Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 5(5): 101528, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38677283

ABSTRACT

Stimulator of interferon genes (STING)-dependent signaling is requisite for effective anti-microbial and anti-tumor activity. STING signaling is commonly defective in cancer cells, which enables tumor cells to evade the immunosurveillance system. We evaluate here whether intrinsic STING signaling in such tumor cells could be reconstituted by creating recombinant herpes simplex viruses (rHSVs) that express components of the STING signaling pathway. We observe that rHSVs expressing STING and/or cGAS replicate inefficiently yet retain in vivo anti-tumor activity, independent of oncolytic activity requisite on the trans-activation of extrinsic STING signaling in phagocytes by engulfed microbial dsDNA species. Accordingly, the in vivo effects of virotherapy could be simulated by nanoparticles incorporating non-coding dsDNA species, which comparably elicit the trans-activation of phagocytes and augment the efficacy of established cancer treatments including checkpoint inhibition and radiation therapy. Our results help elucidate mechanisms of virotherapeutic anti-tumor activity as well as provide alternate strategies to treat cancer.


Subject(s)
DNA , Phagocytes , Animals , Phagocytes/immunology , Phagocytes/metabolism , Humans , Mice , DNA/metabolism , DNA/immunology , DNA/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Signal Transduction , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Cell Line, Tumor , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Neoplasms/genetics , Simplexvirus/genetics , Simplexvirus/immunology , Mice, Inbred C57BL , Oncolytic Virotherapy/methods
2.
Front Oncol ; 11: 695770, 2021.
Article in English | MEDLINE | ID: mdl-34249754

ABSTRACT

Homozygous deletion (HD) of the tumor suppressor gene CDKN2A is the most frequent genetic alteration in malignant pleural mesothelioma and is also frequent in non-small cell lung cancers. This HD is often accompanied by the HD of the type I interferons (IFN I) genes that are located closed to the CDKN2A gene on the p21.3 region of chromosome 9. IFN I genes encode sixteen cytokines (IFN-α, IFN-߅) that are implicated in cellular antiviral and antitumor defense and in the induction of the immune response. In this review, we discuss the potential influence of IFN I genes HD on thoracic cancers therapy and speak in favor of better taking these HD into account in patients monitoring.

3.
Mol Ther Oncolytics ; 18: 573-578, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32995481

ABSTRACT

Malignant pleural mesothelioma (MPM) is a cancer of the pleura that lacks efficient treatment. Oncolytic immunotherapy using oncolytic vaccinia virus (VV) may represent an alternative therapeutic approach for the treatment of this malignancy. Here, we studied the oncolytic activity of VV thymidine kinase (TK)-ribonucleotide reductase (RR)-/green fluorescent protein (GFP) against MPM. This virus is a VV from the Copenhagen strain that is deleted of two genes encoding the TK (J2R) and the RR (I4L) and that express the GFP. First, we show in vitro that VVTK-RR-/GFP efficiently infects and kills the twenty-two human MPM cell lines used in this study. We also show that the virus replicates in all eight tested MPM cell lines, however, with approximately a 10-fold difference in the amplification level from one cell line to another. Then, we studied the therapeutic efficiency of VVTK-RR-/GFP in non-obese diabetic (NOD) severe combined immunodeficient (SCID) mice that bear peritoneal human MPM tumors. One intraperitoneal infection of VVTK-RR-/GFP reduces the tumor burden and significantly increases mice survival compared to untreated animals. Thus, VVTK-RR - may be a promising oncolytic virus (OV) for the oncolytic immunotherapy of MPM.

5.
J Thorac Oncol ; 15(5): 827-842, 2020 05.
Article in English | MEDLINE | ID: mdl-31945495

ABSTRACT

INTRODUCTION: Oncolytic immunotherapy is based on the use of nonpathogenic replicative oncolytic viruses that infect and kill tumor cells exclusively. Recently, we found that the spontaneous oncolytic activity of the Schwarz strain of measles virus (MV) against human malignant pleural mesothelioma (MPM) depends on defects in the antiviral type I interferon (IFN-I) response in tumor cells. METHODS: In this study, we studied three independent human MPM bio-collections to identify the defects in the IFN-I responses in tumor cells. RESULTS: We show that the most frequent defect is the homozygous deletions (HDs) of all the 14 IFN-I genes (IFN-α and IFN-ß) that we found in more than half of MV-sensitive MPM cell lines. These HDs occur together with the HDs of the tumor suppressor gene CDKN2A also located in the 9p21.3 chromosome region. Therefore, the IFN-I-/- MPM cell lines develop a partial and weak IFN-I response when they are exposed to the virus compared with that of normal cells and MV-resistant MPM cell lines. This response consists of the expression of a restricted number of IFN-stimulated genes that do not depend on the presence of IFN-I. In addition, the IFN-I-/- MPM cell lines infected by MV also develop a pro-inflammatory response associated with stress of the endoplasmic reticulum. CONCLUSION: Our study emphasizes the link between HDs of IFN-I encoding genes and the CDKN2A gene in MPM and sensitivity to MV oncolytic immunotherapy.


Subject(s)
Interferon Type I , Lung Neoplasms , Mesothelioma , Oncolytic Virotherapy , Oncolytic Viruses , Cell Line, Tumor , Homozygote , Humans , Interferon Type I/genetics , Measles virus/genetics , Mesothelioma/genetics , Mesothelioma/therapy , Oncolytic Viruses/genetics , Sequence Deletion
6.
Cancer Lett ; 472: 29-39, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31838086

ABSTRACT

Toll-like receptor 3 (TLR3) is an immune receptor that behaves like a death receptor in tumor cells, thereby providing an original target for cancer therapy. The therapeutic potential of TLR3 targeting in malignant mesothelioma, an aggressive and incurable neoplasia of the pleura and peritoneum, has so far not been addressed. We investigated TLR3 expression and sensitivity of human mesothelioma cell lines to the synthetic dsRNA Poly(I:C), alone or in combination with cisplatin, the gold standard chemotherapy in mesothelioma. Activation of TLR3 by Poly(I:C) induced apoptosis of 4/8 TLR3-positive cell lines but not of TLR3-negative cell lines. The combined cisplatin/Poly(I:C) treatment enhanced apoptosis of 3/4 Poly(I:C)-sensitive cell lines and overcame resistance to Poly(I:C) or cisplatin alone in 2/4 cell lines. Efficacy of the combined treatment relied on cisplatin-induced downregulation of c-FLIP, the main regulator of the extrinsic apoptotic pathway, leading to an enhanced caspase-8-mediated pathway. Of note, 6/6 primary cell samples isolated from patients with peritoneal mesothelioma expressed TLR3. Patient-derived cells were sensitive to Poly(I:C) alone while the combined cisplatin/Poly(I:C) treatment induced dramatic cell death. Our findings demonstrate that TLR3 targeting in combination with cisplatin presents an innovative therapeutic strategy in mesothelioma.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Toll-Like Receptor 3/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Caspase 8/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Mesothelioma/genetics , Mesothelioma/physiopathology , Mesothelioma, Malignant , Poly I-C/pharmacology , Signal Transduction/drug effects
7.
Methods Mol Biol ; 2058: 127-132, 2020.
Article in English | MEDLINE | ID: mdl-31486035

ABSTRACT

Oncolytic immunotherapy efficacy relies partially on the induction of immunogenic tumor cell death following infection with oncolytic viruses (OV) to induce an antitumor immune response. Here, we describe a method to determine if an OV is able to induce such an immunogenic tumor cell death. This method consists in testing whether tumor cells lysed by an OV are able to induce the maturation of human monocyte-derived immature dendritic cells (Mo-iDC).


Subject(s)
Genetic Vectors/genetics , Immunomodulation , Neoplasms/immunology , Oncolytic Viruses/genetics , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Death/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Genetic Therapy/methods , Humans , Immunophenotyping , Monocytes/immunology , Monocytes/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology
8.
Biol Chem ; 399(9): 1099-1105, 2018 09 25.
Article in English | MEDLINE | ID: mdl-29498930

ABSTRACT

We recently reported that human melanoma cells, but not benign melanocytes, aberrantly express kallikrein-related peptidase 7 (KLK7). Here, we show a KLK7 overexpression-mediated decrease of cell adhesion to extracellular matrix binding proteins, associated with downregulation of α5/ß1/αv/ß3 integrin expression. We also report an up-regulation of MCAM/CD146 and an increase in spheroid formation of these cells. Our results demonstrate that aberrant KLK7 expression leads to a switch to a more malignant phenotype suggesting a potential role of KLK7 in melanoma invasion. Thus, KLK7 may represent a biomarker for melanoma progression and may be a potential therapeutic target for melanoma.


Subject(s)
Kallikreins/genetics , Kallikreins/metabolism , Melanoma/genetics , Melanoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Adhesion/genetics , Down-Regulation , Humans , Integrins/biosynthesis , Melanoma/metabolism , Phenotype
9.
Oncoimmunology ; 7(3): e1407897, 2018.
Article in English | MEDLINE | ID: mdl-29399408

ABSTRACT

Oncolytic immunotherapy using oncolytic viruses (OV) has been shown to stimulate the antitumor immune response by inducing the release of tumor-associated antigens (TAA) and danger signals from the dying infected tumor cells. In this study, we sought to determine if the lysis of tumor cells induced by different OV: measles virus, vaccinia virus, vesicular stomatitis virus, herpes simplex type I virus, adenovirus or enterovirus, has consequences on the capacity of tumor cells to present TAA, such as NY-ESO-1. We show that the co-culture of NY-ESO-1neg/HLA-DP4pos melanoma cells with NY-ESO-1pos/HLA-DP4neg melanoma cells infected and killed by different OV induces an intercellular transfer of NY-ESO-1 that allows the recognition of NY-ESO-1neg/HLA-DP4pos tumor cells by an HLA-DP4/NY-ESO-1(157-170)-specific CD4+ cytotoxic T cell clone, NY67. We then confirmed this result in a second model with an HLA-DP4+ melanoma cell line that expresses a low amount of NY-ESO-1. Recognition of this cell line by the NY67 clone is largely increased in the presence of OV productive infection. Altogether, our results show for the first time another mechanism of stimulation of the anti-tumor immune response by OV, via the loading of tumor cells with TAA that sensitizes them for direct recognition by specific effector CD4+ T cells, supporting the use of OV for cancer immunotherapy.

10.
Mol Oncol ; 11(10): 1330-1347, 2017 10.
Article in English | MEDLINE | ID: mdl-28636767

ABSTRACT

Members of the tissue kallikrein-related peptidase (KLK) family not only regulate several important physiological functions, but aberrant expression has also been associated with various malignancies. Clinically, KLKs have been suggested as promising biomarkers for diagnosis and prognosis in many types of cancer. As of yet, expression of KLKs and their role in skin cancers are, however, poorly addressed. Malignant melanoma is an aggressive disease associated with poor prognosis. Hence, diagnostic biomarkers to monitor melanoma progression are needed. Herein, we demonstrate that although mRNA of several KLKs are aberrantly expressed in melanoma cell lines, only the KLK7 protein is highly secreted in vitro. In line with these findings, ectopic expression of KLK7 in human melanomas and its absence in benign nevi were demonstrated by immunohistochemistry in vivo. Interestingly, overexpression of KLK7 induced a significant reduction in melanoma cell proliferation and colony formation. Moreover, KLK7 overexpression triggered an increase in cell motility and invasion associated with decreased expression of E-cadherin and an upregulation of MCAM/CD146. Our results demonstrate, for the first time, that aberrant KLK7 expression leads to a switch from proliferative to invasive phenotype, suggesting a potential role of KLK7 in melanoma progression. Thus, we hypothesize that KLK7 may represent a potential biomarker for melanoma progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Kallikreins/genetics , Melanoma/genetics , Neoplasm Invasiveness/genetics , Skin Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Humans , Kallikreins/analysis , Kallikreins/metabolism , MAP Kinase Signaling System , Melanoma/metabolism , Melanoma/pathology , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
11.
Oncotarget ; 6(42): 44892-904, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26539644

ABSTRACT

Attenuated measles virus (MV) is currently being evaluated as an oncolytic virus in clinical trials and could represent a new therapeutic approach for malignant pleural mesothelioma (MPM). Herein, we screened the sensitivity to MV infection and replication of twenty-two human MPM cell lines and some healthy primary cells. We show that MV replicates in fifteen of the twenty-two MPM cell lines. Despite overexpression of CD46 by a majority of MPM cell lines compared to healthy cells, we found that the sensitivity to MV replication did not correlate with this overexpression. We then evaluated the antiviral type I interferon (IFN) responses of MPM cell lines and healthy cells. We found that healthy cells and the seven insensitive MPM cell lines developed a type I IFN response in presence of the virus, thereby inhibiting replication. In contrast, eleven of the fifteen sensitive MPM cell lines were unable to develop a complete type I IFN response in presence of MV. Finally, we show that addition of type I IFN onto MV sensitive tumor cell lines inhibits replication. These results demonstrate that defects in type I IFN response are frequent in MPM and that MV takes advantage of these defects to exert oncolytic activity.


Subject(s)
Interferon Type I/metabolism , Measles virus/growth & development , Mesothelioma/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/growth & development , Pleural Neoplasms/therapy , Virus Replication , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Host-Pathogen Interactions , Humans , Interferon Type I/immunology , Measles virus/immunology , Measles virus/metabolism , Membrane Cofactor Protein/metabolism , Mesothelioma/immunology , Mesothelioma/metabolism , Mesothelioma/virology , Oncolytic Viruses/immunology , Oncolytic Viruses/metabolism , Pleural Neoplasms/immunology , Pleural Neoplasms/metabolism , Pleural Neoplasms/virology , Receptors, Cell Surface/metabolism , Signal Transduction , Signaling Lymphocytic Activation Molecule Family Member 1 , Time Factors
12.
Oncolytic Virother ; 4: 133-40, 2015.
Article in English | MEDLINE | ID: mdl-27512676

ABSTRACT

Cancer virotherapy is an attractive alternative to conventional treatments because it offers a wide range of antitumor effects due to 1) the diversity of the oncolytic viruses that are now available and 2) their multifaceted activities against both tumor cells and tumor vessels, in addition to their ability to induce antitumor immune responses. In this review, we summarize preclinical and clinical data regarding the targeting of malignant mesothelioma (MM) by oncolytic viruses. We also discuss the potential of other oncolytic viruses that have already shown antitumor effects against several malignancies in advanced clinical trials but are yet to be tested against MM cells. Finally, we review how the activation of the immune system and combinations with other types of anticancer treatments could support the development of oncolytic virotherapy for the treatment of MM.

SELECTION OF CITATIONS
SEARCH DETAIL
...