1
|
Sepahi A, Ho HE, Vyas P, Umiker B, Kis-Toth K, Wiederschain D, Radigan L, Cunningham-Rundles C. ICOS agonist vopratelimab modulates follicular helper T cells and improves B cell function in common variable immunodeficiency. Clin Immunol 2024; 264:110217. [PMID: 38621471 DOI: 10.1016/j.clim.2024.110217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
Common variable immunodeficiency (CVID) is an immune defect characterized by hypogammaglobulinemia and impaired development of B cells into plasma cells. As follicular helper T cells (TFH) play a central role in humoral immunity, we examined TFH cells in CVID, and investigated whether an inducible T cell co-stimulator (ICOS) agonist, vopratelimab, could modulate TFH, B cell interactions and enhance immunoglobulin production. CVID subjects had decreased TFH17 and increased TFH1 subsets; this was associated with increased transitional B cells and decreased IgG+ B and IgD-IgM-CD27+ memory B cells. ICOS expression on CVID CD4+ T cells was also decreased. However, ICOS activation of CD4+ T cells by vopratelimab significantly increased total CVID TFH, TFH2, cell numbers, as well as IL-4, IL-10 and IL-21 secretion in vitro. Vopratelimab treatment also increased plasma cells, IgG+ B cells, reduced naïve & transitional B cells and significantly increased IgG1 secretion by CVID B cells. Interestingly, vopratelimab treatment also restored IgA secretion in PBMCs from several CVID patients who had a complete lack of endogenous serum IgA. Our data demonstrate the potential of TFH modulation in restoring TFH and enhancing B cell maturation in CVID. The effects of an ICOS agonist in antibody defects warrants further investigation. This biologic may also be of therapeutic interest in other clinical settings of antibody deficiency.
Collapse
Affiliation(s)
- Ali Sepahi
- PharmaEssentia Innovation Research Center, Bedford, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Hsi-En Ho
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Prapti Vyas
- ReNAgade Therapeutics, Cambridge, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Benjamin Umiker
- AstraZeneca, Cambridge, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Katalin Kis-Toth
- NextPoint Therapeutics, Inc., Cambridge, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Dmitri Wiederschain
- Crossbow Therapeutics, Cambridge, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Lin Radigan
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Charlotte Cunningham-Rundles
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
2
|
Philips EA, Liu J, Kvalvaag A, Mørch AM, Tocheva AS, Ng C, Liang H, Ahearn IM, Pan R, Luo CC, Leithner A, Qin Z, Zhou Y, Garcia-España A, Mor A, Littman DR, Dustin ML, Wang J, Kong XP. Transmembrane domain-driven PD-1 dimers mediate T cell inhibition. Sci Immunol 2024; 9:eade6256. [PMID: 38457513 PMCID: PMC11166110 DOI: 10.1126/sciimmunol.ade6256] [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: 08/28/2022] [Accepted: 02/15/2024] [Indexed: 03/10/2024]
Abstract
Programmed cell death-1 (PD-1) is a potent immune checkpoint receptor on T lymphocytes. Upon engagement by its ligands, PD-L1 or PD-L2, PD-1 inhibits T cell activation and can promote immune tolerance. Antagonism of PD-1 signaling has proven effective in cancer immunotherapy, and conversely, agonists of the receptor may have a role in treating autoimmune disease. Some immune receptors function as dimers, but PD-1 has been considered monomeric. Here, we show that PD-1 and its ligands form dimers as a consequence of transmembrane domain interactions and that propensity for dimerization correlates with the ability of PD-1 to inhibit immune responses, antitumor immunity, cytotoxic T cell function, and autoimmune tissue destruction. These observations contribute to our understanding of the PD-1 axis and how it can potentially be manipulated for improved treatment of cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Elliot A. Philips
- Departments of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jia Liu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Audun Kvalvaag
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
| | - Alexander M. Mørch
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Anna S. Tocheva
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, New York, NY 10029, USA
| | - Charles Ng
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Hong Liang
- Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Ian M. Ahearn
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Ruimin Pan
- Departments of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Christina C. Luo
- Departments of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alexander Leithner
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Zhihua Qin
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Yong Zhou
- Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Antonio Garcia-España
- Research Unit, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
| | - Adam Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Dan R. Littman
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Michael L. Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Jun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Xiang-Peng Kong
- Departments of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
3
|
Saha S, Sparkes A, Matus EI, Lee P, Gariépy J. The IgV domain of the poliovirus receptor alone is immunosuppressive and binds to its receptors with comparable affinity. Sci Rep 2023; 13:4609. [PMID: 36944702 PMCID: PMC10030575 DOI: 10.1038/s41598-023-30999-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023] Open
Abstract
PVR (poliovirus receptor) functions as a ligand that signals through TIGIT and CD96 to induce suppression of T-cell and NK-cell responses. Alternatively, PVR binds to CD226, resulting in a co-stimulatory signal. To date, TIGIT antibody antagonists have been developed to restore immune functions and allow PVR to signal though CD226 in the context of cancer immunotherapy. Due to PVR receptor heterogeneity, agonizing either of these pathways with a recombinant form of the PVR extracellular domain represents a therapeutic strategy for either immunosuppression or activation. Here, we developed a minimal murine PVR-Fc fusion construct, consisting of only the IgV domain of PVR (vdPVR-Fc), and assessed its ability to dampen inflammatory responses in a murine model of psoriasis. vdPVR-Fc and PVR-Fc containing the full-length extracellular domain bound to TIGIT, CD96 and CD226 with similar low nanomolar affinities as defined by surface plasmon resonance. vdPVR-Fc was also able to suppress the in-vitro proliferation of murine CD4+ and CD8+ T-cells in mixed splenocyte cultures. Importantly, vdPVR-Fc delayed the onset, and reduced inflammatory responses (scaling and thickness) in a murine model of psoriasis. Collectively, our results suggest that the minimal IgV domain of PVR is sufficient to dampen immune responses in-vitro and attenuate symptoms of psoriasis in-vivo.
Collapse
Affiliation(s)
- Shrayasee Saha
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
| | - Amanda Sparkes
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Esther I Matus
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Peter Lee
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Jean Gariépy
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada.
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.
- Physical Sciences, Sunnybrook Research Institute, 2075 Bayview Ave., Room M7-434, Toronto, ON, M4N 3M5, Canada.
| |
Collapse
|
4
|
Arbitman L, Furie R, Vashistha H. B cell-targeted therapies in systemic lupus erythematosus. J Autoimmun 2022; 132:102873. [PMID: 35963808 DOI: 10.1016/j.jaut.2022.102873] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 10/15/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease of unknown etiology that primarily affects women of childbearing age. There is no disease more heterogeneous than SLE as patients experience a myriad of manifestations and unpredictable periods of heightened disease activity. This heterogeneity not only makes it difficult for treatment decisions and prognostication, but has made drug development quite challenging. Despite these challenges, belimumab, voclosporin, and anifromulab, approved by the United States Food and Drug Administration (FDA) to treat SLE or lupus nephritis (LN), enhanced our armamentarium of traditional therapies, such as hydroxychloroquine, corticosteroids, and immunosuppressives. However, there remains a dire need to develop therapies that offer greater efficacy and safety. Patients with SLE produce excessive amounts of autoantibodies and cytokines that result in inflammation and organ damage. While a considerable number of potential drug development targets exist, there has been much attention focused on B cells. Strategies have included direct B cell killing, modulation of B cell function, inhibition of molecules essential to B cell growth and survival, and acceleration of autoantibody clearance, to name just a few. In this article, we review SLE clinical trials evaluating experimental agents that target B cells or plasma cells.
Collapse
Affiliation(s)
- Leah Arbitman
- Harpur College of Arts and Sciences, Binghamton University, Binghamton, NY, USA
| | - Richard Furie
- Division of Rheumatology Northwell Health and Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | - Himanshu Vashistha
- Division of Rheumatology, Department of Medicine, Northwell Health, Great Neck, NY, USA.
| |
Collapse
|
5
|
Boggio E, Gigliotti CL, Moia R, Scotta A, Crespi I, Boggione P, De Paoli L, Deambrogi C, Garzaro M, Vidali M, Chiocchetti A, Stoppa I, Rolla R, Dianzani C, Monge C, Clemente N, Gaidano G, Dianzani U. Inducible T-cell co-stimulator (ICOS) and ICOS ligand are novel players in the multiple-myeloma microenvironment. Br J Haematol 2021; 196:1369-1380. [PMID: 34954822 DOI: 10.1111/bjh.17968] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/19/2022]
Abstract
The inducible T-cell co-stimulator (ICOS) is a T-cell receptor that, once bound to ICOS ligand (ICOSL) expressed on several cell types including the B-cell lineage, plays a decisive role in adaptive immunity by regulating the interplay between B and T cells. In addition to its immunomodulatory functions, we have shown that ICOS/ICOSL signalling can inhibit the activity of osteoclasts, unveiling a novel mechanism of lymphocyte-bone cells interactions. ICOS and ICOSL can also be found as soluble forms, namely sICOS and sICOSL. Here we show that: (i) levels of sICOS and sICOSL are increased in multiple myeloma (MM) compared to monoclonal gammopathy of undetermined significance and smouldering MM; (ii) levels of sICOS and sICOSL variably correlate with several markers of tumour burden; and (iii) sICOS levels tend to be higher in Durie-Salmon stage II/III versus stage I MM and correlate with overall survival as an independent variable. Moreover, surface ICOS and ICOSL are expressed in both myeloma cells and normal plasma cells, where they probably regulate different functional stages. Finally, ICOSL triggering inhibits the migration of myeloma cell lines in vitro and the growth of ICOSL+ MOPC-21 myeloma cells in vivo. These results suggest that ICOS and ICOSL represent novel markers and therapeutic targets for MM.
Collapse
Affiliation(s)
- Elena Boggio
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Casimiro Luca Gigliotti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | | | - Ilaria Crespi
- Maggiore della Carità University Hospital, Novara, Italy
| | - Paola Boggione
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Lorenzo De Paoli
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Clara Deambrogi
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Massimiliano Garzaro
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Matteo Vidali
- Maggiore della Carità University Hospital, Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Ian Stoppa
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Roberta Rolla
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Chiara Monge
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Nausicaa Clemente
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| |
Collapse
|
6
|
Goenka R, Xu Z, Samayoa J, Banach D, Beam C, Bose S, Dooner G, Forsyth CM, Lu X, Medina L, Sadhukhan R, Sielaff B, Sousa S, Tao Q, Touw D, Wu F, Kingsbury GA, Akamatsu Y. CTLA4-Ig-Based Bifunctional Costimulation Inhibitor Blocks CD28 and ICOS Signaling to Prevent T Cell Priming and Effector Function. THE JOURNAL OF IMMUNOLOGY 2021; 206:1102-1113. [PMID: 33495237 DOI: 10.4049/jimmunol.2001100] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/21/2020] [Indexed: 11/19/2022]
Abstract
CTLA4-Ig/abatacept dampens activation of naive T cells by blocking costimulation via CD28. It is an approved drug for rheumatoid arthritis but failed to deliver efficacy in a number of other autoimmune diseases. One explanation is that activated T cells rely less on CD28 signaling and use alternate coreceptors for effector function. ICOS is critical for activation of T-dependent humoral immune responses, which drives pathophysiology of IgG-mediated autoimmune diseases. In this study, we asked whether CD28 and ICOS play nonredundant roles for maintenance of T-dependent responses in mouse models. Using a hapten-protein immunization model, we show that during an ongoing germinal center response, combination treatment with CTLA4-Ig and ICOS ligand (ICOSL) blocking Ab completely dissolves ongoing germinal center responses, whereas single agents show only partial activity. Next, we took two approaches to engineer a therapeutic molecule that blocks both pathways. First, we engineered CTLA4-Ig to enhance binding to ICOSL while retaining affinity to CD80/CD86. Using a library approach, binding affinity of CTLA4-Ig to human ICOSL was increased significantly from undetectable to 15-42 nM; however, the affinity was still insufficient to completely block binding of ICOSL to ICOS. Second, we designed a bispecific costimulation inhibitor with high-affinity CTLA4 extracellular domains fused to anti-ICOSL Ab termed bifunctional costimulation inhibitor. With this bispecific approach, we achieved complete inhibition of CD80 and CD86 binding to CD28 as well as ICOS binding to ICOSL. Such bispecific molecules may provide greater therapeutic benefit in IgG-mediated inflammatory diseases compared with CTLA4-Ig alone.
