Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Trends Cancer ; 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39341696

RÉSUMÉ

Ovarian cancer (OC) represents ecosystems of highly diverse tumor microenvironments (TMEs). The presence of tumor-infiltrating lymphocytes (TILs) is linked to enhanced immune responses and long-term survival. In this review we present emerging evidence suggesting that cellular crosstalk tightly regulates the distribution of TILs within the TME, underscoring the need to better understand key cellular networks that promote or impede T cell infiltration in OC. We also capture the emergent methodologies and computational techniques that enable the dissection of cell-cell crosstalk. Finally, we present innovative ex vivo TME models that can be leveraged to map and perturb cellular communications to enhance T cell infiltration and immune reactivity.

2.
Nat Rev Clin Oncol ; 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39232212

RÉSUMÉ

Despite documented evidence that ovarian cancer cells express immune-checkpoint molecules, such as PD-1 and PD-L1, and of a positive correlation between the presence of tumour-infiltrating lymphocytes and favourable overall survival outcomes in patients with this tumour type, the results of trials testing immune-checkpoint inhibitors (ICIs) in these patients thus far have been disappointing. The lack of response to ICIs can be attributed to tumour heterogeneity as well as inherent or acquired resistance associated with the tumour microenvironment (TME). Understanding tumour immunobiology, discovering biomarkers for patient selection and establishing optimal treatment combinations remains the hope but also a key challenge for the future application of immunotherapy in ovarian cancer. In this Review, we summarize results from trials testing ICIs in patients with ovarian cancer. We propose the implementation of a systematic CD8+ T cell-based immunophenotypic classification of this malignancy, followed by discussions of the preclinical data providing the basis to treat such immunophenotypes with combination immunotherapies. We posit that the integration of an accurate TME immunophenotype characterization with genetic data can enable the design of tailored therapeutic approaches and improve patient recruitment in clinical trials. Lastly, we propose a roadmap incorporating tissue-based profiling to guide future trials testing adoptive cell therapy approaches and assess novel immunotherapy combinations while promoting collaborative research.

3.
Nature ; 629(8011): 426-434, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38658764

RÉSUMÉ

Expansion of antigen-experienced CD8+ T cells is critical for the success of tumour-infiltrating lymphocyte (TIL)-adoptive cell therapy (ACT) in patients with cancer1. Interleukin-2 (IL-2) acts as a key regulator of CD8+ cytotoxic T lymphocyte functions by promoting expansion and cytotoxic capability2,3. Therefore, it is essential to comprehend mechanistic barriers to IL-2 sensing in the tumour microenvironment to implement strategies to reinvigorate IL-2 responsiveness and T cell antitumour responses. Here we report that prostaglandin E2 (PGE2), a known negative regulator of immune response in the tumour microenvironment4,5, is present at high concentrations in tumour tissue from patients and leads to impaired IL-2 sensing in human CD8+ TILs via the PGE2 receptors EP2 and EP4. Mechanistically, PGE2 inhibits IL-2 sensing in TILs by downregulating the IL-2Rγc chain, resulting in defective assembly of IL-2Rß-IL2Rγc membrane dimers. This results in impaired IL-2-mTOR adaptation and PGC1α transcriptional repression, causing oxidative stress and ferroptotic cell death in tumour-reactive TILs. Inhibition of PGE2 signalling to EP2 and EP4 during TIL expansion for ACT resulted in increased IL-2 sensing, leading to enhanced proliferation of tumour-reactive TILs and enhanced tumour control once the cells were transferred in vivo. Our study reveals fundamental features that underlie impairment of human TILs mediated by PGE2 in the tumour microenvironment. These findings have therapeutic implications for cancer immunotherapy and cell therapy, and enable the development of targeted strategies to enhance IL-2 sensing and amplify the IL-2 response in TILs, thereby promoting the expansion of effector T cells with enhanced therapeutic potential.


Sujet(s)
Lymphocytes T CD8+ , Prolifération cellulaire , Dinoprostone , Interleukine-2 , Lymphocytes TIL , Mitochondries , Transduction du signal , Animaux , Humains , Souris , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Dinoprostone/métabolisme , Régulation négative , Ferroptose , Sous-unité gamma commune aux récepteurs des interleukines/biosynthèse , Sous-unité gamma commune aux récepteurs des interleukines/déficit , Sous-unité gamma commune aux récepteurs des interleukines/métabolisme , Interleukine-2/antagonistes et inhibiteurs , Interleukine-2/immunologie , Interleukine-2/métabolisme , Sous-unité bêta du récepteur à l'interleukine-2/métabolisme , Lymphocytes TIL/cytologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Mitochondries/métabolisme , Stress oxydatif , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/antagonistes et inhibiteurs , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/métabolisme , Microenvironnement tumoral/immunologie
4.
Nature ; 629(8011): 417-425, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38658748

RÉSUMÉ

Cancer-specific TCF1+ stem-like CD8+ T cells can drive protective anticancer immunity through expansion and effector cell differentiation1-4; however, this response is dysfunctional in tumours. Current cancer immunotherapies2,5-9 can promote anticancer responses through TCF1+ stem-like CD8+ T cells in some but not all patients. This variation points towards currently ill-defined mechanisms that limit TCF1+CD8+ T cell-mediated anticancer immunity. Here we demonstrate that tumour-derived prostaglandin E2 (PGE2) restricts the proliferative expansion and effector differentiation of TCF1+CD8+ T cells within tumours, which promotes cancer immune escape. PGE2 does not affect the priming of TCF1+CD8+ T cells in draining lymph nodes. PGE2 acts through EP2 and EP4 (EP2/EP4) receptor signalling in CD8+ T cells to limit the intratumoural generation of early and late effector T cell populations that originate from TCF1+ tumour-infiltrating CD8+ T lymphocytes (TILs). Ablation of EP2/EP4 signalling in cancer-specific CD8+ T cells rescues their expansion and effector differentiation within tumours and leads to tumour elimination in multiple mouse cancer models. Mechanistically, suppression of the interleukin-2 (IL-2) signalling pathway underlies the PGE2-mediated inhibition of TCF1+ TIL responses. Altogether, we uncover a key mechanism that restricts the IL-2 responsiveness of TCF1+ TILs and prevents anticancer T cell responses that originate from these cells. This study identifies the PGE2-EP2/EP4 axis as a molecular target to restore IL-2 responsiveness in anticancer TILs to achieve cancer immune control.


Sujet(s)
Lymphocytes T CD8+ , Prolifération cellulaire , Dinoprostone , Lymphocytes TIL , Tumeurs , Cellules souches , Échappement de la tumeur à la surveillance immunitaire , Animaux , Femelle , Humains , Mâle , Souris , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Différenciation cellulaire , Lignée cellulaire tumorale , Dinoprostone/métabolisme , Modèles animaux de maladie humaine , Facteur nucléaire hépatocytaire HNF-1 alpha/métabolisme , Interleukine-2 , Noeuds lymphatiques/cytologie , Noeuds lymphatiques/immunologie , Lymphocytes TIL/cytologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Souris de lignée C57BL , Tumeurs/immunologie , Tumeurs/prévention et contrôle , Sous-type EP2 des récepteurs des prostaglandines E/déficit , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/déficit , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Transduction du signal , Cellules souches/cytologie , Cellules souches/immunologie , Cellules souches/métabolisme , Échappement de la tumeur à la surveillance immunitaire/immunologie
5.
Sci Immunol ; 9(92): eadg7995, 2024 Feb 02.
Article de Anglais | MEDLINE | ID: mdl-38306416

RÉSUMÉ

Adoptive cell therapy (ACT) using ex vivo-expanded tumor-infiltrating lymphocytes (TILs) can eliminate or shrink metastatic melanoma, but its long-term efficacy remains limited to a fraction of patients. Using longitudinal samples from 13 patients with metastatic melanoma treated with TIL-ACT in a phase 1 clinical study, we interrogated cellular states within the tumor microenvironment (TME) and their interactions. We performed bulk and single-cell RNA sequencing, whole-exome sequencing, and spatial proteomic analyses in pre- and post-ACT tumor tissues, finding that ACT responders exhibited higher basal tumor cell-intrinsic immunogenicity and mutational burden. Compared with nonresponders, CD8+ TILs exhibited increased cytotoxicity, exhaustion, and costimulation, whereas myeloid cells had increased type I interferon signaling in responders. Cell-cell interaction prediction analyses corroborated by spatial neighborhood analyses revealed that responders had rich baseline intratumoral and stromal tumor-reactive T cell networks with activated myeloid populations. Successful TIL-ACT therapy further reprogrammed the myeloid compartment and increased TIL-myeloid networks. Our systematic target discovery study identifies potential T-myeloid cell network-based biomarkers that could improve patient selection and guide the design of ACT clinical trials.


Sujet(s)
Immunothérapie adoptive , Mélanome , Humains , Mélanome/génétique , Lymphocytes TIL/métabolisme , Protéomique , Lymphocytes T CD8+/métabolisme , Microenvironnement tumoral
6.
Cancer Cell ; 39(12): 1623-1642.e20, 2021 12 13.
Article de Anglais | MEDLINE | ID: mdl-34739845

RÉSUMÉ

The mechanisms regulating exhaustion of tumor-infiltrating lymphocytes (TIL) and responsiveness to PD-1 blockade remain partly unknown. In human ovarian cancer, we show that tumor-specific CD8+ TIL accumulate in tumor islets, where they engage antigen and upregulate PD-1, which restrains their functions. Intraepithelial PD-1+CD8+ TIL can be, however, polyfunctional. PD-1+ TIL indeed exhibit a continuum of exhaustion states, with variable levels of CD28 costimulation, which is provided by antigen-presenting cells (APC) in intraepithelial tumor myeloid niches. CD28 costimulation is associated with improved effector fitness of exhausted CD8+ TIL and is required for their activation upon PD-1 blockade, which also requires tumor myeloid APC. Exhausted TIL lacking proper CD28 costimulation in situ fail to respond to PD-1 blockade, and their response may be rescued by local CTLA-4 blockade and tumor APC stimulation via CD40L.


Sujet(s)
Cellules présentatrices d'antigène/métabolisme , Antigène CD28/métabolisme , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Cellules myéloïdes/métabolisme , Tumeurs/traitement médicamenteux , Niche de cellules souches/génétique , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs/immunologie
7.
J Immunother Cancer ; 8(2)2020 08.
Article de Anglais | MEDLINE | ID: mdl-32817208

RÉSUMÉ

BACKGROUND: Novel therapeutic strategies in ovarian cancer (OC) are needed as the survival rate remains dismally low. Although dendritic cell-based cancer vaccines are effective in eliciting therapeutic responses, their complex and costly manufacturing process hampers their full clinical utility outside specialized clinics. Here, we describe a novel approach of generating a rapid and effective cancer vaccine using ascites-derived monocytes for treating OC. METHODS: Using the ID8 mouse ovarian tumor model and OC patient samples, we isolated ascites monocytes and evaluated them with flow cytometry, Luminex cytokine and chemokine array analysis, ex vivo cocultures with T cells, in vivo tumor challenge and T cell transfer experiments, RNA-sequencing and mass spectrometry. RESULTS: We demonstrated the feasibility of isolating ascites monocytes and restoring their ability to function as bona fide antigen-presenting cells (APCs) with Toll-like receptor (TLR) 4 lipopolysaccharide and TLR9 CpG-oligonucleotides, and a blocking antibody to interleukin-10 receptor (IL-10R Ab) in the ID8 model. The ascites monocytes were laden with tumor antigens at a steady state in vivo. After a short 48 hours activation, they upregulated maturation markers (CD80, CD86 and MHC class I) and demonstrated strong ex vivo T cell stimulatory potential and effectively suppressed tumor and malignant ascites in vivo. They also induced protective long-term T cell memory responses. To evaluate the translational potential of this approach, we isolated ascites monocytes from stage III/IV chemotherapy-naïve OC patients. Similarly, the human ascites monocytes presented tumor-associated antigens (TAAs), including MUC1, ERBB2, mesothelin, MAGE, PRAME, GPC3, PMEL and TP53 at a steady state. After a 48-hour treatment with TLR4 and IL-10R Ab, they efficiently stimulated oligoclonal tumor-associated lymphocytes (TALs) with strong reactivity against TAAs. Importantly, the activated ascites monocytes retained their ability to activate TALs in the presence of ascitic fluid. CONCLUSIONS: Ascites monocytes are naturally loaded with tumor antigen and can perform as potent APCs following short ex vivo activation. This novel ascites APC vaccine can be rapidly prepared in 48 hours with a straightforward and affordable manufacturing process, and would be an attractive therapeutic vaccine for OC.


Sujet(s)
Ascites/physiopathologie , Vaccins anticancéreux/immunologie , Monocytes/métabolisme , Tumeurs de l'ovaire/immunologie , Récepteurs de type Toll/immunologie , Animaux , Femelle , Humains , Mésothéline , Souris , Tumeurs de l'ovaire/mortalité , Analyse de survie
8.
Curr Opin Biotechnol ; 65: 190-196, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32334152

RÉSUMÉ

New treatments are urgently needed in patients with ovarian cancer (OC), as diagnosis is delayed in many instances, resulting in 85% recurrence of the disease following surgery and standard chemotherapy. OC is considered to be an immunological type of cancer, despite its limited response to current immunotherapy options, including vaccination. Thus, additional interventions may improve their efficacy. Dendritic cells (DCs) are the most widely used cellular vaccination therapy in patients with OC due to their crucial role in the initiation and development of immune response. There are viable options for DC-vaccination with a favorable toxicity profile, but specific alternatives should consider the limited therapeutic effectiveness of DC-vaccination in OC treatment. In this respect, B-cells and macrophages provide additional possibilities that may be explored for immunotherapy. Here we consider the current state-of-the-art of immunotherapy strategies for OC treatment and evaluate their potential for future improvements.


Sujet(s)
Vaccins anticancéreux , Tumeurs de l'ovaire , Vaccins , Vaccins anticancéreux/usage thérapeutique , Cellules dendritiques , Femelle , Humains , Immunothérapie , Tumeurs de l'ovaire/thérapie
9.
Cancer Cell ; 35(6): 885-900.e10, 2019 06 10.
Article de Anglais | MEDLINE | ID: mdl-31185212

RÉSUMÉ

We investigated the role of chemokines in regulating T cell accumulation in solid tumors. CCL5 and CXCL9 overexpression was associated with CD8+ T cell infiltration in solid tumors. T cell infiltration required tumor cell-derived CCL5 and was amplified by IFN-γ-inducible, myeloid cell-secreted CXCL9. CCL5 and CXCL9 coexpression revealed immunoreactive tumors with prolonged survival and response to checkpoint blockade. Loss of CCL5 expression in human tumors was associated with epigenetic silencing through DNA methylation. Reduction of CCL5 expression caused tumor-infiltrating lymphocyte (TIL) desertification, whereas forced CCL5 expression prevented Cxcl9 expression and TILs loss, and attenuated tumor growth in mice through IFN-γ. The cooperation between tumor-derived CCL5 and IFN-γ-inducible CXCR3 ligands secreted by myeloid cells is key for orchestrating T cell infiltration in immunoreactive and immunoresponsive tumors.


Sujet(s)
Lymphocytes T CD8+/métabolisme , Chimiotaxie des leucocytes , Cytokines/métabolisme , Cellules dendritiques/métabolisme , Activation des lymphocytes , Lymphocytes TIL/métabolisme , Macrophages/métabolisme , Tumeurs de l'ovaire/métabolisme , Animaux , Antinéoplasiques immunologiques/pharmacologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Chimiokine CCL5/génétique , Chimiokine CCL5/immunologie , Chimiokine CCL5/métabolisme , Chimiokine CXCL9/génétique , Chimiokine CXCL9/immunologie , Chimiokine CXCL9/métabolisme , Chimiotaxie des leucocytes/effets des médicaments et des substances chimiques , Techniques de coculture , Cytokines/génétique , Cytokines/immunologie , Méthylation de l'ADN , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Épigenèse génétique , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Immunothérapie/méthodes , Interféron gamma/génétique , Interféron gamma/immunologie , Interféron gamma/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/immunologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Souris de lignée C57BL , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/thérapie , Communication paracrine , Récepteurs CXCR3/génétique , Récepteurs CXCR3/immunologie , Récepteurs CXCR3/métabolisme , Transduction du signal
10.
Vaccine ; 34(21): 2453-2459, 2016 05 05.
Article de Anglais | MEDLINE | ID: mdl-27016652

RÉSUMÉ

Subunit vaccines, employing purified protein antigens rather than intact pathogens, require the addition of adjuvants for enhanced immunogenicity with a correct balance between strong activation of the immune system and low toxicity. Here we show that the endogenous (i.e., autologous) non-toxic TLR4 agonist extra domain A type III repeat of fibronectin (FNIII EDA) can synergize with the exogenous (i.e., bacterial), toxic-at-high-dose, TLR9 agonist CpG to induce efficient cellular immune responses while keeping the dose of CpG low. The efficacy of the combined TLR agonists, even at half-doses, led to stronger dendritic cell activation, enhanced cytotoxic T lymphocyte activation as well as stronger humoral response, compared to the individual agonists given at full doses. Immune cells induced after vaccination with the co-adjuvanted formulation could mediate tumor regression in an E.G7-OVA tumor model, and eradicate circulating hepatitis B virus (HBV) in a transgenic HBV model. Together, these results show that endogenous TLR agonists, such as variants of FNIII EDA, can synergize with exogenous TLR ligands, such as CpG, and strongly enhance cellular immune responses, while improving their safety profile.


Sujet(s)
Vaccins anticancéreux/immunologie , Fibronectines/immunologie , Vaccins anti-hépatite B/immunologie , Oligodésoxyribonucléotides/immunologie , Lymphocytes T cytotoxiques/immunologie , Lymphocytes auxiliaires Th1/immunologie , Récepteur de type Toll-4/agonistes , Adjuvants immunologiques/administration et posologie , Animaux , Vaccins anticancéreux/administration et posologie , Cellules dendritiques/immunologie , Modèles animaux de maladie humaine , Fibronectines/composition chimique , Hépatite B/immunologie , Hépatite B/virologie , Vaccins anti-hépatite B/administration et posologie , Virus de l'hépatite B/immunologie , Immunité cellulaire , Immunité humorale , Souris , Souris transgéniques , Récepteurs de reconnaissance de motifs moléculaires , Récepteur de type Toll-4/immunologie , Récepteur-9 de type Toll-like/agonistes , Récepteur-9 de type Toll-like/immunologie , Vaccination
11.
Sci Rep ; 5: 15907, 2015 Oct 29.
Article de Anglais | MEDLINE | ID: mdl-26511151

RÉSUMÉ

New approaches based on induction of antigen-specific immunological tolerance are being explored for treatment of autoimmunity and prevention of immunity to protein drugs. Antigens associated with apoptotic debris are known to be processed tolerogenically in vivo. Our group is exploring an approach toward antigen-specific tolerization using erythrocyte-binding antigens, based on the premise that as the erythrocytes circulate, age and are cleared, the erythrocyte surface-bound antigen payload will be cleared tolerogenically along with the eryptotic debris. Here, we characterized the phenotypic signatures of CD8+ T cells undergoing tolerance in response to soluble and erythrocyte-targeted antigen. Signaling through programmed death-1/programmed death ligand-1 (PD-1/PD-L1), but not through cytotoxic T lymphocyte antigen 4 (CTLA4), was shown to be required for antigen-specific T cell deletion, anergy and expression of regulatory markers. Generation of CD25+FOXP3+ regulatory T cells in response to erythrocyte-targeted antigens but not soluble antigen at an equimolar dose was observed, and these cells were required for long-term maintenance of immune tolerance in both the CD4+ and CD8+ T cell compartments. Evidence of infectious tolerance was observed, in that tolerance to a one antigenic epitope was able to regulate responses to other epitopes in the same protein antigen.


Sujet(s)
Antigènes/immunologie , Érythrocytes/immunologie , Tolérance immunitaire , Mémoire immunologique , Lymphocytes T régulateurs/immunologie , Animaux , Lymphocytes T CD8+/immunologie , Souris
12.
Curr Opin Immunol ; 35: 80-8, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26163377

RÉSUMÉ

Unwanted immunity develops in response to many protein drugs, in autoimmunity, in allergy, and in transplantation. Approaches to induce immunological tolerance aim to either prevent these responses or reverse them after they have already taken place. We present here recent developments in approaches, based on engineered peptides, proteins and biomaterials, that harness mechanisms of peripheral tolerance both prophylactically and therapeutically to induce antigen-specific immunological tolerance. These mechanisms are based on responses of B and T lymphocytes to other cells in their immune environment that result in cellular deletion or ignorance to particular antigens, or in development of active immune regulatory responses. Several of these approaches are moving toward clinical development, and some are already in early stages of clinical testing.


Sujet(s)
Lymphocytes B/immunologie , Tolérance périphérique , Lymphocytes T/immunologie , Animaux , Présentation d'antigène , Apoptose , Épitopes/métabolisme , Humains , Ingénierie des protéines , Interactions entre récepteurs , Récepteurs pour l'antigène des lymphocytes B/métabolisme , Spécificité antigénique des récepteurs des lymphocytes T
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE