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.
Acta Neuropathol Commun ; 8(1): 221, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33308315

ABSTRACT

Cancer-associated fibroblasts (CAFs) are activated fibroblasts constituting the major stromal components in many types of cancer. CAFs contribute to hallmarks of cancer such as proliferation, invasion and immunosuppressive tumor microenvironment, and are associated with poor prognosis of patients with cancer. However, in glioblastoma (GBM), the most common and aggressive primary malignant brain tumor, our knowledge about CAFs or CAF-like stromal cells is limited. Here, using commonly accepted CAF markers, we characterized CAF-like cell populations in clinical glioma specimens and datasets along with mouse models of GBM. We found that tumor-associated pericytes marked by co-expression of fibroblast activation protein α (FAP) and PDGFRß represent major stromal cell subsets in both human GBM and mouse GBM models, while a fraction of mesenchymal neoplastic cells also express FAP in patient tumors. Since oncolytic viruses can kill cancer cells and simultaneously modulate the tumor microenvironment by impacting non-neoplastic populations such as immune cells and tumor vasculature, we further investigated the ability of oncolytic viruses to target GBM-associated stromal cells. An oncolytic adenovirus, ICOVIR15, carrying ∆24-E1A and an RGD-fiber, infects and depletes FAP+ pericytes as well as GBM cells in murine GBM. Our study thus identifies FAP+/PDGFRß+ pericytes as a major CAF-like stromal cell population in GBM, and highlights the unique property of this oncolytic adenovirus to target both GBM cells and GBM-associated stromal FAP+ cells.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Endopeptidases/metabolism , Glioblastoma/metabolism , Membrane Proteins/metabolism , Oncolytic Viruses , Pericytes/metabolism , Animals , Cancer-Associated Fibroblasts/cytology , Cancer-Associated Fibroblasts/virology , Disease Models, Animal , Glioblastoma/pathology , Humans , Mice , Oncolytic Virotherapy , Pericytes/cytology , Pericytes/virology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/virology , Tumor Microenvironment
2.
Pharmacol Res ; 161: 105094, 2020 11.
Article in English | MEDLINE | ID: mdl-32795509

ABSTRACT

Oncolytic virus (OV) immunotherapy has demonstrated to be a promising approach in cancer treatment due to tumor-specific oncolysis. However, their clinical use so far has been largely limited due to the lack of suitable delivery strategies with high efficacy. Direct 'intratumoral' injection is the way to cross the hurdles of systemic toxicity, while providing local effects. Progress in this field has enabled the development of alternative way using 'systemic' oncolytic virotherapy for producing better results. One major potential roadblock to systemic OV delivery is the low virus persistence in the face of hostile immune system. The delivery challenge is even greater when attempting to target the oncolytic viruses into the entire tumor mass, where not all tumor cells are equally exposed to exactly the same microenvironment. The microenvironment of many tumors is known to be massively infiltrated with various types of leucocytes in both primary and metastatic sites. Interestingly, this intratumoral immune cell heterogeneity exhibits a degree of organized distribution inside the tumor bed as evidenced, for example, by the hypoxic tumor microenviroment where predominantly recruits tumor-associated macrophages. Although in vivo OV delivery seems complicated and challenging, recent results are encouraging for decreasing the limitations of systemically administered oncolytic viruses and an improved efficiency of oncolytic viral therapy in targeting cancerous tissues in vitro. Here, we review the latest developments of carrier cell-based oncolytic virus delivery using tumor-infiltrating immune cells with a focus on the main features of each cellular vehicle.


Subject(s)
Cancer-Associated Fibroblasts/virology , Cytokine-Induced Killer Cells/virology , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/virology , Monocytes/virology , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/pathogenicity , T-Lymphocytes/virology , Animals , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/transplantation , Cytokine-Induced Killer Cells/immunology , Cytokine-Induced Killer Cells/transplantation , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Monocytes/immunology , Monocytes/transplantation , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/virology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/virology , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Phenotype , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Tumor Hypoxia , Tumor Microenvironment
3.
Cancer Lett ; 478: 93-106, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32160975

