1
|
Crescioli S, Jatiani S, Moise L. With great power, comes great responsibility: the importance of broadly measuring Fc-mediated effector function early in the antibody development process. MAbs 2025; 17:2453515. [PMID: 39819511 PMCID: PMC11810086 DOI: 10.1080/19420862.2025.2453515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
The field of antibody therapeutics is rapidly growing, with over 210 antibodies currently approved or in regulatory review and ~ 1,250 antibodies in clinical development. Antibodies are highly versatile molecules that, with strategic design of their antigen-binding domain (Fab) and the domain responsible for mediating effector functions (Fc), can be used in a wide range of therapeutic indications. Building on many years of progress, the biopharmaceutical industry is now advancing innovative research and development by exploring new targets and new formats and using antibody engineering to fine-tune functions tailored to specific disease requirements. In addition to considering the target and the disease context, however, the unique features of each therapeutic antibody trigger a diverse set of Fc-mediated effector functions. To avoid unexpected results on safety and efficacy outcomes during the later stages of the development process, it is crucial to measure the impact of antibody design on Fc-mediated effector function early in the antibody development process. Given the breadth of effector functions antibodies can deploy and the close interplay between the antibody Fab and Fc functional domains, it is important to conduct a comprehensive evaluation of Fc-mediated functions using an array of antigen-specific biophysical and cell-mediated functional assays. Here, we review antibody and Fc receptor properties that influence Fc effector functions and discuss their implications on development of safe and efficacious antibody therapeutics.
Collapse
|
2
|
Loyau J, Monney T, Montefiori M, Bokhovchuk F, Streuli J, Blackburn M, Goepfert A, Caro LN, Chakraborti S, De Angelis S, Grandclément C, Blein S, Mbow ML, Srivastava A, Perro M, Sammicheli S, Zhukovsky EA, Dyson M, Dreyfus C. Biparatopic binding of ISB 1442 to CD38 in trans enables increased cell antibody density and increased avidity. MAbs 2025; 17:2457471. [PMID: 39882744 PMCID: PMC11784651 DOI: 10.1080/19420862.2025.2457471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 01/31/2025] Open
Abstract
ISB 1442 is a bispecific biparatopic antibody in clinical development to treat hematological malignancies. It consists of two adjacent anti-CD38 arms targeting non-overlapping epitopes that preferentially drive binding to tumor cells and a low-affinity anti-CD47 arm to enable avidity-induced blocking of proximal CD47 receptors. We previously reported the pharmacology of ISB 1442, designed to reestablish synthetic immunity in CD38+ hematological malignancies. Here, we describe the discovery, optimization and characterization of the ISB 1442 antigen binding fragment (Fab) arms, their assembly to 2 + 1 format, and present the high-resolution co-crystal structures of the two anti-CD38 Fabs, in complex with CD38. This, with biophysical and functional assays, elucidated the underlying mechanism of action of ISB 1442. In solution phase, ISB 1442 forms a 2:2 complex with CD38 as determined by size-exclusion chromatography with multi-angle light scattering and electron microscopy. The predicted antibody-antigen stoichiometries at different CD38 surface densities were experimentally validated by surface plasmon resonance and cell binding assays. The specific design and structural features of ISB 1442 enable: 1) enhanced trans binding to adjacent CD38 molecules to increase Fc density at the cancer cell surface; 2) prevention of avid cis binding to monomeric CD38 to minimize blockade by soluble shed CD38; and 3) greater binding avidity, with a slower off-rate at high CD38 density, for increased specificity. The superior CD38 targeting of ISB 1442, at both high and low receptor densities, by its biparatopic design, will enhance proximal CD47 blockade and thus counteract a major tumor escape mechanism in multiple myeloma patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mario Perro
- Ichnos Glenmark Innovation, New York, NY, USA
| | | | | | | | | |
Collapse
|
3
|
Troisi M, Fabbrini M, Stazzoni S, Viviani V, Carboni F, Abbiento V, Fontana LE, Tomei S, Audagnotto M, Santini L, Spagnuolo A, Antonelli G, Paciello I, Vacca F, Cardamone D, Marini E, Mokhtary P, Finetti F, Giusti F, Bodini M, Torricelli G, Limongi C, Del Vecchio M, Favaron S, Tavarini S, Sammicheli C, Rossi A, Mandelli AP, Fortini P, Caffarelli C, Gonnelli S, Nuti R, Efron A, Baldari CT, Sala C, Tagliabue A, Savino S, Brunelli B, Norais N, Frigimelica E, Bardelli M, Pizza M, Margarit I, Delany I, Finco O, Andreano E, Rappuoli R. Human monoclonal antibodies targeting subdominant meningococcal antigens confer cross-protection against gonococcus. Sci Transl Med 2025; 17:eadv0969. [PMID: 40397716 DOI: 10.1126/scitranslmed.adv0969] [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: 12/05/2024] [Accepted: 04/01/2025] [Indexed: 05/23/2025]
Abstract
Gonococcus, a bacterium resistant to most antibiotics, causes more than 80 million cases of gonorrhea annually and is considered a high-priority pathogen by the World Health Organization. Recently, vaccine development prospects were boosted by reports that licensed meningococcus serogroup B (MenB) vaccines provided partial protection against gonococcal infection. To determine antigens responsible for cross-protection, memory B cells isolated from 4CMenB-vaccinated volunteers were single cell-sorted to identify antibodies that kill gonococcus in a bactericidal assay. Nine different antibodies, all deriving from the IGHV4-34 germline and carrying an unusually long heavy-chain complementarity-determining region 3, recognized the PorB protein; four others recognized the lipooligosaccharide; and another four had unknown specificity. One of the PorB-specific antibodies provided protection in a mouse model of gonococcus infection. The identification of PorB and lipooligosaccharide as key antigens of gonococcal and meningococcal immunity provides a mechanistic explanation of the cross-protection observed in the clinic and shows that isolating human monoclonal antibodies from vaccinees can be instrumental for bacterial antigen discovery.
Collapse
Affiliation(s)
- Marco Troisi
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | | | - Samuele Stazzoni
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | | | | | - Valentina Abbiento
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | | | | | | | | | | | - Giada Antonelli
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Ida Paciello
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Fabiola Vacca
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Dario Cardamone
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
- University of Turin, Turin, Italy
| | - Eleonora Marini
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Pardis Mokhtary
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | | | | | | | | | | | | | - Sara Favaron
- GSK, Siena, Italy
- Department of Chemistry, Materials and Chemical Engineering, Politecnico di Milano, Milan, Italy
| | | | | | | | | | - Pietro Fortini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Carla Caffarelli
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Stefano Gonnelli
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Ranuccio Nuti
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Adriana Efron
- Instituto Nacional de Enfermedades Infecciosas-ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| | | | - Claudia Sala
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Aldo Tagliabue
- Institute Biomedical Technologies, National Research Council, Segrate, Milan, Italy
| | | | | | | | | | | | - Mariagrazia Pizza
- GSK, Siena, Italy
- Imperial College London, South Kensington Campus, London, UK
| | | | | | | | - Emanuele Andreano
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Rino Rappuoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Fondazione Biotecnopolo di Siena, Siena, Italy
| |
Collapse
|
4
|
Bick MV, Puig E, Beauparlant D, Nedellec R, Burton I, Ardaghi K, Zalunardo TR, Bastidas R, Li X, Guenaga J, Lee WH, Wyatt R, Zhu W, Crispin M, Ozorowski G, Ward AB, Burton DR, Hangartner L. Molecular parameters governing antibody FcγR signaling and effector functions in the context of HIV envelope. Cell Rep 2025; 44:115331. [PMID: 40158219 DOI: 10.1016/j.celrep.2025.115331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 10/29/2024] [Accepted: 01/28/2025] [Indexed: 04/02/2025] Open
Abstract
Antibody effector functions contribute to the immune response to pathogens and can influence the efficacy of antibodies as therapeutics. To date, however, there is limited information on the molecular parameters that govern fragment crystallizable (Fc) effector functions. In this study, using AI-assisted protein design, the influences of binding kinetics, epitope location, and stoichiometry of binding on cellular Fc effector functions were investigated using engineered HIV-1 envelope as a model antigen. For this antigen, stoichiometry of binding was found to be the primary molecular determinant of FcγRIIIa signaling, antibody-dependent cellular cytotoxicity, and antibody-dependent cellular phagocytosis, while epitope location and antibodybinding kinetics, at least in the ranges investigated, were of no substantial impact. These findings are of importance for informing the development of vaccination strategies against HIV-1 and, possibly, other viral pathogens.
Collapse
Affiliation(s)
- Michael V Bick
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - Eduard Puig
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - David Beauparlant
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - Rebecca Nedellec
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - Iszac Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - Keihvan Ardaghi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - Thea R Zalunardo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - Raiza Bastidas
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - Xuduo Li
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - Javier Guenaga
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92109, USA
| | - Richard Wyatt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Wenwen Zhu
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92109, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Andrew B Ward
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92109, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dennis R Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Lars Hangartner
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92109, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
5
|
Vezzani G, Viviani V, Audagnotto M, Rossi A, Cinelli P, Pacchiani N, Limongi C, Santini L, Giusti F, Tomei S, Torricelli G, Faenzi E, Sammicheli C, Tavarini S, Efron A, Biolchi A, Finco O, Delany I, Frigimelica E. Isolation of human monoclonal antibodies from 4CMenB vaccinees reveals PorB and LOS as the main OMV components inducing cross-strain protection. Front Immunol 2025; 16:1565862. [PMID: 40308602 PMCID: PMC12040683 DOI: 10.3389/fimmu.2025.1565862] [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: 01/23/2025] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction The 4CMenB vaccine licensed against serogroup B Neisseria meningitidis (MenB) contains three recombinant proteins and Outer Membrane Vesicles (OMV) from a New Zealand epidemic strain. The protective response mediated on differentmeningococcal strains has been historically ascribed to one of the four main vaccine antigens fHbp, NHBA, NadA, and PorA nominated as the immunodominant antigen of the OMV component. It is however accepted that the extensive cross-protection observed after vaccination may be attributed to other proteins in the OMV. Here we interrogate the B cell responses elicited in humans to the OMV component after 4CMenB vaccination to elucidate the contribution of additional OMV antigens to meningococcal cross-protection. Methods Following the isolation of plasmablasts from vaccinees, the OMV-specific human monoclonal antibodies (HumAbs) were recombinantly expressed and characterized for their binding and functional activity on a panel of MenB strains. Their target specificity was assessed through a tailor-made protein array and Western blot. Results We found that 18 HumAbs showing bactericidal activity were PorB-specific, 1 was LOS-specific and 4 functional HumAbs remain with unknown targets. We identified three functional classes within the PorB HumAbs, through binding and in silico docking experiments, likely to be elicited from distinct epitopes on PorB and highlighting this antigen as a multi-epitope immunogenic OMV component responsible for distinct cross-protection across multiple MenB strains. Interestingly three of the PorB HumAbs and the LOS-specific HumAb showed bactericidal activity also against gonococcus. Discussion We identified PorB and LOS as antigens on the OMV that may be implicated in the real-world observations of moderate protection against gonorrhea infection after OMV-based vaccinations.
Collapse
Affiliation(s)
- Giacomo Vezzani
- GSK Vaccines, Siena, Italy
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Adriana Efron
- Departamento de Bacteriología, Instituto Nacional de Enfermedades Infecciosas-ANLIS “Dr. Carlos G. Malbrán”, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
6
|
Kang N, Duan Q, Min X, Li T, Li Y, Gao J, Liu W. Multifaceted function of B cells in tumorigenesis. Front Med 2025; 19:297-317. [PMID: 40119025 DOI: 10.1007/s11684-025-1127-5] [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] [Accepted: 12/16/2024] [Indexed: 03/24/2025]
Abstract
B lymphocytes (B cells) play a complex and paradoxical role in tumorigenesis. They can recognize tumor-associated antigens, present these antigens to T cells, and produce antibodies that directly target and eliminate tumor cells. This makes B cells a potentially powerful ally in combating cancer. However, B cells also exhibit immunosuppressive functions, secreting cytokines like IL-10 or generating tumor-promoting antibodies that dampen the anti-tumor immune response, and some tumor cells have even been shown to exploit B cells to promote their growth and metastasis. This dual nature of B cells presents both opportunities and challenges for tumor immunotherapy. In this review, we summarize the mechanisms underlying the multifaceted functions of B cells and their current applications in cancer immunotherapy. Furthermore, we also explore the key issues and future directions in this field, emphasizing the need for further research to fully harness the anti-tumor potential of B cells in the fight against cancer.
Collapse
Affiliation(s)
- Na Kang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Qinghui Duan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Xin Min
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Tong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Yuxin Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- School of Management and Engineering, Nanjing University, Nanjing, 210008, China
- Postdoctoral Workstation, Govtor Capital Co., Ltd., Nanjing, 210013, China
| | - Ji Gao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| |
Collapse
|
7
|
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
|
8
|
Kaneko MK, Suzuki H, Ohishi T, Nakamura T, Yanaka M, Tanaka T, Kato Y. Antitumor Activities of a Humanized Cancer-Specific Anti-HER2 Monoclonal Antibody, humH 2Mab-250 in Human Breast Cancer Xenografts. Int J Mol Sci 2025; 26:1079. [PMID: 39940848 PMCID: PMC11817376 DOI: 10.3390/ijms26031079] [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: 12/29/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Monoclonal antibody (mAb) and cell-based immunotherapies represent cutting-edge strategies for cancer treatment. However, safety concerns persist due to the potential targeting of normal cells that express reactive antigens. Therefore, it is crucial to develop cancer-specific mAbs (CasMabs) that can bind to cancer-specific antigens and exhibit antitumor activity in vivo, thereby reducing the risk of adverse effects. We previously screened mAbs targeting human epidermal growth factor receptor 2 (HER2) and successfully developed a cancer-specific anti-HER2 mAb, H2Mab-250/H2CasMab-2 (mouse IgG1, kappa). In this study, we assessed both the in vitro and in vivo antitumor efficacy of the humanized H2Mab-250 (humH2Mab-250). Although humH2Mab-250 showed lower reactivity to HER2-overexpressed Chinese hamster ovary-K1 (CHO/HER2) and breast cancer cell lines (BT-474 and SK-BR-3) than trastuzumab in flow cytometry, both humH2Mab-250 and trastuzumab showed similar antibody-dependent cellular cytotoxicity (ADCC) against CHO/HER2 and the breast cancer cell lines in the presence of effector splenocytes. In addition, humH2Mab-250 exhibited significant complement-dependent cellular cytotoxicity (CDC) in CHO/HER2 and the breast cancer cell lines compared to trastuzumab. Furthermore, humH2Mab-250 possesses compatible in vivo antitumor effects against CHO/HER2 and breast cancer xenografts with trastuzumab. These findings highlight the distinct roles of ADCC and CDC in the antitumor effects of humH2Mab-250 and trastuzumab and suggest a potential direction for the clinical development of humH2Mab-250 for HER2-positive tumors.
Collapse
MESH Headings
- Humans
- Animals
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/antagonists & inhibitors
- Breast Neoplasms/drug therapy
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Female
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Mice
- CHO Cells
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Cricetulus
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Trastuzumab/pharmacology
- Antibodies, Monoclonal/pharmacology
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.K.K.); (T.N.); (M.Y.); (T.T.)
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.K.K.); (T.N.); (M.Y.); (T.T.)
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, 18-24 Miyamoto, Numazu 410-0301, Shizuoka, Japan;
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku 141-0021, Tokyo, Japan
| | - Takuro Nakamura
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.K.K.); (T.N.); (M.Y.); (T.T.)
| | - Miyuki Yanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.K.K.); (T.N.); (M.Y.); (T.T.)
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.K.K.); (T.N.); (M.Y.); (T.T.)
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.K.K.); (T.N.); (M.Y.); (T.T.)
| |
Collapse
|
9
|
Goldberg BS, Ackerman ME. Underappreciated layers of antibody-mediated immune synapse architecture and dynamics. mBio 2025; 16:e0190024. [PMID: 39660921 PMCID: PMC11708040 DOI: 10.1128/mbio.01900-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
The biologic activities of antibody drugs are dictated by structure-function relationships-emerging from the kind, composition, and degree of interactions with a target antigen and with soluble and cellular antibody receptors of the innate immune system. These activities are canonically understood to be both modular: antigen recognition is driven by the heterodimeric antigen-binding fragment, and innate immune recruitment by the homodimeric constant/crystallizable fragment. The model that treats these domains with a high degree of independence has served the field well but is not without limitations. Here, we consider how new insights, particularly from structural studies, complicate the model of neat biophysical separation between these domains and shape our understanding of antibody effector functions. The emerging model endeavors to explain the phenotypic impact of both antibody intrinsic characteristics and extrinsic features-fitting them within a spatiotemporal paradigm that better accounts for observed antibody activities. In this review, we will use insights from recent models of classical complement complexes and T cell immune synapse formation to explore how structural differences in antibody-mediated immune synapses may relate to their functional diversity.
Collapse
Affiliation(s)
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
10
|
Pande AH, Sandeep, Shinde SH. Polybodies: Next-generation clinical antibodies. Drug Discov Today 2024; 29:104198. [PMID: 39369985 DOI: 10.1016/j.drudis.2024.104198] [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: 08/29/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Conventional antibodies [full-length and fragments: F(ab')2, fragment antigen-binding (Fab), single-chain variable fragment (scFv), variable heavy domain of heavy chain antibody (VHH)] are monospecific, first-generation antibodies, that have dominated the biopharmaceuticals field. However, protein engineering approaches has led to the advent of the next-generation antibodies (polybodies), which are significant improvement over the conventional antibodies. Polybodies comprise polyspecific and/or polyvalent antibodies that enable a single antibody to target multiple specific antigens simultaneously. Polybodies are superior to first-generation antibodies (more efficacious, broad-spectrum, resistance resilient, customizable, etc.) and provide a cost-effective healthcare solution. This review addresses recent developments in polybodies, highlighting their superiority over conventional antibodies and offering future perspectives to encourage the generation of innovative immunotherapies.
Collapse
Affiliation(s)
- Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| | - Sandeep
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Suraj H Shinde
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| |
Collapse
|
11
|
van der Horst HJ, Mutis T. Enhancing Fc-mediated effector functions of monoclonal antibodies: The example of HexaBodies. Immunol Rev 2024; 328:456-465. [PMID: 39275983 PMCID: PMC11659923 DOI: 10.1111/imr.13394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
Since the approval of the CD20-targeting monoclonal antibody (mAb) rituximab for the treatment of lymphoma in 1997, mAb therapy has significantly transformed cancer treatment. With over 90 FDA-approved mAbs for the treatment of various hematological and solid cancers, modern cancer treatment relies heavily on these therapies. The overwhelming success of mAbs as cancer therapeutics is attributed to their broad applicability, high safety profile, and precise targeting of cancer-associated surface antigens. Furthermore, mAbs can induce various anti-tumor cytotoxic effector mechanisms including antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC), all of which are mediated via their fragment crystallizable (Fc) domain. Over the past decades, these effector mechanisms have been substantially improved through Fc domain engineering. In this review, we will outline the different approaches to enhance Fc effector functions via Fc engineering of mAbs, with a specific emphasis on the so-called "HexaBody" technology, which is designed to enhance the hexamerization of mAbs on the target cell surface, thereby inducing greater complement activation, CDC, and receptor clustering. The review will summarize the development, preclinical, and clinical testing of several HexaBodies designed for the treatment of B-cell malignancies, as well as the potential use of the HexaBody technology beyond Fc-mediated effector functions.
Collapse
Affiliation(s)
- Hilma J. van der Horst
- Department of HematologyCancer Center Amsterdam, Amsterdam UMC, VU Medical CenterAmsterdamThe Netherlands
- Present address:
Department of Fundamental Oncology, Ludwig Institute for Cancer ResearchUniversity of LausanneEpalingesSwitzerland
| | - Tuna Mutis
- Department of HematologyCancer Center Amsterdam, Amsterdam UMC, VU Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
12
|
Whitehead CA, Wines BD, Davies AM, McDonnell JM, Trist HM, Esparon SE, Hogarth PM. Stellabody: A novel hexamer-promoting mutation for improved IgG potency. Immunol Rev 2024; 328:438-455. [PMID: 39364646 PMCID: PMC11659935 DOI: 10.1111/imr.13400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Advances in antibody engineering are being directed at the development of next generation immunotherapeutics with improved potency. Hexamerisation of IgG is a normal physiological aspect of IgG biology and recently described mutations that facilitate this process have a substantial impact upon monoclonal antibody behavior resulting in the elicitation of dramatically enhanced complement-dependent cytotoxicity, Fc receptor function, and enhanced antigen binding effects, such as targeted receptor agonism or microbe neutralization. Whereas the discovery of IgG hexamerisation enhancing mutations has largely focused on residues with exposure at the surface of the Fc-Fc and CH2-CH3 interfaces, our unique approach is the engineering of the mostly buried residue H429 in the CH3 domain. Selective substitution at position 429 forms the basis of Stellabody technology, where the choice of amino acid results in distinct hexamerisation outcomes. H429F results in monomeric IgG that hexamerises after target binding, so called "on-target" hexamerisation, while the H429Y mutant forms pH-sensitive hexamers in-solution prior to antigen binding. Moreover, Stellabody technologies are broadly applicable across the family of antibody-based biologic therapeutics, including conventional mAbs, bispecific mAbs, and Ig-like biologics such as Fc-fusions, with applications in diverse diseases.
Collapse
Affiliation(s)
- Clarissa A. Whitehead
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Bruce D. Wines
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Anna M. Davies
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's HouseLondonUK
| | - James M. McDonnell
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's HouseLondonUK
| | - Halina M. Trist
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
| | | | - P. Mark Hogarth
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
- Department of Clinical PathologyThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
13
|
Ishikawa K, Suzuki H, Ohishi T, Nakamura T, Yanaka M, Li G, Tanaka T, Ohkoshi A, Kawada M, Kaneko MK, Katori Y, Kato Y. Antitumor activities of anti‑CD44 monoclonal antibodies in mouse xenograft models of esophageal cancer. Oncol Rep 2024; 52:147. [PMID: 39219278 PMCID: PMC11391255 DOI: 10.3892/or.2024.8806] [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: 04/18/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
CD44 is a type I transmembrane glycoprotein associated with poor prognosis in various solid tumors. Since CD44 plays a critical role in tumor development by regulating cell adhesion, survival, proliferation and stemness, it has been considered a target for tumor therapy. Anti‑CD44 monoclonal antibodies (mAbs) have been developed and applied to antibody‑drug conjugates and chimeric antigen receptor‑T cell therapy. Anti-pan‑CD44 mAbs, C44Mab‑5 and C44Mab‑46, which recognize both CD44 standard (CD44s) and variant isoforms were previously developed. The present study generated a mouse IgG2a version of the anti‑pan‑CD44 mAbs (5‑mG2a and C44Mab‑46‑mG2a) to evaluate the antitumor activities against CD44‑positive cells. Both 5‑mG2a and C44Mab‑46‑mG2a recognized CD44s‑overexpressed CHO‑K1 (CHO/CD44s) cells and esophageal tumor cell line (KYSE770) in flow cytometry. Furthermore, both 5‑mG2a and C44Mab‑46‑mG2a could activate effector cells in the presence of CHO/CD44s cells and exhibited complement-dependent cytotoxicity against both CHO/CD44s and KYSE770 cells. Furthermore, the administration of 5‑mG2a and C44Mab‑46‑mG2a significantly suppressed CHO/CD44s and KYSE770 xenograft tumor development compared with the control mouse IgG2a. These results indicate that 5‑mG2a and C44Mab‑46‑mG2a could exert antitumor activities against CD44‑positive cancers and be a promising therapeutic regimen for tumors.
Collapse
Affiliation(s)
- Kenichiro Ishikawa
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu, Shizuoka 410‑0301, Japan
| | - Takuro Nakamura
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Miyuki Yanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Guanjie Li
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Akira Ohkoshi
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, Shinagawa‑ku, Tokyo 141‑0021, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Yukio Katori
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| |
Collapse
|
14
|
Frischauf N, Strasser J, Borg EG, Labrijn AF, Beurskens FJ, Preiner J. Complement activation by IgG subclasses is governed by their ability to oligomerize upon antigen binding. Proc Natl Acad Sci U S A 2024; 121:e2406192121. [PMID: 39436656 PMCID: PMC11536094 DOI: 10.1073/pnas.2406192121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/05/2024] [Indexed: 10/23/2024] Open
Abstract
Complement activation through antibody-antigen complexes is crucial in various pathophysiological processes and utilized in immunotherapies to eliminate infectious agents, regulatory immune cells, or cancer cells. The tertiary structures of the four IgG antibody subclasses are largely comparable, with the most prominent difference being the hinge regions connecting the Fab and Fc domains, providing them with unique structural flexibility. Complement recruitment and activation depend strongly on IgG subclass, which is commonly rationalized by differences in hinge flexibility and the respective affinities for C1, the first component of the classical complement pathway. However, a unifying mechanism of how these different IgG subclass properties combine to modulate C1 activation has not yet been proposed. We here demonstrate that complement activation is determined by their varying ability to form IgG oligomers on antigenic surfaces large enough to multivalently bind and activate C1. We directly visualize the resulting IgG oligomer structures and characterize their distribution by means of high-speed atomic force microscopy, quantify their complement recruitment efficiency from quartz crystal microbalance experiments, and characterize their ability to activate complement on tumor cell lines as well as in vesicle-based complement lysis assays. We present a mechanistic model of the multivalent interactions that govern C1 binding to IgG oligomers and use it to extract kinetic rate constants from real-time interaction data from which we further calculate equilibrium dissociation constants. Together, we provide a comprehensive view on the parameters that govern complement activation by the different IgG subclasses, which may inform the design of future antibody therapies.
Collapse
Affiliation(s)
- Nikolaus Frischauf
- Medical Engineering, Nano Structuring and Bio-Analytics, University of Applied Sciences Upper Austria, Linz4020, Austria
| | - Jürgen Strasser
- Medical Engineering, Nano Structuring and Bio-Analytics, University of Applied Sciences Upper Austria, Linz4020, Austria
| | | | | | | | - Johannes Preiner
- Medical Engineering, Nano Structuring and Bio-Analytics, University of Applied Sciences Upper Austria, Linz4020, Austria
| |
Collapse
|
15
|
Long H, Simmons A, Mayorga A, Burgess B, Nguyen T, Budda B, Rychkova A, Rhinn H, Tassi I, Ward M, Yeh F, Schwabe T, Paul R, Kenkare-Mitra S, Rosenthal A. Preclinical and first-in-human evaluation of AL002, a novel TREM2 agonistic antibody for Alzheimer's disease. Alzheimers Res Ther 2024; 16:235. [PMID: 39444037 PMCID: PMC11515656 DOI: 10.1186/s13195-024-01599-1] [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: 08/26/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Variants of the gene triggering receptor expressed on myeloid cells-2 (TREM2) increase the risk of Alzheimer's disease (AD) and other neurodegenerative disorders. Signaling by TREM2, an innate immune receptor expressed by microglia, is thought to enhance phagocytosis of amyloid beta (Aβ) and other damaged proteins, promote microglial proliferation, migration, and survival, and regulate inflammatory signaling. Thus, TREM2 activation has potential to alter the progression of AD. AL002 is an investigational, engineered, humanized monoclonal immunoglobulin G1 (IgG1) antibody designed to target TREM2. In AD mouse models, an AL002 murine variant has been previously shown to induce microglial proliferation and reduce filamentous Aβ plaques and neurite dystrophy. METHODS Preclinical studies assessed the safety, tolerability, pharmacokinetics, and pharmacodynamics of AL002 in cynomolgus monkeys. INVOKE-1 (NCT03635047) was a first-in-human phase 1, randomized, placebo-controlled, double-blind study assessing the safety, tolerability, PK, and PD of AL002 administered as single ascending doses (SAD) in healthy volunteers. RESULTS In cynomolgus monkeys, weekly intravenous injections of AL002 for 4 weeks were well tolerated, dose-dependently decreased soluble TREM2 (sTREM2) in cerebrospinal fluid (CSF) and total TREM2 in hippocampus and frontal cortex, and increased biomarkers of TREM2 signaling in CSF and brain. In the phase 1 study of 64 healthy volunteers, a single intravenous infusion of AL002 demonstrated brain target engagement based on a dose-dependent reduction of sTREM2 in CSF and parallel increases in biomarkers of TREM2 signaling and microglia recruitment. Single-dose AL002 showed central nervous system penetrance and was well tolerated, with no treatment-related serious adverse events over 12 weeks. CONCLUSIONS These findings support the continued clinical development of AL002 for AD and other neurodegenerative diseases in which TREM2 activation may be beneficial. AL002 is currently being tested in a phase 2, randomized, double-blind, placebo-controlled study in early AD. TRIAL REGISTRATION Clinicaltrials.gov, NCT03635047. Registered on August 15, 2018, https://www. CLINICALTRIALS gov/study/NCT03635047 .
Collapse
Affiliation(s)
- Hua Long
- Alector, Inc., South San Francisco, CA, 94080, USA
| | - Adam Simmons
- Alector, Inc., South San Francisco, CA, 94080, USA.
| | | | | | - Tuan Nguyen
- Alector, Inc., South San Francisco, CA, 94080, USA
| | | | | | - Herve Rhinn
- Alector, Inc., South San Francisco, CA, 94080, USA
- Leal Therapeutics, Worcester, MA, USA
| | - Ilaria Tassi
- Alector, Inc., South San Francisco, CA, 94080, USA
- Deep Apple Therapeutics, Inc., San Francisco, CA, USA
| | - Michael Ward
- Alector, Inc., South San Francisco, CA, 94080, USA
- Independent Consultant, San Francisco, CA, USA
| | - Felix Yeh
- Alector, Inc., South San Francisco, CA, 94080, USA
- Genentech, Inc., South San Francisco, CA, USA
| | - Tina Schwabe
- Alector, Inc., South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, USA
| | - Robert Paul
- Alector, Inc., South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
16
|
Noone DP, Isendoorn MME, Hamers SMWR, Keizer ME, Wulffelé J, van der Velden TT, Dijkstra DJ, Trouw LA, Filippov DV, Sharp TH. Structural basis for surface activation of the classical complement cascade by the short pentraxin C-reactive protein. Proc Natl Acad Sci U S A 2024; 121:e2404542121. [PMID: 39240968 PMCID: PMC11406272 DOI: 10.1073/pnas.2404542121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/04/2024] [Indexed: 09/08/2024] Open
Abstract
Human C-reactive protein (CRP) is a pentameric complex involved in immune defense and regulation of autoimmunity. CRP is also a therapeutic target, with both administration and depletion of serum CRP being pursued as a possible treatment for autoimmune and cardiovascular diseases, among others. CRP binds to phosphocholine (PC) moieties on membranes to activate the complement system via the C1 complex, but it is unknown how CRP, or any pentraxin, binds to C1. Here, we present a cryoelectron tomography (cryoET)-derived structure of CRP bound to PC ligands and the C1 complex. To gain control of CRP binding, a synthetic mimotope of PC was synthesized and used to decorate cell-mimetic liposome surfaces. Structure-guided mutagenesis of CRP yielded a fully active complex able to bind PC-coated liposomes that was ideal for cryoET and subtomogram averaging. In contrast to antibodies, which form Fc-mediated hexameric platforms to bind and activate the C1 complex, CRP formed rectangular platforms assembled from four laterally associated CRP pentamers that bind only four of the six available globular C1 head groups. Potential residues mediating lateral association of CRP were identified from interactions between unit cells in existing crystal structures, which rationalized previously unexplained mutagenesis data regarding CRP-mediated complement activation. The structure also enabled interpretation of existing biochemical data regarding interactions mediating C1 binding and identified additional residues for further mutagenesis studies. These structural data therefore provide a possible mechanism for regulation of complement by CRP, which limits complement progression and has consequences for how the innate immune system influences autoimmunity.
Collapse
Affiliation(s)
- Dylan P. Noone
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Marjolein M. E. Isendoorn
- Leiden Institute of Chemistry, Gorlaeus Laboratory, Leiden University, 2333 CCLeiden, The Netherlands
| | - Sebastiaan M. W. R. Hamers
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Mariska E. Keizer
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Jip Wulffelé
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Tijn T. van der Velden
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
| | - Douwe J. Dijkstra
- Department of Immunology, Leiden University Medical Center, 2333 ZALeiden, The Netherlands
| | - Leendert A. Trouw
- Department of Immunology, Leiden University Medical Center, 2333 ZALeiden, The Netherlands
| | - Dmitri V. Filippov
- Leiden Institute of Chemistry, Gorlaeus Laboratory, Leiden University, 2333 CCLeiden, The Netherlands
| | - Thomas H. Sharp
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RCLeiden, The Netherlands
- School of Biochemistry, University of Bristol, BristolBS8 1TD, United Kingdom
| |
Collapse
|
17
|
Ishikawa K, Suzuki H, Ohishi T, Li G, Tanaka T, Kawada M, Ohkoshi A, Kaneko MK, Katori Y, Kato Y. Anti-CD44 Variant 10 Monoclonal Antibody Exerts Antitumor Activity in Mouse Xenograft Models of Oral Squamous Cell Carcinomas. Int J Mol Sci 2024; 25:9190. [PMID: 39273139 PMCID: PMC11395228 DOI: 10.3390/ijms25179190] [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: 08/02/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
CD44 regulates cell adhesion, proliferation, survival, and stemness and has been considered a tumor therapy target. CD44 possesses the shortest CD44 standard (CD44s) and a variety of CD44 variant (CD44v) isoforms. Since the expression of CD44v is restricted in epithelial cells and carcinomas compared to CD44s, CD44v has been considered a promising target for monoclonal antibody (mAb) therapy. We previously developed an anti-CD44v10 mAb, C44Mab-18 (IgM, kappa), to recognize the variant exon 10-encoded region. In the present study, a mouse IgG2a version of C44Mab-18 (C44Mab-18-mG2a) was generated to evaluate the antitumor activities against CD44-positive cells compared with the previously established anti-pan CD44 mAb, C44Mab-46-mG2a. C44Mab-18-mG2a exhibited higher reactivity compared with C44Mab-46-mG2a to CD44v3-10-overexpressed CHO-K1 (CHO/CD44v3-10) and oral squamous cell carcinoma cell lines (HSC-2 and SAS) in flow cytometry. C44Mab-18-mG2a exerted a superior antibody-dependent cellular cytotoxicity (ADCC) against CHO/CD44v3-10. In contrast, C44Mab-46-mG2a showed a superior complement-dependent cytotoxicity (CDC) against CHO/CD44v3-10. A similar tendency was observed in ADCC and CDC against HSC-2 and SAS. Furthermore, administering C44Mab-18-mG2a or C44Mab-46-mG2a significantly suppressed CHO/CD44v3-10, HSC-2, and SAS xenograft tumor growth compared with the control mouse IgG2a. These results indicate that C44Mab-18-mG2a could be a promising therapeutic regimen for CD44v10-positive tumors.
Collapse
Affiliation(s)
- Kenichiro Ishikawa
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (A.O.); (Y.K.)
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, 18-24 Miyamoto, Numazu-shi 410-0301, Shizuoka, Japan;
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan;
| | - Guanjie Li
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan;
| | - Akira Ohkoshi
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (A.O.); (Y.K.)
| | - Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| | - Yukio Katori
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (A.O.); (Y.K.)
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| |
Collapse
|
18
|
Suzuki H, Ohishi T, Tanaka T, Kaneko MK, Kato Y. Anti-HER2 Cancer-Specific mAb, H 2Mab-250-hG 1, Possesses Higher Complement-Dependent Cytotoxicity than Trastuzumab. Int J Mol Sci 2024; 25:8386. [PMID: 39125956 PMCID: PMC11313270 DOI: 10.3390/ijms25158386] [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: 04/22/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Cancer-specific monoclonal antibodies (CasMabs) that recognize cancer-specific antigens with in vivo antitumor efficacy are innovative therapeutic strategies for minimizing adverse effects. We previously established a cancer-specific anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody (mAb), H2Mab-250/H2CasMab-2. In flow cytometry and immunohistochemistry, H2Mab-250 reacted with HER2-positive breast cancer cells but did not show reactivity to normal epithelial cells. In contrast, a clinically approved anti-HER2 mAb, trastuzumab, strongly recognizes both breast cancer and normal epithelial cells in flow cytometry. The human IgG1 version of H2Mab-250 (H2Mab-250-hG1) possesses compatible in vivo antitumor effects against breast cancer xenografts to trastuzumab despite the lower affinity and effector activation than trastuzumab in vitro. This study compared the antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cellular cytotoxicity (CDC) between H2Mab-250-hG1 and trastuzumab. Both H2Mab-250-hG1 and trastuzumab showed ADCC activity against HER2-overexpressed Chinese hamster ovary -K1 and breast cancer cell lines (BT-474 and SK-BR-3) in the presence of human natural killer cells. Some tendency was observed where trastuzumab showed a more significant ADCC effect compared to H2Mab-250-hG1. Importantly, H2Mab-250-hG1 exhibited superior CDC activity in these cells compared to trastuzumab. Similar results were obtained in the mouse IgG2a types of both H2Mab-250 and trastuzumab. These results suggest the different contributions of ADCC and CDC activities to the antitumor effects of H2Mab-250-hG1 and trastuzumab, and indicate a future direction for the clinical development of H2Mab-250-hG1 against HER2-positive tumors.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, 18-24 Miyamoto, Numazu-shi, Shizuoka 410-0301, Japan;
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| | - Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| |
Collapse
|
19
|
Hamers SMWR, Abendstein L, Boyle AL, Jongkees SAK, Sharp TH. Selection and characterization of a peptide-based complement modulator targeting C1 of the innate immune system. RSC Chem Biol 2024; 5:787-799. [PMID: 39092440 PMCID: PMC11289891 DOI: 10.1039/d4cb00081a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/24/2024] [Indexed: 08/04/2024] Open
Abstract
The human complement pathway plays a pivotal role in immune defence, homeostasis, and autoimmunity regulation, and complement-based therapeutics have emerged as promising interventions, with both antagonistic and agonistic approaches being explored. The classical pathway of complement is initiated when the C1 complex binds to hexameric antibody platforms. Recent structural data revealed that C1 binds to small, homogeneous interfaces at the periphery of the antibody platforms. Here, we have developed a novel strategy for complement activation using macrocyclic peptides designed to mimic the interface between antibodies and the C1 complex. In vitro selection utilizing the RaPID system identified a cyclic peptide (cL3) that binds to the C1 complex via the globular head domains of C1q. Notably, when immobilized on surfaces, cL3 effectively recruits C1 from human serum, activates C1s proteases, and induces lysis of cell-mimetic lipid membranes. This represents the first instance of a peptide capable of activating complement by binding C1 when immobilized. Further characterization and synthesis of deletion mutants revealed a critical cycle size of cL3 essential for C1 binding and efficient complement activation. Importantly, cL3 also demonstrated the ability to inhibit complement-mediated lysis without affecting C1 binding, highlighting its potential as a therapeutic modality to prevent complement-dependent cytotoxicity whilst promoting cellular phagocytosis and cell clearance. In summary, this study introduces the concept of "Peptactins" - peptide-based activators of complement - and underscores the potential of macrocyclic peptides for complement modulation, offering potential advantages over traditional biologicals in terms of size, production, and administration.
Collapse
Affiliation(s)
- Sebastiaan M W R Hamers
- Department of Cell and Chemical Biology, Leiden University Medical Centre 2300 RC Leiden The Netherlands
| | - Leoni Abendstein
- Department of Cell and Chemical Biology, Leiden University Medical Centre 2300 RC Leiden The Netherlands
| | - Aimee L Boyle
- Leiden Institute of Chemistry, Leiden University 2333 CC Leiden The Netherlands
- School of Chemistry, University of Bristol Bristol BS8 1QU UK
| | - Seino A K Jongkees
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam 1081 HV Amsterdam The Netherlands
| | - Thomas H Sharp
- Department of Cell and Chemical Biology, Leiden University Medical Centre 2300 RC Leiden The Netherlands
- School of Biochemistry, University of Bristol Bristol BS8 1TD UK
| |
Collapse
|
20
|
Hamers SMWR, Boyle AL, Sharp TH. Engineering Agonistic Bispecifics to Investigate the Influence of Distance on Surface-Mediated Complement Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:235-243. [PMID: 38819221 PMCID: PMC11215631 DOI: 10.4049/jimmunol.2400091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/12/2024] [Indexed: 06/01/2024]
Abstract
The development of agonists capable of activating the human complement system by binding to the C1 complex presents a novel approach for targeted cell killing. Bispecific nanobodies and Abs can successfully use C1 for this purpose; however, efficacy varies significantly between epitopes, Ab type, and bispecific design. To address this variability, we investigated monomeric agonists of C1 in the form of bispecific nanobodies, which lack Fc domains that lead to oligomerization in Abs. These therefore offer an ideal opportunity to explore the geometric parameters crucial for C1 activation. In this study, we explored the impact of linker length as a metric for Ag and epitope location. DNA nanotechnology and protein engineering allowed us to design linkers with controlled lengths and flexibilities, revealing a critical range of end-to-end distances for optimal complement activation. We discovered that differences in complement activation were not caused by differential C1 activation or subsequent cleavage of C4, but instead impacted C4b deposition and downstream membrane lysis. Considering the importance of Ab class and subclass, this study provides insights into the structural requirements of C1 binding and activation, highlighting linker and hinge engineering as a potential strategy to enhance potency over specific cellular targets. Additionally, using DNA nanotechnology to modify geometric parameters demonstrated the potential for synthetic biology in complement activation. Overall, this research offers valuable insights into the design and optimization of agonists for targeted cell killing through complement activation.
Collapse
Affiliation(s)
| | - Aimee L. Boyle
- Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
- School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Thomas H. Sharp
- Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, the Netherlands
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
21
|
Bobrowicz M, Kusowska A, Krawczyk M, Zhylko A, Forcados C, Slusarczyk A, Barankiewicz J, Domagala J, Kubacz M, Šmída M, Dostalova L, Marhelava K, Fidyt K, Pepek M, Baranowska I, Szumera-Cieckiewicz A, Inderberg EM, Wälchli S, Granica M, Graczyk-Jarzynka A, Majchrzak M, Poreba M, Gehlert CL, Peipp M, Firczuk M, Prochorec-Sobieszek M, Winiarska M. CD20 expression regulates CD37 levels in B-cell lymphoma - implications for immunotherapies. Oncoimmunology 2024; 13:2362454. [PMID: 38846084 PMCID: PMC11155707 DOI: 10.1080/2162402x.2024.2362454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Rituximab (RTX) plus chemotherapy (R-CHOP) applied as a first-line therapy for lymphoma leads to a relapse in approximately 40% of the patients. Therefore, novel approaches to treat aggressive lymphomas are being intensively investigated. Several RTX-resistant (RR) cell lines have been established as surrogate models to study resistance to R-CHOP. Our study reveals that RR cells are characterized by a major downregulation of CD37, a molecule currently explored as a target for immunotherapy. Using CD20 knockout (KO) cell lines, we demonstrate that CD20 and CD37 form a complex, and hypothesize that the presence of CD20 stabilizes CD37 in the cell membrane. Consequently, we observe a diminished cytotoxicity of anti-CD37 monoclonal antibody (mAb) in complement-dependent cytotoxicity in both RR and CD20 KO cells that can be partially restored upon lysosome inhibition. On the other hand, the internalization rate of anti-CD37 mAb in CD20 KO cells is increased when compared to controls, suggesting unhampered efficacy of antibody drug conjugates (ADCs). Importantly, even a major downregulation in CD37 levels does not hamper the efficacy of CD37-directed chimeric antigen receptor (CAR) T cells. In summary, we present here a novel mechanism of CD37 regulation with further implications for the use of anti-CD37 immunotherapies.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antigens, CD20/immunology
- Antigens, CD20/metabolism
- Antigens, CD20/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cell Line, Tumor
- Cyclophosphamide/pharmacology
- Cyclophosphamide/therapeutic use
- Doxorubicin/pharmacology
- Doxorubicin/administration & dosage
- Drug Resistance, Neoplasm/drug effects
- Gene Expression Regulation, Neoplastic
- Immunotherapy/methods
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/drug therapy
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Rituximab/pharmacology
- Rituximab/therapeutic use
- Tetraspanins/genetics
- Tetraspanins/metabolism
- Vincristine/pharmacology
- Vincristine/therapeutic use
Collapse
Affiliation(s)
| | - Aleksandra Kusowska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Doctoral School, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Marta Krawczyk
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Doctoral School of Translational Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Andriy Zhylko
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Doctoral School, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Christopher Forcados
- Translational Research Unit, Department of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Aleksander Slusarczyk
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Barankiewicz
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
- Faculty of Medicine, Lazarski University, Warsaw, Poland
| | - Joanna Domagala
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Matylda Kubacz
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Michal Šmída
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Lenka Dostalova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | | | - Klaudyna Fidyt
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Monika Pepek
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Iwona Baranowska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Szumera-Cieckiewicz
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Biobank, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Else Marit Inderberg
- Translational Research Unit, Department of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Department of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Monika Granica
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Doctoral School, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Graczyk-Jarzynka
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Martyna Majchrzak
- Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Marcin Poreba
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
- Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Carina Lynn Gehlert
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Matthias Peipp
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Malgorzata Firczuk
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Monika Prochorec-Sobieszek
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Magdalena Winiarska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
22
|
La Guidara C, Adamo R, Sala C, Micoli F. Vaccines and Monoclonal Antibodies as Alternative Strategies to Antibiotics to Fight Antimicrobial Resistance. Int J Mol Sci 2024; 25:5487. [PMID: 38791526 PMCID: PMC11122364 DOI: 10.3390/ijms25105487] [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: 03/30/2024] [Revised: 05/04/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Antimicrobial resistance (AMR) is one of the most critical threats to global public health in the 21st century, causing a large number of deaths every year in both high-income and low- and middle-income countries. Vaccines and monoclonal antibodies can be exploited to prevent and treat diseases caused by AMR pathogens, thereby reducing antibiotic use and decreasing selective pressure that favors the emergence of resistant strains. Here, differences in the mechanism of action and resistance of vaccines and monoclonal antibodies compared to antibiotics are discussed. The state of the art for vaccine technologies and monoclonal antibodies are reviewed, with a particular focus on approaches validated in clinical studies. By underscoring the scope and limitations of the different emerging technologies, this review points out the complementary of vaccines and monoclonal antibodies in fighting AMR. Gaps in antigen discovery for some pathogens, as well as challenges associated with the clinical development of these therapies against AMR pathogens, are highlighted.
Collapse
Affiliation(s)
- Chiara La Guidara
- Magnetic Resonance Center CERM, University of Florence, 50019 Florence, Italy
- Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Florence, Italy
| | | | - Claudia Sala
- Monoclonal Antibody Discovery Laboratory, Fondazione Toscana Life Sciences, 53100 Siena, Italy
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health S.R.L. (GVGH), 53100 Siena, Italy
| |
Collapse
|
23
|
Abendstein L, Noteborn WEM, Veenman LS, Dijkstra DJ, van de Bovenkamp FS, Trouw LA, Sharp TH. DNA Nanostructure-Templated Antibody Complexes Provide Insights into the Geometric Requirements of Human Complement Cascade Activation. J Am Chem Soc 2024; 146:13455-13466. [PMID: 38703132 PMCID: PMC11099972 DOI: 10.1021/jacs.4c02772] [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: 02/24/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024]
Abstract
The classical complement pathway is activated by antigen-bound IgG antibodies. Monomeric IgG must oligomerize to activate complement via the hexameric C1q complex, and hexamerizing mutants of IgG appear as promising therapeutic candidates. However, structural data have shown that it is not necessary to bind all six C1q arms to initiate complement, revealing a symmetry mismatch between C1 and the hexameric IgG complex that has not been adequately explained. Here, we use DNA nanotechnology to produce specific nanostructures to template antigens and thereby spatially control IgG valency. These DNA-nanotemplated IgG complexes can activate complement on cell-mimetic lipid membranes, which enabled us to determine the effect of IgG valency on complement activation without the requirement to mutate antibodies. We investigated this using biophysical assays together with 3D cryo-electron tomography. Our data revealed the importance of interantigen distance on antibody-mediated complement activation, and that the cleavage of complement component C4 by the C1 complex is proportional to the number of ideally spaced antigens. Increased IgG valency also translated to better terminal pathway activation and membrane attack complex formation. Together, these data provide insights into how nanopatterning antigen-antibody complexes influence the activation of the C1 complex and suggest routes to modulate complement activation by antibody engineering. Furthermore, to our knowledge, this is the first time DNA nanotechnology has been used to study the activation of the complement system.
Collapse
Affiliation(s)
- Leoni Abendstein
- Department
of Cell and Chemical Biology, Leiden University
Medical Center, Leiden 2300 RC, The Netherlands
| | - Willem E. M. Noteborn
- Department
of Cell and Chemical Biology, Leiden University
Medical Center, Leiden 2300 RC, The Netherlands
| | - Luc S. Veenman
- Department
of Cell and Chemical Biology, Leiden University
Medical Center, Leiden 2300 RC, The Netherlands
| | - Douwe J. Dijkstra
- Department
of Immunology, Leiden University Medical
Center, Leiden 2333 ZA, The Netherlands
| | | | - Leendert A. Trouw
- Department
of Immunology, Leiden University Medical
Center, Leiden 2333 ZA, The Netherlands
| | - Thomas H. Sharp
- Department
of Cell and Chemical Biology, Leiden University
Medical Center, Leiden 2300 RC, The Netherlands
- School
of Biochemistry, University of Bristol, Bristol BS8 1TD, U.K.
| |
Collapse
|
24
|
Sandeep, Shinde SH, Ahmed S, Sharma SS, Pande AH. Engineered polyspecific antibodies: A new frontier in the field of immunotherapeutics. Immunology 2024; 171:464-496. [PMID: 38140855 DOI: 10.1111/imm.13743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
The 21st-century beginning remarked with the huge success of monospecific MAbs, however, in the last couple of years, polyspecific MAbs (PsAbs) have been an interesting topic and show promise of being biobetter than monospecific MAbs. Polyspecificity, in which a single antibody serves multiple specific target binding, has been hypothesized to contribute to the development of a highly effective antibody repertoire for immune defence. This polyspecific MAb trend represents an explosion that is gripping the whole pharmaceutical industry. This review is concerned with the current development and quality enforcement of PsAbs. All provided literature on monospecific MAbs and polyspecific MAbs (PsAbs) were searched using various electronic databases such as PubMed, Google Scholar, Web of Science, Elsevier, Springer, ACS, Google Patent and books via the keywords Antibody engineering, Polyspecific antibody, Conventional antibody, non-conventional antibody, and Single domain antibody. In the literature, there are more than 100 different formats to construct PsAb by quadroma technology, chemical conjugation and genetic engineering. Till March 2023, nine PsAb have been approved around the world, and around 330 are in advanced developmental stages, showing the dominancy of PsAb in the growing health sector. Recent advancements in protein engineering techniques and the fusion of non-conventional antibodies have made it possible to create complex PsAbs that demonstrate higher stability and enhanced potency. This marks the most significant achievement for cancer immunotherapy, in which PsAbs have immense promise. It is worth mentioning that seven out of the nine PsAbs have been approved as anti-cancer therapy. As PsAbs continue to acquire prominence, they could pave the way for the development of novel immunotherapies for multiple diseases.
Collapse
Affiliation(s)
- Sandeep
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Suraj H Shinde
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Sakeel Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| |
Collapse
|
25
|
Cleary SJ, Seo Y, Tian JJ, Kwaan N, Bulkley DP, Bentlage AE, Vidarsson G, Boilard É, Spirig R, Zimring JC, Looney MR. IgG hexamers initiate complement-dependent acute lung injury. J Clin Invest 2024; 134:e178351. [PMID: 38530369 PMCID: PMC11142733 DOI: 10.1172/jci178351] [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: 12/15/2023] [Accepted: 03/20/2024] [Indexed: 03/28/2024] Open
Abstract
Antibodies can initiate lung injury in a variety of disease states such as autoimmunity, in reactions to transfusions, or after organ transplantation, but the key factors determining in vivo pathogenicity of injury-inducing antibodies are unclear. Harmful antibodies often activate the complement cascade. A model for how IgG antibodies trigger complement activation involves interactions between IgG Fc domains driving the assembly of IgG hexamer structures that activate C1 complexes. The importance of IgG hexamers in initiating injury responses was not clear, so we tested their relevance in a mouse model of alloantibody- and complement-mediated acute lung injury. We used 3 approaches to block alloantibody hexamerization (antibody carbamylation, the K439E Fc mutation, or treatment with domain B from staphylococcal protein A), all of which reduced acute lung injury. Conversely, Fc mutations promoting spontaneous hexamerization made a harmful alloantibody into a more potent inducer of acute lung injury and rendered an innocuous alloantibody pathogenic. Treatment with a recombinant Fc hexamer "decoy" therapeutic protected mice from lung injury, including in a model with transgenic human FCGR2A expression that exacerbated pathology. These results indicate an in vivo role of IgG hexamerization in initiating acute lung injury and the potential for therapeutics that inhibit or mimic hexamerization to treat antibody-mediated diseases.
Collapse
Affiliation(s)
| | | | | | | | - David P. Bulkley
- Department of Biochemistry and Biophysics, UCSF, San Francisco, California, USA
| | | | | | - Éric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec – Université Laval, Québec, Quebec, Canada
| | - Rolf Spirig
- CSL Behring, Research, CSL Behring Biologics Research Center, Bern, Switzerland
| | - James C. Zimring
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | |
Collapse
|
26
|
Foss S, Sakya SA, Aguinagalde L, Lustig M, Shaughnessy J, Cruz AR, Scheepmaker L, Mathiesen L, Ruso-Julve F, Anthi AK, Gjølberg TT, Mester S, Bern M, Evers M, Bratlie DB, Michaelsen TE, Schlothauer T, Sok D, Bhattacharya J, Leusen J, Valerius T, Ram S, Rooijakkers SHM, Sandlie I, Andersen JT. Human IgG Fc-engineering for enhanced plasma half-life, mucosal distribution and killing of cancer cells and bacteria. Nat Commun 2024; 15:2007. [PMID: 38453922 PMCID: PMC10920689 DOI: 10.1038/s41467-024-46321-9] [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: 04/07/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Monoclonal IgG antibodies constitute the fastest growing class of therapeutics. Thus, there is an intense interest to design more potent antibody formats, where long plasma half-life is a commercially competitive differentiator affecting dosing, frequency of administration and thereby potentially patient compliance. Here, we report on an Fc-engineered variant with three amino acid substitutions Q311R/M428E/N434W (REW), that enhances plasma half-life and mucosal distribution, as well as allows for needle-free delivery across respiratory epithelial barriers in human FcRn transgenic mice. In addition, the Fc-engineered variant improves on-target complement-mediated killing of cancer cells as well as both gram-positive and gram-negative bacteria. Hence, this versatile Fc technology should be broadly applicable in antibody design aiming for long-acting prophylactic or therapeutic interventions.
Collapse
Affiliation(s)
- Stian Foss
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Siri A Sakya
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Leire Aguinagalde
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marta Lustig
- Section for Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Jutamas Shaughnessy
- Department of Medicine, Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ana Rita Cruz
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lisette Scheepmaker
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Line Mathiesen
- Department of Public Health, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fulgencio Ruso-Julve
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Aina Karen Anthi
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Torleif Tollefsrud Gjølberg
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Simone Mester
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Malin Bern
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Mitchell Evers
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Diane B Bratlie
- Infection Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Terje E Michaelsen
- Infection Immunology, Norwegian Institute of Public Health, Oslo, Norway
- Department of Chemical Pharmacy, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Munich, Germany
| | - Devin Sok
- International AIDS Vaccine Initiative (IAVI), New York, NY, USA
| | - Jayanta Bhattacharya
- Antibody Translational Research Program, Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Jeanette Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Thomas Valerius
- Section for Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Sanjay Ram
- Department of Medicine, Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Suzan H M Rooijakkers
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway.
| |
Collapse
|
27
|
Cleary SJ, Seo Y, Tian JJ, Kwaan N, Bulkley DP, Bentlage AEH, Vidarsson G, Boilard É, Spirig R, Zimring JC, Looney MR. IgG hexamers initiate acute lung injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.577129. [PMID: 38328049 PMCID: PMC10849723 DOI: 10.1101/2024.01.24.577129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Antibodies can initiate lung injury in a variety of disease states such as autoimmunity, transfusion reactions, or after organ transplantation, but the key factors determining in vivo pathogenicity of injury-inducing antibodies are unclear. A previously overlooked step in complement activation by IgG antibodies has been elucidated involving interactions between IgG Fc domains that enable assembly of IgG hexamers, which can optimally activate the complement cascade. Here, we tested the in vivo relevance of IgG hexamers in a complement-dependent alloantibody model of acute lung injury. We used three approaches to block alloantibody hexamerization (antibody carbamylation, the K439E Fc mutation, or treatment with domain B from Staphylococcal protein A), all of which reduced acute lung injury. Conversely, Fc mutations promoting spontaneous hexamerization made a harmful alloantibody into a more potent inducer of acute lung injury and rendered an innocuous alloantibody pathogenic. Treatment with a recombinant Fc hexamer 'decoy' therapeutic protected mice from lung injury, including in a model with transgenic human FCGR2A expression that exacerbated pathology. These results indicate a direct in vivo role of IgG hexamerization in initiating acute lung injury and the potential for therapeutics that inhibit or mimic hexamerization to treat antibody-mediated diseases.
Collapse
Affiliation(s)
- Simon J. Cleary
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Yurim Seo
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Jennifer J. Tian
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Nicholas Kwaan
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - David P. Bulkley
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), CA, USA
| | | | | | - Éric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Rolf Spirig
- CSL Behring, Research, CSL Behring Biologics Research Center, Bern, Switzerland
| | - James C. Zimring
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Cvijić T, Horvat M, Plahutnik J, Golob A, Marušič J. Multivariate quantitative analysis of glycan impact on IgG1 effector functions. MAbs 2024; 16:2430295. [PMID: 39572418 PMCID: PMC11587841 DOI: 10.1080/19420862.2024.2430295] [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: 08/14/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024] Open
Abstract
Development of novel therapeutic proteins and biosimilars requires a thorough understanding of the relationship between their structure and function. Particularly, how IgG glycosylation affects its effector functions is a point increasingly underscored in guidelines by the World Health Organization and regulatory agencies. Our results show that just a 1% decrease in Fc fucosylation can lead to a more than 25% increase in antibody-dependent cell-mediated cytotoxicity. The intercorrelated nature of glycan patterns, combined with the low variability and lack of well-defined glycan patterns in process development and manufacture samples, makes studying the effects of individual glycan structures challenging. The conventional approach to structure-function studies often relies on a suboptimal set of tools, such as the one-factor-at-a-time method for experimental planning and univariate data analysis. Here, we introduce a systematic approach to understanding and prediction of the impact of Fc glycans on effector functions, using a combination of the design of experiment, multivariate data analysis, and in-vitro glycoengineering. This approach adheres to quality-by-design principles and aligns with regulatory agency guidelines. A variety of analytical assays, including binding and cell-based assays, were applied to investigate the effect of individual glycans of the IgG1 molecule. The regression models developed here provide a quantitative explanation and prediction of the impact of individual glycan features on the binding to FcγRs and bioactivity of the therapeutic protein. To the best of our knowledge, this is the first report of a systematic approach to quantitatively understand the multivariate impact of glycosylation on the effector functionality of therapeutic monoclonal antibodies, providing valuable tools for advancing therapeutic protein development.
Collapse
Affiliation(s)
- Tamara Cvijić
- Lek d.d. Part of Sandoz, Biopharma Technical Development, Ljubljana, Slovenia
- Biotechnical faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Matej Horvat
- Lek d.d. Part of Sandoz, Biopharma Technical Development, Ljubljana, Slovenia
| | - Jakob Plahutnik
- Lek d.d. Part of Sandoz, Biopharma Technical Development, Ljubljana, Slovenia
| | - Ana Golob
- Lek d.d. Part of Sandoz, Biopharma Technical Development, Ljubljana, Slovenia
| | - Jaka Marušič
- Lek d.d. Part of Sandoz, Biopharma Technical Development, Ljubljana, Slovenia
| |
Collapse
|
31
|
Damelang T, de Taeye SW, Rentenaar R, Roya-Kouchaki K, de Boer E, Derksen NIL, van Kessel K, Lissenberg-Thunnissen S, Rooijakkers SHM, Jongerius I, Mebius MM, Schuurman J, Labrijn AF, Vidarsson G, Rispens T. The Influence of Human IgG Subclass and Allotype on Complement Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1725-1735. [PMID: 37843500 PMCID: PMC10656437 DOI: 10.4049/jimmunol.2300307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
Complement activation via the classical pathway is initiated when oligomeric Igs on target surfaces are recognized by C1 of the complement cascade. The strength of this interaction and activation of the complement system are influenced by structural variation of the Ab, including Ab isotype, subclass, and glycosylation profile. Polymorphic variants of IgG have also been described to influence Fc-dependent effector functions. Therefore, we assessed complement binding, deposition, and complement-dependent cytotoxicity (CDC) of 27 known IgG allotypes with anti-trinitrophenyl specificity. Differences between allotypes within subclasses were minor for IgG1, IgG3, and IgG4 allotypes, and more substantial for IgG2. Allelic variant IGHG2*06, containing a unique serine at position 378 in the CH3 domain, showed less efficient complement activation and CDC compared with other IgG2 polymorphisms. We also observed variable cell lysis between IgG1 and IgG3, with IgG3 being superior in lysis of human RBCs and Ramos cells, and IgG1 being superior in lysis of Raji and Wien133 cells, demonstrating that a long-standing conundrum in the literature depends on cellular context. Furthermore, we compared IgG1 and IgG3 under different circumstances, showing that Ag density and Ab hinge length, but not complement regulators, define the context dependency of Ab-mediated CDC activity. Our results point toward a variation in the capacity of IgG subclasses to activate complement due to single amino acid changes and hinge length differences of allotypes to activate complement, which might give new insights on susceptibility to infectious, alloimmune, or autoimmune diseases and aid the design of Ab-based therapeutics.
Collapse
Affiliation(s)
- Timon Damelang
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Steven W. de Taeye
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
| | - Rosa Rentenaar
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kasra Roya-Kouchaki
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
| | - Esther de Boer
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ninotska I. L. Derksen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kok van Kessel
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Suzan H. M. Rooijakkers
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ilse Jongerius
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | | | | | | | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
32
|
Bauer-Smith H, Sudol ASL, Beers SA, Crispin M. Serum immunoglobulin and the threshold of Fc receptor-mediated immune activation. Biochim Biophys Acta Gen Subj 2023; 1867:130448. [PMID: 37652365 PMCID: PMC11032748 DOI: 10.1016/j.bbagen.2023.130448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Antibodies can mediate immune recruitment or clearance of immune complexes through the interaction of their Fc domain with cellular Fc receptors. Clustering of antibodies is a key step in generating sufficient avidity for efficacious receptor recognition. However, Fc receptors may be saturated with prevailing, endogenous serum immunoglobulin and this raises the threshold by which cellular receptors can be productively engaged. Here, we review the factors controlling serum IgG levels in both healthy and disease states, and discuss how the presence of endogenous IgG is encoded into the functional activation thresholds for low- and high-affinity Fc receptors. We discuss the circumstances where antibody engineering can help overcome these physiological limitations of therapeutic antibodies. Finally, we discuss how the pharmacological control of Fc receptor saturation by endogenous IgG is emerging as a feasible mechanism for the enhancement of antibody therapeutics.
Collapse
Affiliation(s)
- Hannah Bauer-Smith
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK; Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Abigail S L Sudol
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Stephen A Beers
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK.
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
33
|
Muts RM, den Boer MA, Bardoel BW, Aerts PC, de Haas CJC, Heck AJR, Rooijakkers SHM, Heesterbeek DAC. Artificial surface labelling of Escherichia coli with StrepTagII antigen to study how monoclonal antibodies drive complement-mediated killing. Sci Rep 2023; 13:18836. [PMID: 37914798 PMCID: PMC10620216 DOI: 10.1038/s41598-023-46026-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023] Open
Abstract
Antibodies play a key role in the immune defence against Gram-negative bacteria. After binding to bacterial surface antigens, IgG and IgM can activate the complement system and trigger formation of lytic membrane attack complex (MAC) pores. Molecular studies to compare functional activity of antibodies on bacteria are hampered by the limited availability of well-defined antibodies against bacterial surface antigens. Therefore, we genetically engineered E. coli by expressing the StrepTagII antigen into outer membrane protein X (OmpX) and validated that these engineered bacteria were recognised by anti-StrepTagII antibodies. We then combined this antigen-antibody system with a purified complement assay to avoid interference of serum components and directly compare MAC-mediated bacterial killing via IgG1 and pentameric IgM. While both IgG1 and IgM could induce MAC-mediated killing, we show that IgM has an increased capacity to induce complement-mediated killing of E. coli compared to IgG1. While Fc mutations that enhance IgG clustering after target binding could not improve MAC formation, mutations that cause formation of pre-assembled IgG hexamers enhanced the complement activating capacity of IgG1. Altogether, we here present a system to study antibody-dependent complement activation on E. coli and show IgM's enhanced capacity over IgG to induce complement-mediated lysis of E. coli.
Collapse
Affiliation(s)
- Remy M Muts
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Maurits A den Boer
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584 CH, Utrecht, The Netherlands
- Netherlands Proteomic Center, 3584 CH, Utrecht, The Netherlands
| | - Bart W Bardoel
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Piet C Aerts
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Carla J C de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584 CH, Utrecht, The Netherlands
- Netherlands Proteomic Center, 3584 CH, Utrecht, The Netherlands
| | - Suzan H M Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Dani A C Heesterbeek
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
34
|
Lee SM, Min SW, Kwon HS, Bae GD, Jung JH, Park HI, Lee SH, Lim CS, Ko BJ, Lee JC, Jung ST. Effective clearance of rituximab-resistant tumor cells by breaking the mirror-symmetry of immunoglobulin G and simultaneous binding to CD55 and CD20. Sci Rep 2023; 13:18275. [PMID: 37880350 PMCID: PMC10600224 DOI: 10.1038/s41598-023-45491-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023] Open
Abstract
Complement-dependent cytotoxicity (CDC), which eliminates aberrant target cells through the assembly and complex formation of serum complement molecules, is one of the major effector functions of anticancer therapeutic antibodies. In this study, we discovered that breaking the symmetry of natural immunoglobulin G (IgG) antibodies significantly increased the CDC activity of anti-CD20 antibodies. In addition, the expression of CD55 (a checkpoint inhibitor in the CDC cascade) was significantly increased in a rituximab-resistant cell line generated in-house, suggesting that CD55 overexpression might be a mechanism by which cancer cells acquire rituximab resistance. Based on these findings, we developed an asymmetric bispecific antibody (SBU-CD55 × CD20) that simultaneously targets both CD55 and CD20 to effectively eliminate rituximab-resistant cancer cells. In various cancer cell lines, including rituximab-resistant lymphoma cells, the SBU-CD55 × CD20 antibody showed significantly higher CDC activity than either anti-CD20 IgG antibody alone or a combination of anti-CD20 IgG antibody and anti-CD55 IgG antibody. Furthermore, the asymmetric bispecific antibody (SBU-CD55 × CD20) exhibited significantly higher CDC activity against rituximab-resistant cancer cells compared to other bispecific antibodies with symmetric features. These results demonstrate that enhancing CDC with an asymmetric CD55-binding bispecific antibody could be a new strategy for developing therapeutics to treat patients with relapsed or refractory cancers.
Collapse
Affiliation(s)
- Sang Min Lee
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Department of Applied Chemistry, Kookmin University, 77, Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea
| | - Sung-Won Min
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Hyeong Sun Kwon
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Gong-Deuk Bae
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Ji Hae Jung
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Hye In Park
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Seung Hyeon Lee
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Chung Su Lim
- New Drug Development Center, Osong Medical Innovation Foundation 123, Cheongju, Chungcheongbuk-do, 28160, Republic of Korea
| | - Byoung Joon Ko
- School of Biopharmaceutical and Medical Science, Sungshin Women's University, 55, Dobonng-Ro 76ga-gil, Gangbuk, Seoul, 01133, Republic of Korea
| | - Ji Chul Lee
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea.
| | - Sang Taek Jung
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.
- Institute of Human Genetics, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
- Biomedical Research Center, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| |
Collapse
|
35
|
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
|
36
|
Lee W, Lee SM, Jung ST. Unlocking the Power of Complement-Dependent Cytotoxicity: Engineering Strategies for the Development of Potent Therapeutic Antibodies for Cancer Treatments. BioDrugs 2023; 37:637-648. [PMID: 37486566 DOI: 10.1007/s40259-023-00618-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
The complement system is a crucial part of the innate immune response, providing defense against invading pathogens and cancer cells. Recently, it has become evident that the complement system plays a significant role in anticancer activities, particularly through complement-dependent cytotoxicity (CDC), alongside antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cell-mediated phagocytosis (ADCP). With the discovery of new roles for serum complement molecules in the human immune system, various approaches are being pursued to develop CDC-enhanced antibody therapeutics. In this review, we focus on successful antibody engineering strategies for enhancing CDC, analyzing the lessons learned and the limitations of each approach. Furthermore, we outline potential pathways for the development of antibody therapeutics specifically aimed at enhancing CDC for superior therapeutic efficacy in the future.
Collapse
Affiliation(s)
- Wonju Lee
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sang Min Lee
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Applied Chemistry, Kookmin University, Seoul, 02707, Republic of Korea
| | - Sang Taek Jung
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.
- Biomedical Research Center, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| |
Collapse
|
37
|
She T, Yang F, Chen S, Yang H, Tao Z, Xing H, Chen J, Chang H, Lu H, Su T, Jin Y, Zhong Y, Cheng J, Zhu H, Lu X. Snoopligase-catalyzed molecular glue enables efficient generation of hyperoligomerized TRAIL variant with enhanced antitumor effect. J Control Release 2023; 361:856-870. [PMID: 37516318 DOI: 10.1016/j.jconrel.2023.07.042] [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: 04/14/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Clinical application of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is predominantly limited by its inefficient apoptosis induction in tumor cells, which might be improved by using molecular superglue-mediated hyperoligomerization to increase its valency. Here, the minimal superglue peptide pairs, including Snoopligase-catalyzed SnoopTagJr/SnoopDogTag and SpyStapler-catalyzed SpyTag/SpyBDTag, were individually fused at the N- or C-terminus of the TRAIL promoter to produce superglue-fusion TRAIL variants. Similar to native trivalent TRAIL, these superglue-fusion TRAIL variants were highly expressed in Escherichia coli (E. coli) and spontaneously trimerized. In the presence of Snoopligase or SpyStapler, the trivalent superglue-fusion TRAIL variants were predominantly crosslinked into hexavalent TRAIL variants. Nevertheless, Snoopligase was more efficient than SpyStapler in the production of hexavalent TRAIL variants. In particular, Snoopligase-catalyzed trivalent TRAIL variants with N-terminal fusion of SnoopTagJr/SnoopDogTag produced hexavalent SnHexaTR with the highest yield (∼70%). The in vitro cytotoxicity of SnHexaTR was 10-40 times greater than that of TRAIL in several tumor cells. In addition, compared to trivalent TRAIL, hexavalent SnHexaTR showed a longer serum half-life and greater tumor uptake, which resulted in eradication of 50% of tumor xenografts of TRAIL-sensitive COLO 205. In mice bearing TRAIL-resistant HT-29 tumor xenografts, hexavalent SnHexaTR combined with bortezomib encapsulated in liposomes also showed robust tumor growth suppression, indicating that hyperoligomerization mediated by minimal molecular superglue significantly increased the cytotoxicity and antitumor effect of TRAIL. As a novel anticancer agent candidate, the hexavalent SnHexaTR has great potential for clinical application in cancer therapy.
Collapse
Affiliation(s)
- Tianshan She
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fen Yang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiyuan Chen
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Yang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ze Tao
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huimin Xing
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Chen
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huansheng Chang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyu Lu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Su
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youmei Jin
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Zhong
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingqiu Cheng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaofeng Lu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
38
|
Hiemstra IH, Santegoets KCM, Janmaat ML, De Goeij BECG, Ten Hagen W, van Dooremalen S, Boross P, van den Brakel J, Bosgra S, Andringa G, van Kessel-Welmers B, Verzijl D, Hibbert RG, Frerichs KA, Mutis T, van de Donk NWCJ, Ahmadi T, Satijn D, Sasser AK, Breij ECW. Preclinical anti-tumour activity of HexaBody-CD38, a next-generation CD38 antibody with superior complement-dependent cytotoxic activity. EBioMedicine 2023; 93:104663. [PMID: 37379657 DOI: 10.1016/j.ebiom.2023.104663] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND HexaBody®-CD38 (GEN3014) is a hexamerization-enhanced human IgG1 that binds CD38 with high affinity. The E430G mutation in its Fc domain facilitates the natural process of antibody hexamer formation upon binding to the cell surface, resulting in increased binding of C1q and potentiated complement-dependent cytotoxicity (CDC). METHODS Co-crystallization studies were performed to identify the binding interface of HexaBody-CD38 and CD38. HexaBody-CD38-induced CDC, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), trogocytosis, and apoptosis were assessed using flow cytometry assays using tumour cell lines, and MM patient samples (CDC). CD38 enzymatic activity was measured using fluorescence spectroscopy. Anti-tumour activity of HexaBody-CD38 was assessed in patient-derived xenograft mouse models in vivo. FINDINGS HexaBody-CD38 binds a unique epitope on CD38 and induced potent CDC in multiple myeloma (MM), acute myeloid leukaemia (AML), and B-cell non-Hodgkin lymphoma (B-NHL) cells. Anti-tumour activity was confirmed in patient-derived xenograft models in vivo. Sensitivity to HexaBody-CD38 correlated with CD38 expression level and was inversely correlated with expression of complement regulatory proteins. Compared to daratumumab, HexaBody-CD38 showed enhanced CDC in cell lines with lower levels of CD38 expression, without increasing lysis of healthy leukocytes. More effective CDC was also confirmed in primary MM cells. Furthermore, HexaBody-CD38 efficiently induced ADCC, ADCP, trogocytosis, and apoptosis after Fc-crosslinking. Moreover, HexaBody-CD38 strongly inhibited CD38 cyclase activity, which is hypothesized to relieve immune suppression in the tumour microenvironment. INTERPRETATION Based on these preclinical studies, a clinical trial was initiated to assess the clinical safety of HexaBody-CD38 in patients with MM. FUNDING Genmab.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Kristine A Frerichs
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tuna Mutis
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Niels W C J van de Donk
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | |
Collapse
|
39
|
Goldberg BS, Spencer DA, Pandey S, Ordonez T, Barnette P, Yu Y, Gao L, Dufloo J, Bruel T, Schwartz O, Ackerman ME, Hessell AJ. Complement contributes to antibody-mediated protection against repeated SHIV challenge. Proc Natl Acad Sci U S A 2023; 120:e2221247120. [PMID: 37155897 PMCID: PMC10193994 DOI: 10.1073/pnas.2221247120] [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: 12/14/2022] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
The first clinical efficacy trials of a broadly neutralizing antibody (bNAb) resulted in less benefit than expected and suggested that improvements are needed to prevent HIV infection. While considerable effort has focused on optimizing neutralization breadth and potency, it remains unclear whether augmenting the effector functions elicited by broadly neutralizing antibodies (bNAbs) may also improve their clinical potential. Among these effector functions, complement-mediated activities, which can culminate in the lysis of virions or infected cells, have been the least well studied. Here, functionally modified variants of the second-generation bNAb 10-1074 with ablated and enhanced complement activation profiles were used to examine the role of complement-associated effector functions. When administered prophylactically against simian-HIV challenge in rhesus macaques, more bNAb was required to prevent plasma viremia when complement activity was eliminated. Conversely, less bNAb was required to protect animals from plasma viremia when complement activity was enhanced. These results suggest that complement-mediated effector functions contribute to in vivo antiviral activity, and that their engineering may contribute to the further improvements in the efficacy of antibody-mediated prevention strategies.
