapse
Affiliation(s)
- Ryte Poskute
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Laura Sewell
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Giordana Lepore
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Rebecca Shrubsall
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Lydia Dewis
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Yasunori Watanabe
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Vanessa Wong
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Rahul Mishra
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Alexander Holt
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Susie Sou
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Claire Harris
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Cristina Moreno Rodriguez
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Ayca Cankorur-Cetinkaya
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Jennifer Smith
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Nikola Lonska
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Adam Powell
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Tingting Cui
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Matthew Cheeks
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Viv Lindo
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
28
|
Sandeep, Shinde SH, Ahmed S, Sharma SS, Pande AH. Engineered polyspecific antibodies: A new frontier in the field of immunotherapeutics. Immunology 2024; 171:464-496. [PMID: 38140855 DOI: 10.1111/imm.13743] [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: 06/02/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
The 21st-century beginning remarked with the huge success of monospecific MAbs, however, in the last couple of years, polyspecific MAbs (PsAbs) have been an interesting topic and show promise of being biobetter than monospecific MAbs. Polyspecificity, in which a single antibody serves multiple specific target binding, has been hypothesized to contribute to the development of a highly effective antibody repertoire for immune defence. This polyspecific MAb trend represents an explosion that is gripping the whole pharmaceutical industry. This review is concerned with the current development and quality enforcement of PsAbs. All provided literature on monospecific MAbs and polyspecific MAbs (PsAbs) were searched using various electronic databases such as PubMed, Google Scholar, Web of Science, Elsevier, Springer, ACS, Google Patent and books via the keywords Antibody engineering, Polyspecific antibody, Conventional antibody, non-conventional antibody, and Single domain antibody. In the literature, there are more than 100 different formats to construct PsAb by quadroma technology, chemical conjugation and genetic engineering. Till March 2023, nine PsAb have been approved around the world, and around 330 are in advanced developmental stages, showing the dominancy of PsAb in the growing health sector. Recent advancements in protein engineering techniques and the fusion of non-conventional antibodies have made it possible to create complex PsAbs that demonstrate higher stability and enhanced potency. This marks the most significant achievement for cancer immunotherapy, in which PsAbs have immense promise. It is worth mentioning that seven out of the nine PsAbs have been approved as anti-cancer therapy. As PsAbs continue to acquire prominence, they could pave the way for the development of novel immunotherapies for multiple diseases.
Collapse
Affiliation(s)
- Sandeep
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Suraj H Shinde
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Sakeel Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| |
Collapse
|
29
|
Wirchnianski AS, Nyakatura EK, Herbert AS, Kuehne AI, Abbasi SA, Florez C, Storm N, McKay LGA, Dailey L, Kuang E, Abelson DM, Wec AZ, Chakraborti S, Holtsberg FW, Shulenin S, Bornholdt ZA, Aman MJ, Honko AN, Griffiths A, Dye JM, Chandran K, Lai JR. Design and characterization of protective pan-ebolavirus and pan-filovirus bispecific antibodies. PLoS Pathog 2024; 20:e1012134. [PMID: 38603762 PMCID: PMC11037526 DOI: 10.1371/journal.ppat.1012134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 04/23/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Monoclonal antibodies (mAbs) are an important class of antiviral therapeutics. MAbs are highly selective, well tolerated, and have long in vivo half-life as well as the capacity to induce immune-mediated virus clearance. Their activities can be further enhanced by integration of their variable fragments (Fvs) into bispecific antibodies (bsAbs), affording simultaneous targeting of multiple epitopes to improve potency and breadth and/or to mitigate against viral escape by a single mutation. Here, we explore a bsAb strategy for generation of pan-ebolavirus and pan-filovirus immunotherapeutics. Filoviruses, including Ebola virus (EBOV), Sudan virus (SUDV), and Marburg virus (MARV), cause severe hemorrhagic fever. Although there are two FDA-approved mAb therapies for EBOV infection, these do not extend to other filoviruses. Here, we combine Fvs from broad ebolavirus mAbs to generate novel pan-ebolavirus bsAbs that are potently neutralizing, confer protection in mice, and are resistant to viral escape. Moreover, we combine Fvs from pan-ebolavirus mAbs with those of protective MARV mAbs to generate pan-filovirus protective bsAbs. These results provide guidelines for broad antiviral bsAb design and generate new immunotherapeutic candidates.
Collapse
MESH Headings
- Animals
- Mice
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/therapeutic use
- Ebolavirus/immunology
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Hemorrhagic Fever, Ebola/virology
- Antibodies, Viral/immunology
- Humans
- Filoviridae/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Monoclonal/immunology
- Female
- Mice, Inbred BALB C
- Filoviridae Infections/immunology
- Filoviridae Infections/therapy
- Filoviridae Infections/prevention & control
Collapse
Affiliation(s)
- Ariel S. Wirchnianski
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Elisabeth K. Nyakatura
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Andrew S. Herbert
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
- The Geneva Foundation, Tacoma, Washington, United States of America
| | - Ana I. Kuehne
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Shawn A. Abbasi
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
- The Geneva Foundation, Tacoma, Washington, United States of America
| | - Catalina Florez
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
- The Geneva Foundation, Tacoma, Washington, United States of America
| | - Nadia Storm
- Department of Virology, Immunology, and Microbiology; and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Lindsay G. A. McKay
- Department of Virology, Immunology, and Microbiology; and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Leandrew Dailey
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Erin Kuang
- Mapp Biopharmaceutical Inc., San Diego, California, United States of America
| | - Dafna M. Abelson
- Mapp Biopharmaceutical Inc., San Diego, California, United States of America
| | - Anna Z. Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Srinjoy Chakraborti
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | | | - Sergey Shulenin
- Integrated BioTherapeutics, Inc., Rockville, Maryland, United States of America
| | | | - M. Javad Aman
- Integrated BioTherapeutics, Inc., Rockville, Maryland, United States of America
| | - Anna N. Honko
- Department of Virology, Immunology, and Microbiology; and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Anthony Griffiths
- Department of Virology, Immunology, and Microbiology; and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - John M. Dye
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Jonathan R. Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
30
|
Driscoll CL, Keeble AH, Howarth MR. SpyMask enables combinatorial assembly of bispecific binders. Nat Commun 2024; 15:2403. [PMID: 38493197 PMCID: PMC10944524 DOI: 10.1038/s41467-024-46599-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 03/04/2024] [Indexed: 03/18/2024] Open
Abstract
Bispecific antibodies are a successful and expanding therapeutic class. Standard approaches to generate bispecifics are complicated by the need for disulfide reduction/oxidation or specialized formats. Here we present SpyMask, a modular approach to bispecifics using SpyTag/SpyCatcher spontaneous amidation. Two SpyTag-fused antigen-binding modules can be precisely conjugated onto DoubleCatcher, a tandem SpyCatcher where the second SpyCatcher is protease-activatable. We engineer a panel of structurally-distinct DoubleCatchers, from which binders project in different directions. We establish a generalized methodology for one-pot assembly and purification of bispecifics in 96-well plates. A panel of binders recognizing different HER2 epitopes were coupled to DoubleCatcher, revealing unexpected combinations with anti-proliferative or pro-proliferative activity on HER2-addicted cancer cells. Bispecific activity depended sensitively on both binder orientation and DoubleCatcher scaffold geometry. These findings support the need for straightforward assembly in different formats. SpyMask provides a scalable tool to discover synergy in bispecific activity, through modulating receptor organization and geometry.
Collapse
Affiliation(s)
- Claudia L Driscoll
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Anthony H Keeble
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Mark R Howarth
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK.
| |
Collapse
|
31
|
Rong Y, Chen IL, Larrabee L, Sawant MS, Fuh G, Koenig P. An Engineered Mouse Model That Generates a Diverse Repertoire of Endogenous, High-Affinity Common Light Chain Antibodies. Antibodies (Basel) 2024; 13:14. [PMID: 38390875 PMCID: PMC10885109 DOI: 10.3390/antib13010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Bispecific antibodies have gained increasing popularity as therapeutics as they enable novel activities that cannot be achieved with monospecific antibodies. Some of the most popular bispecific formats are molecules in which two Fab arms with different antigen specificities are combined into one IgG-like molecule. One way to produce these bispecific molecules requires the discovery of antibodies against the two antigens of interest that share a common light chain. Here, we present the generation and characterization of a common light chain mouse model, in which the endogenous IGKJ cluster is replaced with a prearranged, modified murine IGKV10-96/IGKJ1 segment. We demonstrate that genetic modification does not impact B-cell development. Upon immunization with ovalbumin, the animals generate an antibody repertoire with VH gene segment usage of a similar diversity to wildtype mice, while the light chain diversity is restricted to antibodies derived from the prearranged IGKV10-96/IGKJ1 germline. We further show that the clonotype diversity of the common light chain immune repertoire matches the diversity of immune repertoire isolated from wildtype mice. Finally, the common light chain anti-ovalbumin antibodies have only slightly lower affinities than antibodies isolated from wildtype mice, demonstrating the suitability of these animals for antibody discovery for bispecific antibody generation.
Collapse
Affiliation(s)
- Yinghui Rong
- 23andMe, Inc. Therapeutics, 349 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - I-Ling Chen
- 23andMe, Inc. Therapeutics, 349 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - Lance Larrabee
- 23andMe, Inc. Therapeutics, 349 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - Manali S Sawant
- 23andMe, Inc. Therapeutics, 349 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - Germaine Fuh
- 23andMe, Inc. Therapeutics, 349 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - Patrick Koenig
- 23andMe, Inc. Therapeutics, 349 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| |
Collapse
|
32
|
Barron N, Dickgiesser S, Fleischer M, Bachmann AN, Klewinghaus D, Hannewald J, Ciesielski E, Kusters I, Hammann T, Krause V, Fuchs SW, Siegmund V, Gross AW, Mueller-Pompalla D, Krah S, Zielonka S, Doerner A. A Generic Approach for Miniaturized Unbiased High-Throughput Screens of Bispecific Antibodies and Biparatopic Antibody-Drug Conjugates. Int J Mol Sci 2024; 25:2097. [PMID: 38396776 PMCID: PMC10889805 DOI: 10.3390/ijms25042097] [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: 11/20/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The toolbox of modern antibody engineering allows the design of versatile novel functionalities exceeding nature's repertoire. Many bispecific antibodies comprise heterodimeric Fc portions recently validated through the approval of several bispecific biotherapeutics. While heterodimerization methodologies have been established for low-throughput large-scale production, few approaches exist to overcome the bottleneck of large combinatorial screening efforts that are essential for the identification of the best possible bispecific antibody. This report presents a novel, robust and miniaturized heterodimerization process based on controlled Fab-arm exchange (cFAE), which is applicable to a variety of heterodimeric formats and compatible with automated high-throughput screens. Proof of applicability was shown for two therapeutic molecule classes and two relevant functional screening read-outs. First, the miniaturized production of biparatopic anti-c-MET antibody-drug conjugates served as a proof of concept for their applicability in cytotoxic screenings on tumor cells with different target expression levels. Second, the automated workflow enabled a large unbiased combinatorial screening of biparatopic antibodies and the identification of hits mediating potent c-MET degradation. The presented workflow utilizes standard equipment and may serve as a facile, efficient and robust method for the discovery of innovative therapeutic agents in many laboratories worldwide.
Collapse
Affiliation(s)
- Nadine Barron
- Protein and Cell Sciences, EMD Serono, 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Stephan Dickgiesser
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Markus Fleischer
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | | | - Daniel Klewinghaus
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Jens Hannewald
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Elke Ciesielski
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Ilja Kusters
- Protein Engineering and Antibody Technologies, EMD Serono, 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Til Hammann
- Discovery Pharmacology, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Volker Krause
- Discovery Pharmacology, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | | | - Vanessa Siegmund
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Alec W. Gross
- Protein Engineering and Antibody Technologies, EMD Serono, 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Dirk Mueller-Pompalla
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Simon Krah
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Stefan Zielonka
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Achim Doerner
- NBE Technologies, Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| |
Collapse
|
33
|
Gandhi PS, Zivkovic M, Østergaard H, Bonde AC, Elm T, Løvgreen MN, Schluckebier G, Johansson E, Olsen OH, Olsen EHN, de Bus IA, Bloem K, Alskär O, Rea CJ, Bjørn SE, Schutgens RE, Sørensen B, Urbanus RT, Faber JH. A bispecific antibody approach for the potential prophylactic treatment of inherited bleeding disorders. NATURE CARDIOVASCULAR RESEARCH 2024; 3:166-185. [PMID: 39196196 PMCID: PMC11358003 DOI: 10.1038/s44161-023-00418-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 12/19/2023] [Indexed: 08/29/2024]
Abstract
Inherited bleeding disorders such as Glanzmann thrombasthenia (GT) lack prophylactic treatment options. As a result, serious bleeding episodes are treated acutely with blood product transfusions or frequent, repeated intravenous administration of recombinant activated coagulation factor VII (rFVIIa). Here we describe HMB-001, a bispecific antibody designed to bind and accumulate endogenous FVIIa and deliver it to sites of vascular injury by targeting it to the TREM (triggering receptor expressed on myeloid cells)-like transcript-1 (TLT-1) receptor that is selectively expressed on activated platelets. In healthy nonhuman primates, HMB-001 prolonged the half-life of endogenous FVIIa, resulting in its accumulation. Mouse bleeding studies confirmed antibody-mediated potentiation of FVIIa hemostatic activity by TLT-1 targeting. In ex vivo models of GT, HMB-001 localized FVIIa on activated platelets and potentiated fibrin-dependent platelet aggregation. Taken together, these results indicate that HMB-001 has the potential to offer subcutaneous prophylactic treatment to prevent bleeds in people with GT and other inherited bleeding disorders, with a low-frequency dosing regimen.
Collapse
Affiliation(s)
| | - Minka Zivkovic
- Center for Benign Haematology, Thrombosis and Haemostasis, Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | | | | | | | | | | | - Ole H Olsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Karien Bloem
- Sanquin Diagnostic Services, Amsterdam, Netherlands
| | | | | | | | - Roger E Schutgens
- Center for Benign Haematology, Thrombosis and Haemostasis, Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | - Rolf T Urbanus
- Center for Benign Haematology, Thrombosis and Haemostasis, Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.
| | | |
Collapse
|
34
|
Madsen AV, Pedersen LE, Kristensen P, Goletz S. Design and engineering of bispecific antibodies: insights and practical considerations. Front Bioeng Biotechnol 2024; 12:1352014. [PMID: 38333084 PMCID: PMC10850309 DOI: 10.3389/fbioe.2024.1352014] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Bispecific antibodies (bsAbs) have attracted significant attention due to their dual binding activity, which permits simultaneous targeting of antigens and synergistic binding effects beyond what can be obtained even with combinations of conventional monospecific antibodies. Despite the tremendous therapeutic potential, the design and construction of bsAbs are often hampered by practical issues arising from the increased structural complexity as compared to conventional monospecific antibodies. The issues are diverse in nature, spanning from decreased biophysical stability from fusion of exogenous antigen-binding domains to antibody chain mispairing leading to formation of antibody-related impurities that are very difficult to remove. The added complexity requires judicious design considerations as well as extensive molecular engineering to ensure formation of high quality bsAbs with the intended mode of action and favorable drug-like qualities. In this review, we highlight and summarize some of the key considerations in design of bsAbs as well as state-of-the-art engineering principles that can be applied in efficient construction of bsAbs with diverse molecular formats.
Collapse
Affiliation(s)
- Andreas V. Madsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lasse E. Pedersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Peter Kristensen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
35
|
Yao X, Xie M, Ben Y, Zhu Y, Yang G, Kwong SCW, Zhang Z, Chiu ML. Large scale controlled Fab exchange GMP process to prepare bispecific antibodies. Front Bioeng Biotechnol 2024; 11:1298890. [PMID: 38283167 PMCID: PMC10812119 DOI: 10.3389/fbioe.2023.1298890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/20/2023] [Indexed: 01/30/2024] Open
Abstract
Objective: Bispecific antibodies (BsAbs) have demonstrated significant therapeutic impacts for the treatment of a broad spectrum of diseases that include oncology, auto-immune, and infectious diseases. However, the large-scale production of clinical batches of bispecific antibodies still has many challenges that include having low yield, poor stability, and laborious downstream purification processes. To address such challenges, we describe the optimization of the controlled Fab arm exchange (cFAE) process for the efficient generation of BsAbs. Methods: The process optimization of a large-scale good manufacturing practice (GMP) cFAE strategy to prepare BsAbs was based on screening the parameters of temperature, reduction, oxidation, and buffer exchange. We include critical quality standards for the reducing agent cysteamine hydrochloride. Results: This large-scale production protocol enabled the generation of bispecific antibodies with >90% exchange yield and at >95% purity. The subsequent downstream processing could use typical mAb procedures. Furthermore, we demonstrated that the bispecific generation protocol can be scaled up to ∼60 L reaction scale using parental monoclonal antibodies that were expressed in a 200 L bioreactor. Conclusion: We presented a robust development strategy for the cFAE process that can be used for a larger scale GMP BsAb production.
Collapse
Affiliation(s)
- Xia Yao
- Tavotek Biotherapeutics, Suzhou, China
| | | | | | - Yixiang Zhu
- Bioworkshops (Suzhou) Limited, Suzhou, China
| | | | | | | | - Mark L. Chiu
- Tavotek Biotherapeutics, Suzhou, China
- Tavotek Biotherapeutics, Lower Gwynedd, PA, United States
| |
Collapse
|
36
|
Damelang T, Brinkhaus M, van Osch TLJ, Schuurman J, Labrijn AF, Rispens T, Vidarsson G. Impact of structural modifications of IgG antibodies on effector functions. Front Immunol 2024; 14:1304365. [PMID: 38259472 PMCID: PMC10800522 DOI: 10.3389/fimmu.2023.1304365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Immunoglobulin G (IgG) antibodies are a critical component of the adaptive immune system, binding to and neutralizing pathogens and other foreign substances. Recent advances in molecular antibody biology and structural protein engineering enabled the modification of IgG antibodies to enhance their therapeutic potential. This review summarizes recent progress in both natural and engineered structural modifications of IgG antibodies, including allotypic variation, glycosylation, Fc engineering, and Fc gamma receptor binding optimization. We discuss the functional consequences of these modifications to highlight their potential for therapeutical applications.
Collapse
Affiliation(s)
- Timon Damelang
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Sanquin Research, Department of Immunopathology, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Maximilian Brinkhaus
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Thijs L. J. van Osch
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Janine Schuurman
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Aran F. Labrijn
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Theo Rispens
- Sanquin Research, Department of Immunopathology, Amsterdam, Netherlands
| | - Gestur Vidarsson
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
37
|
van der Wulp W, Luu W, Ressing ME, Schuurman J, van Kasteren SI, Guelen L, Hoeben RC, Bleijlevens B, Heemskerk MHM. Antibody-epitope conjugates deliver immunogenic T-cell epitopes more efficiently when close to cell surfaces. MAbs 2024; 16:2329321. [PMID: 38494955 PMCID: PMC10950288 DOI: 10.1080/19420862.2024.2329321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/07/2024] [Indexed: 03/19/2024] Open
Abstract
Antibody-mediated delivery of immunogenic viral CD8+ T-cell epitopes to redirect virus-specific T cells toward cancer cells is a promising new therapeutic avenue to increase the immunogenicity of tumors. Multiple strategies for viral epitope delivery have been shown to be effective. So far, most of these have relied on a free C-terminus of the immunogenic epitope for extracellular delivery. Here, we demonstrate that antibody-epitope conjugates (AECs) with genetically fused epitopes to the N-terminus of the antibody can also sensitize tumors for attack by virus-specific CD8+ T cells. AECs carrying epitopes genetically fused at the N-terminus of the light chains of cetuximab and trastuzumab demonstrate an even more efficient delivery of the T-cell epitopes compared to AECs with the epitope fused to the C-terminus of the heavy chain. We demonstrate that this increased efficiency is not caused by the shift in location of the cleavage site from the N- to the C-terminus, but by its increased proximity to the cell surface. We hypothesize that this facilitates more efficient epitope delivery. These findings not only provide additional insights into the mechanism of action of AECs but also broaden the possibilities for genetically fused AECs as an avenue for the redirection of multiple virus-specific T cells toward tumors.
Collapse
Affiliation(s)
- W. van der Wulp
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - W. Luu
- Genmab, Utrecht, The Netherlands
| | - M. E. Ressing
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - S. I. van Kasteren
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | | - R. C. Hoeben
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - M. H. M. Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
38
|
Evers A, Krah S, Demir D, Gaa R, Elter D, Schroeter C, Zielonka S, Rasche N, Dotterweich J, Knuehl C, Doerner A. Engineering hydrophobicity and manufacturability for optimized biparatopic antibody-drug conjugates targeting c-MET. MAbs 2024; 16:2302386. [PMID: 38214660 PMCID: PMC10793681 DOI: 10.1080/19420862.2024.2302386] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024] Open
Abstract
Optimal combinations of paratopes assembled into a biparatopic antibody have the capacity to mediate high-grade target cross-linking on cell membranes, leading to degradation of the target, as well as antibody and payload delivery in the case of an antibody-drug conjugate (ADC). In the work presented here, molecular docking suggested a suitable paratope combination targeting c-MET, but hydrophobic patches in essential binding regions of one moiety necessitated engineering. In addition to rational design of HCDR2 and HCDR3 mutations, site-specific spiking libraries were generated and screened in yeast and mammalian surface display approaches. Comparative analyses revealed similar positions amendable for hydrophobicity reduction, with a broad combinatorial diversity obtained from library outputs. Optimized variants showed high stability, strongly reduced hydrophobicity, retained affinities supporting the desired functionality and enhanced producibility. The resulting biparatopic anti-c-MET ADCs were comparably active on c-MET expressing tumor cell lines as REGN5093 exatecan DAR6 ADC. Structural molecular modeling of paratope combinations for preferential inter-target binding combined with protein engineering for manufacturability yielded deep insights into the capabilities of rational and library approaches. The methodologies of in silico hydrophobicity identification and sequence optimization could serve as a blueprint for rapid development of optimal biparatopic ADCs targeting further tumor-associated antigens in the future.
Collapse
Affiliation(s)
- Andreas Evers
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Simon Krah
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Deniz Demir
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Ramona Gaa
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Desislava Elter
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Stefan Zielonka
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Nicolas Rasche
- ADC and Targeted Therapeutics, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Christine Knuehl
- Research Unit Oncology, Merck Healthcare KGaA, Darmstadt, Germany
| | - Achim Doerner
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
39
|
Condado-Morales I, Dingfelder F, Waibel I, Turnbull OM, Patel B, Cao Z, Rose Bjelke J, Nedergaard Grell S, Bennet A, Hummer AM, Raybould MIJ, Deane CM, Egebjerg T, Lorenzen N, Arosio P. A comparative study of the developability of full-length antibodies, fragments, and bispecific formats reveals higher stability risks for engineered constructs. MAbs 2024; 16:2403156. [PMID: 39364796 PMCID: PMC11457596 DOI: 10.1080/19420862.2024.2403156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/16/2024] [Accepted: 09/07/2024] [Indexed: 10/05/2024] Open
Abstract
Engineered antibody formats, such as antibody fragments and bispecifics, have the potential to offer improved therapeutic efficacy compared to traditional full-length monoclonal antibodies (mAbs). However, the translation of these non-natural molecules into successful therapeutics can be hampered by developability challenges. Here, we systematically analyzed 64 different antibody constructs targeting Tumor Necrosis Factor (TNF) which cover 8 distinct molecular format families, encompassing full-length antibodies, various types of single chain variable fragments, and bispecifics. We measured 15 biophysical properties related to activity, manufacturing, and stability, scoring variants with a flag-based risk approach and a recent in silico developability profiler. Our comparative assessment revealed that overall developability is higher for the natural full-length antibody format. Bispecific antibodies, antibodies with scFv fragments at the C-terminus of the light chain, and single-chain Fv antibody fragments (scFvs) have intermediate developability properties, while more complicated formats, such as scFv- scFv, bispecific mAbs with one Fab exchanged with a scFv, and diabody formats are collectively more challenging. In particular, our study highlights the propensity for fragmentation and aggregation, both in bulk and at interfaces, for many current engineered formats.
Collapse
Affiliation(s)
- Itzel Condado-Morales
- Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Fabian Dingfelder
- Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Isabel Waibel
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | - Bhargav Patel
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Zheng Cao
- Department of Bioanalysis, Beijing Novo Nordisk Pharmaceutical Science & Technology Co. Ltd (Novo Nordisk R&D China), Beijing, China
| | - Jais Rose Bjelke
- Department of Purification Technologies, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | | | - Anja Bennet
- Department of Kidney Biology, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | | | | | | | - Thomas Egebjerg
- Department of Mammalian Expression, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | - Nikolai Lorenzen
- Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| |
Collapse
|
40
|
Fawcett C, Tickle JR, Coles CH. Facilitating high throughput bispecific antibody production and potential applications within biopharmaceutical discovery workflows. MAbs 2024; 16:2311992. [PMID: 39674918 DOI: 10.1080/19420862.2024.2311992] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 12/17/2024] Open
Abstract
A major driver for the recent investment surge in bispecific antibody (bsAb) platforms and products is the multitude of distinct mechanisms of action that bsAbs offer compared to a combination of two monoclonal antibodies. Four bsAb products were granted first regulatory approvals in the US or EU during 2023 and the biopharmaceutical industry pipeline is brimming with bsAb candidates across a broad range of therapeutic applications. In previously reported bsAb discovery campaigns, following a hypothesis-based choice of two specific target proteins, selections and screening activities have often been performed in mono-specific formats. The conversion to bispecific modalities has usually been positioned toward the end of the discovery process and has involved small numbers of lead molecules, largely due to challenges in expressing, purifying, and analyzing large numbers of bsAbs. In this review, we discuss emerging strategies to facilitate the production of expanded bsAb panels, focusing particularly upon combinatorial methods to generate bsAb matrices. Such technologies will enable screening in. bispecific formats at earlier stages of discovery campaigns, not only widening the accessible protein space to maximize chances of success, but also advancing empirical bi-target validation activities to assess initial target selection hypotheses.
Collapse
Affiliation(s)
- Caitlin Fawcett
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Joseph R Tickle
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
| | - Charlotte H Coles
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
| |
Collapse
|
41
|
Surowka M, Klein C. A pivotal decade for bispecific antibodies? MAbs 2024; 16:2321635. [PMID: 38465614 PMCID: PMC10936642 DOI: 10.1080/19420862.2024.2321635] [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: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/12/2024] Open
Abstract
Bispecific antibodies (bsAbs) are a class of antibodies that can mediate novel mechanisms of action compared to monospecific monoclonal antibodies (mAbs). Since the discovery of mAbs and their adoption as therapeutic agents in the 1980s and 1990s, the development of bsAbs has held substantial appeal. Nevertheless, only three bsAbs (catumaxomab, blinatumomab, emicizumab) were approved through the end of 2020. However, since then, 11 bsAbs received regulatory agency approvals, of which nine (amivantamab, tebentafusp, mosunetuzumab, cadonilimab, teclistamab, glofitamab, epcoritamab, talquetamab, elranatamab) were approved for the treatment of cancer and two (faricimab, ozoralizumab) in non-oncology indications. Notably, of the 13 currently approved bsAbs, two, emicizumab and faricimab, have achieved blockbuster status, showing the promise of this novel class of therapeutics. In the 2020s, the approval of additional bsAbs can be expected in hematological malignancies, solid tumors and non-oncology indications, establishing bsAbs as essential part of the therapeutic armamentarium.
Collapse
Affiliation(s)
- Marlena Surowka
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| |
Collapse
|
42
|
Guo X, Wu Y, Xue Y, Xie N, Shen G. Revolutionizing cancer immunotherapy: unleashing the potential of bispecific antibodies for targeted treatment. Front Immunol 2023; 14:1291836. [PMID: 38106416 PMCID: PMC10722299 DOI: 10.3389/fimmu.2023.1291836] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/08/2023] [Indexed: 12/19/2023] Open
Abstract
Recent progressions in immunotherapy have transformed cancer treatment, providing a promising strategy that activates the immune system of the patient to find and eliminate cancerous cells. Bispecific antibodies, which engage two separate antigens or one antigen with two distinct epitopes, are of tremendous concern in immunotherapy. The bi-targeting idea enabled by bispecific antibodies (BsAbs) is especially attractive from a medical standpoint since most diseases are complex, involving several receptors, ligands, and signaling pathways. Several research look into the processes in which BsAbs identify different cancer targets such angiogenesis, reproduction, metastasis, and immune regulation. By rerouting cells or altering other pathways, the bispecific proteins perform effector activities in addition to those of natural antibodies. This opens up a wide range of clinical applications and helps patients with resistant tumors respond better to medication. Yet, further study is necessary to identify the best conditions where to use these medications for treating tumor, their appropriate combination partners, and methods to reduce toxicity. In this review, we provide insights into the BsAb format classification based on their composition and symmetry, as well as the delivery mode, focus on the action mechanism of the molecule, and discuss the challenges and future perspectives in BsAb development.
Collapse
Affiliation(s)
- Xiaohan Guo
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yi Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ying Xue
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Guobo Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
43
|
Rispens T, Huijbers MG. The unique properties of IgG4 and its roles in health and disease. Nat Rev Immunol 2023; 23:763-778. [PMID: 37095254 PMCID: PMC10123589 DOI: 10.1038/s41577-023-00871-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
IgG4 is the least abundant subclass of IgG in human serum and has unique functional features. IgG4 is largely unable to activate antibody-dependent immune effector responses and, furthermore, undergoes Fab (fragment antigen binding)-arm exchange, rendering it bispecific for antigen binding and functionally monovalent. These properties of IgG4 have a blocking effect, either on the immune response or on the target protein of IgG4. In this Review, we discuss the unique structural characteristics of IgG4 and how these contribute to its roles in health and disease. We highlight how, depending on the setting, IgG4 responses can be beneficial (for example, in responses to allergens or parasites) or detrimental (for example, in autoimmune diseases, in antitumour responses and in anti-biologic responses). The development of novel models for studying IgG4 (patho)physiology and understanding how IgG4 responses are regulated could offer insights into novel treatment strategies for these IgG4-associated disease settings.
Collapse
Affiliation(s)
- Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Maartje G Huijbers
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
44
|
Abdeldaim DT, Schindowski K. Fc-Engineered Therapeutic Antibodies: Recent Advances and Future Directions. Pharmaceutics 2023; 15:2402. [PMID: 37896162 PMCID: PMC10610324 DOI: 10.3390/pharmaceutics15102402] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Monoclonal therapeutic antibodies have revolutionized the treatment of cancer and other diseases. Fc engineering aims to enhance the effector functions or half-life of therapeutic antibodies by modifying their Fc regions. Recent advances in the Fc engineering of modern therapeutic antibodies can be considered the next generation of antibody therapy. Various strategies are employed, including altering glycosylation patterns via glycoengineering and introducing mutations to the Fc region, thereby enhancing Fc receptor or complement interactions. Further, Fc engineering strategies enable the generation of bispecific IgG-based heterodimeric antibodies. As Fc engineering techniques continue to evolve, an expanding portfolio of Fc-engineered antibodies is advancing through clinical development, with several already approved for medical use. Despite the plethora of Fc-based mutations that have been analyzed in in vitro and in vivo models, we focus here in this review on the relevant Fc engineering strategies of approved therapeutic antibodies to finetune effector functions, to modify half-life and to stabilize asymmetric bispecific IgGs.
Collapse
Affiliation(s)
- Dalia T. Abdeldaim
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
| |
Collapse
|
45
|
Shah D, Soper B, Shopland L. Cytokine release syndrome and cancer immunotherapies - historical challenges and promising futures. Front Immunol 2023; 14:1190379. [PMID: 37304291 PMCID: PMC10248525 DOI: 10.3389/fimmu.2023.1190379] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Cancer is the leading cause of death worldwide. Cancer immunotherapy involves reinvigorating the patient's own immune system to fight against cancer. While novel approaches like Chimeric Antigen Receptor (CAR) T cells, bispecific T cell engagers, and immune checkpoint inhibitors have shown promising efficacy, Cytokine Release Syndrome (CRS) is a serious adverse effect and remains a major concern. CRS is a phenomenon of immune hyperactivation that results in excessive cytokine secretion, and if left unchecked, it may lead to multi-organ failure and death. Here we review the pathophysiology of CRS, its occurrence and management in the context of cancer immunotherapy, and the screening approaches that can be used to assess CRS and de-risk drug discovery earlier in the clinical setting with more predictive pre-clinical data. Furthermore, the review also sheds light on the potential immunotherapeutic approaches that can be used to overcome CRS associated with T cell activation.
