Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Physiol Pharmacol ; 72(3)2021 Jun.
Article in English | MEDLINE | ID: mdl-34810290

ABSTRACT

This study aimed to investigate the effects of gastric cancer interstitial fluid (GCIF) on tumors and explore the possible mechanism of Xiaotan Sanjie decoction (XTSJ) on treatment of gastric cancer from the view of regulating microRNA-21 (miR-21) expression. The GCIF was extracted and identified by measuring the levels of interleukin-8 (IL-8), intercellular adhesion molecule 1 (ICAM-1) and miR-21. The effects of GCIF on the proliferation of SGC-7901 cells and tumor growing were assessed by cell counting kit-8 (CCK-8) assay and subcutaneously transplanted tumor-bearing nude mice model, respectively. Additionally, inhibition effect of XTSJ decoction on proliferation of SGC-7901 cells intervened by GCIF were assessed in vitro and anti-cancer effect of it was further assessed using orthotopic transplanted tumor-bearing nude mice model. The concentration of SGC-7901 gastric cancer cells were dependent on the concentration of the added GCIF. After 72 hours of continuous culture, the interstitial fluid had an obvious proliferative effect on the SGC-7901 tumor cells, which was the most significant in the high concentration group. XTSJ decoction could inhibit the growth-promoting effect (P < 0.01) of GCIF on gastric cancer cells. Intervention of the GCIF might promote the growth (P < 0.05) of the subcutaneously transplanted tumors in nude mice and decrease the net weight of the tumor-bearing nude mice (P < 0.05) after tumor removal. The GCIF was able to up-regulate the expression (P < 0.001) of miR-21 in the subcutaneously transplanted tumors. XTSJ decoction could downregulate the expression (P < 0.05) of miR-21 in SGC-7901 orthotopically transplanted tumors. XTSJ decoction can inhibit the multiplicative effect of GCIF on gastric cancer cells, growth of gastric tumor and promotion effect of GCIF on tumors, probably due to the down-regulating miR-21 expression in tumor tissues.


Subject(s)
MicroRNAs , Stomach Neoplasms , Animals , Cell Line, Tumor , Cell Proliferation , Extracellular Fluid , Gene Expression Regulation, Neoplastic , Medicine, Chinese Traditional , Mice , Mice, Nude , MicroRNAs/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
2.
Gene Ther ; 9(12): 793-803, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12040461

ABSTRACT

Chemokine gene transfer represents a promising approach in the treatment of malignancies. Macrophage-derived chemokine (MDC) (CCL22) belongs to the CC chemokine family and is a strong chemoattractant for dendritic cells (DC), NK cells and T cells. Using adenoviral vectors, human MDC gene was transferred in vivo to investigate its efficacy to induce an antitumor response and to determine the immunologic mechanisms involved. We observed that intratumoral injection of recombinant adenovirus encoding human MDC (AdMDC) resulted in marked tumor regression in a murine model with pre-established subcutaneous 3LL lung carcinoma and induced significant CTL activity. The antitumor response was demonstrated to be CD4+ T cell- and CD8+ T cell-dependent. Administration of AdMDC induced chemoattraction of DC to the tumor site, facilitated DC migration to draining lymph nodes or spleen, and finally activated DC to produce high levels of IL-12. Furthermore, a significant increase of IL-4 production within the tumors was observed early after the AdMDC administration and was followed by the increase of IL-12 and IL-2 production. The levels of IL-2, IL-12 and IFN-gamma in serum, lymph nodes and spleen were also found to be higher in mice treated with AdMDC as compared with that in AdLacZ- or PBS-treated mice. The antitumor response induced by AdMDC was markedly impaired in IL-4 knockout mice, suggesting an important role of IL-4 in the induction of antitumor immunity by MDC. These results suggest that MDC gene transfer might elicit significant antitumor effects through efficient induction of antitumor immunity and might be of therapeutic potentials for cancer.


Subject(s)
Chemokines, CC/genetics , Genetic Therapy/methods , Lung Neoplasms/therapy , Adenoviridae/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL22 , Dendritic Cells/immunology , Genetic Vectors/administration & dosage , Injections, Intralesional , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-2/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lung Neoplasms/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Transfection
3.
Gene Ther ; 8(7): 542-50, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11319621

ABSTRACT

Antibody-targeted superantigen C215Fab-SEA is a fusion protein of staphylococcal enterotoxin A (SEA) and the Fab region of the tumor-reactive C215 mAb. It can trigger CTL against C215 antigen-positive tumor cells and induce tumor-suppressive cytokines. However, the antitumor effect of C215Fab-SEA is not satisfactory because of suboptimal production of Th1 cytokines after repeated administration. Interleukin 18 (IL-18) is a novel cytokine with profound effects on Th1 cellular response. In this study, we showed that adenovirus-mediated intratumoral IL-18 gene transfer strongly improved the therapeutic efficacy of C215Fab-SEA in the pre-established C215 antigen-expressing B16 melanoma murine model. More significant tumor inhibition and prolonged survival time were observed in tumor-bearing mice received combined therapy of C215Fab-SEA and Ad IL-18 than those of mice treated with C215Fab-SEA or AdIL-18 alone. Combination therapy augmented NK and CTL activities of tumor-bearing mice more markedly. The production of IL-2 and IFN-gamma also increased more significantly. More potent antitumor effect of combined therapy was observed in IL-10 KO mice with enhanced Th1 response. Our data demonstrated that the antitumor effect of C215Fab-SEA immunotherapy could be potentiated significantly by combination with intratumoral IL-18 gene transfer through more efficient activation of Th1 immune responses.


Subject(s)
Genetic Therapy/methods , Interleukin-18/genetics , Melanoma, Experimental/therapy , Superantigens/therapeutic use , Adenoviridae/genetics , Animals , Antigens, Neoplasm/immunology , Combined Modality Therapy , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Enterotoxins/immunology , Female , Immunoglobulin Fab Fragments/immunology , Interleukin-18/immunology , Killer Cells, Natural/immunology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/therapeutic use , Spleen/immunology , Staphylococcus aureus/immunology , Superantigens/immunology , Survival Rate , T-Lymphocytes, Cytotoxic/immunology
4.
Cancer Res ; 61(9): 3735-40, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11325846

ABSTRACT

Dendritic cell (DC)-based tumor vaccine represents a promising approach to the immunotherapy of malignant tumors. We prepared a novel type of DC-based vaccine, stable conjugates of DCs and EL4 cells transduced with cDNA of OVA (E.G7). Immunization with DC-E.G7 conjugates led to generation of T helper (Th) 1 cytokine-producing cells, antigen-specific CD8(+) T cells, and strong antitumor immunity that is dependent on both CD4(+) T cells and CD8(+) T cells. To further increase the potency of the vaccine, interleukin 18-transfected DCs were used to prepare the IL18DC-E.G7 conjugates. Immunization with such conjugates significantly increased the production of Th1 cytokine-producing cells and the number of antigen-specific CD8(+) T cells, as well as stronger antitumor immunity. Furthermore, the increased Th1 cytokine production and stronger antitumor effect were not observed in mice depleted of IFN-gamma. These data indicated that DC-tumor cell conjugates are a potent tumor vaccine. Interleukin 18 can be administrated using gene-transfected cells and enhances antitumor immunity, which is mainly mediated by IFN-gamma.


Subject(s)
Dendritic Cells/immunology , Interleukin-18/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , DNA, Complementary/genetics , Dendritic Cells/cytology , Epitopes, T-Lymphocyte/immunology , Female , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-18/genetics , Interleukin-2/biosynthesis , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Th1 Cells/immunology , Th1 Cells/metabolism , Thymoma/immunology , Thymoma/pathology , Transfection
5.
J Cancer Res Clin Oncol ; 127(2): 101-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11216910

ABSTRACT

PURPOSE: Tumor suppressor gene therapy and cytokine gene therapy have limited antitumor effects when used alone. Thus, in the present study, we investigated the antitumor potentials of the combined transfer of the p16 tumor suppressor gene and the murine granulocyte-macrophage colony-stimulating factor (GM-CSF) gene. METHODS: The adenovirus-harboring p16 gene (Adp16) and adenovirus-harboring GM-CSF (AdGMCSF) gene were utilized for the treatment of established tumors in vivo. The mice were inoculated s.c. with Renca renal carcinoma cells and 3 days later received an intratumoral injection of Adp16 in combination with AdGMCSF. RESULTS: The results demonstrated that tumor-bearing mice treated with Adp16 and Ad-GMCSF showed more potent inhibition of tumor growth and a prolonged survival period than mice treated with Adp16. AdGMCSF, adenovirus-expressing beta-galactosidase or PBS (P<0.01). Treatments of the mice with Adp16 alone or AdGMCSF alone also showed obvious antitumor effects as compared with those mice treated with PBS (P<0.05). After combined p16 and AdGMCSF gene therapy, the expression of H2Kd and Fas molecules on freshly isolated tumor cells increased markedly, and more CD(4)+ T cells and CD(8)+ T cells infiltrated in the tumor sites. The cytotoxicity of natural killer cells and specific cytotoxic T lymphocytes increased more significantly after the combined therapy. CONCLUSIONS: Our results demonstrated that combination p16 gene and GM-CSF gene therapy could inhibit the growth of established tumors in mice more significantly through efficient induction of antitumor immunity.


Subject(s)
Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Genes, p16 , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Adenoviridae , Animals , Blotting, Western , Carcinoma, Renal Cell/genetics , Female , Gene Transfer Techniques , Mice , Mice, Inbred BALB C , Transfection , Tumor Cells, Cultured
6.
Gene Ther ; 7(19): 1672-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11083476

ABSTRACT

To increase the antitumor effects of cytosine deaminase (AdCD) gene therapy and induce more potent antitumor immunity, Th1 cytokine interleukin-18 encoded adenovirus (AdIL18) was combined with adenovirus encoding CD (AdCD) for the therapy of established murine B16 melanoma. Combination therapy of the tumor-bearing mice with AdIL 18 and AdCD/5FC inhibited the growth of the subcutaneous B16 tumors more significantly, compared with AdIL 18 or AdCD/5FC alone. In vivo depletion analysis with anti-CD4, anti-CD8 or anti-NK 1.1 McAb illustrated that both CD8+ T cells and CD4+ T cells played key roles in the augmented antitumor response of the combined therapy. Peptide/MHC tetramer represents a powerful and general tool for rapid, highly sensitive, and direct analysis of antigen-specific T cells. In this study, we prepared H-2Kb/TRP-2180-188 tetramer, which was demonstrated to bind H-2Kb-restricted, B16 melanoma-specific CD8+ T cells. B16 specific H-2Kb/TRP2180-188 tetramer was used to stain the tumor-specific CD8+ T cells and the results showed that CD8+ tetramer+ T cells were about 3-5% of the splenic CD8+ T cells derived from tumor-bearing mice after combined therapy. The CTL cytotoxicity was markedly induced in mice after combined therapy, suggesting efficient induction of tumor-specific CD8+ T cells after combined gene therapy with AdCD/5FC/AdIL18. IL-18 gene transfer could significantly augment the cytotoxicity of NK cells and macrophages, and increase the production of interleukin-2 and interferon-gamma, as compared with treatments with AdCD/5FC, AdlacZ/5FC or PBS. These data suggested that in vivo IL-18 gene transfer could augment the antitumor effects of CD suicide gene therapy through efficient induction of antitumor immunity.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Interleukin-18/genetics , Melanoma, Experimental/therapy , Nucleoside Deaminases/genetics , Adenoviridae/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytosine Deaminase , Female , Genetic Vectors/administration & dosage , Interferon-gamma/immunology , Interleukin-2/immunology , Killer Cells, Natural/immunology , Macrophages/immunology , Male , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL
7.
Gene Ther ; 7(8): 707-13, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10800095

ABSTRACT

Adoptive transfer of activated macrophages, being both effector cells and antigen-presenting cells, represents a promising approach to immunotherapy of cancer. In order to get activated macrophages with increased antitumor potential, in the present study, murine peritoneal macrophages were transduced with human macrophage colony-stimulating factor (M-CSF) and murine interferon-gamma (IFNgamma) by recombinant adenovirus infection. The results demonstrate that M-CSF and IFNgamma gene-modified macrophages exhibited higher expression of MHC-II, B7.1 and ICAM-1, increased antigen-presenting activity and cytotoxicity. It was also shown that they secreted more tumor necrosis factor, interleukin-1 and nitric oxide. In vivo experiments showed that in previously initiated murine pulmonary metastatic melanoma, tumor lysate-pulsed, M-CSF and IFNgamma gene-modified macrophages elicited more potent antitumor effects than tumor lysate pulsed M-CSF or IFNgamma gene-modified macrophages. Cytotoxic T lymphocyte (CTL) activity, IFNgamma and tumor-necrosis factor production of the splenocytes increased significantly in mice after intravenous injection of the gene-modified macrophages. M-CSF and IFNgamma gene-modified macrophages may act as activated effector and antigen-presenting cells, thus eliciting a more potent antitumor response.


Subject(s)
Immunotherapy, Adoptive/methods , Interferon-gamma/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages, Peritoneal/immunology , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Animals , Antigen Presentation , Genetic Engineering/methods , Humans , Interferon-gamma/genetics , Interleukin-1/metabolism , Macrophage Colony-Stimulating Factor/genetics , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Skin Neoplasms/immunology , Tumor Necrosis Factor-alpha/metabolism
8.
Cancer Gene Ther ; 7(2): 177-86, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10770625

ABSTRACT

Suicide gene therapy has been studied intensively for the treatment of cancer. A limited antitumoral effect was obtained by intratumoral injection of adenovirus harboring Escherichia coli cytosine deaminase gene (AdCD) in tumor-bearing mice followed by continuous administration of 5-fluorocytosine (5FC). To address the drawbacks of the limited potential for the induction of antitumoral immunity by CD suicide gene therapy, we hypothesized that antigen-presenting cells (APCs) might contribute to the efficient induction of an antitumoral immune response in tumor-bearing mice undergoing suicide gene therapy. We preinjected the mice with murine stem cell factor (SCF)-encoding adenovirus (AdSCF) and murine granulocyte-macrophage colony-stimulating factor (GM-CSF)-encoding adenovirus (AdGM-CSF); after 7 days, the mice were inoculated with CT26 colon adenocarcinoma. AdCD was injected intratumorally into tumor-bearing mice followed by 5FC administration. The results showed that AdSCF/AdGM-CSF treatment could increase the number, surface molecule expression, and function of APCs efficiently. A more significant growth inhibition of established tumors and a prolongation of the survival period were observed in tumor-bearing mice after AdSCF/AdGM-CSF pretreatment in combination with AdCD/5FC therapy when compared with mice treated with AdSCF or AdGM-CSF in combination with AdCD/5FC, or AdCD/5FC alone (P < .01). Cytotoxic T-lymphocyte activity was induced efficiently after the combined therapy, and mRNA of tumor necrosis factor-alpha, interleukin-4, interferon-gamma, and interleukin-2 was present in the tumor mass after combined therapy, suggesting that a more potent antitumoral response was induced by enhanced APCs. Our results demonstrated that AdSCF/AdGM-CSF pretreatment could activate APCs, and that these APCs could present the tumor antigens released from AdCD/5FC-killed tumor cells and activate the antitumoral response of the host, thus increasing the therapeutic efficiency of suicide gene therapy.


Subject(s)
Adenoviridae/enzymology , Adenoviridae/genetics , Adjuvants, Immunologic/genetics , Antigen-Presenting Cells/cytology , Gene Transfer Techniques , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Nucleoside Deaminases/genetics , Stem Cell Factor/genetics , Adenoviridae/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Cancer Vaccines/therapeutic use , Cell Count , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Combined Modality Therapy , Cytokines/biosynthesis , Cytosine Deaminase , Female , Humans , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured
9.
Gene Ther ; 7(4): 329-38, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10694814

ABSTRACT

Lymphotactin (Ltn) is the sole member of C chemokines which attracts T cells and NK cells specially. Ltn gene was transferred in vivo to improve the antitumor efficacy of cytosine deaminase (CD) gene therapy. Upregulation of CD80 and CD54 on murine CT26 colon carcinoma cells was observed after combined transfection with adenovirus encoding CD (AdCD) and adenovirus encoding murine Ltn (AdLtn) followed by administration of 5-fluorocytosine (5FC) in vitro. AdCD/5FC treatment also increased the expression of CD95 and induced obvious apoptosis of CT26 cells. After combined treatment with AdLtn and AdCD/5FC, the pre-established murine model with subcutaneous CT26 colon carcinoma exhibited most significant tumor growth inhibition, and four of eight tumor-bearing mice were tumor free, while tumors in other mice grew more progressively. Examination of lymphocyte infiltration and cytokine gene expression in tumor tissue revealed that tumors from AdLtn/AdCD/5FC-or AdLtn-treated mice were heavily infiltrated with CD4+, CD8+ T cells and NK cells, and IL-2 and IFN-gamma mRNA expression were present in parallel with T cell and NK cell infiltration. Splenic NK and CTL activities increased significantly after the combination therapy. In vivo depletion analysis showed that NK cells, CD4+ T cells and CD8+T cells participated in the antitumor effect of the host with CD8+T cells being the main T cell subset responsible for the enhanced antitumor immune response. These findings suggested that increased immunogenicity and induction of apoptosis of the tumor cells, and efficient induction of local and systemic antitumor immunity of the host might contribute to the enhanced antitumor effects of the combined Ltn and CD suicide therapy. Gene Therapy (2000) 7, 329-338.


Subject(s)
Adenoviridae/genetics , Chemokines, C , Colonic Neoplasms/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Nucleoside Deaminases/genetics , Animals , Apoptosis , Colonic Neoplasms/pathology , Cytosine Deaminase , Female , Flow Cytometry , Immunity, Active/genetics , Lymphokines/genetics , Male , Mice , Mice, Inbred BALB C , Nucleoside Deaminases/therapeutic use , RNA, Messenger/metabolism , Sialoglycoproteins/genetics , Transfection/genetics , Tumor Cells, Cultured
10.
J Exp Clin Cancer Res ; 18(2): 191-200, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10464706

ABSTRACT

Interleukin 2 (IL-2) expressing plasmid and interleukin 6 (IL-6)-expressing plasmid were encapsulated in liposome and administrated intratumoraly into tumor-bearing mice 4 days after subcutaneous inoculation of B16F10 melanoma cells. The results showed that treatment of tumor-bearing mice with IL-2 gene or IL-6 gene transfer inhibited the growth of subcutaneous tumor and prolonged the survival of tumor-bearing mice significantly when compared with the treatment of PBS or control gene transfer mediated by liposome (P < 0.01). Combined transfer of IL-2 gene and IL-6 gene was found to elicit inhibitory effects on the growth of B16F10 tumor more significantly and prolonged the survival period of tumor-bearing mice more obviously. We investigated the local immunity in tumor microenvironment and found that IL-2 and IL-6 gene transfer could significantly increase the expression of lymphocyte function-associated antigen-1 on tumor infiltrating lymphocytes (TIL) and MHC-I molecule on tumor cells freshly isolated from the tumor mass. The NK and CTL activity of TIL increased markedly after the combined transfer of these two cytokine genes. We also observed the systemic antitumor immune response in the tumor-bearing mice and demonstrated that NK and CTL activity of splenocytes and the production of IL-2, tumor necrosis factor and interferon-gamma from splenocytes increased obviously in mice after the combined transfer of IL-2 and IL-6 gene. In conclusion, local and systemic antitumor immunity of the tumor-bearing host could be induced efficiently after the combined gene transfer. The enhanced specific and non-specific antitumor immunity might be responsible for the more potent antitumor effects of the combined gene therapy.


Subject(s)
Gene Transfer Techniques , Interleukin-2/administration & dosage , Interleukin-2/genetics , Interleukin-6/administration & dosage , Interleukin-6/genetics , Liposomes/metabolism , Melanoma, Experimental/immunology , Muscle Neoplasms/immunology , Animals , Cytokines/biosynthesis , Genetic Therapy , Immunity, Cellular/genetics , Injections, Intralesional , Liposomes/genetics , Lymphocyte Function-Associated Antigen-1/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Muscle Neoplasms/genetics , Muscle Neoplasms/therapy , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology
11.
Transplantation ; 67(9): 1242-50, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10342316

ABSTRACT

BACKGROUND: Bone marrow transplantation (BMT) and gene therapy are potent approaches to the recovery of bone marrow depression and induction of antitumor immunity after chemotherapy for the treatment of malignancies. In the present study, enhanced antitumor effect of BMT in combination with fibroblast-mediated interleukin (IL)-2 and IL-3 gene therapy was observed in tumor-bearing mice after chemotherapy. METHODS: BALB/c mice were inoculated s.c. with J558L plasmacytoma cells and injected i.p. with cyclophosphamide 300 mg/kg 3 days later. 24 hours after chemotherapy syngeneic bone marrow cells in combination with NIH3T3 fibroblast cells engineered to produce IL-2 (NIH3T3-IL-2) and/or NIH3T3 cells engineered to produce IL-3 (NIH3T3-IL-3) were implanted into the tumor-bearing mice. RESULTS: BMT in combination with implantation of either NIH3T3-IL-2 or NIH3T3-IL-3 cells exerted significant inhibition on the growth of J558L tumors and prolonged the survival period of the tumor-bearing mice as compared with the treatments with Hanks solution, BMT alone, or BMT plus implantation of NIH3T3 cells transduced with Neo gene. Synergistic antitumor effect was observed in mice after combined BMT and cytokine gene therapy. The cytotoxicities of natural killer cells, cytotoxic T lymphocytes, and macrophages in mice increased markedly after the combined treatment. Recovery of CFU-GM, CFU-MK and CFU-E formation in mice after combined therapy was accelerated obviously in mice after combined therapy. CONCLUSIONS: BMT in combination with fibroblast-mediated IL-2 and IL-3 gene therapy elicited augmented antitumor effects synergistically in tumor-bearing mice after chemotherapy mainly through induction of antitumor immune response and accelerated recovery of hematopoiesis.


