Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Clin Exp Med ; 15(2): 127-35, 2015 May.
Article in English | MEDLINE | ID: mdl-24619299

ABSTRACT

The pathogenic mechanism of malignant melanoma involves the dynamic interplay of transformed cell and normal host cell, but cancer treatments always target each partition separately. In the tumor microenvironment, milk fat globule epidermal growth factor-8 (MFG-E8) is a secreted glycoprotein highly expressed in the vertical growth phase of melanoma, leading to tumor progression through coordinated αvß3 and αvß5 integrin signaling in tumor cells and host cells. Doxorubicin (Dox) is one of the most widely used antitumor drugs against a lot of solid tumors, including melanoma. In this work, Dox was used to combine with down-regulation of MFG-E8 by RNA interference (RNAi) in order to determine the synergistic effect of the antitumor activity in vivo. And the possible mechanisms were investigated. Results showed that combination group (MFG-E8 RNAi plus Dox) could inhibit the growth of melanoma more effectively than monotherapy or control groups. We found that the combination treatment induced more tumor cell apoptosis and inhibited more neovascularization than other groups. Moreover, this combination treatment attenuated CD4(+) CD25(+) Foxp3(+) Treg cells in tumor-infiltrating lymphocytes compared with other groups. Our findings suggested that MFG-E8 down-regulation enhanced the antitumor function of chemotherapy through coordinated cell apoptosis and immune-mediated mechanisms, which might be a feasible way for cancer therapy.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Combined Modality Therapy , Down-Regulation , Doxorubicin/administration & dosage , Melanoma/therapy , Milk Proteins/antagonists & inhibitors , Animals , Antigens, Surface/genetics , Apoptosis , Disease Models, Animal , Female , Gene Knockdown Techniques , Immunity, Cellular , Melanoma/pathology , Mice, Inbred C57BL , Milk Proteins/genetics , RNA Interference , Treatment Outcome
2.
J Cancer Res Clin Oncol ; 140(11): 1815-23, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24927808

ABSTRACT

PURPOSE: Lipopolysaccharide (LPS) is a major component of the outer surface membrane of Gram-negative bacteria which has been proved an effective immune enhancer. Here, we investigated the anti-tumor effect of irradiated tumor cells that stimulated by LPS in mouse xenografts models. METHODS: Tumor cells were irradiated after stimulation with 1 µg/mL LPS for 48 h. The C57BL/6 mice were immunized subcutaneously with irradiated tumor cells. The anti-tumor effect of lymphocytes of immunized mice was investigated. The cytotoxicity of spleen lymphocytes from immunized mice was determined by a standard (51)Cr-release assay. The roles of immune cell subsets in anti-tumor activity were assessed by injected intraperitoneally with monoclonal antibodies. RESULTS: We observed that the vaccine of irradiated tumor cell with LPS-stimulated elicited a stronger protective anti-tumor immunity than other controls. Adoptive transfer of lymphocytes of immunized mice showed that the cellular immune response was involved in the anti-tumor effect. And this effect was achieved by activation of antigen-specific CD8(+) T cell response and reduction of myeloid-derived suppressor cells (MDSCs, Gr1(+) CD11b (+) ), which were confirmed by depletion of immune cell subsets and flow cytometry analysis. CONCLUSIONS: In summary, our study showed that stimulation of LPS was able to enhance anti-tumor immunity of vaccination with tumor cells after irradiation treatment, which might be a new strategy for cancer therapy.


Subject(s)
Cancer Vaccines/immunology , Lipopolysaccharides/pharmacology , Melanoma, Experimental/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/radiation effects , Cell Line, Tumor , Female , Hybridomas , Immunotherapy, Adoptive , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Spleen/immunology , Tumor Burden/immunology , Vaccination , Xenograft Model Antitumor Assays
3.
J Cancer Res Clin Oncol ; 139(5): 797-807, 2013 May.
Article in English | MEDLINE | ID: mdl-23385883

ABSTRACT

PURPOSE: The overexpression of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) has been proved to inhibit tumor growth and metastasis through degradation of prostaglandin E2 (PGE2), which is often overexpressed in various cancers and accelerates tumor progression. Cyclooxygenase-2 (COX-2), a synthase of PGE2, actively produces much PGE2 to counteract the 15-PGDH-induced antitumor efficacy. Here, we investigated the combinational effect by using pcDNA3.1(+) encoding mouse 15-PGDH gene therapy and celecoxib, a COX-2 inhibitor, in mouse breast cancers. METHODS: Mice bearing 4T1 were treated with short-term administration of the COX-2 inhibitor celecoxib (40 mg/kg/day) plus liposome-encapsulated mouse 15-PGDH in order to determine their synergistic antitumor activity in vivo. And the possible mechanisms were investigated. RESULTS: We observed that the combination treatment of 15-PGDH and celecoxib significantly inhibited tumor growth and lung metastases than monotherapy or controls. Moreover, the effect of combination treatment was associated with significant reduction of PGE2 in serum, which resulted from increased 15-PDGH and decreased COX-2 in tumor tissues. The tumor tissues in combination treatment presented more apoptotic cells and less microvessel density. Notably, the number of myeloid-derived suppressor cells in the spleen was also significantly decreased in the combination treatment than others. CONCLUSIONS: Our findings suggested that celecoxib increased the antitumor activity of 15-PGDH by synergistically blocking PGE2 pathway, which might be a new feasible way for cancer therapy.


Subject(s)
Cyclooxygenase 2 Inhibitors/administration & dosage , Genetic Therapy , Hydroxyprostaglandin Dehydrogenases/genetics , Mammary Neoplasms, Experimental/therapy , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Animals , Apoptosis/drug effects , Apoptosis/genetics , Celecoxib , Cell Line, Tumor , Combined Modality Therapy , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/adverse effects , Dinoprostone/metabolism , Disease Models, Animal , Female , Genetic Therapy/adverse effects , Humans , Hydroxyprostaglandin Dehydrogenases/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Pyrazoles/adverse effects , Spleen/immunology , Spleen/pathology , Sulfonamides/adverse effects , Tumor Burden/drug effects , Tumor Burden/genetics
4.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(5): 594-8, 2011 Sep.
Article in Chinese | MEDLINE | ID: mdl-22007479

ABSTRACT

OBJECTIVE: To study molecular mechanisms underlying the extravasation of mice melanoma cells during lung metastasis. METHODS: B16-RED melanoma cell line was established which stably express the red fluorescent protein. B16-RED cells were compared with B16 cells in ability of proliferation and lung metastasis. A mouse lung metastasis model was established with B16-RED melanoma cells. FITC-dextran was injected i.v. and CD31 indirect immunoflourescence (IIF) staining was made to identify the location of the tumor cells and the time of tumor cell extravasation. Finally, at 48 hours post cell injection, the lung and a normal lung were removed and used for 32K mice microarray analysis. RESULTS: B16-RED was consistent with B16 in cell shape and ability of proliferation and lung metastasis. 52.7% of B16-RED melanoma cells completed the extravasation within 48 hours in mouse lung metastasis model. Many important signal pathways were involved during lung metastasis, including leukocyte transendothelial migration, MAPK signaling pathway, neuroactive ligand-receptor interaction, focal adhesion, cytokine-cytokine receptor interaction, regulation of actin cytoskeleton, axon guidance, calcium signaling pathway, tight junction, etc. CONCLUSION: The extravasation during metastasis is a complex and multiple-steps process, in which many important signal pathways in host tissues were involved.


Subject(s)
Cell Movement , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Neoplastic Cells, Circulating/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/physiopathology
5.
Cancer Biol Ther ; 12(6): 523-30, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21743297

ABSTRACT

The use of survivinT34A mutant targeted disruption of survivin, the strongest inhibitor of apoptosis protein overexpressed in tumors, has proved a promising strategy for advanced cancers. However, hyperthermia, as a cytotoxic enhancer, regularly activates the expression of survivin to counteract the heat-induced antitumor activity. Here, we investigated the combinational antitumor effect by using liposome-encapsulated mouse survivinT34A and hyperthermia in mouse models. We observed that the combination treatment of surivinT34A and hyperthermia significantly increased the growth inhibition and apoptosis of tumor cells in vitro compared with single treatment or other controls, which was similar to the effect of survivin silencing in combination with hyperthermia. Moreover, the inhibition of tumor growth in vivo was also remarkably enhanced by combination of surivinT34A and hyperthermia when compared with other treatments. Naturally, the tumor tissues in combination treatment presented the larger necrosis-like areas, more apoptotic cells and less microvessel density. Our findings suggest that the antitumor efficacy of survivin disruption can be enhanced by hyperthermia, which might be a new feasible approach for cancer therapy.


Subject(s)
Genetic Therapy , Hyperthermia, Induced , Inhibitor of Apoptosis Proteins/genetics , Mutant Proteins/genetics , Neoplasms/therapy , Repressor Proteins/genetics , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Combined Modality Therapy , Female , Mice , Mice, Inbred BALB C , Mutation, Missense , Neoplasm Transplantation , Neoplasms/genetics , Survivin
6.
PLoS One ; 6(7): e21768, 2011.
Article in English | MEDLINE | ID: mdl-21765912

ABSTRACT

Tumor neovascularization is a highly complex process including multiple steps. Understanding this process, especially the initial stage, has been limited by the difficulties of real-time visualizing the neovascularization embedded in tumor tissues in living animal models. In the present study, we have established a xenograft model in zebrafish by implanting mammalian tumor cells into the perivitelline space of 48 hours old Tg(Flk1:EGFP) transgenic zebrafish embryos. With this model, we dynamically visualized the process of tumor neovascularization, with unprecedented high-resolution, including new sprouts from the host vessels and the origination from VEGFR2(+) individual endothelial cells. Moreover, we quantified their contributions during the formation of vascular network in tumor. Real-time observations revealed that angiogenic sprouts in tumors preferred to connect each other to form endothelial loops, and more and more endothelial loops accumulated into the irregular and chaotic vascular network. The over-expression of VEGF165 in tumor cells significantly affected the vascularization in xenografts, not only the number and size of neo-vessels but the abnormalities of tumor vascular architecture. The specific inhibitor of VEGFR2, SU5416, significantly inhibited the vascularization and the growth of melanoma xenografts, but had little affects to normal vessels in zebrafish. Thus, this zebrafish/tumor xenograft model not only provides a unique window to investigate the earliest events of tumoral neoangiogenesis, but is sensitive to be used as an experimental platform to rapidly and visually evaluate functions of angiogenic-related genes. Finally, it also offers an efficient and cost-effective means for the rapid evaluation of anti-angiogenic chemicals.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Xenograft Model Antitumor Assays , Zebrafish/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Indoles/pharmacology , Mice , Microvessels/drug effects , Microvessels/pathology , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Pyrroles/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 41(4): 563-6, 2010 Jul.
Article in Chinese | MEDLINE | ID: mdl-20848769

ABSTRACT

OBJECTIVE: To develop a novel anti-angiogenesis strategy based on a DNA vaccine coding both human and mouse soluble VEGFR2. METHODS: The gene fragments coding human and mouse sVEGFR2 were amplified with PCR and cloned into pVITRO2 to generate pVITRO2-hm-sVEGFR2 recombinant. The in vitro VEGF blocking effect of the pVITRO2-hm-sVEGFR2 expression products on HUVEC cells were evaluated. The anti-tumor effect of pVITRO2-hm-sVEGFR2 was studied in mouse B16 model. The microvessels were stained by using CD31 antibody. RESULTS: The co-expressing vector pVITRO2-hm-sVEGFR2 was constructed successfully, confirmed by the restriction endonuclease digestion and sequencing. The expressing products of pVITRO2-hm-sVEGFR2 could obviously block the function of VEGF on promoting the proliferation of HUVEC in vitro. The tumor growth in mice was also significantly inhibited by pVITRO2-hm-sVEGFR2 expression. CD31 staining demonstrated that the microvessel density obviously decreased in tumor tissues treated with pVITRO2-hm-sVEGFR2. Both anti-tumor and anti-angiogenesis effects of pVITRO2-hm-sVEGFR2 were stronger than that of plasmids which coding only human or mouse sVEGFR2. CONCLUSION: pVITRO2-hm-sVEGFR2 could be a novel DNA vaccine for the anti-tumor therapy by inhibiting angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cancer Vaccines/immunology , Neovascularization, Pathologic/prevention & control , Vaccines, DNA/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Angiogenesis Inhibitors/metabolism , Animals , Cancer Vaccines/genetics , Cell Line , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Vectors/genetics , Humans , Melanoma, Experimental/therapy , Mice , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vascular Endothelial Growth Factor Receptor-2/genetics
8.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 41(4): 567-70, 594, 2010 Jul.
Article in Chinese | MEDLINE | ID: mdl-20848770

ABSTRACT

OBJECTIVE: To determine the enhancement effects of IL-15 to the tumor whole cell vaccine in tumor immunotherapy. METHODS: CT26 colon carcinoma model was established with BALB/c mice. Thirty two mice with CT26 colon carcinoma were divided randomly into four groups, which were subcutaneously injected at several spots with PBS (200 microL), CT26 whole cell vaccine (2 x 10(6) cells in 200 microL PBS), mIL-15 (20 microg, encapsulated with liposome in 200 microL 5% glucose solution) and CT26 whole cell vaccine (2 x 10(6) cells in 100 microL PBS) plus mIL-15 (20 microg, encapsulated with liposome in 100 microL 5% glucose solution) respectively every three days for six doses, the plasmid was injected beside the vaccine injecting spots. The size of tumors was measured every four days. All mice were sacrificed 30 days after tumor implantation. The pathologic observation and apoptotic analysis of tumors were preceded. RESULTS: CT26 whole cell vaccine combined with mIL-15 inhibited tumor growth by 45% compared with that of control group, the differences between CT26 + mIL-15 group and the other three groups were significant (P < 0. 05). The HE staining showed that the necrosis areas in tumors of the CT26 + mIL-15 group were larger than those of other three groups. The apoptotic index of tumors from the CT26 + mIL-15 group was (46.7 +/- 7.2)%, higher than that of the other three groups obviously (P < 0.05). CONCLUSION: IL-15 could enhance the therapeutic effects of CT26 whole cell vaccine in tumor immunotherapy.


Subject(s)
Cancer Vaccines/therapeutic use , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Interleukin-15/therapeutic use , Animals , Cancer Vaccines/immunology , Colonic Neoplasms/pathology , Drug Synergism , Immunotherapy/methods , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Random Allocation
9.
J Biosci ; 35(2): 209-16, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20689177

ABSTRACT

Anti-apoptosis plays an important role in tumour formation and development. Survivin is a member of the inhibitor of apoptosis (IAP) family, which is a target for anti-cancer drug exploitation was replaced as development. We investigated the role of the homo dominant-negative mutant Survivin-T34A in suppressing human lung adenocarcinomas (A549). The anti-tumour activity of HSurvivinT34A plasmid was evaluated in the A549 cell line and nude mice bearing A549 subcutaneous tumours. Low-dose systemic administration was continuously used. The HSurvivinT34A plasmid (5 meu g/one) complexed with a cationic liposome (DOTAP/Chol) significantly inhibited tumour growth in our model. We observed microvessel density degradation by CD31 immunohistochemistry and apoptotic cell increase by TUNEL assay, PI staining and flow cytometric analysis in the treated group. The present findings suggest that the HSurvivinT34A plasmid complexed with a cationic liposome may provide an effective approach to inhibit the growth of human lung adenocarcinomas in vitro and in vivo.


Subject(s)
Adenocarcinoma/therapy , Lung Neoplasms/therapy , Microtubule-Associated Proteins/genetics , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Female , Gene Transfer Techniques , Genetic Therapy , Humans , Inhibitor of Apoptosis Proteins , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Plasmids , Survivin , Xenograft Model Antitumor Assays
10.
Cancer Biol Ther ; 10(3): 276-81, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20543584

ABSTRACT

Basic fibroblast growth factor (bFGF) is a mitogen for endothelial cells, which participates in tumor angiogenesis. Active immunity against bFGF could be a promising approach for the biotherapy of cancer. Because bFGF is abundant in normal and malignant tissues, it is presumably difficult for normal bFGF to induce immunity due to self-tolerance. In addition, previous studies have shown that a complex consisting of a cationic liposome and a non-coding plasmid DNA can be used to stimulate innate immunity. This stimulation initiates a potent cytokine response, which can inhibit tumor growth. To investigate the effects of immunity against bFGF on murine colon carcinomas, we employed an N-, C-terminally truncated basic fibroblast growth factor (tbFGF, of human origin) as an antigen and a liposome-DNA complex as an adjuvant. After six immunizations, a robust bFGF-specific immune response was elicited. Subsequently, inhibition of tumor growth and a significant reduction in tumor vasculature were observed. The antitumor effect was confirmed by adoptive therapy of activated spleen cells from the immunized mice. In vitro, a CTL assay revealed that bFGF-specific cytotoxic T lymphocytes (CTL) resulted in the lysis of mouse microvascular endothelial cells (MS1) rather than that of the CT26 colorectal cancer cells. These results suggest that anti-angiogenesis treatment induced by a bFGF-specific CTLs against microvascular endothelial cells may be a useful method for cancer therapy.


Subject(s)
Cancer Vaccines/administration & dosage , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Fibroblast Growth Factor 2/immunology , Amino Acid Sequence , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , DNA/administration & dosage , Female , Fibroblast Growth Factor 2/administration & dosage , Fibroblast Growth Factor 2/genetics , Genetic Vectors/genetics , Humans , Immunity, Innate/immunology , Liposomes/administration & dosage , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Plasmids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...