Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Cancer Immunol Res ; 11(6): 777-791, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37040466

ABSTRACT

High levels of IL1ß can result in chronic inflammation, which in turn can promote tumor growth and metastasis. Inhibition of IL1ß could therefore be a promising therapeutic option in the treatment of cancer. Here, the effects of IL1ß blockade induced by the mAbs canakinumab and gevokizumab were evaluated alone or in combination with docetaxel, anti-programmed cell death protein 1 (anti-PD-1), anti-VEGFα, and anti-TGFß treatment in syngeneic and humanized mouse models of cancers of different origin. Canakinumab and gevokizumab did not show notable efficacy as single-agent therapies; however, IL1ß blockade enhanced the effectiveness of docetaxel and anti-PD-1. Accompanying these effects, blockade of IL1ß alone or in combination induced significant remodeling of the tumor microenvironment (TME), with decreased numbers of immune suppressive cells and increased tumor infiltration by dendritic cells (DC) and effector T cells. Further investigation revealed that cancer-associated fibroblasts (CAF) were the cell type most affected by treatment with canakinumab or gevokizumab in terms of change in gene expression. IL1ß inhibition drove phenotypic changes in CAF populations, particularly those with the ability to influence immune cell recruitment. These results suggest that the observed remodeling of the TME following IL1ß blockade may stem from changes in CAF populations. Overall, the results presented here support the potential use of IL1ß inhibition in cancer treatment. Further exploration in ongoing clinical studies will help identify the best combination partners for different cancer types, cancer stages, and lines of treatment.


Subject(s)
Interleukin-1beta , Neoplasms , Tumor Microenvironment , Animals , Mice , Cell Line, Tumor , Docetaxel/pharmacology , Immunity , Immunotherapy , Neoplasms/drug therapy , Interleukin-1beta/antagonists & inhibitors
2.
Nat Commun ; 11(1): 6315, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33298926

ABSTRACT

Despite the increasing interest in targeting stromal elements of the tumor microenvironment, we still face tremendous challenges in developing adequate therapeutics to modify the tumor stromal landscape. A major obstacle to this is our poor understanding of the phenotypic and functional heterogeneity of stromal cells in tumors. Herein, we perform an unbiased interrogation of tumor mesenchymal cells, delineating the co-existence of distinct subsets of cancer-associated fibroblasts (CAFs) in the microenvironment of murine carcinomas, each endowed with unique phenotypic features and functions. Furthermore, our study shows that neutralization of TGFß in vivo leads to remodeling of CAF dynamics, greatly reducing the frequency and activity of the myofibroblast subset, while promoting the formation of a fibroblast population characterized by strong response to interferon and heightened immunomodulatory properties. These changes correlate with the development of productive anti-tumor immunity and greater efficacy of PD1 immunotherapy. Along with providing the scientific rationale for the evaluation of TGFß and PD1 co-blockade in the clinical setting, this study also supports the concept of plasticity of the stromal cell landscape in tumors, laying the foundation for future investigations aimed at defining pathways and molecules to program CAF composition for cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cancer-Associated Fibroblasts/immunology , Carcinoma/drug therapy , Interferon-beta/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer-Associated Fibroblasts/drug effects , Carcinoma/immunology , Carcinoma/pathology , Cell Line, Tumor/transplantation , Cell Plasticity/drug effects , Cell Plasticity/immunology , Disease Models, Animal , Drug Synergism , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Stromal Cells/drug effects , Stromal Cells/immunology , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
3.
Cancer Immunol Res ; 6(12): 1472-1485, 2018 12.
Article in English | MEDLINE | ID: mdl-30266714

ABSTRACT

Cancer-associated fibroblasts (CAFs) are generally associated with poor clinical outcome. CAFs support tumor growth in a variety of ways and can suppress antitumor immunity and response to immunotherapy. However, a precise understanding of CAF contributions to tumor growth and therapeutic response is lacking. Discrepancies in this field of study may stem from heterogeneity in the composition and function of fibroblasts in the tumor microenvironment. Furthermore, it remains unclear whether CAFs directly interact with and suppress T cells. Here, mouse and human breast tumors were used to examine stromal cells expressing fibroblast activation protein (FAP), a surface marker for CAFs. Two discrete populations of FAP+ mesenchymal cells were identified on the basis of podoplanin (PDPN) expression: a FAP+PDPN+ population of CAFs and a FAP+PDPN- population of cancer-associated pericytes (CAPs). Although both subsets expressed extracellular matrix molecules, the CAF transcriptome was enriched in genes associated with TGFß signaling and fibrosis compared with CAPs. In addition, CAFs were enriched at the outer edge of the tumor, in close contact with T cells, whereas CAPs were localized around vessels. Finally, FAP+PDPN+ CAFs suppressed the proliferation of T cells in a nitric oxide-dependent manner, whereas FAP+PDPN- pericytes were not immunosuppressive. Collectively, these findings demonstrate that breast tumors contain multiple populations of FAP-expressing stromal cells of dichotomous function, phenotype, and location.


Subject(s)
Breast Neoplasms/pathology , Gelatinases/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Stromal Cells/metabolism , Tumor Microenvironment/immunology , Animals , Breast Neoplasms/immunology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Proliferation , Endopeptidases , Female , Gene Expression Regulation , Humans , Membrane Glycoproteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide/metabolism , Pericytes/metabolism , Pericytes/pathology , Stromal Cells/pathology , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL