Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Zhonghua Nei Ke Za Zhi ; 60(9): 784-790, 2021 Sep 01.
Article in Chinese | MEDLINE | ID: mdl-34445813

ABSTRACT

Lupus nephritis (LN) refers to renal involvement in systemic lupus erythematosus and is characterized by hematuria, proteinuria, edema, hypertension and renal insufficiency. The complete remission rate of proliferative LN remains low using the current induction protocols and LN tends to flare. Scientific and standardized diagnosis and therapy are crucial for the treatment of LN. Therefore, based on the current international and domestic experiences and guidelines, the Chinese Rheumatology Association developed the recommendations of diagnosis and therapy for LN, with the purpose of enhancing efficacy, reducing flare, halting renal progression and improving outcome of LN.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , China , Humans , Kidney , Lupus Nephritis/diagnosis , Lupus Nephritis/therapy , Remission Induction
2.
Biochem Pharmacol ; 91(1): 18-30, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24976507

ABSTRACT

BACKGROUND: Resistance of cancer cells towards chemotherapeutics represents a major cause of therapy failure. The objective of our study was to evaluate cellular defense strategies in response to the novel vacuolar H(+)-ATPase inhibitor, archazolid B. EXPERIMENTAL APPROACH: The effects of archazolid B on T24 bladder carcinoma cells were investigated by combining "omics" technologies (transcriptomics (mRNA and miRNA) and proteomics). Free cholesterol distribution was determined by filipin staining using flow cytometry and fluorescence microscopy. Flow cytometry was performed for LDLR surface expression studies. Uptake of LDL cholesterol was visualized by confocal microscopy. SREBP activation was determined performing Western Blotting. The efficiency of archazolid B/fluvastatin combination was tested by cytotoxicity assays. RESULTS: Archazolid B led to accumulation of free cholesterol within intracellular compartments and drastic disturbances in cholesterol homeostasis resulting in activation of SREBP-2 (sterol regulatory element-binding protein 2) and up-regulation of target genes including HMGCR (HMG-CoA reductase), the key enzyme of cholesterol biosynthesis. LDLR surface expression was reduced and LDL uptake was completely inhibited after 24h, indicating newly synthesized cholesterol to be the main source of cholesterol in archazolid B treated cells. By combining archazolid B with the HMGCR inhibitor fluvastatin, cholesterol was reduced and cell viability decreased by about 20% compared to archazolid B treatment alone. CONCLUSIONS: Our study revealed cholesterol biosynthesis as an important resistance mechanism in T24 cells after archazolid B treatment. The combination of archazolid B with statins may be an attractive strategy to potentiate archazolid B induced cell killing by affecting cholesterol biosynthesis.


Subject(s)
Cholesterol/biosynthesis , Drug Resistance, Neoplasm/drug effects , Macrolides/pharmacology , Thiazoles/pharmacology , Urinary Bladder Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Endocytosis/drug effects , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Lipoproteins, LDL/metabolism , MicroRNAs , Real-Time Polymerase Chain Reaction , Receptors, LDL/metabolism , Reproducibility of Results , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
3.
Gene Ther ; 21(5): 457-67, 2014 May.
Article in English | MEDLINE | ID: mdl-24572790

ABSTRACT

Dendritic cell (DC)-based vaccine approaches are being actively evaluated for developing immunotherapeutic agents against cancers. In this study, we investigated the use of engineered DCs expressing transgenic tumor-associated antigen hgp100 and the regulatory cytokine interleukin-21, namely DC-hgp100/mIL-21, as a therapeutic vaccine against melanoma. Tumor-bearing mice were injected intratumorally with transgenic DCs followed by three booster injections. Transgenic DC-hgp100/mIL-21 showed significant reduction in primary tumor growth and metastasis compared with DC-hgp100 alone and DC-mIL-21 alone. In vivo depletion of specific immune cell types (CD8(+) T, CD4(+) T and Natural killer (NK)-1.1(+) cells) effectively blocked the protective effect of this combinational vaccine. In adoptive transfer experiments, a survival rate of nearly 90% was observed at 60 days post-tumor inoculation for the combinational vaccine group. In contrast, all mice in the DC-hgp100 and DC-mIL-21-only groups died within 43-46 days after tumor challenge. Considerably increased levels of interferon (IFN)-γ, tumor necrosis factor (TNF)-α, granulocyte macrophage colony-stimulating factor (GM-CSF) and cytotoxic T lymphocytes (CTLs) were detected with the combination vaccine group compared with other individual treatment groups. In comparison with the DC-hgp100 or mIL-21 groups, the combinational DC-hgp100/mIL-21 vaccine also drastically suppressed the myeloid-derived suppressor cells (MDSCs) and T-regulatory (Treg) cell populations. Our findings suggest that a combinational DC- and gene-based hgp100 and mIL-21 vaccine therapy strategy warrants further evaluation as a clinically relevant cancer vaccine approach for human melanoma patients.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/transplantation , Interleukins/immunology , Melanoma, Experimental/immunology , Adoptive Transfer , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Cell Proliferation , Cell- and Tissue-Based Therapy , Dendritic Cells/cytology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , Interleukins/genetics , Killer Cells, Natural/immunology , Lymphocyte Depletion , Melanoma, Experimental/mortality , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis/immunology , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Vaccines, Synthetic/immunology
4.
Gene Ther ; 16(11): 1329-39, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19626052

ABSTRACT

The induction of strong cell-mediated immunity against targeted cancer cells is difficult, and often requires specific vaccination schema and the appropriate adjuvants to be effective. The chemokine RANTES has been studied as a vaccine adjuvant in cancer therapy, but specific applications remain to be determined. For gene-based vaccination against B16 melanoma in C57BL/6JNarl mice, initial priming with mouse RANTES cDNA followed 24 h later by human gp100 DNA vaccination, and later boosting with a viral vector expressing mRANTES and hgp100 strongly suppressed B16/hgp100 primary tumors and lung metastasis. The inclusion of mRANTES in this vaccination regimen gave significantly better suppression of tumor growth, substantially enhanced mouse survival, and led to greater cytotoxic activity of splenocytes against B16/hgp100 cells than vaccination against hgp100 alone. B16/hgp100 melanoma cells were resistant to the ligands TRAIL and FasL in vitro but sensitized to them in vivo owing to the priming effect of cytokines in response to vaccination. Our data demonstrate that co-vaccination with chemokine (mRANTES) and tumor-specific (hgp100) genes in a specific time sequence is more effective at suppressing tumor growth and metastasis than hgp100 alone, and this effect may be mediated by sensitization of tumor cells to death ligands.


Subject(s)
Cancer Vaccines , Chemokine CCL5/metabolism , Genetic Therapy/methods , Melanoma, Experimental/prevention & control , Membrane Glycoproteins/metabolism , Adjuvants, Immunologic/genetics , Animals , Cancer Vaccines/immunology , Chemokine CCL5/genetics , Cytotoxicity, Immunologic , DNA, Complementary/genetics , Female , Gene Expression , Humans , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Skin/immunology , Transgenes , Tumor Cells, Cultured , Vaccines, DNA/immunology , gp100 Melanoma Antigen
5.
Gene Ther ; 11(1): 100-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14681703

ABSTRACT

The augmentation and optimization of specific targeted transgene expression systems are important strategies for clinical research into gene therapy and DNA vaccination, due to safety considerations. In this study, we introduced 3' untranslated regions and transcriptional control modifications and direct tandem or combinational vector design strategies into a number of specific cytokine cDNA expression plasmids. The experiments were performed in parallel using both in vivo and in vitro transgene expression systems. In vivo studies were carried out using gene gun delivery of test vectors into mouse skin tissues. A combination of specific cell lines and fresh cell explants were used for in vitro and ex vivo transgene expression assay systems. The results from these comparative experiments demonstrated that a number of molecular biology manipulations can be readily adapted to define and significantly enhance the level or/and duration of transgene expression for a group of clinically relevant cytokine genes, with very similar effects for both in vivo and in vitro test systems. This cytokine transgene expression system may offer a favorable means for improving the efficiency of cytokine gene therapy and DNA vaccines in future clinical studies.


Subject(s)
Cytokines/genetics , Genetic Therapy/methods , Immunotherapy, Active/methods , Neoplasms/therapy , Promoter Regions, Genetic , 3' Untranslated Regions , Animals , Biolistics , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Carcinoma/immunology , Carcinoma/therapy , Cell Line, Tumor , Cells, Cultured , Cytokines/analysis , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interferon-gamma/genetics , Interleukin-2/genetics , Lymphocytes/immunology , Lymphoma, T-Cell/immunology , Melanoma/immunology , Melanoma/therapy , Mice , Neoplasms/immunology , Skin/immunology , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Transgenes
6.
Plant Cell Rep ; 21(10): 993-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12835910

ABSTRACT

The present protocol was aimed at establishing a routine transformation procedure via Agrobacterium tumefaciens for an important Oncidium orchid cultivar. An expression vector containing hptII and gusA genes driven by the cauliflower mosaic virus (CaMV) 35S promoter was successfully introduced into the Oncidium orchid genome by A. tumefaciens. Protocorm-like bodies (PLBs) derived from protocorms, were the target explants for transformation. The transformation was performed through two stages of cocultivation, the first stage occurring on antibiotic-free medium for 3 days, and the subsequent stage on medium containing 100 mg/l timentin for 1 month. Among 1,000 inoculated PLBs, 108 putatively transformed PLBs were proliferated on 5 mg/l hygromycin selection medium. A total of 28 independent transgenic orchid plants were obtained, from which six transgenic lines that were positive for beta-glucuronidase were randomly selected and confirmed by Southern, northern and western blot analyses. These results indicated that the foreign DNA was successfully integrated into the orchid genome and expressed transcriptionally and translationally in these orchid plants. The present transformation method reported is suitable for improving the Oncidium orchid through genetic engineering.


Subject(s)
Agrobacterium tumefaciens/genetics , Cinnamates , Hygromycin B/analogs & derivatives , Orchidaceae/genetics , Orchidaceae/metabolism , Transformation, Genetic , Agrobacterium tumefaciens/physiology , Dose-Response Relationship, Drug , Gene Expression , Hygromycin B/pharmacology , Orchidaceae/drug effects , Plants, Genetically Modified , Selection, Genetic
7.
Hum Gene Ther ; 13(14): 1711-21, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12396624

ABSTRACT

The primary objective of this phase I study was to determine the safety of an autologous tumor vaccine given by intradermal injection of lethally irradiated granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected autologous melanoma and sarcoma cells. Secondary objectives included validation of the gene delivery technology (particle-mediated gene transfer), determining the host immune response to the tumor after vaccination, and monitoring patients for evidence of antitumor response. Sixteen patients were treated with either of two different doses of GM-CSF-treated tumor cells. One patient received treatment with both doses of tumor cells. No treatment-related local or systemic toxicity was noted in any patient. Patients administered 100% treated cells (i.e., with a preparation of tumor cells that had all been exposed to GM-CSF DNA transfection) had a more extensive lymphocytic infiltrate at the vaccine site than did patients given 10% treated cells (a preparation of tumor cells in which 10% had been exposed to GM-CSF transfection) or nontreated tumor. The generation of a systemic immune response to autologous tumor by a delayed-type hypersensitivity response to the intradermal placement of nontransfected tumor cells was noted in one patient. One patient had a transient partial response of metastatic tumor sites. The entire procedure, from tumor removal to vaccine placement, was accomplished in less than 6 hr in all patients. Four of 17 patient tumor preparations produced greater than 3.0 ng of GM-CSF per 10(6) cells per 24 hr in vitro. The one patient with greater than 30 ng of GM-CSF per 10(6) cells per 24 hr in vitro had positive DTH, a significant histologic inflammatory response, and clinically stable disease. This technique of gene transfer was safe and feasible, but resulted in clinically relevant levels of gene expression in only a minority of patients.


Subject(s)
Biolistics , Cancer Vaccines/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunologic Factors/genetics , Melanoma/therapy , Sarcoma/therapy , Soft Tissue Neoplasms/therapy , Vaccines, DNA/therapeutic use , Cancer Vaccines/administration & dosage , DNA, Complementary/genetics , Dose-Response Relationship, Immunologic , Feasibility Studies , Gold , Humans , Hypersensitivity, Delayed/immunology , Injections, Intradermal , Leiomyosarcoma/pathology , Leiomyosarcoma/therapy , Liposarcoma/pathology , Liposarcoma/therapy , Melanoma/pathology , Melanoma/secondary , Safety , Sarcoma/pathology , Soft Tissue Neoplasms/pathology , Treatment Outcome , Tumor Cells, Cultured/transplantation , Vaccines, DNA/administration & dosage
8.
J Agric Food Chem ; 49(7): 3420-4, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11453785

ABSTRACT

The antioxidant activity of extracts from bark and heartwood of Acacia confusa was evaluated by various antioxidant assays, including free radical and superoxide radical scavenging assays and lipid peroxidation assay as well as hydroxyl radical-induced DNA strand scission assay. In addition, an ex vivo antioxidant assay using a flow cytometric technique was also employed in this study. The results indicate that both bark and heartwood extracts clearly have strong antioxidant effects. Similar inhibitory activities for each test sample were found for both 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical generation and lipid peroxidation. As for the superoxide radical scavenging activity, the heartwood extract was more effective than the bark extract. Furthermore, the heartwood extract protected PhiX174 supercoiled DNA against strand scission induced by ultraviolet photolysis of H2O2, and it reduced the amounts of intracellular hydrogen peroxide, a reactive oxygen species, when it was co-incubated with human promyelocytic leukemia (HL-60) cells under oxidative stress.


Subject(s)
Acacia/chemistry , Antioxidants/pharmacology , Lipid Peroxidation/drug effects , Plant Extracts/pharmacology , Antioxidants/chemistry , Flow Cytometry , Free Radical Scavengers , Plant Extracts/chemistry , Reactive Oxygen Species
9.
Clin Cancer Res ; 7(4): 952-61, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309346

ABSTRACT

Particle-mediated gene delivery was used to immunize mice against melanoma. Mice were immunized with a plasmid cDNA coding for the human melanoma-associated antigen, gp100. Murine B16 melanoma, stably transfected with human gp100 expression plasmid, was used as a tumor model. Particle-mediated delivery of gp100 plasmid into the skin of naïve mice resulted in significant protection from a subsequent tumor challenge. Co-delivery of murine granulocyte-macrophage colony-stimulating factor (GM-CSF) expression plasmid together with the gp100 plasmid consistently resulted in a greater level of protection from tumor challenge. The inclusion of the GM-CSF plasmid with the gp100 DNA vaccine allowed a reduction in the gp100 plasmid dose required for antitumor efficacy. Protection from tumor challenge was achieved with as little as 62.5 ng of gp100 DNA per vaccination. Tumor protection induced by the gp100 + GM-CSF gene combination was T cell mediated, because it was abrogated in vaccinated mice treated with anti-CD4 and anti-CD8 monoclonal antibodies. In addition, administration of the gp100 + GM-CSF DNA vaccine to mice bearing established 7-day tumors resulted in significant suppression of tumor growth. These results indicate that inclusion of GM-CSF DNA augments the efficacy of particle-mediated vaccination with gp100 DNA, and this form of combined gp100 + GM-CSF DNA vaccine warrants clinical evaluation in melanoma patients.


Subject(s)
DNA/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Melanoma, Experimental/prevention & control , Membrane Glycoproteins/therapeutic use , Neoplasm Proteins/therapeutic use , Animals , DNA/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Melanoma, Experimental/drug therapy , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Neoplasm Proteins/genetics , Plasmids/genetics , Recurrence , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Vaccination , gp100 Melanoma Antigen
10.
Curr Protoc Hum Genet ; Chapter 12: Unit 12.6, 2001 May.
Article in English | MEDLINE | ID: mdl-18428251

ABSTRACT

Particle-mediated or "gene gun" technology has been developed as a nonviral method for gene transfer into various mammalian tissues. Gene delivery is achieved by physical force: a strong shock wave is generated that accelerates DNA-coated gold particles to high speeds, providing them with the momentum needed to penetrate the targeted cells. This unit describes general procedures for in vivo and in vitro DNA and RNA transfections by particle-mediated delivery. The Basic Protocol and an alternate protocol address in vivo delivery to mouse skin. In vitro delivery to cryopreserved and adherent cells is also described.


Subject(s)
Biolistics/methods , Animals , Cell Adhesion , Cells, Cultured , DNA, Recombinant/administration & dosage , DNA, Recombinant/genetics , Genetics, Medical , Humans , In Vitro Techniques , Mice , RNA/administration & dosage , RNA/genetics , Skin , Transfection
11.
Mol Pharmacol ; 58(5): 1057-66, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11040054

ABSTRACT

Effects of piperlactam S (C(17)H(13)NO(4); mol. wt. 295) isolated from Piper kadsura on phytohemagglutinin (PHA) stimulated cell proliferation were studied in primary culture of human T cells. The results showed that piperlactam S suppressed T cell proliferation at about 0 to 12 h after stimulation with PHA. Synthesis of total cellular proteins and RNA in activated cell cultures was also suppressed. The inhibitory action of piperlactam S was not through direct cytotoxicity. Cell cycle analysis indicated that piperlactam S arrested the cell cycle progression of activated T cells from the G(1) transition to the S phase. In an attempt to further localize the point in the cell cycle at which arrest occurred, a set of key regulatory events leading to the G(1)/S boundary, including gene expression of cytokines and c-Fos protein synthesis, was examined. Piperlactam S suppressed, in activated T lymphocytes, the production and mRNA expression of cytokines such as interleukin-2 (IL-2), IL-4, and interferon-gamma in a dose-dependent manner. In addition, Western blot analysis indicated that c-Fos protein expressed in activated T lymphocytes was decreased by piperlactam S. Results of kinetic study indicated that inhibitory effects of piperlactam S on IL-2 mRNA expressed in T cells might be related to blocking c-Fos protein synthesis. Thus, the suppressant effects of piperlactam S on proliferation of T cells activated by PHA seemed to be mediated, at least in part, through inhibition of early transcripts of T cells, especially those of important cytokines, IL-2, IL-4, and arresting cell cycle progression in the cells.


Subject(s)
Cell Cycle/drug effects , Cytokines/metabolism , Gene Expression Regulation/drug effects , Lactams/pharmacology , Magnoliopsida/chemistry , T-Lymphocytes/drug effects , Adult , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Division/drug effects , Cytokines/genetics , Humans , In Vitro Techniques , Male , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology
12.
Cancer Gene Ther ; 7(6): 826-38, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10880012

ABSTRACT

In our previous studies using gene gun-mediated delivery of interleukin 12 (IL-12) cDNA in vivo, we observed T-cell-mediated regression of established murine tumors and demonstrated the induction of systemic immunity in test animals. In this study, we further characterized the antitumoral and anti-metastatic effect of this gene therapy approach by employing two murine metastatic mammary tumor models: the immunogenic TS/A adenocarcinoma and the weakly immunogenic 4T1 adenocarcinoma. In the TS/A model, gene transfer into the skin overlying an established intradermal tumor with an IL-12 cDNA expression vector resulted in complete tumor regression in 50% of mice followed by the development of immunological memory. In contrast, the growth of the intradermal 4T1 tumors was not affected by the IL-12 gene therapy protocol. However, this treatment resulted in a substantial reduction of spontaneous metastases in the lungs of 4T1 tumor-bearing mice and significantly prolonged their survival time. T cells were not required for this anti-metastatic effect, because it was also observed in nude mice and in mice depleted of CD4+ and CD8+ T cells. Tumor-draining lymph node cells obtained from 4T1 tumor-bearing mice treated with IL-12 cDNA exhibited increased natural killer (NK) activity and produced enhanced levels of interferon-gamma (IFN-gamma) compared with similar mice treated with luciferase cDNA. In addition, in vivo depletion of NK cells or neutralization of IFN-gamma resulted in partial suppression of the anti-metastatic effect of IL-12 gene therapy, suggesting the involvement of both NK cells and IFN-gamma in this effect.


Subject(s)
Adenocarcinoma/prevention & control , Genetic Therapy , Interleukin-12/genetics , Lung Neoplasms/prevention & control , Mammary Neoplasms, Experimental/therapy , T-Lymphocytes/immunology , Adenocarcinoma/immunology , Adenocarcinoma/secondary , Animals , Female , Gene Transfer Techniques , Genetic Vectors , Interferon-gamma/immunology , Interleukin-12/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lymphocyte Count , Lymphocyte Depletion , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Survival Rate , Tumor Cells, Cultured
14.
Methods Mol Med ; 35: 331-44, 2000.
Article in English | MEDLINE | ID: mdl-21390815

ABSTRACT

During the past several years, particle-mediated delivery techniques have been developed as a nonviral technology for gene transfer (1-7). For mammalian somatic tissues, this technology, popularly known as the gene gun method, has been shown effective for transfection of skin, liver, pancreas, muscle, spleen, and other organs in vivo (3,4), brain, mammary, and leukocyte primary cultures or tissue explants ex vivo (2,5-7), and a wide range of cell lines in vitro (3,6,7). In this chapter, we describe the general principles, mechanisms, protocols, and uses of the particle-mediated gene transfer technology for in vivo gene transfer, mainly into skin tissues. Specific applications of this technology to basic studies in molecular biology as well as to gene therapy and genetic immunization against cancer are addressed.

15.
Proc Natl Acad Sci U S A ; 96(23): 13351-6, 1999 Nov 09.
Article in English | MEDLINE | ID: mdl-10557324

ABSTRACT

We report here that a cancer gene therapy protocol using a combination of IL-12, pro-IL-18, and IL-1beta converting enzyme (ICE) cDNA expression vectors simultaneously delivered via gene gun can significantly augment antitumor effects, evidently by generating increased levels of bioactive IL-18 and consequently IFN-gamma. First, we compared the levels of IFN-gamma secreted by mouse splenocytes stimulated with tumor cells transfected with various test genes, including IL-12 alone; pro-IL-18 alone; pro-IL-18 and ICE; IL-12 and pro-IL-18; and IL-12, pro-IL-18, and ICE. Among these treatments, the combination of IL-12, pro-IL-18, and ICE cDNA resulted in the highest level of IFN-gamma production from splenocytes in vitro, and similar results were obtained when these same treatments were delivered to the skin of a mouse by gene gun and IFN-gamma levels were measured at the skin transfection site in vivo. Furthermore, the triple gene combinatorial gene therapy protocol was the most effective among all tested groups at suppressing the growth of TS/A (murine mammary adenocarcinoma) tumors previously implanted intradermally at the skin site receiving DNA transfer by gene gun on days 6, 8, 10, and 12 after tumor implantation. Fifty percent of mice treated with the combined three-gene protocol underwent complete tumor regression. In vivo depletion experiments showed that this antitumor effect was CD8(+) T cell-mediated and partially IFN-gamma-dependent. These results suggest that a combinatorial gene therapy protocol using a mixture of IL-12, pro-IL-18, and ICE cDNAs can confer potent antitumor activities against established TS/A tumors via cytotoxic CD8(+) T cells and IFN-gamma-dependent pathways.


Subject(s)
Adenocarcinoma/pathology , Genetic Therapy , Interleukin-12/genetics , Interleukin-18/genetics , Mammary Neoplasms, Experimental/pathology , Serpins/genetics , Viral Proteins , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Animals , Base Sequence , Biolistics , DNA Primers , DNA, Complementary , Disease Models, Animal , Female , Gene Transfer Techniques , Interferon-gamma/biosynthesis , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Protein Precursors/genetics , RNA, Messenger/genetics , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
16.
Cancer Gene Ther ; 6(4): 331-9, 1999.
Article in English | MEDLINE | ID: mdl-10419051

ABSTRACT

Human papillomaviruses are associated with >90% of all cases of uterine cervical tumors. The E6 and E7 oncoproteins of human papillomavirus are potentially ideal targets of immune therapy for cervical cancer, because their expression is necessary for cellular transformation. Although both E6 and E7 proteins contain numerous predicted cytotoxic T lymphocyte (CTL) epitopes that are capable of binding to human leukocyte antigens, the majority of earlier in vivo tumor rejection studies have focused on E7. We show here that gene gun-mediated skin transfection of plasmid vector encoding the nontransforming, amino-terminal half of E6 resulted in the induction of E6-specific CTL activity and tumor rejection in a murine model. The use of recombinant murine interleukin-12 (rmIL-12) as a vaccine adjuvant has been shown to result in both an enhancement and suppression of immune responses, depending upon the doses of rmIL-12 and the experimental systems used. We demonstrate here that local expression of transgenic mIL-12 at the E6 DNA vaccination site potentiated E6-specific CTL responses and increased vaccine-induced antitumor therapeutic efficacy. Our results indicate that transfection of the mIL-12 gene at the vaccination site may represent an attractive adjuvant for cancer gene immunotherapy.


Subject(s)
Interleukin-12/genetics , Oncogene Proteins, Viral/genetics , Repressor Proteins , Skin/metabolism , Uterine Cervical Neoplasms/therapy , Vaccines, DNA/therapeutic use , Animals , Cancer Vaccines/therapeutic use , DNA, Complementary , Female , Genetic Vectors , Humans , Mice , Mice, Inbred BALB C , Papillomavirus Vaccines , T-Lymphocytes, Cytotoxic/immunology , Transfection , Uterine Cervical Neoplasms/immunology , Viral Vaccines/therapeutic use
17.
Cancer Gene Ther ; 6(1): 26-36, 1999.
Article in English | MEDLINE | ID: mdl-10078961

ABSTRACT

Tumor vaccines and gene therapy have received significant attention as means of increasing cellular and humoral immune responses to cancer. We conducted a pilot study of seven research dogs to determine whether intradermal injection of canine tumor cells transfected via the Accell particle-mediated gene transfer device with the cDNA for human granulocyte-macrophage colony-stimulating factor (hGM-CSF) would generate biologically relevant levels of protein and result in demonstrable histological changes at sites of vaccination. Tumor cell vaccines of 10(7) irradiated canine melanoma cells were nontoxic, safe, and well tolerated. No significant alterations in blood chemistry values or hematological profiles were detected. A histological review of control vaccine sites revealed inflammatory responses predominated by eosinophils, whereas vaccine sites with hGM-CSF-transfected tumor cells had an influx of neutrophils and macrophages. Enzyme-linked immunosorbent assays of skin biopsies from vaccine sites had local hGM-CSF production (8.68-16.82 ng/site of injection) at 24 hours after injection and detectable levels (0.014-0.081 ng/site) for < or =2 weeks following vaccination. Flow cytometric analysis of hGM-CSF-transfected cells demonstrated < or =25% transfection efficiency, and hGM-CSF levels obtained during time-course assays demonstrated biologically relevant levels for both irradiated and nonirradiated samples. These data demonstrate the in vivo biological activity of irradiated hGM-CSF-transfected canine tumor cells and help provide evidence for a valid translational research model of spontaneous tumors.


Subject(s)
Cancer Vaccines/therapeutic use , Gene Transfer Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Melanoma/genetics , Animals , Cell Death , Dogs , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Enzyme-Linked Immunosorbent Assay , Eosinophils/immunology , Genes, Reporter , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Luciferases/metabolism , Pilot Projects , Transfection , Tumor Cells, Cultured , beta-Galactosidase/metabolism
18.
J Immunother ; 22(2): 135-44, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10093038

ABSTRACT

Using three murine tumor models, we compared the antitumor efficacy and certain physiological effects of an in vivo interleukin-12 (IL-12) gene therapy protocol and a systemic IL-12 protein therapy protocol. An IL-12 cDNA gene construct was administered in situ into skin tissue via gene gun delivery, and recombinant IL-12 protein was administered subcutaneously at a dose of 1 microgram/mouse/treatment. Both treatment regimes induced a comparable level of regression of established intradermal MethA sarcomas. In B16 melanoma and P815 mastocytoma models, antitumor efficacy of IL-12 protein therapy appeared to be slightly higher than that of IL-12 gene therapy; however, the protein therapy protocol in this comparative study resulted in a high level of mortality of mice. It was also demonstrated that IL-12 gene therapy, in contrast to the IL-12 protein therapy, was not associated with weight loss, splenomegaly, increased Ly6 antigen expression in the spleen, or visible signs of toxicity, such as fur ruffling and lethargy. Moreover, serum levels of interferon-gamma (IFN-gamma) induced in response to IL-12 gene therapy were 300-1000 times lower than those induced by the systemic IL-12 protein administration. Together, these results suggest that gene gunmediated in vivo delivery of IL-12 cDNA may be considered as a safer alternative to IL-12 protein therapy for certain human cancers.


Subject(s)
Biolistics , Genetic Therapy , Interleukin-12/genetics , Interleukin-12/therapeutic use , Neoplasms, Experimental/therapy , Animals , Antigens, Ly/blood , Female , Interferon-gamma/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Splenomegaly , Weight Loss
19.
Hum Gene Ther ; 9(15): 2197-205, 1998 Oct 10.
Article in English | MEDLINE | ID: mdl-9794204

ABSTRACT

Induction or short-term transgenic expression of specific cytokines, growth factors, or other candidate therapeutic genes in hematopoietic progenitor or stem cells is potentially applicable to gene therapy for cancer. In this study, we explored the application of a gene gun technique, as an alternative to viral vectors, for ex vivo gene transfer into and transient gene expression in highly enriched CD34+ cells derived from human umbilical cord blood. Twenty-four hours posttransfection, 32.6 to 1500 pg/l x 10(6) CD34+ cells of transient gene expression was routinely obtained for specific cytokine and reporter genes. Transgene expression at the single-cell level was revealed by X-Gal staining of lacZ cDNA-transfected CD34+ cells. Expression of four candidate therapeutic genes, namely human granulocyte-macrophage colony-stimulating factor, tumor necrosis factor alpha, interleukin 2, and interferon gamma, was detectable for 4 to 7 days in CD34+ cells. A human elongation factor 1alpha promoter/intron 1 transcription unit was identified as a strong cellular promoter for CD34+ cells, exhibiting strength similar to that of the commonly employed cytomegalovirus immediate-early promoter. These results suggest that the nonviral, gene gun technique offers an efficient alternative approach for transient transgenic studies of hematopoietic cells and may provide new possibilities for certain cancer gene therapy strategies using CD34+ cells.


Subject(s)
Antigens, CD34/analysis , Biolistics , Cytokines/genetics , Hematopoietic Stem Cells/metabolism , Transfection , Transgenes , Cell Survival , Cells, Cultured , Cytokines/biosynthesis , Fetal Blood/cytology , Flow Cytometry , Gene Expression , Genes, Reporter , Genetic Therapy/methods , Hematopoietic Stem Cells/immunology , Humans , Luciferases/metabolism , Promoter Regions, Genetic , Time Factors
20.
Hum Gene Ther ; 9(13): 1851-61, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9741424

ABSTRACT

Cytokine gene-engineered tumor vaccines are currently an area of intense investigation in both basic research and clinical medicine. Our efforts to utilize tumor vaccines in an immunotherapeutic manner involve canines with spontaneous tumors. We hypothesized that canine tumor cells, transfected with human granulocyte-macrophage colony-stimulating factor (hGM-CSF) cDNA in a plasmid vector, would prove nontoxic following intradermal administration, generate biologically relevant levels of protein, effect local histological changes at the sites of vaccination, and create a systemic antitumor response. Sixteen tumor-bearing dogs were admitted to a study of ex vivo gene therapy. Tumor tissue was surgically removed, enzymatically and mechanically dissociated, irradiated, transfected, and intradermally injected back into the patients. The dogs were vaccinated with primary autologous tumor cells transfected with hGM-CSF or a reporter control gene. hGM-CSF protein was detected (0.07 to 14.15 ng/vaccination site) at 24 hr postinjection and dramatic histological changes were observed, characterized by neutrophil and macrophage infiltration at the sites of injection of hGM-CSF-transfected tumor cells. This was in stark contrast to the lesser neutrophilic and eosinophilic infiltrates found at control vaccination sites. Objective evidence of an antitumor response was observed in three animals. These data, in a large animal translational model of spontaneous tumors, demonstrate in vivo biological activity of hGM-CSF-transfected autologous tumor cell vaccines.


Subject(s)
Cancer Vaccines/therapeutic use , Dog Diseases/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Neoplasms/veterinary , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Transplantation/veterinary , DNA, Complementary , Dog Diseases/pathology , Dogs , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Male , Neoplasm Transplantation/veterinary , Neoplasms/pathology , Neoplasms/therapy , Transfection/methods , Tumor Cells, Cultured , Vaccination/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL
...