Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 5280, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902261

ABSTRACT

The regulatory circuits dictating CD8+ T cell responsiveness versus exhaustion during anti-tumor immunity are incompletely understood. Here we report that tumor-infiltrating antigen-specific PD-1+ TCF-1- CD8+ T cells express the immunosuppressive cytokine Fgl2. Conditional deletion of Fgl2 specifically in mouse antigen-specific CD8+ T cells prolongs CD8+ T cell persistence, suppresses phenotypic and transcriptomic signatures of T cell exhaustion, and improves control of the tumor. In a mouse model of chronic viral infection, PD-1+ CD8+ T cell-derived Fgl2 also negatively regulates virus-specific T cell responses. In humans, CD8+ T cell-derived Fgl2 is associated with poorer survival in patients with melanoma. Mechanistically, the dampened responsiveness of WT Fgl2-expressing CD8+ T cells, when compared to Fgl2-deficient CD8+ T cells, is underpinned by the cell-intrinsic interaction of Fgl2 with CD8+ T cell-expressed FcγRIIB and concomitant caspase 3/7-mediated apoptosis. Our results thus illuminate a cell-autonomous regulatory axis by which PD-1+ CD8+ T cells both express the receptor and secrete its ligand in order to mediate suppression of anti-tumor and anti-viral immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Melanoma , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Receptors, IgG , Animals , Female , Humans , Mice , Apoptosis , CD8-Positive T-Lymphocytes/immunology , Melanoma/immunology , Melanoma/genetics , Mice, Knockout , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Receptors, IgG/metabolism , Receptors, IgG/genetics , Receptors, IgG/immunology
2.
Sci Transl Med ; 15(710): eadd1868, 2023 08 23.
Article in English | MEDLINE | ID: mdl-37611081

ABSTRACT

Checkpoint inhibition using Fc-containing monoclonal antibodies has emerged as a powerful therapeutic approach to augment antitumor immunity. We recently showed that FcγRIIB, the only inhibitory IgG-Fc receptor, is expressed on a population of highly differentiated effector CD8+ T cells in the tumors of mice and humans, raising the possibility that CD8+ T cell responses may be directly modulated by checkpoint inhibitor binding to T cell-expressed FcγRIIB. Here, we show that despite exhibiting strong proliferative and cytokine responses at baseline, human FcγRIIBpos CD8+ T cells exhibited reduced responsiveness to both PD-1 and CTLA-4 checkpoint inhibition as compared with FcγRIIBneg CD8+ T cells in vitro. Moreover, frequencies of FcγRIIBpos CD8+ T cells were reduced after treatment of patients with melanoma with nivolumab in vivo. This reduced responsiveness was FcγRIIB dependent, because conditional genetic deletion of FcγRIIB on tumor-specific CD8+ T cells improved response to checkpoint blockade in B16 and LLC mouse models of cancer. The limited responsiveness of FcγRIIBpos CD8+ T cells was also dependent on an intact Fc region of the checkpoint inhibitor, in that treatment with Fc-devoid anti-PD-1 F(ab) fragments resulted in increased proliferation of FcγRIIBpos CD8+ T cells, without altering the response of FcγRIIBneg CD8+ T cells. Last, the addition of FcγRIIB blockade improved efficacy of PD-1 checkpoint inhibition in mouse models of melanoma, lung, and colon cancer. These results illuminate an FcγRIIB-mediated, cell-autonomous mechanism of CD8+ T cell suppression, which limits the efficacy of checkpoint inhibitors during antitumor immune responses in vivo.


Subject(s)
Colonic Neoplasms , Melanoma , Animals , Humans , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , CD8-Positive T-Lymphocytes , Cytokines , Disease Models, Animal , Immunoglobulin Fc Fragments , Melanoma/drug therapy , Receptors, IgG
3.
J Immunol ; 206(10): 2412-2419, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33911005

ABSTRACT

Sepsis induces significant immune dysregulation characterized by lymphocyte apoptosis and alterations in the cytokine milieu. Because cancer patients exhibit a 10-fold greater risk of developing sepsis compared with the general population, we aimed to understand how pre-existing malignancy alters sepsis-induced immune dysregulation. To address this question, we assessed the impact of tumor-specific CD8+ T cells on the immune response in a mouse model of cecal ligation and puncture (CLP)-induced sepsis. Tumor-bearing animals containing Thy1.1+ tumor-specific CD8+ T cells were subjected to CLP, and groups of animals received anti-Thy1.1 mAb to deplete tumor-specific CD8+ T cells or isotype control. Results indicated that depleting tumor-specific T cells significantly improved mortality from sepsis. The presence of tumor-specific CD8+ T cells resulted in increased expression of the 2B4 coinhibitory receptor and increased apoptosis of endogenous CD8+ T cells. Moreover, tumor-specific T cells were not reduced in number in the tumors during sepsis but did exhibit impaired IFN-γ production in the tumor, tumor draining lymph node, and spleen 24 h after CLP. Our research provides novel insight into the mechanisms by which pre-existing malignancy contributes to increased mortality during sepsis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity , Lung Neoplasms/complications , Lung Neoplasms/immunology , Melanoma, Experimental/complications , Melanoma, Experimental/immunology , Sepsis/complications , Sepsis/immunology , Skin Neoplasms/complications , Skin Neoplasms/immunology , Animals , Apoptosis/immunology , Cell Line, Tumor , Cytokines/blood , Interferon-gamma/metabolism , Lung Neoplasms/blood , Lymph Nodes/immunology , Male , Melanoma, Experimental/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Sepsis/blood , Sepsis/mortality , Skin Neoplasms/blood , Spleen/immunology , Thy-1 Antigens/genetics
4.
Exp Cell Res ; 403(1): 112567, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33812866

ABSTRACT

We chose to evaluate Hypoxanthine Guanine Phosphoribosyltransferase (HPRT) as a possible biomarker for prostate cancer due to its involvement in nucleotide synthesis and cell cycle progression. We utilized two prostate cancer cell lines (PC3 and DU145) along with patient tissue and knockdowns to evaluate overall HPRT expression. The surface localization of HPRT was determined utilizing flow cytometry, confocal microscopy, and scanning electron microscopy followed by ADCC to evaluate targeting potential. We found significant upregulation of HPRT within malignant samples with approximately 47% of patients had elevated levels of HPRT compared to normal controls. We also observed a significant association between HPRT and the plasma membrane of DU145 cells (p = 0.0004), but found no presence on PC3 cells (p = 0.14). This was confirmed with scanning electron microscopy and confocal microscopy. ADCC experiments were performed to determine whether HPRT could be used as a target antigen for selective cell-mediated killing. We found that DU145 cells treated with HPRT antibodies had a significantly higher incidence of cell death than both isotype treated samples and PC3 cells treated with the same concentrations of HPRT antibody. Finally, we determined that p53 had a significant impact on HPRT expression both internally and on the surface of cancer cells. These results suggest HPRT as a possible biomarker target for the treatment of patients with prostate cancer.


Subject(s)
Cell Division/physiology , Cytotoxicity, Immunologic/immunology , Hypoxanthine Phosphoribosyltransferase/metabolism , Prostatic Neoplasms/metabolism , Cell Line , Cell Membrane/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/immunology , Male , Prostatic Neoplasms/immunology , Tumor Suppressor Protein p53/metabolism
5.
JCI Insight ; 6(4)2021 02 22.
Article in English | MEDLINE | ID: mdl-33616086

ABSTRACT

In the setting of cancer, T cells upregulate coinhibitory molecules that attenuate TCR signaling and lead to the loss of proliferative capacity and effector function. Checkpoint inhibitors currently in clinical use have dramatically improved mortality from melanoma yet are not effective in all patients, suggesting that additional pathways may contribute to suppression of tumor-specific CD8+ T cell responses in melanoma. Here, we show that FcγRIIB, an inhibitory Fc receptor previously thought to be exclusively expressed on B cells and innate immune cells, is upregulated on tumor-infiltrating effector CD8+ T cells in an experimental melanoma model and expressed on CD8+ T cells in patients with melanoma. Genetic deficiency of Fcgr2b resulted in enhanced tumor-infiltrating CD8+ T cell responses and significantly reduced tumor burden. Adoptive transfer experiments of Fcgr2b-/- tumor antigen-specific T cells into FcγRIIB-sufficient hosts resulted in an increased frequency of tumor-infiltrating CD8+ T cells with greater effector function. Finally, FcγRIIB was expressed on CD8+ memory T cells isolated from patients with melanoma. These data illuminate a cell-intrinsic role for the FcγRIIB checkpoint in suppressing tumor-infiltrating CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Adaptive Immunity , Adoptive Transfer , Adult , Aged , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Female , Humans , Immunologic Memory , Male , Memory T Cells , Mice , Mice, Inbred C57BL , Middle Aged , T-Lymphocytes , Transcriptome
6.
Immunol Res ; 68(1): 63-70, 2020 02.
Article in English | MEDLINE | ID: mdl-32096010

ABSTRACT

With several different CAR T cell therapies under advanced phases of clinical trials, and the first FDA-approved CAR treatments in 2017 (Yescarta and Kymriah), CAR T cell therapy has become one of the most promising therapies for the treatment of certain types of cancer. This success has bred an opportunity to optimize the production of CAR T cells for easier patient access. CAR T cell therapy is a rather expensive and personalized process that requires expensive measures to collect cells from patients, engineer those cells, and re-infuse the cells into the patient with adequate quality controls at each phase. With this in mind, significant attempts at creating a "universal" CAR T cell are underway in order to create an "off-the-shelf" product that would reduce the expense and time required for traditional CAR T cell treatment. The primary obstacle facing this endeavor is avoiding graft-versus-host disease that accompanies allogeneic transplants between genetically dissimilar individuals. With the advent of CRISPR and TALEN technology, editing the genome of allogeneic cells has become very possible, and several groups have provided initial data analyzing the effects of CAR T cells that have been edited to avoid host rejection and avoid endogenous TCR alloreactivity. These engineered cells not only have to avoid GVHD but also have to retain their anti-tumor efficacy in vivo. Here, we expand on the recent efforts and strides that have been made in the design and testing of universal allogeneic CAR T cells.


Subject(s)
Graft vs Host Disease/prevention & control , Immunotherapy, Adoptive/methods , Isoantigens/metabolism , Neoplasms/therapy , T-Lymphocytes/physiology , Animals , CRISPR-Cas Systems , Genetic Engineering , Humans , Isoantigens/genetics , Isoantigens/immunology , Neoplasms/immunology , Precision Medicine , T-Lymphocytes/transplantation , Transcription Activator-Like Effector Nucleases/metabolism , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...