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.
Gut ; 72(10): 1927-1941, 2023 10.
Article in English | MEDLINE | ID: mdl-37230755

ABSTRACT

OBJECTIVE: To better understand the immune microenvironment of pancreatic ductal adenocarcinomas (PDACs), here we explored the relevance of T and B cell compartmentalisation into tertiary lymphoid structures (TLSs) for the generation of local antitumour immunity. DESIGN: We characterised the functional states and spatial organisation of PDAC-infiltrating T and B cells using single-cell RNA sequencing (scRNA-seq), flow cytometry, multicolour immunofluorescence, gene expression profiling of microdissected TLSs, as well as in vitro assays. In addition, we performed a pan-cancer analysis of tumour-infiltrating T cells using scRNA-seq and sc T cell receptor sequencing datasets from eight cancer types. To evaluate the clinical relevance of our findings, we used PDAC bulk RNA-seq data from The Cancer Genome Atlas and the PRINCE chemoimmunotherapy trial. RESULTS: We found that a subset of PDACs harbours fully developed TLSs where B cells proliferate and differentiate into plasma cells. These mature TLSs also support T cell activity and are enriched with tumour-reactive T cells. Importantly, we showed that chronically activated, tumour-reactive T cells exposed to fibroblast-derived TGF-ß may act as TLS organisers by producing the B cell chemoattractant CXCL13. Identification of highly similar subsets of clonally expanded CXCL13 + tumour-infiltrating T cells across multiple cancer types further indicated a conserved link between tumour-antigen recognition and the allocation of B cells within sheltered hubs in the tumour microenvironment. Finally, we showed that the expression of a gene signature reflecting mature TLSs was enriched in pretreatment biopsies from PDAC patients with longer survival after receiving different chemoimmunotherapy regimens. CONCLUSION: We provided a framework for understanding the biological role of PDAC-associated TLSs and revealed their potential to guide the selection of patients for future immunotherapy trials.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Tertiary Lymphoid Structures , Humans , Tertiary Lymphoid Structures/metabolism , Tertiary Lymphoid Structures/pathology , Pancreatic Neoplasms/genetics , Carcinoma, Pancreatic Ductal/genetics , Immunity , Tumor Microenvironment , Pancreatic Neoplasms
2.
Front Immunol ; 12: 782852, 2021.
Article in English | MEDLINE | ID: mdl-34925363

ABSTRACT

In recent years, it became apparent that cancers either associated with viral infections or aberrantly expressing endogenous retroviral elements (EREs) are more immunogenic, exhibiting an intense intra-tumor immune cell infiltration characterized by a robust cytolytic apparatus. On the other hand, epigenetic regulation of EREs is crucial to maintain steady-state conditions and cell homeostasis. In line with this, epigenetic disruptions within steady-state cells can lead to cancer development and trigger the release of EREs into the cytoplasmic compartment. As such, detection of viral molecules by intracellular innate immune sensors leads to the production of type I and type III interferons that act to induce an antiviral state, thus restraining viral replication. This knowledge has recently gained momentum due to the possibility of triggering intratumoral activation of interferon responses, which could be used as an adjuvant to elicit strong anti-tumor immune responses that ultimately lead to a cascade of cytokine production. Accordingly, several therapeutic approaches are currently being tested using this rationale to improve responses to cancer immunotherapies. In this review, we discuss the immune mechanisms operating in viral infections, show evidence that exogenous viruses and endogenous retroviruses in cancer may enhance tumor immunogenicity, dissect the epigenetic control of EREs, and point to interferon pathway activation in the tumor milieu as a promising molecular predictive marker and immunotherapy target. Finally, we briefly discuss current strategies to modulate these responses within tumor tissues, including the clinical use of innate immune receptor agonists and DNA demethylating agents.


