Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Tumour Biol ; 39(4): 1010428317695949, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28381173

RESUMO

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.


Assuntos
Neoplasias dos Ductos Biliares/terapia , Elementos de DNA Transponíveis , Proteínas Ligadas por GPI/imunologia , Imunoterapia/métodos , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Neoplasias dos Ductos Biliares/patologia , Células Cultivadas , Humanos , Mesotelina , Camundongos , Proteínas Recombinantes de Fusão
2.
Apoptosis ; 21(10): 1179-90, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27472927

RESUMO

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.


Assuntos
Adenoviridae/genética , Antineoplásicos/administração & dosagem , Daunorrubicina/administração & dosagem , Leucemia/terapia , Vírus Oncolíticos/genética , Receptores Imunológicos/genética , Adenoviridae/fisiologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Terapia Genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Leucemia/tratamento farmacológico , Leucemia/fisiopatologia , Leucemia/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Receptores Imunológicos/metabolismo
3.
Acta Pharmacol Sin ; 36(10): 1237-45, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25937637

RESUMO

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.


Assuntos
Proteínas Argonautas/genética , Carcinoma Hepatocelular/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Neovascularização Patológica/genética , PTEN Fosfo-Hidrolase/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Linhagem Celular Tumoral , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Camundongos Nus , MicroRNAs/genética , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , RNA Interferente Pequeno/genética , Terapêutica com RNAi , Transdução de Sinais
4.
J Hepatol ; 59(4): 762-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23714159

RESUMO

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.


Assuntos
Genes p53 , Proteína HMGB1/metabolismo , Neoplasias Hepáticas Experimentais/etiologia , Animais , Linhagem Celular , Dietilnitrosamina/toxicidade , Técnicas de Inativação de Genes , Hepatite Crônica/etiologia , Hepatite Crônica/metabolismo , Hepatite Crônica/patologia , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Ratos , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo
5.
J Cell Mol Med ; 16(6): 1298-309, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21794078

RESUMO

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.


Assuntos
Adenoviridae/genética , Genes Supressores de Tumor , Terapia Genética/métodos , Vetores Genéticos , Vírus Oncolíticos/genética , Animais , Apoptose , Linhagem Celular Tumoral , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/patologia , RNA Interferente Pequeno/genética , Proteínas Repressoras , Survivina , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Biochim Biophys Acta ; 1794(10): 1433-40, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19520192

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Compostos Organoplatínicos/uso terapêutico , Proteoma/metabolismo , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/patologia , Eletroforese em Gel Bidimensional , Humanos , Immunoblotting , Proteínas de Neoplasias/isolamento & purificação , Proteínas de Neoplasias/metabolismo , Oxaliplatina , Proteoma/efeitos dos fármacos , Proteoma/isolamento & purificação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem , Proteína Tumoral 1 Controlada por Tradução
7.
Zhonghua Zhong Liu Za Zhi ; 32(2): 84-7, 2010 Feb.
Artigo em Zh | MEDLINE | ID: mdl-20403235

RESUMO

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.


Assuntos
Neoplasias Abdominais/secundário , Adenocarcinoma/patologia , Linhagem Celular Tumoral/patologia , Neoplasias da Vesícula Biliar/patologia , Neoplasias Abdominais/metabolismo , Neoplasias Abdominais/patologia , Parede Abdominal , Adenocarcinoma/metabolismo , Adenocarcinoma/secundário , Animais , Antígeno CA-19-9/metabolismo , Linhagem Celular Tumoral/metabolismo , Feminino , Neoplasias da Vesícula Biliar/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Camundongos Nus , Camundongos SCID , Pessoa de Meia-Idade , Transplante de Neoplasias
8.
Mol Oncol ; 14(3): 657-668, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31899582

RESUMO

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.


Assuntos
Antígenos de Diferenciação/metabolismo , Antígeno CD47/imunologia , Imunoglobulina G/metabolismo , Imunoterapia/métodos , Macrófagos/imunologia , Neoplasias Ovarianas/imunologia , Receptores Imunológicos/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Antígenos de Diferenciação/genética , Antígeno CD47/genética , Antígeno CD47/metabolismo , Pontos de Checagem do Ciclo Celular/imunologia , Morte Celular/imunologia , Linhagem Celular Tumoral , Testes Imunológicos de Citotoxicidade , Feminino , Humanos , Imunoglobulina G/genética , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Fagocitose/genética , Fagocitose/imunologia , Receptores Imunológicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Telomerase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cancer Sci ; 100(4): 678-83, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19298599

RESUMO

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.


Assuntos
Adenoviridae/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/terapia , Terapia Genética/métodos , Terapia Viral Oncolítica/métodos , Adenoviridae/fisiologia , Animais , Apoptose/efeitos dos fármacos , Benzimidazóis/metabolismo , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Corantes/metabolismo , Efeito Citopatogênico Viral/genética , Efeito Citopatogênico Viral/fisiologia , Amarelo de Eosina-(YS)/metabolismo , Feminino , Corantes Fluorescentes/metabolismo , Expressão Gênica , Vetores Genéticos , Violeta Genciana/metabolismo , Células HCT116 , Hematoxilina/metabolismo , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas/métodos , Camundongos , Camundongos Nus , Sais de Tetrazólio/metabolismo , Tiazóis/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Apoptosis ; 14(9): 1086-94, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19551515

RESUMO

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.


Assuntos
Adenoviridae/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/uso terapêutico , Terapia Genética , Leucemia/terapia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/uso terapêutico , Vírus Oncolíticos/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Apoptose , Linhagem Celular Tumoral , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Leucemia/genética , Leucemia/patologia , Camundongos , Mutagênese Insercional , Terapia Viral Oncolítica , Transgenes
11.
Zhonghua Yi Xue Za Zhi ; 89(20): 1372-6, 2009 May 26.
Artigo em Zh | MEDLINE | ID: mdl-19671324

RESUMO

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.


Assuntos
Adenoviridae/genética , Neoplasias do Colo/terapia , Terapia Genética , Interleucina-12/uso terapêutico , Animais , Linhagem Celular Tumoral , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Interleucina-12/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mifepristona , Neoplasias Experimentais/terapia , Proteínas Recombinantes de Fusão , Transdução Genética , Transfecção
12.
World J Gastroenterol ; 14(8): 1274-9, 2008 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-18300357

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Terapia Viral Oncolítica/métodos , Telomerase/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Fibroblastos/metabolismo , Humanos , Microscopia de Fluorescência/métodos , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Telomerase/biossíntese , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Replicação Viral
13.
Cell Death Dis ; 9(2): 177, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29415996

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/terapia , Receptores ErbB/imunologia , Imunoterapia Adotiva/métodos , Neoplasias Pulmonares/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/transplante , Animais , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Camundongos , Distribuição Aleatória , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Chin Med Sci J ; 22(2): 119-22, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17763585

RESUMO

OBJECTIVE: To explore the feasibility and safety of gene transfer into porcine myocardium via the pericardial cavity by a homemade easy device. METHODS: Replication-deficient recombinant adenoviral vector carrying LacZ report gene (Ad-LacZ) was constructed by the calcium phosphate precipitation method. Twelve healthy Chinese mini-swine were randomly divided into experimental group (n = 6) and control group (n = 6). Acute myocardial infarction (AMI) model was established by balloon occlusion of the distal part of D1 branch of left anterior descending (LAD) artery, at the same time the intrapericardial cavity injections were performed through the small incision of the abdominal wall below the xyphoid appendix using a homemade device. Then gene transfer was performed using a central venous catheter. The pericardium was pretreated with injection of a mixture of collagenase (1,200 U) and hyaluronidase (3,000 U) in both groups. Then 2.0 x 10(9) plaque formation unit (PFU) Ad-LacZ was injected into the pericardial cavity in experimental group, while 1 mL of normal saline was injected in the control group. The beta-galactosidase activity detection and X-gal staining of the ischemic myocardium were performed on the 3rd, 7th, and 28th day after injection. RESULTS: The LAD artery was occluded completely and infarction and ischemia were detected by histological assessment In experimental group, the X-gal staining positive cells and beta-galactosidase activity quantification were detectable on the 3rd day after injection, increased markedly on the 7th day, and then declined on the 28th day. The transfer efficiencies indicated by the positive myocardial cells were 16.7%, 45.6% , 22.8% on the 3rd, 7th, 28th day, respectively. In control group, no positive cells and beta-galactosidase activity were observed. CONCLUSION: Adenovirus can be transferred into ischemic myocardium and express target gene in the AMI model for four weeks with the homemade easy device via pericardial cavity pretreated by collagenase and hyaluronidase.


Assuntos
Técnicas de Transferência de Genes , Coração , Óperon Lac , Pericárdio/fisiologia , Punções/métodos , Adenoviridae/genética , Animais , Animais Geneticamente Modificados , Genes Reporter , Vetores Genéticos , Miocárdio/enzimologia , Suínos , Porco Miniatura , beta-Galactosidase/genética
15.
J Cancer ; 8(2): 199-206, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28243324

RESUMO

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.

16.
World J Gastroenterol ; 12(47): 7613-20, 2006 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-17171789

RESUMO

AIM: To construct a tumor-selective replication-competent adenovirus (RCAd), SG300, using a modified promoter of human telomerase reverse transcriptase (hTERT). METHODS: The antitumor efficacy of SG300 in hepatocellular carcinoma was assessed in vitro and in vivo. In vitro cell viability by MTT assay was used to assess the tumor-selective oncolysis and safety features of SG300, and in vivo antitumor activity of SG300 was assessed in established hepatocellular carcinoma models in nude mice. RESULTS: SG300 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. Both in Hep3B and SMMC-7721 xenograft models of hepatocellular carcinoma, SG300 had an obvious antitumor effect, resulting in a decrease in tumor volume. Its selective oncolysis to tumor cells and safety to normal cells was also superior to that of ONYX-015. Pathological examination of tumor specimens showed that SG300 replicated selectively in cancer cells and resulted in apoptosis and necrosis of cancer cells. CONCLUSION: hTERT promoter-regulated replicative adenovirus SG300 has a better cancer-selective replication-competent ability, and can specifically kill a wide range of cancer cells with positive telomerase activity, and thus has better potential for targeting therapy of hepatocellular carcinoma.


Assuntos
Adenoviridae/genética , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Terapia Viral Oncolítica/métodos , Telomerase/genética , Animais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Vírus Oncolíticos/genética , Regiões Promotoras Genéticas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Int J Biol Sci ; 12(6): 718-29, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27194949

RESUMO

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.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias/imunologia , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/isolamento & purificação , Linfócitos T/imunologia , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Humanos , Neoplasias/metabolismo , Linfócitos T/metabolismo
19.
Chin Med J (Engl) ; 118(3): 179-85, 2005 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-15740644

RESUMO

BACKGROUND: The expression of therapeutic gene and its anti-tumor effects will be augmented and a synergism of oncolytic virus with the therapeutic gene is speculated. This study was undertaken to assess the anti-tumor effects of a novel gene-viral therapeutic system CNHK300-mEndostatin (CNHK300-mE) in hepatocellular carcinoma (HCC). METHODS: A novel gene-viral therapeutic system named CNHK300-mE was constructed using the human telomerase reverse transcriptase (hTERT) promoter to drive the expression of the adenovirus E1A gene and cloning the therapeutic gene mouse endostatin into the adenovirus genome. By the tissue culture infectious dose 50 (TCID50) method and cytoviability assay, the replicative and cytolytic capabilities of CNHK300-mE in two HCC lines (HepGII and Hep3B) and one normal cell line (MRC-5) were analyzed, and the transgene expressions of mouse endostatin in vitro and in vivo were detected by Western blotting and ELISA assay. Tumor growth suppression and anti-angiogenesis effects in vivo were investigated using nude mice xenografts model derived from SMMC-7721 HCC cells. RESULTS: The 3296-fold replicating capacity of CNHK300-mE in HCC cell lines versus in the normal cell line at 96 hours post infection and the 25-fold effective dose for killing 50% cells (ED50) in the normal cell line versus HCC cell lines, which were both superior to ONYX-015, were observed. Tumor growth suppression of CNHK300-mE superior to either Ad-mE or ONYX-015 was demonstrated (P < 0.01) and the anti-angiogenic effects in vivo superior to Ad-mE were also observed with immunohistochemical staining of von Willebrand factor. In comparison with non-replicative adenovirus Ad-mE, the transgene expression of mE mediated by CNHK300-mE was significantly higher in vitro (P < 0.005) and in vivo (P < 0.05). CONCLUSION: Being capable of replicating in and lysing the telomerase-positive HCC cells and mediating effective expression of the therapeutic gene in vitro and in vivo, the novel gene-viral therapeutic system CNHK300-mE is potentially effective in the treatment of HCC.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Terapia Genética , Neoplasias Hepáticas Experimentais/terapia , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Transplante Heterólogo , Replicação Viral
20.
Zhonghua Zhong Liu Za Zhi ; 27(2): 69-72, 2005 Feb.
Artigo em Zh | MEDLINE | ID: mdl-15946541

RESUMO

OBJECTIVE: To develop a novel vector system, which combines the advantages of the gene therapy, antiangiogenic therapy and virus therapy, and to observe its effect on lung cancer. METHODS: Human angiostatin gene hA(k1-5) was inserted into the genome of the replicative virus specific for the tumor cells by virus recombination technology. The expression of hA(k1-5), its effect on tumor growth in vitro and in vivo were studied. RESULTS: A new kind of gene-viral vector system, designated as CNHK200-hA(k1-5), in which the E1b55 000 gene was deleted but the E1a gene of adenovirus preserved, was constructed. The novel vector system possessed the same property as the replicative virus ONYX-015, which replicates in p53- tumor cells but not in normal cells, thus specifically kills tumor cells. In vitro, CNHK200-hA and Ad-hA both could kill A549 tumor cells but the latter needed 100 times more MOI to achieve the same amplitude of cell killing. In vivo, the therapeutic effect of CNHK200-hA on human lung cancer A549 xenograft in nude mice was significantly better than that of Ad-hA and that of tumor-replicative virus ONYX-015. CONCLUSION: CNHK200-hA(k1-5), a novel vector is constructed in which the angiostatin gene is inserted into the genome of the replicative adenovirus cytotoxic to p53-negative tumor cells. It has the advantages of specific tumor targeting, high level gene expression in tumor cells, and potent tumoricidal activity.


Assuntos
Adenoviridae/genética , Angiostatinas/fisiologia , Terapia Genética , Neoplasias Pulmonares/terapia , Proteínas E1A de Adenovirus/genética , Angiostatinas/biossíntese , Angiostatinas/genética , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Vetores Genéticos , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA