Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Immunity ; 56(7): 1631-1648.e10, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37392737

ABSTRACT

CD137 (4-1BB)-activating receptor represents a promising cancer immunotherapeutic target. Yet, the cellular program driven by CD137 and its role in cancer immune surveillance remain unresolved. Using T cell-specific deletion and agonist antibodies, we found that CD137 modulates tumor infiltration of CD8+-exhausted T (Tex) cells expressing PD1, Lag-3, and Tim-3 inhibitory receptors. T cell-intrinsic, TCR-independent CD137 signaling stimulated the proliferation and the terminal differentiation of Tex precursor cells through a mechanism involving the RelA and cRel canonical NF-κB subunits and Tox-dependent chromatin remodeling. While Tex cell accumulation induced by prophylactic CD137 agonists favored tumor growth, anti-PD1 efficacy was improved with subsequent CD137 stimulation in pre-clinical mouse models. Better understanding of T cell exhaustion has crucial implications for the treatment of cancer and infectious diseases. Our results identify CD137 as a critical regulator of Tex cell expansion and differentiation that holds potential for broad therapeutic applications.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Cell Differentiation , Cell Proliferation , Receptors, Antigen, T-Cell
2.
Blood ; 144(12): 1271-1283, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-38875515

ABSTRACT

ABSTRACT: The promising results obtained with immunotherapeutic approaches for multiple myeloma (MM) call for a better stratification of patients based on immune components. The most pressing being cytotoxic lymphocytes such as natural killer (NK) cells that are mandatory for MM surveillance and therapy. Here, we performed a single-cell RNA sequencing analysis of NK cells from 10 patients with MM and 10 age/sex-matched healthy donors that revealed important transcriptomic changes in the NK cell landscape affecting both the bone marrow (BM) and peripheral blood compartment. The frequency of mature cytotoxic CD56dim NK cell subsets was reduced in patients with MM at the advantage of late-stage NK cell subsets expressing NF-κB and interferon-I inflammatory signatures. These NK cell subsets accumulating in patients with MM were characterized by low CD16 and CD226 expression and poor cytotoxic functions. MM CD16/CD226Lo NK cells also had adhesion defects with reduced lymphocyte function-associated antigen 1 (LFA-1) integrin activation and actin polymerization that may account for their limited effector functions in vitro. Finally, analysis of BM-infiltrating NK cells in a retrospective cohort of 177 patients with MM from the Intergroupe Francophone du Myélome (IFM) 2009 trial demonstrated that a high frequency of NK cells and their low CD16 and CD226 expression were associated with a shorter overall survival. Thus, CD16/CD226Lo NK cells with reduced effector functions accumulate along MM development and negatively affect patients' clinical outcomes. Given the growing interest in harnessing NK cells to treat myeloma, this improved knowledge around MM-associated NK cell dysfunction will stimulate the development of more efficient immunotherapeutic drugs against MM.


Subject(s)
Cell Adhesion , Killer Cells, Natural , Multiple Myeloma , Humans , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Prognosis , Female , Male , Cytotoxicity, Immunologic , Antigens, Differentiation, T-Lymphocyte/metabolism , Middle Aged , Aged , Retrospective Studies , Lymphocyte Function-Associated Antigen-1/metabolism , Receptors, IgG , GPI-Linked Proteins
3.
Trends Immunol ; 43(3): 180-194, 2022 03.
Article in English | MEDLINE | ID: mdl-35090787

ABSTRACT

The T cell receptor (TCR) endows T cells with antigen specificity and is central to nearly all aspects of T cell function. Each naïve T cell has a unique TCR sequence that is stably maintained during cell division. In this way, the TCR serves as a molecular barcode that tracks processes such as migration, differentiation, and proliferation of T cells. Recent technological advances have enabled sequencing of the TCR from single cells alongside deep molecular phenotypes on an unprecedented scale. In this review, we discuss strengths and limitations of TCR sequences as molecular barcodes and their application to study immune responses following Programmed Death-1 (PD-1) blockade in cancer. Additionally, we consider applications of TCR data beyond use as a barcode.


