Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Clin Exp Rheumatol ; 33(6): 839-43, 2015.
Article in English | MEDLINE | ID: mdl-26315300

ABSTRACT

OBJECTIVES: Selectively targeting signalling pathways represents a promising pharmacological approach in rheumatoid arthritis (RA). Abundant levels of epidermal growth factor receptor (EGFR) are expressed in the synovial lining layers, and the anti-arthritis effect of erlotinib and lapatinib, small-molecule EGFR tyrosine kinase inhibitors (TKIs), has been demonstrated through the systemic administration on experimental arthritis models. Nevertheless, their therapeutic responses by the intra-articular (i.a.) route remain to be explored in rheumatoid joint. METHODS: The administration of an EGFR TKI (a gefitinib analogue) was explored in two in vivo models of collagen-induced arthritis (CIA) and in vitro experiments by using synovial fibroblasts (SF) from RA patients and CIA rats. RESULTS: There was a significant reduction of arthritis scores in CIA mice receiving the daily intraperitoneal injection. After the onset of arthritis in CIA rats, ankle joints receiving a single i.a. injection had significant lower articular indexes with reduced synovial inflammation, pannus formation and erosion on cartilage and bone as well as total histological scores by histopathological analyses. In CIASF or RASF, upon in vitro human EGF stimulation, there was a dose-dependent increase in cell proliferation and Akt activation with suppressed responses under the EGFR TKI treatment. CONCLUSIONS: These findings demonstrate the effect of i.a. injection of an EGFR TKI on amelioration of rheumatoid joint through the suppression of synovial inflammation, pannus formation and erosion on cartilage and bone in experimental arthritis, implicating targeting the i.a. EGFR signalling transduction as a pharmacological strategy.


Subject(s)
Arthritis, Rheumatoid , ErbB Receptors/antagonists & inhibitors , Quinazolines/pharmacology , Synovial Membrane , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Culture Techniques , Dose-Response Relationship, Drug , Drug Monitoring , Fibroblasts/drug effects , Fibroblasts/metabolism , Gefitinib , Humans , Injections, Intra-Articular , Mice , Protein Kinase Inhibitors/pharmacology , Rats , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology , Treatment Outcome
2.
Gene Ther ; 22(4): 305-15, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25588741

ABSTRACT

Most solid tumors undergo hypoxia, leading to rapid cell division, metastasis and expansion of a cell population with hallmarks of cancer stem cells (CSCs). Tumor-selective replication of oncolytic adenoviruses may be hindered by oxygen deprivation in tumors. It is desirable to develop a potent oncolytic adenovirus, retaining its antitumor activity even in a hypoxic environment. We have previously generated an Oct4-dependent oncolytic adenovirus, namely Ad9OC, driven by nine copies of the Oct4 response element (ORE) for specifically killing Oct4-overexpressing bladder tumors. Here, we developed a novel Oct4 and hypoxia dual-regulated oncolytic adenovirus, designated AdLCY, driven by both hypoxia response element (HRE) and ORE. We showed that hypoxia-inducible factor (HIF)-2α and Oct4 were frequently overexpressed in hypoxic bladder cancer cells, and HIF-2α was involved in HRE-dependent and Oct4 transactivation. AdLCY exhibited higher cytolytic activities than Ad9OC against hypoxic bladder cancer cells, while sparing normal cells. AdLCY exerted potent antitumor effects in mice bearing human bladder tumor xenografts and syngeneic bladder tumors. It could target hypoxic CD44- and CD133-positive bladder tumor cells. Therefore, AdLCY may have therapeutic potential for targeting hypoxic bladder tumors and CSCs. As Oct4 is expressed in various cancers, AdLCY may be further explored as a broad-spectrum anticancer agent.


Subject(s)
Antineoplastic Agents/metabolism , Octamer Transcription Factor-3/metabolism , Oncolytic Viruses/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Cell Line, Tumor , Heterografts , Humans , Mice
3.
Cancer Gene Ther ; 19(4): 255-62, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22281757

ABSTRACT

Tc-99m-HL91 is a hypoxia imaging biomarker. The aim of this study was to investigate the value of Tc-99m-HL91 imaging for hypoxia-induced cytosine deaminase (CD)/5-fluorocytosine (5-FC) gene therapy in a murine lung tumor model. C57BL/6 mice were implanted with Lewis lung carcinoma cells transduced with the hypoxia-inducible promoter-driven CD gene (LL2/CD) or luciferase gene (LL2/Luc) serving as the control. When tumor volumes reached 100 mm(3), pretreatment images were acquired after injection of Tc-99m-HL91. The mice were divided into low and high hypoxic groups based on the tumor-to-non-tumor ratio of Tc-99m-HL91. They were injected daily with 5-FC (500 mg kg(-1)) or the vehicle for 1 week. When tumor volumes reached 1000 mm(3), autoradiography and histological examinations were performed. Treatment with 5-FC delayed tumor growth and enhanced the survival of mice bearing high hypoxic LL2/CD tumors. The therapeutic effect of hypoxia-induced CD/5-FC gene therapy was more pronounced in high hypoxic tumors than in low hypoxic tumors. This study provides the first evidence that Tc-99m-HL91 can serve as an imaging biomarker for predicting the treatment responses of hypoxia-regulated CD/5-FC gene therapy in animal tumor models. Our results suggest that hypoxia imaging using Tc-99m-HL91 has the predictive value for the success of hypoxia-directed treatment regimens.


Subject(s)
Antimetabolites/therapeutic use , Carcinoma, Lewis Lung/therapy , Cytosine Deaminase/genetics , Flucytosine/therapeutic use , Organotechnetium Compounds , Oximes , Radiopharmaceuticals , Animals , Antimetabolites/toxicity , Body Weight , Carcinoma, Lewis Lung/diagnostic imaging , Carcinoma, Lewis Lung/genetics , Cell Hypoxia , Cell Line , Cytosine Deaminase/metabolism , Flucytosine/toxicity , Genetic Therapy , Male , Mice , Mice, Inbred C57BL , Radionuclide Imaging , Tumor Burden/genetics
4.
Gene Ther ; 19(7): 752-60, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22089492

ABSTRACT

Knockdown of Toll-like receptors (TLRs) is a novel therapeutic strategy in treating patients with rheumatoid arthritis (RA). We examined the effects of lentiviral vector-mediated delivery of TLR7 short hairpin RNA gene (Lt.shTLR7) on collagen-induced arthritis (CIA). After being immunized on days 0 and 7, Sprague-Dawley rats received intra-articular (i.a.) injection of Lt.shTLR7 or scramble control vector on days 7 and 10. The therapeutic effects were evaluated by measuring ankle circumferences, articular index, and radiographic and histological scores on killing on day 16. Microvessel densities, vascular endothelial growth factor (VEGF) levels, pro-inflammatory cytokine concentrations and T-cell numbers within the synovial tissues were measured. Moreover, VEGF and pro-inflammatory cytokine concentrations in culture supernatants from TLR7-transfected synovial fibroblasts (SFs) stimulated with imiquimod or endogenous ligands were examined. There were significant reduction in ankle circumferences, articular indexes, and radiographic and histological scores. Microvessel densities, VEGF concentrations, interleukin (IL)-1ß and IL-6 levels and T-cell densities within synovial tissues were significantly lower. Induction of VEGF, IL-1ß and IL-6 production from stimulated SFs was significantly suppressed. Taken together, these data demonstrate the effects of i.a. lentiviral vector-mediated delivery of shTLR7 RNA gene on inhibition of CIA, and implicate the manipulation of TLR7 as a potential therapeutic strategy in RA patients.


Subject(s)
Arthritis, Experimental/therapy , RNA, Small Interfering/pharmacology , Toll-Like Receptor 7/genetics , Animals , Ankle Joint/drug effects , Arthritis, Experimental/pathology , Cytokines/biosynthesis , Genetic Vectors , Injections, Intra-Articular , Lentivirus/genetics , Male , Microcirculation/drug effects , Rats , Rats, Sprague-Dawley , Synovial Membrane/metabolism , Vascular Endothelial Growth Factor A/analysis
5.
Gene Ther ; 17(10): 1225-33, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20520649

ABSTRACT

Different members of the galectin family may have inhibitory or stimulatory roles in controlling immune responses and regulating inflammatory reactions in autoimmune diseases such as rheumatoid arthritis (RA). A hypothetical model of a cross talk between galectin-1 and galectin-3 has been established in the circumstance of rheumatoid joints. As galectin-3 is a positive regulator and galectin-1 is a negative regulator of inflammation and autoimmune responses, in this study we evaluated the effects of local knockdown of galectin-3 or overexpression of galectin-1 on ameliorating collagen-induced arthritis (CIA) in rats. Lentiviral vectors encoding galectin-3 small hairpin RNA (shRNA) and galectin-1, as well as two control vectors expressing luciferase shRNA and green fluorescent protein, were individually injected intra-articularly into the ankle joints of rats with CIA, and their treatment responses were monitored by measuring the clinical, radiological and histological changes. Our results show that both knockdown of galectin-3 and overexpression of galectin-1 induced higher percentages of antigen-induced T-cell death in the lymph node cells from arthritic rats. Furthermore, these treatments significantly reduced articular index scores, radiographic scores and histological scores, accompanied with decreased T-cell infiltrates and reduced microvessel density in the ankle joints. Our findings implicate galectin-3 and galectin-1 as potential therapeutic targets for the treatment of RA.


Subject(s)
Arthritis, Experimental/therapy , Galectin 1/genetics , Galectin 3/genetics , Genetic Vectors/administration & dosage , Lentivirus/genetics , RNA, Small Interfering/genetics , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/therapy , Cells, Cultured , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Injections, Intra-Articular , Rats , Transfection
6.
Cancer Gene Ther ; 17(1): 37-48, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19543243

ABSTRACT

The objective of this study was to develop an HER2-targeted, envelope-modified Moloney murine leukemia virus (MoMLV)-based gammaretroviral vector carrying interleukin (IL)-12 gene for bladder cancer therapy. It displayed a chimeric envelope protein containing a single-chain variable fragment (scFv) antibody to the HER2 receptor and carried the mouse IL-12 gene. The fragment of anti-erbB2scFv was constructed into the proline-rich region of the viral envelope of the packaging vector lacking a transmembrane subunit of the carboxyl terminal region of surface subunit. As compared with envelope-unmodified gammaretroviruses, envelope-modified ones had extended viral tropism to human HER2-expressing bladder cancer cell lines, induced apoptosis, and affected cell cycle progression despite lower viral titers. Moreover, animal studies showed that envelope-modified gammaretroviruses carrying IL-12 gene exerted higher antitumor activity in terms of retarding tumor growth and prolonging the survival of tumor-bearing mice than unmodified ones, which were associated with enhanced tumor cell apoptosis as well as increased intratumoral levels of IL-12, interferon-gamma, IL-1beta, and tumor necrosis factor-alpha proteins. Therefore, the antitumor activity of gammaretroviruses carrying the IL-12 gene was enhanced through genetic modification of the envelope targeting HER2 receptor, which may be a promising strategy for bladder cancer therapy.


Subject(s)
Interleukin-12/genetics , Moloney murine leukemia virus/physiology , Oncolytic Virotherapy/methods , Receptor, ErbB-2/genetics , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/virology , Animals , Apoptosis/genetics , Cell Line, Tumor , Genetic Therapy/methods , Genetic Vectors , Humans , Interleukin-12/metabolism , Mice , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/metabolism , Receptor, ErbB-2/metabolism , Transduction, Genetic , Transfection , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/genetics , Viral Tropism
7.
Cancer Gene Ther ; 15(8): 526-34, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18497852

ABSTRACT

Cervical cancer is the second most common type of malignant tumor among women worldwide. When the disease is confined locally, it can be controlled with surgical resection and radiotherapy. However, patients with recurrent or metastatic disease often have a poor prognosis. Measurement of serum levels of squamous cell carcinoma (SCC) antigens has been widely used as serological markers for SCC of uterine cervix. Recently, it has been demonstrated that cervical cancer patients with elevated squamous cell carcinoma antigen-2 (SCCA2) expression in tumor cells carry a poor prognosis. Here, by using a luciferase reporter assay, we show that SCCA2 promoter was active in SCCA2-producing human cervical cancer cell lines, including Cx, Cxwj, SiHa and HeLa cells, but relatively quiescent in normal cervical epithelial cells. We then developed a conditionally replicating adenovirus, designated Ad-KFH, under the transcriptional control of the SCCA2 promoter. This E1B-55 kDa-deleted oncolytic adenovirus replicated specifically in and lysed SCCA2-producing cervical cancer cells. Furthermore, in a peritoneal metastatic tumor model, Ad-KFH retarded Cxwj tumor growth in NOD/severe combined immunodeficient mice and prolonged survival of tumor-bearing mice, especially when combined with cisplatin. These results suggest that Ad-KFH may provide a new strategy of gene therapy for advanced or recurrent uterine cervical cancer.


