Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 147
Filtrer
1.
Cell ; 187(11): 2690-2702.e17, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38723627

RÉSUMÉ

The quality and quantity of tumor-infiltrating lymphocytes, particularly CD8+ T cells, are important parameters for the control of tumor growth and response to immunotherapy. Here, we show in murine and human cancers that these parameters exhibit circadian oscillations, driven by both the endogenous circadian clock of leukocytes and rhythmic leukocyte infiltration, which depends on the circadian clock of endothelial cells in the tumor microenvironment. To harness these rhythms therapeutically, we demonstrate that efficacy of chimeric antigen receptor T cell therapy and immune checkpoint blockade can be improved by adjusting the time of treatment during the day. Furthermore, time-of-day-dependent T cell signatures in murine tumor models predict overall survival in patients with melanoma and correlate with response to anti-PD-1 therapy. Our data demonstrate the functional significance of circadian dynamics in the tumor microenvironment and suggest the importance of leveraging these features for improving future clinical trial design and patient care.


Sujet(s)
Lymphocytes T CD8+ , Immunothérapie , Lymphocytes TIL , Souris de lignée C57BL , Microenvironnement tumoral , Animaux , Humains , Souris , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Horloges circadiennes , Rythme circadien , Cellules endothéliales/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Immunothérapie/méthodes , Lymphocytes TIL/immunologie , Mélanome/immunologie , Mélanome/thérapie , Mélanome/anatomopathologie , Microenvironnement tumoral/immunologie
3.
Adv Sci (Weinh) ; 11(15): e2309026, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38342608

RÉSUMÉ

Tumor-associated macrophages (TAM) are a diverse population of myeloid cells that are often abundant and immunosuppressive in human cancers. CXCL9Hi TAM has recently been described to have an antitumor phenotype and is linked to immune checkpoint response. Despite the emerging understanding of the unique antitumor TAM phenotype, there is a lack of TAM-specific therapeutics to exploit this new biological understanding. Here, the discovery and characterization of multiple small-molecule enhancers of chemokine ligand 9 (CXCL9) and their targeted delivery in a TAM-avid systemic nanoformulation is reported. With this strategy, it is efficient encapsulation and release of multiple drug loads that can efficiently induce CXCL9 expression in macrophages, both in vitro and in vivo in a mouse tumor model. These observations provide a window into the molecular features that define TAM-specific states, an insight a novel therapeutic anticancer approach is used to discover.


Sujet(s)
Tumeurs , Macrophages associés aux tumeurs , Animaux , Humains , Souris , Chimiokine CXCL9/génétique , Chimiokine CXCL9/métabolisme , Macrophages/métabolisme , Tumeurs/anatomopathologie , Phénotype
4.
Immunity ; 56(10): 2218-2230, 2023 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-37708889

RÉSUMÉ

In cancer patients, dendritic cells (DCs) in tumor-draining lymph nodes can present antigens to naive T cells in ways that break immunological tolerance. The clonally expanded progeny of primed T cells are further regulated by DCs at tumor sites. Intratumoral DCs can both provide survival signals to and drive effector differentiation of incoming T cells, thereby locally enhancing antitumor immunity; however, the paucity of intratumoral DCs or their expression of immunoregulatory molecules often limits antitumor T cell responses. Here, we review the current understanding of DC-T cell interactions at both priming and effector sites of immune responses. We place emerging insights into DC functions in tumor immunity in the context of DC development, ontogeny, and functions in other settings and propose that DCs control at least two T cell-associated checkpoints of the cancer immunity cycle. Our understanding of both checkpoints has implications for the development of new approaches to cancer immunotherapy.

5.
Science ; 381(6657): 515-524, 2023 08 04.
Article de Anglais | MEDLINE | ID: mdl-37535729

RÉSUMÉ

Tumor microenvironments (TMEs) influence cancer progression but are complex and often differ between patients. Considering that microenvironment variations may reveal rules governing intratumoral cellular programs and disease outcome, we focused on tumor-to-tumor variation to examine 52 head and neck squamous cell carcinomas. We found that macrophage polarity-defined by CXCL9 and SPP1 (CS) expression but not by conventional M1 and M2 markers-had a noticeably strong prognostic association. CS macrophage polarity also identified a highly coordinated network of either pro- or antitumor variables, which involved each tumor-associated cell type and was spatially organized. We extended these findings to other cancer indications. Overall, these results suggest that, despite their complexity, TMEs coordinate coherent responses that control human cancers and for which CS macrophage polarity is a relevant yet simple variable.


Sujet(s)
Polarité de la cellule , Chimiokine CXCL9 , Tumeurs de la tête et du cou , Macrophages , Ostéopontine , Carcinome épidermoïde de la tête et du cou , Microenvironnement tumoral , Humains , Chimiokine CXCL9/analyse , Chimiokine CXCL9/métabolisme , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/anatomopathologie , Macrophages/immunologie , Ostéopontine/analyse , Ostéopontine/métabolisme , Pronostic , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Polarité de la cellule/immunologie
6.
Int J Cancer ; 153(9): 1568-1578, 2023 11 01.
Article de Anglais | MEDLINE | ID: mdl-37306359

RÉSUMÉ

The spatial distribution of tumor-infiltrating lymphocytes (TIL) predicts breast cancer outcome and response to systemic therapy, highlighting the importance of an intact tissue structure for characterizing tumors. Here, we present ST-FFPE, a spatial transcriptomics method for the analysis of formalin-fixed paraffin-embedded samples, which opens the possibility of interrogating archival tissue. The method involves extraction, exome capture and sequencing of RNA from different tumor compartments microdissected by laser-capture, and can be used to study the cellular composition of tumor microenvironment. Focusing on triple-negative breast cancer (TNBC), we characterized T cells, B cells, dendritic cells, fibroblasts and endothelial cells in both stromal and intra-epithelial compartments. We found a highly variable spatial distribution of immune cell subsets among tumors. This analysis revealed that the immune repertoires of intra-epithelial T and B cells were consistently less diverse and more clonal than those of stromal T and B cells. T-cell receptor (TCR) sequencing confirmed a reduced diversity and higher clonality of intra-epithelial T cells relative to the corresponding stromal T cells. Analysis of the top 10 dominant clonotypes in the two compartments showed a majority of shared but also some unique clonotypes both in stromal and intra-epithelial T cells. Hyperexpanded clonotypes were more abundant among intra-epithelial than stromal T cells. These findings validate the ST-FFPE method and suggest an accumulation of antigen-specific T cells within tumor core. Because ST-FFPE is applicable for analysis of previously collected tissue samples, it could be useful for rapid assessment of intratumoral cellular heterogeneity in multiple disease and treatment settings.


Sujet(s)
Tumeurs du sein , Humains , Femelle , Tumeurs du sein/génétique , Cellules endothéliales , Transcriptome , Récepteurs aux antigènes des cellules T , Analyse de profil d'expression de gènes , Lymphocytes TIL , Microenvironnement tumoral/génétique
7.
Immunity ; 56(5): 1152, 2023 May 09.
Article de Anglais | MEDLINE | ID: mdl-37163985
8.
J Immunother Cancer ; 11(5)2023 05.
Article de Anglais | MEDLINE | ID: mdl-37208130

RÉSUMÉ

BACKGROUND: Although immune checkpoint inhibitors have been a breakthrough in clinical oncology, these therapies fail to produce durable responses in a significant fraction of patients. This lack of long-term efficacy may be due to a poor pre-existing network linking innate and adaptive immunity. Here, we present an antisense oligonucleotide (ASO)-based strategy that dually targets toll-like receptor 9 (TLR9) and programmed cell death ligand 1 (PD-L1), aiming to overcome resistance to anti-PD-L1 monoclonal therapy. METHODS: We designed a high-affinity immunomodulatory IM-TLR9:PD-L1-ASO antisense oligonucleotide (hereafter, IM-T9P1-ASO) targeting mouse PD-L1 messenger RNA and activating TLR9. Then, we performed in vitro and in vivo studies to validate the IM-T9P1-ASO activity, efficacy, and biological effects in tumors and draining lymph nodes. We also performed intravital imaging to study IM-T9P1-ASO pharmacokinetics in the tumor. RESULTS: IM-T9P1-ASO therapy, unlike PD-L1 antibody therapy, results in durable antitumor responses in multiple mouse cancer models. Mechanistically, IM-T9P1-ASO activates a state of tumor-associated dendritic cells (DCs), referred to here as DC3s, which have potent antitumor potential but express the PD-L1 checkpoint. IM-T9P1-ASO has two roles: it triggers the expansion of DC3s by engaging with TLR9 and downregulates PD-L1, thereby unleashing the antitumor functions of DC3s. This dual action leads to tumor rejection by T cells. The antitumor efficacy of IM-T9P1-ASO depends on the antitumor cytokine interleukin-12 (IL-12), produced by DC3s, and Batf3, a transcription factor required for DC development. CONCLUSIONS: By simultaneously targeting TLR9 and PD-L1, IM-T9P1-ASO amplifies antitumor responses via DC activation, leading to sustained therapeutic efficacy in mice. By highlighting differences and similarities between mouse and human DCs, this study could serve to develop similar therapeutic strategies for patients with cancer.


