Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Cell ; 187(19): 5336-5356.e30, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39137777

ABSTRACT

Tumors growing in metabolically challenged environments, such as glioblastoma in the brain, are particularly reliant on crosstalk with their tumor microenvironment (TME) to satisfy their high energetic needs. To study the intricacies of this metabolic interplay, we interrogated the heterogeneity of the glioblastoma TME using single-cell and multi-omics analyses and identified metabolically rewired tumor-associated macrophage (TAM) subpopulations with pro-tumorigenic properties. These TAM subsets, termed lipid-laden macrophages (LLMs) to reflect their cholesterol accumulation, are epigenetically rewired, display immunosuppressive features, and are enriched in the aggressive mesenchymal glioblastoma subtype. Engulfment of cholesterol-rich myelin debris endows subsets of TAMs to acquire an LLM phenotype. Subsequently, LLMs directly transfer myelin-derived lipids to cancer cells in an LXR/Abca1-dependent manner, thereby fueling the heightened metabolic demands of mesenchymal glioblastoma. Our work provides an in-depth understanding of the immune-metabolic interplay during glioblastoma progression, thereby laying a framework to unveil targetable metabolic vulnerabilities in glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Myelin Sheath , Tumor Microenvironment , Humans , Myelin Sheath/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Animals , Mice , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Cholesterol/metabolism , Liver X Receptors/metabolism , Macrophages/metabolism , Cell Line, Tumor , ATP Binding Cassette Transporter 1/metabolism , Female , Male
2.
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
3.
Cell ; 181(7): 1454-1457, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32589956

ABSTRACT

Despite its success in multiple tumor types, immunotherapy remains poorly efficacious in brain malignancies. In this issue of Cell, Friebel et al. and Klemm et al. provide in-depth insights into the versatile nuances of immune cells in primary and metastatic brain tumors, granting the field with a rich framework to explore novel therapeutic avenues.


Subject(s)
Brain Neoplasms , Brain Neoplasms/therapy , Humans , Immunotherapy
4.
Cell ; 178(4): 807-819.e21, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398338

ABSTRACT

The NRF2 transcription factor controls a cell stress program that is implicated in cancer and there is great interest in targeting NRF2 for therapy. We show that NRF2 activity depends on Fructosamine-3-kinase (FN3K)-a kinase that triggers protein de-glycation. In its absence, NRF2 is extensively glycated, unstable, and defective at binding to small MAF proteins and transcriptional activation. Moreover, the development of hepatocellular carcinoma triggered by MYC and Keap1 inactivation depends on FN3K in vivo. N-acetyl cysteine treatment partially rescues the effects of FN3K loss on NRF2 driven tumor phenotypes indicating a key role for NRF2-mediated redox balance. Mass spectrometry reveals that other proteins undergo FN3K-sensitive glycation, including translation factors, heat shock proteins, and histones. How glycation affects their functions remains to be defined. In summary, our study reveals a surprising role for the glycation of cellular proteins and implicates FN3K as targetable modulator of NRF2 activity in cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Female , Gene Knockdown Techniques , Glucose/metabolism , Glycosylation , HEK293 Cells , Hep G2 Cells , Heterografts , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transduction, Genetic
5.
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
6.
Cell ; 153(2): 449-60, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23562644

ABSTRACT

The p53 tumor suppressor can restrict malignant transformation by triggering cell-autonomous programs of cell-cycle arrest or apoptosis. p53 also promotes cellular senescence, a tumor-suppressive program that involves stable cell-cycle arrest and secretion of factors that modify the tissue microenvironment. In the presence of chronic liver damage, we show that ablation of a p53-dependent senescence program in hepatic stellate cells increases liver fibrosis and cirrhosis associated with reduced survival and enhances the transformation of adjacent epithelial cells into hepatocellular carcinoma. p53-expressing senescent stellate cells release factors that skew macrophage polarization toward a tumor-inhibiting M1-state capable of attacking senescent cells in culture, whereas proliferating p53-deficient stellate cells secrete factors that stimulate polarization of macrophages into a tumor-promoting M2-state and enhance the proliferation of premalignant cells. Hence, p53 can act non-cell autonomously to suppress tumorigenesis by promoting an antitumor microenvironment, in part, through secreted factors that modulate macrophage function.


Subject(s)
Cell Transformation, Neoplastic , Cellular Senescence , Hepatic Stellate Cells/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Cellular Microenvironment , Fibrosis/pathology , Hepatic Stellate Cells/cytology , Humans , Inflammation/metabolism , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/cytology , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , NF-kappa B
7.
Nature ; 595(7869): 730-734, 2021 07.
Article in English | MEDLINE | ID: mdl-34290403

ABSTRACT

Hepatocellular carcinoma (HCC)-the most common form of liver cancer-is an aggressive malignancy with few effective treatment options1. Lenvatinib is a small-molecule inhibitor of multiple receptor tyrosine kinases that is used for the treatment of patients with advanced HCC, but this drug has only limited clinical benefit2. Here, using a kinome-centred CRISPR-Cas9 genetic screen, we show that inhibition of epidermal growth factor receptor (EGFR) is synthetic lethal with lenvatinib in liver cancer. The combination of the EGFR inhibitor gefitinib and lenvatinib displays potent anti-proliferative effects in vitro in liver cancer cell lines that express EGFR and in vivo in xenografted liver cancer cell lines, immunocompetent mouse models and patient-derived HCC tumours in mice. Mechanistically, inhibition of fibroblast growth factor receptor (FGFR)  by lenvatinib treatment leads to feedback activation of the EGFR-PAK2-ERK5 signalling axis, which is blocked by EGFR inhibition. Treatment of 12 patients with advanced HCC who were unresponsive to lenvatinib treatment with the combination of lenvatinib plus gefitinib (trial identifier NCT04642547) resulted in meaningful clinical responses. The combination therapy identified here may represent a promising strategy for the approximately 50% of patients with advanced HCC who have high levels of EGFR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , Animals , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Gefitinib/pharmacology , Humans , Liver Neoplasms/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, Fibroblast Growth Factor , Signal Transduction , Xenograft Model Antitumor Assays
8.
Trends Immunol ; 44(12): 933-935, 2023 12.
Article in English | MEDLINE | ID: mdl-37949785

ABSTRACT

Immunotherapies have thus far led to disappointing outcomes in patients suffering from glioblastoma. Published in Immunity, Chen et al.'s recent study shows the therapeutic potential of an αCTLA-4 antibody (Ab), specifically in murine mesenchymal-like glioblastoma. αCTLA-4 Ab efficacy relied on the distinctive cooperation between CD4+ Th1 T cells and microglia, unleashing a potent antitumor response.


Subject(s)
Glioblastoma , Humans , Mice , Animals , CTLA-4 Antigen , Adaptive Immunity , Antibodies , Immunotherapy , Immunity, Innate
9.
Nature ; 574(7777): 268-272, 2019 10.
Article in English | MEDLINE | ID: mdl-31578521

ABSTRACT

Liver cancer remains difficult to treat, owing to a paucity of drugs that target critical dependencies1,2; broad-spectrum kinase inhibitors such as sorafenib provide only a modest benefit to patients with hepatocellular carcinoma3. The induction of senescence may represent a strategy for the treatment of cancer, especially when combined with a second drug that selectively eliminates senescent cancer cells (senolysis)4,5. Here, using a kinome-focused genetic screen, we show that pharmacological inhibition of the DNA-replication kinase CDC7 induces senescence selectively in liver cancer cells with mutations in TP53. A follow-up chemical screen identified the antidepressant sertraline as an agent that kills hepatocellular carcinoma cells that have been rendered senescent by inhibition of CDC7. Sertraline suppressed mTOR signalling, and selective drugs that target this pathway were highly effective in causing the apoptotic cell death of hepatocellular carcinoma cells treated with a CDC7 inhibitor. The feedback reactivation of mTOR signalling after its inhibition6 is blocked in cells that have been treated with a CDC7 inhibitor, which leads to the sustained inhibition of mTOR and cell death. Using multiple in vivo mouse models of liver cancer, we show that treatment with combined inhibition of of CDC7 and mTOR results in a marked reduction of tumour growth. Our data indicate that exploiting an induced vulnerability could be an effective treatment for liver cancer.


Subject(s)
Apoptosis/drug effects , Cellular Senescence/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Molecular Targeted Therapy , Sertraline/pharmacology , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Female , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sertraline/therapeutic use , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
10.
Genes Dev ; 30(2): 220-32, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26773004

ABSTRACT

Proteases are important for regulating multiple tumorigenic processes, including angiogenesis, tumor growth, and invasion. Elevated protease expression is associated with poor patient prognosis across numerous tumor types. Several multigene protease families have been implicated in cancer, including cysteine cathepsins. However, whether individual family members have unique roles or are functionally redundant remains poorly understood. Here we demonstrate stage-dependent effects of simultaneously deleting cathepsin B (CtsB) and CtsS in a murine pancreatic neuroendocrine tumor model. Early in tumorigenesis, the double knockout results in an additive reduction in angiogenic switching, whereas at late stages, several tumorigenic phenotypes are unexpectedly restored to wild-type levels. We identified CtsZ, which is predominantly supplied by tumor-associated macrophages, as the compensatory protease that regulates the acquired tumor-promoting functions of lesions deficient in both CtsB and CtsS. Thus, deletion of multiple cathepsins can lead to stage-dependent, compensatory mechanisms in the tumor microenvironment, which has potential implications for the clinical consideration of selective versus pan-family cathepsin inhibitors in cancer.


Subject(s)
Carcinoma, Neuroendocrine/enzymology , Cathepsins/genetics , Cathepsins/metabolism , Gene Deletion , Pancreatic Neoplasms/enzymology , Animals , Apoptosis/genetics , Carcinogenesis/genetics , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/physiopathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/physiopathology
11.
Semin Cancer Biol ; 86(Pt 3): 41-56, 2022 11.
Article in English | MEDLINE | ID: mdl-35569742

ABSTRACT

The intricate cross-talks between tumor cells and their microenvironment play a key role in cancer progression and resistance to treatment. In recent years, targeting pro-tumorigenic components of the tumor microenvironment (TME) has emerged as a tantalizing strategy to improve the efficacy of standard-of-care (SOC) treatments, particularly for hard-to-treat cancers such as glioblastoma. In this review, we explore how the distinct microenvironmental niches characteristic of the glioblastoma TME shape response to therapy. In particular, we delve into the interplay between tumor-associated macrophages (TAM) and glioblastoma cells within angiogenic and hypoxic niches, and interrogate their dynamic co-evolution upon SOC therapies that fuels malignancy. Resolving the complexity of therapy-induced alterations in the glioblastoma TME and their impact on disease relapse is a stepping stone to identify targetable pro-tumorigenic pathways and TAM subsets, and may open the way to efficient combination therapies that will improve clinical outcomes.


Subject(s)
Glioblastoma , Humans , Glioblastoma/genetics , Glioblastoma/drug therapy , Tumor Microenvironment/physiology , Macrophages/metabolism , Carcinogenesis/metabolism , Hypoxia/metabolism
12.
Curr Oncol Rep ; 24(3): 311-324, 2022 03.
Article in English | MEDLINE | ID: mdl-35119629

ABSTRACT

PURPOSE OF REVIEW: Glioblastoma is the commonest primary brain cancer in adults whose outcomes are amongst the worst of any cancer. The current treatment pathway comprises surgery and postoperative chemoradiotherapy though unresectable diffusely infiltrative tumour cells remain untreated for several weeks post-diagnosis. Intratumoural heterogeneity combined with increased hypoxia in the postoperative tumour microenvironment potentially decreases the efficacy of adjuvant interventions and fails to prevent early postoperative regrowth, called rapid early progression (REP). In this review, we discuss the clinical implications and biological foundations of post-surgery REP. Subsequently, clinical interventions potentially targeting this phenomenon are reviewed systematically. RECENT FINDINGS: Early interventions include early systemic chemotherapy, neoadjuvant immunotherapy, local therapies delivered during surgery (including Gliadel wafers, nanoparticles and stem cell therapy) and several radiotherapy techniques. We critically appraise and compare these strategies in terms of their efficacy, toxicity, challenges and potential to prolong survival. Finally, we discuss the most promising strategies that could benefit future glioblastoma patients. There is biological rationale to suggest that early interventions could improve the outcome of glioblastoma patients and they should be investigated in future trials.


Subject(s)
Brain Neoplasms , Glioblastoma , Adult , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Carmustine/therapeutic use , Chemoradiotherapy , Glioblastoma/drug therapy , Glioblastoma/therapy , Humans , Tumor Microenvironment
13.
Genes Dev ; 28(19): 2134-50, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25274726

ABSTRACT

During the process of tumor progression, cancer cells can produce the requisite growth- and invasion-promoting factors and can also rely on noncancerous cells in the tumor microenvironment as an alternative, cell-extrinsic source. However, whether the cellular source influences the function of such tumor-promoting factors remains an open question. Here, we examined the roles of the cathepsin Z (CtsZ) protease, which is provided by both cancer cells and macrophages in pancreatic neuroendocrine tumors in humans and mice. We found that tumor proliferation was exclusively regulated by cancer cell-intrinsic functions of CtsZ, whereas tumor invasion required contributions from both macrophages and cancer cells. Interestingly, several of the tumor-promoting functions of CtsZ were not dependent on its described catalytic activity but instead were mediated via the Arg-Gly-Asp (RGD) motif in the enzyme prodomain, which regulated interactions with integrins and the extracellular matrix. Together, these results underscore the complexity of interactions within the tumor microenvironment and indicate that cellular source can indeed impact molecular function.


Subject(s)
Cathepsin Z/metabolism , Extracellular Matrix/metabolism , Macrophages/enzymology , Neoplasms/enzymology , Neoplasms/physiopathology , Animals , Cell Line, Tumor , Integrins/metabolism , Mice, Inbred C57BL , Neoplasm Invasiveness/physiopathology
14.
Proc Natl Acad Sci U S A ; 114(11): 2934-2939, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28246332

ABSTRACT

The genetic and phenotypic diversity of cells within tumors is a major obstacle for cancer treatment. Because of the stochastic nature of genetic alterations, this intratumoral heterogeneity is often viewed as chaotic. Here we show that the altered metabolism of cancer cells creates predictable gradients of extracellular metabolites that orchestrate the phenotypic diversity of cells in the tumor microenvironment. Combining experiments and mathematical modeling, we show that metabolites consumed and secreted within the tumor microenvironment induce tumor-associated macrophages (TAMs) to differentiate into distinct subpopulations according to local levels of ischemia and their position relative to the vasculature. TAMs integrate levels of hypoxia and lactate into progressive activation of MAPK signaling that induce predictable spatial patterns of gene expression, such as stripes of macrophages expressing arginase 1 (ARG1) and mannose receptor, C type 1 (MRC1). These phenotypic changes are functionally relevant as ischemic macrophages triggered tube-like morphogenesis in neighboring endothelial cells that could restore blood perfusion in nutrient-deprived regions where angiogenic resources are most needed. We propose that gradients of extracellular metabolites act as tumor morphogens that impose order within the microenvironment, much like signaling molecules convey positional information to organize embryonic tissues. Unearthing embryology-like processes in tumors may allow us to control organ-like tumor features such as tissue repair and revascularization and treat intratumoral heterogeneity.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Tumor Microenvironment , Cell Line, Tumor , Cluster Analysis , Energy Metabolism , Extracellular Space/metabolism , Gene Expression Profiling , Humans , Hypoxia/metabolism , Lactic Acid/metabolism , MAP Kinase Signaling System , Macrophages/metabolism , Macrophages/pathology , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Oxygen/metabolism , Phenotype , Transcriptome , Tumor Microenvironment/genetics
15.
Proc Natl Acad Sci U S A ; 110(48): 19402-7, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24218566

ABSTRACT

Drastic metabolic alterations, such as the Warburg effect, are found in most if not all types of malignant tumors. Emerging evidence shows that cancer cells benefit from these alterations, but little is known about how they affect noncancerous stromal cells within the tumor microenvironment. Here we show that cancer cells are better adapted to metabolic changes in the microenvironment, leading to the emergence of spatial structure. A clear example of tumor spatial structure is the localization of tumor-associated macrophages (TAMs), one of the most common stromal cell types found in tumors. TAMs are enriched in well-perfused areas, such as perivascular and cortical regions, where they are known to potentiate tumor growth and invasion. However, the mechanisms of TAM localization are not completely understood. Computational modeling predicts that gradients--of nutrients, gases, and metabolic by-products such as lactate--emerge due to altered cell metabolism within poorly perfused tumors, creating ischemic regions of the tumor microenvironment where TAMs struggle to survive. We tested our modeling prediction in a coculture system that mimics the tumor microenvironment. Using this experimental approach, we showed that a combination of metabolite gradients and differential sensitivity to lactic acid is sufficient for the emergence of macrophage localization patterns in vitro. This suggests that cancer metabolic changes create a microenvironment where tumor cells thrive over other cells. Understanding differences in tumor-stroma sensitivity to these alterations may open therapeutic avenues against cancer.


Subject(s)
Glycolysis/physiology , Macrophages/physiology , Models, Biological , Neoplasms/metabolism , Tumor Microenvironment/physiology , Cell Line, Tumor , Fluorescent Antibody Technique , Humans , Hydrogen-Ion Concentration , Image Processing, Computer-Assisted , Macrophages/cytology , Microscopy, Fluorescence , Statistics, Nonparametric
16.
PLoS Pathog ; 9(3): e1003234, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555249

ABSTRACT

Exposure to hepatitis C virus (HCV) typically results in chronic infection that leads to progressive liver disease ranging from mild inflammation to severe fibrosis and cirrhosis as well as primary liver cancer. HCV triggers innate immune signaling within the infected hepatocyte, a first step in mounting of the adaptive response against HCV infection. Persistent inflammation is strongly associated with liver tumorigenesis. The goal of our work was to investigate the initiation of the inflammatory processes triggered by HCV viral proteins in their host cell and their possible link with HCV-related liver cancer. We report a dramatic upregulation of the lymphotoxin signaling pathway and more specifically of lymphotoxin-ß in tumors of the FL-N/35 HCV-transgenic mice. Lymphotoxin expression is accompanied by activation of NF-κB, neosynthesis of chemokines and intra-tumoral recruitment of mononuclear cells. Spectacularly, IKKß inactivation in FL-N/35 mice drastically reduces tumor incidence. Activation of lymphotoxin-ß pathway can be reproduced in several cellular models, including the full length replicon and HCV-infected primary human hepatocytes. We have identified NS5B, the HCV RNA dependent RNA polymerase, as the viral protein responsible for this phenotype and shown that pharmacological inhibition of its activity alleviates activation of the pro-inflammatory pathway. These results open new perspectives in understanding the inflammatory mechanisms linked to HCV infection and tumorigenesis.


Subject(s)
Hepacivirus/enzymology , Liver Neoplasms/metabolism , Lymphocytes/immunology , Lymphotoxin-beta/metabolism , RNA-Dependent RNA Polymerase/metabolism , Animals , Cell Line , Chemokines/metabolism , Chemotaxis, Leukocyte , Hepacivirus/pathogenicity , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Host-Pathogen Interactions , Humans , I-kappa B Kinase/metabolism , Immunity, Innate , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Liver/metabolism , Liver/pathology , Liver/virology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Lymphocyte Activation , Lymphocytes/virology , Male , Mice , Mice, Transgenic , NF-kappa B , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Signal Transduction , Up-Regulation , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism
17.
Cancer Cell ; 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39332398

ABSTRACT

The ability of disseminated cancer cells to colonize the brain is highly dependent on initial survival cues, often evoking early microenvironmental adaptations. In this issue of Cancer Cell, Gan et al. unveil disparate tumor architectures in early stage HER2+ breast cancer and triple-negative breast cancer brain metastases that shape stromal interactions, providing a rationale for subtype-dependent patient stratification.

18.
Nat Rev Cancer ; 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354070

ABSTRACT

Cancer and metabolic disorders have emerged as major global health challenges, reaching epidemic levels in recent decades. Often viewed as separate issues, metabolic disorders are shown by mounting evidence to heighten cancer risk and incidence. The intricacies underlying this connection are still being unraveled and encompass a complex interplay between metabolites, cancer cells and immune cells within the tumour microenvironment (TME). Here, we outline the interplay between metabolic and immune cell dysfunction in the context of three highly prevalent metabolic disorders, namely obesity; two associated liver diseases, metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH); and type 2 diabetes. We focus primarily on macrophages and T cells, the critical roles of which in dictating inflammatory response and immune surveillance in metabolic disorder-associated cancers are widely reported. Moreover, considering the ever-increasing number of patients prescribed with metabolism disorder-altering drugs and diets in recent years, we discuss how these therapies modulate systemic and local immune phenotypes, consequently impacting cancer malignancy. Collectively, unraveling the determinants of metabolic disorder-associated immune landscape and their role in fuelling cancer malignancy will provide a framework essential to therapeutically address these highly prevalent diseases.

19.
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
20.
Cancer Discov ; 14(1): 36-48, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38047596

ABSTRACT

Cancer cells adapt and survive through the acquisition and selection of molecular modifications. This process defines cancer evolution. Building on a theoretical framework based on heritable genetic changes has provided insights into the mechanisms supporting cancer evolution. However, cancer hallmarks also emerge via heritable nongenetic mechanisms, including epigenetic and chromatin topological changes, and interactions between tumor cells and the tumor microenvironment. Recent findings on tumor evolutionary mechanisms draw a multifaceted picture where heterogeneous forces interact and influence each other while shaping tumor progression. A comprehensive characterization of the cancer evolutionary toolkit is required to improve personalized medicine and biomarker discovery. SIGNIFICANCE: Tumor evolution is fueled by multiple enabling mechanisms. Importantly, genetic instability, epigenetic reprogramming, and interactions with the tumor microenvironment are neither alternative nor independent evolutionary mechanisms. As demonstrated by findings highlighted in this perspective, experimental and theoretical approaches must account for multiple evolutionary mechanisms and their interactions to ultimately understand, predict, and steer tumor evolution.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Neoplasms/pathology , Epigenomics , Precision Medicine , Tumor Microenvironment/genetics
SELECTION OF CITATIONS
SEARCH DETAIL