1
|
Dieppois C, Adjemout M, Cretin J, Gallardo F, Torres M, Picard C, Sawadogo SA, Rihet P, Paul P. An Expression Quantitative Trait Locus of Fc Gamma Receptor Genes Is Associated With Antimalarial IgG Responses and Infection Levels in Burkinabe Families. J Infect Dis 2025; 231:1008-1019. [PMID: 39450553 DOI: 10.1093/infdis/jiae528] [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: 08/21/2024] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND The interaction between antibodies and Fcγ receptors (FcγRs) plays a critical role in regulating immune responses to Plasmodium falciparum. Polymorphisms in genes encoding FcγRs influence the host's capacity to control parasite infection. This study investigates whether noncoding variants influencing FcγR expression are associated with antimalarial immunization and infection traits. METHODS We utilized eQTL databases and functional annotations to identify noncoding variants, specifically rs1771575, rs2099684, and rs6700241, within the FCGR gene cluster. In addition, we examined the coding variants rs1801274 (p.His167Arg) and rs1050501 (p.Ile231Thr), which affect the affinity of FcγRIIa and FcγRIIb for IgG. These variants were genotyped in 163 individuals from Burkinabe families. Family-based linear mixed regression and Quantitative Transmission Disequilibrium Tests (QTDT) analyses were performed to assess associations with IgG levels and malaria infection, accounting for relevant covariates. RESULTS Linear mixed models identified rs1771575 as associated with total IgG levels, while both rs1771575 and rs1801274 were linked to IgG2, and rs1050501 to IgG1 levels. A haplotype combining rs2099684 and rs6700241 was positively associated with IgG1. The rs1771575-CC and rs1050501-TT genotypes correlated with higher infection levels in children. QTDT models confirmed the association of rs1771575 with IgG2 and infection in children. CONCLUSIONS Our findings suggest that the intergenic variant rs1771575 serves as an independent marker for IgG levels and blood infection in children. This highlights the interplay between regulatory variants and coding mutations in FCGR, which may influence immune function and antibody production. These results underscore the potential for personalized strategies to monitor humoral responses in malaria-endemic regions.
Collapse
Affiliation(s)
- Christelle Dieppois
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Mathieu Adjemout
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Jules Cretin
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Frederic Gallardo
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Magali Torres
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Christophe Picard
- Anthropologie bio-culturelle, droit, éthique et santé, Établissement Français du Sang, Centre National de la Recherche Scientifique, Aix Marseille University, Marseille, France
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
| | - Serge Aimé Sawadogo
- Unité de Formation en Sciences de la Santé, Université Joseph Ki-Zerbo, Ouagadougou, Burkina Faso
- Centre Pour la Pecherche et l'Pnnovation en Pmmunologie Pédicale de Puagadougou-Nelson Mandela, Ouagadougou, Burkina Faso
| | - Pascal Rihet
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Pascale Paul
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| |
Collapse
|
2
|
Simpson AP, Oldham RJ, Cox KL, Taylor MC, James S, White AL, Bogdanov Y, Glennie MJ, Frendeus B, Cragg MS, Roghanian A. FcγRIIB (CD32B) antibodies enhance immune responses through activating FcγRs. Clin Exp Immunol 2025; 219:uxaf015. [PMID: 40089806 DOI: 10.1093/cei/uxaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/20/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025] Open
Abstract
Fc receptors (FcR) play a key role in coordinating responses from both the innate and adaptive immune system. The inhibitory Fc gamma receptor (FcγRIIB/CD32B; referred to as FcγRII/CD32 in mice) restrains the immune response, specifically through regulating immunoglobulin G (IgG) effector functions. FcγRII-deficient mice demonstrate elevated incidence and severity of autoimmunity and increased responses to immunization and infections. To explore the potential of FcγRIIB as a target for augmenting vaccines, we tested the ability of monoclonal antibodies (mAb) against mouse FcγRII and human FcγRIIB to enhance humoral responses in preclinical models. We used wild-type (WT), FcγR-deficient, and human FcγRIIB transgenic (Tg) mice with either a functional intracellular domain (hFcγRIIB Tg) or lacking immunoreceptor tyrosine-based inhibitory motif (ITIM) signalling capacity (NoTIM). Targeting mouse FcγRII and human FcγRIIB with antibodies significantly augmented humoral immune responses against experimental antigens and enhanced tumour clearance in vivo. Surprisingly, mAbs without a functional Fc (N297Q; referred to as Fc-null) lacked efficacy. Similarly, blocking FcγRII in mice lacking activating FcγRs failed to enhance immune responses. Conversely, blocking both signalling-competent and signalling-defective (NoTIM) FcγRIIB in Tg mice with a WT, but not Fc-null, FcγRIIB mAb equally enhanced immunity. These data indicate the redundancy of inhibitory signalling in potentiating immune responses in vivo. Collectively, our data suggest that mAb-targeting of FcγRIIB stabilizes mAb Fc and enhances immune responses via Fc-mediated crosslinking of activating FcγRs, irrespective of the inhibitory function of FcγRIIB. These findings support a strategy to boost immune responses in immunization protocols.
Collapse
Affiliation(s)
- Alexander P Simpson
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Robert J Oldham
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Kerry L Cox
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Martin C Taylor
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Sonya James
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Ann L White
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Yury Bogdanov
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Martin J Glennie
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Björn Frendeus
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
- BioInvent International AB, Sölvegatan 41, Lund, Sweden
| | - Mark S Cragg
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
- Institute for Life Sciences, University of Southampton, Highfield, Southampton, UK
| | - Ali Roghanian
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
- Institute for Life Sciences, University of Southampton, Highfield, Southampton, UK
| |
Collapse
|
3
|
Bond A, Morrissey MA. Biochemical and biophysical mechanisms macrophages use to tune phagocytic appetite. J Cell Sci 2025; 138:JCS263513. [PMID: 39749603 PMCID: PMC11828473 DOI: 10.1242/jcs.263513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Macrophages phagocytose, or eat, pathogens, dead cells and cancer cells. To activate phagocytosis, macrophages recognize 'eat me' signals like IgG and phosphatidylserine on the target cell surface. Macrophages must carefully adjust their phagocytic appetite to ignore non-specific or transient eat me signal exposure on healthy cells while still rapidly recognizing pathogens and debris. Depending on the context, macrophages can increase their appetite for phagocytosis, to prioritize an effective immune response, or decrease their appetite, to avoid damage to healthy tissue during homeostasis. In this Review, we discuss the biochemical and biophysical mechanisms that macrophages employ to increase or decrease their sensitivity or capacity for phagocytosis. We discuss evidence that macrophages tune their sensitivity via several mechanisms, including altering the balance of activating and inhibitory receptor expression, altering the availability of activating receptors, as well as influencing their clustering and mobility, and modulating inhibitory receptor location. We also highlight how membrane availability limits the capacity of macrophages for phagocytosis and discuss potential mechanisms to promote membrane recycling and increase phagocytic capacity. Overall, this Review highlights recent work detailing the molecular toolkit that macrophages use to alter their appetite.
Collapse
Affiliation(s)
- Annalise Bond
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| | - Meghan A. Morrissey
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
4
|
Frampton S, Smith R, Ferson L, Gibson J, Hollox EJ, Cragg MS, Strefford JC. Fc gamma receptors: Their evolution, genomic architecture, genetic variation, and impact on human disease. Immunol Rev 2024; 328:65-97. [PMID: 39345014 PMCID: PMC11659932 DOI: 10.1111/imr.13401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Fc gamma receptors (FcγRs) are a family of receptors that bind IgG antibodies and interface at the junction of humoral and innate immunity. Precise regulation of receptor expression provides the necessary balance to achieve healthy immune homeostasis by establishing an appropriate immune threshold to limit autoimmunity but respond effectively to infection. The underlying genetics of the FCGR gene family are central to achieving this immune threshold by regulating affinity for IgG, signaling efficacy, and receptor expression. The FCGR gene locus was duplicated during evolution, retaining very high homology and resulting in a genomic region that is technically difficult to study. Here, we review the recent evolution of the gene family in mammals, its complexity and variation through copy number variation and single-nucleotide polymorphism, and impact of these on disease incidence, resolution, and therapeutic antibody efficacy. We also discuss the progress and limitations of current approaches to study the region and emphasize how new genomics technologies will likely resolve much of the current confusion in the field. This will lead to definitive conclusions on the impact of genetic variation within the FCGR gene locus on immune function and disease.
Collapse
Affiliation(s)
- Sarah Frampton
- Cancer Genomics Group, Faculty of Medicine, School of Cancer SciencesUniversity of SouthamptonSouthamptonUK
| | - Rosanna Smith
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer ImmunologyUniversity of SouthamptonSouthamptonUK
| | - Lili Ferson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer SciencesUniversity of SouthamptonSouthamptonUK
| | - Jane Gibson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer SciencesUniversity of SouthamptonSouthamptonUK
| | - Edward J. Hollox
- Department of Genetics, Genomics and Cancer SciencesCollege of Life Sciences, University of LeicesterLeicesterUK
| | - Mark S. Cragg
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer ImmunologyUniversity of SouthamptonSouthamptonUK
| | - Jonathan C. Strefford
- Cancer Genomics Group, Faculty of Medicine, School of Cancer SciencesUniversity of SouthamptonSouthamptonUK
| |
Collapse
|
5
|
Lim SH, Beers SA, Al-Shamkhani A, Cragg MS. Agonist Antibodies for Cancer Immunotherapy: History, Hopes, and Challenges. Clin Cancer Res 2024; 30:1712-1723. [PMID: 38153346 PMCID: PMC7615925 DOI: 10.1158/1078-0432.ccr-23-1014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/31/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
Immunotherapy is among the most promising new treatment modalities to arise over the last two decades; antibody drugs are delivering immunotherapy to millions of patients with many different types of cancer. Initial success with antibody therapeutics came in the form of direct targeting or cytotoxic antibodies, such as rituximab and trastuzumab, which bind directly to tumor cells to elicit their destruction. These were followed by immunomodulatory antibodies that elicit antitumor responses by either stimulating immune cells or relieving tumor-mediated suppression. By far the most successful approach in the clinic to date has been relieving immune suppression, with immune checkpoint blockade now a standard approach in the treatment of many cancer types. Despite equivalent and sometimes even more impressive effects in preclinical models, agonist antibodies designed to stimulate the immune system have lagged behind in their clinical translation. In this review, we document the main receptors that have been targeted by agonist antibodies, consider the various approaches that have been evaluated to date, detail what we have learned, and consider how their anticancer potential can be unlocked.
Collapse
Affiliation(s)
- Sean H. Lim
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, SO16 6YD, UK
| | - Stephen A. Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, SO16 6YD, UK
| | - Aymen Al-Shamkhani
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, SO16 6YD, UK
| | - Mark S. Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
6
|
Knorr DA, Blanchard L, Leidner RS, Jensen SM, Meng R, Jones A, Ballesteros-Merino C, Bell RB, Baez M, Marino A, Sprott D, Bifulco CB, Piening B, Dahan R, Osorio JC, Fox BA, Ravetch JV. FcγRIIB Is an Immune Checkpoint Limiting the Activity of Treg-Targeting Antibodies in the Tumor Microenvironment. Cancer Immunol Res 2024; 12:322-333. [PMID: 38147316 PMCID: PMC10911703 DOI: 10.1158/2326-6066.cir-23-0389] [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: 05/21/2023] [Revised: 10/10/2023] [Accepted: 12/21/2023] [Indexed: 12/27/2023]
Abstract
Preclinical murine data indicate that fragment crystallizable (Fc)-dependent depletion of intratumoral regulatory T cells (Treg) is a major mechanism of action of anti-CTLA-4. However, the two main antibodies administered to patients (ipilimumab and tremelimumab) do not recapitulate these effects. Here, we investigate the underlying mechanisms responsible for the limited Treg depletion observed with these therapies. Using an immunocompetent murine model humanized for CTLA-4 and Fcγ receptors (FcγR), we show that ipilimumab and tremelimumab exhibit limited Treg depletion in tumors. Immune profiling of the tumor microenvironment (TME) in both humanized mice and humans revealed high expression of the inhibitory Fc receptor, FcγRIIB, which limits antibody-dependent cellular cytotoxicity/phagocytosis. Blocking FcγRIIB in humanized mice rescued the Treg-depleting capacity and antitumor activity of ipilimumab. Furthermore, Fc engineering of antibodies targeting Treg-associated targets (CTLA-4 or CCR8) to minimize FcγRIIB binding significantly enhanced Treg depletion, resulting in increased antitumor activity across various tumor models. Our results define the inhibitory FcγRIIB as an immune checkpoint limiting antibody-mediated Treg depletion in the TME, and demonstrate Fc engineering as an effective strategy to overcome this limitation and improve the efficacy of Treg-targeting antibodies.
Collapse
Affiliation(s)
- David A. Knorr
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lucas Blanchard
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, New York
| | - Rom S. Leidner
- Earle A. Chiles Research Institute (a division of Providence Cancer Institute), Portland, Oregon
| | - Shawn M. Jensen
- Earle A. Chiles Research Institute (a division of Providence Cancer Institute), Portland, Oregon
| | - Ryan Meng
- Earle A. Chiles Research Institute (a division of Providence Cancer Institute), Portland, Oregon
| | - Andrew Jones
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, New York
| | | | - Richard B. Bell
- Earle A. Chiles Research Institute (a division of Providence Cancer Institute), Portland, Oregon
| | - Maria Baez
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, New York
| | - Alessandra Marino
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, New York
| | - David Sprott
- Earle A. Chiles Research Institute (a division of Providence Cancer Institute), Portland, Oregon
| | - Carlo B. Bifulco
- Earle A. Chiles Research Institute (a division of Providence Cancer Institute), Portland, Oregon
| | - Brian Piening
- Earle A. Chiles Research Institute (a division of Providence Cancer Institute), Portland, Oregon
| | - Rony Dahan
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Juan C. Osorio
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bernard A. Fox
- Earle A. Chiles Research Institute (a division of Providence Cancer Institute), Portland, Oregon
| | - Jeffrey V. Ravetch
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, New York
| |
Collapse
|
7
|
Rakké YS, Buschow SI, IJzermans JNM, Sprengers D. Engaging stimulatory immune checkpoint interactions in the tumour immune microenvironment of primary liver cancers - how to push the gas after having released the brake. Front Immunol 2024; 15:1357333. [PMID: 38440738 PMCID: PMC10910082 DOI: 10.3389/fimmu.2024.1357333] [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: 12/17/2023] [Accepted: 01/31/2024] [Indexed: 03/06/2024] Open
Abstract
Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA) are the first and second most common primary liver cancer (PLC). For decades, systemic therapies consisting of tyrosine kinase inhibitors (TKIs) or chemotherapy have formed the cornerstone of treating advanced-stage HCC and CCA, respectively. More recently, immunotherapy using immune checkpoint inhibition (ICI) has shown anti-tumour reactivity in some patients. The combination regimen of anti-PD-L1 and anti-VEGF antibodies has been approved as new first-line treatment of advanced-stage HCC. Furthermore, gemcibatine plus cisplatin (GEMCIS) with an anti-PD-L1 antibody is awaiting global approval for the treatment of advanced-stage CCA. As effective anti-tumour reactivity using ICI is achieved in a minor subset of both HCC and CCA patients only, alternative immune strategies to sensitise the tumour microenvironment of PLC are waited for. Here we discuss immune checkpoint stimulation (ICS) as additional tool to enhance anti-tumour reactivity. Up-to-date information on the clinical application of ICS in onco-immunology is provided. This review provides a rationale of the application of next-generation ICS either alone or in combination regimen to potentially enhance anti-tumour reactivity in PLC patients.
Collapse
Affiliation(s)
- Yannick S. Rakké
- Department of Surgery, Erasmus MC-Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Sonja I. Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC-Cancer Institute-University Medical Center, Rotterdam, Netherlands
| | - Jan N. M. IJzermans
- Department of Surgery, Erasmus MC-Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-Cancer Institute-University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
8
|
Galvez-Cancino F, Simpson AP, Costoya C, Matos I, Qian D, Peggs KS, Litchfield K, Quezada SA. Fcγ receptors and immunomodulatory antibodies in cancer. Nat Rev Cancer 2024; 24:51-71. [PMID: 38062252 DOI: 10.1038/s41568-023-00637-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 12/24/2023]
Abstract
The discovery of both cytotoxic T lymphocyte-associated antigen 4 (CTLA4) and programmed cell death protein 1 (PD1) as negative regulators of antitumour immunity led to the development of numerous immunomodulatory antibodies as cancer treatments. Preclinical studies have demonstrated that the efficacy of immunoglobulin G (IgG)-based therapies depends not only on their ability to block or engage their targets but also on the antibody's constant region (Fc) and its interactions with Fcγ receptors (FcγRs). Fc-FcγR interactions are essential for the activity of tumour-targeting antibodies, such as rituximab, trastuzumab and cetuximab, where the killing of tumour cells occurs at least in part due to these mechanisms. However, our understanding of these interactions in the context of immunomodulatory antibodies designed to boost antitumour immunity remains less explored. In this Review, we discuss our current understanding of the contribution of FcγRs to the in vivo activity of immunomodulatory antibodies and the challenges of translating results from preclinical models into the clinic. In addition, we review the impact of genetic variability of human FcγRs on the activity of therapeutic antibodies and how antibody engineering is being utilized to develop the next generation of cancer immunotherapies.
Collapse
Affiliation(s)
- Felipe Galvez-Cancino
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Alexander P Simpson
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Cristobal Costoya
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Ignacio Matos
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Danwen Qian
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, UK
| | - Karl S Peggs
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Kevin Litchfield
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, UK
| | - Sergio A Quezada
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| |
Collapse
|
9
|
Bauer-Smith H, Sudol ASL, Beers SA, Crispin M. Serum immunoglobulin and the threshold of Fc receptor-mediated immune activation. Biochim Biophys Acta Gen Subj 2023; 1867:130448. [PMID: 37652365 PMCID: PMC11032748 DOI: 10.1016/j.bbagen.2023.130448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Antibodies can mediate immune recruitment or clearance of immune complexes through the interaction of their Fc domain with cellular Fc receptors. Clustering of antibodies is a key step in generating sufficient avidity for efficacious receptor recognition. However, Fc receptors may be saturated with prevailing, endogenous serum immunoglobulin and this raises the threshold by which cellular receptors can be productively engaged. Here, we review the factors controlling serum IgG levels in both healthy and disease states, and discuss how the presence of endogenous IgG is encoded into the functional activation thresholds for low- and high-affinity Fc receptors. We discuss the circumstances where antibody engineering can help overcome these physiological limitations of therapeutic antibodies. Finally, we discuss how the pharmacological control of Fc receptor saturation by endogenous IgG is emerging as a feasible mechanism for the enhancement of antibody therapeutics.
Collapse
Affiliation(s)
- Hannah Bauer-Smith
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK; Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Abigail S L Sudol
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Stephen A Beers
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK.
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
10
|
Cretin J, Adjemout M, Dieppois C, Gallardo F, Torres M, Merard Z, Sawadogo SA, Picard C, Rihet P, Paul P. A Non-Coding Fc Gamma Receptor Cis-Regulatory Variant within the 1q23 Gene Cluster Is Associated with Plasmodium falciparum Infection in Children Residing in Burkina Faso. Int J Mol Sci 2023; 24:15711. [PMID: 37958695 PMCID: PMC10650193 DOI: 10.3390/ijms242115711] [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: 08/07/2023] [Revised: 08/27/2023] [Accepted: 08/31/2023] [Indexed: 11/15/2023] Open
Abstract
Antibodies play a crucial role in activating protective immunity against malaria by interacting with Fc-gamma receptors (FcγRs). Genetic variations in genes encoding FcγRs can affect immune cell responses to the parasite. In this study, our aim was to investigate whether non-coding variants that regulate FcγR expression could influence the prevalence of Plasmodium falciparum infection. Through bioinformatics approaches, we selected expression quantitative trait loci (eQTL) for FCGR2A, FCGR2B, FCGR2C, FCGR3A, and FCGR3B genes encoding FcγRs (FCGR), in whole blood. We prioritized two regulatory variants, rs2099684 and rs1771575, located in open genomic regions. These variants were identified using RegVar, ImmuNexUT, and transcription factor annotations specific to immune cells. In addition to these, we genotyped the coding variants FCGR2A/rs1801274 and FCGR2B/rs1050501 in 234 individuals from a malaria-endemic area in Burkina Faso. We conducted age and family-based analyses to evaluate associations with the prevalence of malarial infection in both children and adults. The analysis revealed that the regulatory rs1771575-CC genotype was predicted to influence FCGR2B/FCGR2C/FCGR3A transcripts in immune cells and was the sole variant associated with a higher prevalence of malarial infection in children. In conclusion, this study identifies the rs1771575 cis-regulatory variant affecting several FcγRs in myeloid and neutrophil cells and associates it with the inter-individual capacity of children living in Burkina Faso to control malarial infection.
Collapse
Affiliation(s)
- Jules Cretin
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
- Institut MarMaRa, 13288 Marseille, France
| | - Mathieu Adjemout
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
- Institut MarMaRa, 13288 Marseille, France
| | - Christelle Dieppois
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Frederic Gallardo
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Magali Torres
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Zachary Merard
- ADES UMR, Aix Marseille University, 13288 Marseille, France (C.P.)
| | - Serge Aimé Sawadogo
- Unité de Formation en Sciences de la Santé (UFR/SDS), Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso;
- Centre PrïmO-Nelson Mandela, 84 rue Sao Tomé et Principe, Ouagadougou 09 BP 706, Burkina Faso
| | - Christophe Picard
- ADES UMR, Aix Marseille University, 13288 Marseille, France (C.P.)
- Immunogenetics Laboratory, Etablissement Français du Sang PACA-Corse, 13001 Marseille, France
| | - Pascal Rihet
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Pascale Paul
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| |
Collapse
|
11
|
Bennion KB, Tariq M, Wyatt MM, Duneton C, Baecher KM, Paulos CM, Kudchadkar RR, Lowe MC, Ford ML. FcγRIIB expressed on CD8 + T cells limits responsiveness to PD-1 checkpoint inhibition in cancer. Sci Transl Med 2023; 15:eadd1868. [PMID: 37611081 PMCID: PMC11325091 DOI: 10.1126/scitranslmed.add1868] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/26/2023] [Indexed: 08/25/2023]
Abstract
Checkpoint inhibition using Fc-containing monoclonal antibodies has emerged as a powerful therapeutic approach to augment antitumor immunity. We recently showed that FcγRIIB, the only inhibitory IgG-Fc receptor, is expressed on a population of highly differentiated effector CD8+ T cells in the tumors of mice and humans, raising the possibility that CD8+ T cell responses may be directly modulated by checkpoint inhibitor binding to T cell-expressed FcγRIIB. Here, we show that despite exhibiting strong proliferative and cytokine responses at baseline, human FcγRIIBpos CD8+ T cells exhibited reduced responsiveness to both PD-1 and CTLA-4 checkpoint inhibition as compared with FcγRIIBneg CD8+ T cells in vitro. Moreover, frequencies of FcγRIIBpos CD8+ T cells were reduced after treatment of patients with melanoma with nivolumab in vivo. This reduced responsiveness was FcγRIIB dependent, because conditional genetic deletion of FcγRIIB on tumor-specific CD8+ T cells improved response to checkpoint blockade in B16 and LLC mouse models of cancer. The limited responsiveness of FcγRIIBpos CD8+ T cells was also dependent on an intact Fc region of the checkpoint inhibitor, in that treatment with Fc-devoid anti-PD-1 F(ab) fragments resulted in increased proliferation of FcγRIIBpos CD8+ T cells, without altering the response of FcγRIIBneg CD8+ T cells. Last, the addition of FcγRIIB blockade improved efficacy of PD-1 checkpoint inhibition in mouse models of melanoma, lung, and colon cancer. These results illuminate an FcγRIIB-mediated, cell-autonomous mechanism of CD8+ T cell suppression, which limits the efficacy of checkpoint inhibitors during antitumor immune responses in vivo.
Collapse
Affiliation(s)
- Kelsey B Bennion
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Marvi Tariq
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Megan M Wyatt
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Charlotte Duneton
- Paediatric Nephrology, Robert Debré Hospital, Paris 75019, France
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kirsten M Baecher
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Chrystal M Paulos
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ragini R Kudchadkar
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael C Lowe
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mandy L Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
12
|
Dadas O, Ertay A, Cragg MS. Delivering co-stimulatory tumor necrosis factor receptor agonism for cancer immunotherapy: past, current and future perspectives. Front Immunol 2023; 14:1147467. [PMID: 37180119 PMCID: PMC10167284 DOI: 10.3389/fimmu.2023.1147467] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/27/2023] [Indexed: 05/15/2023] Open
Abstract
The tumor necrosis factor superfamily (TNFSF) and their receptors (TNFRSF) are important regulators of the immune system, mediating proliferation, survival, differentiation, and function of immune cells. As a result, their targeting for immunotherapy is attractive, although to date, under-exploited. In this review we discuss the importance of co-stimulatory members of the TNFRSF in optimal immune response generation, the rationale behind targeting these receptors for immunotherapy, the success of targeting them in pre-clinical studies and the challenges in translating this success into the clinic. The efficacy and limitations of the currently available agents are discussed alongside the development of next generation immunostimulatory agents designed to overcome current issues, and capitalize on this receptor class to deliver potent, durable and safe drugs for patients.
Collapse
Affiliation(s)
- Osman Dadas
- Antibody and Vaccine Group, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ayse Ertay
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Mark S. Cragg
- Antibody and Vaccine Group, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
13
|
Knorr D, Leidner R, Jensen S, Meng R, Jones A, Ballesteros-Merino C, Bell RB, Baez M, Sprott D, Bifulco C, Piening B, Dahan R, Fox BA, Ravetch J. FcyRIIB is a novel immune checkpoint in the tumor microenvironment limiting activity of Treg-targeting antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.522856. [PMID: 36711504 PMCID: PMC9884505 DOI: 10.1101/2023.01.19.522856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Despite pre-clinical murine data supporting T regulatory (Treg) cell depletion as a major mechanism by which anti-CTLA-4 antibodies function in vivo, the two main antibodies tested in patients (ipilimumab and tremelimumab) have failed to demonstrate similar effects. We report analogous findings in an immunocompetent murine model humanized for CTLA-4 and Fcy receptors (hCTLA-4/hFcyR mice), where both ipilimumab and tremelimumab fail to show appreciable Treg depletion. Immune profiling of the tumor microenvironment (TME) in both mice and human samples revealed upregulation of the inhibitory Fcy receptor, FcyRIIB, which limits the ability of the antibody Fc fragment of human anti-CTLA-4 antibodies to induce effective antibody dependent cellular cytotoxicty/phagocytosis (ADCC/ADCP). Blocking FcyRIIB in humanized mice rescues Treg depleting capacity and anti-tumor activity of ipilimumab. For another target, CC motif chemokine receptor 8 (CCR8), which is selectively expressed on tumor infiltrating Tregs, we show that Fc engineering to enhance binding to activating Fc receptors, while limiting binding to the inhibitory Fc receptor, leads to consistent Treg depletion and single-agent activity across multiple tumor models, including B16, MC38 and MB49. These data reveal the importance of reducing engagement to the inhibitory Fc receptor to optimize Treg depletion by TME targeting antibodies. Our results define the inhibitory FcyRIIB receptor as a novel immune checkpoint limiting antibody-mediated Treg depletion in tumors, and demonstrate Fc variant engineering as a means to overcome this limitation and augment efficacy for a repertoire of antibodies currently in use or under clinical evaluation in oncology.
Collapse
Affiliation(s)
- David Knorr
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rom Leidner
- Earle A. Chiles Research Institute, a division of Providence Cancer Institute, Portland, OR
| | - Shawn Jensen
- Earle A. Chiles Research Institute, a division of Providence Cancer Institute, Portland, OR
| | - Ryan Meng
- Earle A. Chiles Research Institute, a division of Providence Cancer Institute, Portland, OR
| | - Andrew Jones
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY
| | | | - R. Bryan Bell
- Earle A. Chiles Research Institute, a division of Providence Cancer Institute, Portland, OR
| | - Maria Baez
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY
| | - David Sprott
- Earle A. Chiles Research Institute, a division of Providence Cancer Institute, Portland, OR
| | - Carlo Bifulco
- Earle A. Chiles Research Institute, a division of Providence Cancer Institute, Portland, OR
| | - Brian Piening
- Earle A. Chiles Research Institute, a division of Providence Cancer Institute, Portland, OR
| | - Rony Dahan
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Bernard A. Fox
- Earle A. Chiles Research Institute, a division of Providence Cancer Institute, Portland, OR
| | - Jeffrey Ravetch
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY
| |
Collapse
|