Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
Front Immunol ; 12: 772555, 2021.
Article in English | MEDLINE | ID: mdl-34925341

ABSTRACT

The combination of radiotherapy (RT) with immunotherapy represents a promising treatment modality for non-small cell lung cancer (NSCLC) patients. As only a minority of patients shows a persistent response today, a spacious optimization window remains to be explored. Previously we showed that fractionated RT can induce a local immunosuppressive profile. Based on the evolving concept of an immunomodulatory role for vagal nerve stimulation (VNS), we tested its therapeutic and immunological effects alone and in combination with fractionated RT in a preclinical-translational study. Lewis lung carcinoma-bearing C57Bl/6 mice were treated with VNS, fractionated RT or the combination while a patient cohort with locally advanced NSCLC receiving concurrent radiochemotherapy (ccRTCT) was enrolled in a clinical trial to receive either sham or effective VNS daily during their 6 weeks of ccRTCT treatment. Preclinically, VNS alone or with RT showed no therapeutic effect yet VNS alone significantly enhanced the activation profile of intratumoral CD8+ T cells by upregulating their IFN-γ and CD137 expression. In the periphery, VNS reduced the RT-mediated rise of splenic, but not blood-derived, regulatory T cells (Treg) and monocytes. In accordance, the serological levels of protumoral CXCL5 next to two Treg-attracting chemokines CCL1 and CCL22 were reduced upon VNS monotherapy. In line with our preclinical findings on the lack of immunological changes in blood circulating immune cells upon VNS, immune monitoring of the peripheral blood of VNS treated NSCLC patients (n=7) did not show any significant changes compared to ccRTCT alone. As our preclinical data do suggest that VNS intensifies the stimulatory profile of the tumor infiltrated CD8+ T cells, this favors further research into non-invasive VNS to optimize current response rates to RT-immunotherapy in lung cancer patients.


Subject(s)
Carcinoma, Lewis Lung/radiotherapy , Carcinoma, Lewis Lung/therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/radiotherapy , Lung Neoplasms/therapy , Vagus Nerve Stimulation , Aged , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Combined Modality Therapy , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice, Inbred C57BL , Middle Aged , Tumor Burden
4.
J Immunother Cancer ; 9(2)2021 02.
Article in English | MEDLINE | ID: mdl-33589525

ABSTRACT

BACKGROUND: Modulation and depletion strategies of regulatory T cells (Tregs) constitute valid approaches in antitumor immunotherapy but suffer from severe adverse effects due to their lack of selectivity for the tumor-infiltrating (ti-)Treg population, indicating the need for a ti-Treg specific biomarker. METHODS: We employed single-cell RNA-sequencing in a mouse model of non-small cell lung carcinoma (NSCLC) to obtain a comprehensive overview of the tumor-infiltrating T-cell compartment, with a focus on ti-Treg subpopulations. These findings were validated by flow cytometric analysis of both mouse (LLC-OVA, MC38 and B16-OVA) and human (NSCLC and melanoma) tumor samples. We generated two CCR8-specific nanobodies (Nbs) that recognize distinct epitopes on the CCR8 extracellular domain. These Nbs were formulated as tetravalent Nb-Fc fusion proteins for optimal CCR8 binding and blocking, containing either an antibody-dependent cell-mediated cytotoxicity (ADCC)-deficient or an ADCC-prone Fc region. The therapeutic use of these Nb-Fc fusion proteins was evaluated, either as monotherapy or as combination therapy with anti-programmed cell death protein-1 (anti-PD-1), in both the LLC-OVA and MC38 mouse models. RESULTS: We were able to discern two ti-Treg populations, one of which is characterized by the unique expression of Ccr8 in conjunction with Treg activation markers. Ccr8 is also expressed by dysfunctional CD4+ and CD8+ T cells, but the CCR8 protein was only prominent on the highly activated and strongly T-cell suppressive ti-Treg subpopulation of mouse and human tumors, with no major CCR8-positivity found on peripheral Tregs. CCR8 expression resulted from TCR-mediated Treg triggering in an NF-κB-dependent fashion, but was not essential for the recruitment, activation nor suppressive capacity of these cells. While treatment of tumor-bearing mice with a blocking ADCC-deficient Nb-Fc did not influence tumor growth, ADCC-prone Nb-Fc elicited antitumor immunity and reduced tumor growth in synergy with anti-PD-1 therapy. Importantly, ADCC-prone Nb-Fc specifically depleted ti-Tregs in a natural killer (NK) cell-dependent fashion without affecting peripheral Tregs. CONCLUSIONS: Collectively, our findings highlight the efficacy and safety of targeting CCR8 for the depletion of tumor-promoting ti-Tregs in combination with anti-PD-1 therapy.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Lewis Lung/therapy , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lymphocyte Depletion , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, CCR8/deficiency , Skin Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Combined Modality Therapy , Databases, Genetic , Female , Gene Expression Profiling , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Phenotype , Programmed Cell Death 1 Receptor/metabolism , RNA-Seq , Receptors, CCR8/genetics , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , T-Lymphocytes, Regulatory/metabolism
5.
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
6.
Front Immunol ; 9: 2250, 2018.
Article in English | MEDLINE | ID: mdl-30349530