Subject(s)
Epigenesis, Genetic/immunology , Immunotherapy/methods , Interferon Type I/metabolism , Interferons/metabolism , Neoplasms/therapy , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials as Topic , DNA Demethylation/drug effects , Endogenous Retroviruses/genetics , Endogenous Retroviruses/immunology , Epigenesis, Genetic/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunity, Innate/genetics , Neoplasms/genetics , Neoplasms/immunology , Oncolytic Viruses/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Interferon Lambda
3.
Front Cell Dev Biol ; 9: 678127, 2021.
Article in English | MEDLINE | ID: mdl-34164398

ABSTRACT

The immune system plays a crucial role in cancer development either by fostering tumor growth or destroying tumor cells, which has open new avenues for cancer immunotherapy. It was only over the last decade that the role of B cells in controlling anti-tumor immune responses in the tumor milieu has begun to be appreciated. B and plasma cells can exert anti-tumor effects through antibody-dependent cell cytotoxicity (ADCC) and activation of the complement cascade, even though their effector functions extend beyond the classical humoral immunity. In tumor tissues, B cells can be found in lymphoid aggregates, known as tertiary lymphoid structures (TLSs), well-organized non-encapsulated structures composed of immune and stromal cells. These structures reflect a process of lymphoid neogenesis occurring in peripheral tissues upon long-lasting exposure to inflammatory signals. The TLS provides an area of intense B cell antigen presentation that can lead to optimal T cell activation and effector functions, as well as the generation of effector B cells, which can be further differentiated in either antibody-secreting plasma cells or memory B cells. Of clinical interest, the crosstalk between B cells and antigen-experienced and exhausted CD8+ T cells within mature TLS was recently associated with improved response to immune checkpoint blockade (ICB) in melanoma, sarcoma and lung cancer. Otherwise, B cells sparsely distributed in the tumor microenvironment or organized in immature TLSs were found to exert immune-regulatory functions, inhibiting anti-tumor immunity through the secretion of anti-inflammatory cytokines. Such phenotype might arise when B cells interact with malignant cells rather than T and dendritic cells. Differences in the spatial distribution likely underlie discrepancies between the role of B cells inferred from human samples or mouse models. Many fast-growing orthotopic tumors develop a malignant cell-rich bulk with reduced stroma and are devoid of TLSs, which highlights the importance of carefully selecting pre-clinical models. In summary, strategies that promote TLS formation in close proximity to tumor cells are likely to favor immunotherapy responses. Here, the cellular and molecular programs coordinating B cell development, activation and organization within TLSs will be reviewed, focusing on their translational relevance to cancer immunotherapy.

4.
Sci Immunol ; 4(42)2019 12 13.
Article in English | MEDLINE | ID: mdl-31836669

ABSTRACT

General control nonderepressible 2 (GCN2) is an environmental sensor controlling transcription and translation in response to nutrient availability. Although GCN2 is a putative therapeutic target for immuno-oncology, its role in shaping the immune response to tumors is poorly understood. Here, we used mass cytometry, transcriptomics, and transcription factor-binding analysis to determine the functional impact of GCN2 on the myeloid phenotype and immune responses in melanoma. We found that myeloid-lineage deletion of GCN2 drives a shift in the phenotype of tumor-associated macrophages and myeloid-derived suppressor cells (MDSCs) that promotes antitumor immunity. Time-of-flight mass cytometry (CyTOF) and single-cell RNA sequencing showed that this was due to changes in the immune microenvironment with increased proinflammatory activation of macrophages and MDSCs and interferon-γ expression in intratumoral CD8+ T cells. Mechanistically, GCN2 altered myeloid function by promoting increased translation of the transcription factor CREB-2/ATF4, which was required for maturation and polarization of macrophages and MDSCs in both mice and humans, whereas targeting Atf4 by small interfering RNA knockdown reduced tumor growth. Last, analysis of patients with cutaneous melanoma showed that GCN2-dependent transcriptional signatures correlated with macrophage polarization, T cell infiltrates, and overall survival. Thus, these data reveal a previously unknown dependence of tumors on myeloid GCN2 signals for protection from immune attack.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Macrophages/immunology , Melanoma/immunology , Myeloid-Derived Suppressor Cells/immunology , Protein Serine-Threonine Kinases/immunology , Tumor Microenvironment/immunology , Animals , Cells, Cultured , Humans , Mice
5.
Nature ; 563(7732): 579-583, 2018 11.
Article in English | MEDLINE | ID: mdl-30429608

ABSTRACT

The use of liquid biopsies for cancer detection and management is rapidly gaining prominence1. Current methods for the detection of circulating tumour DNA involve sequencing somatic mutations using cell-free DNA, but the sensitivity of these methods may be low among patients with early-stage cancer given the limited number of recurrent mutations2-5. By contrast, large-scale epigenetic alterations-which are tissue- and cancer-type specific-are not similarly constrained6 and therefore potentially have greater ability to detect and classify cancers in patients with early-stage disease. Here we develop a sensitive, immunoprecipitation-based protocol to analyse the methylome of small quantities of circulating cell-free DNA, and demonstrate the ability to detect large-scale DNA methylation changes that are enriched for tumour-specific patterns. We also demonstrate robust performance in cancer detection and classification across an extensive collection of plasma samples from several tumour types. This work sets the stage to establish biomarkers for the minimally invasive detection, interception and classification of early-stage cancers based on plasma cell-free DNA methylation patterns.


Subject(s)
Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/metabolism , DNA Methylation , DNA, Neoplasm/blood , DNA, Neoplasm/metabolism , Early Detection of Cancer/methods , Neoplasms/classification , Neoplasms/genetics , Adenocarcinoma/blood , Adenocarcinoma/genetics , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , DNA Mutational Analysis , Epigenesis, Genetic , Female , Heterografts , Humans , Liquid Biopsy , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplasms/blood , Organ Specificity , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/genetics
6.
Res Vet Sci ; 107: 220-227, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27473999

ABSTRACT

Salmonella enterica serovar Gallinarum biovar Gallinarum (SG) causes fowl typhoid in chickens, a septicemic infection which results in high mortality rates. This disease causes high economic impact to the poultry industry worldwide because of the mortality or elimination of positive flocks to control bacterial dissemination. Live vaccines are used in the fields, however the characterization of immune mechanisms important for protection are being studied to improve the efficacy of vaccination schemes. In this study, we evaluated the immune response in brown layer-hens, vaccinated or not, during the most critical period of infection. Cellular and humoral immunity were extensively evaluated until 7 days post-infection (DPI), by flow cytometry and ELISA, respectively. Furthermore, we evaluated the expression of important pro-inflammatory cytokines after infection of bone marrow derived macrophages (BMDMs) with the live attenuated SG vaccine and with the wild SG strain. The results showed an increasing production of IgG and IgM during the first week post-infection, in vaccinated layer-hens, which was absent in unvaccinated birds. The population of CD8(+)CD44(+) and CD4(+)CD44(+) T cells in spleen and cecal tonsils constantly decreased in unvaccinated birds in comparison with vaccinated layers. The expression of IFN-γ and TNF-α in BMDMs was induced by both SG strains (attenuated and wild) at similar levels (p>0.05). Vaccination with live SG vaccine reduced systemic infection by challenge strain of SG and prevented the mortality rate of 85% that occurred in unvaccinated layer-hens during 30 dpi. Furthermore, the immunization enhanced the proliferation of effector CD4(+) and CD8(+) T cells after challenge.


Subject(s)
Chickens , Poultry Diseases/prevention & control , Salmonella Infections, Animal/prevention & control , Salmonella Vaccines/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Chickens/immunology , Female , Immunity, Humoral , Immunization , Poultry Diseases/microbiology , Salmonella Infections, Animal/microbiology , Salmonella enterica/immunology , Tumor Necrosis Factor-alpha , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...