1
|
Zhu J, Naulaerts S, Boudhan L, Martin M, Gatto L, Van den Eynde BJ. Tumour immune rejection triggered by activation of α2-adrenergic receptors. Nature 2023:10.1038/s41586-023-06110-8. [PMID: 37286594 DOI: 10.1038/s41586-023-06110-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 04/20/2023] [Indexed: 06/09/2023]
Abstract
Immunotherapy based on immunecheckpoint blockade (ICB) using antibodies induces rejection of tumours and brings clinical benefit in patients with various cancer types1. However, tumours often resist immune rejection. Ongoing efforts trying to increase tumour response rates are based on combinations of ICB with compounds that aim to reduce immunosuppression in the tumour microenvironment but usually have little effect when used as monotherapies2,3. Here we show that agonists of α2-adrenergic receptors (α2-AR) have very strong anti-tumour activity when used as monotherapies in multiple immunocompetent tumour models, including ICB-resistant models, but not in immunodeficient models. We also observed marked effects in human tumour xenografts implanted in mice reconstituted with human lymphocytes. The anti-tumour effects of α2-AR agonists were reverted by α2-AR antagonists, and were absent in Adra2a-knockout (encoding α2a-AR) mice, demonstrating on-target action exerted on host cells, not tumour cells. Tumours from treated mice contained increased infiltrating T lymphocytes and reduced myeloid suppressor cells, which were more apoptotic. Single-cell RNA-sequencing analysis revealed upregulation of innate and adaptive immune response pathways in macrophages and T cells. To exert their anti-tumour effects, α2-AR agonists required CD4+ T lymphocytes, CD8+ T lymphocytes and macrophages. Reconstitution studies in Adra2a-knockout mice indicated that the agonists acted directly on macrophages, increasing their ability to stimulate T lymphocytes. Our results indicate that α2-AR agonists, some of which are available clinically, could substantially improve the clinical efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Jingjing Zhu
- Ludwig Institute for Cancer Research, Brussels, Belgium.
- de Duve Institute, UCLouvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium.
| | - Stefan Naulaerts
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
| | - Loubna Boudhan
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
| | - Manon Martin
- de Duve Institute, UCLouvain, Brussels, Belgium
- Computational Biology and Bioinformatics, UCLouvain, Brussels, Belgium
| | - Laurent Gatto
- de Duve Institute, UCLouvain, Brussels, Belgium
- Computational Biology and Bioinformatics, UCLouvain, Brussels, Belgium
| | - Benoit J Van den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
2
|
Petit PF, Bombart R, Desimpel PH, Naulaerts S, Thouvenel L, Collet JF, Van den Eynde BJ, Zhu J. T-cell mediated targeted delivery of anti-PD-L1 nanobody overcomes poor antibody penetration and improves PD-L1 blocking at the tumor site. Cancer Immunol Res 2022; 10:713-727. [PMID: 35439300 DOI: 10.1158/2326-6066.cir-21-0801] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/09/2022] [Accepted: 04/15/2022] [Indexed: 11/16/2022]
Abstract
Monoclonal antibodies blocking immune checkpoints such as PD-L1 have yielded strong clinical benefits in many cancer types. Still, the current limitations are the lack of clinical response in a majority of patients and the development of immune-related adverse events in some. As an alternative to PD-L1-specific antibody injection, we have developed an approach based on the engineering of tumor-targeting T cells to deliver intratumorally an anti-PD-L1 nanobody. In the MC38-OVA model, our strategy enhanced tumor control as compared to injection of PD-L1-specific antibody combined with adoptive transfer of tumor-targeting T cells. As a possible explanation for this, we demonstrated that PD-L1-specific antibody massively occupied PD-L1 in the periphery but failed to penetrate to PD-L1-expressing cells at the tumor site. In sharp contrast, locally delivered anti-PD-L1 nanobody improved PD-L1 blocking at the tumor site while avoiding systemic exposure. Our approach appears promising to overcome the limitations of immunotherapy based on PD-L1-specific antibody treatment.
Collapse
Affiliation(s)
| | - Raphaele Bombart
- Ludwig Institute for Cancer Research, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Stefan Naulaerts
- Ludwig Institute for Cancer Research, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Laurie Thouvenel
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Benoit J Van den Eynde
- Ludwig Institute for Cancer Research, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Jingjing Zhu
- Ludwig Institute for Cancer Research, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
3
|
Canè S, Van Snick J, Uyttenhove C, Pilotte L, Van den Eynde BJ. TGFβ1 neutralization displays therapeutic efficacy through both an immunomodulatory and a non-immune tumor-intrinsic mechanism. J Immunother Cancer 2021; 9:e001798. [PMID: 33637600 PMCID: PMC7919595 DOI: 10.1136/jitc-2020-001798] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Transforming growth factor-β (TGFβ) is emerging as a promising target for cancer therapy, given its ability to promote progression of advanced tumors and to suppress anti-tumor immune responses. However, TGFβ also plays multiple roles in normal tissues, particularly during organogenesis, raising toxicity concerns about TGFβ blockade. Dose-limiting cardiovascular toxicity was observed, possibly due to the blockade of all three TGFβ isoforms. The dominant isoform in tumors is TGFβ1, while TGFβ2 and TGFβ3 seem to be more involved in cardiovascular development. Recent data indicated that selective targeting of TGFβ1 promoted the efficacy of checkpoint inhibitor anti-PD1 in transplanted preclinical tumor models, without cardiovascular toxicity. METHODS To further explore the therapeutic potential of isoform-specific TGFβ blockade, we developed neutralizing mAbs targeting mature TGFβ1 or TGFβ3, and tested them, in parallel with anti-panTGFβ mAb 1D11, in two preclinical models: the transplanted colon cancer model CT26, and the autochthonous melanoma model TiRP. RESULTS We observed that the blockade of TGFβ1, but not that of TGFβ3, increased the efficacy of a prophylactic cellular vaccine against colon cancer CT26. This effect was similar to pan-TGFβ blockade, and was associated with increased infiltration of activated CD8 T cells in the tumor, and reduced levels of regulatory T cells and myeloid-derived suppressor cells. In contrast, in the autochthonous TiRP melanoma model, we observed therapeutic efficacy of the TGFβ1-specific mAb as a single agent, while the TGFβ3 mAb was inactive. In this model, the anti-tumor effect of TGFβ1 blockade was tumor intrinsic rather than immune mediated, as it was also observed in T-cell depleted mice. Mechanistically, TGFβ1 blockade increased mouse survival by delaying the phenotype switch, akin to epithelial-to-mesenchymal transition (EMT), which transforms initially pigmented tumors into highly aggressive unpigmented tumors. CONCLUSIONS Our results confirm TGFβ1 as the relevant isoform to target for cancer therapy, not only in combination with checkpoint inhibitors, but also with other immunotherapies such as cancer vaccines. Moreover, TGFβ1 blockade can also act as a monotherapy, through a tumor-intrinsic effect blocking the EMT-like transition. Because human melanomas that resist therapy often express a gene signature that links TGFβ1 with EMT-related genes, these results support the clinical development of TGFβ1-specific mAbs in melanoma.
Collapse
Affiliation(s)
- Stefania Canè
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- Department of Medicine, Immunology Section, University of Verona, Verona, Italy
| | - Jacques Van Snick
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
| | - Catherine Uyttenhove
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
| | - Luc Pilotte
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- WELBIO, UCLouvain, Brussels, Belgium
| | - Benoit J Van den Eynde
- Ludwig Institute for Cancer Research, De Duve Institute, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- WELBIO, UCLouvain, Brussels, Belgium
| |
Collapse
|
4
|
Resistance to cancer immunotherapy mediated by apoptosis of tumor-infiltrating lymphocytes. Nat Commun 2017; 8:1404. [PMID: 29123081 PMCID: PMC5680273 DOI: 10.1038/s41467-017-00784-1] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 07/27/2017] [Indexed: 12/15/2022] Open
Abstract
Despite impressive clinical success, cancer immunotherapy based on immune checkpoint blockade remains ineffective in many patients due to tumoral resistance. Here we use the autochthonous TiRP melanoma model, which recapitulates the tumoral resistance signature observed in human melanomas. TiRP tumors resist immunotherapy based on checkpoint blockade, cancer vaccines or adoptive T-cell therapy. TiRP tumors recruit and activate tumor-specific CD8+ T cells, but these cells then undergo apoptosis. This does not occur with isogenic transplanted tumors, which are rejected after adoptive T-cell therapy. Apoptosis of tumor-infiltrating lymphocytes can be prevented by interrupting the Fas/Fas-ligand axis, and is triggered by polymorphonuclear-myeloid-derived suppressor cells, which express high levels of Fas-ligand and are enriched in TiRP tumors. Blocking Fas-ligand increases the anti-tumor efficacy of adoptive T-cell therapy in TiRP tumors, and increases the efficacy of checkpoint blockade in transplanted tumors. Therefore, tumor-infiltrating lymphocytes apoptosis is a relevant mechanism of immunotherapy resistance, which could be blocked by interfering with the Fas/Fas-ligand pathway. Cancer immunotherapy is ineffective in a subset of patients. Here the authors show that, in a mouse model of melanoma, resistance to immune checkpoint inhibitors relies on loss of tumor-specific T cells through FasL-mediated apoptosis triggered by polymorphonuclear-myeloid-derived suppressor cells.
Collapse
|
5
|
Frick M, Mouchacca P, Verdeil G, Hamon Y, Billaudeau C, Buferne M, Fallet M, Auphan-Anezin N, Schmitt-Verhulst AM, Boyer C. Distinct patterns of cytolytic T-cell activation by different tumour cells revealed by Ca 2+ signalling and granule mobilization. Immunology 2016; 150:199-212. [PMID: 27716898 DOI: 10.1111/imm.12679] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/26/2016] [Accepted: 09/30/2016] [Indexed: 12/22/2022] Open
Abstract
Cancer-germline genes in both humans and mice have been shown to encode antigens susceptible to targeting by cytotoxic CD8 T effector cells (CTL). We analysed the ability of CTL to kill different tumour cell lines expressing the same cancer-germline gene P1A (Trap1a). We previously demonstrated that CTL expressing a T-cell receptor specific for the P1A35-43 peptide associated with H-2Ld , although able to induce regression of P1A-expressing P815 mastocytoma cells, were much less effective against P1A-expressing melanoma cells. Here, we analysed parameters of the in vitro interaction between P1A-specific CTL and mastocytoma or melanoma cells expressing similar levels of the P1A gene and of surface H-2Ld . The mastocytoma cells were more sensitive to cytolysis than the melanoma cells in vitro. Analysis by video-microscopy of early events required for target cell killing showed that similar patterns of increase in cytoplasmic Ca2+ concentration ([Ca2+ ]i) were induced by both types of P1A-expressing tumour cells. However, the use of CTL expressing a fluorescent granzyme B (GZMB-Tom) showed a delay in the migration of cytotoxic granules to the tumour interaction site, as well as a partially deficient GZMB-Tom exocytosis in response to the melanoma cells. Among surface molecules possibly affecting tumour-CTL interactions, the mastocytoma cells were found to express intercellular adhesion molecule-1, the ligand for LFA-1, which was not detected on the melanoma cells.
