1
|
Van Der Byl W, Nüssing S, Peters TJ, Ahn A, Li H, Ledergor G, David E, Koh AS, Wagle MV, Deguit CDT, de Menezes MN, Travers A, Sampurno S, Ramsbottom KM, Li R, Kallies A, Beavis PA, Jungmann R, Bastings MMC, Belz GT, Goel S, Trapani JA, Crabtree GR, Chang HY, Amit I, Goodnow CC, Luciani F, Parish IA. The CD8 + T cell tolerance checkpoint triggers a distinct differentiation state defined by protein translation defects. Immunity 2024; 57:1324-1344.e8. [PMID: 38776918 PMCID: PMC11807353 DOI: 10.1016/j.immuni.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/01/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Peripheral CD8+ T cell tolerance is a checkpoint in both autoimmune disease and anti-cancer immunity. Despite its importance, the relationship between tolerance-induced states and other CD8+ T cell differentiation states remains unclear. Using flow cytometric phenotyping, single-cell RNA sequencing (scRNA-seq), and chromatin accessibility profiling, we demonstrated that in vivo peripheral tolerance to a self-antigen triggered a fundamentally distinct differentiation state separate from exhaustion, memory, and functional effector cells but analogous to cells defectively primed against tumors. Tolerant cells diverged early and progressively from effector cells, adopting a transcriptionally and epigenetically distinct state within 60 h of antigen encounter. Breaching tolerance required the synergistic actions of strong T cell receptor (TCR) signaling and inflammation, which cooperatively induced gene modules that enhanced protein translation. Weak TCR signaling during bystander infection failed to breach tolerance due to the uncoupling of effector gene expression from protein translation. Thus, tolerance engages a distinct differentiation trajectory enforced by protein translation defects.
Collapse
Affiliation(s)
- Willem Van Der Byl
- The Kirby Institute for Infection and Immunity, UNSW, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine, UNSW, Sydney, NSW, Australia
| | - Simone Nüssing
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Timothy J Peters
- Garvan Institute of Medical Research, Sydney, NSW, Australia; University of New South Wales Sydney, Sydney, NSW, Australia
| | - Antonio Ahn
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Hanjie Li
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Guy Ledergor
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Andrew S Koh
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Mayura V Wagle
- Garvan Institute of Medical Research, Sydney, NSW, Australia; John Curtin School of Medical Research, ANU, Canberra, ACT, Australia
| | | | - Maria N de Menezes
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Avraham Travers
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Shienny Sampurno
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Kelly M Ramsbottom
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rui Li
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia; Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany; Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Maartje M C Bastings
- Institute of Materials, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Interfaculty Bioengineering Institute, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Gabrielle T Belz
- The Frazer Institute, The University of Queensland, Brisbane, QLD, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Shom Goel
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Gerald R Crabtree
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA; Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Chris C Goodnow
- School of Medical Sciences, Faculty of Medicine, UNSW, Sydney, NSW, Australia; Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Fabio Luciani
- The Kirby Institute for Infection and Immunity, UNSW, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine, UNSW, Sydney, NSW, Australia.
| | - Ian A Parish
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia; John Curtin School of Medical Research, ANU, Canberra, ACT, Australia.
| |
Collapse
|
2
|
May JF, Kelly RG, Suen AYW, Kim J, Kim J, Anderson CC, Rayat GR, Baldwin TA. Establishment of CD8+ T Cell Thymic Central Tolerance to Tissue-Restricted Antigen Requires PD-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:271-283. [PMID: 37982696 DOI: 10.4049/jimmunol.2200775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/02/2023] [Indexed: 11/21/2023]
Abstract
Highly self-reactive T cells are censored from the repertoire by both central and peripheral tolerance mechanisms upon receipt of high-affinity TCR signals. Clonal deletion is considered a major driver of central tolerance; however, other mechanisms such as induction of regulatory T cells and functional impairment have been described. An understanding of the interplay between these different central tolerance mechanisms is still lacking. We previously showed that impaired clonal deletion to a model tissue-restricted Ag did not compromise tolerance. In this study, we determined that murine T cells that failed clonal deletion were rendered functionally impaired in the thymus. Programmed cell death protein 1 (PD-1) was induced in the thymus and was required to establish cell-intrinsic tolerance to tissue-restricted Ag in CD8+ thymocytes independently of clonal deletion. In bone marrow chimeras, tolerance was not observed in PD-L1-deficient recipients, but tolerance was largely maintained following adoptive transfer of tolerant thymocytes or T cells to PD-L1-deficient recipients. However, CRISPR-mediated ablation of PD-1 in tolerant T cells resulted in broken tolerance, suggesting different PD-1 signaling requirements for establishing versus maintaining tolerance. Finally, we showed that chronic exposure to high-affinity Ag supported the long-term maintenance of tolerance. Taken together, our study identifies a critical role for PD-1 in establishing central tolerance in autoreactive T cells that escape clonal deletion. It also sheds light on potential mechanisms of action of anti-PD-1 pathway immune checkpoint blockade and the development of immune-related adverse events.
Collapse
Affiliation(s)
- Julia F May
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Rees G Kelly
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander Y W Suen
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Jeongbee Kim
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Jeongwoo Kim
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Colin C Anderson
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Gina R Rayat
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Ray Rajotte Surgical-Medical Research Institute, AB Diabetes and Transplant Institutes, University of Alberta, Edmonton, Alberta, Canada
| | - Troy A Baldwin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
Carbone FR, Mackay LK. Functional T cell tolerance by peripheral tissue-based checkpoint control. Nat Immunol 2023; 24:1224-1225. [PMID: 37474656 DOI: 10.1038/s41590-023-01574-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Affiliation(s)
- Francis R Carbone
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia.
| | - Laura K Mackay
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
4
|
Damo M, Hornick NI, Venkat A, William I, Clulo K, Venkatesan S, He J, Fagerberg E, Loza JL, Kwok D, Tal A, Buck J, Cui C, Singh J, Damsky WE, Leventhal JS, Krishnaswamy S, Joshi NS. PD-1 maintains CD8 T cell tolerance towards cutaneous neoantigens. Nature 2023; 619:151-159. [PMID: 37344588 PMCID: PMC10989189 DOI: 10.1038/s41586-023-06217-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 05/12/2023] [Indexed: 06/23/2023]
Abstract
The peripheral T cell repertoire of healthy individuals contains self-reactive T cells1,2. Checkpoint receptors such as PD-1 are thought to enable the induction of peripheral tolerance by deletion or anergy of self-reactive CD8 T cells3-10. However, this model is challenged by the high frequency of immune-related adverse events in patients with cancer who have been treated with checkpoint inhibitors11. Here we developed a mouse model in which skin-specific expression of T cell antigens in the epidermis caused local infiltration of antigen-specific CD8 T cells with an effector gene-expression profile. In this setting, PD-1 enabled the maintenance of skin tolerance by preventing tissue-infiltrating antigen-specific effector CD8 T cells from (1) acquiring a fully functional, pathogenic differentiation state, (2) secreting significant amounts of effector molecules, and (3) gaining access to epidermal antigen-expressing cells. In the absence of PD-1, epidermal antigen-expressing cells were eliminated by antigen-specific CD8 T cells, resulting in local pathology. Transcriptomic analysis of skin biopsies from two patients with cutaneous lichenoid immune-related adverse events showed the presence of clonally expanded effector CD8 T cells in both lesional and non-lesional skin. Thus, our data support a model of peripheral T cell tolerance in which PD-1 allows antigen-specific effector CD8 T cells to co-exist with antigen-expressing cells in tissues without immunopathology.
Collapse
Affiliation(s)
- Martina Damo
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Noah I Hornick
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Aarthi Venkat
- Departments of Genetics and of Computer Science, Yale University School of Medicine, New Haven, CT, USA
| | - Ivana William
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kathryn Clulo
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Srividhya Venkatesan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jiaming He
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Eric Fagerberg
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jennifer L Loza
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Darwin Kwok
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Aya Tal
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jessica Buck
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Can Cui
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jaiveer Singh
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - William E Damsky
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Jonathan S Leventhal
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Smita Krishnaswamy
- Departments of Genetics and of Computer Science, Yale University School of Medicine, New Haven, CT, USA
| | - Nikhil S Joshi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
5
|
Zhao Z, Grégoire C, Oliveira B, Chung K, Melenhorst JJ. Challenges and opportunities of CAR T cell therapies for CLL. Semin Hematol 2023; 60:25-33. [PMID: 37080707 DOI: 10.1053/j.seminhematol.2023.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 02/10/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have transformed the treatment landscape of blood cancers. These engineered receptors which endow T cells with antibody-like target cell recognition combined with the typical T cell target cell lysis abilities. Introduced into the clinic in the 2010s, CAR T-cells have shown efficacy in chronic B lymphocytic leukemia (CLL), but a majority of patients do not achieve sustained remission. Here we discuss the current treatment landscape in CLL using small molecules and allogeneic stem cell transplantation, the niche CAR T-cells filled in this context, and what we have learned from biomarker and mechanistic studies. Several product parameters and improvements are introduced as examples of how the bedside-to-bench is translated into improved CAR T-cells for CLL. We hope to convey to our readers the crucial role translational medicine plays in transforming the treatment outcomes for patients with CLL and how this line of research is an essential component of modern medicine.