Collapse
Affiliation(s)
- Deep Shah
- In vivo Services, The Jackson Laboratory, Sacramento, CA, United States
| | - Brian Soper
- Technical Information Services, The Jackson Laboratory, Bar Harbor, ME, United States
| | - Lindsay Shopland
- In vivo Services, The Jackson Laboratory, Sacramento, CA, United States
| |
Collapse
|
46
|
Radić L, Sliepen K, Yin V, Brinkkemper M, Capella-Pujol J, Schriek AI, Torres JL, Bangaru S, Burger JA, Poniman M, Bontjer I, Bouhuijs JH, Gideonse D, Eggink D, Ward AB, Heck AJ, Van Gils MJ, Sanders RW, Schinkel J. Bispecific antibodies combine breadth, potency, and avidity of parental antibodies to neutralize sarbecoviruses. iScience 2023; 26:106540. [PMID: 37063468 PMCID: PMC10065043 DOI: 10.1016/j.isci.2023.106540] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/07/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
SARS-CoV-2 variants evade current monoclonal antibody therapies. Bispecific antibodies (bsAbs) combine the specificities of two distinct antibodies taking advantage of the avidity and synergy provided by targeting different epitopes. Here we used controlled Fab-arm exchange to produce bsAbs that neutralize SARS-CoV and SARS-CoV-2 variants, including Omicron and its subvariants, by combining potent SARS-CoV-2-specific neutralizing antibodies with broader antibodies that also neutralize SARS-CoV. We demonstrated that the parental antibodies rely on avidity for neutralization using bsAbs containing one irrelevant Fab arm. Using mass photometry to measure the formation of antibody:spike complexes, we determined that bsAbs increase binding stoichiometry compared to corresponding cocktails, without a loss of binding affinity. The heterogeneous binding pattern of bsAbs to spike, observed by negative-stain electron microscopy and mass photometry provided evidence for both intra- and inter-spike crosslinking. This study highlights the utility of cross-neutralizing antibodies for designing bivalent agents to combat circulating and future SARS-like coronaviruses.
Collapse
Affiliation(s)
- Laura Radić
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Kwinten Sliepen
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Victor Yin
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
- Netherlands Proteomics Center, 3584 CH Utrecht, the Netherlands
| | - Mitch Brinkkemper
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Joan Capella-Pujol
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Angela I. Schriek
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Jonathan L. Torres
- Department of Structural Biology and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sandhya Bangaru
- Department of Structural Biology and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Judith A. Burger
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Meliawati Poniman
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Ilja Bontjer
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Joey H. Bouhuijs
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - David Gideonse
- Center for Infectious Disease Control, WHO COVID-19 reference laboratory, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | - Dirk Eggink
- Center for Infectious Disease Control, WHO COVID-19 reference laboratory, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | - Andrew B. Ward
- Department of Structural Biology and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Albert J.R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
- Netherlands Proteomics Center, 3584 CH Utrecht, the Netherlands
| | - Marit J. Van Gils
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Rogier W. Sanders
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Janke Schinkel
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| |
Collapse
|
47
|
He Y, Ma H, Wang C, Ai Z, Wu Q, Chen H, Lu D. A novel brick for bispecific antibody construction. Proteins 2023. [PMID: 36964928 DOI: 10.1002/prot.26492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 03/02/2023] [Accepted: 03/14/2023] [Indexed: 03/27/2023]
Abstract
In recent years, the development of bispecific antibodies (bsAbs) has become a major trend in the biopharmaceutical industry. By simultaneously engaging two molecular targets, bsAbs have exhibited unique mechanisms of action that could lead to clinical benefits unattainable by conventional monoclonal antibodies. The type of structure used to construct a bsAb directly influences the distance, angle, degree of freedom, and affinity between the two antibody binding sites and the interaction between the two antigens or the cells where the antigens are located, which have been bound by the antibody. Consequently, the structure of the bsAb is one of the most vital factors affecting its function. Herein, we reported for the first time a novel basic module bsAb format, VFV (Variable domain-Fab-Variable domain). And then, the feasibility of the VFV format was demonstrated by constructing a series of engager-like basic module bsAbs. Next, a series of VFV bsAbs containing Fc (VFV-Ig), Fab (VFV-Fab), or Hinge (VFV-Hinge) were developed based on Hxb module, and all of them had adequate purity and activity. Finally, a T cell engager bsAb with the potential to overcome on-target off-tumor activity was constructed according to the structural characteristics of VFV, which validated that the VFV module can be used as a new brick for the construction of various bsAbs. In a word, the successful construction of this bsAb format for the first time not only enriches the arsenal of the bsAb format, but also provides inspiration for the construction of new bsAbs. Nevertheless, we are fully aware that as a proof-of-concept study, this paper has many shortcomings, and there is still a lot of work to be done to determine whether VFV can serve as a platform for drug development.
Collapse
Affiliation(s)
- Yan He
- School of Life Sciences, Fudan University, Shanghai, China
| | - Haili Ma
- LongBio Pharma Co, Shanghai, China
| | - Cong Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhilong Ai
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiao Wu
- School of Life Sciences, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongyan Chen
- School of Life Sciences, Fudan University, Shanghai, China
| | - Daru Lu
- School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
48
|
Cho BC, Simi A, Sabari J, Vijayaraghavan S, Moores S, Spira A. Amivantamab, an Epidermal Growth Factor Receptor (EGFR) and Mesenchymal-epithelial Transition Factor (MET) Bispecific Antibody, Designed to Enable Multiple Mechanisms of Action and Broad Clinical Applications. Clin Lung Cancer 2023; 24:89-97. [PMID: 36481319 DOI: 10.1016/j.cllc.2022.11.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022]
Abstract
Substantial therapeutic advancements have been made in identifying and treating activating mutations in advanced non-small cell lung cancer (NSCLC); however, resistance to epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition factor (MET) inhibitors remains common with current targeted therapies. Amivantamab, a fully human bispecific antibody targeting EGFR and MET, is approved in the United States and other countries for the treatment of patients with advanced NSCLC with EGFR exon 20 insertion mutations, for whom disease has progressed on or after platinum-based chemotherapy. Preliminary efficacy and safety have also been demonstrated in patients with common EGFR- or MET-mutated NSCLC. Amivantamab employs 3 distinct potential mechanisms of action (MOAs) including ligand blocking, receptor degradation, and immune cell-directing activity, such as antibody-dependent cellular cytotoxicity and trogocytosis. Notably, efficacy with amivantamab does not require all 3 MOAs to occur simultaneously, broadening applicability by using diverse antitumor mechanisms. This review focuses on the molecular characteristics of amivantamab and its unique MOAs leading to in vitro and in vivo efficacy and safety in preclinical and clinical studies.
Collapse
Affiliation(s)
- Byoung Chul Cho
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro Sinchon-dong, Seodaemun-gu, Seoul, South Korea.
| | - Allison Simi
- Janssen Scientific Affairs, LLC, 800 Ridgeview Drive, Horsham, PA
| | - Joshua Sabari
- NYU Langone Health, 160 E 34th St 8th floor, New York, NY
| | | | - Sheri Moores
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA
| | | |
Collapse
|
49
|
Larivière L, Krüger JE, von Hirschheydt T, Schlothauer T, Bray-French K, Bader M, Runza V. End-to-end approach for the characterization and control of product-related impurities in T cell bispecific antibody preparations. Int J Pharm X 2023; 5:100157. [PMID: 36687375 PMCID: PMC9850176 DOI: 10.1016/j.ijpx.2023.100157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/23/2022] [Accepted: 01/01/2023] [Indexed: 01/04/2023] Open
Abstract
Antibody-based T cell-activating biologics are promising therapeutic medicines being developed for a number of indications, mainly in the oncology field. Among those, T cell bispecific antibodies are designed to bind one tumor-specific antigen and the T cell receptor at the same time, leading to a robust T cell response against the tumor. Although their unique format and the versatility of the CrossMab technology allows for the generation of safer molecules in an efficient manner, product-related variants cannot be completely avoided. Therefore, it is of extreme importance that both a manufacturing process that limits or depletes product-related impurities, as well as a thorough analytical characterization are in place, starting from the development of the manufacturing cell line until the assessment of potential toxicities. Here, we describe such an end-to-end approach to minimize, quantify and control impurities and -upon their functional characterization- derive specifications that allow for the release of clinical material.
Collapse
Key Words
- Antibody manufacturing process
- CE-SDS, capillary electrophoresis‑sodium dodecyl sulfate
- CRS, cytokine release syndrome
- ELISA, enzyme-linked immunosorbent assay
- End-to-end approach
- Fc, fragment crystallizable
- Functional characterization
- GMP, good manufacturing process
- HIC, hydrophobic interaction chromatography
- HMW, high molecular weight (species)
- IEX, ion exchange chromatography
- PBS, phosphate buffer saline
- Product-related impurities control
- SEC, size-exclusion chromatography
- SPR, surface plasmon resonance
- TAA, tumor-associated antigen
- TCB, T cell bispecific
- TCR, T cell receptor
Collapse
Affiliation(s)
- Laurent Larivière
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Julia Eva Krüger
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Thomas von Hirschheydt
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Tilman Schlothauer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Katharine Bray-French
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel 4058, Switzerland
| | - Martin Bader
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Valeria Runza
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany,Corresponding author.
| |
Collapse
|
50
|
Koga H, Yamano T, Betancur J, Nagatomo S, Ikeda Y, Yamaguchi K, Nabuchi Y, Sato K, Teranishi-Ikawa Y, Sato M, Hirayama H, Hayasaka A, Torizawa T, Haraya K, Sampei Z, Shiraiwa H, Kitazawa T, Igawa T, Kuramochi T. Efficient production of bispecific antibody by FAST-Ig TM and its application to NXT007 for the treatment of hemophilia A. MAbs 2023; 15:2222441. [PMID: 37339067 PMCID: PMC10283433 DOI: 10.1080/19420862.2023.2222441] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023] Open
Abstract
Efficient production of bispecific antibodies (BsAbs) in single mammalian cells is essential for basic research and industrial manufacturing. However, preventing unwanted pairing of heavy chains (HCs) and light chains (LCs) is a challenging task. To address this, we created an engineering technology for preferential cognate HC/LC and HC/HC paring called FAST-Ig (Four-chain Assembly by electrostatic Steering Technology - Immunoglobulin), and applied it to NXT007, a BsAb for the treatment of hemophilia A. We introduced charged amino-acid substitutions at the HC/LC interface to facilitate the proper assembly for manufacturing a standard IgG-type BsAb. We generated CH1/CL interface-engineered antibody variants that achieved > 95% correct HC/LC pairing efficiency with favorable pharmacological properties and developability. Among these, we selected a design (C3) that allowed us to separate the mis-paired species with an unintended pharmacological profile using ion-exchange chromatography. Crystal structure analysis demonstrated that the C3 design did not affect the overall structure of both Fabs. To determine the final design for HCs-heterodimerization, we compared the stability of charge-based and knobs into hole-based Fc formats in acidic conditions and selected the more stable charge-based format. FAST-Ig was also applicable to stable CHO cell lines for industrial production and demonstrated robust chain pairing with different subclasses of parent BsAbs. Thus, it can be applied to a wide variety of BsAbs both preclinically and clinically.
Collapse
Affiliation(s)
- Hikaru Koga
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Takashi Yamano
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Juan Betancur
- API Process Development Department, Chugai Pharmaceutical Co., Ltd, Ukima, Tokyo, Japan
| | - Satoko Nagatomo
- Analytical Development Department, Chugai Pharmaceutical Co, Ltd, Ukima, Tokyo, Japan
| | - Yousuke Ikeda
- Analytical Development Department, Chugai Pharmaceutical Co, Ltd, Ukima, Tokyo, Japan
| | - Kazuki Yamaguchi
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Yoshiaki Nabuchi
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Kazuki Sato
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | | | - Motohiko Sato
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Hiroyuki Hirayama
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Akira Hayasaka
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Takuya Torizawa
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Kenta Haraya
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Zenjiro Sampei
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Hirotake Shiraiwa
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Takehisa Kitazawa
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Tomoyuki Igawa
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Chuo-Ku, Tokyo, Japan
| | - Taichi Kuramochi
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| |
Collapse
|