ABSTRACT

Therapeutic approaches that engage immune cells to treat cancer are becoming increasingly utilized in the clinics and demonstrated durable clinical benefit in several solid tumor types. Most of the current immunotherapies focus on manipulating T cells, however, the tumor microenvironment (TME) is abundantly infiltrated by a heterogeneous population of tumor-associated myeloid cells, including tumor-associated macrophages (TAMs), tumor-associated dendritic cells (TADCs), tumor-associated neutrophils (TANs), and myeloid-derived suppressor cells (MDSCs). Educated by signals perceived in the TME, these cells often acquire tumor-promoting properties ultimately favoring disease progression. Upon appropriate stimuli, myeloid cells can exhibit cytoxic, phagocytic, and antigen-presenting activities thereby bolstering antitumor immune responses. Thus, depletion, reprogramming or reactivation of myeloid cells to either directly eradicate malignant cells or promote antitumor T-cell responses is an emerging field of interest. In this review, we briefly discuss the tumor-promoting and tumor-suppressive roles of myeloid cells in the TME, and describe potential therapeutic strategies in preclinical and clinical development that aim to target them to further expand the range of current treatment options.


Subject(s)
Immunotherapy/methods , Myeloid Cells/immunology , Neoplasms/immunology , Neoplasms/therapy , Tumor Microenvironment/immunology , Dendritic Cells/immunology , Humans , Macrophages/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/pathology , Neutrophils/immunology , T-Lymphocytes/immunology
7.
Cell Immunol ; 330: 202-215, 2018 08.
Article in English | MEDLINE | ID: mdl-29661474

ABSTRACT

The liver is a major target organ for metastasis of both gastrointestinal and extra-gastrointestinal cancers. Due to its frequently inoperable nature, liver metastasis represents a leading cause of cancer-associated death worldwide. In the past years, the pivotal role of the immune system in this process is being increasingly recognised. In particular, the role of the hepatic macrophages, both recruited monocyte-derived macrophages (Mo-Mfs) and tissue-resident Kupffer cells (KCs), has been shown to be more versatile than initially imagined. However, the lack of tools to easily distinguish between these two macrophage populations has hampered the assignment of particular functionalities to specific hepatic macrophage subsets. In this Review, we highlight the most remarkable findings regarding the origin and functions of hepatic macrophage populations, and we provide a detailed description of their distinct roles in the different phases of the liver metastatic process.


Subject(s)
Kupffer Cells/immunology , Liver Neoplasms/immunology , Liver/immunology , Macrophages/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Hepatocytes/immunology , Hepatocytes/metabolism , Homeostasis/immunology , Humans , Kupffer Cells/pathology , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasm Metastasis
8.
FEBS J ; 285(4): 777-787, 2018 02.
Article in English | MEDLINE | ID: mdl-28834216

ABSTRACT

Tumor-associated macrophages (TAM) are by now established as important regulators of tumor progression by impacting on tumor immunity, angiogenesis, and metastasis. Hence, a multitude of approaches are currently pursued to intervene with TAM's protumor activities, the most advanced of which being a blockade of macrophage-colony stimulating factor (M-CSF)/M-CSF receptor (M-CSFR) signaling. M-CSFR signaling largely impacts on the differentiation of macrophages, including TAM, and hence strongly influences the numbers of these cells in tumors. However, a repolarization of TAM toward a more antitumor phenotype may be more elegant and may yield stronger effects on tumor growth. In this respect, several aspects of TAM behavior could be altered, such as their intratumoral localization, metabolism and regulatory pathways. Intervention strategies could include the use of small molecules but also new generations of biologicals which may complement the current success of immune checkpoint blockers. This review highlights current work on the search for new therapeutic targets in TAM.


Subject(s)
Macrophages/drug effects , Neoplasms/drug therapy , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Humans , Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects
9.
J Leukoc Biol ; 102(2): 317-324, 2017 08.
Article in English | MEDLINE | ID: mdl-28389620

ABSTRACT

Dendritic cells (DCs) are specialized APCs present in all tissues, including tumors. They play a major role in orchestrating immune responses and were shown to occur in various functional states in tumors. In this respect, immunogenic tumor-associated DCs (TADCs) are required to initiate and sustain T cell-dependent anti-cancer immunity, whereas regulatory TADCs harbor robust immunosuppressive potential and accelerate malignant growth. Importantly, the heterogeneity of the DC compartment in tumors has been dissected recently in murine and human cancers and was shown to consist of developmentally distinct subsets, including conventional DC (cDC)1, cDC2, and monocyte-derived DCs (Mo-DCs). TADCs constitute an essential target in efforts to generate therapeutic immunity against cancer, and the understanding of the complexity of the TADC heterogeneity might prove important for therapeutic interventions targeted at specific TADC subsets or their precursors. Hence, this review addresses the differential functional specializations of ontogenically distinct TADC subsets.


Subject(s)
Dendritic Cells/immunology , Neoplasms/immunology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...