Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
Mol Oncol ; 14(3): 657-668, 2020 03.
Article En | MEDLINE | ID: mdl-31899582

Oncolytic viruses armed with therapeutic transgenes of interest show great potential in cancer immunotherapy. Here, a novel oncolytic adenovirus carrying a signal regulatory protein-α (SIRPα)-IgG1 Fc fusion gene (termed SG635-SF) was constructed, which could block the CD47 'don't eat me' signal of cancer cells. A strong promoter sequence (CCAU) was chosen to control the expression of the SF fusion protein, and a 5/35 chimeric fiber was utilized to enhance the efficiency of infection. As a result, SG635-SF was found to specifically proliferate in hTERT-positive cancer cells and largely increased the abundance of the SF gene. The SF fusion protein was effectively detected, and CD47 was successfully blocked in SK-OV3 and HO8910 ovarian cancer cells expressing high levels of CD47. Although the ability to induce cell cycle arrest and cell death was comparable to that of the control empty SG635 oncolytic adenovirus in vitro, the antitumor effect of SG635-SF was significantly superior to that of SG635 in vivo. Furthermore, CD47 was largely blocked and macrophage infiltration distinctly increased in xenograft tissues of SK-OV3 cells but not in those of CD47-negative HepG2 cells, indicating that the enhanced antitumor effect of SG635-SF was CD47-dependent. Collectively, these findings highlight a potent antitumor effect of SG635-SF in the treatment of CD47-positive cancers.


Antigens, Differentiation/metabolism , CD47 Antigen/immunology , Immunoglobulin G/metabolism , Immunotherapy/methods , Macrophages/immunology , Ovarian Neoplasms/immunology , Receptors, Immunologic/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antigens, Differentiation/genetics , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cell Cycle Checkpoints/immunology , Cell Death/immunology , Cell Line, Tumor , Cytotoxicity Tests, Immunologic , Female , Humans , Immunoglobulin G/genetics , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Receptors, Immunologic/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Telomerase/metabolism , Xenograft Model Antitumor Assays
2.
Cell Death Dis ; 9(2): 177, 2018 02 07.
Article En | MEDLINE | ID: mdl-29415996

Effective control of non-small-cell lung cancer (NSCLC) remains clinically challenging, especially during advanced stages of the disease. This study developed an adoptive T-cell treatment through expression of a chimeric antigen receptor (CAR) to target human epidermal growth factor receptor (EGFR) in NSCLC. We optimized the non-viral piggyBac transposon system to engineer human T cells for the expression of EGFR-CAR, consisting of EGFR scFv, transmembrane domain, and intracellular 4-1BB-CD3ζ signaling domains. The modified CAR T cells exhibited expansion capability and anticancer efficacy in a time- and antigen-dependent manner in vitro as well as regression of EGFR-positive human lung cancer xenografts in vivo. EGFR-CAR T therapy is a promising strategy to improve the efficacy and potency of the adoptive immunotherapy in NSCLC. Moreover, EGFR-CAR T therapy could become a clinical application for NSCLC patients in the future.


Carcinoma, Non-Small-Cell Lung/therapy , ErbB Receptors/immunology , Immunotherapy, Adoptive/methods , Lung Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Animals , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Random Allocation , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
3.
Tumour Biol ; 39(4): 1010428317695949, 2017 Apr.
Article En | MEDLINE | ID: mdl-28381173

Chimeric antigen receptor modified T cell-based immunotherapy is revolutionizing the field of cancer treatment. However, its potential in treating bile duct carcinoma has not been fully explored. Herein, we developed the second-generation mesothelin-targeting chimeric antigen receptor-modified T cells with the 4-1BB co-stimulatory module by the piggyBac transposon system. Mesothelin-targeting chimeric antigen receptor was expressed by 66.0% of mesothelin-targeting chimeric antigen receptor-modified T cells post electrophoretic transfection and stimulation with K562-meso cells; the expressions of activation markers were tested by flow cytometry assay and showed greater activation of mesothelin-targeting chimeric antigen receptor-modified T cells than control T cells (CD107α: 71.9% vs 48.6%; CD27: 92.1% vs 61.8%; CD137: 55.5% vs 8.4%; CD28: 98.0% vs 82.1%; CD134: 37.5% vs 10.4%). Furthermore, mesothelin-targeting chimeric antigen receptor-modified T cells exerted cytotoxicity toward mesothelin-expressing EH-CA1b and EH-CA1a cells in an effector-to-target ratio-dependent manner, while leaving mesothelin-negative GSC-SD and EH-GB1 cells and normal liver L02 cells almost unharmed. Mesothelin-targeting chimeric antigen receptor-modified T cells secreted cytokines at higher levels when co-cultured with mesothelin-positive EH-CA1a and EH-CA1b cells than with mesothelin-negative GSC-SD and EH-GB1 cells. Enhanced cytotoxicity and cytokine secretion of mesothelin-targeting chimeric antigen receptor-modified T cells compared to control T cells were also observed when co-cultured with 293-meso cells (interferon γ: 85.1% ± 1.47% vs 8.3% ± 2.50%, p = 0.000; tumor necrosis factor α: 90.9% ± 4.67% vs 18.5% ± 3.62%, p = 0.0004; interleukin 2: 60.8% ± 2.00% vs 15.6% ± 2.06%, p = 0.002; interleukin 6: 6.4% ± 2.95% vs 1.7% ± 0.63%, p = 0.055). In addition, mesothelin-targeting chimeric antigen receptor-modified T cells showed greater inhibitory and proliferative capability than control T cells within EH-CA1a cell xenografts. This study shows the potential of mesothelin-targeting chimeric antigen receptor-modified T cells in treating bile duct carcinoma.


Bile Duct Neoplasms/therapy , DNA Transposable Elements , GPI-Linked Proteins/immunology , Immunotherapy/methods , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Bile Duct Neoplasms/pathology , Cells, Cultured , Humans , Mesothelin , Mice , Recombinant Fusion Proteins
4.
J Cancer ; 8(2): 199-206, 2017.
Article En | MEDLINE | ID: mdl-28243324

Targeting cancer stem cells with oncolytic virus (OV) holds great potential for thorough elimination of cancer cells. Based on our previous studies, we here established 11R-P53 and mGM-CSF carrying oncolytic adenovirus (OAV) SG655-mGMP and investigated its therapeutic effect on hepatocellular carcinoma stem cells Hep3B-C and teratoma stem cells ECCG5. Firstly, the augmenting effect of 11R in our construct was tested and confirmed by examining the expression of EGFP with Fluorescence and FCM assays after transfecting Hep3B-C and ECCG5 cells with OVA SG7605-EGFP and SG7605-11R-EGFP. Secondly, the expressions of 11R-P53 and GM-CSF in Hep3B-C and ECCG5 cells after transfection with OAV SG655-mGMP were detected by Western blot and Elisa assays, respectively. Thirdly, the enhanced growth inhibitory and augmented apoptosis inducing effects of OAV SG655-mGMP on Hep3B-C and ECCG5 cells were tested with FCM assays by comparing with the control, wild type 5 adenovirus, 11R-P53 carrying OVA in vitro. Lastly, the in vivo therapeutic effect of OAV SG655-mGMP toward ECCG5 cell-formed xenografts was studied by measuring tumor volumes post different treatments with PBS, OAV SG655-11R-P53, OAV SG655-mGM-CSF and OAV SG655-mGMP. Treatment with OAV SG655-mGMP induced significant xenograft growth inhibition, inflammation factor AIF1 expression and immune cells infiltration. Therefore, our OAV SG655-mGMP provides a novel platform to arm OVs to target cancer stem cells.

5.
Apoptosis ; 21(10): 1179-90, 2016 10.
Article En | MEDLINE | ID: mdl-27472927

V-set and transmembrane domain-containing 1 (VSTM1), which is downregulated in bone marrow cells from leukemia patients, may provide a diagnostic and treatment target. Here, a triple-regulated oncolytic adenovirus was constructed to carry a VSTM1 gene expression cassette, SG611-VSTM1, and contained the E1a gene with a 24-nucleotide deletion within the CR2 region under control of the human telomerase reverse transcriptase promoter, E1b gene directed by the hypoxia response element, and VSTM1 gene controlled by the cytomegalovirus promoter. Real-time quantitative PCR and Western blot analyses showed that SG611-VSTM1 expressed VSTM1 highly efficiently in the human leukemic cell line K562 compared with SG611. In Cell Counting Kit-8 and flow cytometric assays, SG611-VSTM1 exhibited more potent anti-proliferative and pro-apoptotic effects in leukemic cells compared with SG611 and exerted synergistic cytotoxicity with low-dose daunorubicin (DNR) in vitro. In xenograft models, SG611-VSTM1 intratumorally injected at a dose of 1 × 10(9) plaque forming units combined with intraperitoneally injected low-dose DNR displayed significantly stronger antitumor effects than either treatment alone. Histopathologic examination revealed that SG611-VSTM1 induced apoptosis of leukemic cells. These results implicate an important role for VSTM1 in the pathogenesis of leukemia, and SG611-VSTM1 may be a promising agent for enhancing chemosensitivity in leukemia therapy.


Adenoviridae/genetics , Antineoplastic Agents/administration & dosage , Daunorubicin/administration & dosage , Leukemia/therapy , Oncolytic Viruses/genetics , Receptors, Immunologic/genetics , Adenoviridae/physiology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Combined Modality Therapy , Female , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Leukemia/drug therapy , Leukemia/physiopathology , Leukemia/virology , Mice , Mice, Inbred BALB C , Mice, Nude , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Receptors, Immunologic/metabolism
6.
Int J Biol Sci ; 12(6): 718-29, 2016.
Article En | MEDLINE | ID: mdl-27194949

Recent years, we have witnessed significant progresses in both basic and clinical studies regarding novel therapeutic strategies with genetically engineered T cells. Modification with chimeric antigen receptors (CARs) endows T cells with tumor specific cytotoxicity and thus induce anti-tumor immunity against malignancies. However, targeting solid tumors is more challenging than targeting B-cell malignancies with CAR-T cells because of the histopathological structure features, specific antigens shortage and strong immunosuppressive environment of solid tumors. Meanwhile, the on-target/off-tumor toxicity caused by relative expression of target on normal tissues is another issue that should be reckoned. Optimization of the design of CAR vectors, exploration of new targets, addition of safe switches and combination with other treatments bring new vitality to the CAR-T cell based immunotherapy against solid tumors. In this review, we focus on the major obstacles limiting the application of CAR-T cell therapy toward solid tumors and summarize the measures to refine this new cancer therapeutic modality.


Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/isolation & purification , T-Lymphocytes/immunology , Animals , Cell- and Tissue-Based Therapy/methods , Humans , Neoplasms/metabolism , T-Lymphocytes/metabolism
7.
Sci Rep ; 5: 10043, 2015 Jun 04.
Article En | MEDLINE | ID: mdl-26040985

Nicotinamide phosphoribosyltransferase (NAMPT) is a promising anticancer target. Using high throughput screening system targeting NAMPT, we obtained a potent NAMPT inhibitor MS0 (China Patent ZL201110447488.9) with excellent in vitro activity (IC50 = 9.87 ± 1.15 nM) and anti-proliferative activity against multiple human cancer cell lines including stem-like cancer cells. Structure-activity relationship studies yielded several highly effective analogues. These inhibitors specifically bound NAMPT, rather than downstream NMNAT. We provided the first chemical case using cellular thermal shift assay to explain the difference between in vitro and cellular activity; MS7 showed best in vitro activity (IC50 = 0.93 ± 0.29 nM) but worst cellular activity due to poor target engagement in living cells. Site-directed mutagenesis studies identified important residues for NAMPT catalytic activity and inhibitor binding. The present findings contribute to deep understanding the action mode of NAMPT inhibitors and future development of NAMPT inhibitors as anticancer agents.


Drug Discovery , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Nicotinamide Phosphoribosyltransferase/chemistry , Protein Binding , Small Molecule Libraries , Structure-Activity Relationship
8.
Acta Pharmacol Sin ; 36(10): 1237-45, 2015 Oct.
Article En | MEDLINE | ID: mdl-25937637

AIM: Argonaute2 (AGO2) protein is the active part of RNA-induced silencing complex, cleaving the target mRNA strand complementary to their bound siRNA. An increasing number of miRNAs has been identified as essential to angiogenesis of hepatocellular carcinoma (HCC). In this study we investigated how AGO2 affected HCC angiogenesis. METHODS: Human HCC cell lines HepG2, Hep3B, Huh7, SMMC-7721, Bel-7404, MHCC97-H and LM-3, and human umbilical vein endothelial cells (HUVEC) were tested. The expression of AGO2 in HCC cells was knocked down with siRNA and restored using recombinant adenovirus expressing Ago2. The levels of relevant mRNAs and proteins were examined using RT-PCR, Western blot and EILSA. Nude mice were implanted with Huh7 or SMMC-7721 cells, and tumor volumes were measured. After the mice were euthanized, the xenograft tumors were used for immunohistological analysis. RESULTS: In 6 HCC cell lines, AGO2 protein expression was significantly correlated with VEGF expression (r=+0.79), and with VEGF secretion (r=+0.852). Knockdown of Ago2 in Huh7 cells and SMMC-7721 cells substantially decreased VEGF expression, whereas the restoration of AGO2 reversed both VEGF expression and secretion. Furthermore, knockdown of Ago2 significantly up-regulated the expression of PTEN (a tumor suppressor involved in the inhibition of HCC angiogenesis), and vice versa. Moreover, the specific PTEN inhibitor bisperoxovanadate (7, 14, 28 nmol/L) dose-dependently restored the expression of VEGF and the capacity of HCC cells to induce HUVECs to form capillary tubule structures. In the xenograft nude mice, knockdown of Ago2 markedly suppressed the tumor growth and decreased PTEN expression and CD31-positive microvascular in the xenograft tumors. CONCLUSION: A direct relationship exists between the miRNA processing machinery AGO2 and HCC angiogenesis that is mediated by the AGO2/PTEN/VEGF signaling pathway. The results suggest the high value of Ago2 knockdown in anti-angiogenesis therapy for HCC.


Argonaute Proteins/genetics , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Neovascularization, Pathologic/genetics , PTEN Phosphohydrolase/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice, Nude , MicroRNAs/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , RNA, Small Interfering/genetics , RNAi Therapeutics , Signal Transduction
9.
J Hepatol ; 59(4): 762-8, 2013 Oct.
Article En | MEDLINE | ID: mdl-23714159

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) develops in response to chronic hepatic injury. Although induced cell death is regarded as the major component of p53 tumor-suppressive activity, we recently found that sustained p53 activation subsequent to DNA damage promotes inflammation-associated hepatocarcinogenesis. Here we aim at exploring the mechanism linking p53 activation and hepatic inflammation during hepatocarcinogenesis. METHODS: p53(-/-) hepatocytes expressing inducible p53 and primary wild type hepatocytes were treated to induce p53 expression. The supernatants were collected and analyzed for the presence of released inflammatory cytokines. Ethyl pyruvate was used in a rat model of carcinogen-induced hepatocarcinogenesis to examine its effect on p53-dependent chronic hepatic injury, inflammation, and tumorigenesis. RESULTS: Here we show that cytoplasmic translocation and circulating levels of potent inflammatory molecule high-mobility group protein 1 (HMGB1) were greater in wild type rats than in p53(+/-) rats following carcinogen administration. Restoration of p53 expression in p53-null hepatocytes or induction of endogenous p53 in wild type hepatocytes gives rise to the release of HMGB1. Administration of the HMGB1 release inhibitor ethyl pyruvate, which does not affect p53-mediated hepatic apoptosis, substantially prevented carcinogen-induced cirrhosis and tumorigenesis in rat livers. CONCLUSIONS: These results suggest that although p53 is usually regarded as a tumor suppressor, its constant activation can promote pro-tumorigenic inflammation, at least in part, via inducing HMGB1 release. Application of HMGB1 inhibitors when restoring p53 in cancer therapy might protect against pro-tumorigenic effects while leaving p53-mediated clearance of malignant cells intact.


Genes, p53 , HMGB1 Protein/metabolism , Liver Neoplasms, Experimental/etiology , Animals , Cell Line , Diethylnitrosamine/toxicity , Gene Knockout Techniques , Hepatitis, Chronic/etiology , Hepatitis, Chronic/metabolism , Hepatitis, Chronic/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Rats , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism
10.
J Cell Mol Med ; 16(6): 1298-309, 2012 Jun.
Article En | MEDLINE | ID: mdl-21794078

Cancer Targeting Gene-Viro-Therapy (CTGVT) is a promising cancer therapeutical strategy that strengthens the anti-tumour effect of oncolytic virus by expressing inserted foreign anti-tumour genes. In this work, we constructed a novel adenoviral vector controlled by the tumour-specific survivin promoter on the basis of the ZD55 vector, which is an E1B55KD gene deleted vector we previously constructed. Compared with the original ZD55 vector, this new adenoviral vector (ZD55SP/E1A) showed much better ability of replication and reporter gene expression. We then combined anti-tumour gene interleukine-24 (IL-24) with an RNA polymerase III-dependent U6 promoter driving short hairpin RNA (shRNA) that targets M-phase phosphoprotein 1 (MPHOSPH1, a newly identified oncogene) by inserting the IL-24 and the shRNA of MPHOSPH1 (shMPP1) expression cassettes into the new ZD55SP/E1A vector. Our results demonstrated excellent anti-tumour effect of ZD55SP/E1A-IL-24-shMPP1 in vitro on multiple cancer cell lines such as lung cancer, liver cancer and ovarian caner. At high multiplicity-of-infection (MOI), ZD55SP/E1A-IL-24-shMPP1 triggered post-mitotic apoptosis in cancer cells by inducing prolonged mitotic arrest; while at low MOI, senescence was induced. More importantly, ZD55SP/E1A-IL-24-shMPP1 also showed excellent anti-tumour effects in vivo on SW620 xenograft nude mice. In conclusion, our strategy of constructing an IL-24 and shMPP1 dual gene expressing oncolytic adenoviral vector, which is regulated by the survivin promoter and E1B55KD deletion, could be a promising method of cancer gene therapy.


Adenoviridae/genetics , Genes, Tumor Suppressor , Genetic Therapy/methods , Genetic Vectors , Oncolytic Viruses/genetics , Animals , Apoptosis , Cell Line, Tumor , Female , Gene Deletion , Gene Expression Regulation , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Interleukins/genetics , Interleukins/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/pathology , RNA, Small Interfering/genetics , Repressor Proteins , Survivin , Xenograft Model Antitumor Assays
11.
Oncol Rep ; 27(3): 775-82, 2012 Mar.
Article En | MEDLINE | ID: mdl-22134783

Gallbladder cancer is a fatal neoplasia with an extremely low survival rate. Liver invasion and metastasis are the most common causes of death; however, the metastatic mechanism is still unclear, and no effective treatment methods are available. To provide comprehensive and profound approaches in investigating the metastatic mechanism and treatment methods, new cell lines derived from liver metastasis are urgently needed. A hepatic metastasis lesion was obtained from a 65-year-old patient, and was treated using a primary culture method to establish a novel gallbladder cancer cell line. Different in vitro/in vivo methods were used to characterize the phenotypes of this cell line. The gallbladder cancer cell line was named EH-GB2, with a roughly 48-h doubling time. The cell line represents stronger colony formation and migration abilities than the control group. The cells showed complicated chromosomal abnormalities. EH-GB2 cells showed epithelial-to-mesenchymal transition (EMT) and the mRNA expression levels of E-cadherin and integrin were decreased, and those of vimentin, Snail, Twist, matrix metalloproteinase-1 (MMP-1) and MMP-2 were increased in comparison with control cells. The in vivo study demonstrated that EH-GB2 cells show significant tumorigenicity in nude mice. The EH-GB2 established gallbladder cancer cell line is useful for future studies of gallbladder cancer development, progression, metastasis and therapy.


Cell Line, Tumor/pathology , Gallbladder Neoplasms/pathology , Liver Neoplasms/secondary , Aged , Animals , Cadherins/genetics , Carcinogenicity Tests/methods , Cell Movement/genetics , Gallbladder Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Integrins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 2/genetics , Mice , Mice, Nude , Phenotype , RNA, Messenger/genetics , Snails/genetics , Twist-Related Protein 1/genetics , Vimentin/genetics
13.
Hum Gene Ther ; 22(9): 1109-19, 2011 Sep.
Article En | MEDLINE | ID: mdl-21361790

Aberrant JAK/STAT3 pathway has been reported to be related to hepatocellular carcinoma (HCC) in many cell lines. In this study, a double-regulated oncolytic adenovirus vector that can replicate and induce a cytopathic effect in alpha-fetoprotein (AFP)-positive HCC cell lines with p53 dysfunction was successfully constructed. Two therapeutic genes, suppressor of cytokine signaling 3 (SOCS3) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), were chosen and incorporated into this vector system, respectively. The combined treatment of AFP-D55-SOCS3 and AFP-D55-TRAIL (2:3 ratio) exhibited potent antitumor activity in AFP-positive HCC cell lines compared with any other treatment both in vitro and in vivo. Specific replication and low progeny yield in AFP-positive HCC cell lines rendered these double-regulated oncolytic adenoviruses remarkably safe. Our data demonstrated that restoration of SOCS3, which inhibits the JAK/STAT3 pathway, by AFP-D55-SOCS3 not only could antagonize HCC therapeutic resistance to TRAIL and adenoviruses, but could also induce cell cycle arrest in HCC cell lines. SOCS3 could down-regulate Cyclin D1 and anti-apoptotic proteins such as XIAP, Survivin, Bcl-xL, and Mcl-1, which are responsible for the synergistic inhibitory effects of AFP-D55-SOCS3 and AFP-D55-TRAIL. Dual gene and double-regulated oncolytic adenoviruses may provide safety and excellent antitumor effects for liver cancer, which is the advantage of a cancer-targeting gene virotherapy strategy.


Adenoviridae/genetics , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Oncolytic Viruses/genetics , Suppressor of Cytokine Signaling Proteins/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cytopathogenic Effect, Viral , Female , Gene Expression , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/toxicity , HEK293 Cells , HT29 Cells , HeLa Cells , Hep G2 Cells , Humans , Kaplan-Meier Estimate , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Oncolytic Virotherapy , Promoter Regions, Genetic , Suppressor of Cytokine Signaling Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Burden/genetics , Virus Replication , Xenograft Model Antitumor Assays , alpha-Fetoproteins/genetics
14.
Zhonghua Zhong Liu Za Zhi ; 32(2): 84-7, 2010 Feb.
Article Zh | MEDLINE | ID: mdl-20403235

OBJECTIVE: To establish a human gallbladder carcinoma cell line derived from a metastatic gallbladder carcinoma and identify its biological characteristics. METHODS: Tissue samples were separated from the surgical specimen obtained from a patient with metastatic carcinoma and single-cell suspension was prepared. Then the cells were cultured in DMEM medium supplemented with 15% fetal bovine serum. The morphology of tumor cells was observed under an electron microscope. The cell growth curve was plotted. The tumorigenicity of the cell line was studied by subcutaneous inoculation in SCID mice. The cells were infected by lentiviral vector carrying fluorescent report genes (lenti-GFP and lenti-Red2) separately for expressions of GFP and Red2, respectively. RESULTS: A novel metastatic gallbladder carcinoma cell line was successfully established and named "EH-GB1". It could be passaged for over 20 generations with typical malignant epithelial morphology and a stable growth cycle of 24 h. Tumors were formed in all of the 10 SCID mice inoculated with EH-GB1 cells subcutaneously, and the tumor cells were tumor marker CA19-9-positive. Continuous expressions of fluorescent report genes were observed in EH-GB1 cells infected by lenti-GFP and lenti-Red2. CONCLUSION: EH-GB1 cells might be the first stable cell line of human gallbladder carcinoma established from a metastatic focus of gallbladder carcinoma. This cell line with continuous expressions of GFP and Red2 might be a novel and perfect experimental model for clinical and basic research on gallbladder carcinoma.


Abdominal Neoplasms/secondary , Adenocarcinoma/pathology , Cell Line, Tumor/pathology , Gallbladder Neoplasms/pathology , Abdominal Neoplasms/metabolism , Abdominal Neoplasms/pathology , Abdominal Wall , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Animals , CA-19-9 Antigen/metabolism , Cell Line, Tumor/metabolism , Female , Gallbladder Neoplasms/metabolism , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Nude , Mice, SCID , Middle Aged , Neoplasm Transplantation
15.
Zhonghua Yi Xue Za Zhi ; 89(20): 1372-6, 2009 May 26.
Article Zh | MEDLINE | ID: mdl-19671324

OBJECTIVE: To construct a RU486 inducible recombinant adenovirus of murine IL-12 protein and study its effect and safety on colonic cancer. METHODS: The replication-defective recombinant adenovirus were produced after cotransfection of shutter vector pDC-RUmIL-12 and adenovirus DNA helper plasmid pBHGloxDeltaE1, 3Cre into HEK293 cells. The recombined adenovirus was purified by CsCl density gradient centrifugation and its titer was determined by end point dilution assay. Expression of this regulatable recombinant adenovirus vector in infected C26 colonic carcinoma cells was tested by ELISA kit in vitro. The tumor model was established by hypodermic inoculation of C26 cells. Sixty tumor-bearing mice were randomly divided into 4 groups: Ad-buffer group; Ad-RUmIL-12 group; Ad-RUmIL-12 + RU486 group and Ad-mIL-12 group, and the treatment effects and side effects were evaluated. RESULTS: The adenoviral vector containing murine IL-12 gene was identify by PCR. The viral titer of Ad-RUmIL-12 was 4.62 x 10(10) pfu/ml. The expression of IL-12 protein was induced by the RU486 and the highest expression (516 +/- 43) pg/ml whereas no significant IL-12 protein was detected without inducer or getting rid of the inducer [(38 +/- 3) pg/ml and (42 +/- 5) pg/ml respectively]. The tumor size increased rapidly in group Ad-buffer and group Ad-RUmIL-12 (P > 0.05). Administration of Ad-RUmIL-12 + RU486 and Ad-mIL-12 were showed to delay markedly the growth of transplanted C26 tumor (P > 0.05). Significantly necrosis was observed in both Ad-mIL-12 and Ad-RUmIL-12 + RU486 experimental groups, but the level of the serum alanine transaminase and the rate of side effect was higher in Ad-mIL-12 group (4/15 and 10/15 respectively, P < 0.05). CONCLUSION: A RU486 regulatable recombinant adenoviral vector containing IL-12 gene was successfully constructed. The expression of vector Ad-RUmIL-12, regulated by inducer RU486 in vivo, can obviously improve safety in tumor treatment and provide a good primer for further researches on in vivo gene therapy.


Adenoviridae/genetics , Colonic Neoplasms/therapy , Genetic Therapy , Interleukin-12/therapeutic use , Animals , Cell Line, Tumor , Female , Gene Transfer Techniques , Genetic Vectors , Interleukin-12/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mifepristone , Neoplasms, Experimental/therapy , Recombinant Fusion Proteins , Transduction, Genetic , Transfection
16.
Biochim Biophys Acta ; 1794(10): 1433-40, 2009 Oct.
Article En | MEDLINE | ID: mdl-19520192

Colon cancer is one of the most common malignancies in the world. Oxaliplatin, a third-generation platinum compound, is widely used in clinical chemotherapy of colon cancer. Although the mechanisms of the antitumor effect of Oxaliplatin have been investigated in recent years, the proteomic changes that are associated with the cellular response to this compound are poorly understood. In this study, we performed a comparative proteomic analysis to survey the global changes in protein expression levels after Oxaliplatin treatment in three colon cancer cell lines: HT29, SW620, and LoVo. Two-dimensional gel electrophoresis coupled with MALDI-TOF/TOF mass spectrometry revealed 57, 48, and 53 differentially expressed proteins in the three cell lines (HT29, SW620 and LoVo, respectively) after Oxaliplatin treatment. Of these proteins, 21 overlapped among all three cell lines. These overlapping proteins participate in many cellular processes, such as apoptosis, signal transduction, transcription and translation, cell structural organization, and metabolism. Additionally, the expression levels of ezrin (EZRI), heat-shock protein beta-1 (HSPB1), translationally controlled tumor protein (TCTP), and cell division control protein 2 homolog (CDC2) were confirmed by immunoblotting. This is the first direct proteomic analysis of Oxaliplatin-treated colon cancer cells. Several interesting proteins that we found warrant further investigation owing to their potential significant functions in the antitumor effect of Oxaliplatin.


Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Organoplatinum Compounds/therapeutic use , Proteome/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Electrophoresis, Gel, Two-Dimensional , Humans , Immunoblotting , Neoplasm Proteins/isolation & purification , Neoplasm Proteins/metabolism , Oxaliplatin , Proteome/drug effects , Proteome/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Tumor Protein, Translationally-Controlled 1
17.
Apoptosis ; 14(9): 1086-94, 2009 Sep.
Article En | MEDLINE | ID: mdl-19551515

PDCD5 (programmed cell death 5) accelerates apoptosis of certain tumor cells and the replication-defective Ad-PDCD5 may be a promising agent for enhancing chemosensitivity. In this study, a triple-regulated conditionally replicating adenoviruses (CRAd) carrying PDCD5 gene expression cassette, SG611-PDCD5, was engineered. In SG611-PDCD5, the E1a gene with a deletion of 24 nucleotides within CR2 region is controlled under the human telomerase reverse transcriptase (hTERT) promoter, the E1b gene expression is directed by the hypoxia response element (HRE), whereas the PDCD5 gene is controlled by the cytomegalovirus promoter. The tumor-selective replication of this virus and its antitumor efficacy were characterized in several leukemic cell lines in vitro and in xenograft models of human leukemic cell line in nude mice. It was found by RQ-RT-PCR assay that SG611-PDCD5 expressed PDCD5 efficiently in leukemic cells. In K562 tumor xenograft models, SG611-PDCD5 displayed a tumor killing capacity. At a dose of 1 x 10(9) plaque-forming units, SG611-PDCD5 alone could completely inhibit the tumor growth and more effective than replication-defective Ad-PDCD5. Histopathologic examination revealed that SG611-PDCD5 administration resulted in leukemic cell apoptosis. We concluded that the triple-regulated SG611-PDCD5, as a more potent and safer antitumor therapeutic, could provide a new strategy for leukemia biotherapy.


Adenoviridae/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/therapeutic use , Genetic Therapy , Leukemia/therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/therapeutic use , Oncolytic Viruses/genetics , Xenograft Model Antitumor Assays , Animals , Apoptosis , Cell Line, Tumor , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Leukemia/genetics , Leukemia/pathology , Mice , Mutagenesis, Insertional , Oncolytic Virotherapy , Transgenes
18.
Cancer Sci ; 100(4): 678-83, 2009 Apr.
Article En | MEDLINE | ID: mdl-19298599

Following targeted gene virotherapy, the suppression of tumorigenicity 13 (ST13) gene was inserted into the double-regulated oncolytic adenovirus SG500 to ensure more safety and potent antitumor activity against colorectal cancer in vitro and in vivo. We generated the ST13-expressing oncolytic adenovirus SG500-ST13, with which colorectal carcinoma cell lines SW620 and HCT116, and the lung fibroblast cell line WI38, were infected. Crystal violet staining was carried out to detect the cytopathic effect in cells, and the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method was used to assay cell viability. The effect of apoptosis induced by SG500-ST13 was confirmed by Hoechst staining and the TdT-mediated dUTP-biotin nick-end labeling method. To further identify the antitumor effects of SG500-ST13 on HCT116 xenografts in Balb/c nude mice, the induction of cell death was assessed by hematoxylin-eosin staining. Immunohistochemical study was also carried out.


Adenoviridae/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Genetic Therapy/methods , Oncolytic Virotherapy/methods , Adenoviridae/physiology , Animals , Apoptosis/drug effects , Benzimidazoles/metabolism , Cell Death , Cell Line, Tumor , Cell Survival , Coloring Agents/metabolism , Cytopathogenic Effect, Viral/genetics , Cytopathogenic Effect, Viral/physiology , Eosine Yellowish-(YS)/metabolism , Female , Fluorescent Dyes/metabolism , Gene Expression , Genetic Vectors , Gentian Violet/metabolism , HCT116 Cells , Hematoxylin/metabolism , Humans , Immunohistochemistry , In Situ Nick-End Labeling/methods , Mice , Mice, Nude , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Xenograft Model Antitumor Assays
19.
Nan Fang Yi Ke Da Xue Xue Bao ; 28(12): 2113-6, 2008 Dec.
Article Zh | MEDLINE | ID: mdl-19114334

OBJECTIVE: To construct an inducible eukaryotic vector carrying red fluorescent protein (DsRed) and evaluate the regulation of DsRed gene expression in vitro. METHODS: The vector pRS17-RUDsRed containing DsRed gene, promoter and RU486-inducible system was constructed using molecular biological methods. To minimize potential interference, the two transcriptional elements were spaced with a 1.6 kb insulator. Fluorescence microscopy and flow cytometry were used to observe the activation of this regulatable vector after transfection in MFC cells. RESULTS: The vector was identified by digestion with different restriction enzymes, sequencing and PCR. In the absence of RU486, the cells transfected with the vector exhibited very low DsRed protein expression, and the addition of RU486 induced efficient DsRed expression in the cells. CONCLUSION: The RU486-inducible eukaryotic vector carrying DsRed protein allows effective regulation of the target gene expression in vitro, which provides a useful tool for gene regulation and gene therapy studies.


Genetic Vectors/genetics , Luminescent Proteins/genetics , Mifepristone/pharmacology , Promoter Regions, Genetic/genetics , Stomach Neoplasms/genetics , Gene Expression Regulation , Genetic Therapy/methods , Humans , Luminescent Proteins/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured , Red Fluorescent Protein
20.
World J Gastroenterol ; 14(8): 1274-9, 2008 Feb 28.
Article En | MEDLINE | ID: mdl-18300357

AIM: To evaluate the therapeutic efficiency of replicative adenovirus CNHK300 targeted in telomerase-positive hepatocellular carcinoma. METHODS: CNHK300, ONYX-015 (55 kDa protein deleted adenovirus) and wtAd5 (wild type adenovirus 5) were compared, and virus proliferation assay, cell viability assay, Western blot and fluorescence microscopy were used to evaluate the proliferation and cytolysis selectivity of CNHK300. RESULTS: The replicative multiples in Hep3B and HepG II after 48 h of CNHK300 proliferation were 40625 and 65326 fold, respectively, similar to that of wtAd5. However, CNHK300 exhibited attenuated replicative ability in normal fibroblast cell line BJ. CNHK300 could lyse hepatocellular carcinoma cells at a low multiplicity of infection (MOI), but could not affect growth of normal cells even at a high MOI. CONCLUSION: CNHK300 is a cancer-selective replication-competent adenovirus which can cause oncolysis of liver cancer cells as well as wtAd5 (wild type adenovirus 5), but had severely attenuated replicative and cytolytic ability in normal cells. This novel strategy of cancer treatment offers a promising treatment platform.


Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Oncolytic Virotherapy/methods , Telomerase/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Fibroblasts/metabolism , Humans , Microscopy, Fluorescence/methods , Promoter Regions, Genetic , RNA, Messenger/metabolism , Telomerase/biosynthesis , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Virus Replication
...