Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
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
2.
Cell Mol Biol (Noisy-le-grand) ; 66(2): 36-40, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32415924

ABSTRACT

Hepatocellular carcinoma is known to be a common predominant cancer in adults, especially in eastern countries. Immune response and cancer-associated fibroblasts (CAFs) have significant influences on tumor development. However, the interaction between CAFs and immunotherapy is unclear in hepatocellular carcinoma. We measured the number of activated fibroblasts in hepatocellular carcinoma samples and samples taken from normal liver tissues. A total of 20 patients' fresh hepatocellular carcinoma and normal tissues which were surrounding the tumor were obtained from the surgery and used for evaluating alpha-SMA expression. We investigated the effects of CAFs in anti-tumor immunity in hepatocellular carcinoma animal model. The effects of CAFs in inducing anti-PD-1 treatment resistance were also measured in a preclinical animal model. Activated fibroblasts were highly accumulated in hepatocellular carcinoma tissues but not in surrounding normal tissues. CAFs showed a significant tumor-promoting effect in an immunocompetent model. The infiltration and function of some immune cells like myeloid-derived suppressive cells and T-cells were increased by CAFs. CAFs also reduced the number and activation of tumor-infiltrating cytotoxic T-cell in tumor tissue. In the treatment model, tumors with a higher amount of CAFs had been insensitive to therapy with anti-PD-1. CAFs are potent inducers of immunosuppression in hepatocellular carcinoma. Depleting CAFs rescued the antitumor immunity in the hepatocellular model and could be a novel treatment to combine with the existing immunotherapy.


Subject(s)
Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Actins/metabolism , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Cancer-Associated Fibroblasts/cytology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/transplantation , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cytokines/metabolism , Dasatinib/therapeutic use , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Humans , Immunotherapy , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
3.
Cancer Sci ; 110(5): 1790-1803, 2019 May.
Article in English | MEDLINE | ID: mdl-30843645

ABSTRACT

Heat shock factor 1 (HSF1) is highly expressed in various malignancies and is a potential modulator of tumor progression. Emerging evidence suggests that HSF1 activation in stromal cells is closely related to poor patient prognosis. However, the role of HSF1 in oral squamous cell carcinoma (OSCC) remains elusive. We aimed to investigate the function of HSF1 in cancer-associated fibroblasts (CAFs) of the tumor microenvironment (TME) and in tumor development. In the present study, we found that HSF1 was highly expressed in both CAFs and tumor cells, and was significantly correlated with poor prognosis and overall survival. Moreover, HSF1 overexpression in CAFs resulted in a fibroblast-like phenotype of Cal27 cells, induced epithelial-mesenchymal transition (EMT), and promoted proliferation, migration and invasion in Cal27 cells. HSF1 knockdown attenuated features of CAFs and reduced EMT, proliferation, migration and invasion in Cal27 cells. Furthermore, HSF1 in CAFs promoted tumor growth in nude mice. Taken together, these data suggest that HSF1 expression in CAFs drive OSCC progression, and could serve as an independent prognostic marker of patients with OSCC. Thus, HSF1 is a potent mediator of OSCC malignancy.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/pathology , Heat Shock Transcription Factors/metabolism , Mouth Neoplasms/pathology , Up-Regulation , Adult , Aged , Aged, 80 and over , Animals , Cancer-Associated Fibroblasts/transplantation , Carcinoma, Squamous Cell/metabolism , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , Middle Aged , Mouth Neoplasms/metabolism , Neoplasm Staging , Neoplasm Transplantation , Prognosis , Survival Analysis , Tumor Microenvironment
4.
Clin Cancer Res ; 23(16): 4769-4779, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28420724

ABSTRACT

Purpose: Cancer metastasis can occur at the early stage of tumor development when a primary tumor is at the microscopic size. In particular, the interaction of malignant cells with other cell types including cancer-associated fibroblasts (CAF) in promoting metastasis at the early stage of tumor development remains largely unknown. Here, we investigated the role of CAFs in facilitating the initial events of cancer metastasis when primary tumors were at microscopic sizes.Experimental Design: Multicolor-coded cancer cells and CAFs were coimplanted into the transparent zebrafish body and metastasis at a single-cell level was monitored in living animals. Healthy fibroblasts, tumor factor-educated fibroblasts, and CAFs isolated from various tumors were tested for their ability to facilitate metastasis.Results: We showed that CAFs promoted cancer cell metastasis at the very early stage during primary tumor development. When a primary tumor was at the microscopic size consisting of a few hundred cells, CAFs were able to hijack cancer cells for dissemination from the primary site. Surprisingly, a majority of metastatic cancer cells remained in tight association with CAFs in the circulation. Furthermore, stimulation of non-metastasis-promoting normal fibroblasts with TGF-B, FGF-2, HGF, and PDGF-BB led to acquisition of their metastatic capacity.Conclusions: Cancer metastasis occurs at the very early stage of tumor formation consisting of only a few hundred cells. CAFs are the key cellular determinant for metastasis. Our findings provide novel mechanistic insights on CAFs in promoting cancer metastasis and targeting CAFs for cancer therapy should be aimed at the early stage during cancer development. Clin Cancer Res; 23(16); 4769-79. ©2017 AACR.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Embryo, Nonmammalian/pathology , Neoplasms/pathology , Zebrafish/embryology , A549 Cells , Animals , Animals, Genetically Modified , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/transplantation , Cell Line, Tumor , Cell Movement/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/metabolism , Transplantation, Heterologous , Zebrafish/genetics
5.
Cancer Lett ; 385: 39-45, 2017 01 28.
Article in English | MEDLINE | ID: mdl-27826041

ABSTRACT

Tumor-associated-fibroblasts (TAFs) are the most important host cells in the stroma and take part in extracellular matrix construction and cancer colony development. During cancer colonization, seed cells from primary tumor can reconstruct the microenvironment by recruiting circulating cancer cells and TAFs to the metastasis site. Previous studies have established that SMC1A, a subunit of cohesin, is an important trigger signal for liver metastasis in colorectal cancer. We investigated the particular effects as well as the underlying mechanism of SMC1A on TAFs recruitment during liver metastasis of colorectal cancer. Here, We found that: first, the high expression of SMC1A in colorectal cancer cells promotes the invasiveness and the viability of these cells by recruiting circulating TAFs, facilitating early tumor construction and tumorigenesis; second, different expression levels of SMC1A influenced the reformation of fibroblasts, which assisted tumorigenesis, and third, expression of SMC1A stimulated the secretion of the inflammatory mediators of TNF-α and IL-1ß, and up-regulated the transcriptional expression of MMP2 and VEGF-ß, both of which were involved in the tumor-related gene pathway.


Subject(s)
Adenocarcinoma/metabolism , Cancer-Associated Fibroblasts/metabolism , Cell Cycle Proteins/metabolism , Chemotaxis , Chromosomal Proteins, Non-Histone/metabolism , Colorectal Neoplasms/metabolism , Liver Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Animals , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/transplantation , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Interleukin-1beta/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor B/metabolism
6.
Cancer Lett ; 383(1): 18-27, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27693460

ABSTRACT

Vascular mimicry (VM) describes the phenomenon that tumor cells but not endothelial cells form vascular-like channels, which provide blood perfusion for tumor tissues. VM is associated with tumor growth, metastasis and worse survival of different cancers. The mechanisms of VM formation remain largely unknown. We showed that the conditioned medium of cancer-associated fibroblast (CM-CAF) promoted tumor cells to form capillary-like structure in vitro. Consistently, co-implantation of CAFs with tumor cells significantly enhanced VM formation in mouse xenografts, and higher amount of CAFs was found in VM+ human HCC tissues compared to VM- ones. However, the CM-CAF-promoted VM formation was attenuated when TGF-ß or SDF1 signaling was abrogated. Similar to CM-CAF, recombinant TGF-ß1 and SDF1 induced VM formation. We further disclosed that the CAF-secreted TGF-ß and SDF1 enhanced the expression of VE-cadherin, MMP2 and laminin5γ2 via TGF-ßR1 and CXCR4 in tumor cells, thereby promoted VM formation. Moreover, tumor cells with high activity of self-sustaining TGF-ß signaling displayed strong capability of VM formation. Subsequent investigations showed that miR-101, which was down-regulated in both tumor cells and CAFs, suppressed the CAF-promoted VM formation in vitro and in vivo. Gain- and loss-of-function analyses revealed that miR-101 attenuated TGF-ß signaling transduction by targeting TGF-ßR1 and Smad2 in tumor cells, and simultaneously abrogated SDF1 signaling by suppressing SDF1 expression in CAFs and inhibiting VE-cadherin expression in tumor cells. Our findings suggest that the miR-101-TGF-ß/SDF1-VE-cadherin/MMP2/LAMC2 networks regulate VM formation and represent the potential targets for cancer therapy.


Subject(s)
Biological Mimicry , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Hepatocellular/metabolism , Chemokine CXCL12/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Neovascularization, Pathologic , Paracrine Communication , Transforming Growth Factor beta1/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/transplantation , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Female , Humans , Laminin/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Time Factors , Transfection , Tumor Cells, Cultured
7.
Sci Rep ; 6: 21782, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26899873

ABSTRACT

Cancer-associated fibroblasts (CAFs), the principal components of the tumor stroma, play a central role in cancer development and progression. As an important regulator of the crosstalk between breast cancer cells and CAFs, the cytokine leptin has been associated to breast carcinogenesis. The nuclear Farnesoid X Receptor-(FXR) seems to exert an oncosuppressive role in different tumors, including breast cancer. Herein, we demonstrated, for the first time, that the synthetic FXR agonist GW4064, inhibiting leptin signaling, affects the tumor-promoting activities of CAFs in breast malignancy. GW4064 inhibited growth, motility and invasiveness induced by leptin as well as by CAF-conditioned media in different breast cancer cell lines. These effects rely on the ability of activated FXR to increase the expression of the suppressor of the cytokine signaling 3 (SOCS3) leading to inhibition of leptin-activated signaling and downregulation of leptin-target genes. In vivo xenograft studies, using MCF-7 cells alone or co-injected with CAFs, showed that GW4064 administration markedly reduced tumor growth. Interestingly, GW4064-treated tumors exhibited decreased levels of leptin-regulated proteins along with a strong staining intensity for SOCS3. Thus, FXR ligands might represent an emerging potential anti-cancer therapy able to block the tumor supportive role of activated fibroblasts within the breast microenvironment.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cancer-Associated Fibroblasts/drug effects , Gene Expression Regulation, Neoplastic , Isoxazoles/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/transplantation , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Communication , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Female , Humans , Leptin/genetics , Leptin/metabolism , MCF-7 Cells , Mice, Nude , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL