1
|
Davies AM, Bui TTT, Pacheco‐Gómez R, Vester SK, Beavil AJ, Gould HJ, Sutton BJ, McDonnell JM. The Crystal Structure of Human IgD-Fc Reveals Unexpected Differences With Other Antibody Isotypes. Proteins 2025; 93:786-800. [PMID: 39582378 PMCID: PMC11878202 DOI: 10.1002/prot.26771] [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: 08/05/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
Of the five human antibody isotypes, the function of IgD is the least well-understood, although various studies point to a role for IgD in mucosal immunity. IgD is also the least well structurally characterized isotype. Until recently, when crystal structures were reported for the IgD Fab, the only structural information available was a model for intact IgD based on solution scattering data. We now report the crystal structure of human IgD-Fc solved at 3.0 Å resolution. Although similar in overall architecture to other human isotypes, IgD-Fc displays markedly different orientations of the Cδ3 domains in the Cδ3 domain dimer and the lowest interface area of all the human isotypes. The nature of the residues that form the dimer interface also differs from those conserved in the other isotypes. By contrast, the interface between the Cδ2 and Cδ3 domains in each chain is the largest among the human isotypes. This interface is characterized by two binding pockets, not seen in other isotypes, and points to a potential role for the Cδ2/Cδ3 interface in stabilizing the IgD-Fc homodimer. We investigated the thermal stability of IgD-Fc, alone and in the context of an intact IgD antibody, and found that IgD-Fc unfolds in a single transition. Human IgD-Fc clearly has unique structural features not seen in the other human isotypes, and comparison with other mammalian IgD sequences suggests that these unique features might be widely conserved.
Collapse
Affiliation(s)
- Anna M. Davies
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Tam T. T. Bui
- Centre for Biomolecular SpectroscopyKing's College LondonLondonUK
| | | | - Susan K. Vester
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Andrew J. Beavil
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Hannah J. Gould
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Brian J. Sutton
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - James M. McDonnell
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| |
Collapse
|
2
|
Ogulur I, Mitamura Y, Yazici D, Pat Y, Ardicli S, Li M, D'Avino P, Beha C, Babayev H, Zhao B, Zeyneloglu C, Giannelli Viscardi O, Ardicli O, Kiykim A, Garcia-Sanchez A, Lopez JF, Shi LL, Yang M, Schneider SR, Skolnick S, Dhir R, Radzikowska U, Kulkarni AJ, Imam MB, Veen WVD, Sokolowska M, Martin-Fontecha M, Palomares O, Nadeau KC, Akdis M, Akdis CA. Type 2 immunity in allergic diseases. Cell Mol Immunol 2025; 22:211-242. [PMID: 39962262 PMCID: PMC11868591 DOI: 10.1038/s41423-025-01261-2] [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: 09/16/2024] [Accepted: 01/09/2025] [Indexed: 03/01/2025] Open
Abstract
Significant advancements have been made in understanding the cellular and molecular mechanisms of type 2 immunity in allergic diseases such as asthma, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis (EoE), food and drug allergies, and atopic dermatitis (AD). Type 2 immunity has evolved to protect against parasitic diseases and toxins, plays a role in the expulsion of parasites and larvae from inner tissues to the lumen and outside the body, maintains microbe-rich skin and mucosal epithelial barriers and counterbalances the type 1 immune response and its destructive effects. During the development of a type 2 immune response, an innate immune response initiates starting from epithelial cells and innate lymphoid cells (ILCs), including dendritic cells and macrophages, and translates to adaptive T and B-cell immunity, particularly IgE antibody production. Eosinophils, mast cells and basophils have effects on effector functions. Cytokines from ILC2s and CD4+ helper type 2 (Th2) cells, CD8 + T cells, and NK-T cells, along with myeloid cells, including IL-4, IL-5, IL-9, and IL-13, initiate and sustain allergic inflammation via T cell cells, eosinophils, and ILC2s; promote IgE class switching; and open the epithelial barrier. Epithelial cell activation, alarmin release and barrier dysfunction are key in the development of not only allergic diseases but also many other systemic diseases. Recent biologics targeting the pathways and effector functions of IL4/IL13, IL-5, and IgE have shown promising results for almost all ages, although some patients with severe allergic diseases do not respond to these therapies, highlighting the unmet need for a more detailed and personalized approach.
Collapse
Affiliation(s)
- Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Carina Beha
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Bingjie Zhao
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Can Zeyneloglu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | | | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Asuncion Garcia-Sanchez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Biomedical and Diagnostic Science, School of Medicine, University of Salamanca, Salamanca, Spain
| | - Juan-Felipe Lopez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Li-Li Shi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Minglin Yang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephen Skolnick
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Seed Health Inc., Los Angeles, CA, USA
| | - Raja Dhir
- Seed Health Inc., Los Angeles, CA, USA
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Abhijeet J Kulkarni
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mar Martin-Fontecha
- Departamento de Quimica Organica, Facultad de Optica y Optometria, Complutense University of Madrid, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| |
Collapse
|
3
|
Zhao C, Sun Z, Wang S, Zhang J, Liu J, Chen L, Lu G, Yu Y, Gao Y. IgG4 glycosylation contributes to the pathogenesis of IgG4 Hashimoto's thyroiditis via the complement pathway. Eur Thyroid J 2024; 13:e240156. [PMID: 39316722 PMCID: PMC11558973 DOI: 10.1530/etj-24-0156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/24/2024] [Indexed: 09/26/2024] Open
Abstract
Background To explore whether IgG4 is involved in the pathogenesis of IgG4 HT. Methods Serum TgAb IgG4 and TPOAb IgG4 were measured in IgG4 HT and non-IgG4 HT. C1q, mannose-binding lectin (MBL), Bb, C3d, C4d, and membrane attack complex (MAC) in thyroid tissues from IgG4 HT, non-IgG4 HT, and controls were examined by immunohistochemistry. We assessed IgG4 and MAC deposition in mouse thyroid by immunohistochemistry after injecting purified IgG4 into mice. The glycosylation patterns of TgAb IgG4 from IgG4 HT were identified by MALDI-TOF-MS. The ability of IgG4 to bind to MBL before and after deglycosylation was assessed by ELISA. MBL and MAC fluorescence were detected in thyrocytes after the addition of IgG4 or deglycosylated IgG4. Results Serum TgAb IgG4 and TPOAb IgG4 levels were significantly higher in the IgG4 HT group. MBL, Bb, C3d, C4d, and MAC levels were significantly higher in the thyroid tissues of IgG4 HT than in non-IgG4 HT (all P < 0.001). IgG4 colocalized with MBL by immunofluorescence. In mice, follicular cell structure disruption was observed after the injection of IgG4 from IgG4 HT, as well as the colocalization of IgG4 with MAC. High levels of TgAb IgG4 glycosylation patterns, including monogalactose glycan (G1F), galactose-deficient glycan (G0F), and high-mannose glycan (M5), were detected in IgG4 HT. After deglycosylation, IgG4 reduced its ability to bind to MBL, and there was low MBL and MAC activation in thyrocytes. Conclusion High levels of IgG4 glycosylation patterns, including G1F, G0F, and M5, may activate the complement lectin pathway, thereby participating in the pathogenesis of IgG4 HT.
Collapse
Affiliation(s)
- Chenxu Zhao
- Department of Endocrinology, Peking University First Hospital, Xicheng District, Beijing, China
| | - Zhiming Sun
- Department of Endocrinology, Peking University First Hospital, Xicheng District, Beijing, China
| | - Shuaihang Wang
- Department of Endocrinology, Peking University First Hospital, Xicheng District, Beijing, China
| | - Jixin Zhang
- Department of Pathology, Peking University First Hospital, Xicheng District, Beijing, China
| | - Jumei Liu
- Department of Pathology, Peking University First Hospital, Xicheng District, Beijing, China
| | - Lei Chen
- Department of Ultrasound, Peking University First Hospital, Xicheng District, Beijing, China
| | - Guizhi Lu
- Department of Endocrinology, Peking University First Hospital, Xicheng District, Beijing, China
| | - Yang Yu
- Department of Endocrinology, Peking University First Hospital, Xicheng District, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Xicheng District, Beijing, China
| |
Collapse
|
4
|
Maslinska M, Kostyra-Grabczak K. Immunoglobulin G4 in primary Sjögren's syndrome and IgG4-related disease - connections and dissimilarities. Front Immunol 2024; 15:1376723. [PMID: 39364411 PMCID: PMC11446744 DOI: 10.3389/fimmu.2024.1376723] [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: 01/26/2024] [Accepted: 07/26/2024] [Indexed: 10/05/2024] Open
Abstract
Primary Sjögren's syndrome (pSS) is an autoimmune disease, with B cell hyperactivation and autoantibody production as its immunological hallmarks. Although the distinction between immunoglobulin G4-related disease (IgG4-RD) and pSS, based on the presence or absence of certain autoantibodies, seems easy to make, possibility of elevated serum IgG4 concentration and often similar organ involvement may lead to a misdiagnosis. The increased serum concentration of IgG4 in IgG4-RD is not clearly linked to the pathogenesis of IgG-RD and it has been suggested that it may constitute just an epiphenomenon. The aim of this article is to discuss the presence of IgG4 in pSS and IgG4-RD and its potential significance for these two diseases.
Collapse
Affiliation(s)
- Maria Maslinska
- Early Arthritis Clinic, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Kinga Kostyra-Grabczak
- Early Arthritis Clinic, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
5
|
Li L, Ding P, Dong Y, Shen S, Lv X, Yu J, Li L, Chen J, Wang P, Han B, Xu T, Hu W. CG001, a C3b-targeted complement inhibitor, blocks 3 complement pathways: development and preclinical evaluation. Blood Adv 2024; 8:4181-4193. [PMID: 38865712 PMCID: PMC11334799 DOI: 10.1182/bloodadvances.2024012874] [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: 02/07/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024] Open
Abstract
ABSTRACT Excessively activated or dysregulated complement activation may contribute to the pathogenesis of a wide range of human diseases, thus leading to a surge in complement inhibitors. Herein, we developed a human-derived and antibody-like C3b-targeted fusion protein (CRIg-FH-Fc) x2, termed CG001, that could potently block all 3 complement pathways. Complement receptor of the immunoglobulin superfamily (CRIg) and factor H (FH) bind to distinct sites in C3b and synergistically inhibit complement activation. CRIg occupancy in C3b prevents the recruitment of C3 and C5 substrates, whereas FH occupancy in C3b accelerates the decay of C3/C5 convertases and promotes the factor I-mediated degradation and inactivation of C3b. CG001 also showed therapeutic effects in alternative pathways-induced hemolytic mouse and classical pathways-induced mesangial proliferative glomerulonephritis rat models. In the pharmacological/toxicological evaluation in rats and cynomolgus monkeys, CG001 displayed an antibody-like pharmacokinetic profile, a convincing complement inhibitory effect, and no observable toxic effects. Therefore, CG001 holds substantial potential for human clinical studies.
Collapse
Affiliation(s)
- Ling Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | | | - Shupei Shen
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Xinyue Lv
- ComGen Pharmaceutical Co Ltd, Shanghai, China
| | - Jie Yu
- ComGen Pharmaceutical Co Ltd, Shanghai, China
| | - Luying Li
- ComGen Pharmaceutical Co Ltd, Shanghai, China
| | - Jianfeng Chen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Pilin Wang
- Alphamab Co Ltd., Suzhou, Jiangsu, China
| | - Bing Han
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Ting Xu
- Alphamab Co Ltd., Suzhou, Jiangsu, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Keating SM, Higgins BW. New technologies in therapeutic antibody development: The next frontier for treating infectious diseases. Antiviral Res 2024; 227:105902. [PMID: 38734210 DOI: 10.1016/j.antiviral.2024.105902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Adaptive immunity to viral infections requires time to neutralize and clear viruses to resolve infection. Fast growing and pathogenic viruses are quickly established, are highly transmissible and cause significant disease burden making it difficult to mount effective responses, thereby prolonging infection. Antibody-based passive immunotherapies can provide initial protection during acute infection, assist in mounting an adaptive immune response, or provide protection for those who are immune suppressed or immune deficient. Historically, plasma-derived antibodies have demonstrated some success in treating diseases caused by viral pathogens; nonetheless, limitations in access to product and antibody titer reduce success of this treatment modality. Monoclonal antibodies (mAbs) have proven an effective alternative, as it is possible to manufacture highly potent and specific mAbs against viral targets on an industrial scale. As a result, innovative technologies to discover, engineer and manufacture specific and potent antibodies have become an essential part of the first line of treatment in pathogenic viral infections. However, a mAb targeting a specific epitope will allow escape variants to outgrow, causing new variant strains to become dominant and resistant to treatment with that mAb. Methods to mitigate escape have included combining mAbs into cocktails, creating bi-specific or antibody drug conjugates but these strategies have also been challenged by the potential development of escape mutations. New technologies in developing antibodies made as recombinant polyclonal drugs can integrate the strength of poly-specific antibody responses to prevent mutational escape, while also incorporating antibody engineering to prevent antibody dependent enhancement and direct adaptive immune responses.
Collapse
Affiliation(s)
- Sheila M Keating
- GigaGen, Inc. (A Grifols Company), 75 Shoreway Road, San Carlos, CA, 94070, USA.
| | | |
Collapse
|
7
|
Lenk L, Baccelli I, Laqua A, Heymann J, Reimer C, Dietterle A, Winterberg D, Mary C, Corallo F, Taurelle J, Narbeburu E, Neyton S, Déramé M, Pengam S, Vogiatzi F, Bornhauser B, Bourquin JP, Raffel S, Dovhan V, Schüler T, Escherich G, den Boer ML, Boer JM, Wessels W, Peipp M, Alten J, Antić Ž, Bergmann AK, Schrappe M, Cario G, Brüggemann M, Poirier N, Schewe DM. The IL-7R antagonist lusvertikimab reduces leukemic burden in xenograft ALL via antibody-dependent cellular phagocytosis. Blood 2024; 143:2735-2748. [PMID: 38518105 PMCID: PMC11251409 DOI: 10.1182/blood.2023021088] [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: 05/08/2023] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 03/24/2024] Open
Abstract
ABSTRACT Acute lymphoblastic leukemia (ALL) arises from the uncontrolled proliferation of B-cell precursors (BCP-ALL) or T cells (T-ALL). Current treatment protocols obtain high cure rates in children but are based on toxic polychemotherapy. Novel therapies are urgently needed, especially in relapsed/refractory (R/R) disease, high-risk (HR) leukemias and T-ALL, in which immunotherapy approaches remain scarce. Although the interleukin-7 receptor (IL-7R) plays a pivotal role in ALL development, no IL-7R-targeting immunotherapy has yet reached clinical application in ALL. The IL-7Rα chain (CD127)-targeting IgG4 antibody lusvertikimab (LUSV; formerly OSE-127) is a full antagonist of the IL-7R pathway, showing a good safety profile in healthy volunteers. Here, we show that ∼85% of ALL cases express surface CD127. We demonstrate significant in vivo efficacy of LUSV immunotherapy in a heterogeneous cohort of BCP- and T-ALL patient-derived xenografts (PDX) in minimal residual disease (MRD) and overt leukemia models, including R/R and HR leukemias. Importantly, LUSV was particularly effective when combined with polychemotherapy in a phase 2-like PDX study with CD127high samples leading to MRD-negativity in >50% of mice treated with combination therapy. Mechanistically, LUSV targeted ALL cells via a dual mode of action comprising direct IL-7R antagonistic activity and induction of macrophage-mediated antibody-dependent cellular phagocytosis (ADCP). LUSV-mediated in vitro ADCP levels significantly correlated with CD127 expression levels and the reduction of leukemia burden upon treatment of PDX animals in vivo. Altogether, through its dual mode of action and good safety profile, LUSV may represent a novel immunotherapy option for any CD127+ ALL, particularly in combination with standard-of-care polychemotherapy.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Xenograft Model Antitumor Assays
- Receptors, Interleukin-7/antagonists & inhibitors
- Mice, SCID
- Phagocytosis/drug effects
- Interleukin-7 Receptor alpha Subunit
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Female
- Mice, Inbred NOD
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Cell Line, Tumor
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
Collapse
Affiliation(s)
- Lennart Lenk
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | | | - Anna Laqua
- Department of Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Julia Heymann
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Claas Reimer
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Anna Dietterle
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Dorothee Winterberg
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | | | | | | | | | | | | | | | - Fotini Vogiatzi
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Beat Bornhauser
- Department of Pediatric Hematology/Oncology, University Children's Hospital, Zurich, Switzerland
| | - Jean-Pierre Bourquin
- Department of Pediatric Hematology/Oncology, University Children's Hospital, Zurich, Switzerland
| | - Simon Raffel
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Vladyslava Dovhan
- Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Gabriele Escherich
- Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Judith M. Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Wiebke Wessels
- Department of Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Julia Alten
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Željko Antić
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Anke K. Bergmann
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Martin Schrappe
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Gunnar Cario
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Monika Brüggemann
- Department of Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Denis M. Schewe
- Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
8
|
Spiteri VA, Doutch J, Rambo RP, Bhatt JS, Gor J, Dalby PA, Perkins SJ. Using atomistic solution scattering modelling to elucidate the role of the Fc glycans in human IgG4. PLoS One 2024; 19:e0300964. [PMID: 38557973 PMCID: PMC10984405 DOI: 10.1371/journal.pone.0300964] [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: 09/21/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Human immunoglobulin G (IgG) exists as four subclasses IgG1-4, each of which has two Fab subunits joined by two hinges to a Fc subunit. IgG4 has the shortest hinge with 12 residues. The Fc subunit has two glycan chains, but the importance of glycosylation is not fully understood in IgG4. Here, to evaluate the stability and structure of non-glycosylated IgG4, we performed a multidisciplinary structural study of glycosylated and deglycosylated human IgG4 A33 for comparison with our similar study of human IgG1 A33. After deglycosylation, IgG4 was found to be monomeric by analytical ultracentrifugation; its sedimentation coefficient of 6.52 S was reduced by 0.27 S in reflection of its lower mass. X-ray and neutron solution scattering showed that the overall Guinier radius of gyration RG and its cross-sectional values after deglycosylation were almost unchanged. In the P(r) distance distribution curves, the two M1 and M2 peaks that monitor the two most common distances within IgG4 were unchanged following deglycosylation. Further insight from Monte Carlo simulations for glycosylated and deglycosylated IgG4 came from 111,382 and 117,135 possible structures respectively. Their comparison to the X-ray and neutron scattering curves identified several hundred best-fit models for both forms of IgG4. Principal component analyses showed that glycosylated and deglycosylated IgG4 exhibited different conformations from each other. Within the constraint of unchanged RG and M1-M2 values, the glycosylated IgG4 models showed more restricted Fc conformations compared to deglycosylated IgG4, but no other changes. Kratky plots supported this interpretation of greater disorder upon deglycosylation, also observed in IgG1. Overall, these more variable Fc conformations may demonstrate a generalisable impact of deglycosylation on Fc structures, but with no large conformational changes in IgG4 unlike those seen in IgG1.
Collapse
Affiliation(s)
- Valentina A. Spiteri
- Division of Biosciences, Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - James Doutch
- ISIS Facility, STFC Rutherford Appleton Laboratory, Harwell Campus, Didcot, Oxfordshire, United Kingdom
| | - Robert P. Rambo
- Diamond Light Source Ltd., Diamond House, Harwell Science and Innovation Campus, Chilton, Didcot, Oxfordshire, United Kingdom
| | - Jayesh S. Bhatt
- Division of Biosciences, Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Jayesh Gor
- Division of Biosciences, Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Paul A. Dalby
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Stephen J. Perkins
- Division of Biosciences, Department of Structural and Molecular Biology, University College London, London, United Kingdom
| |
Collapse
|
9
|
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
|
10
|
Rispens T, Huijbers MG. The unique properties of IgG4 and its roles in health and disease. Nat Rev Immunol 2023; 23:763-778. [PMID: 37095254 PMCID: PMC10123589 DOI: 10.1038/s41577-023-00871-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
IgG4 is the least abundant subclass of IgG in human serum and has unique functional features. IgG4 is largely unable to activate antibody-dependent immune effector responses and, furthermore, undergoes Fab (fragment antigen binding)-arm exchange, rendering it bispecific for antigen binding and functionally monovalent. These properties of IgG4 have a blocking effect, either on the immune response or on the target protein of IgG4. In this Review, we discuss the unique structural characteristics of IgG4 and how these contribute to its roles in health and disease. We highlight how, depending on the setting, IgG4 responses can be beneficial (for example, in responses to allergens or parasites) or detrimental (for example, in autoimmune diseases, in antitumour responses and in anti-biologic responses). The development of novel models for studying IgG4 (patho)physiology and understanding how IgG4 responses are regulated could offer insights into novel treatment strategies for these IgG4-associated disease settings.
Collapse
Affiliation(s)
- Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Maartje G Huijbers
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
11
|
Wu Q, Cao C, Wei S, He H, Chen K, Su L, Liu Q, Li S, Lai Y, Li J. Decreasing hydrophobicity or shielding hydrophobic areas of CH2 attenuates low pH-induced IgG4 aggregation. Front Bioeng Biotechnol 2023; 11:1257665. [PMID: 37711444 PMCID: PMC10497874 DOI: 10.3389/fbioe.2023.1257665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/18/2023] [Indexed: 09/16/2023] Open
Abstract
Protein aggregation is a major challenge in the development of therapeutic monoclonal antibodies (mAbs). Several stressors can cause protein aggregation, including temperature shifts, mechanical forces, freezing-thawing cycles, oxidants, reductants, and extreme pH. When antibodies are exposed to low pH conditions, aggregation increases dramatically. However, low pH treatment is widely used in protein A affinity chromatography and low pH viral inactivation procedures. In the development of an IgG4 subclass antibody, mAb1-IgG4 showed a strong tendency to aggregate when temporarily exposed to low pH conditions. Our findings showed that the aggregation of mAb1-IgG4 under low pH conditions is determined by the stability of the Fc. The CH2 domain is the least stable domain in mAb1-IgG4. The L309E, Q311D, and Q311E mutations in the CH2 domain significantly reduced the aggregation propensity, which could be attributed to a reduction in the hydrophobicity of the CH2 domain. Protein stabilizers, such as sucrose and mannose, could also attenuate low pH-induced mAb1-IgG4 aggregation by shielding hydrophobic areas and increasing protein stability. Our findings provide valuable strategies for managing the aggregation of protein therapeutics with a human IgG4 backbone.
Collapse
Affiliation(s)
- Qiang Wu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| | - Chunlai Cao
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
- The United Biotechnology (Zhuhai Hengqin) Co., Ltd., Zhuhai, Guangdong, China
| | - Suzhen Wei
- The United Biotechnology (Zhuhai Hengqin) Co., Ltd., Zhuhai, Guangdong, China
| | - Hua He
- The United Biotechnology (Zhuhai Hengqin) Co., Ltd., Zhuhai, Guangdong, China
| | - Kangyue Chen
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| | - Lijuan Su
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| | - Qiulian Liu
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| | - Shuang Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yongjie Lai
- Department of Microbiology and Immunology, Zunyi Medical University (Zhuhai Campus), Zhuhai, Guangdong, China
| | - Jing Li
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| |
Collapse
|
12
|
Joo V, Petrovas C, de Leval L, Noto A, Obeid M, Fenwick C, Pantaleo G. A CD64/FcγRI-mediated mechanism hijacks PD-1 from PD-L1/2 interaction and enhances anti-PD-1 functional recovery of exhausted T cells. Front Immunol 2023; 14:1213375. [PMID: 37622123 PMCID: PMC10446174 DOI: 10.3389/fimmu.2023.1213375] [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: 04/27/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAb) targeting the immune checkpoint inhibitor programmed cell death protein 1 (PD-1) have achieved considerable clinical success in anti-cancer therapy through relieving T cell exhaustion. Blockade of PD-1 interaction with its ligands PD-L1 and PD-L2 is an important determinant in promoting the functional recovery of exhausted T cells. Here, we show that anti-PD-1 mAbs act through an alternative mechanism leading to the downregulation of PD-1 surface expression on memory CD4+ and CD8+ T cells. PD-1 receptor downregulation is a distinct process from receptor endocytosis and occurs in a CD14+ monocyte dependent manner with the CD64/Fcγ receptor I acting as the primary factor for this T cell extrinsic process. Importantly, downregulation of surface PD-1 strongly enhances antigen-specific functional recovery of exhausted PD-1+CD8+ T cells. Our study demonstrates a novel mechanism for reducing cell surface levels of PD-1 and limiting the inhibitory targeting by PD-L1/2 and thereby enhancing the efficacy of anti-PD-1 Ab in restoring T cell functionality.
Collapse
Affiliation(s)
- Victor Joo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Constantinos Petrovas
- Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alessandra Noto
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Michel Obeid
- Lausanne Center for Immuno-oncology Toxicities (LCIT), Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
13
|
Strobl MR, Demir H, Sánchez Acosta G, Drescher A, Kitzmüller C, Möbs C, Pfützner W, Bohle B. The role of IgG 1 and IgG 4 as dominant IgE-blocking antibodies shifts during allergen immunotherapy. J Allergy Clin Immunol 2023; 151:1371-1378.e5. [PMID: 36657603 DOI: 10.1016/j.jaci.2023.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
BACKGROUND The induction of allergen-specific IgE-blocking antibodies is a hallmark of allergen immunotherapy (AIT). The inhibitory bioactivity has largely been attributed to IgG4; however, our recent studies indicated the dominance of IgG1 early in AIT. OBJECTIVES Here, the IgE-blocking activity and avidity of allergen-specific IgG1 and IgG4 antibodies were monitored throughout 3 years of treatment. METHODS Serum samples from 24 patients were collected before and regularly during AIT with birch pollen. Bet v 1-specific IgG1 and IgG4 levels were determined by ELISA and ImmunoCAP, respectively. Unmodified and IgG1- or IgG4-depleted samples were compared for their inhibition of Bet v 1-induced basophil activation. The stability of Bet v 1-antibody complexes was compared by ELISA and by surface plasmon resonance. RESULTS Bet v 1-specific IgG1 and IgG4 levels peaked at 12 and 24 months of AIT, respectively. Serological IgE-blocking peaked at 6 months and remained high thereafter. In the first year of therapy, depletion of IgG1 clearly diminished the inhibition of basophil activation while the absence of IgG4 hardly reduced IgE-blocking. Then, IgG4 became the main inhibitory isotype in most individuals. Both isotypes displayed high avidity to Bet v 1 ab initio of AIT, which did not increase during treatment. Bet v 1-IgG1 complexes were enduringly more stable than Bet v 1-IgG4 complexes were. CONCLUSIONS In spite of the constant avidity of AIT-induced allergen-specific IgG1 and IgG4 antibodies, their dominance in IgE-blocking shifted in the course of treatment. The blocking activity of allergen-specific IgG1 should not be underestimated, particularly early in AIT.
Collapse
Affiliation(s)
- Maria R Strobl
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Hilal Demir
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gabriela Sánchez Acosta
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Anja Drescher
- Cytiva Europe GmbH, Freiburg, Department of Dermatology and Allergology, Philipps-Universität Marburg, Freiburg and Marburg, Germany
| | - Claudia Kitzmüller
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Christian Möbs
- Clinical and Experimental Allergology, Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Wolfgang Pfützner
- Clinical and Experimental Allergology, Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Barbara Bohle
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
14
|
Oskam N, Damelang T, Streutker M, Ooijevaar-de Heer P, Nouta J, Koeleman C, Van Coillie J, Wuhrer M, Vidarsson G, Rispens T. Factors affecting IgG4-mediated complement activation. Front Immunol 2023; 14:1087532. [PMID: 36776883 PMCID: PMC9910309 DOI: 10.3389/fimmu.2023.1087532] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Of the four human immunoglobulin G (IgG) subclasses, IgG4 is considered the least inflammatory, in part because it poorly activates the complement system. Regardless, in IgG4 related disease (IgG4-RD) and in autoimmune disorders with high levels of IgG4 autoantibodies, the presence of these antibodies has been linked to consumption and deposition of complement components. This apparent paradox suggests that conditions may exist, potentially reminiscent of in vivo deposits, that allow for complement activation by IgG4. Furthermore, it is currently unclear how variable glycosylation and Fab arm exchange may influence the ability of IgG4 to activate complement. Here, we used well-defined, glyco-engineered monoclonal preparations of IgG4 and determined their ability to activate complement in a controlled system. We show that IgG4 can activate complement only at high antigen and antibody concentrations, via the classical pathway. Moreover, elevated or reduced Fc galactosylation enhanced or diminished complement activation, respectively, with no apparent contribution from the lectin pathway. Fab glycans slightly reduced complement activation. Lastly, we show that bispecific, monovalent IgG4 resulting from Fab arm exchange is a less potent activator of complement than monospecific IgG4. Taken together, these results imply that involvement of IgG4-mediated complement activation in pathology is possible but unlikely.
Collapse
Affiliation(s)
- Nienke Oskam
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, Amsterdam, Netherlands
| | - Timon Damelang
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, Amsterdam, Netherlands.,Department of Immunohematology Experimental, Sanquin Research, Amsterdam, Netherlands.,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Marij Streutker
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, Amsterdam, Netherlands
| | - Pleuni Ooijevaar-de Heer
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, Amsterdam, Netherlands
| | - Jan Nouta
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Carolien Koeleman
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Julie Van Coillie
- Department of Immunohematology Experimental, Sanquin Research, Amsterdam, Netherlands.,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Gestur Vidarsson
- Department of Immunohematology Experimental, Sanquin Research, Amsterdam, Netherlands.,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
15
|
Pang X, Huang Z, Zhong T, Zhang P, Wang ZM, Xia M, Li B. Cadonilimab, a tetravalent PD-1/CTLA-4 bispecific antibody with trans-binding and enhanced target binding avidity. MAbs 2023; 15:2180794. [PMID: 36872527 PMCID: PMC10012886 DOI: 10.1080/19420862.2023.2180794] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 03/07/2023] Open
Abstract
Clinical studies have shown that combination therapy of antibodies targeting cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 (PD-1) significantly improves clinical benefit over PD-1 antibody alone. However, broad application of this combination has been limited by toxicities. Cadonilimab (AK104) is a symmetric tetravalent bispecific antibody with a crystallizable fragment (Fc)-null design. In addition to demonstrating biological activity similar to that of the combination of CTLA-4 and PD-1 antibodies, cadonilimab possess higher binding avidity in a high-density PD-1 and CTLA-4 setting than in a low-density PD-1 setting, while a mono-specific anti-PD-1 antibody does not demonstrate this differential activity. With no binding to Fc receptors, cadonilimab shows minimal antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and interleukin-6 (IL-6)/IL-8 release. These features all likely contribute to significantly lower toxicities of cadonilimab observed in the clinic. Higher binding avidity of cadonilimab in a tumor-like setting and Fc-null design may lead to better drug retention in tumors and contribute to better safety while achieving anti-tumor efficacy.
Collapse
Affiliation(s)
- Xinghua Pang
- Research and Development Department, Akeso Biopharma, Inc, Zhongshan, China
| | - Zhaoliang Huang
- Research and Development Department, Akeso Biopharma, Inc, Zhongshan, China
| | - Tingting Zhong
- Research and Development Department, Akeso Biopharma, Inc, Zhongshan, China
| | - Peng Zhang
- Research and Development Department, Akeso Biopharma, Inc, Zhongshan, China
| | | | - Michelle Xia
- Research and Development Department, Akeso Biopharma, Inc, Zhongshan, China
| | - Baiyong Li
- Research and Development Department, Akeso Biopharma, Inc, Zhongshan, China
| |
Collapse
|
16
|
Lin HJ, James I, Hyer CD, Haderlie CT, Zackrison MJ, Bateman TM, Berg M, Park JS, Daley SA, Zuniga Pina NR, Tseng YJJ, Moody JD, Price JC. Quantifying In Situ Structural Stabilities of Human Blood Plasma Proteins Using a Novel Iodination Protein Stability Assay. J Proteome Res 2022; 21:2920-2935. [PMID: 36356215 PMCID: PMC9724711 DOI: 10.1021/acs.jproteome.2c00323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Indexed: 11/12/2022]
Abstract
Many of the diseases that plague society today are driven by a loss of protein quality. One method to quantify protein quality is to measure the protein folding stability (PFS). Here, we present a novel mass spectrometry (MS)-based approach for PFS measurement, iodination protein stability assay (IPSA). IPSA quantifies the PFS by tracking the surface-accessibility differences of tyrosine, histidine, methionine, and cysteine under denaturing conditions. Relative to current methods, IPSA increases protein coverage and granularity to track the PFS changes of a protein along its sequence. To our knowledge, this study is the first time the PFS of human serum proteins has been measured in the context of the blood serum (in situ). We show that IPSA can quantify the PFS differences between different transferrin iron-binding states in near in vivo conditions. We also show that the direction of the denaturation curve reflects the in vivo surface accessibility of the amino acid residue and reproducibly reports a residue-specific PFS. Along with IPSA, we introduce an analysis tool Chalf that provides a simple workflow to calculate the residue-specific PFS. The introduction of IPSA increases the potential to use protein structural stability as a structural quality metric in understanding the etiology and progression of human disease. Data is openly available at Chorusproject.org (project ID 1771).
Collapse
Affiliation(s)
- Hsien-Jung
L. Lin
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Isabella James
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Chad D. Hyer
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Connor T. Haderlie
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Michael J. Zackrison
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Tyler M. Bateman
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Monica Berg
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Ji-Sun Park
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - S. Anisha Daley
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Nathan R. Zuniga Pina
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Yi-Jie J. Tseng
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - James D. Moody
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - John C. Price
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| |
Collapse
|
17
|
Comprehensive overview of autoantibody isotype and subclass distribution. J Allergy Clin Immunol 2022; 150:999-1010. [DOI: 10.1016/j.jaci.2022.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/27/2022] [Accepted: 05/13/2022] [Indexed: 11/06/2022]
|
18
|
Gou M, Qian N, Zhang Y, Yan H, Si H, Wang Z, Dai G. Fruquintinib in Combination With PD-1 Inhibitors in Patients With Refractory Non-MSI-H/pMMR Metastatic Colorectal Cancer: A Real-World Study in China. Front Oncol 2022; 12:851756. [PMID: 35875064 PMCID: PMC9300867 DOI: 10.3389/fonc.2022.851756] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/10/2022] [Indexed: 12/27/2022] Open
Abstract
BackgroundFruquintinib, a vascular endothelial growth factor receptor inhibitor, is a new anticancer drug independently developed in China to treat refractory metastatic colorectal cancer (mCRC). In Japan, regorafenib combined with nivolumab has been demonstrated to be promising in patients with refractory mCRC. Here, in a real-world study, we were aimed to evaluate the efficacy of fruquintinib with various programmed death-1 (PD-1) inhibitors after standard treatment in Chinese non-microsatellite instability-high (MSI-H)/mismatch repair proficient mCRC patients.MethodsA total of 45 patients with refractory mCRC were involved in the study. They received fruquintinib (3 or 5 mg, orally administered once a day for 3 weeks followed by 1 week off in 4-week cycles) and a PD-1 inhibitor(200 mg pembrolizumab, 3 mg/kg nivolumab, 200 mg sintilimab or camrelizumab, intravenously administered on D1 once every 3 weeks). Progression-free survival (PFS), overall survival (OS), disease control rate (DCR), and objective response rate (ORR) were reviewed and evaluated.ResultsAmong the 45 patients, the median age was 54 years (29-85). The ORR was 11.1% (5/45), DCR 62.2% (28/45), median PFS equal 3.8 months, and median OS was 14.9 months. The response duration was 3.4 months. PFS between left and right primary tumors and PFS with or without lung metastases were both not significantly different (p > 0.05), which was inconsistent with the result of REGONIVO study. The multivariate analysis indicated no association of OS benefit in the specified subgroups. No adverse-effect-related deaths were reported.ConclusionsFruquintinib, in combination with anti-PD-1, was observed to have clinical activity in a small population of patients with heavily pretreated mCRC in our center. Further studies are needed to verify this outcome in a large population.
Collapse
Affiliation(s)
- Miaomiao Gou
- Medical Oncology Department, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Niansong Qian
- Sanya Medical Center, Chinese People’s Liberation Army General Hospital, Sanya, China
| | - Yong Zhang
- Medical Oncology Department, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Huan Yan
- Medical Oncology Department, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Haiyan Si
- Medical Oncology Department, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Zhikuan Wang
- Medical Oncology Department, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: Guanghai Dai, ; Zhikuan Wang,
| | - Guanghai Dai
- Medical Oncology Department, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: Guanghai Dai, ; Zhikuan Wang,
| |
Collapse
|
19
|
Partridge MA, Chen J, Karayusuf EK, Sirimanne T, Stefan C, Lai CH, Gathani M, DeStefano L, Rozanski M, McAfee S, Rajadhyaksha M, Andisik MD, Torri A, Sumner G. Pre-existing Reactivity to an IgG4 Fc-Epitope: Characterization and Mitigation of Interference in a Bridging Anti-drug Antibody Assay. AAPS J 2022; 24:78. [DOI: 10.1208/s12248-022-00729-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
AbstractTwenty percent of baseline patient samples exhibited a pre-existing response in a bridging anti-drug antibody (ADA) assay for a human IgG4 monoclonal antibody (mAb) therapeutic. In some cases, assay signals were more than 100-fold higher than background, potentially confounding detection of true treatment-emergent ADA responses. The pre-existing reactivity was mapped by competitive inhibition experiments using recombinant proteins or chimeric human mAbs with IgG4 heavy chain regions swapped for IgG1 sequences. These experiments demonstrated that the majority of the samples had reactivity to an epitope containing leucine 445 in the CH3 domain of human IgG4. The pre-existing reactivity in baseline patient samples was mitigated by replacing the ADA assay capture reagent with a version of the drug containing a wild type IgG1 proline substitution at residue 445 without impacting detection of drug-specific, treatment-emergent ADA. Finally, purification on Protein G or anti-human IgG (H + L) columns indicated the pre-existing response was likely due to immunoglobulins in patient samples.
Graphical abstract
Collapse
|
20
|
Huang Z, Pang X, Zhong T, Qu T, Chen N, Ma S, He X, Xia D, Wang M, Xia M, Li B. Penpulimab, an Fc-Engineered IgG1 Anti-PD-1 Antibody, With Improved Efficacy and Low Incidence of Immune-Related Adverse Events. Front Immunol 2022; 13:924542. [PMID: 35833116 PMCID: PMC9272907 DOI: 10.3389/fimmu.2022.924542] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/30/2022] [Indexed: 12/19/2022] Open
Abstract
Background IgG4 anbibodies are deficient in stability and may contribute to tumor-associated escape from immune surveillance. We developed an IgG1 backbone anti-programmed cell death protein-1 (PD-1) antibody, penpulimab, which is designed to remove crystallizable fragment (Fc) gamma receptor (FcγR) binding that mediates antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and proinflammatory cytokine release. Methods Aggregation of different anti-PD-1 antibodies was tested by size exclusion chromatography, and melting temperature midpoint (Tm) and aggregation temperature onset (Tagg) were also determined. The affinity constants of penpulimab for PD-1 and human FcγRs were measured by surface plasmon resonance and biolayer interferometry. ADCC and ADCP were determined in cellular assays and antibody-dependent cytokine release (ADCR) from human macrophages was detected by ELISA. Binding kinetics of penpulimab to human PD-1 was determined by Biacore, and epitope/paratope mapping of PD-1/penpulimab was investigated using x-ray crystallography. Additionally, patients from six ongoing trials were included for analysis of immune-related adverse events (irAEs). Results Penpulimab demonstrated better stability and a lower level of host-cell protein residue compared with IgG4 backbone anti-PD-1 antibodies. As expected, penpulimab exhibited no apparent binding to FcγRIa, FcγRIIa_H131, FcγRIIIa_V158 and FcγRIIIa_F158, elicited no apparent ADCC and ADCP activities, and induced no remarkable IL-6 and IL-8 release by activated macrophages in vitro. Penpulimab was shown in the co-crystal study to bind to human PD-1 N-glycosylation site at N58 and had a slower off-rate from PD-1 versus nivolumab or pembrolizumab. Four hundred sixty-five patients were analyzed for irAEs. Fifteen (3.2%) patients had grade 3 or above irAEs. No death from irAEs was reported. Conclusions IgG1 backbone anti-PD1 antibody penpulimab has a good stability and reduced host cell protein residue, as well as potent binding to the antigen. Fc engineering has eliminated Fc-mediated effector functions of penpulimab including ADCC, ADCP and reduced ADCR, which may contribute to its more favorable safety profile. Clinical Trial Registration www.ClinicalTrials.gov, identifier: AK105-101: NCT03352531, AK105-201: NCT03722147, AK105-301: NCT03866980, AK105-202:NCT03866967, AK105-203: NCT04172571, AK105-204: NCT04172506.
Collapse
Affiliation(s)
- Zhaoliang Huang
- Research and Development Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| | - Xinghua Pang
- Research and Development Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| | - Tingting Zhong
- Research and Development Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| | - Tailong Qu
- Research and Development Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| | - Na Chen
- Research and Development Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| | - Shun Ma
- Chemical Manufacturing and Control Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| | - Xinrong He
- Chemical Manufacturing and Control Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| | - Dennis Xia
- Manufacturing and Quality Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| | - Max Wang
- Procurement and Sourcing Department and Clinical Operation Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| | | | - Baiyong Li
- Research and Development Department, Akeso Biopharma Co., Ltd., Zhongshan, China
| |
Collapse
|
21
|
Panhuber A, Lamorte G, Bruno V, Cetin H, Bauer W, Höftberger R, Erber AC, Frommlet F, Koneczny I. A systematic review and meta-analysis of HLA class II associations in patients with IgG4 autoimmunity. Sci Rep 2022; 12:9229. [PMID: 35654912 PMCID: PMC9163138 DOI: 10.1038/s41598-022-13042-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/13/2022] [Indexed: 11/22/2022] Open
Abstract
Autoimmune diseases caused by pathogenic IgG4 subclass autoantibodies (IgG4-AID) include diseases like MuSK myasthenia gravis, pemphigus vulgaris or thrombotic thrombocytopenic purpura. Their etiology is still unknown. Polymorphisms in the human leukocyte antigen (HLA) gene locus, particularly in HLA-DRB1, are known genetic susceptibility factors for autoimmune diseases. We hypothesized a similar role for HLA polymorphisms in IgG4-AID and conducted a systematic review and meta-analysis with case-control studies on IgG4-AID based on MOOSE/ HuGENet guidelines. Genotype (G) and allele (A) frequencies of HLA-DQB1*05 (G: OR 3.8; 95% CI 2.44-5.9; p < 0.00001; A: OR 2.54; 95% CI 1.82-3.55; p < 0.00001) and HLA-DRB1*14 (G: OR 4.31; 95% CI 2.82-6.59; p < 0.00001; A: OR 4.78; 95% CI 3.52-6.49; p < 0.00001) and the HLA-DRB1*14-DQB1*05 haplotype (OR 6.3; 95% CI 3.28-12.09; p < 0.00001/OR 4.98; 95% CI 3.8-6.53; p < 0.00001) were increased while HLA-DRB1*13 (G: OR 0.48; 95% CI 0.34-0.68; p < 0.0001; A: OR 0.46; 95% CI 0.34-0.62; p < 0.00001) was decreased in IgG4-AID patients. In conclusion, the HLA-DQB1*05, HLA-DRB1*14 alleles and the HLA-DQB1*05-DRB1*14 haplotype could be genetic risk factors that predispose for the production of pathogenic IgG4 autoantibodies and the HLA-DRB1*13 allele may protect from IgG4 autoimmunity.
Collapse
Affiliation(s)
- Anja Panhuber
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Giovanni Lamorte
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Veronica Bruno
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Hakan Cetin
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Astrid C Erber
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Florian Frommlet
- Center for Medical Statistics Informatics and Intelligent Systems, Section for Medical Statistics, Medical University of Vienna, Vienna, Austria
| | - Inga Koneczny
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
22
|
Koneczny I, Tzartos J, Mané-Damas M, Yilmaz V, Huijbers MG, Lazaridis K, Höftberger R, Tüzün E, Martinez-Martinez P, Tzartos S, Leypoldt F. IgG4 Autoantibodies in Organ-Specific Autoimmunopathies: Reviewing Class Switching, Antibody-Producing Cells, and Specific Immunotherapies. Front Immunol 2022; 13:834342. [PMID: 35401530 PMCID: PMC8986991 DOI: 10.3389/fimmu.2022.834342] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/28/2022] [Indexed: 12/24/2022] Open
Abstract
Organ-specific autoimmunity is often characterized by autoantibodies targeting proteins expressed in the affected tissue. A subgroup of autoimmunopathies has recently emerged that is characterized by predominant autoantibodies of the IgG4 subclass (IgG4-autoimmune diseases; IgG4-AID). This group includes pemphigus vulgaris, thrombotic thrombocytopenic purpura, subtypes of autoimmune encephalitis, inflammatory neuropathies, myasthenia gravis and membranous nephropathy. Although the associated autoantibodies target specific antigens in different organs and thus cause diverse syndromes and diseases, they share surprising similarities in genetic predisposition, disease mechanisms, clinical course and response to therapies. IgG4-AID appear to be distinct from another group of rare immune diseases associated with IgG4, which are the IgG4-related diseases (IgG4-RLD), such as IgG4-related which have distinct clinical and serological properties and are not characterized by antigen-specific IgG4. Importantly, IgG4-AID differ significantly from diseases associated with IgG1 autoantibodies targeting the same organ. This may be due to the unique functional characteristics of IgG4 autoantibodies (e.g. anti-inflammatory and functionally monovalent) that affect how the antibodies cause disease, and the differential response to immunotherapies of the IgG4 producing B cells/plasmablasts. These clinical and pathophysiological clues give important insight in the immunopathogenesis of IgG4-AID. Understanding IgG4 immunobiology is a key step towards the development of novel, IgG4 specific treatments. In this review we therefore summarize current knowledge on IgG4 regulation, the relevance of class switching in the context of health and disease, describe the cellular mechanisms involved in IgG4 production and provide an overview of treatment responses in IgG4-AID.
Collapse
Affiliation(s)
- Inga Koneczny
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - John Tzartos
- Neuroimmunology, Tzartos NeuroDiagnostics, Athens, Greece
- 2nd Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marina Mané-Damas
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Vuslat Yilmaz
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Maartje G. Huijbers
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - Konstantinos Lazaridis
- Department of Immunology, Laboratory of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Pilar Martinez-Martinez
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Socrates Tzartos
- Neuroimmunology, Tzartos NeuroDiagnostics, Athens, Greece
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Frank Leypoldt
- Neuroimmunology, Institute of Clinical Chemistry and Department of Neurology, UKSH Kiel/Lübeck, Kiel University, Kiel, Germany
| |
Collapse
|
23
|
Lai PK, Ghag G, Yu Y, Juan V, Fayadat-Dilman L, Trout BL. Differences in human IgG1 and IgG4 S228P monoclonal antibodies viscosity and self-interactions: Experimental assessment and computational predictions of domain interactions. MAbs 2021; 13:1991256. [PMID: 34747330 PMCID: PMC8583000 DOI: 10.1080/19420862.2021.1991256] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Human/humanized IgG4 antibodies have reduced effector function relative to IgG1 antibodies, which is desirable for certain therapeutic purposes. However, the developability and biophysical properties for IgG4 antibodies are not well understood. This work focuses on the head-to-head comparison of key biophysical properties, such as self-interaction and viscosity, for 14 human/humanized, and chimeric IgG1 and IgG4 S228P monoclonal antibody pairs that contain the identical variable regions. Experimental measurements showed that the IgG4 S228P antibodies have similar or higher self-interaction and viscosity than that of IgG1 antibodies in 20 mM sodium acetate, pH 5.5. We report sequence and structural drivers for the increased viscosity and self-interaction detected in IgG4 S228P antibodies through a combination of experimental data and computational models. Further, we applied and extended a previously established computational model for IgG1 antibodies to predict the self-interaction and viscosity behavior for each antibody pair, providing insight into the structural characteristics and differences of these two isotypes. Interestingly, we observed that the IgG4 S228P swapped variants, where the CH3 domain was swapped for that of an IgG1, showed reduced self-interaction behavior. These domain swapped IgG4 S228P molecules also showed reduced viscosity from experiment and coarse-grained simulations. We also observed that experimental diffusion interaction parameter (kD) values have a high correlation with computational diffusivity prediction for both IgG1 and IgG4 S228P isotypes. Abbreviations: AHc, constant region Hamaker constant; AHv, variable region Hamaker constant; CDRs, Complementarity-determining regions; CG, Coarse-grained model; CH1, Constant heavy chain 1; CH2 Constant heavy chain 2; CH3 Constant heavy chain 3; chgCH3 Effective charge on the CH3 region; CL Constant light chain; cP, Centipoise; DLS, Dynamic light scattering; Fab, Fragment antigen-binding; Fc, Fragment crystallizable; Fv, Variable domaing; (r) Radial distribution function; H1 CDR1 of Heavy Chain; H2 CDR2 of Heavy Chain; H3 CDR3 of Heavy Chain; HVI, High viscosity index; IgG1 human immunoglobulin of IgG1 subclass; IgG4 human immunoglobulin of IgG4 subclass; kD, Diffusion interaction parameter; L1 CDR1 of Light Chain; L2 CDR2 of Light Chain; L3 CDR3 of Light Chain; mAb, Monoclonal antibody; MD, Molecular dynamics; PPI Protein–protein interactions; SCM, Spatial charge map; UP-SEC, Ultra-high-performance size-exclusion chromatography; VH, Variable domain of Heavy Chain; VL, Variable domain of Light Chain
Collapse
Affiliation(s)
- Pin-Kuang Lai
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts USA.,Current Address: Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, Hoboken, New Jersey USA
| | - Gaurav Ghag
- Merck & Co, Discovery Biologics, Protein Sciences Department, South San Francisco, CA , USA
| | - Yao Yu
- Merck & Co, Discovery Biologics, Protein Sciences Department, South San Francisco, CA , USA
| | - Veronica Juan
- Merck & Co, Discovery Biologics, Protein Sciences Department, South San Francisco, CA , USA
| | | | - Bernhardt L Trout
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts USA
| |
Collapse
|
24
|
Spiteri VA, Goodall M, Doutch J, Rambo RP, Gor J, Perkins SJ. Solution structures of human myeloma IgG3 antibody reveal extended Fab and Fc regions relative to the other IgG subclasses. J Biol Chem 2021; 297:100995. [PMID: 34302810 PMCID: PMC8371214 DOI: 10.1016/j.jbc.2021.100995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/08/2021] [Accepted: 07/19/2021] [Indexed: 11/21/2022] Open
Abstract
Human immunoglobulin G subclass 3 (IgG3) possesses a uniquely long hinge region that separates its Fab antigen-binding and Fc receptor-binding regions. Owing to this hinge length, the molecular structure of full-length IgG3 remains elusive, and the role of the two conserved Fc glycosylation sites are unknown. To address these issues, we subjected glycosylated and deglycosylated human myeloma IgG3 to multidisciplinary solution structure studies. Using analytical ultracentrifugation, the elongated structure of IgG3 was determined from the reduced sedimentation coefficients s020,w of 5.82 to 6.29 S for both glycosylated and deglycosylated IgG3. X-ray and neutron scattering showed that the Guinier RG values were 6.95 nm for glycosylated IgG3 and were unchanged after deglycosylation, again indicating an elongated structure. The distance distribution function P(r) showed a maximum length of 25 to 28 nm and three distinct maxima. The molecular structure of IgG3 was determined using atomistic modeling based on molecular dynamics simulations of the IgG3 hinge and Monte Carlo simulations to identify physically realistic arrangements of the Fab and Fc regions. This resulted in libraries containing 135,135 and 73,905 glycosylated and deglycosylated IgG3 structures, respectively. Comparisons with the X-ray and neutron scattering curves gave 100 best-fit models for each form of IgG3 that accounted for the experimental scattering curves. These models revealed the first molecular structures for full-length IgG3. The structures exhibited relatively restricted Fab and Fc conformations joined by an extended semirigid hinge, which explains the potent effector functions of IgG3 relative to the other subclasses IgG1, IgG2, and IgG4.
Collapse
Affiliation(s)
- Valentina A Spiteri
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Margaret Goodall
- Institute for Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - James Doutch
- ISIS Facility, STFC Rutherford Appleton Laboratory, Harwell Campus, Didcot, Oxfordshire, United Kingdom
| | - Robert P Rambo
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, United Kingdom
| | - Jayesh Gor
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Stephen J Perkins
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom.
| |
Collapse
|
25
|
An Overview of the Relevance of IgG4 Antibodies in Allergic Disease with a Focus on Food Allergens. CHILDREN-BASEL 2021; 8:children8050418. [PMID: 34065166 PMCID: PMC8160978 DOI: 10.3390/children8050418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 12/25/2022]
Abstract
Antibodies of the IgG4 isotype are strongly associated with allergic disease but have several properties such as not precipitating with allergens, not activating complement and poor binding to Fcγ receptors that argue against a pro-inflammatory role. In keeping with that, IgG4 antibodies are a striking feature of the response to immunotherapy. In two naturally occurring situations IgG4 antibodies are common with low or absent IgE antibodies. The first example is children raised in a house with a cat and the second is eosinophilic esophagitis (EoE). In many population-based cohorts, the ownership of a cat in early childhood is associated with a decreased prevalence of a cat allergy at age 10. The second example (i.e., EoE) is a novel form of food allergy that is not mediated by IgE and is related to consuming cow’s milk or wheat. In EoE, patients have IgG4 to milk proteins in high > 10 µg/mL or very high > 100 µg/mL titers. Enigmatically these patients are found to have deposits of IgG4 in the wall of their inflamed esophagus. The factors that have given rise to EoE remain unclear; however, changes in food processing over the past 50 years, particularly ultra-heat treatment and the high pressure homogenization of milk, represent a logical hypothesis.
Collapse
|
26
|
Wei B, Gao X, Cadang L, Izadi S, Liu P, Zhang HM, Hecht E, Shim J, Magill G, Pabon JR, Dai L, Phung W, Lin E, Wang C, Whang K, Sanchez S, Oropeza J, Camperi J, Zhang J, Sandoval W, Zhang YT, Jiang G. Fc galactosylation follows consecutive reaction kinetics and enhances immunoglobulin G hexamerization for complement activation. MAbs 2021; 13:1893427. [PMID: 33682619 PMCID: PMC7946005 DOI: 10.1080/19420862.2021.1893427] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Fc galactosylation is a critical quality attribute for anti-tumor recombinant immunoglobulin G (IgG)-based monoclonal antibody (mAb) therapeutics with complement-dependent cytotoxicity (CDC) as the mechanism of action. Although the correlation between galactosylation and CDC has been known, the underlying structure–function relationship is unclear. Heterogeneity of the Fc N-glycosylation produced by Chinese hamster ovary (CHO) cell culture biomanufacturing process leads to variable CDC potency. Here, we derived a kinetic model of galactose transfer reaction in the Golgi apparatus and used this model to determine the correlation between differently galactosylated species from CHO cell culture process. The model was validated by a retrospective data analysis of more than 800 historical samples from small-scale and large-scale CHO cell cultures. Furthermore, using various analytical technologies, we discovered the molecular basis for Fc glycan terminal galactosylation changing the three-dimensional conformation of the Fc, which facilitates the IgG1 hexamerization, thus enhancing C1q avidity and subsequent complement activation. Our study offers insight into the formation of galactosylated species, as well as a novel three-dimensional understanding of the structure–function relationship of terminal galactose to complement activation in mAb therapeutics.
Collapse
Affiliation(s)
- Bingchuan Wei
- Protein Analytical Chemistry, Genentech Inc., South San Francisco,United States.,Small Molecule Analytical Chemistry, Genentech Inc, South San Francisco, United States
| | - Xuan Gao
- Biological Technologies, Genentech Inc., South San Francisco, United States
| | - Lance Cadang
- Protein Analytical Chemistry, Genentech Inc., South San Francisco,United States
| | - Saeed Izadi
- Pharmaceutical Development, Genentech Inc., South San Francisco, United States
| | - Peilu Liu
- Protein Analytical Chemistry, Genentech Inc., South San Francisco,United States.,Department of Chemistry and Biochemistry, Florida State University,Florida, United States
| | - Hui-Min Zhang
- Protein Analytical Chemistry, Genentech Inc., South San Francisco,United States
| | - Elizabeth Hecht
- Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc., South San Francisco, United States
| | - Jeongsup Shim
- Biological Technologies, Genentech Inc., South San Francisco, United States
| | - Gordon Magill
- Department of Cell Culture and Bioprocess Operations, Genentech Inc., South San Francisco, United States
| | - Juan Rincon Pabon
- Protein Analytical Chemistry, Genentech Inc., South San Francisco,United States.,Department of Chemistry, University of Kansas, Lawrence United States
| | - Lu Dai
- Protein Analytical Chemistry, Genentech Inc., South San Francisco,United States
| | - Wilson Phung
- Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc., South San Francisco, United States
| | - Elaine Lin
- Biological Technologies, Genentech Inc., South San Francisco, United States
| | - Christopher Wang
- Biological Technologies, Genentech Inc., South San Francisco, United States
| | - Kevin Whang
- Biological Technologies, Genentech Inc., South San Francisco, United States
| | - Sean Sanchez
- Biological Technologies, Genentech Inc., South San Francisco, United States
| | - Jose Oropeza
- Biological Technologies, Genentech Inc., South San Francisco, United States
| | - Julien Camperi
- Protein Analytical Chemistry, Genentech Inc., South San Francisco,United States
| | - Jennifer Zhang
- Protein Analytical Chemistry, Genentech Inc., South San Francisco,United States
| | - Wendy Sandoval
- Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc., South San Francisco, United States
| | | | - Guoying Jiang
- Biological Technologies, Genentech Inc., South San Francisco, United States
| |
Collapse
|
27
|
Lai PK, Fernando A, Cloutier TK, Gokarn Y, Zhang J, Schwenger W, Chari R, Calero-Rubio C, Trout BL. Machine Learning Applied to Determine the Molecular Descriptors Responsible for the Viscosity Behavior of Concentrated Therapeutic Antibodies. Mol Pharm 2021; 18:1167-1175. [PMID: 33450157 DOI: 10.1021/acs.molpharmaceut.0c01073] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Predicting the solution viscosity of monoclonal antibody (mAb) drug products remains as one of the main challenges in antibody drug design, manufacturing, and delivery. In this work, the concentration-dependent solution viscosity of 27 FDA-approved mAbs was measured at pH 6.0 in 10 mM histidine-HCl. Six mAbs exhibited high viscosity (>30 cP) in solutions at 150 mg/mL mAb concentration. Combining molecular modeling and machine learning feature selection, we found that the net charge in the mAbs and the amino acid composition in the Fv region are key features which govern the viscosity behavior. For mAbs whose behavior was not dominated by charge effects, we observed that high viscosity is correlated with more hydrophilic and fewer hydrophobic residues in the Fv region. A predictive model based on the net charges of mAbs and a high viscosity index is presented as a fast screening tool for classifying low- and high-viscosity mAbs.
Collapse
Affiliation(s)
- Pin-Kuang Lai
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Amendra Fernando
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Theresa K Cloutier
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Yatin Gokarn
- Biologics Development, Sanofi, Framingham, Massachusetts 01701, United States
| | - Jifeng Zhang
- Biologics Development, Sanofi, Framingham, Massachusetts 01701, United States
| | - Walter Schwenger
- Biologics Development, Sanofi, Framingham, Massachusetts 01701, United States
| | - Ravi Chari
- Biologics Development, Sanofi, Framingham, Massachusetts 01701, United States
| | - Cesar Calero-Rubio
- Biologics Development, Sanofi, Framingham, Massachusetts 01701, United States
| | - Bernhardt L Trout
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
28
|
Yamaguchi Y, Barb AW. A synopsis of recent developments defining how N-glycosylation impacts immunoglobulin G structure and function. Glycobiology 2020; 30:214-225. [PMID: 31822882 DOI: 10.1093/glycob/cwz068] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/26/2019] [Accepted: 08/21/2019] [Indexed: 12/21/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are the fastest growing group of drugs with 11 new antibodies or antibody-drug conjugates approved by the Food and Drug Administration in 2018. Many mAbs require effector function for efficacy, including antibody-dependent cell-mediated cytotoxicity triggered following contact of an immunoglobulin G (IgG)-coated particle with activating crystallizable fragment (Fc) γ receptors (FcγRs) expressed by leukocytes. Interactions between IgG1 and the FcγRs require post-translational modification of the Fc with an asparagine-linked carbohydrate (N-glycan). Though the structure of IgG1 Fc and the role of Fc N-glycan composition on disease were known for decades, the underlying mechanism of how the N-glycan affected FcγR binding was not defined until recently. This review will describe the current understanding of how N-glycosylation impacts the structure and function of the IgG1 Fc and describe new techniques that are poised to provide the next critical breakthroughs.
Collapse
Affiliation(s)
| | - Adam W Barb
- Department of Biochemistry and Molecular Biology and Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, 30602
| |
Collapse
|
29
|
Koneczny I. Update on IgG4-mediated autoimmune diseases: New insights and new family members. Autoimmun Rev 2020; 19:102646. [PMID: 32801046 DOI: 10.1016/j.autrev.2020.102646] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/08/2020] [Indexed: 12/23/2022]
Abstract
Antibodies of IgG4 subclass are exceptional players of the immune system, as they are considered to be immunologically inert and functionally monovalent, and as such may be part of classical tolerance mechanisms. IgG4 antibodies are found in a range of different diseases, including IgG4-related diseases, allergy, cancer, rheumatoid arthritis, helminth infection and IgG4 autoimmune diseases, where they may be pathogenic or protective. IgG4 autoimmune diseases are an emerging new group of diseases that are characterized by pathogenic, antigen-specific autoantibodies of IgG4 subclass, such as MuSK myasthenia gravis, pemphigus vulgaris and thrombotic thrombocytopenic purpura. The list of IgG4 autoantigens is rapidly growing and to date contains 29 candidate antigens. Interestingly, IgG4 autoimmune diseases are restricted to four distinct organs: 1) the central and peripheral nervous system, 2) the kidney, 3) the skin and mucous membranes and 4) the vascular system and soluble antigens in the blood circulation. The pathogenicity of IgG4 can be validated using our classification system, and is usually excerted by functional blocking of protein-protein interaction.
Collapse
Affiliation(s)
- Inga Koneczny
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
30
|
Wang H, Xu Q, Zhao C, Zhu Z, Zhu X, Zhou J, Zhang S, Yang T, Zhang B, Li J, Yan M, Liu R, Ma C, Quan Y, Zhang Y, Zhang W, Geng Y, Chen C, Chen S, Liu D, Chen Y, Tian D, Su M, Chen X, Gu J. An immune evasion mechanism with IgG4 playing an essential role in cancer and implication for immunotherapy. J Immunother Cancer 2020; 8:jitc-2020-000661. [PMID: 32819973 PMCID: PMC7443307 DOI: 10.1136/jitc-2020-000661] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Recent impressive advances in cancer immunotherapy have been largely derived from cellular immunity. The role of humoral immunity in carcinogenesis has been less understood. Based on our previous observations we hypothesize that an immunoglobulin subtype IgG4 plays an essential role in cancer immune evasion. METHODS The distribution, abundance, actions, properties and possible mechanisms of IgG4 were investigated with human cancer samples and animal tumor models with an extensive array of techniques both in vitro and in vivo. RESULTS In a cohort of patients with esophageal cancer we found that IgG4-containing B lymphocytes and IgG4 concentration were significantly increased in cancer tissue and IgG4 concentrations increased in serum of patients with cancer. Both were positively related to increased cancer malignancy and poor prognoses, that is, more IgG4 appeared to associate with more aggressive cancer growth. We further found that IgG4, regardless of its antigen specificity, inhibited the classic immune reactions of antibody-dependent cell-mediated cytotoxicity, antibody-dependent cellular phagocytosis and complement-dependent cytotoxicity against cancer cells in vitro, and these effects were obtained through its Fc fragment reacting to the Fc fragments of cancer-specific IgG1 that has been bound to cancer antigens. We also found that IgG4 competed with IgG1 in reacting to Fc receptors of immune effector cells. Therefore, locally increased IgG4 in cancer microenvironment should inhibit antibody-mediated anticancer responses and help cancer to evade local immune attack and indirectly promote cancer growth. This hypothesis was verified in three different immune potent mouse models. We found that local application of IgG4 significantly accelerated growth of inoculated breast and colorectal cancers and carcinogen-induced skin papilloma. We also tested the antibody drug for cancer immunotherapy nivolumab, which was IgG4 in nature with a stabilizing S228P mutation, and found that it significantly promoted cancer growth in mice. This may provide an explanation to the newly appeared hyperprogressive disease sometimes associated with cancer immunotherapy. CONCLUSION There appears to be a previously unrecognized immune evasion mechanism with IgG4 playing an essential role in cancer microenvironment with implications in cancer diagnosis and immunotherapy.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Qian Xu
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Chanyuan Zhao
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Ziqi Zhu
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Xiaoqing Zhu
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Junjie Zhou
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Shuming Zhang
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Tiqun Yang
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Biying Zhang
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Jun Li
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Meiling Yan
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Renming Liu
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Changchun Ma
- Department of Radiation Oncology, Affiliated Cancer Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Yan Quan
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Yongqu Zhang
- Department of Breast Center, Affiliated Cancer Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Weifeng Zhang
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Yiqun Geng
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Chuangzhen Chen
- Department of Radiation Oncology, Affiliated Cancer Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Shaobin Chen
- Department of Thoracic Surgery, Affiliated Cancer Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Ditian Liu
- Department of Thoracic Surgery, Affiliated Cancer Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Yuping Chen
- Department of Thoracic Surgery, Affiliated Cancer Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Dongping Tian
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Min Su
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Xueling Chen
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
| | - Jiang Gu
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
- Jinxin Research Institute for Reproductive Medicine and Genetics, Jinjiang Hospital for Maternal and Child Health Care, Chengdu, China
| |
Collapse
|
31
|
Hsieh SC, Shen CY, Liao HT, Chen MH, Wu CH, Li KJ, Lu CS, Kuo YM, Tsai HC, Tsai CY, Yu CL. The Cellular and Molecular Bases of Allergy, Inflammation and Tissue Fibrosis in Patients with IgG4-related Disease. Int J Mol Sci 2020; 21:ijms21145082. [PMID: 32708432 PMCID: PMC7404109 DOI: 10.3390/ijms21145082] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 12/16/2022] Open
Abstract
IgG4-related disease (IgG4-RD) is a spectrum of complex fibroinflammatory disorder with protean manifestations mimicking malignant neoplasms, infectious or non-infectious inflammatory process. The histopathologic features of IgG4-RD include lymphoplasmacytic infiltration, storiform fibrosis and obliterative phlebitis together with increased in situ infiltration of IgG4 bearing-plasma cells which account for more than 40% of all IgG-producing B cells. IgG4-RD can also be diagnosed based on an elevated serum IgG4 level of more than 110 mg/dL (normal < 86.5 mg/mL in adult) in conjunction with protean clinical manifestations in various organs such as pancreato–hepatobiliary inflammation with/without salivary/lacrimal gland enlargement. In the present review, we briefly discuss the role of genetic predisposition, environmental factors and candidate autoantibodies in the pathogenesis of IgG4-RD. Then, we discuss in detail the immunological paradox of IgG4 antibody, the mechanism of modified Th2 response for IgG4 rather than IgE antibody production and the controversial issues in the allergic reactions of IgG4-RD. Finally, we extensively review the implications of different immune-related cells, cytokines/chemokines/growth factors and Toll-like as well as NOD-like receptors in the pathogenesis of tissue fibro-inflammatory reactions. Our proposals for the future investigations and prospective therapeutic strategies for IgG4-RD are shown in the last part.
Collapse
Affiliation(s)
- Song-Chou Hsieh
- Division of Rheumatology, Immunology & Allergy, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan; (S.-C.H.); (C.-Y.S.); (C.-H.W.); (K.-J.L.); (C.-S.L.); (Y.-M.K.)
| | - Chieh-Yu Shen
- Division of Rheumatology, Immunology & Allergy, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan; (S.-C.H.); (C.-Y.S.); (C.-H.W.); (K.-J.L.); (C.-S.L.); (Y.-M.K.)
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, Taipei 11217, Taiwan; (H.-T.L.); (M.-H.C.); (H.-C.T.)
| | - Ming-Han Chen
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, Taipei 11217, Taiwan; (H.-T.L.); (M.-H.C.); (H.-C.T.)
| | - Cheng-Han Wu
- Division of Rheumatology, Immunology & Allergy, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan; (S.-C.H.); (C.-Y.S.); (C.-H.W.); (K.-J.L.); (C.-S.L.); (Y.-M.K.)
| | - Ko-Jen Li
- Division of Rheumatology, Immunology & Allergy, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan; (S.-C.H.); (C.-Y.S.); (C.-H.W.); (K.-J.L.); (C.-S.L.); (Y.-M.K.)
| | - Cheng-Shiun Lu
- Division of Rheumatology, Immunology & Allergy, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan; (S.-C.H.); (C.-Y.S.); (C.-H.W.); (K.-J.L.); (C.-S.L.); (Y.-M.K.)
| | - Yu-Min Kuo
- Division of Rheumatology, Immunology & Allergy, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan; (S.-C.H.); (C.-Y.S.); (C.-H.W.); (K.-J.L.); (C.-S.L.); (Y.-M.K.)
| | - Hung-Cheng Tsai
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, Taipei 11217, Taiwan; (H.-T.L.); (M.-H.C.); (H.-C.T.)
| | - Chang-Youh Tsai
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, Taipei 11217, Taiwan; (H.-T.L.); (M.-H.C.); (H.-C.T.)
- Correspondence: (C.-Y.T.); (C.-L.Y.); Tel.: +886-2-28712121 (ext. 3366) (C.-Y.T.); +886-2-23123456 (ext. 65011) (C.-L.Y.)
| | - Chia-Li Yu
- Division of Rheumatology, Immunology & Allergy, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan; (S.-C.H.); (C.-Y.S.); (C.-H.W.); (K.-J.L.); (C.-S.L.); (Y.-M.K.)
- Correspondence: (C.-Y.T.); (C.-L.Y.); Tel.: +886-2-28712121 (ext. 3366) (C.-Y.T.); +886-2-23123456 (ext. 65011) (C.-L.Y.)
| |
Collapse
|
32
|
Vankemmelbeke M, McIntosh RS, Chua JX, Kirk T, Daniels I, Patsalidou M, Moss R, Parsons T, Scott D, Harris G, Ramage JM, Spendlove I, Durrant LG. Engineering the Human Fc Region Enables Direct Cell Killing by Cancer Glycan-Targeting Antibodies without the Need for Immune Effector Cells or Complement. Cancer Res 2020; 80:3399-3412. [PMID: 32532823 DOI: 10.1158/0008-5472.can-19-3599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/25/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022]
Abstract
Murine IgG3 glycan-targeting mAb often induces direct cell killing in the absence of immune effector cells or complement via a proinflammatory mechanism resembling oncotic necrosis. This cancer cell killing is due to noncovalent association between Fc regions of neighboring antibodies, resulting in enhanced avidity. Human isotypes do not contain the residues underlying this cooperative binding mode; consequently, the direct cell killing of mouse IgG3 mAb is lost upon chimerization or humanization. Using the Lewisa/c/x -targeting 88mAb, we identified the murine IgG3 residues underlying the direct cell killing and increased avidity via a series of constant region shuffling and subdomain swapping approaches to create improved ("i") chimeric mAb with enhanced tumor killing in vitro and in vivo. Constant region shuffling identified a major CH3 and a minor CH2 contribution, which was further mapped to discontinuous regions among residues 286-306 and 339-378 that, when introduced in 88hIgG1, recapitulated the direct cell killing and avidity of 88mIgG3. Of greater interest was the creation of a sialyl-di-Lewisa-targeting i129G1 mAb via introduction of these selected residues into 129hIgG1, converting it into a direct cell killing mAb with enhanced avidity and significant in vivo tumor control. The human iG1 mAb, termed Avidimabs, retained effector functions, paving the way for the proinflammatory direct cell killing to promote antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity through relief of immunosuppression. Ultimately, Fc engineering of human glycan-targeting IgG1 mAb confers proinflammatory direct cell killing and enhanced avidity, an approach that could be used to improve the avidity of other mAb with therapeutic potential. SIGNIFICANCE: Fc engineering enhances avidity and direct cell killing of cancer-targeting anti-glycan antibodies to create superior clinical candidates for cancer immunotherapy.
Collapse
Affiliation(s)
- Mireille Vankemmelbeke
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom.,Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Richard S McIntosh
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Jia Xin Chua
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom.,Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Thomas Kirk
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom.,Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Ian Daniels
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Marilena Patsalidou
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Robert Moss
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Tina Parsons
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - David Scott
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, United Kingdom
| | - Gemma Harris
- Research Complex at Harwell, Rutherford Appleton Laboratory, Didcot, United Kingdom
| | - Judith M Ramage
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Ian Spendlove
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Lindy G Durrant
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom. .,Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| |
Collapse
|
33
|
Boffa JJ, Esteve E, Buob D. Renal involvement in IgG4-related disease. Presse Med 2020; 49:104017. [PMID: 32234380 DOI: 10.1016/j.lpm.2020.104017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/04/2019] [Accepted: 07/04/2019] [Indexed: 10/24/2022] Open
Abstract
IgG4-RD may affect several organs including kidneys. The kidney is involved in approximately 20% of patient with IgG4-RD. The most common intrinsic kidney disease is tubulointerstitial nephritis (IgG4-TIN). Retroperitoneal fibrosis (IgG4-RPF) may induce obstructive acute renal failure. More rarely, IgG4-RKD can manifest as a glomerular disease, in particular as a membranous nephropathy (MN). It mostly affects middle-aged to elderly men and causes acute or chronic renal dysfunction, multiple hypodense lesions on CT-Scan and various extra-renal lesions. Increased serum IgG4 and hypocomplementemia are the most important serological findings for the diagnosis of IgG4-RD and thus should be systematically assessed when IgG4-RKD is suspected. Specific diagnosis criteria for IgG4-TIN including interstitial infiltration of IgG4-positive plasma cells, storiform fibrosis and tubular basement membrane immune complex deposits have been proposed. Corticosteroids are effective and remain the first-line therapy but relapses or severe forms could respond to immunosuppressive therapy.
Collapse
Affiliation(s)
- Jean-Jacques Boffa
- Inserm UMRS 1155, department of nephrology, Sorbonne université, hôpital Tenon, AP-HP, 75020 Paris, France.
| | - Emmanuel Esteve
- Inserm UMRS 1155, department of nephrology, Sorbonne université, hôpital Tenon, AP-HP, 75020 Paris, France
| | - David Buob
- Inserm UMRS 1155, department of pathology, Sorbonne université, hôpital Tenon, AP-HP, 75020 Paris, France
| |
Collapse
|
34
|
R409K mutation prevents acid-induced aggregation of human IgG4. PLoS One 2020; 15:e0229027. [PMID: 32182240 PMCID: PMC7077836 DOI: 10.1371/journal.pone.0229027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/28/2020] [Indexed: 11/20/2022] Open
Abstract
Human immunoglobulin G isotype 4 (IgG4) antibodies are suitable for use in either the antagonist or agonist format because their low effector functions prevent target cytotoxicity or unwanted cytokine secretion. However, while manufacturing therapeutic antibodies, they are exposed to low pH during purification, and IgG4 is more susceptible to low-pH-induced aggregation than IgG1. Therefore, we investigated the underlying mechanisms of IgG4 aggregation at low pH and engineered an IgG4 with enhanced stability. By swapping the constant regions of IgG1 and IgG4, we determined that the constant heavy chain (CH3) domain is critical for aggregate formation, but a core-hinge-stabilizing S228P mutation in IgG4 is insufficient for preventing aggregation. To identify the aggregation-prone amino acid, we substituted the CH3 domain of IgG4 with that of IgG1, changing IgG4 Arg409 to a Lys, thereby preventing the aggregation of the IgG4 variant as effectively as in IgG1. A stabilizing effect was also recorded with other variable-region variants. Analysis of thermal stability using differential scanning calorimetry revealed that the R409K substitution increased the Tm value of CH3, suggesting that the R409K mutation contributed to the structural strengthening of the CH3-CH3 interaction. The R409K mutation did not influence the binding to antigens/human Fcγ receptors; whereas, the concurrent S228P and R409K mutations in IgG4 suppressed Fab-arm exchange drastically and as effectively as in IgG1, in both in vitro and in vivo in mice models. Our findings suggest that the IgG4 R409K variant represents a potential therapeutic IgG for use in low-effector-activity format that exhibits increased stability.
Collapse
|
35
|
Feng Y, Wang Y, Zhang S, Haneef K, Liu W. Structural and immunogenomic insights into B-cell receptor activation. J Genet Genomics 2020; 47:27-35. [PMID: 32111437 DOI: 10.1016/j.jgg.2019.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/10/2019] [Accepted: 12/09/2019] [Indexed: 02/08/2023]
Abstract
B cells express B-cell receptors (BCRs) which recognize antigen to trigger signaling cascades for B-cell activation and subsequent antibody production. BCR activation has a crucial influence on B-cell fate. How BCR is activated upon encountering antigen remains to be solved, although tremendous progresses have been achieved in the past few years. Here, we summarize the models that have been proposed to explain BCR activation, including the cross-linking model, the conformation-induced oligomerization model, the dissociation activation model, and the conformational change model. Especially, we elucidate the partially resolved structures of antibodies and/or BCRs by far and discusse how these current structural and further immunogenomic messages and more importantly the future studies may shed light on the explanation of BCR activation and the relevant diseases in the case of dysregulation.
Collapse
Affiliation(s)
- Yangyang Feng
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Yu Wang
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Shaocun Zhang
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Kabeer Haneef
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
36
|
Chromatographic assay to probe the binding energy and mechanisms of homologous proteins to surface-bound ligands. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1136:121927. [DOI: 10.1016/j.jchromb.2019.121927] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/08/2019] [Accepted: 12/03/2019] [Indexed: 01/01/2023]
|
37
|
Tian X, Deng Z, Wang S, Wang Y. Basic Research and Clinical Reports Associated with Low Serum IgG4 Concentrations. Int Arch Allergy Immunol 2019; 181:149-158. [PMID: 31805576 DOI: 10.1159/000503967] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/07/2019] [Indexed: 11/19/2022] Open
Abstract
Elevated IgG4 concentrations in serum have received a great deal of attention recently, whereas the significance of decreased IgG4 levels was frequently neglected in spite of its close relation with infectious and noninfectious inflammations. In this review, based on the structural and functional characteristics of IgG4, we bring together case reports and research related to low levels of IgG4 and try to scratch the importance of decreased IgG4 concentrations in serum. As with elevated IgG4 levels, low serum IgG4-related diseases can be involved in multiple systems such as infection in the respiratory system, stroke in the circulatory system, and glomerulonephritis in the urinary system. Both genetic and immune dysregulation can contribute to decreased IgG4 levels. In the light of animal experiments, we believe that the mystery of low IgG4 can be revealed as long as enough attention is acquired.
Collapse
Affiliation(s)
- Xinyu Tian
- Department of Nephrology, Peking University Third Hospital, Beijing, China
| | - Zhenling Deng
- Department of Nephrology, Peking University Third Hospital, Beijing, China
| | - Song Wang
- Department of Nephrology, Peking University Third Hospital, Beijing, China
| | - Yue Wang
- Department of Nephrology, Peking University Third Hospital, Beijing, China,
| |
Collapse
|
38
|
Wright DW, Elliston ELK, Hui GK, Perkins SJ. Atomistic Modeling of Scattering Curves for Human IgG1/4 Reveals New Structure-Function Insights. Biophys J 2019; 117:2101-2119. [PMID: 31708160 PMCID: PMC6895691 DOI: 10.1016/j.bpj.2019.10.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/03/2019] [Accepted: 10/18/2019] [Indexed: 11/11/2022] Open
Abstract
Small angle x-ray and neutron scattering are techniques that give solution structures for large macromolecules. The creation of physically realistic atomistic models from known high-resolution structures to determine joint x-ray and neutron scattering best-fit structures offers a, to our knowledge, new method that significantly enhances the utility of scattering. To validate this approach, we determined scattering curves for two human antibody subclasses, immunoglobulin G (IgG) 1 and IgG4, on five different x-ray and neutron instruments to show that these were reproducible, then we modeled these by Monte Carlo simulations. The two antibodies have different hinge lengths that connect their antigen-binding Fab and effector-binding Fc regions. Starting from 231,492 and 190,437 acceptable conformations for IgG1 and IgG4, respectively, joint x-ray and neutron scattering curve fits gave low goodness-of-fit R factors for 28 IgG1 and 2748 IgG4 structures that satisfied the disulphide connectivity in their hinges. These joint best-fit structures showed that the best-fit IgG1 models had a greater separation between the centers of their Fab regions than those for IgG4, in agreement with their hinge lengths of 15 and 12 residues, respectively. The resulting asymmetric IgG1 solution structures resembled its crystal structure. Both symmetric and asymmetric solution structures were determined for IgG4. Docking simulations with our best-fit IgG4 structures showed greater steric clashes with its receptor to explain its weaker FcγRI receptor binding compared to our best-fit IgG1 structures with fewer clashes and stronger receptor binding. Compared to earlier approaches for fitting molecular antibody structures by solution scattering, we conclude that this joint fit approach based on x-ray and neutron scattering data, combined with Monte Carlo simulations, significantly improved our understanding of antibody solution structures. The atomistic nature of the output extended our understanding of known functional differences in Fc receptor binding between IgG1 and IgG4.
Collapse
Affiliation(s)
- David W Wright
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Emma L K Elliston
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Gar Kay Hui
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Stephen J Perkins
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom.
| |
Collapse
|
39
|
Culver EL, van de Bovenkamp FS, Derksen NIL, Koers J, Cargill T, Barnes E, de Neef LA, Koeleman CAM, Aalberse RC, Wuhrer M, Rispens T. Unique patterns of glycosylation in immunoglobulin subclass G4-related disease and primary sclerosing cholangitis. J Gastroenterol Hepatol 2019; 34:1878-1886. [PMID: 30345709 PMCID: PMC6899843 DOI: 10.1111/jgh.14512] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/03/2018] [Accepted: 10/07/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIM Immunoglobulin subclass G4-related disease (IgG4-RD) is characterized by an abundance of IgG4 antibodies in the serum and tissue. Glycosylation status of antibodies can impact on immune effector functions and disease pathophysiology. We sought to establish glycosylation patterns in a prospective cohort of patients with IgG4-RD and the relationship with disease activity and response to treatment. METHODS We assessed IgG Fc-tail and Fab-arm glycosylation status in patients with IgG4-RD (n = 22), disease controls with primary sclerosing cholangitis (PSC) (n = 22), and healthy controls (n = 22). Serum IgG and subclasses were quantified using ELISA. Fc and Fab glycosylation were analyzed by mass spectrometry and lectin affinity chromatography, respectively. Disease activity, organ damage, and response to treatment were assessed using the IgG4 Responder Index. RESULTS Immunoglobulin G Fab sialylation was increased in IgG4-RD compared with PSC and healthy control (P = 0.01), with a preferential increase in IgG4-specific Fab sialylation, which was independent of IgG4 Fab-arm exchange. There was a reduction in IgG1-specific Fc bisection and hybrid structures in IgG4-RD (P < 0.01), which recovered upon steroid treatment and correlated with disease activity. Overall, IgG Fc galactosylation was reduced in both IgG4-RD and PSC (P < 0.01), with a preferential reduction in IgG1-specific sialylation and enhancement of IgG4-specific bisection in PSC. IgG4 fucosylation and IgG1/2/3 hybrid structures negatively correlated with complement C3 and C4 levels in IgG4-RD (P < 0.01), but not PSC. CONCLUSION We report the first study showing unique antibody glycosylation status in a prospective cohort of IgG4-RD and PSC patients, which may determine modulation of the immune system and contribute to disease pathophysiology.
Collapse
Affiliation(s)
- Emma L Culver
- Translational Gastroenterology Unit and Oxford NIHR Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
| | - Fleur S van de Bovenkamp
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
| | - Ninotska I L Derksen
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
| | - Jana Koers
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
| | - Tamsin Cargill
- Translational Gastroenterology Unit and Oxford NIHR Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
| | - Eleanor Barnes
- Translational Gastroenterology Unit and Oxford NIHR Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
| | - Louise A de Neef
- Center for Proteomics and MetabolomicsLeiden University Medical CenterLeidenThe Netherlands
| | - Carolien A M Koeleman
- Center for Proteomics and MetabolomicsLeiden University Medical CenterLeidenThe Netherlands
| | - Rob C Aalberse
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and MetabolomicsLeiden University Medical CenterLeidenThe Netherlands
| | - Theo Rispens
- Sanquin Research, Department of Immunopathology, and Landsteiner Laboratory, Academic Medical CentreUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
40
|
IgG4 antibodies from patients with asymptomatic bancroftian filariasis inhibit the binding of IgG1 and IgG2 to C1q in a Fc-Fc-dependent mechanism. Parasitol Res 2019; 118:2957-2968. [PMID: 31485865 PMCID: PMC6754495 DOI: 10.1007/s00436-019-06451-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 08/30/2019] [Indexed: 12/24/2022]
Abstract
A striking feature of lymphatic filariasis (LF) is the clinical heterogeneity among exposed individuals. While endemic normals (EN) remain free of infection despite constant exposure to the infective larvae, a small group of patients, generally microfilaria free (Mf-) develops severe pathology (CP) such as lymphedema or hydrocele. Another group of infected individuals remains asymptomatic while expressing large amounts of microfilariae (Mf+). This Mf+ group is characterized by an immune-suppressed profile with high levels of anti-inflammatory cytokines and elevated IgG4. This particular immunoglobulin is unable to activate the complement. The complement system plays a critical role in both innate and adaptive immunity. However, its importance and regulation during LF is not fully understood. Using affinity chromatography and solid-phase-enzyme-immunoassays, we investigated the ability of antibody isotypes from LF clinical groups to bind C1q, the first element of the complement’s classical pathway. The results indicate that while C1q is similarly expressed in all LF clinical groups, IgG1–2 in the plasma from Mf+ individuals presented significantly lower affinity to C1q compared to EN, Mf−, and CP. In addition, selective depletion of IgG4 significantly enhanced the affinity of IgG1–2 to C1q in Mf+ individuals. Strikingly, no effect was seen on the ability of IgG3 to bind C1q in the same conditions. More interestingly, papain-generated IgG4-Fc-portions interacted with Fc portions of IgG1–2 as revealed by far-western blot analysis. These data suggest that while being unable to bind C1q, IgG4 inhibits the first steps of the complement classical pathway by IgG1 or IgG2 via Fc-Fc interactions.
Collapse
|
41
|
Lei D, Liu J, Liu H, Cleveland TE, Marino JP, Lei M, Ren G. Single-Molecule 3D Images of "Hole-Hole" IgG1 Homodimers by Individual-Particle Electron Tomography. Sci Rep 2019; 9:8864. [PMID: 31221961 PMCID: PMC6586654 DOI: 10.1038/s41598-019-44978-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/30/2019] [Indexed: 12/20/2022] Open
Abstract
The engineering of immunoglobulin-G molecules (IgGs) is of wide interest for improving therapeutics, for example by modulating the activity or multiplexing the specificity of IgGs to recognize more than one antigen. Optimization of engineered IgG requires knowledge of three-dimensional (3D) structure of synthetic IgG. However, due to flexible nature of the molecules, their structural characterization is challenging. Here, we use our reported individual-particle electron tomography (IPET) method with optimized negative-staining (OpNS) for direct 3D reconstruction of individual IgG hole-hole homodimer molecules. The hole-hole homodimer is an undesired variant generated during the production of a bispecific antibody using the knob-into-hole heterodimer technology. A total of 64 IPET 3D density maps at ~15 Å resolutions were reconstructed from 64 individual molecules, revealing 64 unique conformations. In addition to the known Y-shaped conformation, we also observed an unusual X-shaped conformation. The 3D structure of the X-shaped conformation contributes to our understanding of the structural details of the interaction between two heavy chains in the Fc domain. The IPET approach, as an orthogonal technique to characterize the 3D structure of therapeutic antibodies, provides insight into the 3D structural variety and dynamics of heterogeneous IgG molecules.
Collapse
Affiliation(s)
- Dongsheng Lei
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Jianfang Liu
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Hongbin Liu
- Protein Analytical Chemistry, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Thomas E Cleveland
- Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology and the University of Maryland, Rockville, MD, 20850, USA
| | - John P Marino
- Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology and the University of Maryland, Rockville, MD, 20850, USA
| | - Ming Lei
- Protein Analytical Chemistry, Genentech Inc., South San Francisco, CA, 94080, USA.
| | - Gang Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
42
|
Xu CA, Feng AZ, Ramineni CK, Wallace MR, Culyba EK, Guay KP, Mehta K, Mabry R, Farrand S, Xu J, Feng J. L 445P mutation on heavy chain stabilizes IgG 4 under acidic conditions. MAbs 2019; 11:1289-1299. [PMID: 31199179 DOI: 10.1080/19420862.2019.1631116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
IgG4, a common type of therapeutic antibody, is less stable during manufacturing processes compared with IgG1. Aggregation and fragmentation are the two main challenges. Here, we report instability of the heavy chain (HC) C-terminal region under acidic conditions, which leads to cleavage and aggregation. Leu445, at the C-terminal region of the HC in IgG4, plays a critical role in its acid-induced fragmentation and subsequent aggregation. We found that mutating HC C-terminal Leu445 to Pro (the corresponding residue in IgG1) in IgG4_CDR-X significantly reduces fragmentation and aggregation, while mutating Pro445 to Leu in IgG1_CDR-X promotes fragmentation and aggregation. HC C-terminal Gly446 cleavage was observed in low pH citrate buffer and resulted in further fragmentation and aggregation, whereas, glycine buffer can completely inhibit the cleavage and aggregation. It is proposed that cleavages occur through acid-induced hydrolysis under acidic conditions and glycine stabilizes IgG4 via two main mechanisms: 1) product feedback inhibition of the hydrolysis reaction, and 2) stabilization of protein conformation by direct interaction with the peptide backbone and charged side chains. Experiments using IgG4 molecules IgG4_CDR-Y and IgG4_CDR-Z with the same CH domains as IgG4_CDR-X, but different complementarity-determining regions (CDRs), indicate that the stability of the HC C-terminal region is also closely related to the sequence of the CDRs. The stability of IgG4_CDR-X is significantly improved when binding to its target. Both observations suggest that there are potential interactions between Fab and CH2-CH3 domains, which could be the key factor affecting the stability of IgG antibodies.
Collapse
Affiliation(s)
| | - Andrew Z Feng
- Chemistry Department, University of Massachusetts Lowell , Lowell , MA , USA
| | - Charan K Ramineni
- Chemistry Department, University of Massachusetts Lowell , Lowell , MA , USA
| | | | | | | | | | - Robert Mabry
- Jounce Therapeutics , Cambridge , MA , USA.,Global Biologics , Cambridge , MA , USA
| | | | - Jin Xu
- Chemistry Department, University of Massachusetts Lowell , Lowell , MA , USA
| | - Jianwen Feng
- Jounce Therapeutics , Cambridge , MA , USA.,MassBiologics, University of Massachusetts Medical School , Mattapan , MA , USA
| |
Collapse
|
43
|
Casaletto JB, Geddie ML, Abu-Yousif AO, Masson K, Fulgham A, Boudot A, Maiwald T, Kearns JD, Kohli N, Su S, Razlog M, Raue A, Kalra A, Håkansson M, Logan DT, Welin M, Chattopadhyay S, Harms BD, Nielsen UB, Schoeberl B, Lugovskoy AA, MacBeath G. MM-131, a bispecific anti-Met/EpCAM mAb, inhibits HGF-dependent and HGF-independent Met signaling through concurrent binding to EpCAM. Proc Natl Acad Sci U S A 2019; 116:7533-7542. [PMID: 30898885 PMCID: PMC6462049 DOI: 10.1073/pnas.1819085116] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Activation of the Met receptor tyrosine kinase, either by its ligand, hepatocyte growth factor (HGF), or via ligand-independent mechanisms, such as MET amplification or receptor overexpression, has been implicated in driving tumor proliferation, metastasis, and resistance to therapy. Clinical development of Met-targeted antibodies has been challenging, however, as bivalent antibodies exhibit agonistic properties, whereas monovalent antibodies lack potency and the capacity to down-regulate Met. Through computational modeling, we found that the potency of a monovalent antibody targeting Met could be dramatically improved by introducing a second binding site that recognizes an unrelated, highly expressed antigen on the tumor cell surface. Guided by this prediction, we engineered MM-131, a bispecific antibody that is monovalent for both Met and epithelial cell adhesion molecule (EpCAM). MM-131 is a purely antagonistic antibody that blocks ligand-dependent and ligand-independent Met signaling by inhibiting HGF binding to Met and inducing receptor down-regulation. Together, these mechanisms lead to inhibition of proliferation in Met-driven cancer cells, inhibition of HGF-mediated cancer cell migration, and inhibition of tumor growth in HGF-dependent and -independent mouse xenograft models. Consistent with its design, MM-131 is more potent in EpCAM-high cells than in EpCAM-low cells, and its potency decreases when EpCAM levels are reduced by RNAi. Evaluation of Met, EpCAM, and HGF levels in human tumor samples reveals that EpCAM is expressed at high levels in a wide range of Met-positive tumor types, suggesting a broad opportunity for clinical development of MM-131.
Collapse
Affiliation(s)
| | - Melissa L Geddie
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Adnan O Abu-Yousif
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Kristina Masson
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Aaron Fulgham
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Antoine Boudot
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Tim Maiwald
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Jeffrey D Kearns
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Neeraj Kohli
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Stephen Su
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Maja Razlog
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Andreas Raue
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139;
| | - Ashish Kalra
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Maria Håkansson
- SARomics Biostructures AB, Medicon Village, SE-223 81 Lund, Sweden
| | - Derek T Logan
- SARomics Biostructures AB, Medicon Village, SE-223 81 Lund, Sweden
| | - Martin Welin
- SARomics Biostructures AB, Medicon Village, SE-223 81 Lund, Sweden
| | | | - Brian D Harms
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Ulrik B Nielsen
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Birgit Schoeberl
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Alexey A Lugovskoy
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Gavin MacBeath
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139;
| |
Collapse
|
44
|
Blech M, Hörer S, Kuhn AB, Kube S, Göddeke H, Kiefer H, Zang Y, Alber Y, Kast SM, Westermann M, Tully MD, Schäfer LV, Garidel P. Structure of a Therapeutic Full-Length Anti-NPRA IgG4 Antibody: Dissecting Conformational Diversity. Biophys J 2019; 116:1637-1649. [PMID: 31023536 DOI: 10.1016/j.bpj.2019.03.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 01/02/2023] Open
Abstract
We report the x-ray crystal structure of intact, full-length human immunoglobulin (IgG4) at 1.8 Å resolution. The data for IgG4 (S228P), an antibody targeting the natriuretic peptide receptor A, show a previously unrecognized type of Fab-Fc orientation with a distorted λ-shape in which one Fab-arm is oriented toward the Fc portion. Detailed structural analysis by x-ray crystallography and molecular simulations suggest that this is one of several conformations coexisting in a dynamic equilibrium state. These results were confirmed by small angle x-ray scattering in solution. Furthermore, electron microscopy supported these findings by preserving molecule classes of different conformations. This study fosters our understanding of IgG4 in particular and our appreciation of antibody flexibility in general. Moreover, we give insights into potential biological implications, specifically for the interaction of human anti-natriuretic peptide receptor A IgG4 with the neonatal Fc receptor, Fcγ receptors, and complement-activating C1q by considering conformational flexibility.
Collapse
Affiliation(s)
- Michaela Blech
- Innovation Unit, Pharmaceutical Development Biologics, Biberach (Riss), Germany.
| | - Stefan Hörer
- Department Lead Identification and Optimization Support, Structural Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | | | - Sebastian Kube
- Innovation Unit, Pharmaceutical Development Biologics, Biberach (Riss), Germany
| | - Hendrik Göddeke
- Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany
| | - Hans Kiefer
- University of Applied Sciences Biberach, Biberach (Riss), Germany
| | - Yuguo Zang
- University of Applied Sciences Biberach, Biberach (Riss), Germany
| | - Yannic Alber
- Physikalische Chemie III, Technische Universität Dortmund, Dortmund, Germany
| | - Stefan M Kast
- Physikalische Chemie III, Technische Universität Dortmund, Dortmund, Germany
| | - Martin Westermann
- Elektronenmikroskopisches Zentrum, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Mark D Tully
- European Synchrotron Radiation Facility, Grenoble, France
| | - Lars V Schäfer
- Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany
| | - Patrick Garidel
- Innovation Unit, Pharmaceutical Development Biologics, Biberach (Riss), Germany.
| |
Collapse
|
45
|
Tolbert WD, Subedi GP, Gohain N, Lewis GK, Patel KR, Barb AW, Pazgier M. From Rhesus macaque to human: structural evolutionary pathways for immunoglobulin G subclasses. MAbs 2019; 11:709-724. [PMID: 30939981 PMCID: PMC6601566 DOI: 10.1080/19420862.2019.1589852] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/15/2019] [Accepted: 02/22/2019] [Indexed: 10/27/2022] Open
Abstract
The Old World monkey, Rhesus macaque (Macaca mulatta, Mm), is frequently used as a primate model organism in the study of human disease and to test new vaccines/antibody treatments despite diverging before chimpanzees and orangutans. Mm and humans share 93% genome identity with substantial differences in the genes of the adaptive immune system that lead to different functional IgG subclass characteristics, Fcγ receptors expressed on innate immune cells, and biological interactions. These differences put limitations on Mm use as a primary animal model in the study of human disease and to test new vaccines/antibody treatments. Here, we comprehensively analyzed molecular properties of the Fc domain of the four IgG subclasses of Rhesus macaque to describe potential mechanisms for their interactions with effector cell Fc receptors. Our studies revealed less diversity in the overall structure among the Mm IgG Fc, with MmIgG1 Fc being the most structurally like human IgG3, although its CH2 loops and N297 glycan mobility are comparable to human IgG1. Furthermore, the Fcs of Mm IgG3 and 4 lack the structural properties typical for their human orthologues that determine IgG3's reduced interaction with the neonatal receptor and IgG4's ability for Fab-arm exchange and its weaker Fcγ receptor interactions. Taken together, our data indicate that MmIgG1-4 are less structurally divergent than the human IgGs, with only MmIgG1 matching the molecular properties of human IgG1 and 3, the most active IgGs in terms of Fcγ receptor binding and Fc-mediated functions. PDB accession numbers for deposited structures are 6D4E, 6D4I, 6D4M, and 6D4N for MmIgG1 Fc, MmIgG2 Fc, MmIgG3 Fc, and MmIgG4 Fc, respectively.
Collapse
Affiliation(s)
- William David Tolbert
- Division of Vaccine Research, Institute of Human Virology of University of Maryland School of Medicine, Baltimore, MD, USA
- Infectious Disease Division, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Ganesh Prasad Subedi
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology of Iowa State University, Ames, IA, USA
| | - Neelakshi Gohain
- Division of Vaccine Research, Institute of Human Virology of University of Maryland School of Medicine, Baltimore, MD, USA
| | - George Kenneth Lewis
- Division of Vaccine Research, Institute of Human Virology of University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kashyap Rajesh Patel
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology of Iowa State University, Ames, IA, USA
| | - Adam Wesley Barb
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology of Iowa State University, Ames, IA, USA
| | - Marzena Pazgier
- Division of Vaccine Research, Institute of Human Virology of University of Maryland School of Medicine, Baltimore, MD, USA
- Infectious Disease Division, Uniformed Services University of the Health Sciences, Bethesda, MD
| |
Collapse
|
46
|
Cao X, Flagg SC, Li X, Chennamsetty N, Balakrishnan G, Das TK. Quadrupole Dalton-Based Controlled Proteolysis Method for Characterization of Higher Order Protein Structure. Anal Chem 2019; 91:5339-5345. [DOI: 10.1021/acs.analchem.9b00306] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Xiang Cao
- Biologics Methods and Analytical Development, Bristol-Myers Squibb, Hopewell, New Jersey 08534, United States
| | - Shannon C. Flagg
- Biologics Methods and Analytical Development, Bristol-Myers Squibb, Hopewell, New Jersey 08534, United States
| | - Xue Li
- Biologics Methods and Analytical Development, Bristol-Myers Squibb, Hopewell, New Jersey 08534, United States
| | - Naresh Chennamsetty
- Biologics Methods and Analytical Development, Bristol-Myers Squibb, Hopewell, New Jersey 08534, United States
| | - Gurusamy Balakrishnan
- Biologics Methods and Analytical Development, Bristol-Myers Squibb, Hopewell, New Jersey 08534, United States
| | - Tapan K. Das
- Biologics Methods and Analytical Development, Bristol-Myers Squibb, Hopewell, New Jersey 08534, United States
| |
Collapse
|
47
|
Kang H, Tolbert TJ, Schöneich C. Photoinduced Tyrosine Side Chain Fragmentation in IgG4-Fc: Mechanisms and Solvent Isotope Effects. Mol Pharm 2018; 16:258-272. [DOI: 10.1021/acs.molpharmaceut.8b00979] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Huan Kang
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, Kansas 66047, United States
| | - Thomas J. Tolbert
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, Kansas 66047, United States
| | - Christian Schöneich
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, Kansas 66047, United States
| |
Collapse
|
48
|
Pu X, Wu L, Su D, Mao W, Fang B. Immunotherapy for non-small cell lung cancers: biomarkers for predicting responses and strategies to overcome resistance. BMC Cancer 2018; 18:1082. [PMID: 30409126 PMCID: PMC6225701 DOI: 10.1186/s12885-018-4990-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/24/2018] [Indexed: 12/26/2022] Open
Abstract
Recent breakthroughs in targeted therapy and immunotherapy have revolutionized the treatment of lung cancer, the leading cause of cancer-related deaths in the United States and worldwide. Here we provide an overview of recent progress in immune checkpoint blockade therapy for treatment of non-small cell lung cancer (NSCLC), and discuss biomarkers associated with the treatment responses, mechanisms underlying resistance and strategies to overcome resistance. The success of immune checkpoint blockade therapies is driven by immunogenicity of tumor cells, which is associated with mutation burden and neoantigen burden in cancers. Lymphocyte infiltration in cancer tissues and interferon-γ-induced PD-L1 expression in tumor microenvironments may serve as surrogate biomarkers for adaptive immune resistance and likelihood of responses to immune checkpoint blockade therapy. In contrast, weak immunogenicity of, and/or impaired antigen presentation in, tumor cells are primary causes of resistance to these therapies. Thus, approaches that increase immunogenicity of cancer cells and/or enhance immune cell recruitment to cancer sites will likely overcome resistance to immunotherapy.
Collapse
Affiliation(s)
- Xingxiang Pu
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital/the affiliated Cancer Hospital of Xiangya school of Medicine, Central South University, 283 Tongzipo Road, Yuelu District, Changsha, 410013 Hunan China
| | - Lin Wu
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital/the affiliated Cancer Hospital of Xiangya school of Medicine, Central South University, 283 Tongzipo Road, Yuelu District, Changsha, 410013 Hunan China
| | - Dan Su
- Department of Pathology, Zhejiang Cancer Hospital, 38 Guanji Road, Banshan Bridge, Hangzhou, 310022 Zejiang China
| | - Weimin Mao
- Department of Thoracic Surgery, Zhejiang Cancer Hospital, 38 Guanji Road, Banshan Bridge, Hangzhou, 310022 Zejiang China
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| |
Collapse
|
49
|
Liu M, Hao M. Unique properties of IgG4 antibody and its clinical application in autoimmune pancreatitis. Scand J Gastroenterol 2018; 53:1121-1131. [PMID: 30175675 DOI: 10.1080/00365521.2018.1476915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Autoimmune pancreatitis (AIP) is defined as a unique form of chronic pancreatitis characterized by clinical presentation with obstructive jaundice, a dense lymphoplasmacytic infiltrate and fibrosis histologically, and a dramatic response to steroids therapeutically. The possible role of IgG4 in driving the pathology of AIP is a controversial subject that has not been addressed satisfactorily. Objective: The purpose of this review is to discuss the unique biology of IgG4 that are important for its role and the clinical applications for serologic detection. METHODS Review of current literature about IgG4 antibody in the clinical application in AIP. RESULTS High serum levels of IgG4 are an important biomarker and broadly used for diagnosis, differentiation from diseases especially pancreatic cancer, and as a parameter to indicate disease activity, extra-pancreatic lesions, and treatment monitoring. However, some controversial studies show it has a limited specificity and sensitivity in these conditions. Conclusion: Although increasing studies have promoted our understanding of the structure and function of IgG4, there is still dilemma between the beneficial and the adverse aspect of IgG4 in the pathogenesis of AIP.
Collapse
Affiliation(s)
- Min Liu
- a Department of Clinical Laboratory , Jinan Dermatosis Prevention and Control Hospital , Jinan , People's Republic of China
| | - Mingju Hao
- b Department of Clinical Laboratory , Qianfo Mountain Hospital of Shandong University , Jinan , People's Republic of China
| |
Collapse
|
50
|
Karahan GE, Claas FHJ, Heidt S. Technical challenges and clinical relevance of single antigen bead C1q/C3d testing and IgG subclass analysis of human leukocyte antigen antibodies. Transpl Int 2018; 31:1189-1197. [DOI: 10.1111/tri.13327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/21/2018] [Accepted: 08/03/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Gonca E. Karahan
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden the Netherlands
| | - Frans H. J. Claas
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden the Netherlands
| | - Sebastiaan Heidt
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden the Netherlands
| |
Collapse
|