Collapse
|
6
|
Shibru B, Fey K, Fricke S, Blaudszun AR, Fürst F, Weise M, Seiffert S, Weyh MK, Köhl U, Sack U, Boldt A. Detection of Immune Checkpoint Receptors - A Current Challenge in Clinical Flow Cytometry. Front Immunol 2021; 12:694055. [PMID: 34276685 PMCID: PMC8281132 DOI: 10.3389/fimmu.2021.694055] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Immunological therapy principles are increasingly determining modern medicine. They are used to treat diseases of the immune system, for tumors, but also for infections, neurological diseases, and many others. Most of these therapies base on antibodies, but small molecules, soluble receptors or cells and modified cells are also used. The development of immune checkpoint inhibitors is amazingly fast. T-cell directed antibody therapies against PD-1 or CTLA-4 are already firmly established in the clinic. Further targets are constantly being added and it is becoming increasingly clear that their expression is not only relevant on T cells. Furthermore, we do not yet have any experience with the long-term systemic effects of the treatment. Flow cytometry can be used for diagnosis, monitoring, and detection of side effects. In this review, we focus on checkpoint molecules as target molecules and functional markers of cells of the innate and acquired immune system. However, for most of the interesting and potentially relevant parameters, there are still no test kits suitable for routine use. Here we give an overview of the detection of checkpoint molecules on immune cells in the peripheral blood and show examples of a possible design of antibody panels.
Collapse
Affiliation(s)
- Benjamin Shibru
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Katharina Fey
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | | | - Friederike Fürst
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Max Weise
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Sabine Seiffert
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Maria Katharina Weyh
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Ulrike Köhl
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Institute for Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Ulrich Sack
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Andreas Boldt
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
7
|
This S, Valbon SF, Lebel MÈ, Melichar HJ. Strength and Numbers: The Role of Affinity and Avidity in the 'Quality' of T Cell Tolerance. Cells 2021; 10:1530. [PMID: 34204485 PMCID: PMC8234061 DOI: 10.3390/cells10061530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 11/17/2022] Open
Abstract
The ability of T cells to identify foreign antigens and mount an efficient immune response while limiting activation upon recognition of self and self-associated peptides is critical. Multiple tolerance mechanisms work in concert to prevent the generation and activation of self-reactive T cells. T cell tolerance is tightly regulated, as defects in these processes can lead to devastating disease; a wide variety of autoimmune diseases and, more recently, adverse immune-related events associated with checkpoint blockade immunotherapy have been linked to a breakdown in T cell tolerance. The quantity and quality of antigen receptor signaling depend on a variety of parameters that include T cell receptor affinity and avidity for peptide. Autoreactive T cell fate choices (e.g., deletion, anergy, regulatory T cell development) are highly dependent on the strength of T cell receptor interactions with self-peptide. However, less is known about how differences in the strength of T cell receptor signaling during differentiation influences the 'function' and persistence of anergic and regulatory T cell populations. Here, we review the literature on this subject and discuss the clinical implications of how T cell receptor signal strength influences the 'quality' of anergic and regulatory T cell populations.
Collapse
Affiliation(s)
- Sébastien This
- Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC H1T 2M4, Canada; (S.T.); (S.F.V.); (M.-È.L.)
- Département de Microbiologie, Immunologie et Infectiologie, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Stefanie F. Valbon
- Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC H1T 2M4, Canada; (S.T.); (S.F.V.); (M.-È.L.)
- Département de Microbiologie, Immunologie et Infectiologie, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Marie-Ève Lebel
- Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC H1T 2M4, Canada; (S.T.); (S.F.V.); (M.-È.L.)
| | - Heather J. Melichar
- Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC H1T 2M4, Canada; (S.T.); (S.F.V.); (M.-È.L.)
- Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Programmed death-1 (PD-1) is an inhibitory receptor that controls T and B cell proliferation and function through interacting with its ligand PD-L1 or PD-L2. PD-1/PD-L1 blockade reboots anti-tumor immunity and is currently used to treat > 15 different types of cancer. However, the response rate is not at 100% and some patients relapse. Importantly, up to 37% of patients treated with PD-1/PD-L1 blocking antibodies develop immune-related adverse events, including overt autoimmunity, such as type 1 diabetes (T1D). Herein, we discuss the role of PD-1, PD-L1, and PD-L2 signaling in pre-clinical models of T1D, including recent work from our laboratory. RECENT FINDINGS We highlight ongoing efforts to harness PD-1/PD-L1 signaling and treat autoimmunity. We also evaluate studies aimed at defining biomarkers that could reliably predict the development of immune-related adverse events after clinical PD-1/PD-L1 blockade. With increasing use of PD-1 blockade in the clinic, onset of autoimmunity is a growing health concern. In this review, we discuss what is known about the role of PD-1 pathway signaling in T1D and comment on ongoing efforts to identify patients at risk of T1D development after PD-1 pathway blockade.
Collapse
Affiliation(s)
- Christopher G Tucker
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, 2101 6th St SE, Wallin Medical Biosciences Building, 3-146, Minneapolis, MN, 55455, USA
| | - Alexander J Dwyer
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, 2101 6th St SE, Wallin Medical Biosciences Building, 3-146, Minneapolis, MN, 55455, USA
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, 2101 6th St SE, Wallin Medical Biosciences Building, 3-146, Minneapolis, MN, 55455, USA.
| | - Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, 2101 6th St SE, Wallin Medical Biosciences Building, 3-146, Minneapolis, MN, 55455, USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Thomas Building, D3-100, Seattle, WA, 98109, USA.
| |
Collapse
|
9
|
Merz V, Gaule M, Zecchetto C, Cavaliere A, Casalino S, Pesoni C, Contarelli S, Sabbadini F, Bertolini M, Mangiameli D, Milella M, Fedele V, Melisi D. Targeting KRAS: The Elephant in the Room of Epithelial Cancers. Front Oncol 2021; 11:638360. [PMID: 33777798 PMCID: PMC7991835 DOI: 10.3389/fonc.2021.638360] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Mutations of the proto-oncogene KRAS are the most frequent gain-of-function alterations found in cancer. KRAS is mutated in about 30% of all human tumors, but it could reach more than 90% in certain cancer types such as pancreatic adenocarcinoma. Although historically considered to be undruggable, a particular KRAS mutation, the G12C variant, has recently emerged as an actionable alteration especially in non-small cell lung cancer (NSCLC). KRASG12C and pan-KRAS inhibitors are being tested in clinical trials and have recently shown promising activity. Due to the difficulties in direct targeting of KRAS, other approaches are being explored. The inhibition of target upstream activators or downstream effectors of KRAS pathway has shown to be moderately effective given the evidence of emerging mechanisms of resistance. Various synthetic lethal partners of KRAS have recently being identified and the inhibition of some of those might prove to be successful in the future. The study of escape mechanisms to KRAS inhibition could support the utility of combination strategies in overcoming intrinsic and adaptive resistance and enhancing clinical benefit of KRASG12C inhibitors. Considering the role of the microenvironment in influencing tumor initiation and promotion, the immune tumor niche of KRAS mutant tumors has been deeply explored and characterized for its unique immunosuppressive skewing. However, a number of aspects remains to be fully understood, and modulating this tumor niche might revert the immunoresistance of KRAS mutant tumors. Synergistic associations of KRASG12C and immune checkpoint inhibitors are being tested.
Collapse
Affiliation(s)
- Valeria Merz
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
- Medical Oncology Unit, Santa Chiara Hospital, Trento, Italy
| | - Marina Gaule
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
- Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| | - Camilla Zecchetto
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
- Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| | - Alessandro Cavaliere
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
- Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| | - Simona Casalino
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
- Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| | - Camilla Pesoni
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
- Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| | - Serena Contarelli
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Fabio Sabbadini
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Monica Bertolini
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Domenico Mangiameli
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Michele Milella
- Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| | - Vita Fedele
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
- Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| |
Collapse
|
10
|
Murray K, Floudas A, Murray C, Fabre A, Crown J, Fearon U, Veale D. First use of tofacitinib to treat an immune checkpoint inhibitor-induced arthritis. BMJ Case Rep 2021; 14:14/2/e238851. [PMID: 33541985 PMCID: PMC7868229 DOI: 10.1136/bcr-2020-238851] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors have revolutionised cancer treatment; however, immune-related adverse events do occur, with up to 7% developing inflammatory arthritis. Common rheumatoid arthritis therapies such as methotrexate, prednisolone and biologics have been used to treat this arthritis in small, uncontrolled case series with varying success. In this case of personalised medicine, we report the first use of tofacitinib, a small molecular inhibitor of the Janus kinase-signal transducer and activator of transcription pathway, to treat checkpoint inhibitor-related inflammatory arthritis. This resulted in a rapid clinical response and complete, sustained remission of the arthritis with associated marked reduction in synovial molecular and cellular immune response.
Collapse
Affiliation(s)
- Kieran Murray
- EULAR Centre for Arthritis and Rheumatic Disease, St. Vincent's University Hospital, Dublin, Ireland
| | - Achilleas Floudas
- EULAR Centre for Arthritis and Rheumatic Disease, St. Vincent's University Hospital, Dublin, Ireland,Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ciara Murray
- Pathology, Oncology and Medicine, University College Dublin, Dublin, Ireland
| | - Aurelie Fabre
- Pathology, Oncology and Medicine, University College Dublin, Dublin, Ireland
| | - John Crown
- Pathology, Oncology and Medicine, University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- EULAR Centre for Arthritis and Rheumatic Disease, St. Vincent's University Hospital, Dublin, Ireland,Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Douglas Veale
- EULAR Centre for Arthritis and Rheumatic Disease, St. Vincent's University Hospital, Dublin, Ireland,Pathology, Oncology and Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
11
|
Wang Y, Lei Y, Gu Y, Kong X, Bian Z, Ji F. Effect of dexmedetomidine on CD4+ T cells and programmed cell death protein-1 in postoperative analgesia: a prospective, randomized, controlled study. Minerva Anestesiol 2021; 87:423-431. [PMID: 33432790 DOI: 10.23736/s0375-9393.20.14581-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Surgical trauma inhibits cellular immunity. Dexmedetomidine produces opioid-sparing effect and an impact on immune response. METHODS Eighty-six surgical patients were enrolled and received postoperative patient-controlled intravenous analgesia (PCIA) with either fentanyl alone (fentanyl group) or combined with dexmedetomidine (dexmedetomidine group). The percentages of T helper cells (Th1, Th2, and Th17) and regulatory T (Treg) cells, expression levels of programmed cell death protein-1 (PD-1) and its ligand (PD-L1) on the CD4+ T cells, and plasma levels of the cytokines were tested. Postoperative pain was measured by numerical rating scale (NRS), including NRS at rest (NRSR) and movement (NRSM). RESULTS In dexmedetomidine group, Th1 cells were increased significantly at 24 and 48 h following surgery (P=0.011 and P=0.013, respectively) and Treg cells were significantly higher at 48 h postoperatively (P=0.013). PD-1 was significantly lower in dexmedetomidine group at 24 h postoperatively (P=0.046) and interleukin 4 (IL-4) and IL-6 were significantly decreased at 48 h postoperatively (P=0.024 and P=0.035, respectively). Compared with fentanyl group, NRSR scores were lower in dexmedetomidine group at 24 h following surgery (P=0.018) and NRSR and NRSM scores were lower at 48 h postoperatively (P=0.007 and P=0.011, respectively). NRSR exhibited negative correlations with Th1 cells in fentanyl group and dexmedetomidine group (P=0.003 and P=0.005, respectively). CONCLUSIONS Dexmedetomidine increases the differentiation of Th1 and Treg cells and reduces the expression of PD-1 on CD4+ T cells. Dexmedetomidine may assist to ameliorate postoperative pain and attenuate proinflammatory response. There might be a negative correlation between pain and Th1 cells.
Collapse
Affiliation(s)
- Yulan Wang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yishan Lei
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanzheng Gu
- Clinical Immunology Institute of Jiangsu Province, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoqi Kong
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhen Bian
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fuhai Ji
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, China -
| |
Collapse
|
12
|
Canavan M, Floudas A, Veale DJ, Fearon U. The PD-1:PD-L1 axis in Inflammatory Arthritis. BMC Rheumatol 2021; 5:1. [PMID: 33423684 PMCID: PMC7798255 DOI: 10.1186/s41927-020-00171-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022] Open
Abstract
The activation of antigen specific T cells during an immune response is a tightly regulated process at the level of both costimulatory and coinhibitory receptors. One such coinhibitory receptor or checkpoint inhibitor which has received much attention in the field of oncology is the programmed cell death protein 1 (PD-1). Blockade of PD-1 or its ligand PD-L1 has proven successful in the treatment of a wide variety of cancers, therefore highlighting an important role for this pathway in anti-tumour immune responses. However, a caveat of PD-1 therapy and boosting anti-tumour immune responses is the development of self-reactive T cells which can lead to the induction of various autoimmune or inflammatory diseases, referred to as immune- related adverse events (irAEs). The emergence of rheumatological irAEs such as Inflammatory Arthritis (IA) in recent years has highlighted the importance of PD-1 in maintaining self-tolerance. Furthermore, the emergence of rheumatology related irAEs raises an important question as to how defects in this pathway can contribute to spontaneous rheumatological disease. In this review, we describe the biological distribution, function and regulation of the PD-1 pathway, its potential role in IA and irAE related IA.
Collapse
Affiliation(s)
- Mary Canavan
- Department of Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
- EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland.
| | - Achilleas Floudas
- Department of Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Douglas J Veale
- EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Department of Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| |
Collapse
|
13
|
Ebelt ND, Zuniga E, Marzagalli M, Zamloot V, Blazar BR, Salgia R, Manuel ER. Salmonella-Based Therapy Targeting Indoleamine 2,3-Dioxygenase Restructures the Immune Contexture to Improve Checkpoint Blockade Efficacy. Biomedicines 2020; 8:E617. [PMID: 33339195 PMCID: PMC7765568 DOI: 10.3390/biomedicines8120617] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/09/2020] [Accepted: 12/13/2020] [Indexed: 12/15/2022] Open
Abstract
Therapeutic options for non-small cell lung cancer (NSCLC) treatment have changed dramatically in recent years with the advent of novel immunotherapeutic approaches. Among these, immune checkpoint blockade (ICB) using monoclonal antibodies has shown tremendous promise in approximately 20% of patients. In order to better predict patients that will respond to ICB treatment, biomarkers such as tumor-associated CD8+ T cell frequency, tumor checkpoint protein status and mutational burden have been utilized, however, with mixed success. In this study, we hypothesized that significantly altering the suppressive tumor immune landscape in NSCLC could potentially improve ICB efficacy. Using sub-therapeutic doses of our Salmonella typhimurium-based therapy targeting the suppressive molecule indoleamine 2,3-dioxygenase (shIDO-ST) in tumor-bearing mice, we observed dramatic changes in immune subset phenotypes that included increases in antigen presentation markers, decreased regulatory T cell frequency and overall reduced checkpoint protein expression. Combination shIDO-ST treatment with anti-PD-1/CTLA-4 antibodies enhanced tumor growth control, compared to either treatment alone, which was associated with significant intratumoral infiltration by CD8+ and CD4+ T cells. Ultimately, we show that increases in antigen presentation markers and infiltration by T cells is correlated with significantly increased survival in NSCLC patients. These results suggest that the success of ICB therapy may be more accurately predicted by taking into account multiple factors such as potential for antigen presentation and immune subset repertoire in addition to markers already being considered. Alternatively, combination treatment with agents such as shIDO-ST could be used to create a more conducive tumor microenvironment for improving responses to ICB.
Collapse
Affiliation(s)
- Nancy D. Ebelt
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| | - Edith Zuniga
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| | - Monica Marzagalli
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| | - Vic Zamloot
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood and Bone Marrow Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Edwin R. Manuel
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| |
Collapse
|
14
|
Cytocidal macrophages in symbiosis with CD4 and CD8 T cells cause acute diabetes following checkpoint blockade of PD-1 in NOD mice. Proc Natl Acad Sci U S A 2020; 117:31319-31330. [PMID: 33229539 DOI: 10.1073/pnas.2019743117] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Autoimmune diabetes is one of the complications resulting from checkpoint blockade immunotherapy in cancer patients, yet the underlying mechanisms for such an adverse effect are not well understood. Leveraging the diabetes-susceptible nonobese diabetic (NOD) mouse model, we phenocopy the diabetes progression induced by programmed death 1 (PD-1)/PD-L1 blockade and identify a cascade of highly interdependent cellular interactions involving diabetogenic CD4 and CD8 T cells and macrophages. We demonstrate that exhausted CD8 T cells are the major cells that respond to PD-1 blockade producing high levels of IFN-γ. Most importantly, the activated T cells lead to the recruitment of monocyte-derived macrophages that become highly activated when responding to IFN-γ. These macrophages acquire cytocidal activity against β-cells via nitric oxide and induce autoimmune diabetes. Collectively, the data in this study reveal a critical role of macrophages in the PD-1 blockade-induced diabetogenesis, providing new insights for the understanding of checkpoint blockade immunotherapy in cancer and infectious diseases.