Subject(s)
Autoimmunity , Neoplasms , Autoimmunity/genetics , Humans , Neoplasms/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes
4.
Mol Cancer ; 21(1): 219, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36514045

ABSTRACT

Adoptive cell therapy (ACT) using tumor infiltrating lymphocytes (TIL) is being studied in multiple tumor types. However, little is known about clonal cell expansion in vitro and persistence of the ACT product in vivo. We performed single-cell RNA and T-Cell Receptor (TCR) sequencing on serial blood and tumor samples from a patient undergoing ACT, who did not respond. We found that clonal expansion varied during preparation of the ACT product, and only one expanded clone was preserved in the ACT product. The TCR of the preserved clone which persisted and remained activated for five months was previously reported as specific for cytomegalovirus and had upregulation of granzyme family genes and genes associated with effector functions (HLA-DQB1, LAT, HLA-DQA1, and KLRD1). Clones that contracted during TIL preparation had features of exhaustion and apoptosis. At disease progression, all previously detected clonotypes were detected. New clonotypes appearing in blood or tumor at disease progression were enriched for genes associated with cytotoxicity or stemness (FGFBP2, GNLY, GZMH, GZMK, IL7R, SELL and KLF2), and these might be harnessed for alternative cellular therapy or cytokine therapy. In-depth single-cell analyses of serial samples from additional ACT-treated patients is warranted, and viral- versus tumor-specificity should be carefully analyzed.


Subject(s)
Melanoma , Humans , Melanoma/genetics , Lymphocytes, Tumor-Infiltrating/pathology , Receptors, Antigen, T-Cell/genetics , Single-Cell Analysis , Treatment Failure , Disease Progression , Cell- and Tissue-Based Therapy , Immunotherapy, Adoptive
5.
Curr Opin Neurol ; 34(6): 827-833, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34569985

ABSTRACT

PURPOSE OF REVIEW: Immunotherapies such as immune checkpoint blockade have revolutionized cancer treatment, but current approaches have failed to improve outcomes in glioblastoma and other brain tumours. T cell dysfunction has emerged as one of the major barriers for the development of central nervous system (CNS)-directed immunotherapy. Here, we explore the unique requirements that T cells must fulfil to ensure immune surveillance in the CNS, and we analyse T cell dysfunction in glioblastoma (GBM) through the prism of CNS-resident immune responses. RECENT FINDINGS: Using comprehensive and unbiased techniques such as single-cell RNA sequencing, multiple studies have dissected the transcriptional state of CNS-resident T cells that patrol the homeostatic brain. A similar approach has revealed that in GBM, tumour-infiltrating T cells lack the hallmarks of antigen-driven exhaustion typical of melanoma and other solid tumours, suggesting the need for better presentation of tumour-derived antigens. Consistently, in a mouse model of GBM, increasing lymphatic drainage to the cervical lymph node was sufficient to promote tumour rejection. SUMMARY: For the success of future immunotherapy strategies, further work needs to explore the natural history of dysfunction in GBM tumour-infiltrating T cells, establish whether these originate from CNS-resident T cells and how they can be manipulated therapeutically.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Brain Neoplasms/therapy , Central Nervous System , Glioblastoma/therapy , Humans , Immunotherapy , Mice , T-Lymphocytes
6.
Blood ; 144(6): 591-592, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39115825
7.
Immunol Rev ; 276(1): 9-25, 2017 03.
Article in English | MEDLINE | ID: mdl-28258696

ABSTRACT

The introduction of immunotherapy with checkpoint receptor blockade has changed the treatment of advanced cancers, at times inducing prolonged remission. Nevertheless, the success rate of the approach is variable across patients and different tumor types, and treatment is often accompanied by severe immune-related side effects, suggesting the importance of co-inhibitory pathway for both prevention of autoimmunity and failure of tumor rejection. A better understanding of how to uncouple anti-tumor activity from loss of self-tolerance is necessary to increase the therapeutic efficacy of checkpoint immunotherapy. In this review, we describe basic concepts of T-cell exhaustion that occur in cancer, highlighting the role of co-inhibitory receptors in contributing to this process while preventing immunopathology. By providing an overview of the current therapeutic success and immune-related burden of secondary effects of checkpoint immunotherapy, we illustrate the "double-edged sword" related to interference with immune-regulatory pathways. Finally, since achieving tumor rejection while preserving self-tolerance is particularly important for the central nervous system, we analyze the case for checkpoint immunotherapy in glioblastoma, the most common adult brain tumor.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain Neoplasms/therapy , Costimulatory and Inhibitory T-Cell Receptors/immunology , Glioblastoma/therapy , Immunotherapy/methods , Self Tolerance , T-Lymphocytes/immunology , Adult , Animals , Brain Neoplasms/immunology , Glioblastoma/immunology , Humans , Immunosuppression Therapy , Tumor Escape
8.
Blood ; 142(21): 1763-1764, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37995107
9.
J Immunol ; 199(11): 3748-3756, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29061767

ABSTRACT

The genetic predisposition to multiple sclerosis (MS) is most strongly conveyed by MHC class II haplotypes, possibly by shaping the autoimmune CD4 T cell repertoire. Whether Ag-processing enzymes contribute to MS susceptibility by editing the peptide repertoire presented by these MHC haplotypes is unclear. Thymus-specific serine protease (TSSP) is expressed by thymic epithelial cells and thymic dendritic cells (DCs) and, in these two stromal compartments, TSSP edits the peptide repertoire presented by class II molecules. We show in this article that TSSP increases experimental autoimmune encephalomyelitis severity by limiting central tolerance to myelin oligodendrocyte glycoprotein. The effect on experimental autoimmune encephalomyelitis severity was MHC class II allele dependent, because the lack of TSSP expression conferred protection in NOD mice but not in C57BL/6 mice. Importantly, although human thymic DCs express TSSP, individuals segregate into two groups having a high or 10-fold lower level of expression. Therefore, the level of TSSP expression by thymic DCs may modify the risk factors for MS conferred by some MHC class II haplotypes.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Epithelial Cells/immunology , Multiple Sclerosis/immunology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Thymus Gland/metabolism , Adolescent , Animals , Cells, Cultured , Central Tolerance , Child , Child, Preschool , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Histocompatibility Antigens Class II/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Myelin-Oligodendrocyte Glycoprotein/immunology
10.
Eur J Immunol ; 46(9): 2247-59, 2016 09.
Article in English | MEDLINE | ID: mdl-27334749

ABSTRACT

T-cell polyspecificity, predicting that individual T cells recognize a continuum of related ligands, implies that multiple antigens can tolerize T cells specific for a given self-antigen. We previously showed in C57BL/6 mice that part of the CD4(+) T-cell repertoire specific for myelin oligodendrocyte glycoprotein (MOG) 35-55 also recognizes the neuronal antigen neurofilament medium (NF-M) 15-35. Such bi-specific CD4(+) T cells are frequent and produce inflammatory cytokines after stimulation. Since T cells recognizing two self-antigens would be expected to be tolerized more efficiently, this finding prompted us to study how polyspecificity impacts tolerance. We found that similar to MOG, NF-M is expressed in the thymus by medullary thymic epithelial cells, a tolerogenic population. Nevertheless, the frequency, phenotype, and capacity to transfer experimental autoimmune encephalomyelitis (EAE) of MOG35-55 -reactive CD4(+) T cells were increased in MOG-deficient but not in NF-M-deficient mice. We found that presentation of NF-M15-35 by I-A(b) on dendritic cells is of short duration, suggesting unstable MHC class II binding. Consistently, introducing an MHC-anchoring residue into NF-M15-35 (NF-M15-35 T20Y) increased its immunogenicity, activating a repertoire able to induce EAE. Our results show that in C57BL/6 mice bi-specific encephalitogenic T cells manage to escape tolerization due to inefficient exposure to two self-antigens.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immune Tolerance , Myelin Proteins/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Neurons/immunology , Animals , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/genetics , Neurofilament Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Cell Antigen Receptor Specificity/genetics , T-Cell Antigen Receptor Specificity/immunology , Thymus Gland/immunology , Thymus Gland/metabolism
11.
Brain ; 139(Pt 5): 1433-46, 2016 05.
Article in English | MEDLINE | ID: mdl-27000832

ABSTRACT

Polyspecific T cells recognizing multiple distinct self-antigens have been identified in multiple sclerosis and other organ-specific autoimmune diseases, but their pathophysiological relevance remains undetermined. Using a mouse model of multiple sclerosis, we show that autoimmune encephalomyelitis induction is strictly dependent on reactivation of pathogenic T cells by a peptide (35-55) derived from myelin oligodendrocyte glycoprotein (MOG). This disease-inducing response wanes after onset. Strikingly, the progression of disease is driven by the in situ activation and expansion of a minority of MOG35-55-specific T cells that also recognize neurofilament-medium (NF-M)15-35, an intermediate filament protein expressed in neurons. This mobilization of bispecific T cells is critical for disease progression as adoptive transfer of NF-M15-35/MOG35-55 bispecific T cell lines caused full-blown disease in wild-type but not NF-M-deficient recipients. Moreover, specific tolerance through injection of NF-M15-35 peptide at the peak of disease halted experimental autoimmune encephalomyelitis progression. Our findings highlight the importance of polyspecific autoreactive T cells in the aggravation and perpetuation of central nervous system autoimmunity.


Subject(s)
Autoantigens/immunology , Autoimmunity , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Peptide Fragments/immunology
12.
J Immunol ; 193(7): 3267-77, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25135834

ABSTRACT

The recognition of multiple ligands by a single TCR is an intrinsic feature of T cell biology, with important consequences for physiological and pathological processes. Polyspecific T cells targeting distinct self-antigens have been identified in healthy individuals as well as in the context of autoimmunity. We have previously shown that the 2D2 TCR recognizes the myelin oligodendrocyte glycoprotein epitope (MOG)35-55 as well as an epitope within the axonal protein neurofilament medium (NF-M15-35) in H-2(b) mice. In this study, we assess whether this cross-reactivity is a common feature of the MOG35-55-specific T cell response. To this end, we analyzed the CD4 T cell response of MOG35-55-immunized C57BL/6 mice for cross-reactivity with NF-M15-35. Using Ag recall responses, we established that an important proportion of MOG35-55-specific CD4 T cells also responded to NF-M15-35 in all mice tested. To study the clonality of this response, we analyzed 22 MOG35-55-specific T cell hybridomas expressing distinct TCR. Seven hybridomas were found to cross-react with NF-M15-35. Using an alanine scan of NF-M18-30 and an in silico predictive model, we dissected the molecular basis of cross-reactivity between MOG35-55 and NF-M15-35. We established that NF-M F24, R26, and V27 proved important TCR contacts. Strikingly, the identified TCR contacts are conserved within MOG38-50. Our data indicate that due to linear sequence homology, part of the MOG35-55-specific T cell repertoire of all C57BL/6 mice also recognizes NF-M15-35, with potential implications for CNS autoimmunity.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Myelin Sheath/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Neurofilament Proteins/immunology , Receptors, Antigen/immunology , Animals , Autoantigens/genetics , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/pathology , CD4-Positive T-Lymphocytes/pathology , Cross Reactions/genetics , Cross Reactions/immunology , Mice , Mice, Knockout , Myelin Sheath/genetics , Myelin-Oligodendrocyte Glycoprotein/genetics , Neurofilament Proteins/genetics , Peptide Fragments/genetics , Peptide Fragments/immunology , Receptors, Antigen/genetics
13.
Res Sq ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38464315

ABSTRACT

Effective anti-tumor immunity is largely driven by cytotoxic CD8+ T cells that can specifically recognize tumor antigens. However, the factors which ultimately dictate successful tumor rejection remain poorly understood. Here we identify a subpopulation of CD8+ T cells which are tumor antigen-specific in patients with melanoma but resemble KIR+CD8+ T cells with a regulatory function (Tregs). These tumor antigen-specific KIR+CD8+ T cells are detectable in both the tumor and the blood, and higher levels of this population are associated with worse overall survival. Our findings therefore suggest that KIR+CD8+ Tregs are tumor antigen-specific but uniquely suppress anti-tumor immunity in patients with melanoma.

14.
J Immunother Cancer ; 11(8)2023 08.
Article in English | MEDLINE | ID: mdl-37586769

ABSTRACT

Immune checkpoint inhibitors (ICIs) are increasingly being used to manage multiple tumor types. Unfortunately, immune-related adverse events affect up to 60% of recipients, often leading to treatment discontinuation in settings where few alternative cancer therapies may be available. Checkpoint inhibitor induced colitis (ICI-colitis) is a common toxicity for which the underlying mechanisms are poorly defined. To better understand the changing colon-specific and peripheral immune environments over the course of progression and treatment of colitis, we collected blood and colon tissue from a patient with Merkel cell carcinoma who developed colitis on treatment with pembrolizumab. We performed single-cell RNA sequencing and T-cell receptor sequencing on samples collected before, during and after pembrolizumab and after various interventions to mitigate toxicity. We report T-cells populations defined by cytotoxicity, memory, and proliferation markers at various stages of colitis. We show preferential depletion of CD8+ T cells with biologic therapy and nominate both circulating and colon-resident T-cell subsets as potential drivers of inflammation and response to immune suppression. Our findings highlight the need for further exploration of the colon immune environment and rationalize future studies evaluating biologics for ICI-colitis, including in the context of ICI re-challenge.


Subject(s)
Colitis , Skin Neoplasms , Humans , Immune Checkpoint Inhibitors/adverse effects , Single-Cell Gene Expression Analysis , Colitis/chemically induced , T-Lymphocyte Subsets
15.
Sci Transl Med ; 14(675): eabl3651, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36516268

ABSTRACT

Genome-wide association studies identifying hundreds of susceptibility loci for autoimmune diseases indicate that genes active in immune cells predominantly mediate risk. However, identification and functional characterization of causal variants remain challenging. Here, we focused on the immunomodulatory role of a protective variant of histone deacetylase 7 (HDAC7). This variant (rs148755202, HDAC7.p.R166H) was identified in a study of low-frequency coding variation in multiple sclerosis (MS). Through transcriptomic analyses, we demonstrate that wild-type HDAC7 regulates genes essential for the function of Foxp3+ regulatory T cells (Tregs), an immunosuppressive subset of CD4 T cells that is generally dysfunctional in patients with MS. Moreover, Treg-specific conditional hemizygous deletion of HDAC7 increased the severity of experimental autoimmune encephalitis (EAE), a mouse model of neuroinflammation. In contrast, Tregs transduced with the protective HDAC7 R166H variant exhibited higher suppressive capacity in an in vitro functional assay, mirroring phenotypes previously observed in patient samples. In vivo modeling of the human HDAC7 R166H variant by generation of a knock-in mouse model bearing an orthologous R150H substitution demonstrated decreased EAE severity linked to transcriptomic alterations of brain-infiltrating Tregs, as assessed by single-cell RNA sequencing. Our data suggest that dysregulation of epigenetic modifiers, a distinct molecular class associated with disease risk, may influence disease onset. Last, our approach provides a template for the translation of genetic susceptibility loci to detailed functional characterization, using in vitro and in vivo modeling.


Subject(s)
Multiple Sclerosis , T-Lymphocytes, Regulatory , Mice , Animals , Humans , Multiple Sclerosis/genetics , Genome-Wide Association Study , CD4-Positive T-Lymphocytes , Histone Deacetylases , Disease Models, Animal
16.
Nat Neurosci ; 25(3): 306-316, 2022 03.
Article in English | MEDLINE | ID: mdl-35260865

ABSTRACT

A key aspect of nearly all single-cell sequencing experiments is dissociation of intact tissues into single-cell suspensions. While many protocols have been optimized for optimal cell yield, they have often overlooked the effects that dissociation can have on ex vivo gene expression. Here, we demonstrate that use of enzymatic dissociation on brain tissue induces an aberrant ex vivo gene expression signature, most prominently in microglia, which is prevalent in published literature and can substantially confound downstream analyses. To address this issue, we present a rigorously validated protocol that preserves both in vivo transcriptional profiles and cell-type diversity and yield across tissue types and species. We also identify a similar signature in postmortem human brain single-nucleus RNA-sequencing datasets, and show that this signature is induced in freshly isolated human tissue by exposure to elevated temperatures ex vivo. Together, our results provide a methodological solution for preventing artifactual gene expression changes during fresh tissue digestion and a reference for future deeper analysis of the potential confounding states present in postmortem human samples.


Subject(s)
Neuroglia , Transcriptome , Brain , Humans , Microglia/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
17.
Sci Immunol ; 6(65): eabf4034, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34739340

ABSTRACT

Immune checkpoint blockade has demonstrated success in treating cancer but can lead to immune-related adverse events (irAEs), illustrating the centrality of these pathways in tolerance. Here, we describe programmed cell death protein 1 (PD-1) control of T cell responses, focusing on its unique restraint of regulatory T cell function. We examine successes and limitations of checkpoint blockade immunotherapy and review clinical and mechanistic features of irAEs. Last, we discuss strategies to modulate PD-1 blockade to enhance antitumor immunity while limiting autoimmunity.


Subject(s)
Autoimmunity , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Humans , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology
18.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33651881

ABSTRACT

Understanding the relationship between tumor and peripheral immune environments could allow longitudinal immune monitoring in cancer. Here, we examined whether T cells that share the same TCRαß and are found in both tumor and blood can be interrogated to gain insight into the ongoing tumor T cell response. Paired transcriptome and TCRαß repertoire of circulating and tumor-infiltrating T cells were analyzed at the single-cell level from matched tumor and blood from patients with metastatic melanoma. We found that in circulating T cells matching clonally expanded tumor-infiltrating T cells (circulating TILs), gene signatures of effector functions, but not terminal exhaustion, reflect those observed in the tumor. In contrast, features of exhaustion are displayed predominantly by tumor-exclusive T cells. Finally, genes associated with a high degree of blood-tumor TCR sharing were overexpressed in tumor tissue after immunotherapy. These data demonstrate that circulating TILs have unique transcriptional patterns that may have utility for the interrogation of T cell function in cancer immunotherapy.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/blood , Melanoma/immunology , Skin Neoplasms/blood , Skin Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Clone Cells , Cytotoxicity, Immunologic/genetics , Humans , Immunotherapy , Melanoma/pathology , Melanoma/therapy , Monitoring, Immunologic/methods , Neoplasm Metastasis , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/genetics , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Transcriptome
19.
Cancers (Basel) ; 13(5)2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33801444

ABSTRACT

Little is known about the subcellular localization and function of programmed cell death 4 (PDCD4) in melanoma. Our past studies suggest PDCD4 interacts with Pleckstrin Homology Domain Containing A5 (PLEKHA5) to influence melanoma brain metastasis outcomes, as high intracranial PDCD4 expression leads to improved survival. We aimed to define the subcellular distribution of PDCD4 in melanoma and in the tumor microenvironment during neoplastic progression and its impact on clinical outcomes. We analyzed multiple tissue microarrays with well-annotated clinicopathological variables using quantitative immunofluorescence and evaluated single-cell RNA-sequencing on a brain metastasis sample to characterize PDCD4+ immune cell subsets. We demonstrate differences in PDCD4 expression during neoplastic progression, with high tumor and stromal PDCD4 levels associated with improved survival in primary melanomas and in intracranial metastases, but not in extracranial metastatic disease. While the expression of PDCD4 is well-documented on CD8+ T cells and natural killer cells, we show that it is also found on B cells and mast cells. PDCD4 expression in the tumor microenvironment is associated with increased immune cell infiltration. Further studies are needed to define the interaction of PDCD4 and PLEKHA5 and to evaluate the utility of this pathway as a therapeutic target in melanoma brain metastasis.

20.
Nat Rev Immunol ; 20(11): 680-693, 2020 11.
Article in English | MEDLINE | ID: mdl-32269380

ABSTRACT

Regulatory T (Treg) cells constitute a dynamic population that is essential for controlling immune responses in health and disease. Defects in Treg cell function and decreases in Treg cell numbers have been observed in patients with autoimmunity and the opposite effects on Treg cells occur in cancer settings. Current research on new therapies for these diseases is focused on modulating Treg cell function to increase or decrease suppressive activity in autoimmunity and cancer, respectively. In this regard, several co-inhibitory receptors that are preferentially expressed by Treg cells under homeostatic conditions have recently been shown to control Treg cell function and stability in different disease settings. These receptors could be amenable to therapeutic targeting aimed at modulating Treg cell function and plasticity. This Review summarizes recent data regarding the role of co-inhibitory molecules in the control of Treg cell function and stability, with a focus on their roles and potential therapeutic use in autoimmunity and cancer.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Costimulatory and Inhibitory T-Cell Receptors/drug effects , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity , Drug Delivery Systems , Humans , Neoplasms/drug therapy , T-Lymphocytes, Regulatory/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL