Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Oncol ; 12(8): 1249-1263, 2018 08.
Article in English | MEDLINE | ID: mdl-29738110

ABSTRACT

We describe a cell damage-induced phenotype in mammary carcinoma cells involving acquisition of enhanced migratory and metastatic properties. Induction of this state by radiation required increased activity of the Ptgs2 gene product cyclooxygenase 2 (Cox2), secretion of its bioactive lipid product prostaglandin E2 (PGE2), and the activity of the PGE2 receptor EP4. Although largely transient, decaying to low levels in a few days to a week, this phenotype was cumulative with damage and levels of cell markers Sca-1 and ALDH1 increased with treatment dose. The Sca-1+ , metastatic phenotype was inhibited by both Cox2 inhibitors and PGE2 receptor antagonists, suggesting novel approaches to radiosensitization.


Subject(s)
Antigens, Ly/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/radiotherapy , Membrane Proteins/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Antigens, Ly/analysis , Cell Line, Tumor , Cell Movement/radiation effects , DNA Damage/radiation effects , Female , Isoenzymes/analysis , Isoenzymes/genetics , Mammary Neoplasms, Animal/pathology , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Retinal Dehydrogenase/analysis , Retinal Dehydrogenase/genetics
2.
J Immunol ; 191(2): 755-63, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772032

ABSTRACT

In the current study, exposure of mammary tumor cells derived from mice transgenic for the polyomavirus middle T oncogene to ionizing radiation resulted in the generation of a tumor cell population that preferentially expressed cancer stem cell markers. In addition, these cells were more resistant to subsequent radiation treatments and appeared to acquire an enhanced capacity for dissemination to the lungs of mice. Therefore, we tested an immunotherapy approach to the treatment of local and disseminated mammary tumor cells in a murine model using a recently developed molecular chaperone-based vaccine that specifically targets the radioresistant subpopulation of tumor cells. Heat shock protein 70-peptide complexes (Hsp70.PC-F) were extracted from fusions of dendritic cells and radiation-enriched tumor cells, and the resulting chaperone vaccines were used to treat mice with pre-existing lung metastases. Immunization of mice with the Hsp70.PC-F vaccine resulted in a T cell-mediated immune response, including a significant increase in CD4 and CD8 T cell proliferation and the induction of effector T cells capable of targeting radioresistant tumor cells. Importantly, the growth of primary tumors was inhibited, and the number of tumor cells metastasizing to lung was reduced significantly by combining chaperone vaccine with radiotherapy. These results indicate that Hsp70.PC-F vaccine can induce specific immunity to radioresistant populations of mammary tumor cells and, thus, can complement radiotherapy, leading to synergistic killing.


Subject(s)
Cancer Vaccines/administration & dosage , HSP70 Heat-Shock Proteins/pharmacology , Immunotherapy/methods , Mammary Neoplasms, Experimental/radiotherapy , Mammary Neoplasms, Experimental/therapy , Radiation Tolerance , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cell Fusion , Cell Proliferation , Combined Modality Therapy , Dendritic Cells/metabolism , Female , HSP70 Heat-Shock Proteins/administration & dosage , Immunity, Cellular/immunology , Immunization , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lymphocyte Activation/immunology , Mammary Tumor Virus, Mouse/immunology , Mammary Tumor Virus, Mouse/radiation effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Chaperones/immunology , Radiation, Ionizing , Retroviridae Infections/immunology , Retroviridae Infections/radiotherapy , Retroviridae Infections/therapy , Tumor Virus Infections/immunology , Tumor Virus Infections/radiotherapy , Tumor Virus Infections/therapy , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
3.
Breast Cancer Res ; 14(1): R18, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22277639

ABSTRACT

INTRODUCTION: It is still uncertain whether metastasis is predominantly an early or late event in tumor progression. The detection of early metastases and cells responsible for the dissemination may therefore have significant clinical implications. METHODS: Lung dissemination and/or metastasis were investigated in mice carrying the polyomavirus middle-T oncogene (PyMT) during different stages of mammary tumorigenesis using the colony forming assay. Immunocytochemical or immunohistochemical staining was used to identify subpopulations of cells responsible for lung dissemination and metastasis. Histological examination was used to show primary and metastatic tumors. The tumor-initiating and metastatic capacity of cells expressing stem cell markers was assessed in syngeneic wild-type (WT) mice whose mammary fat pads were injected with these cells. RESULTS: Metastatic mammary epithelial cells were detected in the lungs of mice carrying the PyMT oncogene (MMT mice). These cells were observed early in breast tumorigenesis when the mammary tree appeared by histological inspection to be normal (or at a premalignant stage), suggesting the possession of disseminating and metastatic capacity even before full malignant transformation. Some of the disseminated cells and lung metastases displayed surface stem cell markers. These findings suggest that stem cells from apparently precancerous primary lesions could be a source of metastasis. Indeed, injection of lung tissue cells from MMT mice into syngeneic WT mice resulted in the formation of mammary tumors. These tumors resembled their parent mammary tumors in the MMT donors as well as grafted tumors derived from mammary tumor cells. Furthermore, when we injected lung tissue cells from GFP MMT mice into the fat pads of recipient WT mice, disseminated or metastatic GFP-expressing cells were detected in the lungs, lymph nodes and blood of the recipient WT mice. We finally identified a subpopulation of mammary epithelial/tumor cells expressing CD44 and Sca1 that was largely responsible for dissemination and metastasis in MMT mice. CONCLUSIONS: The tumorigenic and metastatic potential of a subpopulation of mammary epithelial/tumor cells in MMT mice is endowed relatively early in mammary neoplasms and suggests a potential role for cancer stem cell sub-populations in metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma/secondary , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Neoplastic Stem Cells/metabolism , Animals , Antigens, Ly/metabolism , Antigens, Polyomavirus Transforming/genetics , Carcinoma/metabolism , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Green Fluorescent Proteins/biosynthesis , Hyaluronan Receptors/metabolism , Lung Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucin-1/metabolism , Neoplasm Transplantation , Tumor Cells, Cultured
4.
Int J Cancer ; 129(8): 1990-2001, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21154809

ABSTRACT

The majority of patients with stage III/IV ovarian carcinoma that respond initially to standard therapies ultimately undergo relapse due to the survival of small populations of cells with tumor-initiating potential. These ovarian cancer (OVCA)-initiating cells (OCIC) are sometimes called cancer stem cells (CSC) because they express stem cell markers, and can survive conventional therapies such as chemotherapy, which usually target rapidly replicating tumor cells, and give rise to recurrent tumors that are more chemo-resistant and more aggressive. Thus, it would be desirable to develop a therapy that could selectively target OCIC and be used to complement the conventional therapies. In this study, we isolated a subset of OVCA cells with a CD44(+) phenotype in samples from patients with OVCA that possess CSC properties including the formation of spheroids in culture, self-renewal and the ability to be engrafted in immune-compromised mice. We next explored the use of immunotherapy using fusions of dendritic cells and OCIC to specifically target the OCIC subpopulations. Fusion cells (FCs) prepared in this way activated T cells to express elevated levels of IFN-γ with enhanced killing of CD44(+) OVCA cells. We envision a combined approach where conventional therapies such as chemotherapy kill the bulk of tumor cells, whereas OCIC-reactive cytotoxic T lymphocytes target the resistant OCIC fraction. A combined therapy such as this may represent a promising approach for the treatment of OVCA.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Neoplastic Stem Cells/immunology , Ovarian Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Fusion , Female , Humans , Hyaluronan Receptors/metabolism , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Nude
5.
Breast Cancer Res ; 12(5): R71, 2010.
Article in English | MEDLINE | ID: mdl-20846433

ABSTRACT

INTRODUCTION: Ionizing radiation (IR) is a widely used approach to cancer therapy, ranking second only to surgery in rate of utilization. Responses of cancer patients to radiotherapy depend in part on the intrinsic radiosensitivity of the tumor cells. Thus, promoting tumor cell sensitivity to IR could significantly enhance the treatment outcome and quality of life for patients. METHODS: Mammary tumor cells were treated by a 16-base phosphodiester-linked oligonucleotide homologous to the telomere G-rich sequence TTAGGG (T-oligo: GGTTAGGTGTAGGTTT) or a control-oligo (the partial complement, TAACCCTAACCCTAAC) followed by IR. The inhibition of tumor cell growth in vitro was assessed by cell counting and clonogenic cell survival assay. The tumorigenesis of tumor cells after various treatments was measured by tumor growth in mice. The mechanism underlying the radiosensitization by T-oligo was explored by immunouorescent determination of phosphorylated histone H2AX (γH2AX) foci, ß-galactosidase staining, comet and Terminal deoxynucleotidyl transferase dUTP Nick End Labeling (TUNEL) assays. The efficacy of the combined treatment was assessed in a spontaneous murine mammary tumor model. RESULTS: Pretreatment of tumor cells with T-oligo for 24 hours in vitro enhanced both senescence and apoptosis of irradiated tumor cells and reduced clonogenic potential. Radiosensitization by T-oligo was associated with increased formation and/or delayed resolution of γH2AX DNA damage foci and fragmented DNA. T-oligo also caused radiosensitization in two in vivo mammary tumor models. Indeed, combined T-oligo and IR-treatment in vivo led to a substantial reduction in tumor growth. Of further significance, treatment with T-oligo and IR led to synergistic inhibition of the growth of spontaneous mammary carcinomas. Despite these profound antitumor properties, T-oligo and IR caused no detectable side effects under our experimental conditions. CONCLUSIONS: Pretreatment with T-oligo sensitizes mammary tumor cells to radiation in both in vitro and in vivo settings with minimal or no normal tissue side effects.


Subject(s)
Mammary Neoplasms, Animal/radiotherapy , Oligonucleotides/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Combined Modality Therapy , Comet Assay , DNA Damage/drug effects , DNA Damage/radiation effects , Female , Galactosides/analysis , Histones/metabolism , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Phosphorylation/radiation effects , Radiation, Ionizing , Radiation-Sensitizing Agents/therapeutic use , Telomere/genetics
6.
J Immunol ; 184(1): 488-96, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19949080

ABSTRACT

In previous studies, we have shown that heat shock protein 70-peptide complexes (HSP70.PCs) derived from the fusion of dendritic cells (DCs) to tumor cells (HSP70.PC-F) possess superior properties compared with HSP70.PCs from tumor cells. HSP70.PC-F are more effective in stimulation of DC maturation and induction of CTL that are able to provide protection of mice against challenge with tumor cells. To develop an improved formulation of HSP70.PC-based tumor vaccine for patient use, we extracted HSP70.PC-F from DCs fused to patient-derived ovarian cancer cells or established human breast cancer cells and examined their properties as tumor vaccines. HSP70.PC-F induced T cells that expressed higher levels of IFN-gamma and exhibited increased levels of killing of tumor cells, compared with those induced by HSP70.PC derived from tumor cells. Enhanced immunogenicity of HSP70.PC-F was associated with improved composition of the vaccine, including increased content of tumor Ags and their processed intermediates, and the detection of other heat shock proteins (HSPs) such as HSP90 and HSP110. The present study has therefore provided an alternative approach to preparation of HSP-based vaccines using DC/tumor fusion technology and gentle and rapid isolation of HSP peptide complexes.


Subject(s)
Cancer Vaccines/immunology , Cell Line, Tumor/immunology , Dendritic Cells/immunology , HSP72 Heat-Shock Proteins/immunology , Immunotherapy, Active/methods , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Cell Fusion , Cell Proliferation , Cytotoxicity, Immunologic , Female , Humans , Immunoblotting , Lymphocyte Activation/immunology , Microscopy, Confocal , T-Lymphocytes/immunology
7.
J Immunol ; 183(5): 3092-8, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19641135

ABSTRACT

Heat shock protein (HSP) 70 isolated from tumor-dendritic cell (DC) fusions (HSP70.PC-F) induces potent antitumor immunity and prevents growth of such tumors. In the present study, we have examined mechanisms underlying such antitumor activity of the HSP70.PC-F vaccine. The degree of antitumor immunity induced by HSP70.PC-F depended on intact TLR signaling in immunized animals, and mice in which the tlr2 and tlr4 genes were both inactivated did not respond to the vaccine. The reduced responses to HSP70.PC-F vaccine in such tlr knockout mice were restored by immunization of animals with HSP70.PC-F-pulsed wild-type DC, indicating a key role for this cell type in HSP70.PC-F-mediated immunity. Our studies also indicate a role for the scavenger receptor expressed by endothelial cells-1 (SREC-1) in antitumor immunity induced by HSP70.PC-F. These two receptor types appeared functionally interdependent, as indicated by the finding that tlr2 and tlr4 knockout decreases HSP70 binding in double-knockout DC and reduces SREC-1 expression. In addition, TLR-dependent, tumor cell killing was suppressed by SREC-1 knockdown in DC, suggesting a significant role for this receptor in HSP70.PC-F-mediated tumor immunity.


Subject(s)
Cancer Vaccines/immunology , Endothelial Cells/immunology , HSP70 Heat-Shock Proteins/immunology , Scavenger Receptors, Class F/physiology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/metabolism , Cell Line , Cells, Cultured , Cytotoxicity Tests, Immunologic , Endothelial Cells/metabolism , HSP70 Heat-Shock Proteins/administration & dosage , HSP70 Heat-Shock Proteins/metabolism , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/genetics , Protein Binding/immunology , Scavenger Receptors, Class F/biosynthesis , Scavenger Receptors, Class F/genetics , Signal Transduction/genetics , T-Lymphocytes, Cytotoxic/metabolism , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Tumor Cells, Cultured
8.
J Immunol ; 177(9): 5946-55, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17056519

ABSTRACT

We have developed a molecular chaperone-based tumor vaccine that reverses the immune tolerance of cancer cells. Heat shock protein (HSP) 70 extracted from fusions of dendritic (DC) and tumor cells (HSP70.PC-F) possess superior properties such as stimulation of DC maturation and T cell proliferation over its counterpart from tumor cells. More importantly, immunization of mice with HSP70.PC-F resulted in a T cell-mediated immune response including significant increase of CD8 T cells and induction of the effector and memory T cells that was able to break T cell unresponsiveness to a nonmutated tumor Ag and provide protection of mice against challenge with tumor cells. By contrast, the immune response to vaccination with HSP70-PC derived from tumor cells is muted against such nonmutated tumor Ag. HSP70.PC-F complexes differed from those derived from tumor cells in a number of key manners, most notably, enhanced association with immunologic peptides. In addition, the molecular chaperone HSP90 was found to be associated with HSP70.PC-F as indicated by coimmunoprecipitation, suggesting ability to carry an increased repertoire of antigenic peptides by the two chaperones. Significantly, activation of DC by HSP70.PC-F was dependent on the presence of an intact MyD88 gene, suggesting a role for TLR signaling in DC activation and T cell stimulation. These experiments indicate that HSP70-peptide complexes (PC) derived from DC-tumor fusion cells have increased their immunogenicity and therefore constitute an improved formulation of chaperone protein-based tumor vaccine.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , HSP70 Heat-Shock Proteins/immunology , Multiprotein Complexes/immunology , Neoplasms/prevention & control , Animals , Cell Fusion , HSP70 Heat-Shock Proteins/isolation & purification , Immunity, Active/genetics , Immunization , Mice , Mice, Knockout , Mucin-1/analysis , Multiprotein Complexes/chemistry , Multiprotein Complexes/isolation & purification , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Peptide Fragments/analysis , Peptides/immunology , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...