Collapse
Affiliation(s)
- Melissa Frick
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | - Pierre Mouchacca
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | - Grégory Verdeil
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | - Yannick Hamon
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | - Cyrille Billaudeau
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | - Michel Buferne
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | - Mathieu Fallet
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | - Nathalie Auphan-Anezin
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | - Anne-Marie Schmitt-Verhulst
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | - Claude Boyer
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| |
Collapse
|
6
|
Buferne M, Chasson L, Grange M, Mas A, Arnoux F, Bertuzzi M, Naquet P, Leserman L, Schmitt-Verhulst AM, Auphan-Anezin N. IFNγ producing CD8 + T cells modified to resist major immune checkpoints induce regression of MHC class I-deficient melanomas. Oncoimmunology 2015; 4:e974959. [PMID: 25949872 PMCID: PMC4404920 DOI: 10.4161/2162402x.2014.974959] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/06/2014] [Indexed: 12/24/2022] Open
Abstract
Tumors with reduced expression of MHC class I (MHC-I) molecules may be unrecognized by tumor antigen-specific CD8+ T cells and thus constitute a challenge for cancer immunotherapy. Here we monitored development of autochthonous melanomas in TiRP mice that develop tumors expressing a known tumor antigen as well as a red fluorescent protein (RFP) reporter knock in gene. The latter permits non-invasive monitoring of tumor growth by biofluorescence. One developing melanoma was deficient in cell surface expression of MHC-I, but MHC-I expression could be rescued by exposure of these cells to IFNγ. We show that CD8+ T cells specific for tumor antigen/MHC-I were efficient at inducing regression of the MHC-I-deficient melanoma, provided that the T cells were endowed with properties permitting their migration into the tumor and their efficient production of IFNγ. This was the case for CD8+ T cells transfected to express an active form of STAT5 (STAT5CA). The amount of IFNγ produced ex vivo from T cells present in tumors after adoptive transfer of the CD8+ T cells was correlated with an increase in surface expression of MHC-I molecules by the tumor cells. We also show that these CD8+ T cells expressed PD-1 and upregulated its ligand PDL-1 on melanoma cells within the tumor. Despite upregulation of this immunosuppressive pathway, efficient IFNγ production in the melanoma microenvironment was found associated with resistance of STAT5CA-expressing CD8+ T cells to inhibition both by PD-1/PDL-1 engagement and by TGFβ1, two main immune regulatory mechanisms hampering the efficiency of immunotherapy in patients.
Collapse
Affiliation(s)
- Michel Buferne
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| | - Lionel Chasson
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| | - Magali Grange
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| | - Amandine Mas
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| | - Fanny Arnoux
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| | - Mélanie Bertuzzi
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| | - Philippe Naquet
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| | - Lee Leserman
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| | - Anne-Marie Schmitt-Verhulst
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| | - Nathalie Auphan-Anezin
- Centre d'Immunologie de Marseille-Luminy (CIML); UM2 Aix-Marseille Université ; Marseille, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) ; Marseille; France ; Centre National de la Recherche Scientifique (CNRS) ; Marseille; France
| |
Collapse
|
7
|
Huijbers IJ, Soudja SM, Uyttenhove C, Buferne M, Inderberg-Suso EM, Colau D, Pilotte L, Powis de Tenbossche CG, Chomez P, Brasseur F, Schmitt-Verhulst AM, Van den Eynde BJ. Minimal tolerance to a tumor antigen encoded by a cancer-germline gene. THE JOURNAL OF IMMUNOLOGY 2011; 188:111-21. [PMID: 22140254 DOI: 10.4049/jimmunol.1002612] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Central tolerance toward tissue-restricted Ags is considered to rely on ectopic expression in the thymus, which was also observed for tumor Ags encoded by cancer-germline genes. It is unknown whether endogenous expression shapes the T cell repertoire against the latter Ags and explains their weak immunogenicity. We addressed this question using mouse cancer-germline gene P1A, which encodes antigenic peptide P1A(35-43) presented by H-2L(d). We made P1A-knockout (P1A-KO) mice and asked whether their anti-P1A(35-43) immune responses were stronger than those of wild-type mice and whether P1A-KO mice responded to other P1A epitopes, against which wild-type mice were tolerized. We observed that both types of mice mounted similar P1A(35-43)-specific CD8 T cell responses, although the frequency of P1A(35-43)-specific CD8 T cells generated in response to P1A-expressing tumors was slightly higher in P1A-KO mice. This higher reactivity allowed naive P1A-KO mice to reject spontaneously P1A-expressing tumors, which progressed in wild-type mice. TCR-Vβ usage of P1A(35-43)-specific CD8 cells was slightly modified in P1A-KO mice. Peptide P1A(35-43) remained the only P1A epitope recognized by CD8 T cells in both types of mice, which also displayed similar thymic selection of a transgenic TCR recognizing P1A(35-43). These results indicate the existence of a minimal tolerance to an Ag encoded by a cancer-germline gene and suggest that its endogenous expression only slightly affects diversification of the T cell repertoire against this Ag.
Collapse
|
8
|
Cooperativity of adaptive and innate immunity: implications for cancer therapy. Cancer Immunol Immunother 2011; 60:1061-74. [PMID: 21656157 DOI: 10.1007/s00262-011-1053-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 05/26/2011] [Indexed: 02/07/2023]
Abstract
The dichotomy of immunology into innate and adaptive immunity has created conceptual barriers in appreciating the intrinsic two-way interaction between immune cells. An emerging body of evidence in various models of immune rejection, including cancer, indicates an indispensable regulation of innate effector functions by adaptive immune cells. This bidirectional cooperativity in innate and adaptive immune functions has broad implications for immune responses in general and for regulating the tumor-associated inflammation that overrides the protective antitumor immunity. Mechanistic understanding of this two-way immune cross-talk could provide insights into novel strategies for designing better immunotherapy approaches against cancer and other diseases that normally defy immune control.
Collapse
|
9
|
Han X, Ye P, Luo L, Zheng L, Liu Y, Chen L, Wang S. The development and functions of CD4(+) T cells expressing a transgenic TCR specific for an MHC-I-restricted tumor antigenic epitope. Cell Mol Immunol 2011; 8:333-40. [PMID: 21643003 DOI: 10.1038/cmi.2011.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
It has been reported that the ratio of CD4(+) to CD8(+) T cells has no bias in a few class I major histocompatibility complex (MHC-I)-restricted T-cell receptor (TCR)-transgenic mice specific for alloantigens or autoantigens, in which most CD4(+) T cells express an MHC-I-restricted TCR. In this study, we further showed that more than 50% of CD4(+) T cells in MHC-I-restricted P1A tumor antigen-specific TCR (P1ATCR)-transgenic mice could specifically bind to MHC-I/P1A peptide complex. P1A peptide could stimulate the transgenic CD4(+) T cells to proliferate and secrete both type 1 helper T cell and type 2 helper T cell cytokines. The activated CD4(+) T cells also showed cytotoxicity against P1A-expressing tumor cells. The analysis of TCR α-chains showed that these CD4(+) T cells were selected by co-expressing endogenous TCRs. Our results show that CD4(+) T cells from P1ATCR transgenic mice co-expressed an MHC-I-restricted transgenic TCR and another rearranged endogenous TCRs, both of which were functional.
Collapse
Affiliation(s)
- Xue Han
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
10
|
Soudja SM, Wehbe M, Mas A, Chasson L, de Tenbossche CP, Huijbers I, Van den Eynde B, Schmitt-Verhulst AM. Tumor-initiated inflammation overrides protective adaptive immunity in an induced melanoma model in mice. Cancer Res 2010; 70:3515-25. [PMID: 20406967 DOI: 10.1158/0008-5472.can-09-4354] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We studied the effect of the immune system on two differentially aggressive melanomas developing in mice on conditional deletion of the INK4A/ARF tumor suppressor gene, with concomitant expression of oncogene H-Ras(G12V) and a natural cancer-germline tumor antigen (TA). "Slow progressor" melanomas contained no activated T lymphocytes (TL). In contrast, "aggressive" melanomas were infiltrated by activated TLs lacking effector molecules and expressing high levels of PD-1, indicating an exhausted phenotype. Aggressive melanomas were also infiltrated by immature myeloid cells (IMC). Infiltration was associated with local inflammation and systemic Th2/Th17-oriented chronic inflammation that seemed to impair further activation of TLs, as tumor-specific T cells adoptively transferred into mice bearing aggressive melanomas were poorly activated and failed to infiltrate the melanoma. This immunosuppression also led to the incapacity of these mice to reject inoculated TA-positive tumors, in contrast to slow-progressing melanoma-bearing mice, which were responsive. To test the role of adaptive immunity in tumor progression, we induced melanomas in immunodeficient RagKO compound mice. These mice developed aggressive but not slow-progressing melanomas at a higher frequency and with a shorter latency than immunocompetent mice. Immunodeficient mice also developed abnormal inflammation and infiltration of IMCs in a manner similar to immunocompetent mice, indicating that this phenotype was not dependent on adaptive immunity. Therefore, tumor-intrinsic factors distinguishing the two melanoma types control the initiation of inflammation, which was independent of adaptive immunity. The latter delayed development of aggressive melanomas but was overridden by inflammation.
Collapse
Affiliation(s)
- Saïdi M Soudja
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerranée, Institut National de la Sante et de la Recherche Medicale, U631 Centre National de la Recherche Scientifique, UMR6102, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Shanker A, Buferne M, Schmitt-Verhulst AM. Cooperative action of CD8 T lymphocytes and natural killer cells controls tumour growth under conditions of restricted T-cell receptor diversity. Immunology 2010; 129:41-54. [PMID: 20050329 DOI: 10.1111/j.1365-2567.2009.03150.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In mice expressing a transgenic T-cell receptor (TCR; TCRP1A) of DBA/2 origin with reactivity towards a cancer-germline antigen P1A, the number of TCRP1A CD8+ T cells in lymphoid organs is lower in DBA/2 than in B10.D2 or B10.D2(x DBA/2)F1 mice. This reduction results from haemopoietic cell autonomous differences in the differentiation of the major histocompatibility complex class I-restricted TCRP1A thymocytes controlled by DBA/2 versus B10.D2-encoded genes. We report here that the lower number of TCRP1A CD8+ T cells in DBA/2 mice correlated with their poor resistance to P1A-expressing mastocytoma solid tumours. Functional potency of CD8+ cytolytic T lymphocytes (CTL) from the above strains was not compromised, but their number after expansion appeared to be influenced by their genetic background. Intriguingly, non-transgenic DBA/ 2 mice resisted P1A+ tumours more efficiently despite poor representation of P1A-specific CTL. This was partly the result of their more heterogeneous TCR repertoire, including reactivity to non-P1A tumour antigens because mice that had rejected a P1A+ tumour became resistant to a P1A) variant of the tumour. Such 'cross-resistance' did not develop in the TCRP1A transgenic mice. Nonetheless, reconstitution of RAGo/o mice with TCRP1A CD8+ T cells, with or without CD4+ T cells, or exclusive representation of TCRP1A CD8+ T cells in RAGo/o TCRP1A transgenic mice efficiently resisted the growth of P1A-expressing tumours. Natural killer cells present at a higher number in RAGo/o mice also contributed to tumour resistance, in part through an NKG2D-dependent mechanism. Hence, in the absence of a polyclonal T-cell repertoire, precursor frequencies of natural killer cells and tumour-specific CTL affect tumour resistance.
Collapse
Affiliation(s)
- Anil Shanker
- Centre d'Immunologie de Marseille-Luminy, Université de Méditerranée, Marseille cedex, France.
| | | | | |
Collapse
|
12
|
Koble C, Kyewski B. The thymic medulla: a unique microenvironment for intercellular self-antigen transfer. ACTA ACUST UNITED AC 2009; 206:1505-13. [PMID: 19564355 PMCID: PMC2715082 DOI: 10.1084/jem.20082449] [Citation(s) in RCA: 183] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Central tolerance is shaped by the array of self-antigens expressed and presented by various types of thymic antigen-presenting cells (APCs). Depending on the overall signal quality and/or quantity delivered in these interactions, self-reactive thymocytes either apoptose or commit to the T regulatory cell lineage. The cellular and molecular complexity underlying these events has only recently been appreciated. We analyzed the ex vivo presentation of ubiquitous or tissue-restricted self-antigens by medullary thymic epithelial cells (mTECs) and thymic dendritic cells (DCs), the two major APC types present in the medulla. We found that the ubiquitously expressed nuclear neo-self-antigen ovalbumin (OVA) was efficiently presented via major histocompatibility complex class II by mTECs and thymic DCs. However, presentation by DCs was highly dependent on antigen expression by TECs, and hemopoietic cells did not substitute for this antigen source. Accordingly, efficient deletion of OVA-specific T cells correlated with OVA expression by TECs. Notably, OVA was only presented by thymic but not peripheral DCs. We further demonstrate that thymic DCs are constitutively provided in situ with cytosolic as well as membrane-bound mTEC-derived proteins. The subset of DCs displaying transferred proteins was enriched in activated DCs, with these cells being most efficient in presenting TEC-derived antigens. These data provide evidence for a unique, constitutive, and unidirectional transfer of self-antigens within the thymic microenvironment, thus broadening the cellular base for tolerance induction toward promiscuously expressed tissue antigens.
Collapse
Affiliation(s)
- Christian Koble
- Division of Developmental Immunology, Tumor Immunology Program, German Cancer Research Center, 69120 Heidelberg, Germany
| | | |
Collapse
|
13
|
Dulude G, Cheynier R, Gauchat D, Abdallah A, Kettaf N, Sékaly RP, Gratton S. The magnitude of thymic output is genetically determined through controlled intrathymic precursor T cell proliferation. THE JOURNAL OF IMMUNOLOGY 2008; 181:7818-24. [PMID: 19017971 DOI: 10.4049/jimmunol.181.11.7818] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The thymus plays a crucial role in providing the immune system with naive T cells showing a diverse TCR repertoire. Whereas the diversity of thymic production is mainly ensured by TCR rearrangement at both the TRA and TRB loci, the number of cells reaching the double-positive differentiation stage defines the extent of thymic output. A quantitative analysis of TCR excision circles (TREC; signal-joint TRECs and DJbetaTRECs) produced at different stages of thymopoiesis was performed in nine laboratory mouse strains. The results clearly demonstrate that the magnitude of thymic output is directly proportional to the extent of proliferation in the double-negative 4 thymocyte subset. Strikingly, intrathymic precursor T cell proliferation was found to be strain dependent, thus suggesting a genetic regulation of thymic output. The inherited character of thymic output was further confirmed by the transmission of the phenotype in a recessive fashion in F(1) progeny of the different parental strains. Our results provide the first demonstration of the genetic regulation of thymic output.
Collapse
Affiliation(s)
- Gaël Dulude
- Laboratoire d'Immunologie, Centre de Recherches du Centre Hospitalier de l'Université Montréal, Saint-Luc, Montréal, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
14
|
Restoration of T-box-containing protein expressed in T cells protects against allergen-induced asthma. J Allergy Clin Immunol 2008; 123:479-85. [PMID: 19081613 DOI: 10.1016/j.jaci.2008.10.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Revised: 10/21/2008] [Accepted: 10/22/2008] [Indexed: 12/22/2022]
Abstract
BACKGROUND A T(H)1-specific transcription factor, T-box-containing protein expressed in T cells (T-bet), controls the production of both T(H)1 and T(H)2 cytokines in T(H) cell differentiation by means of distinct mechanisms. T-bet-deficient mice overproduce T(H)2 cytokines and have spontaneous airway inflammation. OBJECTIVES We tested whether T-bet overexpression could protect against the development or progression of asthma. METHODS We generated a T cell-specific and inducible line of T-bet-transgenic mice on a T-bet-deficient genetic background and used it to study the function of T-bet in an ovalbumin (OVA)-induced asthma model. RESULTS Induction of T-bet in a T cell-specific manner in an OVA model of asthma concomitant with OVA injection prevented airway hyperresponsiveness, eosinophilic and lymphocytic inflammation, and IL-5 and IL-13 production in bronchoalveolar lavage fluid and also reduced serum IgE and T(H)2 cytokine production by peripheral T cells. Even when T-bet expression was induced during later stages of asthma progression, T-bet overexpression still attenuated airway hyperresponsiveness and goblet cell hyperplasia, as well as T(H)2 cytokine production. CONCLUSIONS Our results suggest that T-bet expression in T cells can prevent the initiation of airway inflammation and progression of chronic inflammation and might be extrapolated to human asthma.
Collapse
|
15
|
Shanker A, Verdeil G, Buferne M, Inderberg-Suso EM, Puthier D, Joly F, Nguyen C, Leserman L, Auphan-Anezin N, Schmitt-Verhulst AM. CD8 T Cell Help for Innate Antitumor Immunity. THE JOURNAL OF IMMUNOLOGY 2007; 179:6651-62. [DOI: 10.4049/jimmunol.179.10.6651] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
16
|
Huijbers IJ, Krimpenfort P, Chomez P, van der Valk MA, Song JY, Inderberg-Suso EM, Schmitt-Verhulst AM, Berns A, Van den Eynde BJ. An inducible mouse model of melanoma expressing a defined tumor antigen. Cancer Res 2006; 66:3278-86. [PMID: 16540681 DOI: 10.1158/0008-5472.can-05-3216] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapy based on vaccination with defined tumor antigens has not yet shown strong clinical efficacy, despite promising results in preclinical models. This discrepancy might result from the fact that available preclinical models rely on transplantable tumors, which do not recapitulate the long-term host-tumor interplay that occurs in patients during progressive tumor development and results in tumor tolerance. To create a faithful preclinical model for cancer immunotherapy, we generated a transgenic mouse strain developing autologous melanomas expressing a defined tumor antigen recognized by T cells. We chose the antigen encoded by P1A, a well-characterized murine cancer germ line gene. To transform melanocytes, we aimed at simultaneously activating the Ras pathway and inactivating tumor suppressor Ink4a/Arf, thereby reproducing two genetic events frequently observed in human melanoma. The melanomas are induced by s.c. injection of 4-OH-tamoxifen (OHT). By activating a CreER recombinase expressed from a melanocyte-specific promoter, this treatment induces the loss of the conditional Ink4a/Arf gene in melanocytes. Because the CreER gene itself is also flanked by loxP sites, the activation of CreER also induces the deletion of its own coding sequence and thereby allows melanocyte-specific expression of genes H-ras and P1A, which are located downstream on the same transgene. All melanomas induced in those mice with OHT show activation of the Ras pathway and deletion of gene Ink4a/Arf. In addition, these melanomas express P1A and are recognized by P1A-specific T lymphocytes. This model will allow to characterize the interactions between the immune system and naturally occurring tumors and thereby to optimize immunotherapy approaches targeting a defined tumor antigen.
Collapse
Affiliation(s)
- Ivo J Huijbers
- Ludwig Institute for Cancer Research and Cellular Genetics Unit, Université Catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|