Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Int J Cancer ; 136(5): 1085-94, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25046660

ABSTRACT

Dendritic cells (DCs) cross-present antigen (Ag) to initiate T-cell immunity against most infections and tumors. Natural killer (NK) cells are innate cytolytic lymphocytes that have emerged as key modulators of multiple DC functions. Here, we show that human NK cells promote cross-presentation of tumor cell-derived Ag by DC leading to Ag-specific CD8(+) T-cell activation. Surprisingly, cytotoxic function of NK cells was not required. Instead, we highlight a critical and nonredundant role for IFN-γ and TNF-α production by NK cells to enhance cross-presentation by DC using two different Ag models. Importantly, we observed that NK cells promote cell-associated Ag cross-presentation selectively by monocytes-derived DC (Mo-DC) and CD34-derived CD11b(neg) CD141(high) DC subsets but not by myeloid CD11b(+) DC. Moreover, we demonstrate that triggering NK cell activation by monoclonal antibodies (mAbs)-coated tumor cells leads to efficient DC cross-presentation, supporting the concept that NK cells can contribute to therapeutic mAbs efficiency by inducing downstream adaptive immunity. Taken together, our findings point toward a novel role of human NK cells bridging innate and adaptive immunity through selective induction of cell-associated Ag cross-presentation by CD141(high) DC, a process that could be exploited to better harness Ag-specific cellular immunity in immunotherapy.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Antibody-Dependent Cell Cytotoxicity , Dendritic Cells/pathology , Humans , Killer Cells, Natural/pathology , Neoplasms/pathology , Tumor Cells, Cultured
2.
FASEB J ; 28(2): 740-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24225148

ABSTRACT

KLK12, a kallikrein peptidase, is thought to take part in the control of angiogenesis. Our analysis of the secretome of endothelial cells (ECs) that had been treated with KLK12 showed that KLK12 converts the extracellular matrix- or membrane-bound precursor of platelet-derived growth factor B (PDGF-B) into a soluble form. Both PDGF-B and vascular endothelial growth factor A (VEGF-A) take part in the induction of angiogenesis by KLK12 in a coculture model of angiogenesis that mimics endothelial tubule formation. We used a cellular approach to analyze the interplay between KLK12, PDGF-B, and VEGF-A and showed that release of PDGF-B by KLK12 leads to the fibroblast-mediated secretion of VEGF-A. This then stimulates EC differentiation and the formation of capillary tube-like structures. Thus, KLK12 favors the interaction of ECs and stromal cells. The released PDGF-B acts as a paracrine factor that modulates VEGF-A secretion by stromal cells, which ultimately leads to angiogenesis. Moreover, the genes encoding KLK12 and PDGFB are both expressed in ECs and up-regulated in tumor cells kept under hypoxic conditions, which is consistent with the physiological involvement of KLK12 in PDGF-B maturation.


Subject(s)
Kallikreins/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Vascular Endothelial Growth Factor A/metabolism , Blotting, Western , Cell Line , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fluorescent Antibody Technique , Humans , Kallikreins/pharmacology , Mass Spectrometry , Proto-Oncogene Proteins c-sis/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
3.
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
4.
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
5.
EBioMedicine ; 31: 17-24, 2018 May.
Article in English | MEDLINE | ID: mdl-29724655

ABSTRACT

Oncolytic virus (OV) therapy is potentially a game-changing cancer treatment that has garnered significant interest due to its versatility and multi-modal approaches towards tumor eradication. In the field of cancer immunotherapy, the immunological phenotype of the tumor microenvironment (TME) is an important determinant of disease prognosis and therapeutic success. There is accumulating data that OVs are capable of dramatically altering the TME immune landscape, leading to improved antitumor activity alone or in combination with assorted immune modulators. Herein, we review how OVs disrupt the immunosuppressive TME and can be used strategically to create a "pro-immune" microenvironment that enables and promotes potent, long-lasting host antitumor immune responses.


Subject(s)
Neoplasms/immunology , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Tumor Microenvironment/immunology , Animals , Humans , Neoplasms/pathology
6.
Oncoimmunology ; 6(1): e1261240, 2017.
Article in English | MEDLINE | ID: mdl-28197384

ABSTRACT

Attenuated measles virus (MV) is currently being evaluated in clinical trials as an oncolytic therapeutic agent. Originally used for its lytic activity against tumor cells, it is now admitted that the effectiveness of MV also lies in its ability to initiate antitumor immune responses through the activation of dendritic cells (DCs). In this study, we investigated the capacity of oncolytic MV to convert human blood myeloid CD1c+ DCs and plasmacytoid DCs (pDCs) into cytotoxic effectors. We found that MV induces the expression of the cytotoxic protein TNF-related apoptosis-inducing ligand (TRAIL) on the surface of DCs. We demonstrate that the secretion of interferon-α (IFN-α) by DCs in response to MV is responsible for this TRAIL expression. Several types of PRRs (pattern recognition receptors) have been implicated in MV genome recognition, including RLRs (RIG-I-like receptors) and TLRs (Toll-like receptors). We showed that CD1c+ DCs secrete modest amounts of IFN-α and express TRAIL in an RLR-dependent manner upon exposure to MV. In pDCs, MV is recognized by RLRs and also by TLR7, leading to the secretion of high amounts of IFN-α and TRAIL expression. Finally, we showed that MV-stimulated DCs induce TRAIL-mediated cell death of Jurkat cells, confirming their acquisition of cytotoxic functions. Our results demonstrate that MV can activate cytotoxic myeloid CD1c+ DCs and pDCs, which may participate to the antitumor immune response.

7.
Curr Gene Ther ; 16(6): 419-428, 2017.
Article in English | MEDLINE | ID: mdl-28042780

ABSTRACT

BACKGROUND: Oncolytic viruses such as live-attenuated, vaccine strains of measles virus (MV) have recently emerged as promising cancer treatments, having shown significant antitumor activity against a large variety of human tumors. OBJECTIVE: Our study aims at determining which parameters define the sensitivity of human melanoma cells to oncolytic MV infection. METHODS: We analyzed both in vitro and in vivo the oncolytic activity of MV against a panel of human melanoma cell established in our laboratory. We tested whether either type I interferons or the interferon pathway inhibitor Ruxolitinib could modulate the sensitivity of these cells to oncolytic MV infection. RESULTS: Human melanoma cells exhibit varying levels of sensitivity to MV infection in culture and as tumor xenografts. As these differences are not explained by their expression level of the CD46 receptor, we hypothesized that antiviral immune responses may be suppressed in certain cell resulting in their inability to control infection efficiently. By analyzing the type I IFN response, we found that resistant cells had a fully functional pathway that was activated upon MV infection. On the contrary, sensitive cell showed defects in this pathway. When pre-treated with IFN-α and IFN-ß, all but one of the sensitive cell became resistant to MV. Cells resistant to MV were rendered sensitive to MV with Ruxolitinib. CONCLUSION: Type I interferon response is the main determinant for the sensitivity or resistance of melanoma to oncolytic MV infection. This will have to be taken into account for future clinical trials on oncolytic MV.


Subject(s)
Interferon Type I/therapeutic use , Measles virus/genetics , Melanoma/therapy , Oncolytic Virotherapy , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Interferon Type I/genetics , Melanoma/genetics , Melanoma/virology , Membrane Cofactor Protein/genetics , Mice , Oncolytic Viruses/genetics , Xenograft Model Antitumor Assays
8.
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.

9.
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
10.
Biomed Res Int ; 2013: 387362, 2013.
Article in English | MEDLINE | ID: mdl-23586034

ABSTRACT

Lung and colorectal cancers are responsible for approximately 2 million deaths each year worldwide. Despite continual improvements, clinical management of these diseases remains challenging and development of novel therapies with increased efficacy is critical to address these major public health issues. Oncolytic viruses have shown promising results against cancers that are resistant to conventional anticancer therapies. Vaccine strains of measles virus (MV) exhibit such natural antitumor properties by preferentially targeting cancer cells. We tested the ability of live-attenuated Schwarz strain of MV to specifically infect tumor cells derived from human lung and colorectal adenocarcinomas and demonstrated that live-attenuated MV exhibits oncolytic properties against these two aggressive neoplasms. We also showed that Schwarz MV was able to prevent uncontrollable growth of large, established lung and colorectal adenocarcinoma xenografts in nude mice. Moreover, MV oncolysis is associated with in vivo activation of caspase-3 in colorectal cancer model, as shown by immunohistochemical staining. Our results provide new arguments for the use of MV as an antitumor therapy against aggressive human malignancies.


Subject(s)
Adenocarcinoma/therapy , Colorectal Neoplasms/therapy , Lung Neoplasms/therapy , Oncolytic Virotherapy , Vaccines, Attenuated/administration & dosage , Adenocarcinoma/pathology , Adenocarcinoma/virology , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Colorectal Neoplasms/virology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/virology , Measles/prevention & control , Measles/virology , Measles Vaccine/administration & dosage , Measles virus/pathogenicity , Mice , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL