Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Cell ; 186(8): 1627-1651, 2023 04 13.
Article in English | MEDLINE | ID: mdl-36924769

ABSTRACT

Macrophages are versatile and heterogeneous innate immune cells undertaking central functions in balancing immune responses and tissue repair to maintain homeostasis. This plasticity, once co-opted by malignant outgrowth, orchestrates manifold reciprocal interactions within the tumor microenvironment, fueling the evolution of the cancer ecosystem. Here, we review the multilayered sources of influence that jointly underpin and longitudinally shape tumor-associated macrophage (TAM) phenotypic states in solid neoplasms. We discuss how, in response to these signals, TAMs steer tumor evolution in the context of natural selection, biological dispersion, and treatment resistance. A number of research frontiers to be tackled are laid down in this review to therapeutically exploit the complex roles of TAMs in cancer. Building upon knowledge obtained from currently applied TAM-targeting strategies and using next generation technologies, we propose conceptual advances and novel therapeutic avenues to rewire TAM multifaceted regulation of the co-evolving cancer ecosystem.


Subject(s)
Neoplasms , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Neoplasms/pathology , Neoplasms/therapy
2.
Nature ; 614(7948): 555-563, 2023 02.
Article in English | MEDLINE | ID: mdl-36725935

ABSTRACT

Single-cell technologies have enabled the characterization of the tumour microenvironment at unprecedented depth and have revealed vast cellular diversity among tumour cells and their niche. Anti-tumour immunity relies on cell-cell relationships within the tumour microenvironment1,2, yet many single-cell studies lack spatial context and rely on dissociated tissues3. Here we applied imaging mass cytometry to characterize the immunological landscape of 139 high-grade glioma and 46 brain metastasis tumours from patients. Single-cell analysis of more than 1.1 million cells across 389 high-dimensional histopathology images enabled the spatial resolution of immune lineages and activation states, revealing differences in immune landscapes between primary tumours and brain metastases from diverse solid cancers. These analyses revealed cellular neighbourhoods associated with survival in patients with glioblastoma, which we leveraged to identify a unique population of myeloperoxidase (MPO)-positive macrophages associated with long-term survival. Our findings provide insight into the biology of primary and metastatic brain tumours, reinforcing the value of integrating spatial resolution to single-cell datasets to dissect the microenvironmental contexture of cancer.


Subject(s)
Brain Neoplasms , Glioma , Single-Cell Analysis , Tumor Microenvironment , Humans , Brain/immunology , Brain/pathology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Glioblastoma/immunology , Glioblastoma/pathology , Glioma/immunology , Glioma/pathology , Macrophages/enzymology , Tumor Microenvironment/immunology , Neoplasm Metastasis , Datasets as Topic
3.
Nat Commun ; 15(1): 2581, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519484

ABSTRACT

Myeloid cells are abundant and plastic immune cell subsets in the liver, to which pro-tumorigenic, inflammatory and immunosuppressive roles have been assigned in the course of tumorigenesis. Yet several aspects underlying their dynamic alterations in hepatocellular carcinoma (HCC) progression remain elusive, including the impact of distinct genetic mutations in shaping a cancer-permissive tumor microenvironment (TME). Here, in newly generated, clinically-relevant somatic female HCC mouse models, we identify cancer genetics' specific and stage-dependent alterations of the liver TME associated with distinct histopathological and malignant HCC features. Mitogen-activated protein kinase (MAPK)-activated, NrasG12D-driven tumors exhibit a mixed phenotype of prominent inflammation and immunosuppression in a T cell-excluded TME. Mechanistically, we report a NrasG12D cancer cell-driven, MEK-ERK1/2-SP1-dependent GM-CSF secretion enabling the accumulation of immunosuppressive and proinflammatory monocyte-derived Ly6Clow cells. GM-CSF blockade curbs the accumulation of these cells, reduces inflammation, induces cancer cell death and prolongs animal survival. Furthermore, GM-CSF neutralization synergizes with a vascular endothelial growth factor (VEGF) inhibitor to restrain HCC outgrowth. These findings underscore the profound alterations of the myeloid TME consequential to MAPK pathway activation intensity and the potential of GM-CSF inhibition as a myeloid-centric therapy tailored to subsets of HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Animals , Humans , Female , Carcinoma, Hepatocellular/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Liver Neoplasms/metabolism , Tumor Microenvironment/genetics , Vascular Endothelial Growth Factor A , Myeloid Cells/metabolism , Mitogen-Activated Protein Kinases/metabolism , Immunosuppressive Agents , Inflammation/pathology
4.
Nat Cancer ; 4(5): 665-681, 2023 05.
Article in English | MEDLINE | ID: mdl-37081259

ABSTRACT

Glioblastomas are aggressive primary brain tumors with an inherent resistance to T cell-centric immunotherapy due to their low mutational burden and immunosuppressive tumor microenvironment. Here we report that fractionated radiotherapy of preclinical glioblastoma models induce a tenfold increase in T cell content. Orthogonally, spatial imaging mass cytometry shows T cell enrichment in human recurrent tumors compared with matched primary glioblastoma. In glioblastoma-bearing mice, α-PD-1 treatment applied at the peak of T cell infiltration post-radiotherapy results in a modest survival benefit compared with concurrent α-PD-1 administration. Following α-PD-1 therapy, CD103+ regulatory T cells (Tregs) with upregulated lipid metabolism accumulate in the tumor microenvironment, and restrain immune checkpoint blockade response by repressing CD8+ T cell activation. Treg targeting elicits tertiary lymphoid structure formation, enhances CD4+ and CD8+ T cell frequency and function and unleashes radio-immunotherapeutic efficacy. These results support the rational design of therapeutic regimens limiting the induction of immunosuppressive feedback pathways in the context of T cell immunotherapy in glioblastoma.


Subject(s)
Glioblastoma , Mice , Humans , Animals , Glioblastoma/radiotherapy , T-Lymphocytes, Regulatory/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/therapeutic use , Neoplasm Recurrence, Local/metabolism , CD8-Positive T-Lymphocytes , Immunotherapy/methods , Tumor Microenvironment
5.
Neuro Oncol ; 24(9): 1494-1508, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35416251

ABSTRACT

BACKGROUND: Glioblastoma is a treatment-resistant brain cancer. Its hierarchical cellular nature and its tumor microenvironment (TME) before, during, and after treatments remain unresolved. METHODS: Here, we used single-cell RNA sequencing to analyze new and recurrent glioblastoma and the nearby subventricular zone (SVZ). RESULTS: We found 4 glioblastoma neural lineages are present in new and recurrent glioblastoma with an enrichment of the cancer mesenchymal lineage, immune cells, and reactive astrocytes in early recurrences. Cancer lineages were hierarchically organized around cycling oligodendrocytic and astrocytic progenitors that are transcriptomically similar but distinct to SVZ neural stem cells (NSCs). Furthermore, NSCs from the SVZ of patients with glioblastoma harbored glioblastoma chromosomal anomalies. Lastly, mesenchymal cancer cells and TME reactive astrocytes shared similar gene signatures which were induced by radiotherapy in a myeloid-dependent fashion in vivo. CONCLUSION: These data reveal the dynamic, immune-dependent nature of glioblastoma's response to treatments and identify distant NSCs as likely cells of origin.


Subject(s)
Brain Neoplasms , Glioblastoma , Neural Stem Cells , Brain Neoplasms/pathology , Glioblastoma/pathology , Humans , Lateral Ventricles/pathology , Neural Stem Cells/pathology , Single-Cell Analysis , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL