Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Immunother Cancer ; 10(1)2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35091453

RESUMO

BACKGROUND: While stimulator of interferon genes (STING) activation in innate immune cells of the tumor microenvironment can result in CD8 T cell-dependent antitumor immunity, whether STING signaling affects CD4 T-cell responses remains elusive. METHODS: Here, we tested whether STING activation modulated the effector functions of CD4 T cells in vivo by analyzing tumor-infiltrating CD4 T cells and evaluating the contribution of the CD4 T cell-derived cytokines in the antitumor activity of the STING ligand 2'3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) in two mouse tumor models. We performed ex vivo experiments to assess the impact of STING activation on CD4 T-cell differentiation and investigate the underlying molecular mechanisms. Finally, we tested whether STING activation enhances TH9 cell antitumor activity against mouse melanoma upon adoptive transfer. RESULTS: We found that activation of STING signaling cell-intrinsically enhances the differentiation and antitumor functions of TH1 and TH9 cells by increasing their respective production of interferon gamma (IFN-γ) and interleukin-9. IRF3 and type I interferon receptors (IFNARs) are required for the STING-driven enhancement of TH1 cell differentiation. However, STING activation favors TH9 cell differentiation independently of the IFNARs/IRF3 pathway but through mammalian target of rapamycin (mTOR) signaling, underscoring that STING activation differentially affects the fate of distinct CD4 T-cell subsets. The therapeutic effect of STING activation relies on TH1 and TH9-derived cytokines, and STING activation enhances the antitumor activity of TH9 cells upon adoptive transfer. CONCLUSION: Our results reveal the STING signaling pathway as a therapeutic target to boost CD4 T-cell effector functions and antitumor immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Interleucina-9/fisiologia , Proteínas de Membrana/fisiologia , Células Th1/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Diferenciação Celular , Feminino , Fator Regulador 3 de Interferon/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Nucleotídeos Cíclicos/farmacologia , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Células Th1/citologia
2.
Cells ; 9(7)2020 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-32707692

RESUMO

Immune checkpoint inhibitors (ICPi) have shown their superiority over conventional therapies to treat some cancers. ICPi are effective against immunogenic tumors. However, patients with tumors poorly infiltrated with immune cells do not respond to ICPi. Combining ICPi with other anticancer therapies such as chemotherapy, radiation, or vaccines, which can stimulate the immune system and recruit antitumor T cells into the tumor bed, may be a relevant strategy to increase the proportion of responding patients. Such an approach still raises the following questions: What are the immunological features modulated by immunogenic therapies that can be critical to ensure not only immediate but also long-lasting tumor protection? How must the combined treatments be administered to the patients to harness their full potential while limiting adverse immunological events? Here, we address these points by reviewing how immunogenic anticancer therapies can provide novel therapeutic opportunities upon combination with ICPi. We discuss their ability to create a permissive tumor microenvironment through the generation of inflamed tumors and stimulation of memory T cells such as resident (TRM) and stem-cell like (TSCM) cells. We eventually underscore the importance of sequence, dose, and duration of the combined anticancer therapies to design optimal and successful cancer immunotherapy strategies.


Assuntos
Inibidores de Checkpoint Imunológico/uso terapêutico , Animais , Terapia Combinada , Humanos , Memória Imunológica , Imunoterapia , Modelos Biológicos , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia
3.
Front Immunol ; 10: 1181, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31191545