Sujet(s)
Tumeurs , Récepteur-9 de type Toll-like , Humains , Souris , Animaux , Récepteur-9 de type Toll-like/métabolisme , Immunothérapie/méthodes , Tumeurs/traitement médicamenteux , Oligonucléotides antisens , Cellules dendritiques
9.
Cell ; 186(7): 1448-1464.e20, 2023 03 30.
Article de Anglais | MEDLINE | ID: mdl-37001504

RÉSUMÉ

Neutrophils accumulate in solid tumors, and their abundance correlates with poor prognosis. Neutrophils are not homogeneous, however, and could play different roles in cancer therapy. Here, we investigate the role of neutrophils in immunotherapy, leading to tumor control. We show that successful therapies acutely expanded tumor neutrophil numbers. This expansion could be attributed to a Sellhi state rather than to other neutrophils that accelerate tumor progression. Therapy-elicited neutrophils acquired an interferon gene signature, also seen in human patients, and appeared essential for successful therapy, as loss of the interferon-responsive transcription factor IRF1 in neutrophils led to failure of immunotherapy. The neutrophil response depended on key components of anti-tumor immunity, including BATF3-dependent DCs, IL-12, and IFNγ. In addition, we found that a therapy-elicited systemic neutrophil response positively correlated with disease outcome in lung cancer patients. Thus, we establish a crucial role of a neutrophil state in mediating effective cancer therapy.


Sujet(s)
Tumeurs du poumon , Granulocytes neutrophiles , Humains , Tumeurs du poumon/génétique , Transduction du signal/génétique , Immunothérapie , Interférons
10.
Clin Cancer Res ; 29(8): 1605-1619, 2023 04 14.
Article de Anglais | MEDLINE | ID: mdl-36749873

RÉSUMÉ

PURPOSE: Adding losartan (LOS) to FOLFIRINOX (FFX) chemotherapy followed by chemoradiation (CRT) resulted in 61% R0 surgical resection in our phase II trial in patients with locally advanced pancreatic cancer (LAPC). Here we identify potential mechanisms of benefit by assessing the effects of neoadjuvant LOS on the tumor microenvironment. EXPERIMENTAL DESIGN: We performed a gene expression and immunofluorescence (IF) analysis using archived surgical samples from patients treated with LOS+FFX+CRT (NCT01821729), FFX+CRT (NCT01591733), or surgery upfront, without any neoadjuvant therapy. We also conducted a longitudinal analysis of multiple biomarkers in the plasma of treated patients. RESULTS: In comparison with FFX+CRT, LOS+FFX+CRT downregulated immunosuppression and pro-invasion genes. Overall survival (OS) was associated with dendritic cell (DC) and antigen presentation genes for patients treated with FFX+CRT, and with immunosuppression and invasion genes or DC- and blood vessel-related genes for those treated with LOS+FFX+CRT. Furthermore, LOS induced specific changes in circulating levels of IL-8, sTie2, and TGF-ß. IF revealed significantly less residual disease in lesions treated with LOS+FFX+CRT. Finally, patients with a complete/near complete pathologic response in the LOS+FFX+CRT-treated group had reduced CD4+FOXP3+ regulatory T cells (Tregs), fewer immunosuppressive FOXP3+ cancer cells (C-FOXP3), and increased CD8+ T cells in pancreatic ductal adenocarcinoma lesions. CONCLUSIONS: Adding LOS to FFX+CRT reduced pro-invasion and immunosuppression-related genes, which were associated with improved OS in patients with LAPC. Lesions from responders in the LOS+FFX+CRT-treated group had reduced Tregs, decreased C-FOXP3 and increased CD8+ T cells. These findings suggest that LOS may potentiate the benefit of FFX+CRT by reducing immunosuppression.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du pancréas , Humains , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Losartan/usage thérapeutique , Fluorouracil , Leucovorine , Traitement néoadjuvant/méthodes , Immunosuppression thérapeutique , Facteurs de transcription Forkhead/génétique , Microenvironnement tumoral/génétique
11.
Nature ; 614(7946): 136-143, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36470303

RÉSUMÉ

The process of cancer immunosurveillance is a mechanism of tumour suppression that can protect the host from cancer development throughout its lifetime1,2. However, it is unknown whether the effectiveness of cancer immunosurveillance fluctuates over a single day. Here we demonstrate that the initial time of day of tumour engraftment dictates the ensuing tumour size across mouse cancer models. Using immunodeficient mice as well as mice lacking lineage-specific circadian functions, we show that dendritic cells (DCs) and CD8+ T cells exert circadian anti-tumour functions that control melanoma volume. Specifically, we find that rhythmic trafficking of DCs to the tumour draining lymph node governs a circadian response of tumour-antigen-specific CD8+ T cells that is dependent on the circadian expression of the co-stimulatory molecule CD80. As a consequence, cancer immunotherapy is more effective when synchronized with DC functions, shows circadian outcomes in mice and suggests similar effects in humans. These data demonstrate that the circadian rhythms of anti-tumour immune components are not only critical for controlling tumour size but can also be of therapeutic relevance.


Sujet(s)
Lymphocytes T CD8+ , Rythme circadien , Cellules dendritiques , Mélanome , Animaux , Humains , Souris , Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Immunothérapie/méthodes , Mélanome/immunologie , Mélanome/anatomopathologie , Mélanome/thérapie , Souris de lignée C57BL , Antigène CD80 , Antigènes néoplasiques/immunologie , Noeuds lymphatiques , Rythme circadien/immunologie
13.
BMC Bioinformatics ; 23(1): 336, 2022 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-35963997

RÉSUMÉ

BACKGROUND: Single-cell RNA sequencing (scRNA-seq) technologies offer unique opportunities for exploring heterogeneous cell populations. However, in-depth single-cell transcriptomic characterization of complex tissues often requires profiling tens to hundreds of thousands of cells. Such large numbers of cells represent an important hurdle for downstream analyses, interpretation and visualization. RESULTS: We develop a framework called SuperCell to merge highly similar cells into metacells and perform standard scRNA-seq data analyses at the metacell level. Our systematic benchmarking demonstrates that metacells not only preserve but often improve the results of downstream analyses including visualization, clustering, differential expression, cell type annotation, gene correlation, imputation, RNA velocity and data integration. By capitalizing on the redundancy inherent to scRNA-seq data, metacells significantly facilitate and accelerate the construction and interpretation of single-cell atlases, as demonstrated by the integration of 1.46 million cells from COVID-19 patients in less than two hours on a standard desktop. CONCLUSIONS: SuperCell is a framework to build and analyze metacells in a way that efficiently preserves the results of scRNA-seq data analyses while significantly accelerating and facilitating them.


Sujet(s)
COVID-19 , Transcriptome , Analyse de regroupements , Humains , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes
14.
Nat Biotechnol ; 40(11): 1654-1662, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35654978

RÉSUMÉ

Cells in complex organisms undergo frequent functional changes, but few methods allow comprehensive longitudinal profiling of living cells. Here we introduce scission-accelerated fluorophore exchange (SAFE), a method for multiplexed temporospatial imaging of living cells with immunofluorescence. SAFE uses a rapid bioorthogonal click chemistry to remove immunofluorescent signals from the surface of labeled cells, cycling the nanomolar-concentration reagents in seconds and enabling multiple rounds of staining of the same samples. It is non-toxic and functional in both dispersed cells and intact living tissues. We demonstrate multiparameter (n ≥ 14), non-disruptive imaging of murine peripheral blood mononuclear and bone marrow cells to profile cellular differentiation. We also show longitudinal multiplexed imaging of bone marrow progenitor cells as they develop into neutrophils over 6 days and real-time multiplexed cycling of living mouse hepatic tissues. We anticipate that SAFE will find broad utility for investigating physiologic dynamics in living systems.


Sujet(s)
Colorants fluorescents , Agranulocytes , Souris , Animaux , Colorants fluorescents/composition chimique , Coloration et marquage , Imagerie optique/méthodes , Technique d'immunofluorescence
16.
Adv Biol (Weinh) ; 6(8): e2200030, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35675910

RÉSUMÉ

Treatment with checkpoint inhibitors can be extraordinarily effective in a fraction of patients, particularly those whose tumors are pre-infiltrated by T cells. In others, efficacy is considerably lower, which has led to interest in developing strategies for sensitization to immunotherapy. Using various colorectal cancer mouse models, it is shown that the use of Traf2 and Nck-interacting protein kinase inhibitors (TNIKi) unexpectedly increases tumor infiltration by PD-1+ CD8+ T cells, thus contributing to tumor control. This appears to happen by two independent mechanisms, by inducing immunogenic cell death and separately by directly activating CD8. The use of TNIKi achieves complete tumor control in 50% of mice when combined with checkpoint inhibitor targeting PD-1. These findings reveal immunogenic properties of TNIKi and indicate that the proportion of colorectal cancers responding to checkpoint therapy can be increased by combining it with immunogenic kinase inhibitors.


