Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
2.
BMC Cancer ; 16: 331, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27220365

ABSTRACT

BACKGROUND: Distant metastasis resulting from vascular dissemination of cancer cells is the primary cause of mortality from breast cancer. We have previously reported that E-selectin expression on the endothelial cell surface mediates shear-resistant adhesion and migration of circulating cancer cells via interaction with CD44. As a result of shedding, soluble E-selectin (sE-selectin) from the activated endothelium is present in the serum. In this study, we aimed to understand the role of sE-selectin in tumor progression and metastasis. METHODS: We investigated the effect of sE-selectin on shear-resistant adhesion and migration of metastatic breast cancer cells and leukocytes in vitro and in vivo. RESULTS: We found that sE-selectin promoted migration and shear-resistant adhesion of CD44(+) (/high) breast cancer cell lines (MDA-MB-231 and MDA-MB-468) to non-activated human microvessel endothelial cells (ES-HMVECs), but not of CD44(-/low) breast cancer cell lines (MCF-7 and T-47D). This endothelial E-selectin independent, sE-selectin-mediated shear-resistant adhesion was also observed in a leukocyte cell line (HL-60) as well as human peripheral blood mononuclear cells (PBMCs). Additionally, the incubation of MDA-MB-231 cells with sE-selectin triggered FAK phosphorylation and shear-resistant adhesion of sE-selectin-treated cells resulted in increased endothelial permeabilization. However, CD44 knockdown in MDA-MB-231 and HL-60 cells resulted in a significant reduction of sE-selectin-mediated shear-resistant adhesion to non-activated HMVECs, suggesting the involvement of CD44/FAK. Moreover, functional blockade of ICAM-1 in non-activated HMVECs resulted in a marked reduction of sE-selectin-mediated shear-resistant adhesion. Finally, the pre-incubation of CD44(+) 4 T1 murine breast cancer cells with sE-selectin augmented infiltration into the lung in E-selectin K/O mice and infusion of human PBMCs pre-incubated with sE-selectin stimulated MDA-MB-231 xenografted breast tumor growth in NSG mice. CONCLUSIONS: Our data suggest that circulating sE-selectin stimulates a broad range of circulating cells via CD44 and mediates pleiotropic effects that promote migration and shear-resistant adhesion in an endothelial E-selectin independent fashion, in turn accelerating tissue infiltration of leukocytes and cancer cells.


Subject(s)
Breast Neoplasms/secondary , E-Selectin/physiology , Endothelium, Vascular/pathology , Leukocytes, Mononuclear/pathology , Neoplastic Cells, Circulating/pathology , Animals , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Adhesion , Cell Movement , Cell Proliferation , Disease Progression , Endothelium, Vascular/metabolism , Female , Humans , Hyaluronan Receptors/metabolism , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplastic Cells, Circulating/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Cancer Res ; 72(24): 6447-56, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23090116

ABSTRACT

Generating antitumor responses through the inhibition of tumor-derived immune suppression represents a promising strategy in the development of cancer immunotherapeutics. Here, we present a strategy incorporating delivery of the bacterium Salmonella typhimurium (ST), naturally tropic for the hypoxic tumor environment, transformed with a small hairpin RNA (shRNA) plasmid against the immunosuppressive molecule indoleamine 2,3-dioxygenase 1 (shIDO). When systemically delivered into mice, shIDO silences host IDO expression and leads to massive intratumoral cell death that is associated with significant tumor infiltration by polymorphonuclear neutrophils (PMN). shIDO-ST treatment causes tumor cell death independently of host IDO and adaptive immunity, which may have important implications for use in immunosuppressed patients with cancer. Furthermore, shIDO-ST treatment increases reactive oxygen species (ROS) produced by infiltrating PMNs and, conversely, PMN immunodepletion abrogates tumor control. Silencing of host IDO significantly enhances S. typhimurium colonization, suggesting that IDO expression within the tumor controls the immune response to S. typhimurium. In summary, we present a novel approach to cancer treatment that involves the specific silencing of tumor-derived IDO that allows for the recruitment of ROS-producing PMNs, which may act primarily to clear S. typhimurium infection, but in the process also induces apoptosis of surrounding tumor tissue resulting in a vigorous antitumor effect.


Subject(s)
Gene Transfer Techniques , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Neoplasms/therapy , RNA, Small Interfering/administration & dosage , Salmonella typhimurium/growth & development , Salmonella typhimurium/genetics , Transformation, Bacterial , Administration, Intravenous , Animals , Cell Line, Tumor , Cell Proliferation , Colony Count, Microbial , Gene Silencing , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/physiology , HEK293 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/microbiology , Neoplasms/pathology , Organisms, Genetically Modified , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Salmonella typhimurium/immunology , Salmonella typhimurium/physiology , Transformation, Bacterial/physiology
4.
Oncoimmunology ; 1(2): 240-241, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22720256

ABSTRACT

Improving on the limited success of cancer immunotherapy requires new approaches to inhibit immunosuppressive pathways initiated by tumor cells to "escape" protective immunity. One unique approach utilizes Salmonella for systemic delivery of inhibitory RNA, targeting the immunosuppressive molecule Stat3, and a Survivin vaccine to suppress growth of aggressive murine tumors.

5.
Cancer Res ; 71(12): 4183-91, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21527558

ABSTRACT

Cancer vaccine therapies have only achieved limited success when focusing on effector immunity with the goal of eliciting robust tumor-specific T-cell responses. More recently, there is an emerging understanding that effective immunity can only be achieved by coordinate disruption of tumor-derived immunosuppression. Toward that goal, we have developed a potent Salmonella-based vaccine expressing codon-optimized survivin (CO-SVN), referred to as 3342Max. When used alone as a therapeutic vaccine, 3342Max can attenuate growth of aggressive murine melanomas overexpressing SVN. However, under more immunosuppressive conditions, such as those associated with larger tumor volumes, we found that the vaccine was ineffective. Vaccine efficacy could be rescued if tumor-bearing mice were treated initially with Salmonella encoding a short hairpin RNA (shRNA) targeting the tolerogenic molecule STAT3 (YS1646-shSTAT3). In vaccinated mice, silencing STAT3 increased the proliferation and granzyme B levels of intratumoral CD4(+) and CD8(+) T cells. The combined strategy also increased apoptosis in tumors of treated mice, enhancing tumor-specific killing of tumor targets. Interestingly, mice treated with YS1646-shSTAT3 or 3342Max alone were similarly unsuccessful in rejecting established tumors, whereas the combined regimen was highly potent. Our findings establish that a combined strategy of silencing immunosuppressive molecules followed by vaccination can act synergistically to attenuate tumor growth, and they offer a novel translational direction to improve tumor immunotherapy.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy, Active , Inhibitor of Apoptosis Proteins/genetics , Melanoma, Experimental/therapy , RNA, Small Interfering/genetics , Repressor Proteins/genetics , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis , Codon , Cytotoxicity, Immunologic , Genetic Vectors , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , STAT3 Transcription Factor/genetics , Survivin , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...