Collapse
Affiliation(s)
| | - Zhenghai Xu
- AbbVie Redwood City, Redwood City, CA 94306; and
| | | | | | | | - Sahana Bose
- AbbVie Bioresearch Center, Worcester, MA 01605
| | | | | | - Xiaoqing Lu
- AbbVie Cambridge Research Center, Cambridge, MA 02139
| | | | | | | | | | - Qingfeng Tao
- AbbVie Cambridge Research Center, Cambridge, MA 02139
| | - Debra Touw
- AbbVie Bioresearch Center, Worcester, MA 01605
| | - Fei Wu
- AbbVie Bioresearch Center, Worcester, MA 01605
| | | | | |
Collapse
|
7
|
Aragoneses-Fenoll L, Montes-Casado M, Ojeda G, García-Paredes L, Arimura Y, Yagi J, Dianzani U, Portolés P, Rojo JM. Role of endocytosis and trans-endocytosis in ICOS costimulator-induced downmodulation of the ICOS Ligand. J Leukoc Biol 2021; 110:867-884. [PMID: 33527556 PMCID: PMC8597029 DOI: 10.1002/jlb.2a0220-127r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 12/16/2020] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
The interaction between the T-lymphocyte costimulatory molecule ICOS and its ligand (ICOS-L) is needed for efficient immune responses, but expression levels are tightly controlled, as altered expression of ICOS or ICOS-L may lead to immunodeficiency, or favor autoimmune diseases and tumor growth. Using cells of mouse B cell lymphoma (M12.C3) and melanoma (B16), or hamster CHO cells transfected with various forms of mouse ICOS-L, and ICOS+ T cell lines, we show that, within minutes, ICOS induces significant downmodulation of surface ICOS-L that is largely mediated by endocytosis and trans-endocytosis. So, after interaction with ICOS+ cells, ICOS-L was found inside permeabilized cells, or in cell lysates, with significant transfer of ICOS from ICOS+ T cells to ICOS-L-expressing cells, and simultaneous loss of surface ICOS by the T cells. Data from cells expressing ICOS-L mutants show that conserved, functionally important residues in the cytoplasmic domain of mouse ICOS-L (Arg300 , Ser307 and Tyr308 ), or removal of ICOS-L cytoplasmic tail have minor effect on its internalization. Internalization was dependent on temperature, and was partially dependent on actin polymerization, the GTPase dynamin, protein kinase C, or the integrity of lipid rafts. In fact, a fraction of ICOS-L was detected in lipid rafts. On the other hand, proteinase inhibitors had negligible effects on early modulation of ICOS-L from the cell surface. Our data add a new mechanism of control of ICOS-L expression to the regulation of ICOS-dependent responses.
Collapse
Affiliation(s)
- Laura Aragoneses-Fenoll
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - María Montes-Casado
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - Gloria Ojeda
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - Lucía García-Paredes
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain.,Current address: Hospital 12 de Octubre, Departamento de Oncología Médica, Av. de Córdoba, s/n, Madrid, 28041, Spain
| | - Yutaka Arimura
- Host Defense for Animals, Nippon Veterinary and Life Science University, 1-7-1 Kyonan, Musashino, Tokyo, 180-8602, Japan
| | - Junji Yagi
- Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, 108-8639, Japan
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Health Sciences, University of Piemonte Orientale (UPO), Novara, 28100, Italy
| | - Pilar Portolés
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain.,Presidencia, Consejo Superior de Investigaciones Científicas, Madrid, 28006, Spain
| | - José M Rojo
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain
| |
Collapse
|
8
|
Rujas E, Cui H, Sicard T, Semesi A, Julien JP. Structural characterization of the ICOS/ICOS-L immune complex reveals high molecular mimicry by therapeutic antibodies. Nat Commun 2020; 11:5066. [PMID: 33033255 PMCID: PMC7545189 DOI: 10.1038/s41467-020-18828-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022] Open
Abstract
The inducible co-stimulator (ICOS) is a member of the CD28/B7 superfamily, and delivers a positive co-stimulatory signal to activated T cells upon binding to its ligand (ICOS-L). Dysregulation of this pathway has been implicated in autoimmune diseases and cancer, and is currently under clinical investigation as an immune checkpoint blockade. Here, we describe the molecular interactions of the ICOS/ICOS-L immune complex at 3.3 Å resolution. A central FDPPPF motif and residues within the CC' loop of ICOS are responsible for the specificity of the interaction with ICOS-L, with a distinct receptor binding orientation in comparison to other family members. Furthermore, our structure and binding data reveal that the ICOS N110 N-linked glycan participates in ICOS-L binding. In addition, we report crystal structures of ICOS and ICOS-L in complex with monoclonal antibodies under clinical evaluation in immunotherapy. Strikingly, antibody paratopes closely mimic receptor-ligand binding core interactions, in addition to contacting peripheral residues to confer high binding affinities. Our results uncover key molecular interactions of an immune complex central to human adaptive immunity and have direct implications for the ongoing development of therapeutic interventions targeting immune checkpoint receptors.
Collapse
Affiliation(s)
- Edurne Rujas
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada.,Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080, Bilbao, Spain
| | - Hong Cui
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada
| | - Taylor Sicard
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Anthony Semesi
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada. .,Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
9
|
Hanson A, Elpek K, Duong E, Shallberg L, Fan M, Johnson C, Wallace M, Mabry GR, Sazinsky S, Pepper L, Shu CJ, Sathyanarayanan S, Zuerndorfer S, Simpson T, Gostissa M, Briskin M, Law D, Michaelson J, Harvey CJ. ICOS agonism by JTX-2011 (vopratelimab) requires initial T cell priming and Fc cross-linking for optimal T cell activation and anti-tumor immunity in preclinical models. PLoS One 2020; 15:e0239595. [PMID: 32970735 PMCID: PMC7514066 DOI: 10.1371/journal.pone.0239595] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy checkpoint inhibitors, such as antibodies targeting PD-1 and CTLA-4, have demonstrated the potential of harnessing the immune system to treat cancer. However, despite encouraging results particularly with respect to survival, only a minority of patients benefit from these therapies. In clinical studies aimed at understanding changes in the immune system following immunotherapy treatment, ICOS (Inducible T cell CO-Stimulator) was shown to be significantly up-regulated on CD4+ T cells and this was associated with clinical activity, indicating that ICOS stimulatory activity may be beneficial in the treatment of solid tumors. In this report, we describe the generation of specific, species cross-reactive, agonist antibodies to ICOS, including the humanized clinical candidate, JTX-2011 (vopratelimab). Preclinical studies suggest that the ICOS stimulating antibodies require Fc receptor cross-linking for optimal agonistic activity. Notably, the ICOS antibodies do not exhibit superagonist properties but rather require T cell receptor (TCR)-mediated upregulation of ICOS for agonist activity. Treatment with the ICOS antibodies results in robust anti-tumor benefit and long-term protection in preclinical syngeneic mouse tumor models. Additional benefit is observed when the ICOS antibodies are administered in combination with anti-PD-1 and anti-CTLA-4 therapies. Based on the preclinical data, JTX-2011 is currently being developed in the clinical setting for the treatment of solid tumors.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
- CHO Cells
- Cells, Cultured
- Cricetinae
- Cricetulus
- Cross-Priming
- Female
- Humans
- Immunotherapy/methods
- Inducible T-Cell Co-Stimulator Protein/immunology
- Jurkat Cells
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Receptors, Fc/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Amanda Hanson
- Preclinical Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Kutlu Elpek
- Preclinical Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Ellen Duong
- Preclinical Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Lindsey Shallberg
- Preclinical Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Martin Fan
- Preclinical Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Calvin Johnson
- Preclinical Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Matthew Wallace
- Protein Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - George R. Mabry
- Protein Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Stephen Sazinsky
- Protein Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Lauren Pepper
- Protein Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Chengyi J. Shu
- Translational Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Sriram Sathyanarayanan
- Translational Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Sarah Zuerndorfer
- Protein Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Tyler Simpson
- Preclinical Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Monica Gostissa
- Pharmacology, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Michael Briskin
- Research, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Deborah Law
- Research, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Jennifer Michaelson
- Preclinical Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
| | - Christopher J. Harvey
- Preclinical Sciences, Jounce Therapeutics, Inc., Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
10
|
Levin SD, Evans LS, Bort S, Rickel E, Lewis KE, Wu RP, Hoover J, MacNeil S, La D, Wolfson MF, Rixon MW, Dillon SR, Kornacker MG, Swanson R, Peng SL. Novel Immunomodulatory Proteins Generated via Directed Evolution of Variant IgSF Domains. Front Immunol 2020; 10:3086. [PMID: 32038630 PMCID: PMC6985287 DOI: 10.3389/fimmu.2019.03086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/17/2019] [Indexed: 12/30/2022] Open
Abstract
Immunoglobulin superfamily member (IgSF) proteins play a significant role in regulating immune responses with surface expression on all immune cell subsets, making the IgSF an attractive family of proteins for therapeutic targeting in human diseases. We have developed a directed evolution platform capable of engineering IgSF domains to increase affinities for cognate ligands and/or introduce binding to non-cognate ligands. Using this scientific platform, ICOSL domains have been derived with enhanced binding to ICOS and with additional high-affinity binding to the non-cognate receptor, CD28. Fc-fusion proteins containing these engineered ICOSL domains significantly attenuate T cell activation in vitro and in vivo and can inhibit development of inflammatory diseases in mouse models. We also present evidence that engineered ICOSL domains can be formatted to selectively provide costimulatory signals to augment T cell responses. Our scientific platform thus provides a system for developing therapeutic protein candidates with selective biological impact for treatments of a wide array of human disorders including cancer and autoimmune/inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Susan Bort
- Alpine Immune Sciences Inc., Seattle, WA, United States
| | - Erika Rickel
- Alpine Immune Sciences Inc., Seattle, WA, United States
| | | | - Rebecca P Wu
- Alpine Immune Sciences Inc., Seattle, WA, United States
| | - Joseph Hoover
- Alpine Immune Sciences Inc., Seattle, WA, United States
| | - Sean MacNeil
- Alpine Immune Sciences Inc., Seattle, WA, United States
| | - David La
- Department of Biochemistry, University of Washington, Seattle, WA, United States
| | | | - Mark W Rixon
- Alpine Immune Sciences Inc., Seattle, WA, United States
| | | | | | - Ryan Swanson
- Alpine Immune Sciences Inc., Seattle, WA, United States
| | | |
Collapse
|
11
|
Slater BT, Han X, Chen L, Xiong Y. Structural insight into T cell coinhibition by PD-1H (VISTA). Proc Natl Acad Sci U S A 2020; 117:1648-1657. [PMID: 31919279 PMCID: PMC6983362 DOI: 10.1073/pnas.1908711117] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Programmed death-1 homolog (PD-1H), a CD28/B7 family molecule, coinhibits T cell activation and is an attractive immunotherapeutic target for cancer and inflammatory diseases. The molecular basis of its function, however, is unknown. Bioinformatic analyses indicated that PD-1H has a very long Ig variable region (IgV)-like domain and extraordinarily high histidine content, suggesting that unique structural features may contribute to coinhibitory mechanisms. Here we present the 1.9-Å crystal structure of the human PD-1H extracellular domain. It reveals an elongated CC' loop and a striking concentration of histidine residues, located in the complementarity-determining region-like proximal half of the molecule. We show that surface-exposed histidine clusters are essential for robust inhibition of T cell activation. PD-1H exhibits a noncanonical IgV-like topology including an extra "H" β-strand and "clamping" disulfide, absent in known IgV-like structures, that likely restricts its orientation on the cell surface differently from other IgV-like domains. These results provide important insight into a molecular basis of T cell coinhibition by PD-1H.
Collapse
Affiliation(s)
| | - Xue Han
- Department of Immunobiology, Yale University, New Haven, CT 06511
| | - Lieping Chen
- Department of Immunobiology, Yale University, New Haven, CT 06511;
| | - Yong Xiong
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520
| |
Collapse
|
12
|
Roussel L, Landekic M, Golizeh M, Gavino C, Zhong MC, Chen J, Faubert D, Blanchet-Cohen A, Dansereau L, Parent MA, Marin S, Luo J, Le C, Ford BR, Langelier M, King IL, Divangahi M, Foulkes WD, Veillette A, Vinh DC. Loss of human ICOSL results in combined immunodeficiency. J Exp Med 2019; 215:3151-3164. [PMID: 30498080 PMCID: PMC6279397 DOI: 10.1084/jem.20180668] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 09/21/2018] [Accepted: 11/06/2018] [Indexed: 12/30/2022] Open
Abstract
Primary immunodeficiencies represent naturally occurring experimental models to decipher human immunobiology. We report a patient with combined immunodeficiency, marked by recurrent respiratory tract and DNA-based viral infections, hypogammaglobulinemia, and panlymphopenia. He also developed moderate neutropenia but without prototypical pyogenic infections. Using whole-exome sequencing, we identified a homozygous mutation in the inducible T cell costimulator ligand gene (ICOSLG; c.657C>G; p.N219K). Whereas WT ICOSL is expressed at the cell surface, the ICOSLN219K mutation abrogates surface localization: mutant protein is retained in the endoplasmic reticulum/Golgi apparatus, which is predicted to result from deleterious conformational and biochemical changes. ICOSLN219K diminished B cell costimulation of T cells, providing a compelling basis for the observed defect in antibody and memory B cell generation. Interestingly, ICOSLN219K also impaired migration of lymphocytes and neutrophils across endothelial cells, which normally express ICOSL. These defects likely contributed to the altered adaptive immunity and neutropenia observed in the patient, respectively. Our study identifies human ICOSLG deficiency as a novel cause of a combined immunodeficiency.
Collapse
Affiliation(s)
- Lucie Roussel
- Infectious Disease Susceptibility Program, McGill University Health Centre and Research Institute-McGill University Health Centre, Montréal, Québec, Canada
| | - Marija Landekic
- Infectious Disease Susceptibility Program, McGill University Health Centre and Research Institute-McGill University Health Centre, Montréal, Québec, Canada
| | - Makan Golizeh
- Infectious Disease Susceptibility Program, McGill University Health Centre and Research Institute-McGill University Health Centre, Montréal, Québec, Canada
| | - Christina Gavino
- Infectious Disease Susceptibility Program, McGill University Health Centre and Research Institute-McGill University Health Centre, Montréal, Québec, Canada
| | - Ming-Chao Zhong
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
| | - Jun Chen
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
| | - Denis Faubert
- Proteomics Discovery Platform, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
| | - Alexis Blanchet-Cohen
- Bioinformatics, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
| | - Luc Dansereau
- Department of Internal Medicine, Hôpital de l'Archipel, Centre intégré de santé et de services sociaux des Îles, Les Îles-de-la-Madeleine, Québec, Canada
| | - Marc-Antoine Parent
- Department of Family Medicine, Centre intégé de santé et de services sociaux des Îles, Les Îles-de-la-Madeleine, Québec, Canada
| | - Sonia Marin
- Hôpital de l'Archipel, Centre intégré de santé et de services sociaux des Îles, Les Îles-de-la-Madeleine, Québec, Canada
| | - Julia Luo
- Infectious Disease Susceptibility Program, McGill University Health Centre and Research Institute-McGill University Health Centre, Montréal, Québec, Canada
| | - Catherine Le
- Infectious Disease Susceptibility Program, McGill University Health Centre and Research Institute-McGill University Health Centre, Montréal, Québec, Canada
| | - Brinley R Ford
- Infectious Disease Susceptibility Program, McGill University Health Centre and Research Institute-McGill University Health Centre, Montréal, Québec, Canada
| | - Mélanie Langelier
- Infectious Disease Susceptibility Program, McGill University Health Centre and Research Institute-McGill University Health Centre, Montréal, Québec, Canada
| | - Irah L King
- Meakins-Christie Laboratories, Research Institute-McGill University Health Centre, Montréal, Québec, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Research Institute-McGill University Health Centre, Montréal, Québec, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada.,Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - William D Foulkes
- Department of Medical Genetics, Research Institute-McGill University Health Centre, Montréal, Québec, Canada.,Department of Human Genetics, McGill University, Montréal, Québec, Canada
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada.,Department of Medicine, University of Montréal, Montréal, Québec, Canada
| | - Donald C Vinh
- Infectious Disease Susceptibility Program, McGill University Health Centre and Research Institute-McGill University Health Centre, Montréal, Québec, Canada .,Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada.,Department of Human Genetics, McGill University, Montréal, Québec, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada
| |
Collapse
|
13
|
West SM, Deng XA. Considering B7-CD28 as a family through sequence and structure. Exp Biol Med (Maywood) 2019; 244:1577-1583. [PMID: 31208204 DOI: 10.1177/1535370219855970] [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] [Indexed: 12/30/2022] Open
Abstract
With the emergence of immuno-oncology, new therapeutic agents that modulate immune activation and regulation are being used to treat cancer patients with durable response. It is well known that following T-cell receptor (TCR) activation, many co-receptors can augment or suppress the TCR signal, and therapeutically targeting these co-receptors has proven effective. The B7-CD28 family is comprised of such immune-regulatory receptors, and antibodies against its members programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have revolutionized cancer treatment. These therapies promote an immune response against tumor cells, which demonstrated better long-term survival and tolerability compared to traditional cancer treatments. In this review we describe the history of the expanding B7-CD28 family, and by comparison of sequence and structure reveal that it is a non-traditional family. The family has grown to include proteins that share low sequence identity, generally grouped by regulation of immune response, which utilize the common immunoglobulin fold. This low level of commonality has provided additional challenges to the drug discovery process as the mechanisms and therapeutic potency between family members can vary greatly. Impact statement Immunotherapy as a field has dramatically expanded in the last decade in the area of oncology with efficacy demonstrated by PD-1, PD-L1, and CTLA-4 blockade. With all three “checkpoint blockade” receptors being in the B7-CD28 family, there has been increased interest in targeting other members in this family due to redundancy in immune regulation, i.e., the combination of therapeutic agents targeting multiple co-inhibitory receptors may yield additional antitumor efficacy. Therefore significant resources are being dedicated to developing additional B7-CD28 treatment options.
Collapse
Affiliation(s)
- Sean M West
- Bristol-Myers Squibb (BMS), Redwood City, CA 94063, USA
| | - Xiaodi A Deng
- Bristol-Myers Squibb (BMS), Redwood City, CA 94063, USA
| |
Collapse
|
14
|
Structures of Immune Checkpoints: An Overview on the CD28-B7 Family. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1172:63-78. [PMID: 31628651 DOI: 10.1007/978-981-13-9367-9_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The co-stimulation and co-inhibition signal pathways, immune checkpoints, are among the central mechanisms to regulate the T-cell immunity. Optimal signals involve intricate interactions of numerous ligands and receptors. Manipulation of these signals offers great clinical opportunities and has revolutionized the cancer treatment therapies. The 2018 Nobel Prize in Physiology or Medicine was awarded to James P. Allison and Tasuku Honjo in recognition of their discovery of cancer immunotherapy by inhibition of immune checkpoint molecules. Despite the landmark discovery in cancer immunotherapy, the efforts to harness immunity against cancer are also restricted by the limited knowledge on the co-stimulation and co-inhibition signaling networks. Understanding the structures of these molecules, in particular, tackling the interaction paradigms from the structural perspective, help to provide more accurate insights into the signaling mechanisms, which may further facilitate the development of novel biologics and improve the efficacy of the existing biologics against these targets. Here we review our current understanding on the structures of these co-stimulatory and co-inhibitory molecules. Specifically, we focus on the structural basis of several checkpoint molecules among the CD28-B7 family and discuss the therapeutic drugs against these targets for the treatment of human cancers, autoimmune disorders, and transplantation.
Collapse
|
15
|
The promise and challenges of immune agonist antibody development in cancer. Nat Rev Drug Discov 2018; 17:509-527. [PMID: 29904196 DOI: 10.1038/nrd.2018.75] [Citation(s) in RCA: 260] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immune cell functions are regulated by co-inhibitory and co-stimulatory receptors. The first two generations of cancer immunotherapy agents consist primarily of antagonist antibodies that block negative immune checkpoints, such as programmed cell death protein 1 (PD1) and cytotoxic T lymphocyte protein 4 (CTLA4). Looking ahead, there is substantial promise in targeting co-stimulatory receptors with agonist antibodies, and a growing number of these agents are making their way through various stages of development. This Review discusses the key considerations and potential pitfalls of immune agonist antibody design and development, their differentiating features from antagonist antibodies and the landscape of agonist antibodies in clinical development for cancer treatment.
Collapse
|
16
|
Kovaleva M, Johnson K, Steven J, Barelle CJ, Porter A. Therapeutic Potential of Shark Anti-ICOSL VNAR Domains is Exemplified in a Murine Model of Autoimmune Non-Infectious Uveitis. Front Immunol 2017; 8:1121. [PMID: 28993766 PMCID: PMC5622306 DOI: 10.3389/fimmu.2017.01121] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/25/2017] [Indexed: 01/11/2023] Open
Abstract
Induced costimulatory ligand (ICOSL) plays an important role in the activation of T cells through its interaction with the inducible costimulator, ICOS. Suppression of full T cell activation can be achieved by blocking this interaction and has been shown to be an effective means of ameliorating disease in models of autoimmunity and inflammation. In this study, we demonstrated the ability of a novel class of anti-ICOSL antigen-binding single domains derived from sharks (VNARs) to effectively reduce inflammation in a murine model of non-infectious uveitis. In initial selections, specific VNARs that recognized human ICOSL were isolated from an immunized nurse shark phage display library and lead domains were identified following their performance in a series of antigen selectivity and in vitro bioassay screens. High potency in cell-based blocking assays suggested their potential as novel binders suitable for further therapeutic development. To test this hypothesis, surrogate anti-mouse ICOSL VNAR domains were isolated from the same phage display library and the lead VNAR clone selected via screening in binding and ICOS/ICOSL blocking experiments. The VNAR domain with the highest potency in cell-based blocking of ICOS/ICOSL interaction was fused to the Fc portion of human IgG1 and was tested in vivo in a mouse model of interphotoreceptor retinoid-binding protein-induced uveitis. The anti-mICOSL VNAR Fc, injected systemically, resulted in a marked reduction of inflammation in treated mice when compared with untreated control animals. This approach inhibited disease progression to an equivalent extent to that seen for the positive corticosteroid control, cyclosporin A, reducing both clinical and histopathological scores. These results represent the first demonstration of efficacy of a VNAR binding domain in a relevant clinical model of disease and highlight the potential of VNARs for the treatment of auto-inflammatory conditions.
Collapse
Affiliation(s)
| | - Katherine Johnson
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | | | | | - Andrew Porter
- Elasmogen Ltd., Aberdeen, United Kingdom
- Department of Molecular and Cell Biology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
17
|
Lara-Rodríguez C, Alvarado-Vásquez N, Bernal D, Gorocica P, Zenteno E, Lascuraín R. CD3+ICOS+ T cells show differences in the synthesis of nitric oxide, IFN-γ, and IL-10 in patients with pulmonary tuberculosis or in healthy household contacts. Clin Exp Med 2015; 16:481-491. [DOI: 10.1007/s10238-015-0380-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/28/2015] [Indexed: 11/28/2022]
|
18
|
Girard T, Gaucher D, El-Far M, Breton G, Sékaly RP. CD80 and CD86 IgC domains are important for quaternary structure, receptor binding and co-signaling function. Immunol Lett 2014; 161:65-75. [DOI: 10.1016/j.imlet.2014.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 05/09/2014] [Indexed: 11/16/2022]
|
19
|
Commodaro AG, Pedregosa JF, Peron JP, Brandão W, Rizzo LV, Bueno V. The imbalance between Treg and Th17 cells caused by FTY720 treatment in skin allograft rejection. Clinics (Sao Paulo) 2012; 67:805-13. [PMID: 22892927 PMCID: PMC3400173 DOI: 10.6061/clinics/2012(07)17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 03/14/2012] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES FTY720 modulates CD4+T cells by the augmentation of regulatory T cell activity, secretion of suppressive cytokines and suppression of IL-17 secretion by Th17 cells. To further understand the process of graft rejection/acceptance, we evaluated skin allograft survival and associated events after FTY720 treatment. METHODS F1 mice (C57BL/6xBALB/c) and C57BL/6 mice were used as donors for and recipients of skin transplantation, respectively. The recipients were transplanted and either not treated or treated with FTY720 by gavage for 21 days to evaluate the allograft survival. In another set of experiments, the immunological evaluation was performed five days post-transplantation. The spleens, axillary lymph nodes and skin allografts of the recipient mice were harvested for phenotyping (flow cytometry), gene expression (real-time PCR) and cytokine (Bio-Plex) analysis. RESULTS The FTY720 treatment significantly increased skin allograft survival, reduced the number of cells in the lymph nodes and decreased the percentage of Tregs at this site in the C57BL/6 recipients. Moreover, the treatment reduced the number of graft-infiltrating cells and the percentage of CD4+ graft-infiltrating cells. The cytokine analysis (splenocytes) showed decreased levels of IL-10, IL-6 and IL-17 in the FTY720-treated mice. We also observed a decrease in the IL-10, IL-6 and IL-23 mRNA levels, as well as an increase in the IL-27 mRNA levels, in the splenocytes of the treated group. The FTY720-treated mice exhibited increased mRNA levels of IL-10, IL-27 and IL-23 in the skin graft. CONCLUSIONS Our results demonstrated prolonged but not indefinite skin allograft survival by FTY720 treatment. This finding indicates that the drug did not prevent the imbalance between Tr1 and Th17 cells in the graft that led to rejection.