Subject(s)
Adenoviridae/genetics , Adenovirus E1B Proteins/genetics , Antigens, Neoplasm/genetics , Genetic Therapy , Promoter Regions, Genetic , Serpins/genetics , Uterine Cervical Neoplasms/therapy , Virus Replication , Adenoviridae/physiology , Animals , Base Sequence , Cell Line, Tumor , DNA Primers , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
8.
Cancer Gene Ther ; 14(11): 904-17, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17704754

ABSTRACT

DNA vaccine and dendritic cells (DCs)-based vaccine have emerged as promising strategies for cancer immunotherapy. Fms-like tyrosine kinase 3-ligand (Flt3L) and granulocyte-macrophage-colony-stimulating factor (GM-CSF) have been exploited for the expansion of DC. It was reported previously that combination of plasmid encoding GM-CSF with HER2/neu DNA vaccine induced predominantly CD4(+) T-cell-mediated antitumor immune response. In this study, we investigated the modulation of immune responses by murine Flt3L and GM-CSF, which acted as genetic adjuvants in the forms of bicistronic (pFLAG) and monocistronic (pFL and pGM) plasmids for HER2/neu DNA vaccine (pN-neu). Coexpression of Flt3L and GM-CSF significantly enhanced maturation and antigen-presentation abilities of splenic DC. Increased numbers of infiltrating DC at the immunization site, higher interferon-gamma production, and enhanced cytolytic activities by splenocytes were prominent in mice vaccinated with pN-neu in conjunction with pFLAG. Importantly, a potent CD8(+) T-cell-mediated antitumor immunity against bladder tumors naturally overexpressing HER2/neu was induced in the vaccinated mice. Collectively, our results indicate that murine Flt3L and GM-CSF genes coexpressed by a bicistronic plasmid modulate the class of immune responses and may be superior to those codelivered by two separate monocistronic plasmids as the genetic adjuvants for HER2/neu DNA vaccine.


Subject(s)
Cancer Vaccines/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Membrane Proteins/genetics , Receptor, ErbB-2/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Dendritic Cells/immunology , Genetic Therapy , Genetic Vectors/genetics , Mice , Mice, Inbred Strains , Plasmids/genetics , Spleen/immunology , Tumor Burden , Urinary Bladder Neoplasms/pathology , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/therapeutic use
9.
Int J Gynecol Cancer ; 17(1): 174-81, 2007.
Article in English | MEDLINE | ID: mdl-17291250

ABSTRACT

Squamous cell carcinoma antigen (SCCA) is a tumor marker for patients with squamous cell carcinoma of uterine cervix, lung, and esophagus. It was encoded by two highly homologous genes, SCCA1 and SCCA2. However, the relevance of SCCA genes to squamous cell carcinogenesis and patient outcome remains far from clear. In this study, by using laser microdissection and real-time quantitative polymerase chain reaction procedures, the messenger RNA (mRNA) expression of the SCCA1 and SCCA2 genes in normal, dysplastic, and malignant squamous epithelia from uterine cervical tissues were analyzed and correlated with outcome of cancer patients. We found that the SCCA2/A1 mRNA ratios were progressively increased from normal, dysplastic, to cancer cells, and the mean ratio was significantly higher in cancer tissues than that in normal epithelium (P= 0.02). The SCCA2/A1 mRNA ratios were not significantly associated with types of human papillomavirus infection (P > 0.05). High SCCA2/SCCA1 mRNA ratios (ratio >1) were an independent predictor of disease recurrence (relative risk: 3.58; P= 0.003). Of the 38 patients with cervical cancer, 12 patients with high SCCA2/SCCA1 mRNA ratios had a significant lower 2-year disease-free survival of only 50%, while it was 92% in those with low SCCA2/SCCA1 mRNA ratios (P < 0.001). In conclusion, our study indicated that the ratios of SCCA2 to SCCA1 RNA were increased during the process of cervical carcinogenesis, and patients with elevated SCCA2/A1 ratio carried a higher risk for recurrence in early-stage uterine cervical cancer.


Subject(s)
Antigens, Neoplasm/biosynthesis , Biomarkers, Tumor/biosynthesis , Serpins/biosynthesis , Uterine Cervical Neoplasms/immunology , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Female , Follow-Up Studies , Humans , Middle Aged , Prognosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serpins/genetics , Uterine Cervical Neoplasms/genetics
10.
Br J Cancer ; 89(2): 344-9, 2003 Jul 21.
Article in English | MEDLINE | ID: mdl-12865928

ABSTRACT

Constitutively activated signal transducers and activators of transcription (STAT) factors, in particular STAT1, STAT3 and STAT5, have been demonstrated in a variety of human tumours and cancer cell lines. However, data on the expression of these STATs in nasopharyngeal carcinoma (NPC) are limited. In this study, the expression patterns of STAT1, STAT3 and STAT5 were immunohistochemically examined on the archival specimens from 61 patients with NPC. Staining results of each STATs were then correlated with the clinical parameters and prognosis of these patients. The results showed that constitutive activation of STAT3 and STAT5 was detected in the majority, 70.5 and 62.3%, respectively, of the 61 tumour specimens. Furthermore, coexpression of activated STAT3 and STAT5 was found in 54.1% of the specimens. In contrast, constitutive activated STAT1 could only be detected in 8 (13.1%) cases. Surprisingly, following radiotherapy, patients with constitutive STAT5 activation, or activation of both STAT3 and STAT5, had better disease-free survival and overall survival than those without activated STAT5. To our knowledge, this is the first report providing the overall expression patterns and prognostic significance of specific STATs in NPC.


Subject(s)
Carcinoma/genetics , Carcinoma/pathology , DNA-Binding Proteins/biosynthesis , Milk Proteins , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Trans-Activators/biosynthesis , Acute-Phase Proteins , Adult , Biomarkers, Tumor/analysis , Biopsy , Disease-Free Survival , Female , Herpesvirus 4, Human , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , STAT3 Transcription Factor , STAT5 Transcription Factor , Signal Transduction , Survival Analysis
11.
Br J Cancer ; 88(9): 1492-9, 2003 May 06.
Article in English | MEDLINE | ID: mdl-12778082

ABSTRACT

Mutations or loss of heterozygosity of p53 are detected in approximately 50% of bladder cancers. E1B-55 kD-deleted adenovirus has been shown to kill tumour cells with defective p53 function while sparing normal cells. Here, we examined the cytolytic effect and replication of E1B-55 kD-deleted adenovirus, designated Ad5WS1, on human bladder cancer cell lines with various p53 status. Ad5WS1 caused more severe cytolytic effect and replicated more efficiently in J82 and TCC-SUP bladder cancer cells carrying mutant p53 compared with TSGH-8301 and BFTC-905 bladder cancer cells retaining wild-type p53. Introduction of dominant negative p53 into BFTC-905 cells rendered them more susceptible to Ad5WS1-induced cytolysis. Furthermore, cells susceptible to lysis caused by Ad5WS1 were not attributable to their greater infectability by adenovirus. Finally, Ad5WS1 suppressed the growth of TCC-SUP bladder tumour xenografts, which could be augmented when combined with replication-defective adenoviral vector encoding kringles 1-5 of plasminogen (K1-5), an angiogenic inhibitor. Taken together, our results show that E1B-55 kD-deleted adenovirus replicates and hence lyses bladder cancer cells with mutant p53 much more efficient than those with wild-type p53. Thus, E1B-deleted adenovirus may have therapeutic potential, especially in combination with adenoviral vector expressing K1-5, for the treatment of bladder cancer.


Subject(s)
Adenoviridae/genetics , Adenovirus E1B Proteins/genetics , Genetic Therapy/methods , Urinary Bladder Neoplasms/therapy , Virus Replication/physiology , Adenoviridae/physiology , Cell Survival , Gene Deletion , Humans , Recombinant Proteins , Transfection , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/virology
12.
Vaccine ; 19(28-29): 3947-56, 2001 Jul 16.
Article in English | MEDLINE | ID: mdl-11427270

ABSTRACT

Previously, we showed that vaccination with the glycoprotein D (gD) gene of pseudorabies virus (PrV) delivered by Escherichia coli induced protective immune responses. In this study, we report that oral DNA vaccination with attenuated Salmonella choleraesuis carrying the PrV gD gene conferred protective immunity in mice against PrV. Moreover, co-delivery of the prothymosin alpha gene carried by S. choleraesuis enhanced the vaccine efficacy. Our results thus demonstrate for the first time, to our knowledge, the effectiveness of oral DNA vaccination using S. choleraesuis as a delivery vehicle and the potential usefulness of prothymosin alpha as a DNA vaccine adjuvant.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Protein Precursors/administration & dosage , Pseudorabies Vaccines/administration & dosage , Salmonella/genetics , Thymosin/administration & dosage , Vaccines, DNA/administration & dosage , Adjuvants, Immunologic/genetics , Administration, Oral , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Viral/biosynthesis , Female , Genetic Vectors , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/immunology , In Vitro Techniques , Lac Operon , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Plasmids/genetics , Promoter Regions, Genetic , Protein Precursors/genetics , Pseudorabies/immunology , Pseudorabies/prevention & control , Pseudorabies Vaccines/genetics , Salmonella/immunology , T-Lymphocytes, Cytotoxic/immunology , Thymosin/analogs & derivatives , Thymosin/genetics , Vaccines, DNA/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
13.
Vaccine ; 19(23-24): 3277-84, 2001 Apr 30.
Article in English | MEDLINE | ID: mdl-11312026

ABSTRACT

Attenuated intracellular bacteria, such as Salmonella and Shigella, have been exploited to act as gene delivery vectors. In this study, we report that nonpathogenic, live Escherichia coli can be used for the delivery of DNA vaccines in vivo, leading to generation of immune responses against plasmid-encoded foreign antigens. The pseudorabies virus (PrV) DNA vaccine carrying the glycoprotein D (gD) gene delivered by E. coli was able to induce protective immune responses in mice against a lethal PrV challenge. Co-delivery of E. coli carrying plasmid DNA encoding prothymosin alpha enhanced cellular immune responses to the PrV DNA vaccine delivered by E. coli. Our results suggest that nonpathogenic E. coli may be used as a vector for DNA vaccines in veterinary uses.


Subject(s)
Herpesvirus 1, Suid/genetics , Pseudorabies/prevention & control , Viral Envelope Proteins/genetics , 3T3 Cells , Adjuvants, Immunologic/administration & dosage , Animals , Escherichia coli/genetics , Escherichia coli/pathogenicity , Female , Genes, Viral , Genetic Vectors , Herpesvirus 1, Suid/immunology , Herpesvirus 1, Suid/pathogenicity , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Protein Precursors/genetics , Protein Precursors/immunology , Pseudorabies/immunology , Pseudorabies Vaccines/administration & dosage , Pseudorabies Vaccines/genetics , Pseudorabies Vaccines/immunology , Swine , Swine Diseases/immunology , Swine Diseases/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Thymosin/analogs & derivatives , Thymosin/genetics , Thymosin/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Viral Envelope Proteins/immunology
14.
Cancer Gene Ther ; 8(1): 73-81, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11219496

ABSTRACT

The murine MBT-2 bladder tumor model in syngeneic C3H/HeN mice was used to investigate the feasibility of gene therapy based on the delivery of interferon-gamma (IFN-gamma) in vivo by retroviral vectors. We constructed a recombinant retroviral vector pRUFneo/IFN-gamma, which was transfected into a retroviral packaging cell line psiCRE, to produce psiCRE/pRUFneo/IFN-gamma cells. The expressions of the neo and IFN-gamma genes were verified by reverse transcription-polymerase chain reaction and IFN-gamma was detected in the culture supernatant from psiCRE/pRUFneo/IFN-gamma cells. After receiving MBT-2 cells admixed with retroviral pRUFneoIFN-gamma supernatant, C3H/HeN mice exhibited lower tumor incidence, lower tumor mass, and higher survival rate, as well as higher antitumor responses compared to those injected with MBT-2 cells admixed with control retroviral supernatant. Moreover, the retroviral pRUFneoIFN-gamma supernatant was able to suppress the growth of rechallenged tumors in postoperated mice. Although the IFN-gamma protein secreted from psiCRE/pRUFneo/IFN-gamma cells partly contributes to the antitumor effect of retroviral pRUFneoIFN-gamma supernatant, the retroviruses carrying the IFN-gamma gene transduced MBT-2 cells in vivo, which may result in enhancing local IFN-gamma production from tumor cells. Because bladder is suitable for the intravesical instillation of therapeutic agents, in vivo administration of retroviral vectors encoding IFN-gamma may be explored for the treatment of bladder cancer.


Subject(s)
Genetic Therapy , Interferon-gamma/therapeutic use , Lymphoma, T-Cell/therapy , Mastocytosis/therapy , Retroviridae/genetics , Urinary Bladder Neoplasms/therapy , Animals , Cell Division/drug effects , Chromium/analysis , Chromium/metabolism , Cytotoxicity, Immunologic/immunology , DNA Primers/chemistry , Female , Gene Transfer Techniques , Genetic Vectors , Humans , In Vitro Techniques , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-2/metabolism , Kanamycin Kinase/genetics , Kanamycin Kinase/metabolism , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/virology , Mice , Mice, Inbred C3H , Neoplasm Transplantation , Postoperative Care , RNA, Viral/analysis , Recombinant Proteins/metabolism , Retroviridae/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Transfection , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/virology
15.
Hepatology ; 33(1): 277-86, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11124846

ABSTRACT

Several types of naturally occurring pre-S mutants in sera or liver tissues in patients with chronic hepatitis B virus (HBV) infection have been identified. To clarify the prevalence and significance of emergence of pre-S mutants, 140 sera and 18 resected livers from patients with HBV were studied. Replicative status was designated as high, intermediate, and low based on the HBV-DNA levels in serum or the expression of HBV antigens in liver. In vitro transfection and Western blot analysis were performed to characterize expression and secretion of HBsAg by the mutant constructs. Five major types (I to V) of pre-S deletion mutants in serum and liver and 2 types (VI and VII) in liver were identified. Pre-S mutant was 6.4% at high replicative phase, 13% at intermediate, and 37.5% at low or nonreplicative phases in serum. In livers, the same tendency existed: pre-S2 deletion mutants emerged and prevailed at a low replicative phase in hepatocytes that expressed a novel marginal pattern of HBsAg and usually clustered in groups. The deletion sequence of pre-S2 region coincides with human leukocyte antigen-restricted T- and B-cell epitopes. In vitro HBsAg was retained in the hepatocytes and synthesis and secretion of major surface antigen decreased for most of the pre-S mutants. Pre-S mutants prevailed with evolution of chronic HBV, probably under immune pressure. Emergence of pre-S mutants may account for the life-long persistence and discrepancy of HBsAg in serum and liver in HBV and may confer growth advantage in view of the clustering proliferation of hepatocytes harboring pre-S2 mutant.


Subject(s)
Hepatitis B virus/growth & development , Hepatitis B virus/genetics , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/virology , Liver/virology , Mutation/physiology , Viral Proteins/genetics , Blotting, Western , Cell Line , Cloning, Molecular , Densitometry , Extracellular Space/metabolism , Gene Deletion , Hepatitis B Surface Antigens/blood , Hepatitis B Surface Antigens/metabolism , Hepatitis B, Chronic/metabolism , Humans , Liver/metabolism , Membrane Proteins/metabolism , Viral Proteins/blood , Viral Proteins/metabolism , Virus Replication
16.
Gene Ther ; 8(21): 1609-17, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11894999

ABSTRACT

To explore the potential use of prothymosin alpha(ProT), a putative thymic hormone, in gene therapy for bladder cancer, we generated a replication-defective recombinant retroviral vector encoding ProT and tested its antitumor effect on the MBT-2 murine bladder cancer. C3H/HeN mice injected with MBT-2 cells in conjunction with retroviruses encoding ProT exhibited smaller tumor mass, lower tumor incidence and higher survival rate, as well as higher antitumor cytotoxic activities compared with those injected with control viruses. However, such effects were not observed in severe combined immunodeficiency mice, suggesting that ProT exerts antitumor effects through its immunomodulatory activities. Cell growth in monolayer culture and colony formation in soft agar were enhanced in ProT gene-modified MBT-2 clones, and such growth-promoting activities of ProT could be reversed if its nuclear localization signal (NLS) was deleted. To circumvent the proliferation-promoting effect of ProT on tumor cells, a retroviral vector encoding ProT lacking NLS was constructed. Our results showed that retroviruses encoding NLS-deleted ProT was more efficacious than those encoding wild-type ProT in prolonging survival of tumor-bearing mice. This is the first report indicating that ProT, in particular NLS-deleted ProT, delivered by retroviral vectors may be further explored for the treatment of bladder cancer.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Protein Precursors/genetics , Retroviridae/genetics , Thymosin/analogs & derivatives , Thymosin/genetics , Urinary Bladder Neoplasms/therapy , Animals , Cell Division , Female , Mice , Mice, Inbred Strains , Mice, SCID , Neoplasm Transplantation , Urinary Bladder Neoplasms/metabolism
17.
Anticancer Res ; 20(3A): 1557-62, 2000.
Article in English | MEDLINE | ID: mdl-10928070

ABSTRACT

The major purpose of this study was to define if the immunosuppressive effect of a transforming growth factor-beta (TGF-beta)-producing autologous tumor vaccine can be abrogated and rendered immunogenic by suppressing its TGF-beta secretion with antisense strategy. In this study, using a TGF-beta antisense gene modified MBT-2 tumor cell line [MBT-2/TGF-beta(-)#3] which we established by ourselves, we first demonstrated that the amounts of TGF-beta produced by irradiated (IR) and non-irradiated MBT-2/TGF-beta(-) #3 were both significantly decreased when detected after in vitro culture for 48 hours. The result of flow cytometry analysis reveals that decreased production of TGF-beta led to the increased expressions of MHC class I molecule and Fas on the surface of MBT-2 tumor cells. This finding may in part explain why the splenocytes obtained from day 17 tumor bearing mice (D17TBM) immunized with IRMBT-2/TGF-beta(-)#3 on day 26 expressed a higher in vitro cytotoxic activity against MBT-2 tumor cells and hence ensured a better survival of D17TBM when they were rechallenged with a two-fold higher amount of wild-type MBT-2 tumor cells, 48 hours after surgical removal of the primary tumor. Our result implies that decreasing the amount of TGF-beta secreted from the autologous tumor vaccine by antisense strategy may significantly improve its immunogenicity through up-regulation of both MHC class I and Fas expressions. Therefore, this could provide an alternative approach for future active immunotherapy.


Subject(s)
Cancer Vaccines , Histocompatibility Antigens Class I/biosynthesis , Neoplasms, Experimental/drug therapy , Oligonucleotides, Antisense/therapeutic use , Transforming Growth Factor beta/metabolism , fas Receptor/biosynthesis , Animals , Female , Immunization , Mice , Mice, Inbred C3H , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Spleen/immunology , Spleen/pathology , Survival Analysis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Tumor Cells, Cultured , Up-Regulation
18.
Proc Natl Sci Counc Repub China B ; 24(2): 56-62, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10809081

ABSTRACT

This study, using the MBT-2 murine bladder tumor model, mainly investigated the role of interleukin-12 (IL-12) in the specific antitumor immune response of a tumor-bearing host when systemically administrated after surgery. Day 17 tumor-bearing mice (D17TBM) along with non-tumor bearing naive mice were treated with daily intraperitoneal (i.p.) injection of IL-12 (0.25 microg/mouse) from day 18 to day 24 for a total of 7 doses. Their splenocytes were obtained on Day 31 for natural killer cells (NK), lymphokine activated killer cells (LAK) and cytotoxic T lymphocyte (CTL) activity assay and lymphocyte subsets phenotypic analysis. The tumor suppression effect of systemic IL-12 administration was evaluated based on the tumor outgrowth of the higher number of tumor cells rechallenged 24 hours after resectioning of the primary tumor. After evaluation on Day 31, the result of in vitro cytotoxicity assay revealed that systemic administration of IL-12 mainly enhanced the splenic LAK and CTL activities in non-tumor-primed naive mice, and the NK activity in tumor-primed D17TBM, respectively. However, in vivo administration of IL-12 with or without IL-2 failed to upgrade the proportions of either CD4+ CD44+ or CD8+ CD44+ T cells subsets in the spleens and regional inguinal lymph nodes (LNs) of both the D17TBM and naive mice. However, the splenic CD8+ CD44+ T-cell subset in the IL-12-treated D17TBM increased prominently after further culturing in the presence of IL-2 400 units/ml plus IL-12 25 ng/ml for 4 days. The fact that systemic administration of IL-12 significantly suppressed the outgrowth of Day-18 challenged tumor cells, especially in D17TBM, clearly indicates that the established specific antitumor immunity in tumor-primed D17TBM was efficiently augmented. From the results of this study, we conclude that, after surgical resection of a primary tumor, systemic administration of IL-12 can be an effective adjuvant therapy because it demonstrates a significant augmentation effect on the tumor-specific immune response in the tumor-primed host.


Subject(s)
Interleukin-12/administration & dosage , Postoperative Care , Urinary Bladder Neoplasms/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Female , Hyaluronan Receptors/analysis , Immunotherapy , Injections, Intraperitoneal , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Mice , Mice, Inbred C3H , Spleen/immunology , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology
19.
Anal Biochem ; 275(2): 260-2, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10552914
20.
Biol Chem ; 380(3): 277-83, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10223329

ABSTRACT

The core antigen of hepatitis B virus (HBcAg) made in Escherichia coli yields particles that closely resemble the viral nucleocapsid. Extensive modifications can be made to the primary structure of HBcAg without impairing particle assembly. This enables other peptide sequences, including very long sequences, to be added, substituted, or inserted into the nucleocapsid subunit while retaining the ability to form highly immunogenic particles. These also retain the T cell epitopes of HBcAg and constitute powerful delivery systems for a diverse range of immunogenic epitopes and have significant potential for development of multicomponent vaccines.


Subject(s)
Hepatitis B Core Antigens/immunology , Hepatitis B Vaccines/immunology , Peptides/immunology , Vaccines, Synthetic/immunology , Animals , Humans , Recombinant Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...