Sujet(s)
Lymphocytes T CD8+ , Tumeurs colorectales , Inhibiteurs de protéines kinases , Animaux , Lymphocytes T CD8+/métabolisme , Tumeurs colorectales/traitement médicamenteux , Modèles animaux de maladie humaine , Immunothérapie , Souris , Récepteur-1 de mort cellulaire programmée , Inhibiteurs de protéines kinases/pharmacologie
17.
J Exp Med ; 219(6)2022 06 06.
Article de Anglais | MEDLINE | ID: mdl-35522219

RÉSUMÉ

Neutrophils are the first responders to infection and inflammation and are thus a critical component of innate immune defense. Understanding the behavior of neutrophils as they act within various inflammatory contexts has provided insights into their role in sterile and infectious diseases; however, the field of neutrophils in cancer is comparatively young. Here, we summarize key concepts and current knowledge gaps related to the diverse roles of neutrophils throughout cancer progression. We discuss sources of neutrophil heterogeneity in cancer and provide recommendations on nomenclature for neutrophil states that are distinct in maturation and activation. We address discrepancies in the literature that highlight a need for technical standards that ought to be considered between laboratories. Finally, we review emerging questions in neutrophil biology and innate immunity in cancer. Overall, we emphasize that neutrophils are a more diverse population than previously appreciated and that their role in cancer may present novel unexplored opportunities to treat cancer.


Sujet(s)
Tumeurs , Granulocytes neutrophiles , Humains , Immunité innée , Inflammation , Tumeurs/génétique , Phénotype
19.
Nat Rev Clin Oncol ; 19(6): 402-421, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35354979

RÉSUMÉ

In the past decade, substantial advances have been made in understanding the biology of tumour-associated macrophages (TAMs), and their clinical relevance is emerging. A particular aspect that is becoming increasingly clear is that the interaction of TAMs with cancer cells and stromal cells in the tumour microenvironment enables and sustains most of the hallmarks of cancer. Therefore, manipulation of TAMs could enable improved disease control in a substantial fraction of patients across a large number of cancer types. In this Review, we examine the diversity of TAMs in various cancer indications and how this heterogeneity is being revisited with the advent of single-cell technologies, and then explore the current knowledge on the functional roles of different TAM states and the prognostic and predictive value of TAM-related signatures. We also review agents targeting TAMs that are currently being or will soon be tested in clinical trials, and how manipulations of TAMs can improve existing anticancer treatments. Finally, we discuss how TAM-targeting approaches could be further integrated into routine clinical practice, considering a precision oncology approach and viewing TAMs as a dynamic population that can evolve under treatment pressure.


Sujet(s)
Tumeurs , Macrophages associés aux tumeurs , Humains , Macrophages , Tumeurs/anatomopathologie , Tumeurs/thérapie , Médecine de précision , Microenvironnement tumoral
20.
Cancer Immunol Res ; 10(1): 40-55, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34795032

RÉSUMÉ

Macrophages often abound within tumors, express colony-stimulating factor 1 receptor (CSF1R), and are linked to adverse patient survival. Drugs blocking CSF1R signaling have been used to suppress tumor-promoting macrophage responses; however, their mechanisms of action remain incompletely understood. Here, we assessed the lung tumor immune microenvironment in mice treated with BLZ945, a prototypical small-molecule CSF1R inhibitor, using single-cell RNA sequencing and mechanistic validation approaches. We showed that tumor control was not caused by CSF1R+ cell depletion; instead, CSF1R targeting reshaped the CSF1R+ cell landscape, which unlocked cross-talk between antitumoral CSF1R- cells. These cells included IFNγ-producing natural killer and T cells, and an IL12-producing dendritic cell subset, denoted as DC3, which were all necessary for CSF1R inhibitor-mediated lung tumor control. These data indicate that CSF1R targeting can activate a cardinal cross-talk between cells that are not macrophages and that are essential to mediate the effects of T cell-targeted immunotherapies and promote antitumor immunity.See related Spotlight by Burrello and de Visser, p. 4.


Sujet(s)
Cellules dendritiques/immunologie , Immunothérapie/méthodes , Interféron gamma/métabolisme , Interleukine-12/métabolisme , Tumeurs du poumon/thérapie , Animaux , Benzothiazoles/pharmacologie , Lignée cellulaire tumorale , Femelle , Tumeurs du poumon/immunologie , Souris , Souris de lignée C57BL , Acides picoliniques/pharmacologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE