Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 14: 1166180, 2023.
Article in English | MEDLINE | ID: mdl-37622122

ABSTRACT

Conventional dendritic cells (cDCs) are at the forefront of activating the immune system to mount an anti-tumor immune response. Flt3L is a cytokine required for DC development that can increase DC abundance in the tumor when administered therapeutically. However, the impact of Flt3L on the phenotype of distinct cDC subsets in the tumor microenvironment is still largely undetermined. Here, using multi-omic single-cell analysis, we show that Flt3L therapy increases all cDC subsets in orthotopic E0771 and TS/A breast cancer and LLC lung cancer models, but this did not result in a reduction of tumor growth in any of the models. Interestingly, a CD81+migcDC1 population, likely developing from cDC1, was induced upon Flt3L treatment in E0771 tumors as well as in TS/A breast and LLC lung tumors. This CD81+migcDC1 subset is characterized by the expression of both canonical cDC1 markers as well as migratory cDC activation and regulatory markers and displayed a Treg-inducing potential. To shift the cDC phenotype towards a T-cell stimulatory phenotype, CD40 agonist therapy was administered to E0771 tumor-bearing mice in combination with Flt3L. However, while αCD40 reduced tumor growth, Flt3L failed to improve the therapeutic response to αCD40 therapy. Interestingly, Flt3L+αCD40 combination therapy increased the abundance of Treg-promoting CD81+migcDC1. Nonetheless, while Treg-depletion and αCD40 therapy were synergistic, the addition of Flt3L to this combination did not result in any added benefit. Overall, these results indicate that merely increasing cDCs in the tumor by Flt3L treatment cannot improve anti-tumor responses and therefore might not be beneficial for the treatment of cancer, though could still be of use to increase cDC numbers for autologous DC-therapy.


Subject(s)
Lung Neoplasms , T-Lymphocytes, Regulatory , Animals , Mice , Receptors, CCR7 , Lung Neoplasms/drug therapy , Combined Modality Therapy , CD40 Antigens , Tumor Microenvironment
2.
Front Immunol ; 13: 1003975, 2022.
Article in English | MEDLINE | ID: mdl-36531986

ABSTRACT

Junctional adhesion molecule-A (JAM-A), expressed on the surface of myeloid cells, is required for extravasation at sites of inflammation and may also modulate myeloid cell activation. Infiltration of myeloid cells is a common feature of tumors that drives disease progression, but the function of JAM-A in this phenomenon and its impact on tumor-infiltrating myeloid cells is little understood. Here we show that systemic cancer-associated inflammation in mice enhanced JAM-A expression selectively on circulating monocytes in an IL1ß-dependent manner. Using myeloid-specific JAM-A-deficient mice, we found that JAM-A was dispensable for recruitment of monocytes and other myeloid cells to tumors, in contrast to its reported role in inflammation. Single-cell RNA sequencing revealed that loss of JAM-A did not influence the transcriptional reprogramming of myeloid cells in the tumor microenvironment. Overall, our results support the notion that cancer-associated inflammation can modulate the phenotype of circulating immune cells, and we demonstrate that tumors can bypass the requirement of JAM-A for myeloid cell recruitment and reprogramming.


Subject(s)
Junctional Adhesion Molecule A , Mice , Animals , Tumor Microenvironment/genetics , Myeloid Cells/metabolism , Monocytes/metabolism , Inflammation/metabolism
3.
Cancer Res ; 82(20): 3785-3801, 2022 10 17.
Article in English | MEDLINE | ID: mdl-35979635

ABSTRACT

Agonistic αCD40 therapy has been shown to inhibit cancer progression in only a fraction of patients. Understanding the cancer cell-intrinsic and microenvironmental determinants of αCD40 therapy response is therefore crucial to identify responsive patient populations and to design efficient combinatorial treatments. Here, we show that the therapeutic efficacy of αCD40 in subcutaneous melanoma relies on preexisting, type 1 classical dendritic cell (cDC1)-primed CD8+ T cells. However, after administration of αCD40, cDC1s were dispensable for antitumor efficacy. Instead, the abundance of activated cDCs, potentially derived from cDC2 cells, increased and further activated antitumor CD8+ T cells. Hence, distinct cDC subsets contributed to the induction of αCD40 responses. In contrast, lung carcinomas, characterized by a high abundance of macrophages, were resistant to αCD40 therapy. Combining αCD40 therapy with macrophage depletion led to tumor growth inhibition only in the presence of strong neoantigens. Accordingly, treatment with immunogenic cell death-inducing chemotherapy sensitized lung tumors to αCD40 therapy in subcutaneous and orthotopic settings. These insights into the microenvironmental regulators of response to αCD40 suggest that different tumor types would benefit from different combinations of therapies to optimize the clinical application of CD40 agonists. SIGNIFICANCE: This work highlights the temporal roles of different dendritic cell subsets in promoting CD8+ T-cell-driven responses to CD40 agonist therapy in cancer.


Subject(s)
CD40 Antigens , Dendritic Cells , Macrophages , Neoplasms , Animals , CD40 Antigens/agonists , CD8-Positive T-Lymphocytes , Dendritic Cells/metabolism , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Neoplasms/metabolism
4.
Cancer Immunol Res ; 9(3): 309-323, 2021 03.
Article in English | MEDLINE | ID: mdl-33361087

ABSTRACT

IL1ß is a central mediator of inflammation. Secretion of IL1ß typically requires proteolytic maturation by the inflammasome and formation of membrane pores by gasdermin D (GSDMD). Emerging evidence suggests an important role for IL1ß in promoting cancer progression in patients, but the underlying mechanisms are ill-defined. Here, we have shown a key role for IL1ß in driving tumor progression in two distinct mouse tumor models. Notably, activation of the inflammasome, caspase-8, as well as the pore-forming proteins GSDMD and mixed lineage kinase domain-like protein in the host were dispensable for the release of intratumoral bioactive IL1ß. Inflammasome-independent IL1ß release promoted systemic neutrophil expansion and fostered accumulation of T-cell-suppressive neutrophils in the tumor. Moreover, IL1ß was essential for neutrophil infiltration triggered by antiangiogenic therapy, thereby contributing to treatment-induced immunosuppression. Deletion of IL1ß allowed intratumoral accumulation of CD8+ effector T cells that subsequently activated tumor-associated macrophages. Depletion of either CD8+ T cells or macrophages abolished tumor growth inhibition in IL1ß-deficient mice, demonstrating a crucial role for CD8+ T-cell-macrophage cross-talk in the antitumor immune response. Overall, these results support a tumor-promoting role for IL1ß through establishing an immunosuppressive microenvironment and show that inflammasome activation is not essential for release of this cytokine in tumors.


Subject(s)
Interleukin-1beta/metabolism , Neoplasms/immunology , Neutrophils/immunology , Tumor Escape , Tumor Microenvironment/immunology , Animals , Cell Communication/immunology , Disease Models, Animal , Female , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Knockout , Neoplasms/pathology , Neutrophils/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Tumor-Associated Macrophages/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...