Collapse
|
20
|
Srinivasan M, Dunker AK. Proline rich motifs as drug targets in immune mediated disorders. INTERNATIONAL JOURNAL OF PEPTIDES 2012; 2012:634769. [PMID: 22666276 PMCID: PMC3362030 DOI: 10.1155/2012/634769] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 02/15/2012] [Indexed: 12/26/2022]
Abstract
The current version of the human immunome network consists of nearly 1400 interactions involving approximately 600 proteins. Intermolecular interactions mediated by proline-rich motifs (PRMs) are observed in many facets of the immune response. The proline-rich regions are known to preferentially adopt a polyproline type II helical conformation, an extended structure that facilitates transient intermolecular interactions such as signal transduction, antigen recognition, cell-cell communication and cytoskeletal organization. The propensity of both the side chain and the backbone carbonyls of the polyproline type II helix to participate in the interface interaction makes it an excellent recognition motif. An advantage of such distinct chemical features is that the interactions can be discriminatory even in the absence of high affinities. Indeed, the immune response is mediated by well-orchestrated low-affinity short-duration intermolecular interactions. The proline-rich regions are predominantly localized in the solvent-exposed regions such as the loops, intrinsically disordered regions, or between domains that constitute the intermolecular interface. Peptide mimics of the PRM have been suggested as potential antagonists of intermolecular interactions. In this paper, we discuss novel PRM-mediated interactions in the human immunome that potentially serve as attractive targets for immunomodulation and drug development for inflammatory and autoimmune pathologies.
Collapse
Affiliation(s)
- Mythily Srinivasan
- Department of Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, Indiana University Purdue University at Indianapolis 1121 West Michigan Street, DS290, Indianapolis, IN 46268, USA
| | - A. Keith Dunker
- Department of Biochemistry and Molecular Biology and School of Informatics, Indiana University School of Medicine, Indiana University Purdue University at Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
21
|
Yao S, Zhu Y, Zhu G, Augustine M, Zheng L, Goode DJ, Broadwater M, Ruff W, Flies S, Xu H, Flies D, Luo L, Wang S, Chen L. B7-h2 is a costimulatory ligand for CD28 in human. Immunity 2011; 34:729-40. [PMID: 21530327 DOI: 10.1016/j.immuni.2011.03.014] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 01/31/2011] [Accepted: 03/18/2011] [Indexed: 01/23/2023]
Abstract
CD28 and CTLA-4 are cell surface cosignaling molecules essential for the control of T cell activation upon the engagement of their ligands B7-1 and B7-2 from antigen-presenting cells. By employing a receptor array assay, we have demonstrated that B7-H2, best known as the ligand of inducible costimulator, was a ligand for CD28 and CTLA-4 in human, whereas these interactions were not conserved in mouse. B7-H2 and B7-1 or B7-2 interacted with CD28 through distinctive domains. B7-H2-CD28 interaction was essential for the costimulation of human T cells' primary responses to allogeneic antigens and memory recall responses. Similar to B7-1 and B7-2, B7-H2 costimulation via CD28 induced survival factor Bcl-xL, downregulated cell cycle inhibitor p27(kip1), and triggered signaling cascade of ERK and AKT kinase-dependent pathways. Our findings warrant re-evaluation of CD28 and CTLA-4's functions previously attributed exclusively to B7-1 and B7-2 and have important implications in therapeutic interventions against human diseases.
Collapse
Affiliation(s)
- Sheng Yao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Maeda M, Ito Y, Hatanaka T, Hashiguchi S, Torikai M, Nakashima T, Sugimura K. Regulation of T cell response by blocking the ICOS signal with the B7RP-1-specific small antibody fragment isolated from human antibody phage library. MAbs 2009; 1:453-61. [PMID: 20065650 DOI: 10.4161/mabs.1.5.9633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A costimulatory signal is required for the full activation of T cells, in addition to the antigen-specific signal via the T cell receptor. The inducible costimulator, ICOS is one of the costimulatory molecules that play an essential role in this process, particularly in the expansion or the development of effector T cells. As blocking of the interaction between ICOS and its ligand, B7RP-1, suppresses the T cell response, it can be applied to the treatment of allograft rejection or autoimmune diseases. Here, we isolated four scFv clones that were specific to human B7RP-1 by biopanning a human antibody phage library. We found that three of these clones inhibited the interaction between ICOS-Fc and B7RP-1-Fc. These inhibitory clones not only recognized B7RP-1 molecules expressed on B cells, as assessed by FACS, but also exhibited inhibitory activity in a proliferation assay of T cells stimulated with anti-CD3 mAb and B7RP-1-Fc. Finally, the suppression effect of the scFv on the allogenic immune response was examined using a mixed lymphocyte reaction assay, which demonstrated a successful inhibition of the allogenic reaction, in spite of the high dose needed for complete inhibition (360 nM).
Collapse
Affiliation(s)
- Masatoshi Maeda
- Department of Chemistry, Biotechnology and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Kagoshima, Japan
| | | | | | | | | | | | | |
Collapse
|
23
|
Chattopadhyay K, Lazar-Molnar E, Yan Q, Rubinstein R, Zhan C, Vigdorovich V, Ramagopal UA, Bonanno J, Nathenson SG, Almo SC. Sequence, structure, function, immunity: structural genomics of costimulation. Immunol Rev 2009; 229:356-86. [PMID: 19426233 DOI: 10.1111/j.1600-065x.2009.00778.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
SUMMARY Costimulatory receptors and ligands trigger the signaling pathways that are responsible for modulating the strength, course, and duration of an immune response. High-resolution structures have provided invaluable mechanistic insights by defining the chemical and physical features underlying costimulatory receptor:ligand specificity, affinity, oligomeric state, and valency. Furthermore, these structures revealed general architectural features that are important for the integration of these interactions and their associated signaling pathways into overall cellular physiology. Recent technological advances in structural biology promise unprecedented opportunities for furthering our understanding of the structural features and mechanisms that govern costimulation. In this review, we highlight unique insights that have been revealed by structures of costimulatory molecules from the immunoglobulin and tumor necrosis factor superfamilies and describe a vision for future structural and mechanistic analysis of costimulation. This vision includes simple strategies for the selection of candidate molecules for structure determination and highlights the critical role of structure in the design of mutant costimulatory molecules for the generation of in vivo structure-function correlations in a mammalian model system. This integrated 'atoms-to-animals' paradigm provides a comprehensive approach for defining atomic and molecular mechanisms.
Collapse
Affiliation(s)
- Kausik Chattopadhyay
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Jin J, Yang SH, Park J, Lee C, Lee J, Kim SM, Kim YS. Fragile maintenance of allograft tolerance induced by lymphocyte sequestration and co-stimulation blockade. Transpl Immunol 2009; 21:221-7. [PMID: 19539029 DOI: 10.1016/j.trim.2009.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 06/04/2009] [Accepted: 06/04/2009] [Indexed: 10/20/2022]
Abstract
The induction of long-term graft survival has been a goal for the last decade. Nevertheless, the issues of stable maintenance of allograft have not yet been evaluated thoroughly. Here, we studied new approaches for induction of tolerance by lymphocyte sequestration (FTY720) and co-stimulatory blockade (MR1) in skin graft model (DBA/2 to BALB/c), thus evaluating the mechanisms incorporated into the maintenance of allograft in proper function. FTY720+MR1 treatment significantly prolonged graft survival than single agent treatment did, and induced long-term graft survival in 60% of recipients expressing the up-regulation of IL-4 and FoxP3. To assess the stability of graft maintenance, we performed the second transplantation on recipients that had shown long-term graft survival. While recipients accepted the second graft from the same strain of first donor, the recipients not only rejected the third-party skin (C57BL/6) promptly but also rejected the first graft soon after the third-party skin was transplanted. The expression patterns of IL-4 and FoxP3 were changed according to the strains of second graft in lymph nodes and in the first graft. T(reg) cells from tolerant recipients effectively suppressed allo-antigen driven T cell proliferation, but T(reg) cells from recipients primed with third-party antigen had significantly hampered suppressive capacity against previously tolerant antigens. Our data indicate that the combination treatment provides effective tool for the induction of long-term graft survival, and the maintenance of allograft in proper function is an actively regulated process.
Collapse
Affiliation(s)
- Jizhe Jin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
25
|
Bakkour S, Sha WC. Mapping of the ICOS binding surface of murine B7h using an unbiased, cellular library of B7h mutants created by cyclical packaging rescue. J Immunol Methods 2008; 332:151-61. [PMID: 18294651 DOI: 10.1016/j.jim.2008.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 01/12/2008] [Accepted: 01/17/2008] [Indexed: 11/28/2022]
Abstract
Functional studies of immunologically relevant molecules often involve time-consuming generation and cloning of gene mutations prior to introduction into mammalian cells. We describe here an alternative mutagenesis approach that relies solely on transfer of helper-free retroviral supernatants to rapidly create in virtually any cell line of interest a large cellular library that retrovirally expresses a defined number of independent point mutations in a gene of interest. Using this rapid non-cloning approach, we generated a 3T3 cellular library retrovirally expressing 2 x 10(5) mutants of the murine costimulatory B7h gene. Screening of this unbiased cellular library identified six residues of murine B7h that are critical for binding to the ICOS receptor. These residues are located on the same strands of human B7h that were identified by targeted mutagenesis [Chattopadhyay, K., Bhatia, S., Fiser, A., Almo, S.C., Nathenson, S.G. (2006). Structural basis of inducible costimulator ligand costimulatory function: determination of the cell surface oligomeric state and functional mapping of the receptor-binding site of the protein. J. Immunol. 177, 3920], indicating that the ICOS receptor-binding interface is similar in mouse and human B7h. Based on this proof-of-principle study, CPR-based mutagenesis is applicable to studies of gene function in a variety of mammalian cells.
Collapse
Affiliation(s)
- Sonia Bakkour
- Cancer Research Laboratory, 441 LSA, Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3200 United States
| | | |
Collapse
|
26
|
Evolution of GITRL immune function: murine GITRL exhibits unique structural and biochemical properties within the TNF superfamily. Proc Natl Acad Sci U S A 2008; 105:635-40. [PMID: 18182486 DOI: 10.1073/pnas.0710529105] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoid-induced TNF receptor ligand (GITRL), a recently identified member of the TNF superfamily, binds to its receptor, GITR, on both effector and regulatory T cells and generates positive costimulatory signals implicated in a wide range of T cell functions. In contrast to all previously characterized homotrimeric TNF family members, the mouse GITRL crystal structure reveals a previously unrecognized dimeric assembly that is stabilized via a unique "domain-swapping" interaction. Consistent with its crystal structure, mouse GITRL exists as a stable dimer in solution. Structure-guided mutagenesis studies confirmed the determinants responsible for dimerization and support a previously unrecognized receptor-recognition surface for mouse GITRL that has not been observed for any other TNF family members. Taken together, the unique structural and biochemical behavior of mouse GITRL, along with the unusual domain organization of murine GITR, support a previously unrecognized mechanism for signaling within the TNF superfamily.
Collapse
|
27
|
Abstract
We describe an automated method for the modeling of point mutations in protein structures. The protein is represented by all non-hydrogen atoms. The scoring function consists of several types of physical potential energy terms and homology-derived restraints. The optimization method implements a combination of conjugate gradient minimization and molecular dynamics with simulated annealing. The testing set consists of 717 pairs of known protein structures differing by a single mutation. Twelve variations of the scoring function were tested in three different environments of the mutated residue. The best-performing protocol optimizes all the atoms of the mutated residue, with respect to a scoring function that includes molecular mechanics energy terms for bond distances, angles, dihedral angles, peptide bond planarity, and non-bonded atomic contacts represented by Lennard-Jones potential, dihedral angle restraints derived from the aligned homologous structure, and a statistical potential for non-bonded atomic interactions extracted from a large set of known protein structures. The current method compares favorably with other tested approaches, especially when predicting long and flexible side-chains. In addition to the thoroughness of the conformational search, sampled degrees of freedom, and the scoring function type, the accuracy of the method was also evaluated as a function of the flexibility of the mutated side-chain, the relative volume change of the mutated residue, and its residue type. The results suggest that further improvement is likely to be achieved by concentrating on the improvement of the scoring function, in addition to or instead of increasing the variety of sampled conformations.
Collapse
Affiliation(s)
- Eric Feyfant
- Wyeth Research, Chemical and Screening Sciences, Cambridge, Massachusetts 02421, USA
| | | | | |
Collapse
|