1
|
Bartsch YC, Webb NE, Burgess E, Kang J, Lauffenburger DA, Julg BD. Combinatorial Fc modifications for complementary antibody functionality. MAbs 2025; 17:2465391. [PMID: 39950649 PMCID: PMC11834420 DOI: 10.1080/19420862.2025.2465391] [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: 11/26/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) can be functionally enhanced via Fc engineering. To determine whether pairs of mAbs with different Fc modifications can be combined for functional complementarity, we investigated the in vitro activity of two HIV-1 mAb libraries, each equipped with 60 engineered Fc variants. Our findings demonstrate that the impact of Fc engineering on Fc functionality is dependent on the specific Fab clone. Notably, combinations of Fc variants of the same Fab specificity exhibited limited enhancement in functional breadth compared to combinations involving two distinct Fabs. This suggests that the strategic selection of complementary Fc modifications can enhance both functional activity and breadth. Furthermore, while some combinations of Fc variants displayed additive functional effects, others were detrimental, suggesting that the functional outcome of Fc mutations is not easily predicted. Collectively, these results provide preliminary evidence supporting the potential of complementary Fc modifications in mAb combinations. Future studies will be essential to identify the optimal Fc modifications that maximize in vivo efficacy.
Collapse
Affiliation(s)
- Yannic C. Bartsch
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
- Laboratory of Anti-Viral Antibody-Omics, TWINCORE-Institute for Experimental Infection Research, Helmholtz Center for Infection Research (HZI) and Medical School Hannover (MHH) and Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| | - Nicholas E. Webb
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Eleanor Burgess
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Jaewon Kang
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
| | | | - Boris D. Julg
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
2
|
Keith AD, Xu T, Chernova TA, Keen MM, Bogacz M, Nedeljković M, Flowers M, Brown T, Frank F, Ortlund EA, Sundberg EJ. Mapping affinity and allostery in human IgG antibody Fc region-Fc γ receptor interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645945. [PMID: 40236212 PMCID: PMC11996314 DOI: 10.1101/2025.03.28.645945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
IgG antibodies, required for a functional immune system, recognize antigens and neutralize pathogens using their Fab regions, while signaling to the immune system by binding to host Fc γ receptors (FcγRs) through their Fc regions. These FcγR interactions initiate and modulate antibody-mediated effector functions that are essential for host immunity, therapeutic monoclonal antibody effectiveness and IgG-mediated pathologies. FcγRs include both activating and inhibitory receptors and the relative binding affinities of the IgG Fc region to FcγRs that generate opposing signals is a key determinant of the immune response. Substantial research effort has been devoted to understanding and manipulating FcγR interactions to decipher their fundamental biological activities and to develop therapeutic monoclonal antibodies with tailored effector functions. However, a common Fc-FcγR binding interface, the high sequence identity of FcγRs, and the inherent conformational dynamics of the IgG Fc region, have prohibited a full understanding of these interactions, even when employing state-of-the-art biophysical and biological methods. Here, we used site-saturation libraries of the human IgG1 Fc region to determine the effective affinities of more than 98% of all possible single-site amino acid substitutions in the Fc to all human FcγRs, as well as the most common FcγR polymorphisms. We provide a comprehensive analysis of Fc amino acid variations that determine Fc stability, orthosteric control of FcγR binding, and short- and long-range allosteric control of FcγR binding. We also predict the relative activating versus inhibitory effector function capacity of nearly every possible single-site Fc mutation.
Collapse
|
3
|
Ovcinnikovs V, Dijkman K, Zom GG, Beurskens FJ, Trouw LA. Enhancing complement activation by therapeutic anti-tumor antibodies: Mechanisms, strategies, and engineering approaches. Semin Immunol 2025; 77:101922. [PMID: 39742715 DOI: 10.1016/j.smim.2024.101922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 01/04/2025]
Abstract
The complement system plays an integral role in both innate and adaptive immune responses. Beyond its protective function against infections, complement is also known to influence tumor immunity, where its activation can either promote tumor progression or mediate tumor cell destruction, depending on the context. One such context can be provided by antibodies, with their inherent capacity to activate the classical complement pathway. In recent years, our understanding of the mechanisms governing complement activation by IgG and IgM antibodies has expanded significantly. At the same time, preclinical and clinical studies on antibodies such as rituximab, ofatumumab, and daratumumab have provided evidence for the role of complement in therapeutic success, encouraging strategies to further enhance its activity. In this review we examine the main determinants of antibody-mediated complement activation, highlighting the importance of antibody subclass, affinity, valency, and geometry of antigen engagement. We summarize the evidence for complement involvement in anti-tumor activity and challenges of accurately estimating the extent of its contribution to therapeutic efficacy. Furthermore, we explore several engineering approaches designed to enhance complement activation, including increased Fc oligomerization and C1q affinity, bispecific C1q-recruiting antibodies, IgG subclass chimeras, as well as antibody and paratope combinations. Strategies targeting membrane-bound complement regulatory proteins to overcome tumor-associated complement inhibition are also discussed as a method to boost therapeutic efficacy. Finally, we highlight the potential of complement-dependent cellular cytotoxicity (CDCC) and complement-dependent cellular phagocytosis (CDCP) as effector mechanisms that warrant deeper investigation. By integrating advances in antibody and complement biology with insights from efforts to enhance complement activation in therapeutic antibodies, this review aims to provide a comprehensive framework of antibody design and engineering strategies that optimize complement activity for improved anti-tumor efficacy.
Collapse
Affiliation(s)
| | - Karin Dijkman
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Leendert A Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
4
|
Kacar M, Al-Hakim A, Savic S. Sequelae of B-Cell Depleting Therapy: An Immunologist's Perspective. BioDrugs 2025; 39:103-130. [PMID: 39680306 DOI: 10.1007/s40259-024-00696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
B-cell depleting therapy (BCDT) has revolutionised the treatment of B-cell malignancies and autoimmune diseases by targeting specific B-cell surface antigens, receptors, ligands, and signalling pathways. This narrative review explores the mechanisms, applications, and complications of BCDT, focusing on the therapeutic advancements since the introduction of rituximab in 1997. Various monoclonal antibodies and kinase inhibitors are examined for their roles in depleting B cells through antibody-dependent and independent mechanisms. The off-target effects, such as hypogammaglobulinemia, infections, and cytokine release syndrome, are discussed, emphasising the need for immunologists to identify and help manage these complications. The increasing prevalence of BCDT has necessitated the involvement of clinical immunologists in addressing treatment-associated immunological abnormalities, including persistent hypogammaglobulinemia and neutropenia. We highlight the importance of considering underlying inborn errors of immunity (IEI) in patients presenting with these complications. Furthermore, we discuss the impact of BCDT on other immune cell populations and the challenges in predicting and managing long-term immunological sequelae. The potential for novel BCDT agents targeting the BAFF/APRIL-TACI/BCMA axis and B-cell receptor signalling pathways to treat autoimmune disorders is also explored, underscoring the rapidly evolving landscape of B-cell targeted therapies.
Collapse
Affiliation(s)
- Mark Kacar
- Department of Allergy, University Clinic Golnik, Golnik, Slovenia
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Adam Al-Hakim
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Sinisa Savic
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK.
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds, UK.
| |
Collapse
|
5
|
Ellerman DA. The Evolving Applications of Bispecific Antibodies: Reaping the Harvest of Early Sowing and Planting New Seeds. BioDrugs 2025; 39:75-102. [PMID: 39673023 DOI: 10.1007/s40259-024-00691-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 12/15/2024]
Abstract
After decades of gradual progress from conceptualization to early clinical trials (1960-2000), the therapeutic potential of bispecific antibodies (bisp Abs) is now being fully realized. Insights gained from both successful and unsuccessful trials are helping to identify which mechanisms of action, antibody formats, and targets prove most effective, and which may benefit from further refinement. While T-cell engagers remain the most commonly used class of bisp Abs, current efforts aim to increase their effectiveness by co-engaging costimulatory molecules, reducing escape mechanisms, and countering immunosuppression. Strategies to minimize cytokine release syndrome (CRS) are also actively under development. In addition, novel antibody formats that are selectively activated within tumors are an exciting area of research, as is the precise targeting of specific T-cell subsets. Beyond T cells, the recruitment of macrophages and dendritic cells is attracting increasing interest, with researchers exploring various macrophage receptors to promote phagocytosis or to carry out specialized functions, such as the immunologically silent clearance of amyloid-beta plaques in the brain. While bisp Abs targeting B cells are relatively limited, they are primarily aimed at inhibiting B-cell activity in autoimmune diseases. Another evolving application involves the forced interaction between proteins. Beyond the successful development of Hemlibra for hemophilia, bispecific antibodies that mimic cytokine activity are being explored. Additionally, the recruitment of cell surface ubiquitin ligases and other enzymes represents a novel and promising therapeutic strategy. In regard to antibody formats, some applications such as the combination of T-cell engagers with costimulatory molecules are driving the development of trispecific antibodies, at least in preclinical settings. However, the increasing structural complexity of multispecific antibodies often leads to more challenging development paths, and the number of multispecific antibodies in clinical trials remains low. The clinical success of certain applications and well-established production methods position this therapeutic class to expand its benefits into other therapeutic areas.
Collapse
Affiliation(s)
- Diego A Ellerman
- Antibody Engineering Department, Genentech Inc, South San Francisco, USA.
| |
Collapse
|
6
|
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
|
7
|
Eggel A, Pennington LF, Jardetzky TS. Therapeutic monoclonal antibodies in allergy: Targeting IgE, cytokine, and alarmin pathways. Immunol Rev 2024; 328:387-411. [PMID: 39158477 PMCID: PMC11659931 DOI: 10.1111/imr.13380] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
The etiology of allergy is closely linked to type 2 inflammatory responses ultimately leading to the production of allergen-specific immunoglobulin E (IgE), a key driver of many allergic conditions. At a high level, initial allergen exposure disrupts epithelial integrity, triggering local inflammation via alarmins including IL-25, IL-33, and TSLP, which activate type 2 innate lymphoid cells as well as other immune cells to secrete type 2 cytokines IL-4, IL-5 and IL-13, promoting Th2 cell development and eosinophil recruitment. Th2 cell dependent B cell activation promotes the production of allergen-specific IgE, which stably binds to basophils and mast cells. Rapid degranulation of these cells upon allergen re-exposure leads to allergic symptoms. Recent advances in our understanding of the molecular and cellular mechanisms underlying allergic pathophysiology have significantly shaped the development of therapeutic intervention strategies. In this review, we highlight key therapeutic targets within the allergic cascade with a particular focus on past, current and future treatment approaches using monoclonal antibodies. Specific targeting of alarmins, type 2 cytokines and IgE has shown varying degrees of clinical benefit in different allergic indications including asthma, chronic spontaneous urticaria, atopic dermatitis, chronic rhinosinusitis with nasal polyps, food allergies and eosinophilic esophagitis. While multiple therapeutic antibodies have been approved for clinical use, scientists are still working on ways to improve on current treatment approaches. Here, we provide context to understand therapeutic targeting strategies and their limitations, discussing both knowledge gaps and promising future directions to enhancing clinical efficacy in allergic disease management.
Collapse
Affiliation(s)
- Alexander Eggel
- Department for BioMedical ResearchUniversity of BernBernSwitzerland
- Department of Rheumatology and ImmunologyUniversity Hospital BernBernSwitzerland
| | | | - Theodore S. Jardetzky
- Department of Structural BiologyStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
8
|
Lefranc M, Lefranc G. Using IMGT unique numbering for IG allotypes and Fc-engineered variants of effector properties and half-life of therapeutic antibodies. Immunol Rev 2024; 328:473-506. [PMID: 39367563 PMCID: PMC11659927 DOI: 10.1111/imr.13399] [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: 10/06/2024]
Abstract
Therapeutic monoclonal antibodies (mAb) are usually of the IgG1, IgG2, and IgG4 classes, and their heavy chains may be modified by amino acid (aa) changes involved in antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), complement-dependent cytotoxicity (CDC), and/or half-life. Allotypes and Fc-engineered variants are classified using IMGT/HGNC gene nomenclature (e.g., Homo sapiens IGHG1). Allotype names follow the WHO/IMGT nomenclature. IMGT-engineered variant names use the IMGT nomenclature (e.g., Homsap G1v1), which comprises species and gene name (both abbreviated) followed by the letter v (for variant) and a number. Both allotypes and engineered variants are defined by their aa changes and positions, based on the IMGT unique numbering for C domain, identified in sequence motifs, referred to as IMGT topological motifs, as their limits and length are standardized and correspond to a structural feature (e.g., strand or loop). One hundred twenty-six variants are displayed with their type, IMGT numbering, Eu-IMGT positions, motifs before and after changes, and their property and function (effector and half-life). Three motifs characterize effector variants, CH2 1.6-3, 23-BC-41, and the FG loop, whereas three different motifs characterize half-life variants, two on CH2 13-AB-18 and 89-96 with H93, and one on CH3 the FG loop with H115.
Collapse
Affiliation(s)
- Marie‐Paule Lefranc
- IMGT®, the international ImMunoGeneTics information system® (IMGT), Laboratoire d'ImmunoGénétique Moléculaire (LIGM), Institut de Génétique Humaine (IGH), UMR 9002 Centre National de la Recherche Scientifique (CNRS)Université de Montpellier (UM)Montpellier Cedex 5France
| | - Gérard Lefranc
- IMGT®, the international ImMunoGeneTics information system® (IMGT), Laboratoire d'ImmunoGénétique Moléculaire (LIGM), Institut de Génétique Humaine (IGH), UMR 9002 Centre National de la Recherche Scientifique (CNRS)Université de Montpellier (UM)Montpellier Cedex 5France
| |
Collapse
|
9
|
Su X, Yu H, Lei Q, Chen X, Tong Y, Zhang Z, Yang W, Guo Y, Lin L. Systemic lupus erythematosus: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:54. [PMID: 39472388 PMCID: PMC11522254 DOI: 10.1186/s43556-024-00217-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disorder characterized by dysregulated immune responses and autoantibody production, which affects multiple organs and varies in clinical presentation and disease severity. The development of SLE is intricate, encompassing dysregulation within the immune system, a collapse of immunological tolerance, genetic susceptibilities to the disease, and a variety of environmental factors that can act as triggers. This review provides a comprehensive discussion of the pathogenesis and treatment strategies of SLE and focuses on the progress and status of traditional and emerging treatment strategies for SLE. Traditional treatment strategies for SLE have mainly employed non-specific approaches, including cytotoxic and immunosuppressive drugs, antimalarials, glucocorticoids, and NSAIDs. These strategies are effective in mitigating the effects of the disease, but they are not a complete cure and are often accompanied by adverse reactions. Emerging targeted therapeutic drugs, on the other hand, aim to control and treat SLE by targeting B and T cells, inhibiting their activation and function, as well as the abnormal activation of the immune system. A deeper understanding of the pathogenesis of SLE and the exploration of new targeted treatment strategies are essential to advance the treatment of this complex autoimmune disease.
Collapse
Affiliation(s)
- Xu Su
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Hui Yu
- Department of Urology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China
| | - Qingqiang Lei
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400000, China
| | - Xuerui Chen
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Yanli Tong
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, Paris, F-75015, France
| | - Zhongyang Zhang
- Department of Health Technology, The Danish National Research Foundation and Villum Foundation's Center IDUN, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Wenyong Yang
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Department of Neurosurgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China.
| | - Yuanbiao Guo
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| | - Liangbin Lin
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
10
|
Aboul-Ella H, Gohar A, Ali AA, Ismail LM, Mahmoud AEER, Elkhatib WF, Aboul-Ella H. Monoclonal antibodies: From magic bullet to precision weapon. MOLECULAR BIOMEDICINE 2024; 5:47. [PMID: 39390211 PMCID: PMC11467159 DOI: 10.1186/s43556-024-00210-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Monoclonal antibodies (mAbs) are used to prevent, detect, and treat a broad spectrum of non-communicable and communicable diseases. Over the past few years, the market for mAbs has grown exponentially with an expected compound annual growth rate (CAGR) of 11.07% from 2024 (237.64 billion USD estimated at the end of 2023) to 2033 (679.03 billion USD expected by the end of 2033). Ever since the advent of hybridoma technology introduced in 1975, antibody-based therapeutics were realized using murine antibodies which further progressed into humanized and fully human antibodies, reducing the risk of immunogenicity. Some benefits of using mAbs over conventional drugs include a drastic reduction in the chances of adverse reactions, interactions between drugs, and targeting specific proteins. While antibodies are very efficient, their higher production costs impede the process of commercialization. However, their cost factor has been improved by developing biosimilar antibodies as affordable versions of therapeutic antibodies. Along with the recent advancements and innovations in antibody engineering have helped and will furtherly help to design bio-better antibodies with improved efficacy than the conventional ones. These novel mAb-based therapeutics are set to revolutionize existing drug therapies targeting a wide spectrum of diseases, thereby meeting several unmet medical needs. This review provides comprehensive insights into the current fundamental landscape of mAbs development and applications and the key factors influencing the future projections, advancement, and incorporation of such promising immunotherapeutic candidates as a confrontation approach against a wide list of diseases, with a rationalistic mentioning of any limitations facing this field.
Collapse
Affiliation(s)
- Hassan Aboul-Ella
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| | - Asmaa Gohar
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, Suez, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University (ACU), Giza, Egypt
- Egyptian Drug Authority (EDA), Giza, Egypt
| | - Aya Ahmed Ali
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Sinai, Egypt
| | - Lina M Ismail
- Department of Biotechnology and Molecular Chemistry, Faculty of Science, Cairo University, Giza, Egypt
- Creative Egyptian Biotechnologists (CEB), Giza, Egypt
| | | | - Walid F Elkhatib
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, Suez, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Heba Aboul-Ella
- Department of Pharmacognosy, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University (ECU), Cairo, Egypt
- Scientific Research Group in Egypt (SRGE), Cairo, Egypt
| |
Collapse
|
11
|
Li N, Gong N, Duan B, Zhang Y, Jian Y, Xu Y, Liu J, Wang X, Zhang X, Du M, Zhou F, Zhao J, Guan X, Peng X, Wang S, Zhang H, Li X. Reduction of circulating IgE and allergens by a pH-sensitive antibody with enhanced FcγRIIb binding. Mol Ther 2024; 32:3729-3742. [PMID: 39228125 PMCID: PMC11489548 DOI: 10.1016/j.ymthe.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/29/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024] Open
Abstract
Allergen-crosslinked IgE triggers allergy by interacting with its receptor on basophils and mast cells. The anti-IgE monoclonal antibody omalizumab can alleviate allergy by competing with the receptor for IgE binding. However, along with neutralization, omalizumab also inhibits IgE degradation, which is clinically associated with high-dose and total IgE accumulation problems. In this study, we have developed an IgE-eliminating antibody on the basis of omalizumab, which has pH-dependent Fabs and an Fc with high affinity for FcγRIIb. In mice, the antibody rapidly eliminated total serum IgE to baseline levels and caused lower free IgE levels than omalizumab. At low dosages, the antibody also exhibited favorable IgE elimination effects. In addition, the antibody can degrade the corresponding allergen with the removal of IgE, addressing the allergy from its source. Introduction of the M252Y/S254T/T256E (YTE) mutation into this antibody prolongs its serum half-life without reducing potency. Thus, this engineered antibody holds a promising therapeutic option for allergy patients. Mechanistic insights are also included in this study.
Collapse
Affiliation(s)
- Na Li
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Nanxin Gong
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China; College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding 071002, P.R. China
| | - Baoxin Duan
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Yongyan Zhang
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Yi Jian
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Yanqin Xu
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Jinming Liu
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Xiaoqian Wang
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Xiaoqi Zhang
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Mingjuan Du
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, P.R. China
| | - Feilong Zhou
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Jiliang Zhao
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Xiangchen Guan
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Xiangda Peng
- Shanghai Zelixir Biotech, Shanghai 200030, P.R. China
| | - Sheng Wang
- Shanghai Zelixir Biotech, Shanghai 200030, P.R. China
| | - Hongkai Zhang
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China; Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, P.R. China.
| | - Xin Li
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China.
| |
Collapse
|
12
|
Irvine EB, Nikolov A, Khan MZ, Peters JM, Lu R, Sixsmith J, Wallace A, van Woudenbergh E, Shin S, Karpinski W, Hsiao JC, Casadevall A, Bryson BD, Cavacini L, Grace PS, Alter G, Fortune SM. Fc-engineered antibodies promote neutrophil-dependent control of Mycobacterium tuberculosis. Nat Microbiol 2024; 9:2369-2382. [PMID: 39174703 PMCID: PMC11371646 DOI: 10.1038/s41564-024-01777-9] [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: 04/27/2022] [Accepted: 07/09/2024] [Indexed: 08/24/2024]
Abstract
Mounting evidence indicates that antibodies can contribute towards control of tuberculosis (TB). However, the underlying mechanisms of humoral immune protection and whether antibodies can be exploited in therapeutic strategies to combat TB are relatively understudied. Here we engineered the receptor-binding Fc (fragment crystallizable) region of an antibody recognizing the Mycobacterium tuberculosis (Mtb) capsule, to define antibody Fc-mediated mechanism(s) of Mtb restriction. We generated 52 Fc variants that either promote or inhibit specific antibody effector functions, rationally building antibodies with enhanced capacity to promote Mtb restriction in a human whole-blood model of infection. While there is likely no singular Fc profile that universally drives control of Mtb, here we found that several Fc-engineered antibodies drove Mtb restriction in a neutrophil-dependent manner. Single-cell RNA sequencing analysis showed that a restrictive Fc-engineered antibody promoted neutrophil survival and expression of cell-intrinsic antimicrobial programs. These data show the potential of Fc-engineered antibodies as therapeutics able to harness the protective functions of neutrophils to promote control of TB.
Collapse
Affiliation(s)
- Edward B Irvine
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Angel Nikolov
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Mehak Z Khan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Joshua M Peters
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Richard Lu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Jaimie Sixsmith
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Aaron Wallace
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA, USA
| | | | - Sally Shin
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Jeff C Hsiao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bryan D Bryson
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lisa Cavacini
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA, USA
| | - Patricia S Grace
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Division of Infectious Disease, Massachusetts General Hospital, Boston, MA, USA.
| | - Sarah M Fortune
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
13
|
Gil Gonzalez L, Won KD, Tawhidi Z, Cummins E, Cruz-Leal Y, Tundidor Cabado Y, Sachs UJ, Norris PAA, Shan Y, Bhakta V, Li J, Samudio I, Silva-Moreno B, Cerna-Portillo L, Pavon Oro A, Bergqvist P, Chan P, Moorehead A, Sholzberg M, Sheffield WP, Lazarus AH. Human Fc gamma receptor IIIA blockade inhibits platelet destruction in a humanized murine model of ITP. Blood Adv 2024; 8:1869-1879. [PMID: 38330193 PMCID: PMC11007428 DOI: 10.1182/bloodadvances.2023012155] [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: 11/13/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
ABSTRACT Fc gamma receptor (FcγR) IIIA is an important receptor for immunoglobulin G (IgG) and is involved in immune defense mechanisms as well as tissue destruction in some autoimmune diseases including immune thrombocytopenia (ITP). FcγRIIIA on macrophages can trigger phagocytosis of IgG-sensitized platelets, and prior pilot studies observed blockade of FcγRIIIA increased platelet counts in patients with ITP. Unfortunately, although blockade of FcγRIIIA in patients with ITP increased platelet counts, its engagement by the blocking antibody drove serious adverse inflammatory reactions. These adverse events were postulated to originate from the antibody's Fc and/or bivalent nature. The blockade of human FcγRIIIA in vivo with a monovalent construct lacking an active Fc region has not yet been achieved. To effectively block FcγRIIIA in vivo, we developed a high affinity monovalent single-chain variable fragment (scFv) that can bind and block human FcγRIIIA. This scFv (17C02) was expressed in 3 formats: a monovalent fusion protein with albumin, a 1-armed human IgG1 antibody, and a standard bivalent mouse (IgG2a) antibody. Both monovalent formats were effective in preventing phagocytosis of ITP serum-sensitized human platelets. In vivo studies using FcγR-humanized mice demonstrated that both monovalent therapeutics were also able to increase platelet counts. The monovalent albumin fusion protein did not have adverse event activity as assessed by changes in body temperature, whereas the 1-armed antibody induced some changes in body temperature even though the Fc region function was impaired by the Leu234Ala and Leu235Ala mutations. These data demonstrate that monovalent blockade of human FcγRIIIA in vivo can potentially be a therapeutic strategy for patients with ITP.
Collapse
Affiliation(s)
- Lazaro Gil Gonzalez
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Kevin D. Won
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Zoya Tawhidi
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | | | - Yoelys Cruz-Leal
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
| | - Yaima Tundidor Cabado
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Ulrich J. Sachs
- Institute for Clinical Immunology, Transfusion Medicine, and Haemostasis, Justus Liebig University, Giessen, Germany
- Department of Thrombosis and Haemostasis, Giessen University Hospital, Giessen, Germany
| | - Peter A. A. Norris
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Yuexin Shan
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Varsha Bhakta
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
| | - Janessa Li
- adMare BioInnovations, Vancouver, BC, Canada
| | | | | | | | - Alequis Pavon Oro
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | | | | | - Amy Moorehead
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Michelle Sholzberg
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - William P. Sheffield
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
| | - Alan H. Lazarus
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
| |
Collapse
|
14
|
Beckmann K, Reitinger C, Yan X, Carle A, Blümle E, Jurkschat N, Paulmann C, Prassl S, Kazandjian LV, Loré K, Nimmerjahn F, Fischer S. Fcγ-Receptor-Independent Controlled Activation of CD40 Canonical Signaling by Novel Therapeutic Antibodies for Cancer Therapy. Antibodies (Basel) 2024; 13:31. [PMID: 38651411 PMCID: PMC11036229 DOI: 10.3390/antib13020031] [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: 01/19/2024] [Revised: 03/04/2024] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
The activation of CD40-mediated signaling in antigen-presenting cells is a promising therapeutic strategy to promote immune responses against tumors. Most agonistic anti-CD40 antibodies currently in development require the Fcγ-receptor (FcγR)-mediated crosslinking of CD40 molecules for a meaningful activation of CD40 signaling but have limitations due to dose-limiting toxicities. Here we describe the identification of CD40 antibodies which strongly stimulate antigen-presenting cells in an entirely FcγR-independent manner. These Fc-silenced anti-CD40 antibodies induce an efficient upregulation of costimulatory receptors and cytokine release by dendritic cells. Finally, the most active identified anti-CD40 antibody shows activity in humanized mice. More importantly, there are no signs of obvious toxicities. These studies thus demonstrate the potent activation of antigen-presenting cells with anti-CD40 antibodies lacking FcγR-binding activity and open the possibility for an efficacious and safe combination therapy for cancer patients.
Collapse
Affiliation(s)
| | - Carmen Reitinger
- Division of Genetics, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Xianglei Yan
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Visionsgatan 4, BioClinicum J7:30, 171 64, Stockholm, Sweden
- Center of Molecular Medicine, 171 76, Stockholm, Sweden
| | - Anna Carle
- Biontech SE, Forstenrieder Str. 8-14, 82061 Neuried, Germany
| | - Eva Blümle
- Biontech SE, Forstenrieder Str. 8-14, 82061 Neuried, Germany
| | | | | | - Sandra Prassl
- Biontech SE, Forstenrieder Str. 8-14, 82061 Neuried, Germany
| | | | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Visionsgatan 4, BioClinicum J7:30, 171 64, Stockholm, Sweden
- Center of Molecular Medicine, 171 76, Stockholm, Sweden
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, 91058 Erlangen, Germany
- FAU Profile Centre Immunomedicine, 91054 Erlangen, Germany
| | | |
Collapse
|
15
|
Stone CA, Spiller BW, Smith SA. Engineering therapeutic monoclonal antibodies. J Allergy Clin Immunol 2024; 153:539-548. [PMID: 37995859 PMCID: PMC11437839 DOI: 10.1016/j.jaci.2023.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/05/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023]
Abstract
The use of human antibodies as biologic therapeutics has revolutionized patient care throughout fields of medicine. As our understanding of the many roles antibodies play within our natural immune responses continues to advance, so will the number of therapeutic indications for which an mAb will be developed. The great breadth of function, long half-life, and modular structure allow for nearly limitless therapeutic possibilities. Human antibodies can be rationally engineered to enhance their desired immune functions and eliminate those that may result in unwanted effects. Antibody therapeutics now often start with fully human variable regions, either acquired from genetically engineered humanized mice or from the actual human B cells. These variable genes can be further engineered by widely used methods for optimization of their specificity through affinity maturation, random mutagenesis, targeted mutagenesis, and use of in silico approaches. Antibody isotype selection and deliberate mutations are also used to improve efficacy and tolerability by purposeful fine-tuning of their immune effector functions. Finally, improvements directed at binding to the neonatal Fc receptor can endow therapeutic antibodies with unbelievable extensions in their circulating half-life. The future of engineered antibody therapeutics is bright, with the global mAb market projected to exhibit compound annual growth, forecasted to reach a revenue of nearly half a trillion dollars in 2030.
Collapse
Affiliation(s)
- Cosby A Stone
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn
| | - Benjamin W Spiller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tenn; Department of Pharmacology, Vanderbilt University, Nashville, Tenn
| | - Scott A Smith
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tenn.
| |
Collapse
|
16
|
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
|
17
|
Hale G, Davy AD, Wilkinson I. Systematic analysis of Fc mutations designed to enhance binding to Fc-gamma receptors. MAbs 2024; 16:2406539. [PMID: 39306747 PMCID: PMC11418285 DOI: 10.1080/19420862.2024.2406539] [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: 07/31/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/25/2024] Open
Abstract
A critical attribute of therapeutic antibodies is their ability to engage with humoral or cellular effector mechanisms, and this depends on the ability of the Fc region to bind to complement (C1q) or Fc receptors. Investigators have sought to optimize these effects by engineering the Fc region to bind to a greater or lesser extent to individual receptors. Different approaches have been used in the clinic, but they have not been systematically compared. We have now produced a matched set of anti-CD20 antibodies representing a range of variants and compared their activity in cell-based assays for complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity, and antibody-dependent phagocytosis using a range of individual Fc receptors. We have also compared the thermal stability of the variants by differential scanning fluorimetry (DSF). The results reveal a spectrum of activities which may be appropriate for different applications.
Collapse
|
18
|
Heisler J, Kovner D, Izadi S, Zarzar J, Carter PJ. Modulation of the high concentration viscosity of IgG 1 antibodies using clinically validated Fc mutations. MAbs 2024; 16:2379560. [PMID: 39028186 PMCID: PMC11262234 DOI: 10.1080/19420862.2024.2379560] [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: 04/04/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024] Open
Abstract
The self-association of therapeutic antibodies can result in elevated viscosity and create problems in manufacturing and formulation, as well as limit delivery by subcutaneous injection. The high concentration viscosity of some antibodies has been reduced by variable domain mutations or by the addition of formulation excipients. In contrast, the impact of Fc mutations on antibody viscosity has been minimally explored. Here, we studied the effect of a panel of common and clinically validated Fc mutations on the viscosity of two closely related humanized IgG1, κ antibodies, omalizumab (anti-IgE) and trastuzumab (anti-HER2). Data presented here suggest that both Fab-Fab and Fab-Fc interactions contribute to the high viscosity of omalizumab, in a four-contact model of self-association. Most strikingly, the high viscosity of omalizumab (176 cP) was reduced 10.7- and 2.2-fold by Fc modifications for half-life extension (M252Y:S254T:T256E) and aglycosylation (N297G), respectively. Related single mutations (S254T and T256E) each reduced the viscosity of omalizumab by ~6-fold. An alternative half-life extension Fc mutant (M428L:N434S) had the opposite effect in increasing the viscosity of omalizumab by 1.5-fold. The low viscosity of trastuzumab (8.6 cP) was unchanged or increased by ≤ 2-fold by the different Fc variants. Molecular dynamics simulations provided mechanistic insight into the impact of Fc mutations in modulating electrostatic and hydrophobic surface properties as well as conformational stability of the Fc. This study demonstrates that high viscosity of some IgG1 antibodies can be mitigated by Fc mutations, and thereby offers an additional tool to help design future antibody therapeutics potentially suitable for subcutaneous delivery.
Collapse
Affiliation(s)
- Joel Heisler
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA, USA
| | - Daniel Kovner
- Department of Pharmaceutical Development, Genentech, Inc, South San Francisco, CA, USA
| | - Saeed Izadi
- Department of Pharmaceutical Development, Genentech, Inc, South San Francisco, CA, USA
| | - Jonathan Zarzar
- Department of Pharmaceutical Development, Genentech, Inc, South San Francisco, CA, USA
| | - Paul J. Carter
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
19
|
Chen C, Wang Z, Kang M, Lee KB, Ge X. High-fidelity large-diversity monoclonal mammalian cell libraries by cell cycle arrested recombinase-mediated cassette exchange. Nucleic Acids Res 2023; 51:e113. [PMID: 37941133 PMCID: PMC10711435 DOI: 10.1093/nar/gkad1001] [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: 02/08/2023] [Revised: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 11/10/2023] Open
Abstract
Mammalian cells carrying defined genetic variations have shown great potentials in both fundamental research and therapeutic development. However, their full use was limited by lack of a robust method to construct large monoclonal high-quality combinatorial libraries. This study developed cell cycle arrested recombinase-mediated cassette exchange (aRMCE), able to provide monoclonality, precise genomic integration and uniform transgene expression. Via optimized nocodazole-mediated mitotic arrest, 20% target gene replacement efficiency was achieved without antibiotic selection, and the improved aRMCE efficiency was applicable to a variety of tested cell clones, transgene targets and transfection methods. As a demonstration of this versatile method, we performed directed evolution of fragment crystallizable (Fc), for which error-prone libraries of over 107 variants were constructed and displayed as IgG on surface of CHO cells. Diversities of constructed libraries were validated by deep sequencing, and panels of novel Fc mutants were identified showing improved binding towards specific Fc gamma receptors and enhanced effector functions. Due to its large cargo capacity and compatibility with different mutagenesis approaches, we expect this mammalian cell platform technology has broad applications for directed evolution, multiplex genetic assays, cell line development and stem cell engineering.
Collapse
Affiliation(s)
- Chuan Chen
- Department of Chemical and Environmental Engineering, University of California Riverside, Riverside, CA 92521, USA
| | - Zening Wang
- Department of Chemical and Environmental Engineering, University of California Riverside, Riverside, CA 92521, USA
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Minhyo Kang
- Department of Chemical and Environmental Engineering, University of California Riverside, Riverside, CA 92521, USA
| | - Ki Baek Lee
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xin Ge
- Department of Chemical and Environmental Engineering, University of California Riverside, Riverside, CA 92521, USA
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
20
|
Merrill JT, Guthridge J, Smith M, June J, Koumpouras F, Machua W, Askanase A, Khosroshahi A, Sheikh SZ, Rathi G, Burington B, Foster P, Matijevic M, Arora S, Wang X, Gao M, Wax S, James JA, Zack DJ. Obexelimab in Systemic Lupus Erythematosus With Exploration of Response Based on Gene Pathway Co-Expression Patterns: A Double-Blind, Randomized, Placebo-Controlled, Phase 2 Trial. Arthritis Rheumatol 2023; 75:2185-2194. [PMID: 37459248 DOI: 10.1002/art.42652] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/17/2023] [Accepted: 07/11/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVE Obexelimab is an investigational, bifunctional, noncytolytic monoclonal antibody that binds CD19 and FcyRIIb to inhibit B cells, plasmablasts, and plasma cells. This trial evaluated the efficacy and safety of obexelimab in the treatment of patients with systemic lupus erythematosus (SLE). METHODS During screening, patients with active, non-organ-threatening SLE received corticosteroid injections to ameliorate symptoms while immunosuppressants were withdrawn (≤10 mg/day prednisone equivalent and ≤400 mg/day hydroxychloroquine allowed). Patients with improved disease activity were randomized 1:1 to obexelimab 5 mg/kg intravenously or placebo once every 2 weeks until week 32 or loss of improvement (LOI). RESULTS In this study, 104 patients were randomized. Analysis of the primary endpoint, proportion of patients reaching week 32 without LOI, used an efficacy-evaluable (EE) population defined as patients who completed the study or withdrew for flare or treatment-related toxicity. This endpoint did not reach statistical significance: 21 of 50 obexelimab-treated patients (42.0%) versus 12 of 42 patients (28.6%) treated with a placebo (P = 0.183). Time to LOI was increased in obexelimab-treated patients versus patients treated with a placebo in the EE (hazard ratio [HR] 0.53, P = 0.025) and intention-to-treat (HR 0.59, P = 0.062) populations. In obexelimab-treated patients, B cells decreased approximately 50%, and trough concentration and inclusion in baseline gene expression clusters with high B cell pathway modules were associated with increased time to LOI. Obexelimab was associated with infusion reactions but was generally safe and well-tolerated. CONCLUSION Although the primary endpoint was not reached, secondary analysis showed time to LOI was significantly increased in obexelimab-treated patients, and analysis of patient subsets defined by gene expression patterns at baseline suggests a responding subpopulation.
Collapse
Affiliation(s)
- Joan T Merrill
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Joel Guthridge
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Miles Smith
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Joshua June
- Great Lakes Center of Rheumatology, Lansing, Michigan
| | | | | | - Anca Askanase
- Columbia University Medical Center, New York City, New York
| | | | - Saira Z Sheikh
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | | | | - Judith A James
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | | |
Collapse
|
21
|
Singh R, Chandley P, Rohatgi S. Recent Advances in the Development of Monoclonal Antibodies and Next-Generation Antibodies. Immunohorizons 2023; 7:886-897. [PMID: 38149884 PMCID: PMC10759153 DOI: 10.4049/immunohorizons.2300102] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/07/2023] [Indexed: 12/28/2023] Open
Abstract
mAbs are highly indispensable tools for diagnostic, prophylactic, and therapeutic applications. The first technique, hybridoma technology, was based on fusion of B lymphocytes with myeloma cells, which resulted in generation of single mAbs against a specific Ag. Along with hybridoma technology, several novel and alternative methods have been developed to improve mAb generation, ranging from electrofusion to the discovery of completely novel technologies such as B cell immortalization; phage, yeast, bacterial, ribosome, and mammalian display systems; DNA/RNA encoded Abs; single B cell technology; transgenic animals; and artificial intelligence/machine learning. This commentary outlines the evolution, methodology, advantages, and limitations of various mAb production techniques. Furthermore, with the advent of next-generation Ab technologies such as single-chain variable fragments, nanobodies, bispecific Abs, Fc-engineered Abs, Ab biosimilars, Ab mimetics, and Ab-drug conjugates, the healthcare and pharmaceutical sectors have become resourceful to develop highly specific mAb treatments against various diseases such as cancer and autoimmune and infectious diseases.
Collapse
Affiliation(s)
- Rohit Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, India
| | - Pankaj Chandley
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, India
| | - Soma Rohatgi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, India
| |
Collapse
|
22
|
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
|
23
|
Beneduce C, Nguyen S, Washburn N, Schaeck J, Meccariello R, Holte K, Ortiz D, Manning AM, Bosques CJ, Kurtagic E. Inhibitory Fc-Gamma IIb Receptor Signaling Induced by Multivalent IgG-Fc Is Dependent on Sialylation. Cells 2023; 12:2130. [PMID: 37681862 PMCID: PMC10486564 DOI: 10.3390/cells12172130] [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: 07/11/2023] [Revised: 08/05/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023] Open
Abstract
Immunoglobulin (IgG) Fc glycosylation has been shown to be important for the biological activity of antibodies. Fc sialylation is important for the anti-inflammatory activity of IgGs. However, evaluating the structure-activity relationship (SAR) of antibody Fc glycosylation has been hindered using simplified in vitro models in which antibodies are often displayed in monomeric forms. Presenting antibodies in monomeric forms may not accurately replicate the natural environment of the antibodies when binding their antigen in vivo. To address these limitations, we used different Fc-containing molecules, displaying their Fc domains in monovalent and multivalent fashion. Given the inhibitory role of Fc gamma receptor IIb (FcγRIIb) in autoimmune and inflammatory diseases, we focused on evaluating the impact of Fc sialylation on the activation of FcγRIIb. We report for the first time that in human cellular systems, sialic acid mediates the induction of FcγRIIb phosphorylation by IgG-Fc when the IgG-Fc is displayed in a multivalent fashion. This effect was observed with different types of therapeutic agents such as sialylated anti-TNFα antibodies, sialylated IVIg and sialylated recombinant multivalent Fc products. These studies represent the first report of the specific effects of Fc sialylation on FcγRIIb signaling on human immune cells and may help in the characterization of the anti-inflammatory activity of Fc-containing therapeutic candidates.
Collapse
Affiliation(s)
- Christopher Beneduce
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| | | | - Nathaniel Washburn
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| | - John Schaeck
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | - Robin Meccariello
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| | | | - Daniel Ortiz
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | | | | | - Elma Kurtagic
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| |
Collapse
|
24
|
Johnson SN, Brucks SD, Apley KD, Farrell MP, Berkland CJ. Multivalent Scaffolds to Promote B cell Tolerance. Mol Pharm 2023; 20:3741-3756. [PMID: 37410969 DOI: 10.1021/acs.molpharmaceut.3c00039] [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: 07/08/2023]
Abstract
Autoimmune diseases are characterized by aberrant immune responses toward self-antigens. Current treatments lack specificity, promoting adverse effects by broadly suppressing the immune system. Therapies that specifically target the immune cells responsible for disease are a compelling strategy to mitigate adverse effects. Multivalent formats that display numerous binding epitopes off a single scaffold may enable selective immunomodulation by eliciting signals through pathways unique to the targeted immune cells. However, the architecture of multivalent immunotherapies can vary widely, and there is limited clinical data with which to evaluate their efficacy. Here, we set forth to review the architectural properties and functional mechanisms afforded by multivalent ligands and evaluate four multivalent scaffolds that address autoimmunity by altering B cell signaling pathways. First, we address both synthetic and natural polymer backbones functionalized with a variety of small molecule, peptide, and protein ligands for probing the effects of valency and costimulation. Then, we review nanoparticles composed entirely from immune signals which have been shown to be efficacious. Lastly, we outline multivalent liposomal nanoparticles capable of displaying high numbers of protein antigens. Taken together, these examples highlight the versatility and desirability of multivalent ligands for immunomodulation and illuminate strengths and weaknesses of multivalent scaffolds for treating autoimmunity.
Collapse
Affiliation(s)
- Stephanie N Johnson
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Spencer D Brucks
- Department of Chemistry, Harvey Mudd College, Claremont, California 91711, United States
| | - Kyle D Apley
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Mark P Farrell
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Cory J Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, United States
- Bioengineering Program, University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
25
|
Perugino CA, Wallace ZS, Zack DJ, Quinn SM, Poma A, Fernandes AD, Foster P, DeMattos S, Burington B, Liu H, Allard-Chamard H, Smith N, Kai X, Xing K, Pillai S, Stone JH. Evaluation of the safety, efficacy, and mechanism of action of obexelimab for the treatment of patients with IgG4-related disease: an open-label, single-arm, single centre, phase 2 pilot trial. THE LANCET. RHEUMATOLOGY 2023; 5:e442-e450. [PMID: 38251576 DOI: 10.1016/s2665-9913(23)00157-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Obexelimab is a bifunctional, non-cytolytic, humanised monoclonal antibody that binds CD19 and Fc gamma receptor IIb to inhibit B cells, plasmablasts, and CD19-expressing plasma cells. We aimed to evaluate the safety, clinical efficacy, and pharmacodynamic effects of obexelimab in patients with active IgG4-related disease. METHODS We conducted an open-label, single-arm, single centre, phase 2 pilot trial at the Massachusetts General Hospital in Boston, MA, USA. Eligible patients were aged 18-80 years and had active IgG4-related disease confirmed by an IgG4-related disease responder index score of 3 or more. Patients received 5 mg/kg of obexelimab intravenously every 2 weeks for 24 weeks. Patients on glucocorticoids at baseline were expected to discontinue usage within 2 months following enrolment. The primary endpoint was the proportion of patients with a decrease of 2 or more from baseline in the IgG4-related disease responder index at day 169 (ie, primary responders). Patients who achieved a decrease of 2 or more at any visit were designated as responders. Adverse events were graded on a scale of 1-5 (ie, mild, moderate, severe, life-threatening, or death) according to the Common Terminology Criteria for Adverse Events grading scale (version 4.3). Exploratory analyses were quantification of B-cell CD19 receptor occupancy, plasmablast, total B-cell and CD4+ cytotoxic T-cell count by flow cytometry, and immunoglobulin concentrations by nephelometry. This study is registered with ClinicalTrials.gov, NCT02725476. FINDINGS Between Feb 24, 2016, and Dec 21, 2016, we enrolled 15 patients. The median age was 63 years (IQR 52-65). Ten (67%) of 15 patients were male, five (33%) were female, and 12 (80%) were White. At baseline, 12 (80%) of 15 patients had an elevated median serum IgG4 concentration of 220 mg/dL (IQR 124-441), and the median IgG4-related disease responder index score was 12 (IQR 7-13). 12 (80%) of 15 patients achieved the primary endpoint (ie, primary responders), 14 (93%) were defined as responders. Reductions from baseline in serum B cells and plasmablasts were observed following treatment with obexelimab. However, in most patients with follow-up data, serum B cells recovered to 75% of baseline concentrations within 42 days of the final obexelimab dose. 13 (87%) of 15 patients reported adverse events, one of which (an infusion reaction) resulted in treatment discontinuation. INTERPRETATION All patients except for one had clinical responses to obexelimab treatment. Both reductions in circulating B cells without evidence of apoptosis during obexelimab treatment and their rapid rebound after treatment discontinuation suggest that obexelimab might lead to B-cell sequestration in lymphoid organs or the bone marrow. These results support the continued development of obexelimab for the treatment of IgG4-related disease. FUNDING Xencor, Zenas BioPharma, National Institute of Arthritis and Musculoskeletal and Skin Diseases, and National Institute of Allergy and Infectious Diseases.
Collapse
Affiliation(s)
- Cory A Perugino
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA, USA
| | - Zachary S Wallace
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Allen Poma
- Clinical Development, Zenas BioPharma, Waltham, MA, USA
| | - Ana D Fernandes
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Hang Liu
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA, USA
| | - Hugues Allard-Chamard
- Division of Rheumatology, Faculté de médecine et des sciences de la santé de l'Université de Sherbrooke et Centre de Recherche Clinique Étienne-Le Bel, Sherbrooke, QC, Canada
| | - Nathan Smith
- Penn State College of Medicine, Hershey, PA, USA
| | - Xin Kai
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA, USA
| | - Kelly Xing
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA, USA
| | - Shiv Pillai
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA, USA
| | - John H Stone
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Boyles JS, Sadowski D, Potter S, Vukojicic A, Parker J, Chang WY, Ma YL, Chambers MG, Nelson J, Barmettler B, Smith EM, Kersjes K, Himes ER, Lin C, Lucchesi J, Brahmbhatt J, Sina R, Martin JA, Maestri E, Wiethoff CM, Dyas GL, Linnik MD, Na S, Witcher DR, Budelsky A, Rubtsova K. A nondepleting anti-CD19 antibody impairs B cell function and inhibits autoimmune diseases. JCI Insight 2023; 8:e166137. [PMID: 37427592 PMCID: PMC10371335 DOI: 10.1172/jci.insight.166137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/19/2023] [Indexed: 07/11/2023] Open
Abstract
B cells contribute to multiple aspects of autoimmune disorders, and B cell-targeting therapies, including B cell depletion, have been proven to be efficacious in treatment of multiple autoimmune diseases. However, the development of novel therapies targeting B cells with higher efficacy and a nondepleting mechanism of action is highly desirable. Here we describe a nondepleting, high-affinity anti-human CD19 antibody LY3541860 that exhibits potent B cell inhibitory activities. LY3541860 inhibits B cell activation, proliferation, and differentiation of primary human B cells with high potency. LY3541860 also inhibits human B cell activities in vivo in humanized mice. Similarly, our potent anti-mCD19 antibody also demonstrates improved efficacy over CD20 B cell depletion therapy in multiple B cell-dependent autoimmune disease models. Our data indicate that anti-CD19 antibody is a highly potent B cell inhibitor that may have potential to demonstrate improved efficacy over currently available B cell-targeting therapies in treatment of autoimmune conditions without causing B cell depletion.
Collapse
Affiliation(s)
- Jeffrey S. Boyles
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Dorota Sadowski
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Scott Potter
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Aleksandra Vukojicic
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - James Parker
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - William Y. Chang
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Yanfei L. Ma
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Mark G. Chambers
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - James Nelson
- Biotechnology Discovery Research, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Barbra Barmettler
- Biotechnology Discovery Research, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Eric M. Smith
- Biotechnology Discovery Research, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Kara Kersjes
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Evan R. Himes
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Chaohua Lin
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jonathan Lucchesi
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jaladhi Brahmbhatt
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Ramtin Sina
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jennifer A. Martin
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Evan Maestri
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Christopher M. Wiethoff
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Gregory L. Dyas
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Matthew D. Linnik
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Songqing Na
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Derrick R. Witcher
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Alison Budelsky
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Kira Rubtsova
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| |
Collapse
|
27
|
Kibler A, Seifert M, Budeus B. Age-related changes of the human splenic marginal zone B cell compartment. Immunol Lett 2023; 256-257:59-65. [PMID: 37044264 DOI: 10.1016/j.imlet.2023.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/24/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023]
Abstract
In this review, we will summarize the growing body of knowledge on the age-related changes of human splenic B cell composition and molecular evidence of immune maturation and discuss the contribution of these changes on splenic protective function. From birth on, the splenic marginal zone (sMZ) contains a specialized B cell subpopulation, which recruits and archives memory B cells from immune responses throughout the organism. The quality of sMZ B cell responses is augmented by germinal center (GC)-dependent maturation of memory B cells during childhood, however, in old age, these mechanisms likely contribute to waning of splenic protective function.
Collapse
Affiliation(s)
- Artur Kibler
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| | - Marc Seifert
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany; Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, Düsseldorf, Germany.
| | - Bettina Budeus
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
28
|
Boudreau CM, Burke JS, Roederer AL, Gorman MJ, Mundle S, Lingwood D, Delagrave S, Sridhar S, Ross TM, Kleanthous H, Alter G. Pre-existing Fc profiles shape the evolution of neutralizing antibody breadth following influenza vaccination. Cell Rep Med 2023; 4:100975. [PMID: 36921600 PMCID: PMC10040413 DOI: 10.1016/j.xcrm.2023.100975] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 12/08/2022] [Accepted: 02/19/2023] [Indexed: 03/16/2023]
Abstract
Under the ever-present threat of a pandemic influenza strain, the evolution of a broadly reactive, neutralizing, functional, humoral immune response may hold the key to protection against both circulating and emerging influenza strains. We apply a systems approach to profile hemagglutinin- and neuraminidase-specific humoral signatures that track with the evolution of broad immunity in a cohort of vaccinated individuals and validate these findings in a second longitudinal cohort. Multivariate analysis reveals the presence of a unique pre-existing Fcγ-receptor-binding antibody profile in individuals that evolved broadly reactive hemagglutination inhibition activity (HAI), marked by the presence of elevated levels of pre-existing FCGR2B-binding antibodies. Moreover, vaccination with FCGR2B-binding antibody-opsonized influenza results in enhanced antibody titers and HAI activity in a murine model. Together, these data suggest that pre-existing FCGR2B binding antibodies are a key correlate of the evolution of broadly protective influenza-specific antibodies, providing insight for the design of next-generation influenza vaccines.
Collapse
Affiliation(s)
- Carolyn M Boudreau
- PhD Program in Virology, Division of Medical Sciences, Harvard University, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - John S Burke
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Alexander L Roederer
- PhD Program in Virology, Division of Medical Sciences, Harvard University, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Matthew J Gorman
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Sophia Mundle
- Discovery North America, Sanofi-Pasteur, Inc., Cambridge, MA 02139, USA
| | - Daniel Lingwood
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | - Saranya Sridhar
- Discovery North America, Sanofi-Pasteur, Inc., Cambridge, MA 02139, USA
| | - Ted M Ross
- University of Georgia, Athens, GA 30602, USA
| | | | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
29
|
Iwata Y, Katada H, Okuda M, Doi Y, Ching TJ, Harada A, Takeiri A, Honda M, Mishima M. Preclinical in vitro evaluation of immune suppression induced by GYM329, Fc-engineered sweeping antibody. J Toxicol Sci 2023; 48:399-409. [PMID: 37394653 DOI: 10.2131/jts.48.399] [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: 07/04/2023]
Abstract
Fc-engineering is commonly used to improve the therapeutic potency of antibody (Ab) treatments. Because FcγRIIb is the only inhibitory FcγR that contains an immunoreceptor tyrosine-based inhibition motif (ITIM), Fc-engineered Abs with enhanced binding affinity to FcγRIIb might provide immune suppression in clinical contexts. GYM329 is an anti-latent myostatin Fc-engineered Ab with increased affinity to FcγRIIb which is expected to improve muscle strength in patients with muscular disorders. Cross-linking of FcγRIIb by immune complex (IC) results in phosphorylation of ITIM to inhibit immune activation and apoptosis in B cells. We examined whether the IC of Fc-engineered Abs with enhanced binding affinity to FcγRIIb causes phosphorylation of ITIM or B cell apoptosis using GYM329 and its Fc variant Abs in human and cynomolgus-monkey (cyno) immune cells in vitro. IC of GYM329 with enhanced binding affinity to human FcγRIIb (×5) induced neither ITIM phosphorylation nor B cell apoptosis. As for GYM329, FcγRIIb should work as an endocytic receptor of small IC to sweep latent myostatin, so it is preferable that GYM329 induces neither ITIM phosphorylation nor B cell apoptosis to prevent immune suppression. In contrast, IC of myo-HuCy2b, the Ab with enhanced binding affinity to human FcγRIIb (×4), induced ITIM phosphorylation and B cell apoptosis. The result of the present study demonstrated that Fc-engineered Abs with similar binding affinity to FcγRIIb had different effects. Thus, it is important to also investigate FcγR-mediated immune functions other than binding to fully understand the biological effects of Fc-engineered Abs.
Collapse
Affiliation(s)
- Yoshika Iwata
- Translational Research Division, Chugai Pharmaceutical Co., Ltd
| | | | | | - Yoshiaki Doi
- Research Division, Chugai Pharmaceutical Co., Ltd
| | | | - Asako Harada
- Translational Research Division, Chugai Pharmaceutical Co., Ltd
| | - Akira Takeiri
- Translational Research Division, Chugai Pharmaceutical Co., Ltd
| | - Masaki Honda
- Translational Research Division, Chugai Pharmaceutical Co., Ltd
| | | |
Collapse
|
30
|
Lefranc MP, Lefranc G. IMGT ® Nomenclature of Engineered IGHG Variants Involved in Antibody Effector Properties and Formats. Antibodies (Basel) 2022; 11:65. [PMID: 36278618 PMCID: PMC9624366 DOI: 10.3390/antib11040065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
The constant region of the immunoglobulin (IG) or antibody heavy gamma chain is frequently engineered to modify the effector properties of the therapeutic monoclonal antibodies. These variants are classified in regards to their effects on effector functions, antibody-dependent cytotoxicity (ADCC), antibody-dependent phagocytosis (ADCP), complement-dependent cytotoxicity (CDC) enhancement or reduction, B cell inhibition by the coengagement of antigen and FcγR on the same cell, on half-life increase, and/or on structure such as prevention of IgG4 half-IG exchange, hexamerisation, knobs-into-holes and the heteropairing H-H of bispecific antibodies, absence of disulfide bridge inter H-L, absence of glycosylation site, and site-specific drug attachment engineered cysteine. The IMGT engineered variant identifier is comprised of the species and gene name (and eventually allele), the letter 'v' followed by a number (assigned chronologically), and for each concerned domain (e.g, CH1, h, CH2 and CH3), the novel AA (single letter abbreviation) and IMGT position according to the IMGT unique numbering for the C-domain and between parentheses, the Eu numbering. IMGT engineered variants are described with detailed amino acid changes, visualized in motifs based on the IMGT numbering bridging genes, sequences, and structures for higher order description.
Collapse
Affiliation(s)
- Marie-Paule Lefranc
- IMGT®, The International ImMunoGeneTics Information System®, Laboratoire d’ImmunoGénétique Moléculaire (LIGM), Institut de Génétique Humaine (IGH), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier (UM), UMR 9002 CNRS-UM, CEDEX 5, 34396 Montpellier, France
| | - Gérard Lefranc
- IMGT®, The International ImMunoGeneTics Information System®, Laboratoire d’ImmunoGénétique Moléculaire (LIGM), Institut de Génétique Humaine (IGH), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier (UM), UMR 9002 CNRS-UM, CEDEX 5, 34396 Montpellier, France
| |
Collapse
|
31
|
Yin Y, Romei MG, Sankar K, Pal LR, Hon Hoi K, Yang Y, Leonard B, De Leon Boenig G, Kumar N, Matsumoto M, Payandeh J, Harris SF, Moult J, Lazar GA. Antibody Interfaces Revealed Through Structural Mining. Comput Struct Biotechnol J 2022; 20:4952-4968. [PMID: 36147680 PMCID: PMC9474289 DOI: 10.1016/j.csbj.2022.08.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/15/2022] Open
Abstract
Antibodies are fundamental effectors of humoral immunity, and have become a highly successful class of therapeutics. There is increasing evidence that antibodies utilize transient homotypic interactions to enhance function, and elucidation of such interactions can provide insights into their biology and new opportunities for their optimization as drugs. Yet the transitory nature of weak interactions makes them difficult to investigate. Capitalizing on their rich structural data and high conservation, we have characterized all the ways that antibody fragment antigen-binding (Fab) regions interact crystallographically. This approach led to the discovery of previously unrealized interfaces between antibodies. While diverse interactions exist, β-sheet dimers and variable-constant elbow dimers are recurrent motifs. Disulfide engineering enabled interactions to be trapped and investigated structurally and functionally, providing experimental validation of the interfaces and illustrating their potential for optimization. This work provides first insight into previously undiscovered oligomeric interactions between antibodies, and enables new opportunities for their biotherapeutic optimization.
Collapse
|
32
|
Liu L, Wu Y, Ye K, Cai M, Zhuang G, Wang J. Antibody-Targeted TNFRSF Activation for Cancer Immunotherapy: The Role of FcγRIIB Cross-Linking. Front Pharmacol 2022; 13:924197. [PMID: 35865955 PMCID: PMC9295861 DOI: 10.3389/fphar.2022.924197] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/30/2022] [Indexed: 12/19/2022] Open
Abstract
Co-stimulation signaling in various types of immune cells modulates immune responses in physiology and disease. Tumor necrosis factor receptor superfamily (TNFRSF) members such as CD40, OX40 and CD137/4-1BB are expressed on myeloid cells and/or lymphocytes, and they regulate antigen presentation and adaptive immune activities. TNFRSF agonistic antibodies have been evaluated extensively in preclinical models, and the robust antitumor immune responses and efficacy have encouraged continued clinical investigations for the last two decades. However, balancing the toxicities and efficacy of TNFRSF agonistic antibodies remains a major challenge in the clinical development. Insights into the co-stimulation signaling biology, antibody structural roles and their functionality in immuno-oncology are guiding new advancement of this field. Leveraging the interactions between antibodies and the inhibitory Fc receptor FcγRIIB to optimize co-stimulation agonistic activities dependent on FcγRIIB cross-linking selectively in tumor microenvironment represents the current frontier, which also includes cross-linking through tumor antigen binding with bispecific antibodies. In this review, we will summarize the immunological roles of TNFRSF members and current clinical studies of TNFRSF agonistic antibodies. We will also cover the contribution of different IgG structure domains to these agonistic activities, with a focus on the role of FcγRIIB in TNFRSF cross-linking and clustering bridged by agonistic antibodies. We will review and discuss several Fc-engineering approaches to optimize Fc binding ability to FcγRIIB in the context of proper Fab and the epitope, including a cross-linking antibody (xLinkAb) model and its application in developing TNFRSF agonistic antibodies with improved efficacy and safety for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Yi Wu
- Lyvgen Biopharma, Shanghai, China
| | - Kaiyan Ye
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meichun Cai
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglei Zhuang
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | |
Collapse
|
33
|
Oostindie SC, Lazar GA, Schuurman J, Parren PWHI. Avidity in antibody effector functions and biotherapeutic drug design. Nat Rev Drug Discov 2022; 21:715-735. [PMID: 35790857 PMCID: PMC9255845 DOI: 10.1038/s41573-022-00501-8] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 12/16/2022]
Abstract
Antibodies are the cardinal effector molecules of the immune system and are being leveraged with enormous success as biotherapeutic drugs. A key part of the adaptive immune response is the production of an epitope-diverse, polyclonal antibody mixture that is capable of neutralizing invading pathogens or disease-causing molecules through binding interference and by mediating humoral and cellular effector functions. Avidity - the accumulated binding strength derived from the affinities of multiple individual non-covalent interactions - is fundamental to virtually all aspects of antibody biology, including antibody-antigen binding, clonal selection and effector functions. The manipulation of antibody avidity has since emerged as an important design principle for enhancing or engineering novel properties in antibody biotherapeutics. In this Review, we describe the multiple levels of avidity interactions that trigger the overall efficacy and control of functional responses in both natural antibody biology and their therapeutic applications. Within this framework, we comprehensively review therapeutic antibody mechanisms of action, with particular emphasis on engineered optimizations and platforms. Overall, we describe how affinity and avidity tuning of engineered antibody formats are enabling a new wave of differentiated antibody drugs with tailored properties and novel functions, promising improved treatment options for a wide variety of diseases.
Collapse
Affiliation(s)
- Simone C Oostindie
- Genmab, Utrecht, Netherlands.,Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Greg A Lazar
- Department of Antibody Engineering, Genentech, San Francisco, CA, USA
| | | | - Paul W H I Parren
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands. .,Sparring Bioconsult, Odijk, Netherlands. .,Lava Therapeutics, Utrecht, Netherlands.
| |
Collapse
|
34
|
Advances of research of Fc-fusion protein that activate NK cells for tumor immunotherapy. Int Immunopharmacol 2022; 109:108783. [PMID: 35561479 DOI: 10.1016/j.intimp.2022.108783] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/02/2022] [Accepted: 04/14/2022] [Indexed: 12/21/2022]
Abstract
The rapid development of bioengineering technology has introduced Fc-fusion proteins, representing a novel kind of recombinant protein, as promising biopharmaceutical products in tumor therapy. Numerous related anti-tumor Fc-fusion proteins have been investigated and are in different stages of development. Fc-fusion proteins are constructed by fusing the Fc-region of the antibody with functional proteins or peptides. They retain the bioactivity of the latter and partial properties of the former. This structural and functional advantage makes Fc-fusion proteins an effective tool in tumor immunotherapy, especially for the recruitment and activation of natural killer (NK) cells, which play a critical role in tumor immunotherapy. Even though tumor cells have developed mechanisms to circumvent the cytotoxic effect of NK cells or induce defective NK cells, Fc-fusion proteins have been proven to effectively activate NK cells to kill tumor cells in different ways, such as antibody-dependent cell-mediated cytotoxicity (ADCC), activate NK cells in different ways in order to promote killing of tumor cells. In this review, we focus on NK cell-based immunity for cancers and current research progress of the Fc-fusion proteins for anti-tumor therapy by activating NK cells.
Collapse
|
35
|
Delidakis G, Kim JE, George K, Georgiou G. Improving Antibody Therapeutics by Manipulating the Fc Domain: Immunological and Structural Considerations. Annu Rev Biomed Eng 2022; 24:249-274. [PMID: 35363537 PMCID: PMC9648538 DOI: 10.1146/annurev-bioeng-082721-024500] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interactions between the crystallizable fragment (Fc) domain of antibodies and a plethora of cellular Fc receptors (FcRs) or soluble proteins form a critical link between humoral and innate immunity. In particular, the immunoglobulin G Fc domain is critical for the clearance of target cells by processes that include (a) cytotoxicity, phagocytosis, or complement lysis; (b) modulation of inflammation; (c) antigen presentation; (d) antibody-mediated receptor clustering; and (e) cytokine release. More than 30 Fc-engineered antibodies aimed primarily at tailoring these effects for optimal therapeutic outcomes are in clinical evaluation or have already been approved. Nonetheless, our understanding of how FcR engagement impacts various immune cell phenotypes is still largely incomplete. Recent insights into FcR biology coupled with advances in Fc:FcR structural analysis, Fc engineering, and mouse models that recapitulate human biology are helping to fill in existing knowledge gaps. These advances will provide a blueprint on how to fine-tune the Fc domain to achieve optimal therapeutic efficacy. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA;
| | - Jin Eyun Kim
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Katia George
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA; .,Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA.,Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
36
|
Heckel F, Turaj AH, Fisher H, Chan HTC, Marshall MJE, Dadas O, Penfold CA, Inzhelevskaya T, Mockridge CI, Alvarado D, Tews I, Keler T, Beers SA, Cragg MS, Lim SH. Agonistic CD27 antibody potency is determined by epitope-dependent receptor clustering augmented through Fc-engineering. Commun Biol 2022; 5:229. [PMID: 35288635 PMCID: PMC8921514 DOI: 10.1038/s42003-022-03182-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Agonistic CD27 monoclonal antibodies (mAb) have demonstrated impressive anti-tumour efficacy in multiple preclinical models but modest clinical responses. This might reflect current reagents delivering suboptimal CD27 agonism. Here, using a novel panel of CD27 mAb including a clinical candidate, we investigate the determinants of CD27 mAb agonism. Epitope mapping and in silico docking analysis show that mAb binding to membrane-distal and external-facing residues are stronger agonists. However, poor epitope-dependent agonism could partially be overcome by Fc-engineering, using mAb isotypes that promote receptor clustering, such as human immunoglobulin G1 (hIgG1, h1) with enhanced affinity to Fc gamma receptor (FcγR) IIb, or hIgG2 (h2). This study provides the critical knowledge required for the development of agonistic CD27 mAb that are potentially more clinically efficacious.
Collapse
Affiliation(s)
- Franziska Heckel
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Anna H Turaj
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Hayden Fisher
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton, SO17 1BJ, UK
- Biological Sciences, University of Southampton, Highfield Campus, Southampton, SO17 1BJ, UK
| | - H T Claude Chan
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Michael J E Marshall
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Osman Dadas
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Christine A Penfold
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Tatyana Inzhelevskaya
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - C Ian Mockridge
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | | | - Ivo Tews
- Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton, SO17 1BJ, UK
- Biological Sciences, University of Southampton, Highfield Campus, Southampton, SO17 1BJ, UK
| | - Tibor Keler
- Celldex Therapeutics, Inc., Hampton, NJ, 08827, USA
| | - Stephen A Beers
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Mark S Cragg
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Sean H Lim
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK.
- Cancer Research UK Research Centre, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK.
| |
Collapse
|
37
|
Kamat V, Boutot C, Rafique A, Granados C, Wang J, Badithe A, Torres M, Chatterjee I, Olsen O, Olson W, Huang T. High affinity human Fc specific monoclonal antibodies for capture kinetic analyses of antibody-antigen interactions. Anal Biochem 2022; 640:114455. [PMID: 34788604 DOI: 10.1016/j.ab.2021.114455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022]
Abstract
We recently demonstrated that capturing human monoclonal antibodies (hmAbs) using high affinity anti-human Fc (AHC) antibodies allows reliable characterization of antibody-antigen interactions. Here, we characterized six human Fc specific mouse monoclonal antibodies (mAbs) and compared their binding profiles with three previously characterized goat AHC polyclonal antibodies (pAbs), exhibiting properties of a good capture reagent. All six mouse AHC mAbs specifically bound with high affinity to the Fc region of hIgG1, hIgG2, hIgG4 and to 43 different hIgG variants, containing substitutions and/or mutations in the hinge and/or Fc region, that have been reported to exhibit modified antibody effector function and/or pharmacokinetics. Biacore sensor surfaces individually derivatized with mouse AHC mAbs exhibited >2.5-fold higher hIgG binding capacity compared to the three goat AHC pAb surfaces and reproducibly captured hIgG over 300 capture-regeneration cycles. The results of the capture kinetic analyses performed on 31 antibody-antigen interactions using surfaces derivatized with either of the two highest affinity AHC mAbs (REGN7942 or REGN7943) were in concordance with those performed using goat AHC pAb surfaces. Our data demonstrate that AHC mAbs such as REGN7942 and REGN7943 that have properties superior than the three goat AHC pAbs are highly valuable research reagents, especially to perform capture kinetic analyses of antibody-antigen interactions on optical biosensors.
Collapse
Affiliation(s)
- Vishal Kamat
- Therapeutic Proteins, Regeneron Pharmaceuticals, USA.
| | | | | | | | - Jing Wang
- Therapeutic Proteins, Regeneron Pharmaceuticals, USA
| | - Ashok Badithe
- Therapeutic Proteins, Regeneron Pharmaceuticals, USA
| | | | | | - Olav Olsen
- Therapeutic Proteins, Regeneron Pharmaceuticals, USA
| | - William Olson
- Therapeutic Proteins, Regeneron Pharmaceuticals, USA
| | - Tammy Huang
- Therapeutic Proteins, Regeneron Pharmaceuticals, USA
| |
Collapse
|
38
|
Chen J, Liao S, Zhou H, Yang L, Guo F, Chen S, Li A, Pan Q, Yang C, Liu HF, Pan Q. Humanized Mouse Models of Systemic Lupus Erythematosus: Opportunities and Challenges. Front Immunol 2022; 12:816956. [PMID: 35116040 PMCID: PMC8804209 DOI: 10.3389/fimmu.2021.816956] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
Animal models have played a crucial role in the understanding of the mechanisms and treatments of human diseases; however, owing to the large differences in genetic background and disease-specific characteristics, animal models cannot fully simulate the occurrence and progression of human diseases. Recently, humanized immune system mice, based on immunodeficient mice, have been developed that allow for the partial reconstruction of the human immune system and mimic the human in vivo microenvironment. Systemic lupus erythematosus (SLE) is a complex disease characterized by the loss of tolerance to autoantigens, overproduction of autoantibodies, and inflammation in multiple organ systems. The detailed immunological events that trigger the onset of clinical manifestations in patients with SLE are still not well known. Two methods have been adopted for the development of humanized SLE mice. They include transferring peripheral blood mononuclear cells from patients with SLE to immunodeficient mice or transferring human hematopoietic stem cells to immunodeficient mice followed by intraperitoneal injection with pristane to induce lupus. However, there are still several challenges to be overcome, such as how to improve the efficiency of reconstruction of the human B cell immune response, how to extend the lifespan and improve the survival rate of mice to extend the observation period, and how to improve the development of standardized commercialized models and use them. In summary, there are opportunities and challenges for the development of humanized mouse models of SLE, which will provide novel strategies for understanding the mechanisms and treatments of SLE.
Collapse
Affiliation(s)
- Jiaxuan Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuzhen Liao
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huimin Zhou
- Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Lawei Yang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengbiao Guo
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuxian Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Aifen Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Quanren Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chen Yang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- *Correspondence: Hua-feng Liu, ; Qingjun Pan,
| | - Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- *Correspondence: Hua-feng Liu, ; Qingjun Pan,
| |
Collapse
|
39
|
Hori Y, Ohmine K, Katada H, Noguchi Y, Sato K, Nambu T, Adeline LR, Wan GS, Haraya K, Ozeki K, Nanami M, Tachibana T, Sampei Z, Kuramochi T, Nezu J, Hattori K, Igawa T. Elimination of plasma soluble antigen in cynomolgus monkeys by combining pH-dependent antigen binding and novel Fc engineering. MAbs 2022; 14:2068213. [PMID: 35482905 PMCID: PMC9067469 DOI: 10.1080/19420862.2022.2068213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
A conventional antibody targeting a soluble antigen in circulation typically requires a huge dosage and frequent intravenous administration to neutralize the antigen. This is because antigen degradation is reduced by the formation of antigen–antibody immune complexes, which escape from lysosomal degradation using neonatal Fc receptor (FcRn)-mediated recycling. To address this, we developed an antigen-sweeping antibody that combines pH-dependent antigen binding and Fc engineering to enhance Fc receptor binding. The sweeping antibody actively eliminates the plasma antigens by increasing the cellular uptake of the immune complex and dissociating the antigens in the acidic endosome for degradation. Strong antigen sweeping can reduce the dosage, potentially achieve higher efficacy, and expand the scope of antigen space available for targeting by antibodies. In this study, to further improve the sweeping efficacy, we developed a novel antibody Fc variant by enhancing Fcγ receptor IIb (FcγRIIb) binding and modulating charge characteristics for increased cellular uptake of the immune complex, together with enhancing FcRn binding for efficient salvage of the antigen-free antibodies. Our Fc variant achieved strong antigen sweeping in cynomolgus monkeys with antibody pharmacokinetics comparable to a wild-type human IgG1 antibody. The positive-charge substitutions enhanced uptake of the immune complex by FcγRIIb-expressing cells in vitro, which was completely inhibited by an anti-FcγRIIb antibody. This suggests that the strong in vivo sweeping efficacy improved by the charge engineering is more likely achieved by FcγRIIb-dependent uptake of the immune complex rather than nonspecific uptake. We expect this novel Fc engineering can maximize the antigen sweeping efficacy even in humans and create novel therapeutic antibodies that meet unmet medical needs for patients.
Collapse
Affiliation(s)
- Yuji Hori
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Ken Ohmine
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | | | - Yuki Noguchi
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Kazuki Sato
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | | | | | | | - Kenta Haraya
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Kazuhisa Ozeki
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Masahiko Nanami
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | | | - Zenjiro Sampei
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Taichi Kuramochi
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Junichi Nezu
- Research Division, Chugai Pharmaceutical Co, Ltd, Chuo-ku, Tokyo, Japan
| | - Kunihiro Hattori
- Research Division, Chugai Pharmaceutical Co, Ltd, Kamakura, Japan
| | - Tomoyuki Igawa
- Research Division, Chugai Pharmaceutical Co, Ltd, Chuo-ku, Tokyo, Japan
| |
Collapse
|
40
|
Chalayer E, Gramont B, Zekre F, Goguyer-Deschaumes R, Waeckel L, Grange L, Paul S, Chung AW, Killian M. Fc receptors gone wrong: A comprehensive review of their roles in autoimmune and inflammatory diseases. Autoimmun Rev 2021; 21:103016. [PMID: 34915182 DOI: 10.1016/j.autrev.2021.103016] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/08/2021] [Indexed: 12/16/2022]
Abstract
Systemic autoimmune and inflammatory diseases have a complex and only partially known pathophysiology with various abnormalities involving all the components of the immune system. Among these components, antibodies, and especially autoantibodies are key elements contributing to autoimmunity. The interaction of antibody fragment crystallisable (Fc) and several distinct receptors, namely Fc receptors (FcRs), have gained much attention during the recent years, with possible major therapeutic perspectives for the future. The aim of this review is to comprehensively describe the known roles for FcRs (activating and inhibitory FcγRs, neonatal FcR [FcRn], FcαRI, FcεRs, Ro52/tripartite motif containing 21 [Ro52/TRIM21], FcδR, and the novel Fc receptor-like [FcRL] family) in systemic autoimmune and inflammatory disorders, namely rheumatoid arthritis, Sjögren's syndrome, systemic lupus erythematosus, systemic sclerosis, idiopathic inflammatory myopathies, mixed connective tissue disease, Crohn's disease, ulcerative colitis, immunoglobulin (Ig) A vasculitis, Behçet's disease, Kawasaki disease, IgG4-related disease, immune thrombocytopenia, autoimmune hemolytic anemia, antiphospholipid syndrome and heparin-induced thrombocytopenia.
Collapse
Affiliation(s)
- Emilie Chalayer
- Department of Hematology and Cell Therapy, Institut de Cancérologie Lucien Neuwirth, Saint-Etienne, France; INSERM U1059-Sainbiose, dysfonction vasculaire et hémostase, Université de Lyon, Saint-Etienne, France
| | - Baptiste Gramont
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Franck Zekre
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Pediatrics, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Roman Goguyer-Deschaumes
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France
| | - Louis Waeckel
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Immunology, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Lucile Grange
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Immunology, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Amy W Chung
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Martin Killian
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France.
| |
Collapse
|
41
|
Wines BD, Trist HM, Esparon S, Impey RE, Mackay GA, Andrews RK, Soares da Costa TP, Pietersz GA, Baker RI, Hogarth PM. Fc Binding by FcγRIIa Is Essential for Cellular Activation by the Anti-FcγRIIa mAbs 8.26 and 8.2. Front Immunol 2021; 12:666813. [PMID: 34759915 PMCID: PMC8573391 DOI: 10.3389/fimmu.2021.666813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 10/05/2021] [Indexed: 11/21/2022] Open
Abstract
FcγR activity underpins the role of antibodies in both protective immunity and auto-immunity and importantly, the therapeutic activity of many monoclonal antibody therapies. Some monoclonal anti-FcγR antibodies activate their receptors, but the properties required for cell activation are not well defined. Here we examined activation of the most widely expressed human FcγR; FcγRIIa, by two non-blocking, mAbs, 8.26 and 8.2. Crosslinking of FcγRIIa by the mAb F(ab’)2 regions alone was insufficient for activation, indicating activation also required receptor engagement by the Fc region. Similarly, when mutant receptors were inactivated in the Fc binding site, so that intact mAb was only able to engage receptors via its two Fab regions, again activation did not occur. Mutation of FcγRIIa in the epitope recognized by the agonist mAbs, completely abrogated the activity of mAb 8.26, but mAb 8.2 activity was only partially inhibited indicating differences in receptor recognition by these mAbs. FcγRIIa inactivated in the Fc binding site was next co-expressed with the FcγRIIa mutated in the epitope recognized by the Fab so that each mAb 8.26 molecule can contribute only three interactions, each with separate receptors, one via the Fc and two via the Fab regions. When the Fab and Fc binding were thus segregated onto different receptor molecules receptor activation by intact mAb did not occur. Thus, receptor activation requires mAb 8.26 Fab and Fc interaction simultaneously with the same receptor molecules. Establishing the molecular nature of FcγR engagement required for cell activation may inform the optimal design of therapeutic mAbs.
Collapse
Affiliation(s)
- Bruce D Wines
- Immune Therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Halina M Trist
- Immune Therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
| | - Sandra Esparon
- Immune Therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
| | - Rachael E Impey
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Graham A Mackay
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, Australia
| | - Robert K Andrews
- Department Cancer Biology and Therapeutics, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Tatiana P Soares da Costa
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Geoffrey A Pietersz
- Immune Therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia.,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Ross I Baker
- Perth Blood Institute, Murdoch University, Perth, WA, Australia.,Western Australian Centre for Thrombosis and Haemostasis, Murdoch University, Murdoch, WA, Australia
| | - P Mark Hogarth
- Immune Therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
42
|
Kitanaga Y, Yamajuku D, Kubo S, Nakamura K, Maeda M, Seki M, Kaneko Y, Kinugasa F, Morokata T, Kondo Y, Yoshinari H, Nakayamada S, Sumida T, Tanaka Y. Discovery of a novel Igβ and FcγRIIB cross-linking antibody, ASP2713, and its potential application in the treatment of systemic lupus erythematosus. Int Immunopharmacol 2021; 101:108343. [PMID: 34781122 DOI: 10.1016/j.intimp.2021.108343] [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: 09/06/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 11/17/2022]
Abstract
B cell-targeted therapies have evolved as established therapies for systemic lupus erythematosus (SLE); however, existing approaches still do not thoroughly satisfy clinical requirements due to limited efficacy against memory B cells, autoantibody-producing plasmablasts and disease heterogeneity. To provide a new treatment option for SLE, we created a novel anti-Igβ antibody with enhanced affinity for Fc gamma receptor (FcγR) IIB called ASP2713. ASP2713 cross-reacted with both human and cynomolgus monkey Igβ and showed increased binding affinity for human and monkey FcγRIIB compared to native human IgG1. This binding property allows dominant B cell binding and induction of intrinsic negative feedback signals. In human B cells, ASP2713 significantly and concentration-dependently induced FcγRIIB ITIM phosphorylation, while suppressing proliferation under B cell receptor stimulation. This pharmacological effect was also confirmed in in vitro B cell proliferation and antibody production assays using peripheral B cells isolated from patients with SLE. In a cynomolgus monkey tetanus toxoid-induced antibody production model, ASP2713 almost completely inhibited the increase in antigen-specific antibodies with superior efficacy to rituximab. Additionally, ASP2713 significantly suppressed recall antibody production in response to secondary tetanus toxoid immunization, indicating the memory B cell- and plasmablast-targeting potential of ASP2713. Our results suggest that ASP2713 may have therapeutic potential as a treatment for SLE, where B cells play a pathogenic role.
Collapse
Affiliation(s)
- Yukihiro Kitanaga
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan; First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan.
| | - Daisuke Yamajuku
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Satoshi Kubo
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Koji Nakamura
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Masashi Maeda
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Mutsumi Seki
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Yoko Kaneko
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Fumitaka Kinugasa
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Tatsuaki Morokata
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Yuya Kondo
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroko Yoshinari
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Shingo Nakayamada
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
43
|
Gunn BM, Bai S. Building a better antibody through the Fc: advances and challenges in harnessing antibody Fc effector functions for antiviral protection. Hum Vaccin Immunother 2021; 17:4328-4344. [PMID: 34613865 PMCID: PMC8827636 DOI: 10.1080/21645515.2021.1976580] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
Antibodies can provide antiviral protection through neutralization and recruitment of innate effector functions through the Fc domain. While neutralization has long been appreciated for its role in antibody-mediated protection, a growing body of work indicates that the antibody Fc domain also significantly contributes to antiviral protection. Recruitment of innate immune cells such as natural killer cells, neutrophils, monocytes, macrophages, dendritic cells and the complement system by antibodies can lead to direct restriction of viral infection as well as promoting long-term antiviral immunity. Monoclonal antibody therapeutics against viruses are increasingly incorporating Fc-enhancing features to take advantage of the Fc domain, uncovering a surprising breadth of mechanisms through which antibodies can control viral infection. Here, we review the recent advances in our understanding of antibody-mediated innate immune effector functions in protection from viral infection and review the current approaches and challenges to effectively leverage innate immune cells via antibodies.
Collapse
Affiliation(s)
- Bronwyn M. Gunn
- Paul G. Allen School of Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Shuangyi Bai
- Paul G. Allen School of Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
44
|
Filbert EL, Björck PK, Srivastava MK, Bahjat FR, Yang X. APX005M, a CD40 agonist antibody with unique epitope specificity and Fc receptor binding profile for optimal therapeutic application. Cancer Immunol Immunother 2021; 70:1853-1865. [PMID: 33392713 PMCID: PMC8195934 DOI: 10.1007/s00262-020-02814-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022]
Abstract
Targeting CD40 with agonist antibodies is a promising approach to cancer immunotherapy. CD40 acts as a master regulator of immunity by mobilizing multiple arms of the immune system to initiate highly effective CD8 + T-cell-mediated responses against foreign pathogens and tumors. The clinical development of CD40 agonist antibodies requires careful optimization of the antibody to maximize therapeutic efficacy while minimizing adverse effects. Both epitope specificity and isotype are critical for CD40 agonist antibody mechanism of action and potency. We developed a novel antibody, APX005M, which binds with high affinity to the CD40 ligand-binding site on CD40 and is optimized for selective interaction with Fcγ receptors to enhance agonistic potency while limiting less desirable Fc-effector functions like antibody-dependent cellular cytotoxicity of CD40-expressing immune cells. APX005M is a highly potent inducer of innate and adaptive immune effector responses and represents a promising CD40 agonist antibody for induction of an effective anti-tumor immune response with a favorable safety profile.