ABSTRACT

Several reports have demonstrated that Epstein-Barr virus (EBV) encoded latent membrane protein 1 (LMP1), which is transferred by extracellular vesicles (EVs) or exosomes, can promote cancer progression. However, its mechanism is still not fully understood. In the present study, we demonstrated that EV packaged LMP1 can activate normal fibroblasts (NFs) into cancer-associated fibroblasts (CAFs). The NF-κB p65 pathway is the key signal that promotes the activation of NFs to CAFs in nasopharyngeal carcinoma (NPC). In activated CAFs, aerobic glycolysis and autophagy were increased. Moreover, glucose uptake and lactate production were decreased, and mitochondrial activity in tumor cells was enhanced, which supported the Reverse Warburg Effect (RWE). During this process, upregulation of MCT4 in CAFs and MCT1 in tumor cells was observed. The NF-κB p65 pathway also plays an important role in the regulation of MCT4. Furthermore, co-culture with CAFs promoted the proliferation, migration and radiation resistance of NPC cells. And EV packaged LMP1 promoted tumor proliferation and pre-metastatic niche formation by activating CAFs in vivo. Our findings indicate that EV packaged LMP1-activated CAFs promote tumor progression via autophagy and stroma-tumor metabolism coupling.


Subject(s)
Extracellular Vesicles/metabolism , Glucose/metabolism , Herpesvirus 4, Human/metabolism , Lactic Acid/metabolism , Nasopharyngeal Neoplasms/virology , Viral Matrix Proteins/metabolism , Autophagy , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/virology , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Coculture Techniques , Disease Progression , Humans , Mitochondria/metabolism , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Nasopharyngeal Neoplasms/metabolism , Symporters/metabolism
4.
Mol Cancer Res ; 15(10): 1410-1420, 2017 10.
Article in English | MEDLINE | ID: mdl-28679779

ABSTRACT

The tumor microenvironment (TME) is a relevant target for novel biological therapies. MV-m-uPA and MV-h-uPA are fully retargeted, species-specific, oncolytic measles viruses (MV) directed against murine or human urokinase receptor (PLAUR/uPAR), expressed in tumor and stromal cells. The effects of stromal-selective targeting by uPAR-retargeted MVs were investigated. In vitro infection, virus-induced GFP expression, and cytotoxicity by MV-h-uPA and MV-m-uPA were demonstrated in human and murine cancer cells and cancer-associated fibroblasts in a species-specific manner. In a murine fibroblast/human breast cancer 3D coculture model, selective fibroblast targeting by MV-m-uPA inhibited breast cancer cell growth. Systemic administration of murine-specific MV-m-uPA in mice bearing human MDA-MB-231 xenografts was associated with a significant delay in tumor progression and improved survival compared with controls. Experiments comparing tumor (MV-h-uPA) versus stromal (MV-m-uPA) versus combined virus targeting showed that tumor and stromal targeting was associated with improved tumor control over the other groups. Correlative studies confirmed in vivo viral targeting of tumor stroma by MV-m-uPA, increased apoptosis, and virus-induced differential regulation of murine stromal genes associated with inflammatory, angiogenesis, and survival pathways, as well as indirect regulation of human cancer pathways, indicating viral-induced modulation of tumor-stroma interactions. These data demonstrate the feasibility of stromal-selective targeting by an oncolytic MV, virus-induced modulation of tumor-stroma pathways, and subsequent tumor growth delay. These findings further validate the critical role of stromal uPAR in cancer progression and the potential of oncolytic viruses as antistromal agents.Implications: The current report demonstrates for the first time the biological, in vitro, and in vivo antitumor and molecular effects of stromal selective targeting by an oncolytic virus. Mol Cancer Res; 15(10); 1410-20. ©2017 AACR.


Subject(s)
Breast Neoplasms/therapy , Measles virus/physiology , Oncolytic Viruses/physiology , Receptors, Urokinase Plasminogen Activator/genetics , Stromal Cells/cytology , Animals , Breast Neoplasms/genetics , Cancer-Associated Fibroblasts/cytology , Cancer-Associated Fibroblasts/virology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Female , HT29 Cells , Humans , Mice , Oncolytic Virotherapy , Stromal Cells/virology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...