Collapse
Affiliation(s)
| | - David A. Spencer
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| | - Shilpi Pandey
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| | - Tracy Ordonez
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| | - Philip Barnette
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| | - Yun Yu
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health and Science University, Portland, OR97239
- Biostatistics & Bioinformatics Core, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR97006
| | - Lina Gao
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health and Science University, Portland, OR97239
- Biostatistics & Bioinformatics Core, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR97006
| | - Jérémy Dufloo
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015Paris, France
- Université de Paris, École doctorale BioSPC 562, 75013Paris, France
| | - Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015Paris, France
- Vaccine Research Institute, 94000Créteil, France
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015Paris, France
- Vaccine Research Institute, 94000Créteil, France
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH03755
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH03755
| | - Ann J. Hessell
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR97006
| |
Collapse
|
40
|
Dadas O, Ertay A, Cragg MS. Delivering co-stimulatory tumor necrosis factor receptor agonism for cancer immunotherapy: past, current and future perspectives. Front Immunol 2023; 14:1147467. [PMID: 37180119 PMCID: PMC10167284 DOI: 10.3389/fimmu.2023.1147467] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/27/2023] [Indexed: 05/15/2023] Open
Abstract
The tumor necrosis factor superfamily (TNFSF) and their receptors (TNFRSF) are important regulators of the immune system, mediating proliferation, survival, differentiation, and function of immune cells. As a result, their targeting for immunotherapy is attractive, although to date, under-exploited. In this review we discuss the importance of co-stimulatory members of the TNFRSF in optimal immune response generation, the rationale behind targeting these receptors for immunotherapy, the success of targeting them in pre-clinical studies and the challenges in translating this success into the clinic. The efficacy and limitations of the currently available agents are discussed alongside the development of next generation immunostimulatory agents designed to overcome current issues, and capitalize on this receptor class to deliver potent, durable and safe drugs for patients.
Collapse
Affiliation(s)
- Osman Dadas
- Antibody and Vaccine Group, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ayse Ertay
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Mark S. Cragg
- Antibody and Vaccine Group, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
41
|
Yang H, Li H, Yang F, Tao Z, Shi Q, She T, Feng Y, Li Z, Chen J, Zhong Y, Su T, Zeng W, Zhang Y, Wang S, Li L, Long T, Long D, Cheng J, Zhu H, Lu X. Molecular superglue-mediated higher-order assembly of TRAIL variants with superior apoptosis induction and antitumor activity. Biomaterials 2023; 295:121994. [PMID: 36775789 DOI: 10.1016/j.biomaterials.2023.121994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 02/12/2023]
Abstract
Prompting higher-order death receptor (DR) clustering by increasing the valency of DR agonist is efficient to induce apoptosis of tumor cells. As an attractive DR agonist with superior biosafety, the trimeric tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exerts limited antitumor effect in patients, which is predominantly attributed to its low DR clustering ability and short serum half-life. Previous antibody scaffolds-based engineering strategies to increase the valency and/or prolong the serum half-life of TRAIL improve apoptosis induction, however, often produce large proteins with poor tumor penetration. Covalent protein ligation mediated by small molecular superglues such as SpyTag/SpyCatcher might be a novel strategy to assemble higher-order TRAIL variants. Upon fusion to TRAIL promotor, SpyTag/SpyCatcher molecular superglue preferentially ligated two trimeric TRAIL to produce a hexameric TRAIL variant, HexaTR, exhibiting a significantly increased apoptosis induction. In addition, an albumin-binding HexaTR, ABD-HexaTR, with a prolonged serum half-life by binding to endogenous albumin was also produced using the same strategy. Compared to the trimeric TRAIL, the hexameric HexaTR and ABD-HexaTR showed 20-50 times greater in vivo antitumor effect, resulting in eradication of several types of large (150-300 mm3) tumor xenografts. Combination with bortezomib carried by liposome further improved the antitumor effects of the hexavalent HexaTR and ABD-HexaTR in refractory cancer. Our results indicate that the superglue-mediated higher-order assembly is promising to improve the DR clustering and proapoptotic signaling of TRAIL, showing great advantages in constructing the next generation of DR agonists for cancer therapy.
Collapse
Affiliation(s)
- Hao Yang
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Heng Li
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fen Yang
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ze Tao
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuxiao Shi
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tianshan She
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanru Feng
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhao Li
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Chen
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Zhong
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Su
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wengjuan Zeng
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yong Zhang
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shisheng Wang
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lan Li
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tingting Long
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dan Long
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingqiu Cheng
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xiaofeng Lu
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
42
|
Aguinagalde Salazar L, den Boer MA, Castenmiller SM, Zwarthoff SA, de Haas C, Aerts PC, Beurskens FJ, Schuurman J, Heck AJR, van Kessel K, Rooijakkers SHM. Promoting Fc-Fc interactions between anti-capsular antibodies provides strong immune protection against Streptococcus pneumoniae. eLife 2023; 12:e80669. [PMID: 36947116 PMCID: PMC10032657 DOI: 10.7554/elife.80669] [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: 05/30/2022] [Accepted: 03/09/2023] [Indexed: 03/23/2023] Open
Abstract
Streptococcus pneumoniae is the leading cause of community-acquired pneumonia and an important cause of childhood mortality. Despite the introduction of successful vaccines, the global spread of both non-vaccine serotypes and antibiotic-resistant strains reinforces the development of alternative therapies against this pathogen. One possible route is the development of monoclonal antibodies (mAbs) that induce killing of bacteria via the immune system. Here, we investigate whether mAbs can be used to induce killing of pneumococcal serotypes for which the current vaccines show unsuccessful protection. Our study demonstrates that when human mAbs against pneumococcal capsule polysaccharides (CPS) have a poor capacity to induce complement activation, a critical process for immune protection against pneumococci, their activity can be strongly improved by hexamerization-enhancing mutations. Our data indicate that anti-capsular antibodies may have a low capacity to form higher-order oligomers (IgG hexamers) that are needed to recruit complement component C1. Indeed, specific point mutations in the IgG-Fc domain that strengthen hexamerization strongly enhance C1 recruitment and downstream complement activation on encapsulated pneumococci. Specifically, hexamerization-enhancing mutations E430G or E345K in CPS6-IgG strongly potentiate complement activation on S. pneumoniae strains that express capsular serotype 6 (CPS6), and the highly invasive serotype 19A strain. Furthermore, these mutations improve complement activation via mAbs recognizing CPS3 and CPS8 strains. Importantly, hexamer-enhancing mutations enable mAbs to induce strong opsonophagocytic killing by human neutrophils. Finally, passive immunization with CPS6-IgG1-E345K protected mice from developing severe pneumonia. Altogether, this work provides an important proof of concept for future optimization of antibody therapies against encapsulated bacteria.
Collapse
Affiliation(s)
| | - Maurits A den Boer
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht UniversityUtrechtNetherlands
- Netherlands Proteomics CenterUtrechtNetherlands
| | - Suzanne M Castenmiller
- Medical Microbiology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Seline A Zwarthoff
- Medical Microbiology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Carla de Haas
- Medical Microbiology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Piet C Aerts
- Medical Microbiology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | | | | | - Albert JR Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht UniversityUtrechtNetherlands
- Netherlands Proteomics CenterUtrechtNetherlands
| | - Kok van Kessel
- Medical Microbiology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Suzan HM Rooijakkers
- Medical Microbiology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| |
Collapse
|
43
|
An anti-HER2 biparatopic antibody that induces unique HER2 clustering and complement-dependent cytotoxicity. Nat Commun 2023; 14:1394. [PMID: 36914633 PMCID: PMC10011572 DOI: 10.1038/s41467-023-37029-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a receptor tyrosine kinase that plays an oncogenic role in breast, gastric and other solid tumors. However, anti-HER2 therapies are only currently approved for the treatment of breast and gastric/gastric esophageal junction cancers and treatment resistance remains a problem. Here, we engineer an anti-HER2 IgG1 bispecific, biparatopic antibody (Ab), zanidatamab, with unique and enhanced functionalities compared to both trastuzumab and the combination of trastuzumab plus pertuzumab (tras + pert). Zanidatamab binds adjacent HER2 molecules in trans and initiates distinct HER2 reorganization, as shown by polarized cell surface HER2 caps and large HER2 clusters, not observed with trastuzumab or tras + pert. Moreover, zanidatamab, but not trastuzumab nor tras + pert, elicit potent complement-dependent cytotoxicity (CDC) against high HER2-expressing tumor cells in vitro. Zanidatamab also mediates HER2 internalization and downregulation, inhibition of both cell signaling and tumor growth, antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP), and also shows superior in vivo antitumor activity compared to tras + pert in a HER2-expressing xenograft model. Collectively, we show that zanidatamab has multiple and distinct mechanisms of action derived from the structural effects of biparatopic HER2 engagement.
Collapse
|
44
|
Di Cristofano F, George A, Tajiknia V, Ghandali M, Wu L, Zhang Y, Srinivasan P, Strandberg J, Hahn M, Sanchez Sevilla Uruchurtu A, Seyhan AA, Carneiro BA, Zhou L, Huntington KE, El-Deiry WS. Therapeutic targeting of TRAIL death receptors. Biochem Soc Trans 2023; 51:57-70. [PMID: 36629496 PMCID: PMC9988005 DOI: 10.1042/bst20220098] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/26/2022] [Accepted: 12/07/2022] [Indexed: 01/12/2023]
Abstract
The discovery of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) along with its potent and selective antitumor effects initiated a decades-long search for therapeutic strategies to target the TRAIL pathway. First-generation approaches were focused on the development of TRAIL receptor agonists (TRAs), including recombinant human TRAIL (rhTRAIL) and TRAIL receptor-targeted agonistic antibodies. While such TRAIL pathway-targeted therapies showed promise in preclinical data and clinical trials have been conducted, none have advanced to FDA approval. Subsequent second-generation approaches focused on improving upon the specific limitations of first-generation approaches by ameliorating the pharmacokinetic profiles and agonistic abilities of TRAs as well as through combinatorial approaches to circumvent resistance. In this review, we summarize the successes and shortcomings of first- and second-generation TRAIL pathway-based therapies, concluding with an overview of the discovery and clinical introduction of ONC201, a compound with a unique mechanism of action that represents a new generation of TRAIL pathway-based approaches. We discuss preclinical and clinical findings in different tumor types and provide a unique perspective on translational directions of the field.
Collapse
Affiliation(s)
- Francesca Di Cristofano
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Andrew George
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Vida Tajiknia
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Maryam Ghandali
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Laura Wu
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Yiqun Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Praveen Srinivasan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Jillian Strandberg
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Marina Hahn
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Ashley Sanchez Sevilla Uruchurtu
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Benedito A. Carneiro
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital and Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Kelsey E. Huntington
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, U.S.A
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
- Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital and Brown University, Providence, RI 02903, U.S.A
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, U.S.A
| |
Collapse
|
45
|
Abstract
As a natural function, antibodies defend the host from infected cells and pathogens by recognizing their pathogenic determinants. Antibodies (Abs) gained wide acceptance with an enormous impact on human health and have predominantly captured the arena of bio-therapeutics and bio-diagnostics. The scope of Ab-based biologics is vast, and it is likely to solve many unmet clinical needs in future. The majority of attention is now devoted to developing innovative technologies for manufacturing and engineering Abs, better suited to satisfy human needs. The advent of Ab engineering technologies (AET) led to phenomenal developments leading to the generation of Abs-/Ab-derived molecules with desirable functional properties proportional to their expanding requirements. Evolution brought by AET, from the naturally occurring Ab forms to several advanced Ab formats and derivatives, was much needed as it is of great interest to the pharmaceutical industry. Thus, numerous advancements in AET have propelled success in therapeutic Ab development, along with the potential for ever-increasing improvements. Unique characteristics of Abs, such as its diversity, specificity, structural integrity and an array of possible applications, together inspire continuous innovation in the field. Overall, the AET could assist in conquer of several limitations of Abs in terms of their applicability in the field of therapeutics, diagnostics and research; AET has so far led to the production of next-generation Abs, which have revolutionized these arenas. Here in this review, we discuss the various distinguished engineering platforms for Ab development and the progress in modern therapeutics by the so-called "next-generation Abs."
Collapse
Affiliation(s)
- Divya Kandari
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.,Banaras Hindu University, Varanasi, India.,Amity University Rajasthan, Jaipur, India
| |
Collapse
|
46
|
Buhre JS, Pongracz T, Künsting I, Lixenfeld AS, Wang W, Nouta J, Lehrian S, Schmelter F, Lunding HB, Dühring L, Kern C, Petry J, Martin EL, Föh B, Steinhaus M, von Kopylow V, Sina C, Graf T, Rahmöller J, Wuhrer M, Ehlers M. mRNA vaccines against SARS-CoV-2 induce comparably low long-term IgG Fc galactosylation and sialylation levels but increasing long-term IgG4 responses compared to an adenovirus-based vaccine. Front Immunol 2023; 13:1020844. [PMID: 36713457 PMCID: PMC9877300 DOI: 10.3389/fimmu.2022.1020844] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 12/09/2022] [Indexed: 01/15/2023] Open
Abstract
Background The new types of mRNA-containing lipid nanoparticle vaccines BNT162b2 and mRNA-1273 and the adenovirus-based vaccine AZD1222 were developed against SARS-CoV-2 and code for its spike (S) protein. Several studies have investigated short-term antibody (Ab) responses after vaccination. Objective However, the impact of these new vaccine formats with unclear effects on the long-term Ab response - including isotype, subclass, and their type of Fc glycosylation - is less explored. Methods Here, we analyzed anti-S Ab responses in blood serum and the saliva of SARS-CoV-2 naïve and non-hospitalized pre-infected subjects upon two vaccinations with different mRNA- and adenovirus-based vaccine combinations up to day 270. Results We show that the initially high mRNA vaccine-induced blood and salivary anti-S IgG levels, particularly IgG1, markedly decrease over time and approach the lower levels induced with the adenovirus-based vaccine. All three vaccines induced, contrary to the short-term anti-S IgG1 response with high sialylation and galactosylation levels, a long-term anti-S IgG1 response that was characterized by low sialylation and galactosylation with the latter being even below the corresponding total IgG1 galactosylation level. Instead, the mRNA, but not the adenovirus-based vaccines induced long-term IgG4 responses - the IgG subclass with inhibitory effector functions. Furthermore, salivary anti-S IgA levels were lower and decreased faster in naïve as compared to pre-infected vaccinees. Predictively, age correlated with lower long-term anti-S IgG titers for the mRNA vaccines. Furthermore, higher total IgG1 galactosylation, sialylation, and bisection levels correlated with higher long-term anti-S IgG1 sialylation, galactosylation, and bisection levels, respectively, for all vaccine combinations. Conclusion In summary, the study suggests a comparable "adjuvant" potential of the newly developed vaccines on the anti-S IgG Fc glycosylation, as reflected in relatively low long-term anti-S IgG1 galactosylation levels generated by the long-lived plasma cell pool, whose induction might be driven by a recently described TH1-driven B cell response for all three vaccines. Instead, repeated immunization of naïve individuals with the mRNA vaccines increased the proportion of the IgG4 subclass over time which might influence the long-term Ab effector functions. Taken together, these data shed light on these novel vaccine formats and might have potential implications for their long-term efficacy.
Collapse
Affiliation(s)
- Jana Sophia Buhre
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Tamas Pongracz
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Inga Künsting
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Anne S. Lixenfeld
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Jan Nouta
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Selina Lehrian
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Franziska Schmelter
- Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Hanna B. Lunding
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Lara Dühring
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Carsten Kern
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Janina Petry
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Emily L. Martin
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Bandik Föh
- Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Moritz Steinhaus
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany,Department of Anesthesiology and Intensive Care, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Vera von Kopylow
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Christian Sina
- Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Tobias Graf
- Medical Department 2, University Heart Center of Schleswig-Holstein, Lübeck, Germany
| | - Johann Rahmöller
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany,Department of Anesthesiology and Intensive Care, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Manfred Wuhrer, ; Marc Ehlers,
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany,Airway Research Center North (ARCN), University of Lübeck, German Center for Lung Research (DZL), Lübeck, Germany,*Correspondence: Manfred Wuhrer, ; Marc Ehlers,
| |
Collapse
|
47
|
Cable J, Saphire EO, Hayday AC, Wiltshire TD, Mousa JJ, Humphreys DP, Breij ECW, Bruhns P, Broketa M, Furuya G, Hauser BM, Mahévas M, Carfi A, Cantaert T, Kwong PD, Tripathi P, Davis JH, Brewis N, Keyt BA, Fennemann FL, Dussupt V, Sivasubramanian A, Kim PM, Rawi R, Richardson E, Leventhal D, Wolters RM, Geuijen CAW, Sleeman MA, Pengo N, Donnellan FR. Antibodies as drugs-a Keystone Symposia report. Ann N Y Acad Sci 2023; 1519:153-166. [PMID: 36382536 PMCID: PMC10103175 DOI: 10.1111/nyas.14915] [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] [Indexed: 11/17/2022]
Abstract
Therapeutic antibodies have broad indications across diverse disease states, such as oncology, autoimmune diseases, and infectious diseases. New research continues to identify antibodies with therapeutic potential as well as methods to improve upon endogenous antibodies and to design antibodies de novo. On April 27-30, 2022, experts in antibody research across academia and industry met for the Keystone symposium "Antibodies as Drugs" to present the state-of-the-art in antibody therapeutics, repertoires and deep learning, bispecific antibodies, and engineering.
Collapse
Affiliation(s)
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA.,Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Adrian C Hayday
- Peter Gorer Department of Immunobiology, King's College London, London, UK.,Cancer Research UK Cancer Immunotherapy Accelerator, London, UK.,Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | | | - Jarrod J Mousa
- Department of Infectious Diseases and Center for Vaccines and Immunology, College of Veterinary Medicine, Athens, Georgia, USA.,Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, Georgia, USA.,Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Esther C W Breij
- Translational Research and Precision Medicine, Genmab BV, Utrecht, the Netherlands
| | - Pierre Bruhns
- Institut Pasteur, Université de Paris, Unit of Antibodies in Therapy and Pathology, Paris, France
| | - Matteo Broketa
- Institut Pasteur, Université de Paris, Unit of Antibodies in Therapy and Pathology, Paris, France
| | - Genta Furuya
- Department of Preventive Medicine and Department of Pathology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Blake M Hauser
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Matthieu Mahévas
- Service de Médecine Interne, Centre de Référence des Cytopénies Auto-immunes de l'adulte, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris-Est Créteil, Créteil, France
| | - Andrea Carfi
- Moderna Inc., Cambridge, Massachusetts, USA.,Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Prabhanshu Tripathi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Bruce A Keyt
- IGM Biosciences, Inc., Mountainview, California, USA
| | | | - Vincent Dussupt
- Emerging Infectious Diseases Branch, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | | | - Philip M Kim
- Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Reda Rawi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Eve Richardson
- Department of Statistics, University of Oxford, Oxford, UK
| | | | - Rachael M Wolters
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | | | |
Collapse
|
48
|
Lipinski B, Unmuth L, Arras P, Becker S, Bauer C, Toleikis L, Krah S, Doerner A, Yanakieva D, Boje AS, Klausz K, Peipp M, Siegmund V, Evers A, Kolmar H, Pekar L, Zielonka S. Generation and engineering of potent single domain antibody-based bispecific IL-18 mimetics resistant to IL-18BP decoy receptor inhibition. MAbs 2023; 15:2236265. [PMID: 37469014 PMCID: PMC10361135 DOI: 10.1080/19420862.2023.2236265] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
Here, we generated bispecific antibody (bsAb) derivatives that mimic the function of interleukin (IL)-18 based on single domain antibodies (sdAbs) specific to IL-18 Rα and IL-18 Rβ. For this, camelids were immunized, followed by yeast surface display (YSD)-enabled discovery of VHHs targeting the individual receptor subunits. Upon reformatting into a strictly monovalent (1 + 1) bispecific sdAb architecture, several bsAbs triggered dose-dependent IL-18 R downstream signaling on IL-18 reporter cells, as well as IFN-γ release by peripheral blood mononuclear cells in the presence of low-dose IL-12. However, compared with IL-18, potencies and efficacies were considerably attenuated. By engineering paratope valencies and the spatial orientation of individual paratopes within the overall design architecture, we were able to generate IL-18 mimetics displaying significantly augmented functionalities, resulting in bispecific cytokine mimetics that were more potent than IL-18 in triggering proinflammatory cytokine release. Furthermore, generated IL-18 mimetics were unaffected from inhibition by IL-18 binding protein decoy receptor. Essentially, we demonstrate that this strategy enables the generation of IL-18 mimetics with tailor-made cytokine functionalities.
Collapse
Affiliation(s)
- Britta Lipinski
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Laura Unmuth
- Early Protein Supply and Characterization (EPSC), Merck Healthcare KGaA, Darmstadt, Germany
| | - Paul Arras
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Stefan Becker
- Early Protein Supply and Characterization (EPSC), Merck Healthcare KGaA, Darmstadt, Germany
| | - Christina Bauer
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Lars Toleikis
- Early Protein Supply and Characterization (EPSC), Merck Healthcare KGaA, Darmstadt, Germany
| | - Simon Krah
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Achim Doerner
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Desislava Yanakieva
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Ammelie Svea Boje
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University Kiel, Kiel, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University Kiel, Kiel, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University Kiel, Kiel, Germany
| | - Vanessa Siegmund
- Early Protein Supply and Characterization (EPSC), Merck Healthcare KGaA, Darmstadt, Germany
| | - Andreas Evers
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Lukas Pekar
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Stefan Zielonka
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
49
|
Troisi M, Marini E, Abbiento V, Stazzoni S, Andreano E, Rappuoli R. A new dawn for monoclonal antibodies against antimicrobial resistant bacteria. Front Microbiol 2022; 13:1080059. [PMID: 36590399 PMCID: PMC9795047 DOI: 10.3389/fmicb.2022.1080059] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial resistance (AMR) is a quickly advancing threat for human health worldwide and almost 5 million deaths are already attributable to this phenomenon every year. Since antibiotics are failing to treat AMR-bacteria, new tools are needed, and human monoclonal antibodies (mAbs) can fill this role. In almost 50 years since the introduction of the first technology that led to mAb discovery, enormous leaps forward have been made to identify and develop extremely potent human mAbs. While their usefulness has been extensively proved against viral pathogens, human mAbs have yet to find their space in treating and preventing infections from AMR-bacteria and fully conquer the field of infectious diseases. The novel and most innovative technologies herein reviewed can support this goal and add powerful tools in the arsenal of weapons against AMR.
Collapse
Affiliation(s)
- Marco Troisi
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Eleonora Marini
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Valentina Abbiento
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Samuele Stazzoni
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Emanuele Andreano
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Rino Rappuoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Fondazione Biotecnopolo di Siena, Siena, Italy
| |
Collapse
|
50
|
Buhre JS, Becker M, Ehlers M. IgG subclass and Fc glycosylation shifts are linked to the transition from pre- to inflammatory autoimmune conditions. Front Immunol 2022; 13:1006939. [PMID: 36405742 PMCID: PMC9669588 DOI: 10.3389/fimmu.2022.1006939] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
A crucial factor for the development of inflammatory autoimmune diseases is the occurrence of antibodies directed against self-tissues and structures, which leads to damage and inflammation. While little is known about the cause of the development of mis-directed, disease-specific T and B cells and resulting IgG autoantibody responses, there is increasing evidence that their induction can occur years before disease symptoms appear. However, a certain proportion of healthy individuals express specific IgG autoantibodies without disease symptoms and not all subjects who generate autoantibodies may develop disease symptoms. Thus, the development of inflammatory autoimmune diseases seems to involve two steps. Increasing evidence suggests that harmless self-directed T and B cell and resulting IgG autoantibody responses in the pre-autoimmune disease stage might switch to more inflammatory T and B cell and IgG autoantibody responses that trigger the inflammatory autoimmune disease stage. Here, we summarize findings on the transition from the pre-disease to the disease stage and vice versa, e.g. by pregnancy and treatment, with a focus on low-/anti-inflammatory versus pro-inflammatory IgG autoantibody responses, including IgG subclass and Fc glycosylation features. Characterization of biomarkers that identify the transition from the pre-disease to the disease stage might facilitate recognition of the ideal time point of treatment initiation and the development of therapeutic strategies for re-directing inflammatory autoimmune conditions.
Collapse
Affiliation(s)
- Jana Sophia Buhre
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Mareike Becker
- Department of Dermatology, Allergology, and Venereology, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
- Airway Research Center North, German Center for Lung Research (DZL), University of Lübeck, Lübeck, Germany
- *Correspondence: Marc Ehlers,
| |
Collapse
|