RESUMO

The unprecedented clinical activity of checkpoint blockade in several types of cancers has formally demonstrated that anti-tumor immune responses are crucial in cancer therapy. Durable responses seen in patients treated with immune checkpoint inhibitors (ICI) show that they can trigger the establishment of long-lasting immunologic memory. This beneficial outcome is however achieved for a limited number of patients. In addition, late relapses are emerging suggesting the development of acquired resistances that compromise the anticancer efficacy of ICI. How can this be prevented through combination therapies? We here review the functions of immune checkpoints, the successes of ICI in treating cancer and their therapeutic limits. We discuss how conventional cancer therapies can be properly selected to set up combinatorial approaches with ICI leading to treatment improvement. We finally summarize clinical data showing the ongoing progress in cancer treatment involving ICI and chemotherapy combination strategies.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno CTLA-4/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Imunomodulação/efeitos dos fármacos , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Antineoplásicos Imunológicos/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígeno B7-H1/imunologia , Antígeno CTLA-4/imunologia , Ensaios Clínicos como Assunto , Sinergismo Farmacológico , Fluoruracila/administração & dosagem , Fluoruracila/farmacologia , Humanos , Ipilimumab/uso terapêutico , Leucovorina/administração & dosagem , Leucovorina/farmacologia , Ativação Linfocitária , Linfocinas/metabolismo , Camundongos , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Compostos Organoplatínicos/administração & dosagem , Compostos Organoplatínicos/farmacologia , Receptor de Morte Celular Programada 1/imunologia , Subpopulações de Linfócitos T/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
4.
Immunol Rev ; 280(1): 175-193, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29027217

RESUMO

Endogenous danger signals are molecules normally present in a given cell compartment that are rapidly released following cell stress and induce immune responses. We and others have shown that dying tumor cells treated with some chemotherapies are able to induce anticancer immune responses, which rely on their release of danger signals such as the nuclear protein HMGB1. DNA can also be released from chemotherapy-treated tumor cells, act as a danger signal, and boost anticancer immunity. While the immunostimulatory properties of DNA have been identified for decades, the recent discovery of a novel family of receptors, cytosolic DNA sensors, has provided a novel impetus not only to understand how chemotherapy can trigger anticancer immune responses but also to exploit DNA-derived molecules for therapeutic use. We will here discuss the molecular characteristics of endogenous danger signals released from chemotherapy-treated tumor cells and focus on the clinical relevance of using these danger signals in chemoimmunotherapeutic strategies against cancer.


Assuntos
Vacinas Anticâncer/imunologia , DNA/imunologia , Tratamento Farmacológico/métodos , Proteína HMGB1/imunologia , Imunoterapia/métodos , Neoplasias/imunologia , Animais , Terapia Combinada , Humanos , Imunidade Inata , Terapia de Alvo Molecular , Neoplasias/terapia , Transdução de Sinais
5.
Nat Commun ; 8(1): 559, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28916785

RESUMO

Autophagy, a catabolic mechanism that involves degradation of cellular components, is essential for cell homeostasis. Although autophagy favours the lineage stability of regulatory T cells, the contribution of autophagy to the differentiation of effector CD4 T cells remains unclear. Here we show that autophagy selectively represses T helper 9 (TH9) cell differentiation. CD4 T cells lacking Atg3 or Atg5 have increased interleukin-9 (IL-9) expression upon differentiation into TH9 cells relative to Atg3- or Atg5-expressing control cells. In addition, the TH9 cell transcription factor, PU.1, undergoes K63 ubiquitination and degradation through p62-dependent selective autophagy. Finally, the blockade of autophagy enhances TH9 cell anticancer functions in vivo, and mice with T cell-specific deletion of Atg5 have reduced tumour outgrowth in an IL-9-dependent manner. Overall, our findings reveal an unexpected function of autophagy in the modulation of TH9 cell differentiation and antitumour activity, and prompt potential autophagy-dependent modulations of TH9 activity for cancer immunotherapy.Autophagy is a cellular process for recycling cell constituents, and is essential for T cell activation, but its function in T cell polarization is still unclear. Here the authors show that autophagy induces the degradation of transcription factor PU.1 to negatively modulate TH9 homeostasis and antitumour immunity.


Assuntos
Autofagia/imunologia , Interleucina-9/imunologia , Linfopoese/imunologia , Neoplasias/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Transativadores/metabolismo , Animais , Proteína 5 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Diferenciação Celular/imunologia , Imunoterapia , Camundongos , Neoplasias/terapia , Enzimas de Conjugação de Ubiquitina
6.
Curr Opin Pharmacol ; 35: 83-88, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28551360

RESUMO

Impressive remissions driven by immunological checkpoint blockade in cancer patients have prompted the scientific community to investigate afresh the crosstalk between cancer cells and the patient's immune system. Preclinical and clinical studies have highlighted that the anticancer efficacy of some conventional chemotherapeutics is based on their ability to restore anticancer immune responses. The current challenge is to understand and circumvent immune resistance mechanisms to chemo- and immunotherapies to design relevant immunotherapy and chemotherapy combinations. In this review, we will summarize which immunological processes are involved in the anticancer action of some cytotoxic agents and discuss the recently unraveled contribution of the gut microbiota into the immunogenicity of anticancer drugs. We will eventually introduce the scientific rationale for therapeutic strategies combining chemotherapeutic drugs and immune checkpoint blockers.


Assuntos
Antineoplásicos/uso terapêutico , Imunoterapia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Animais , Microbioma Gastrointestinal/imunologia , Humanos
7.
Eur J Cancer ; 75: 86-97, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28219022

RESUMO

The efficacy of checkpoint inhibitor therapy illustrates that cancer immunotherapy, which aims to foster the host immune response against cancer to achieve durable anticancer responses, can be successfully implemented in a routine clinical practice. However, a substantial proportion of patients does not benefit from this treatment, underscoring the need to identify alternative strategies to defeat cancer. Despite the demonstration in the 1990's that the detection of danger signals, including the nucleic acids DNA and RNA, by dendritic cells (DCs) in a cancer setting is essential for eliciting host defence, the molecular sensors responsible for recognising these danger signals and eliciting anticancer immune responses remain incompletely characterised, possibly explaining the disappointing results obtained so far upon the clinical implementation of DC-based cancer vaccines. In 2008, STING (stimulator of interferon genes), was identified as a protein that is indispensable for the recognition of cytosolic DNA. The central role of STING in controlling anticancer immune responses was exemplified by observations that spontaneous and radiation-induced adaptive anticancer immunity was reduced in the absence of STING, illustrating the potential of STING-targeting for cancer immunotherapy. Here, we will discuss the relevance of manipulating the STING signalling pathway for cancer treatment and integrating STING-targeting based strategies into combinatorial therapies to obtain long-lasting anticancer immune responses.


Assuntos
Imunoterapia/métodos , Interferons/genética , Proteínas de Membrana/fisiologia , Neoplasias/terapia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Carcinogênese/imunologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , DNA/efeitos dos fármacos , DNA/imunologia , Células Dendríticas/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Proteínas de Membrana/agonistas , Proteínas de Membrana/metabolismo , Camundongos , Neoplasias/genética , Transdução de Sinais
8.
Semin Immunopathol ; 39(1): 39-46, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27832300

RESUMO

IL-9 was initially identified as a T cell growth factor with a potential oncogenic activity. Accordingly, IL-9 drives tumor growth in most hematological cancers. However, the links between IL-9 and cancer progression have been recently revisited following the discovery of TH9 cells. TH9 cells, which have been characterized in 2008 as a proinflammatory CD4 T cell subset that promotes protection against parasites and drives tissue inflammation in colitis, actually harbor potent IL-9-dependent anti-cancer properties in solid tumors and especially melanoma. While the molecular mechanisms underlying these observations are still being investigated, TH9 cells were demonstrated to activate both innate and adaptive immune responses, thereby favoring anti-cancer immunity and tumor elimination. Human TH9 cells have also been identified in cancer tissues, but their functions remain elusive. The present review aims to discuss the anti-cancer potential of TH9 cells and their possible clinical relevance for cancer immunotherapy.


Assuntos
Imunidade , Interleucina-9/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Imunidade Adaptativa , Animais , Citocinas/genética , Citocinas/metabolismo , Citotoxicidade Imunológica , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade Inata , Imunomodulação , Interleucina-9/genética , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Neoplasias/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA