Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
2.
Sci Rep ; 6: 32351, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27571893

ABSTRACT

Genetic modification of whole-cell cancer vaccines to augment their efficacies has a history of over two and a half decades. Various genes and gene combinations, targeting different aspects of immune responses have been tested in pursuit of potent adjuvant effects. Here we show that co-expression of two cytokine members of the common cytokine receptor γ-chain family, IL-21 and IL-7, in whole-cell cancer vaccines boosts antitumor immunity in a CD4(+) and CD8(+) T cell-dependent fashion. It also generates effective immune memory. The vaccine-elicited short-term effects positively correlated with enhanced infiltration of CD4(+) and CD8(+) effector T cells, and the long-term effects positively correlated with enhanced infiltration of effector memory T cells, especially CD8(+) effector memory T cells. Preliminary data suggested that the vaccine exhibited good safety profile in murine models. Taken together, the combination of IL-21 and IL-7 possesses potent adjuvant efficacy in whole-cell vaccines. This finding warrants future development of IL-21 and IL-7 co-expressing whole-cell cancer vaccines and their relevant combinatorial regimens.


Subject(s)
Cancer Vaccines/immunology , Immunity, Cellular/genetics , Interleukin-7/immunology , Interleukins/immunology , Neoplasms/prevention & control , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Gene Expression Regulation, Neoplastic/immunology , Humans , Interleukin-7/genetics , Interleukins/genetics , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control
3.
Sci Rep ; 6: 23764, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-27026065

ABSTRACT

Overexpression of folate receptor alpha (FRα) and high telomerase activity are considered to be the characteristics of ovarian cancers. In this study, we developed FRα-targeted lipoplexes loaded with an hTERT promoter-regulated plasmid that encodes a matrix protein (MP) of the vesicular stomatitis virus, F-LP/pMP(2.5), for application in ovarian cancer treatment. We first characterized the pharmaceutical properties of F-LP/pMP(2.5). The efficient expression of the MP-driven hTERT promoter in SKOV-3 cells was determined after an in-vitro transfection assay, which was significantly increased compared with a non-modified LP/pMP(2.5) group. F-LP/pMP(2.5) treatment significantly inhibited the growth of tumors and extended the survival of mice in a SKOV-3 tumor model compared with other groups. Such an anti-tumor effect was due to the increased expression of MP in tumor tissue, which led to the induction of tumor cell apoptosis, inhibition of tumor cell proliferation and suppression of tumor angiogenesis. Furthermore, a preliminary safety evaluation demonstrated a good safety profile of F-LP/pMP(2.5) as a gene therapy agent. Therefore, FRα-targeted lipoplexes with therapeutic gene expression regulated by an hTERT promoter might be a promising gene therapy agent and a potential translational candidate for the clinical treatment of ovarian cancer.


Subject(s)
Ovarian Neoplasms/therapy , Animals , Cell Line, Tumor , Female , Genetic Therapy , Humans , Liposomes , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Plasmids/genetics , Transfection , Vesiculovirus/genetics , Viral Matrix Proteins/genetics
4.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 46(4): 542-7, 2015 Jul.
Article in Chinese | MEDLINE | ID: mdl-26480655

ABSTRACT

OBJECTIVE: To determine the role of infiltrated T cells in the accumulation of myeloid-derived suppressor cells (MDSCs) in tumor microenvironment. METHODS: T-cell-deficient nude mice models were established using BALB/c mice. Growth of tumors was compared between those with and without adoptive transfer of T cells. Pathological changes of the tumors were examined with HE histological analysis. The levels of MDSCs were detected with flow cytometry (FACS). RESULTS: Tumor growth was promoted in T-cell-deficient nude mice, which was accompanied with lower levels of MDSCs compared with BALB/c mice (P < 0.05). T cell transfer increased the level of MDSCs significantly (P < 0.05). T cells depletion decreased the level of MDSCs (P < 0.05). CONCLUSION: Infiltrated T cells induce the accumulation of MDSCs in tumor microenvironment, and influence tumor growth.


Subject(s)
Myeloid Cells/cytology , Neoplasms/pathology , T-Lymphocytes/cytology , Tumor Microenvironment , Animals , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Nude
5.
J Breast Cancer ; 17(1): 25-32, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24744794

ABSTRACT

PURPOSE: The universal organic solvent dimethyl sulfoxide (DMSO) can be used as a differentiation inducer of many cancer cells and has been widely used as a solvent in laboratories. However, its effects on breast cancer cells are not well understood. The aim of this study is to investigate the effect and associated mechanisms of DMSO on mouse breast cancer. METHODS: We applied DMSO to observe the effect on tumors in a mouse breast cancer model. Tumor-associated macrophages (TAMs) were tested by flow cytometry. Ex vivo tumor microenvironment was imitated by 4T1 cultured cell conditioned medium. Enzyme-linked immunosorbent assays were performed to detect interleukin (IL)-10 and IL-12 expression in medium. To investigate the cytotoxicity of DMSO on TAMs, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays were performed. RESULTS: We found that DMSO produced tumor retardation when injected into mouse peritoneal cavities in a certain concentration range (0.5-1.0 mg/g). Furthermore, as detected by flow cytometry, TAM subtypes were found to be transformed. We further imitated a tumor microenvironment in vitro by using 4T1 cultured cell conditioned medium. Similarly, by using low concentration DMSO (1.0%-2.0% v/v), TAMs were induced to polarize to the classically activated macrophage (M1-type) and inhibited from polarizing into the alternatively activated macrophage (M2-type) in the conditioned medium. IL-10 expression in tumors was reduced, while IL-12 was increased compared with the control. Furthermore, we reported that 2.0% (v/v) DMSO could lead to cytotoxicity in peritoneal macrophages after 48 hours in MTT assays. CONCLUSION: Our findings suggest that DMSO could exert antitumor effects in 4T1 cancer-bearing mice by reversing TAM orientation and polarization from M2- to M1-type TAMs. These data may provide novel insight into studying breast cancer immunotherapy.

6.
FEBS Lett ; 588(10): 1921-9, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24713430

ABSTRACT

Here, we investigated the molecular mechanism underlying the changes in the distribution of nucleolin. Our study identified PI3K/Akt signaling as an essential pathway regulating the distribution of nucleolin. Furthermore, nucleolin can interact with phospho-PI3K-p55, and changes in the distribution of nucleolin were related to its phosphorylation. Subsequently, we analyzed the correlation of VEGF and nucleolin, and found that distribution of nucleolin related to metastatic potential. Finally, blocking cell surface nucleolin influences the process of epithelial-mesenchymal transitions. This indicates that nucleolin may be a novel cancer therapy target and a predictive marker for tumor migration in colorectal carcinoma.


Subject(s)
Colorectal Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Blotting, Western , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , HCT116 Cells , Humans , Male , Microscopy, Fluorescence , Middle Aged , Neoplasm Metastasis , Phosphorylation , Protein Binding , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Nucleolin
7.
Bioorg Med Chem Lett ; 24(5): 1426-31, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24491459

ABSTRACT

A series of novel benzamide derivatives were prepared and evaluated using cell-based measurements. Among these compounds, 10f significantly inhibited Hedgehog signaling and showed equivalent or more potency than GDC-0449 in different tests. Furthermore, compound 10f potently inhibited the proliferation of Daoy, a medulloblastoma cell line that is reported to be resistant to GDC-0449, which indicated a promising prospect in the treatment of Hedgehog signaling pathway related cancer in clinical trial.


Subject(s)
Benzamides/chemical synthesis , Benzamides/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Sulfones/chemical synthesis , Sulfones/pharmacology , Animals , Benzamides/chemistry , Cell Line, Tumor , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor , Structure-Activity Relationship , Sulfones/chemistry , Zebrafish
8.
Clin Exp Med ; 14(2): 203-13, 2014 May.
Article in English | MEDLINE | ID: mdl-23263406

ABSTRACT

A de novo VEGFR2-inhibited compound SKLB1002 which is independently developed in our laboratory has been described for antiangiogenesis and displays a potent antitumor activity in vivo and in vitro. In the present investigation, we aim to prove that combination therapy of SKLB1002 with hyperthermia plays a synergy as an antitumor agent in solid tumor. In this study, we analyzed their synergetic inhibitory action on human umbilical vein endothelial cells (HUVEC), murine mammary cancer 4T1, murine colon carcinoma CT26 in vitro. Multiply-table tournament was performed to detect cell proliferation in vitro. 4T1 implantation and CT26 implantation in BALB/c mice were used to examine the activity of combination therapy of SKLB1002 with hyperthermia in vivo. Vascular density was determined by CD31 immunohistochemistry. TUNEL was used to measure apoptosis in tumor tissue. Metastasis assay was investigated via measurement of pulmonary metastasis nodules under the microscope. Potential toxicity of combination therapy was observed by histologic analysis of main organs stained with H&E. In vitro, the combination therapy significantly inhibited cell proliferation of HUVEC, 4T1 and CT26. In vivo, 4T1 and CT26 model experiments showed that combination therapy remarkably inhibited tumor growth and prolonged life span. When compared with controls, combination therapy reached 61 % inhibition index of tumor growth against CT26 and 51 % against 4T1. Moreover, it reduced angiogenesis and increased tumor apoptosis and necrosis. It was further found that combination therapy could efficiently prevent tumor from metastasizing to lung. Importantly, it had no toxicity to main organs including heart, liver, spleen, lung and kidney. Combination treatment has been proved to be a novel and strong strategy in clinical antitumor therapy. Our findings suggest that the combination therapy of SKLB1002 with hyperthermia has a synergistic antiangiogenesis, anticancer and promotion of apoptosis efficacy compared with controls. These findings could pave a new way in clinical tumor therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Hyperthermia, Induced/methods , Quinazolines/therapeutic use , Thiadiazoles/therapeutic use , Angiogenesis Inhibitors/adverse effects , Animals , Antineoplastic Agents/adverse effects , Apoptosis , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Colonic Neoplasms/pathology , Combined Modality Therapy , Disease Models, Animal , Female , Histocytochemistry , Hyperthermia, Induced/adverse effects , Immunohistochemistry , Mice, Inbred BALB C , Necrosis , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Quinazolines/adverse effects , Thiadiazoles/adverse effects , Treatment Outcome
9.
Oncol Res ; 20(10): 473-82, 2013.
Article in English | MEDLINE | ID: mdl-24308158

ABSTRACT

Heme oxygenase-1 [HO-1, also called heat shot protein 32 (HSP32)] can specifically metabolize heme to carbon monoxide, biliverdin, and ferrous iron and plays an important role in the processes of anti-inflammation, tissue protection, and antioxidative stress reaction. It has been reported that HO-1 can promote tumorigenesis and metastasis of many tumors. However, the detailed mechanisms of how HO-1 affects tumor progress are not clear. Here, we used ZnPPIX (a specific inhibitor of HO-1) to evaluate its potential effects on mouse breast cancer and tumor-associated macrophages (TAMs). We found out that mouse 4T1 breast cancer growth can be effectively suppressed through inhibition of HO-1 in vitro and in vivo. Moreover, in the 4T1 mouse model, when HO-1 was suppressed in TAMs, alternatively activated macrophages (M2 type) switched to classically activated macrophages (M1 type). In conclusion, 4T1 breast cancer growth was modulated by HO-1 expression. Furthermore, inhibition of HO-1 may induce tumor-associated immune response by activating TAMs' alternative proliferation. These data suggest that HO-1 may be an important target of breast cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Heme Oxygenase-1/antagonists & inhibitors , Macrophages/drug effects , Mammary Neoplasms, Experimental/drug therapy , Membrane Proteins/antagonists & inhibitors , Protoporphyrins/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Heme Oxygenase-1/metabolism , Macrophage Activation/drug effects , Macrophages/enzymology , Macrophages/pathology , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Necrosis , Time Factors , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...