Collapse
|
15
|
Nüssing S, Trapani JA, Parish IA. Revisiting T Cell Tolerance as a Checkpoint Target for Cancer Immunotherapy. Front Immunol 2020; 11:589641. [PMID: 33072137 PMCID: PMC7538772 DOI: 10.3389/fimmu.2020.589641] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/04/2020] [Indexed: 12/30/2022] Open
Abstract
Immunotherapy has revolutionized the treatment of cancer. Nevertheless, the majority of patients do not respond to therapy, meaning a deeper understanding of tumor immune evasion strategies is required to boost treatment efficacy. The vast majority of immunotherapy studies have focused on how treatment reinvigorates exhausted CD8+ T cells within the tumor. In contrast, how therapies influence regulatory processes within the draining lymph node is less well studied. In particular, relatively little has been done to examine how tumors may exploit peripheral CD8+ T cell tolerance, an under-studied immune checkpoint that under normal circumstances prevents detrimental autoimmune disease by blocking the initiation of T cell responses. Here we review the therapeutic potential of blocking peripheral CD8+ T cell tolerance for the treatment of cancer. We first comprehensively review what has been learnt about the regulation of CD8+ T cell peripheral tolerance from the non-tumor models in which peripheral tolerance was first defined. We next consider how the tolerant state differs from other states of negative regulation, such as T cell exhaustion and senescence. Finally, we describe how tumors hijack the peripheral tolerance immune checkpoint to prevent anti-tumor immune responses, and argue that disruption of peripheral tolerance may contribute to both the anti-cancer efficacy and autoimmune side-effects of immunotherapy. Overall, we propose that a deeper understanding of peripheral tolerance will ultimately enable the development of more targeted and refined cancer immunotherapy approaches.
Collapse
Affiliation(s)
- Simone Nüssing
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Joseph A Trapani
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Ian A Parish
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
16
|
Robust Iterative Stimulation with Self-Antigens Overcomes CD8 + T Cell Tolerance to Self- and Tumor Antigens. Cell Rep 2020; 28:3092-3104.e5. [PMID: 31533033 PMCID: PMC6874401 DOI: 10.1016/j.celrep.2019.08.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/24/2019] [Accepted: 08/09/2019] [Indexed: 12/16/2022] Open
Abstract
The immune system adapts to constitutive antigens to preserve self-tolerance, which is a major barrier for anti-tumor immunity. Antigen-specific reversal of tolerance constitutes a major goal to spur therapeutic applications. Here, we show that robust, iterative, systemic stimulation targeting tissue-specific antigens in the context of acute infections reverses established CD8+ T cell tolerance to self, including in T cells that survive negative selection. This strategy results in large numbers of circulating and resident memory self-specific CD8+ T cells that are widely distributed and can be co-opted to control established malignancies bearing self-antigen without concomitant autoimmunity. Targeted expansion of both self- and tumor neoantigen-specific T cells acts synergistically to boost anti-tumor immunity and elicits protection against aggressive melanoma. Our findings demonstrate that T cell tolerance can be re-adapted to responsiveness through robust antigenic exposure, generating self-specific CD8+ T cells that can be used for cancer treatment.
Collapse
|
17
|
Symonds AL, Zheng W, Miao T, Wang H, Wang T, Kiome R, Hou X, Li S, Wang P. Egr2 and 3 control inflammation, but maintain homeostasis, of PD-1 high memory phenotype CD4 T cells. Life Sci Alliance 2020; 3:3/9/e202000766. [PMID: 32709717 PMCID: PMC7391068 DOI: 10.26508/lsa.202000766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 01/13/2023] Open
Abstract
PD-1high memory CD4 T cells are pathogenic in autoimmune disease; here they show their expression of Egr2 is defective in rheumatoid arthritis and Egr2 & 3 control their inflammation and homeostasis. The transcription factors Egr2 and 3 are essential for controlling inflammatory autoimmune responses of memory phenotype (MP) CD4 T cells. However, the mechanism is still unclear. We have now found that the Egr2+ subset (PD-1high MP) of MP CD4 T cells expresses high levels of checkpoint molecules (PD-1 and Lag3) and also markers of effector T cells (CXCR3 and ICAM-1). Egr2/3 are not required for PD-1high MP CD4 cell development but mediate a unique transcriptional programme that effectively controls their inflammatory responses, while promoting homeostatic proliferation and adaptive responses. Egr2 negative PD-1high MP CD4 T cells are impaired in homeostatic proliferation and adaptive responses against viral infection but display inflammatory responses to innate stimulation such as IL-12. PD-1high MP CD4 T cells have recently been implicated in rheumatoid arthritis pathogenesis, and we have now found that Egr2 expression is reduced in PD-1high MP CD4 T cells from patients with active rheumatoid arthritis compared with healthy controls. These findings demonstrate that Egr2/3 control the inflammatory responses of PD-1high MP CD4 T cells and maintain their adaptive immune fitness.
Collapse
Affiliation(s)
- Alistair Lj Symonds
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Wei Zheng
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tizong Miao
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Haiyu Wang
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - TieShang Wang
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ruth Kiome
- Bioscience, Brunel University, Uxbridge, UK
| | - Xiujuan Hou
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Suling Li
- Bioscience, Brunel University, Uxbridge, UK
| | - Ping Wang
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
18
|
Martinov T, Fife BT. Type 1 diabetes pathogenesis and the role of inhibitory receptors in islet tolerance. Ann N Y Acad Sci 2020; 1461:73-103. [PMID: 31025378 PMCID: PMC6994200 DOI: 10.1111/nyas.14106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/25/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes (T1D) affects over a million Americans, and disease incidence is on the rise. Despite decades of research, there is still no cure for this disease. Exciting beta cell replacement strategies are being developed, but in order for such approaches to work, targeted immunotherapies must be designed. To selectively halt the autoimmune response, researchers must first understand how this response is regulated and which tolerance checkpoints fail during T1D development. Herein, we discuss the current understanding of T1D pathogenesis in humans, genetic and environmental risk factors, presumed roles of CD4+ and CD8+ T cells as well as B cells, and implicated autoantigens. We also highlight studies in non-obese diabetic mice that have demonstrated the requirement for CD4+ and CD8+ T cells and B cells in driving T1D pathology. We present an overview of central and peripheral tolerance mechanisms and comment on existing controversies in the field regarding central tolerance. Finally, we discuss T cell- and B cell-intrinsic tolerance mechanisms, with an emphasis on the roles of inhibitory receptors in maintaining islet tolerance in humans and in diabetes-prone mice, and strategies employed to date to harness inhibitory receptor signaling to prevent or reverse T1D.
Collapse
Affiliation(s)
- Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
19
|
Falcone M, Fousteri G. Role of the PD-1/PD-L1 Dyad in the Maintenance of Pancreatic Immune Tolerance for Prevention of Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:569. [PMID: 32973682 PMCID: PMC7466754 DOI: 10.3389/fendo.2020.00569] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
The human pancreas, like almost all organs in the human body, is immunologically tolerated despite the presence of innate and adaptive immune cells that promptly mediate protective immune responses against pathogens in situ. The PD-1/PD-L1 inhibitory pathway seems to play a key role in the maintenance of immune tolerance systemically and within the pancreatic tissue. Tissue resident memory T cells (TRM), T regulatory cells (Treg), macrophages and even β cells exhibit PD-1 or PD-L1 expression that contributes in controlling pancreatic immune homeostasis and tolerance. Dysregulation of the PD-1/PD-L1 axis as shown by animal studies and our recent experience with checkpoint inhibitory blockade in humans can lead to immune dysfunctions leading to chronic inflammatory disease and to type 1 diabetes (T1D) in genetically susceptible individuals. In this review, we discuss the role of the PD-1/PD-L1 axis in pancreatic tissue homeostasis and tolerance, speculate how genetic and environmental factors can regulate the PD-1/PD-L1 pathway, and discuss PD-1/PD-L1-based therapeutic approaches for pancreatic islet transplantation and T1D treatment.
Collapse
|
20
|
Fiege JK, Stone IA, Dumm RE, Waring BM, Fife BT, Agudo J, Brown BD, Heaton NS, Langlois RA. Long-term surviving influenza infected cells evade CD8+ T cell mediated clearance. PLoS Pathog 2019; 15:e1008077. [PMID: 31557273 PMCID: PMC6782110 DOI: 10.1371/journal.ppat.1008077] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 10/08/2019] [Accepted: 09/11/2019] [Indexed: 01/02/2023] Open
Abstract
Influenza A virus (IAV) is a seasonal pathogen with the potential to cause devastating pandemics. IAV infects multiple epithelial cell subsets in the respiratory tract, eliciting damage to the lungs. Clearance of IAV is primarily dependent on CD8+ T cells, which must balance control of the infection with immunopathology. Using a virus expressing Cre recombinase to permanently label infected cells in a Cre-inducible reporter mouse, we previously discovered infected club cells that survive both lytic virus replication and CD8+ T cell-mediated clearance. In this study, we demonstrate that ciliated epithelial cells, type I and type II alveolar cells can also become survivor cells. Survivor cells are stable in the lung long-term and demonstrate enhanced proliferation compared to uninfected cells. When we investigated how survivor cells evade CD8+ T cell killing we observed that survivor cells upregulated the inhibitory ligand PD-L1, but survivor cells did not use PD-L1 to evade CD8+ T cell killing. Instead our data suggest that survivor cells are not inherently resistant to CD8+ T cell killing, but instead no longer present IAV antigen and cannot be detected by CD8+ T cells. Finally, we evaluate the failure of CD8+ T cells to kill these previously infected cells. This work demonstrates that additional cell types can survive IAV infection and that these cells robustly proliferate and are stable long term. By sparing previously infected cells, the adaptive immune system may be minimizing pathology associated with IAV infection.
Collapse
Affiliation(s)
- Jessica K. Fiege
- University of Minnesota, Department of Microbiology and Immunology and the Center for Immunology, Minneapolis, Minnesota, United States of America
| | - Ian A. Stone
- University of Minnesota, Department of Microbiology and Immunology and the Center for Immunology, Minneapolis, Minnesota, United States of America
| | - Rebekah E. Dumm
- Duke University School of Medicine, Department of Molecular Genetics and Microbiology, Durham, North Carolina, United States of America
| | - Barbara M. Waring
- University of Minnesota, Department of Microbiology and Immunology and the Center for Immunology, Minneapolis, Minnesota, United States of America
| | - Brian T. Fife
- University of Minnesota, Department of Medicine and the Center for Immunology, Minneapolis, Minnesota, United States of America
| | - Judith Agudo
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York City, New York, United States of America
| | - Brian D. Brown
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York City, New York, United States of America
| | - Nicholas S. Heaton
- Duke University School of Medicine, Department of Molecular Genetics and Microbiology, Durham, North Carolina, United States of America
| | - Ryan A. Langlois
- University of Minnesota, Department of Microbiology and Immunology and the Center for Immunology, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
21
|
Martinov T, Swanson LA, Breed ER, Tucker CG, Dwyer AJ, Johnson JK, Mitchell JS, Sahli NL, Wilson JC, Singh LM, Hogquist KA, Spanier JA, Fife BT. Programmed Death-1 Restrains the Germinal Center in Type 1 Diabetes. THE JOURNAL OF IMMUNOLOGY 2019; 203:844-852. [PMID: 31324724 DOI: 10.4049/jimmunol.1801535] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 06/18/2019] [Indexed: 01/22/2023]
Abstract
Programmed death-1 (PD-1) inhibits T and B cell function upon ligand binding. PD-1 blockade revolutionized cancer treatment, and although numerous patients respond, some develop autoimmune-like symptoms or overt autoimmunity characterized by autoantibody production. PD-1 inhibition accelerates autoimmunity in mice, but its role in regulating germinal centers (GC) is controversial. To address the role of PD-1 in the GC reaction in type 1 diabetes, we used tetramers to phenotype insulin-specific CD4+ T and B cells in NOD mice. PD-1 or PD-L1 deficiency, and PD-1 but not PD-L2 blockade, unleashed insulin-specific T follicular helper CD4+ T cells and enhanced their survival. This was concomitant with an increase in GC B cells and augmented insulin autoantibody production. The effect of PD-1 blockade on the GC was reduced when mice were treated with a mAb targeting the insulin peptide:MHC class II complex. This work provides an explanation for autoimmune side effects following PD-1 pathway inhibition and suggests that targeting the self-peptide:MHC class II complex might limit autoimmunity arising from checkpoint blockade.
Collapse
Affiliation(s)
- Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Linnea A Swanson
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Elise R Breed
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Christopher G Tucker
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Alexander J Dwyer
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Jenna K Johnson
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Jason S Mitchell
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Nathanael L Sahli
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Joseph C Wilson
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Lovejot M Singh
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Kristin A Hogquist
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Justin A Spanier
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| |
Collapse
|
22
|
Shergold AL, Millar R, Nibbs RJ. Understanding and overcoming the resistance of cancer to PD-1/PD-L1 blockade. Pharmacol Res 2019; 145:104258. [DOI: 10.1016/j.phrs.2019.104258] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/01/2019] [Accepted: 05/01/2019] [Indexed: 12/22/2022]
|
23
|
Dirice E, Kahraman S, De Jesus DF, El Ouaamari A, Basile G, Baker RL, Yigit B, Piehowski PD, Kim MJ, Dwyer AJ, Ng RWS, Schuster C, Vethe H, Martinov T, Ishikawa Y, Teo AKK, Smith RD, Hu J, Haskins K, Serwold T, Qian WJ, Fife BT, Kissler S, Kulkarni RN. Increased β-cell proliferation before immune cell invasion prevents progression of type 1 diabetes. Nat Metab 2019; 1:509-518. [PMID: 31423480 PMCID: PMC6696912 DOI: 10.1038/s42255-019-0061-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) is characterized by pancreatic islet infiltration by autoreactive immune cells and a near-total loss of β-cells1. Restoration of insulin-producing β-cells coupled with immunomodulation to suppress the autoimmune attack has emerged as a potential approach to counter T1D2-4. Here we report that enhancing β-cell mass early in life, in two models of female NOD mice, results in immunomodulation of T-cells, reduced islet infiltration and lower β-cell apoptosis, that together protect them from developing T1D. The animals displayed altered β-cell antigens, and islet transplantation studies showed prolonged graft survival in the NOD-LIRKO model. Adoptive transfer of splenocytes from the NOD-LIRKOs prevented development of diabetes in pre-diabetic NOD mice. A significant increase in the splenic CD4+CD25+FoxP3+ regulatory T-cell (Treg) population was observed to underlie the protected phenotype since Treg depletion rendered NOD-LIRKO mice diabetic. The increase in Tregs coupled with activation of TGF-β/SMAD3 signaling pathway in pathogenic T-cells favored reduced ability to kill β-cells. These data support a previously unidentified observation that initiating β-cell proliferation, alone, prior to islet infiltration by immune cells alters the identity of β-cells, decreases pathologic self-reactivity of effector cells and increases Tregs to prevent progression of T1D.
Collapse
Affiliation(s)
- Ercument Dirice
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Sevim Kahraman
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Dario F. De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
- Graduate Program in Areas of Basic and Applied Biology
(GABBA), Abel Salazar Biomedical Sciences Institute, University of Porto, Porto,
Portugal
| | - Abdelfattah El Ouaamari
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Giorgio Basile
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Rocky L. Baker
- Department of Immunology, School of Medicine, University of
Colorado, Aurora, CO, USA
| | - Burcu Yigit
- Division of Immunology, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, MA, USA
| | - Paul D. Piehowski
- Biological Sciences Division, Pacific Northwest National
Laboratory, Richland, WA, USA
| | - Mi-Jeong Kim
- Section for Immunobiology, Joslin Diabetes Center, Boston,
MA, USA
| | - Alexander J. Dwyer
- University of Minnesota, Center for Immunology, Department
of Medicine, Minneapolis, MN, USA
| | - Raymond W. S. Ng
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Heidrun Vethe
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
| | - Tijana Martinov
- University of Minnesota, Center for Immunology, Department
of Medicine, Minneapolis, MN, USA
| | - Yuki Ishikawa
- Section for Immunobiology, Joslin Diabetes Center, Boston,
MA, USA
| | - Adrian Kee Keong Teo
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard D. Smith
- Biological Sciences Division, Pacific Northwest National
Laboratory, Richland, WA, USA
| | - Jiang Hu
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn Haskins
- Department of Immunology, School of Medicine, University of
Colorado, Aurora, CO, USA
| | - Thomas Serwold
- Section for Immunobiology, Joslin Diabetes Center, Boston,
MA, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National
Laboratory, Richland, WA, USA
| | - Brian T. Fife
- University of Minnesota, Center for Immunology, Department
of Medicine, Minneapolis, MN, USA
| | - Stephan Kissler
- Section for Immunobiology, Joslin Diabetes Center, Boston,
MA, USA
| | - Rohit N. Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Boston, MA, USA
| |
Collapse
|
24
|
Thompson EA, Mitchell JS, Beura LK, Torres DJ, Mrass P, Pierson MJ, Cannon JL, Masopust D, Fife BT, Vezys V. Interstitial Migration of CD8αβ T Cells in the Small Intestine Is Dynamic and Is Dictated by Environmental Cues. Cell Rep 2019; 26:2859-2867.e4. [PMID: 30865878 PMCID: PMC6754515 DOI: 10.1016/j.celrep.2019.02.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 08/05/2018] [Accepted: 02/08/2019] [Indexed: 02/08/2023] Open
Abstract
The migratory capacity of adaptive CD8αβ T cells dictates their ability to locate target cells and exert cytotoxicity, which is the basis of immune surveillance for the containment of microbes and disease. The small intestine (SI) is the largest mucosal surface and is a primary site of pathogen entrance. Using two-photon laser scanning microscopy, we found that motility of antigen (Ag)-specific CD8αβ T cells in the SI is dynamic and varies with the environmental milieu. Pathogen-specific CD8αβ T cell movement differed throughout infection, becoming locally confined at memory. Motility was not dependent on CD103 but was influenced by micro-anatomical locations within the SI and by inflammation. CD8 T cells responding to self-protein were initially affected by the presence of self-Ag, but this was altered after complete tolerance induction. These studies identify multiple factors that affect CD8αβ T cell movement in the intestinal mucosa and show the adaptability of CD8αβ T cell motility.
Collapse
Affiliation(s)
- Emily A Thompson
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jason S Mitchell
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lalit K Beura
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - David J Torres
- Department of Mathematics and Physical Science, Northern New Mexico College, Espanola, NM 87532, USA
| | - Paulus Mrass
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Mark J Pierson
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Judy L Cannon
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131, USA
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Brian T Fife
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
25
|
Kong BY, Bolton H, Kim JW, Silveira PA, Fromm PD, Clark GJ. On the Other Side: Manipulating the Immune Checkpoint Landscape of Dendritic Cells to Enhance Cancer Immunotherapy. Front Oncol 2019; 9:50. [PMID: 30788290 PMCID: PMC6372550 DOI: 10.3389/fonc.2019.00050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/17/2019] [Indexed: 12/26/2022] Open
Abstract
Monoclonal antibodies targeting co-inhibitory immune checkpoint molecules have been successful in clinical trials of both solid and hematological malignancies as acknowledged by the 2018 Nobel Prize in Medicine, however improving clinical response rates is now key to expanding their efficacy in areas of unmet medical need. Antibodies to checkpoint inhibitors target molecules on either T cells or tumor cells to stimulate T cells or remove tumor mediated immunosuppression, respectively. However, many of the well-characterized T cell immune checkpoint receptors have their ligands on antigen presenting cells or exert direct effects on those cells. Dendritic cells are the most powerful antigen presenting cells; they possess the ability to elicit antigen-specific responses and have important roles in regulation of immune tolerance. Despite their theoretical benefits in cancer immunotherapy, the translation of DC therapies into the clinic is yet to be fully realized and combining DC-based immunotherapy with immune checkpoint inhibitors is an attractive strategy. This combination takes advantage of the antigen presenting capability of DC to maximize specific immune responses to tumor antigens whilst removing tumor-associated immune inhibitory mechanisms with immune checkpoint inhibition. Here we review the expression and functional effects of immune checkpoint molecules on DC and identify rational combinations for DC vaccination to enhance antigen-specific T cell responses, cytokine production, and promotion of long-lasting immunological memory.
Collapse
Affiliation(s)
- Benjamin Y Kong
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia.,Department of Medical Oncology, Concord Repatriation General Hospital, Concord, NSW, Australia
| | - Holly Bolton
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Julius W Kim
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Pablo A Silveira
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Phillip D Fromm
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Georgina J Clark
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
26
|
Reprogramming responsiveness to checkpoint blockade in dysfunctional CD8 T cells. Proc Natl Acad Sci U S A 2019; 116:2640-2645. [PMID: 30679280 DOI: 10.1073/pnas.1810326116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Established T cell dysfunction is a barrier to antitumor responses, and checkpoint blockade presumably reverses this. Many patients fail to respond to treatment and/or develop autoimmune adverse events. The underlying reason for T cell responsiveness remains elusive. Here, we show that susceptibility to checkpoint blockade is dependent on the activation status of T cells. Newly activated self-specific CD8 T cells respond to checkpoint blockade and cause autoimmunity, which is mitigated by inhibiting the mechanistic target of rapamycin. However, once tolerance is established, self-specific CD8 T cells display a gene signature comparable to tumor-specific CD8 T cells in a fixed state of dysfunction. Tolerant self-specific CD8 T cells do not respond to single or combinatorial dosing of anti-CTLA4, anti-PD-L1, anti-PD-1, anti-LAG-3, and/or anti-TIM-3. Despite this, T cell responsiveness can be induced by vaccination with cognate antigen, which alters the previously fixed transcriptional signature and increases antigen-sensing machinery. Antigenic reeducation of tolerant T cells synergizes with checkpoint blockade to generate functional CD8 T cells, which eliminate tumors without concomitant autoimmunity and are transcriptionally distinct from classic effector T cells. These data demonstrate that responses to checkpoint blockade are dependent on the activation state of a T cell and show that checkpoint blockade-insensitive CD8 T cells can be induced to respond to checkpoint blockade with robust antigenic stimulation to participate in tumor control.
Collapse
|
27
|
Tipping the balance: inhibitory checkpoints in intestinal homeostasis. Mucosal Immunol 2019; 12:21-35. [PMID: 30498201 DOI: 10.1038/s41385-018-0113-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/28/2018] [Accepted: 11/07/2018] [Indexed: 02/04/2023]
Abstract
The small intestinal and colonic lamina propria are populated with forkhead box P3 (FOXP3)+CD4+ regulatory T cells (Tregs) and interleukin-10-producing T cells that orchestrate intestinal tolerance to harmless microbial and food antigens. Expression of co-inhibitory receptors such as CTLA-4 and PD-1 serve as checkpoints to these cells controlling their T-cell receptor (TCR)-mediated and CD28-mediated activation and modulating the phenotype of neighboring antigen presenting cells. Recent discoveries on the diversity of co-inhibitory receptors and their selective cellular expression has shed new light on their tissue-dependent function. In this review, we provide an overview of the co-inhibitory pathways and checkpoints of Treg and effector T cells and their mechanisms of action in intestinal homeostasis. Better understanding of these inhibitory checkpoints is desired as their blockade harbors clinical potential for the treatment of cancer and their stimulation may offer new opportunities to treat chronic intestinal inflammation such as inflammatory bowel disease.
Collapse
|
28
|
Lewinsky H, Barak AF, Huber V, Kramer MP, Radomir L, Sever L, Orr I, Mirkin V, Dezorella N, Shapiro M, Cohen Y, Shvidel L, Seiffert M, Herishanu Y, Becker-Herman S, Shachar I. CD84 regulates PD-1/PD-L1 expression and function in chronic lymphocytic leukemia. J Clin Invest 2018; 128:5465-5478. [PMID: 30277471 DOI: 10.1172/jci96610] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/26/2018] [Indexed: 12/21/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by clonal proliferation and progressive accumulation of mature B lymphocytes in the peripheral blood, lymphoid tissues, and bone marrow. CLL is characterized by profound immune defects leading to severe infectious complications. T cells are numerically, phenotypically, and functionally highly abnormal in CLL, with only limited ability to exert antitumor immune responses. Exhaustion of T cells has also been suggested to play an important role in antitumor responses. CLL-mediated T cell exhaustion is achieved by the aberrant expression of several inhibitory molecules on CLL cells and their microenvironment, prominently the programmed cell death ligand 1/programmed cell death 1 (PD-L1/PD-1) receptors. Previously, we showed that CD84, a member of the SLAM family of receptors, bridges between CLL cells and their microenvironment. In the current study, we followed CD84 regulation of T cell function. We showed that cell-cell interaction mediated through human and mouse CD84 upregulates PD-L1 expression on CLL cells and in their microenvironment and PD-1 expression on T cells. This resulted in suppression of T cell responses and activity in vitro and in vivo. Thus, our results demonstrate a role for CD84 in the regulation of immune checkpoints by leukemia cells and identify CD84 blockade as a therapeutic strategy to reverse tumor-induced immune suppression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Irit Orr
- Life Sciences Core Facilities, Department of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Vita Mirkin
- Hematology Institute, Kaplan Medical Center, Rehovot, Israel
| | - Nili Dezorella
- Department of Hematology, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Mika Shapiro
- Department of Hematology, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yosef Cohen
- Sanz Medical Center, Laniado Medical Center, Netanya, Israel
| | - Lev Shvidel
- Hematology Institute, Kaplan Medical Center, Rehovot, Israel
| | - Martina Seiffert
- Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yair Herishanu
- Hematology Institute, Kaplan Medical Center, Rehovot, Israel
| | | | | |
Collapse
|
29
|
Cullis J, Das S, Bar-Sagi D. Kras and Tumor Immunity: Friend or Foe? Cold Spring Harb Perspect Med 2018; 8:cshperspect.a031849. [PMID: 29229670 DOI: 10.1101/cshperspect.a031849] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With the recent breakthroughs in immunotherapy as curative treatments in certain tumor types, there has been renewed interest in the relationship between immunity and tumor growth. Although we are gaining a greater understanding of the complex interplay of immune modulating components in the tumor microenvironment, the specific role that tumor cells play in shaping the immune milieu is still not well characterized. In this review, we focus on how mutant Kras tumor cells contribute to tumor immunity, with a specific focus on processes induced directly or indirectly by the oncogene.
Collapse
Affiliation(s)
- Jane Cullis
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016
| | - Shipra Das
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
30
|
Osum KC, Burrack AL, Martinov T, Sahli NL, Mitchell JS, Tucker CG, Pauken KE, Papas K, Appakalai B, Spanier JA, Fife BT. Interferon-gamma drives programmed death-ligand 1 expression on islet β cells to limit T cell function during autoimmune diabetes. Sci Rep 2018; 8:8295. [PMID: 29844327 PMCID: PMC5974126 DOI: 10.1038/s41598-018-26471-9] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes is caused by autoreactive T cell-mediated β cell destruction. Even though co-inhibitory receptor programmed death-1 (PD-1) restrains autoimmunity, the expression and regulation of its cognate ligands on β cell remains unknown. Here, we interrogated β cell-intrinsic programmed death ligand-1 (PD-L1) expression in mouse and human islets. We measured a significant increase in the level of PD-L1 surface expression and the frequency of PD-L1+ β cells as non-obese diabetic (NOD) mice aged and developed diabetes. Increased β cell PD-L1 expression was dependent on T cell infiltration, as β cells from Rag1-deficient mice lacked PD-L1. Using Rag1-deficient NOD mouse islets, we determined that IFN-γ promotes β cell PD-L1 expression. We performed analogous experiments using human samples, and found a significant increase in β cell PD-L1 expression in type 1 diabetic samples compared to type 2 diabetic, autoantibody positive, and non-diabetic samples. Among type 1 diabetic samples, β cell PD-L1 expression correlated with insulitis. In vitro experiments with human islets from non-diabetic individuals showed that IFN-γ promoted β cell PD-L1 expression. These results suggest that insulin-producing β cells respond to pancreatic inflammation and IFN-γ production by upregulating PD-L1 expression to limit self-reactive T cells.
Collapse
Affiliation(s)
- Kevin C Osum
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Adam L Burrack
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Nathanael L Sahli
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Jason S Mitchell
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Christopher G Tucker
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Kristen E Pauken
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Klearchos Papas
- Department of Surgery, University of Arizona, Tucson, AZ, USA
| | | | - Justin A Spanier
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
31
|
Liu QZ, Ma WT, Yang JB, Zhao ZB, Yan K, Yao Y, Li L, Miao Q, Gershwin ME, Lian ZX. The CXC Chemokine Receptor 3 Inhibits Autoimmune Cholangitis via CD8 + T Cells but Promotes Colitis via CD4 + T Cells. Front Immunol 2018; 9:1090. [PMID: 29868034 PMCID: PMC5966573 DOI: 10.3389/fimmu.2018.01090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/01/2018] [Indexed: 11/13/2022] Open
Abstract
CXC chemokine receptor 3 (CXCR3), a receptor for the C-X-C motif chemokines (CXCL) CXCL9, CXCL10, and CXCL11, which not only plays a role in chemotaxis but also regulates differentiation and development of memory and effector T cell populations. Herein, we explored the function of CXCR3 in the modulation of different organ-specific autoimmune diseases in interleukin (IL)-2 receptor deficiency (CD25-/-) mice, a murine model for both cholangitis and colitis. We observed higher levels of CXCL9 and CXCL10 in the liver and colon and higher expression of CXCR3 on T cells of the CD25-/- mice compared with control animals. Deletion of CXCR3 resulted in enhanced liver inflammation but alleviated colitis. These changes in liver and colon pathology after CXCR3 deletion were associated with increased numbers of hepatic CD4+ and CD8+ T cells, in particular effector memory CD8+ T cells, as well as decreased T cells in mesenteric lymph nodes and colon lamina propria. In addition, increased interferon-γ response and decreased IL-17A response was observed in both liver and colon after CXCR3 deletion. CXCR3 modulated the functions of T cells involved in different autoimmune diseases, whereas the consequence of such modulation was organ-specific regarding to their effects on disease severity. Our findings emphasize the importance of extra caution in immunotherapy for organ-specific autoimmune diseases, as therapeutic interventions aiming at a target such as CXCR3 for certain disease could result in adverse effects in an unrelated organ.
Collapse
Affiliation(s)
- Qing-Zhi Liu
- Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Chronic Disease Laboratory, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Wen-Tao Ma
- Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei, China.,College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, China
| | - Jing-Bo Yang
- Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zhi-Bin Zhao
- Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Chronic Disease Laboratory, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Kai Yan
- Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Chronic Disease Laboratory, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Yuan Yao
- Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Chronic Disease Laboratory, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Liang Li
- Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Chronic Disease Laboratory, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Qi Miao
- Department of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Zhe-Xiong Lian
- Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Chronic Disease Laboratory, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| |
Collapse
|
32
|
Arneth BM. Activation of CD4 and CD8 T cell receptors and regulatory T cells in response to human proteins. PeerJ 2018; 6:e4462. [PMID: 29568705 PMCID: PMC5846456 DOI: 10.7717/peerj.4462] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/15/2018] [Indexed: 11/20/2022] Open
Abstract
This study assessed in detail the influence of four different human proteins on the activation of CD4+ and CD8+ T lymphocytes and on the formation of regulatory T cells. Human whole-blood samples were incubated with four different human proteins. The effects of these proteins on the downstream immune-system response, on the expression of extracellular activation markers on and intracellular cytokines in T lymphocytes, and on the number of regulatory T cells (T-reg cells) were investigated via flow cytometry. Incubation with β-actin or glyceraldehyde 3-phosphate dehydrogenase (GAPDH), which are cytoplasmic proteins, increased the expression of both extracellular activation markers (CD69 and HLA-DR) and intracellular cytokines but did not significantly affect the number of T-reg cells. In contrast, incubation with human albumin or insulin, which are serum proteins, reduced both extracellular activation markers and intracellular cytokine expression and subsequently increased the number of T-reg cells. These findings may help to explain the etiological basis of autoimmune diseases.
Collapse
Affiliation(s)
- Borros M Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, University Hospital of the Universities of Giessen and Marburg UKGM, Justus Liebig University Giessen, Giessen, Hessen, Germany
| |
Collapse
|
33
|
Che YM, Zhang Y, Li M, Li XP, Zhang LL. In vitro and in vivo effect of PD-1/PD-L1 blockade on microglia/macrophage activation and T cell subset balance in cryptococcal meningitis. J Cell Biochem 2017; 119:3044-3057. [PMID: 29058791 DOI: 10.1002/jcb.26432] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/18/2017] [Indexed: 11/11/2022]
Abstract
This study aimed to investigate the PD-1/ PD-L1 signaling pathway and its effects the activation of microglia/macrophage and balancing T cell subsets in cryptococcal meningitis (CM). A total of 126 CM patients and 126 healthy individuals were recruited for the study. The CM patients were treated with amphotericin B (AmB). Seventy five C57BL/6 mice were grouped into the normal control, CM model, CM + AmB, sham, and CM + PD-1 antibodies (Ab) groups. CD4+ and CD8+ T cells as well as microglia/macrophages were analyzed by means of flow cytometry. Ionized calcium-binding adaptor molecule 1 (Ibal) expression was detected using western blotting and immunohistochemistry techniques. And the expression of Rab5 and Rab11 were detected using an immunofluorescence assay. Both PD-1 and PD-L1 mRNA and protein expression among the mice in the study were evaluated by qRT-PCR and western blotting methods. Compared to the CM model group, the CM + AmB and CM + PD-1 Ab groups exhibited increased levels of Th1 cytokines and chemokines expression, and reduced levels of Th2 cytokines expressions. Elevated cell purity and viability of CD4+ T cell were recorded as well as increases in microglia, however, there were reductions in the number of CD8+ T cells. Depleted expressions of Ibal, Rab5, and Rab11 as well as reduced mRNA expressions of PD-1 and PD-L1 in CD4+ , microglia, and macrophage cells. The findings suggested that suppression of the PD-1/PD-L1 signaling pathway restricts the proliferation of CM by down-regulating the expressions of Th2 cells and suppressing microglia and macrophage activation.
Collapse
Affiliation(s)
- Yuan-Mei Che
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Yi Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Ming Li
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Xiao-Peng Li
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Lun-Li Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China
| |
Collapse
|
34
|
Xu-Monette ZY, Zhang M, Li J, Young KH. PD-1/PD-L1 Blockade: Have We Found the Key to Unleash the Antitumor Immune Response? Front Immunol 2017; 8:1597. [PMID: 29255458 PMCID: PMC5723106 DOI: 10.3389/fimmu.2017.01597] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/06/2017] [Indexed: 12/13/2022] Open
Abstract
PD-1-PD-L1 interaction is known to drive T cell dysfunction, which can be blocked by anti-PD-1/PD-L1 antibodies. However, studies have also shown that the function of the PD-1-PD-L1 axis is affected by the complex immunologic regulation network, and some CD8+ T cells can enter an irreversible dysfunctional state that cannot be rescued by PD-1/PD-L1 blockade. In most advanced cancers, except Hodgkin lymphoma (which has high PD-L1/L2 expression) and melanoma (which has high tumor mutational burden), the objective response rate with anti-PD-1/PD-L1 monotherapy is only ~20%, and immune-related toxicities and hyperprogression can occur in a small subset of patients during PD-1/PD-L1 blockade therapy. The lack of efficacy in up to 80% of patients was not necessarily associated with negative PD-1 and PD-L1 expression, suggesting that the roles of PD-1/PD-L1 in immune suppression and the mechanisms of action of antibodies remain to be better defined. In addition, important immune regulatory mechanisms within or outside of the PD-1/PD-L1 network need to be discovered and targeted to increase the response rate and to reduce the toxicities of immune checkpoint blockade therapies. This paper reviews the major functional and clinical studies of PD-1/PD-L1, including those with discrepancies in the pathologic and biomarker role of PD-1 and PD-L1 and the effectiveness of PD-1/PD-L1 blockade. The goal is to improve understanding of the efficacy of PD-1/PD-L1 blockade immunotherapy, as well as enhance the development of therapeutic strategies to overcome the resistance mechanisms and unleash the antitumor immune response to combat cancer.
Collapse
Affiliation(s)
- Zijun Y. Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianyong Li
- Department of Hematology, JiangSu Province Hospital, The First Affiliated Hospital of NanJing Medical University, NanJing, JiangSu Province, China
| | - Ken H. Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Graduate School of Biomedical Science, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
35
|
|
36
|
CD4+ T cells in patients with chronic inflammatory rheumatic disorders show distinct levels of exhaustion. J Allergy Clin Immunol 2016; 138:586-589.e10. [DOI: 10.1016/j.jaci.2016.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 04/07/2016] [Accepted: 04/20/2016] [Indexed: 11/23/2022]
|
37
|
Spanier JA, Frederick DR, Taylor JJ, Heffernan JR, Kotov DI, Martinov T, Osum KC, Ruggiero JL, Rust BJ, Landry SJ, Jenkins MK, McLachlan JB, Fife BT. Efficient generation of monoclonal antibodies against peptide in the context of MHCII using magnetic enrichment. Nat Commun 2016; 7:11804. [PMID: 27292946 PMCID: PMC4909947 DOI: 10.1038/ncomms11804] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 05/02/2016] [Indexed: 11/09/2022] Open
Abstract
Monoclonal antibodies specific for foreign antigens, auto-antigens, allogeneic antigens and tumour neo-antigens in the context of major histocompatibility complex II (MHCII) are highly desirable as novel immunotherapeutics. However, there is no standard protocol for the efficient generation of monoclonal antibodies that recognize peptide in the context of MHCII, and only a limited number of such reagents exist. In this report, we describe an approach for the generation and screening of monoclonal antibodies specific for peptide bound to MHCII. This approach exploits the use of recombinant peptide:MHC monomers as immunogens, and subsequently relies on multimers to pre-screen and magnetically enrich the responding antigen-specific B cells before fusion and validation, thus saving significant time and reagents. Using this method, we have generated two antibodies enabling us to interrogate antigen presentation and T-cell activation. This methodology sets the standard to generate monoclonal antibodies against the peptide–MHCII complexes. Generating antibodies specific for the peptide–MHCII complexes has been challenging, with only a handful made to date. Here, the authors develop a more efficient approach to generate these antibodies, and demonstrate their potential in research and therapeutic applications.
Collapse
Affiliation(s)
- Justin A Spanier
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - Daniel R Frederick
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Justin J Taylor
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - James R Heffernan
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - Dmitri I Kotov
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - Kevin C Osum
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - Jenna L Ruggiero
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - Blake J Rust
- Department of Biochemistry, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Samuel J Landry
- Department of Biochemistry, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Marc K Jenkins
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - James B McLachlan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
38
|
Hamilton-Williams EE, Bergot AS, Reeves PLS, Steptoe RJ. Maintenance of peripheral tolerance to islet antigens. J Autoimmun 2016; 72:118-25. [PMID: 27255733 DOI: 10.1016/j.jaut.2016.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 01/04/2023]
Abstract
Reestablishment of immune tolerance to the insulin-producing beta cells is the desired goal for type 1 diabetes (T1D) treatment and prevention. Immune tolerance to multiple islet antigens is defective in individuals with T1D, but the mechanisms involved are multifaceted and may involve loss of thymic and peripheral tolerance. In this review we discuss our current understanding of the varied mechanisms by which peripheral tolerance to islet antigens is maintained in healthy individuals where genetic protection from T1D is present and how this fails in those with genetic susceptibility to disease. Novel findings in regards to expression of neo-islet antigens, non-classical regulatory cell subsets and the impact of specific genetic variants on tolerance induction are discussed.
Collapse
Affiliation(s)
- Emma E Hamilton-Williams
- The University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia.
| | - Anne-Sophie Bergot
- The University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Peta L S Reeves
- The University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Raymond J Steptoe
- The University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
39
|
Intestinal APCs of the endogenous nanomineral pathway fail to express PD-L1 in Crohn's disease. Sci Rep 2016; 6:26747. [PMID: 27226337 PMCID: PMC4880906 DOI: 10.1038/srep26747] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 05/03/2016] [Indexed: 01/06/2023] Open
Abstract
Crohn’s disease is a chronic inflammatory condition most commonly affecting the ileum and colon. The aetiology of Crohn’s disease is complex and may include defects in peptidoglycan recognition, and/or failures in the establishment of intestinal tolerance. We have recently described a novel constitutive endogenous delivery system for the translocation of nanomineral-antigen-peptidoglycan (NAP) conjugates to antigen presenting cells (APCs) in intestinal lymphoid patches. In mice NAP conjugate delivery to APCs results in high surface expression of the immuno-modulatory molecule programmed death receptor ligand 1 (PD-L1). Here we report that NAP conjugate positive APCs in human ileal tissues from individuals with ulcerative colitis and intestinal carcinomas, also have high expression of PD-L1. However, NAP-conjugate positive APCs in intestinal tissue from patients with Crohn’s disease show selective failure in PD-L1 expression. Therefore, in Crohn’s disease intestinal antigen taken up by lymphoid patch APCs will be presented without PD-L1 induced tolerogenic signalling, perhaps initiating disease.
Collapse
|
40
|
Martinov T, Spanier JA, Pauken KE, Fife BT. PD-1 pathway-mediated regulation of islet-specific CD4 + T cell subsets in autoimmune diabetes. ACTA ACUST UNITED AC 2016; 3. [PMID: 27656680 PMCID: PMC5027981 DOI: 10.14800/ie.1164] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type 1 diabetes (T1D) is a CD4+ T cell-driven autoimmune disease resulting from the destruction of insulin-producing pancreatic beta cells. Clinical evidence and studies in non-obese diabetic (NOD) mice suggest that insulin is a major autoantigen. With this in mind, we developed insulin B10-23:IAg7 tetramer reagents to track insulin-specific CD4+ T cells in mice and interrogated the role of Programmed death-1 (PD-1) for peripheral tolerance. PD-1 is a T cell inhibitory receptor necessary to maintain tolerance and prevent T1D in NOD mice. PD-1 pathway inhibitors are increasingly used in the clinic for treating malignancies, and while many patients benefit, some develop adverse autoimmune events, including T1D. We therefore sought to understand the role of PD-1 in maintaining islet-specific tolerance in diabetes-resistant strains. B6.g7 mice express the same MHC Class II allele as NOD mice, have predominantly naïve insulin-specific CD4+ T cells in the periphery, and remain diabetes-free even after PD-1 pathway blockade. Here, we examined the trafficking potential of insulin-specific CD4+ T cells in NOD and B6.g7 mice with or without anti-PD-L1 treatment, and found that PD-L1 blockade preferentially increased the number of CD44highCXCR3+ insulin-specific cells in NOD but not B6.g7 mice. Additionally, we investigated whether pancreatic islets in NOD and B6.g7 mice expressed CXCL10, a lymphocyte homing chemokine and ligand for CXCR3. Anti-PD-L1 treated and control NOD mice had detectable CXCL10 expression in the islets, while B6.g7 islets did not. These data suggest that islet tolerance may be in part attributed to the pancreatic environment and in the absence of pancreas inflammation, chemotactic cytokines may be missing. This, together with our previous data showing that PD-1 pathway blockade preferentially affects effector but not anergic self-specific T cells has implications for the use of checkpoint blockade in treating tumor patients. Our work suggests that determining tumor- and self-specific CD4+ T cell activation status (naïve, effector or anergic) prior to initiation of immunotherapy would likely help to stratify individuals who would benefit from this therapy versus those who might have adverse effects or incomplete tumor control.
Collapse
Affiliation(s)
- Tijana Martinov
- Center for Immunology, Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Justin A Spanier
- Center for Immunology, Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Kristen E Pauken
- Center for Immunology, Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Brian T Fife
- Center for Immunology, Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|