Collapse
Affiliation(s)
- Erin L Filbert
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA
| | - Pia K Björck
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA
| | - Minu K Srivastava
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA
| | - Frances R Bahjat
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA
| | - Xiaodong Yang
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA.
| |
Collapse
|
45
|
On the Use of Surface Plasmon Resonance Biosensing to Understand IgG-FcγR Interactions. Int J Mol Sci 2021; 22:ijms22126616. [PMID: 34205578 PMCID: PMC8235063 DOI: 10.3390/ijms22126616] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 01/01/2023] Open
Abstract
Surface plasmon resonance (SPR)-based optical biosensors offer real-time and label-free analysis of protein interactions, which has extensively contributed to the discovery and development of therapeutic monoclonal antibodies (mAbs). As the biopharmaceutical market for these biologics and their biosimilars is rapidly growing, the role of SPR biosensors in drug discovery and quality assessment is becoming increasingly prominent. One of the critical quality attributes of mAbs is the N-glycosylation of their Fc region. Other than providing stability to the antibody, the Fc N-glycosylation influences immunoglobulin G (IgG) interactions with the Fcγ receptors (FcγRs), modulating the immune response. Over the past two decades, several studies have relied on SPR-based assays to characterize the influence of N-glycosylation upon the IgG-FcγR interactions. While these studies have unveiled key information, many conclusions are still debated in the literature. These discrepancies can be, in part, attributed to the design of the reported SPR-based assays as well as the methodology applied to SPR data analysis. In fact, the SPR biosensor best practices have evolved over the years, and several biases have been pointed out in the development of experimental SPR protocols. In parallel, newly developed algorithms and data analysis methods now allow taking into consideration complex biomolecular kinetics. In this review, we detail the use of different SPR biosensing approaches for characterizing the IgG-FcγR interactions, highlighting their merit and inherent experimental complexity. Furthermore, we review the latest SPR-derived conclusions on the influence of the N-glycosylation upon the IgG-FcγR interactions and underline the differences and similarities across the literature. Finally, we explore new avenues taking advantage of novel computational analysis of SPR results as well as the latest strategies to control the glycoprofile of mAbs during production, which could lead to a better understanding and modelling of the IgG-FcγRs interactions.
Collapse
|
46
|
Abstract
The host immune system is highly compromised in case of viral infections and relapses are very common. The capacity of the virus to destroy the host cell by liberating its own DNA or RNA and replicating inside the host cell poses challenges in the development of antiviral therapeutics. In recent years, many new technologies have been explored for diagnosis, prevention, and treatment of viral infections. Nanotechnology has emerged as one of the most promising technologies on account of its ability to deal with viral diseases in an effective manner, addressing the limitations of traditional antiviral medicines. It has not only helped us to overcome problems related to solubility and toxicity of drugs, but also imparted unique properties to drugs, which in turn has increased their potency and selectivity toward viral cells against the host cells. The initial part of the paper focuses on some important proteins of influenza, Ebola, HIV, herpes, Zika, dengue, and corona virus and those of the host cells important for their entry and replication into the host cells. This is followed by different types of nanomaterials which have served as delivery vehicles for the antiviral drugs. It includes various lipid-based, polymer-based, lipid-polymer hybrid-based, carbon-based, inorganic metal-based, surface-modified, and stimuli-sensitive nanomaterials and their application in antiviral therapeutics. The authors also highlight newer promising treatment approaches like nanotraps, nanorobots, nanobubbles, nanofibers, nanodiamonds, nanovaccines, and mathematical modeling for the future. The paper has been updated with the recent developments in nanotechnology-based approaches in view of the ongoing pandemic of COVID-19.Graphical abstract.
Collapse
Affiliation(s)
- Malobika Chakravarty
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Amisha Vora
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
47
|
Cargill T, Culver EL. The Role of B Cells and B Cell Therapies in Immune-Mediated Liver Diseases. Front Immunol 2021; 12:661196. [PMID: 33936097 PMCID: PMC8079753 DOI: 10.3389/fimmu.2021.661196] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
B cells form a branch of the adaptive immune system, essential for the body’s immune defense against pathogens. B cell dysfunction has been implicated in the pathogenesis of immune mediated liver diseases including autoimmune hepatitis, IgG4-related hepatobiliary disease, primary biliary cholangitis and primary sclerosing cholangitis. B cells may initiate and maintain immune related liver diseases in several ways including the production of autoantibodies and the activation of T cells via antigen presentation or cytokine production. Here we comprehensively review current knowledge on B cell mechanisms in immune mediated liver diseases, exploring disease pathogenesis, B cell therapies, and novel treatment targets. We identify key areas where future research should focus to enable the development of targeted B cell therapies.
Collapse
Affiliation(s)
- Tamsin Cargill
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Emma L Culver
- Oxford Liver Unit, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
48
|
Gunn BM, Lu R, Slein MD, Ilinykh PA, Huang K, Atyeo C, Schendel SL, Kim J, Cain C, Roy V, Suscovich TJ, Takada A, Halfmann PJ, Kawaoka Y, Pauthner MG, Momoh M, Goba A, Kanneh L, Andersen KG, Schieffelin JS, Grant D, Garry RF, Saphire EO, Bukreyev A, Alter G. A Fc engineering approach to define functional humoral correlates of immunity against Ebola virus. Immunity 2021; 54:815-828.e5. [PMID: 33852832 PMCID: PMC8111768 DOI: 10.1016/j.immuni.2021.03.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 01/28/2021] [Accepted: 03/16/2021] [Indexed: 01/31/2023]
Abstract
Protective Ebola virus (EBOV) antibodies have neutralizing activity and induction of antibody constant domain (Fc)-mediated innate immune effector functions. Efforts to enhance Fc effector functionality often focus on maximizing antibody-dependent cellular cytotoxicity, yet distinct combinations of functions could be critical for antibody-mediated protection. As neutralizing antibodies have been cloned from EBOV disease survivors, we sought to identify survivor Fc effector profiles to help guide Fc optimization strategies. Survivors developed a range of functional antibody responses, and we therefore applied a rapid, high-throughput Fc engineering platform to define the most protective profiles. We generated a library of Fc variants with identical antigen-binding fragments (Fabs) from an EBOV neutralizing antibody. Fc variants with antibody-mediated complement deposition and moderate natural killer (NK) cell activity demonstrated complete protective activity in a stringent in vivo mouse model. Our findings highlight the importance of specific effector functions in antibody-mediated protection, and the experimental platform presents a generalizable resource for identifying correlates of immunity to guide therapeutic antibody design.
Collapse
Affiliation(s)
- Bronwyn M Gunn
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Richard Lu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Matthew D Slein
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Jiyoung Kim
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Caitlin Cain
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Ayato Takada
- Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Peter J Halfmann
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Matthias G Pauthner
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Mambu Momoh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
| | - Augustine Goba
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
| | - Lansana Kanneh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
| | - Kristian G Andersen
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA
| | - John S Schieffelin
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Donald Grant
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone; Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Robert F Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
49
|
Farley CR, Morris AB, Tariq M, Bennion KB, Potdar S, Kudchadkar R, Lowe MC, Ford ML. FcγRIIB is a T cell checkpoint in antitumor immunity. JCI Insight 2021; 6:135623. [PMID: 33616086 PMCID: PMC7934918 DOI: 10.1172/jci.insight.135623] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/20/2021] [Indexed: 01/02/2023] Open
Abstract
In the setting of cancer, T cells upregulate coinhibitory molecules that attenuate TCR signaling and lead to the loss of proliferative capacity and effector function. Checkpoint inhibitors currently in clinical use have dramatically improved mortality from melanoma yet are not effective in all patients, suggesting that additional pathways may contribute to suppression of tumor-specific CD8+ T cell responses in melanoma. Here, we show that FcγRIIB, an inhibitory Fc receptor previously thought to be exclusively expressed on B cells and innate immune cells, is upregulated on tumor-infiltrating effector CD8+ T cells in an experimental melanoma model and expressed on CD8+ T cells in patients with melanoma. Genetic deficiency of Fcgr2b resulted in enhanced tumor-infiltrating CD8+ T cell responses and significantly reduced tumor burden. Adoptive transfer experiments of Fcgr2b–/– tumor antigen-specific T cells into FcγRIIB-sufficient hosts resulted in an increased frequency of tumor-infiltrating CD8+ T cells with greater effector function. Finally, FcγRIIB was expressed on CD8+ memory T cells isolated from patients with melanoma. These data illuminate a cell-intrinsic role for the FcγRIIB checkpoint in suppressing tumor-infiltrating CD8+ T cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Ragini Kudchadkar
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Michael C Lowe
- Department of Surgery and.,Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | | |
Collapse
|
50
|
Turman JM, Cheplowitz AM, Tiwari C, Thomas T, Joshi D, Bhat M, Wu Q, Pong E, Chu SY, Szymkowski DE, Sharma A, Seveau S, Robinson JM, Kwiek JJ, Burton D, Rajaram MVS, Kim J, Hangartner L, Ganesan LP. Accelerated Clearance and Degradation of Cell-Free HIV by Neutralizing Antibodies Occurs via FcγRIIb on Liver Sinusoidal Endothelial Cells by Endocytosis. THE JOURNAL OF IMMUNOLOGY 2021; 206:1284-1296. [PMID: 33568400 DOI: 10.4049/jimmunol.2000772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/05/2021] [Indexed: 01/19/2023]
Abstract
Neutralizing Abs suppress HIV infection by accelerating viral clearance from blood circulation in addition to neutralization. The elimination mechanism is largely unknown. We determined that human liver sinusoidal endothelial cells (LSEC) express FcγRIIb as the lone Fcγ receptor, and using humanized FcγRIIb mouse, we found that Ab-opsonized HIV pseudoviruses were cleared considerably faster from circulation than HIV by LSEC FcγRIIb. Compared with humanized FcγRIIb-expressing mice, HIV clearance was significantly slower in FcγRIIb knockout mice. Interestingly, a pentamix of neutralizing Abs cleared HIV faster compared with hyperimmune anti-HIV Ig (HIVIG), although the HIV Ab/Ag ratio was higher in immune complexes made of HIVIG and HIV than pentamix and HIV. The effector mechanism of LSEC FcγRIIb was identified to be endocytosis. Once endocytosed, both Ab-opsonized HIV pseudoviruses and HIV localized to lysosomes. This suggests that clearance of HIV, endocytosis, and lysosomal trafficking within LSEC occur sequentially and that the clearance rate may influence downstream events. Most importantly, we have identified LSEC FcγRIIb-mediated endocytosis to be the Fc effector mechanism to eliminate cell-free HIV by Abs, which could inform development of HIV vaccine and Ab therapy.
Collapse
Affiliation(s)
- James M Turman
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Alana M Cheplowitz
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Charu Tiwari
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Thushara Thomas
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Dhruvi Joshi
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Menakshi Bhat
- Center for Retrovirus Research, Department of Microbiology, The Ohio State University, Columbus, OH 43210
| | - Qian Wu
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | | | | | | | - Amit Sharma
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210.,Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Stephanie Seveau
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - John M Robinson
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210
| | - Jesse J Kwiek
- Center for Retrovirus Research, Department of Microbiology, The Ohio State University, Columbus, OH 43210
| | - Dennis Burton
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Murugesan V S Rajaram
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115
| | - Lars Hangartner
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Latha P Ganesan
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210;
| |
Collapse
|