Subject(s)
3T3 Cells/transplantation , Bone Marrow Transplantation , Genetic Therapy/methods , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-3/genetics , Interleukin-3/immunology , Plasmacytoma/therapy , 3T3 Cells/metabolism , Animals , Bone Marrow Cells/immunology , Combined Modality Therapy , Cyclophosphamide/pharmacology , Female , Hematopoiesis/physiology , Immunosuppressive Agents/pharmacology , Interferon-gamma/biosynthesis , Interleukin-2/metabolism , Interleukin-3/metabolism , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Macrophage Activation , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred BALB C , Monocytes/immunology , Neutrophils/immunology , Plasmacytoma/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Cytotoxic/immunology , Transduction, Genetic
12.
Chin Med J (Engl) ; 112(2): 162-5, 1999 Feb.
Article in English | MEDLINE | ID: mdl-11593586

ABSTRACT

OBJECTIVE: To investigate the antitumor effect of combined adenovirus encoding E. coli cytosine deaminase (AdCD) and adenovirus encoding murine interleukin 2 (AdIL-2) on murine melanoma. METHODS: C57BL/6 mice were inoculated s.c. with B16F10 melanoma cells and 3 days later received injections of AdCD and/or AdIL-2 at the site of tumor inoculation followed by administration of 5-flurocytosine (5FC) 300 mg/kg per day for 10 days. RESULTS: Mice receiving AdCD/5FC/AdIL2 therapy developed tumors more slowly and survived much longer when compared with mice treated with AdCD/5FC, AdIL2, AdlacZ/5FC, or PBS. Immunological analysis illustrated that combined treatment could enhance NK activity and CTL activity. Flow cytometry demonstrated that AdCD/5FC/AdIL2 therapy increased the expression of MHC-1 and CD80 molecules on freshly isolated tumor cells. The CD4+ and CD8+ T cell infiltration in the tumor increased significantly after the combined therapy. CONCLUSIONS: Our data showed that combined transfer of CD suicide gene and IL-2 gene could inhibit the tumor growth more significantly. The increased specific and non-specific antitumor immunity might be responsible for the enhanced therapeutic effect.


Subject(s)
Flucytosine/therapeutic use , Genetic Therapy , Interleukin-2/genetics , Melanoma, Experimental/therapy , Nucleoside Deaminases/genetics , Adenoviruses, Human/genetics , Animals , Cytosine Deaminase , Escherichia coli/genetics , Female , Gene Transfer Techniques , Genes, Tumor Suppressor , Interleukin-2/therapeutic use , Male , Mice , Mice, Inbred C57BL , Nucleoside Deaminases/therapeutic use
13.
J Mol Med (Berl) ; 76(11): 782-9, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9826123

ABSTRACT

Fibroblast-mediated cytokine gene therapy has proven to be a promising strategy for restoring hematopoiesis following repeated chemotherapy. Interleukin 3 (IL-3) and interleukin 6 (IL-6) can synergistically promote the recovery of hematopoiesis following chemotherapy. In this investigation, combined use of fibroblast-mediated IL-3 and IL-6 gene therapy was tested for hematopoietic effects on mice with or without 5-fluorouracil administration. The results demonstrated that combined therapy with IL-3 gene-modified NIH3T3 cell (NIH3T3-IL-3) and IL-6 gene-modified fibroblast NIH3T3 cell (NIH3T3-IL-6) implantation achieves obvious stimulation of hematopoiesis in normal mice and accelerates recovery of hematopoiesis. In normal mice the quantities of platelets, neutrophils, and total white blood cells in peripheral blood increased significantly after the combined implantation of NIH3T3-IL-3 and NIH3T3-IL-6 cells. The numbers of colony-forming unit (CFU) granulocyte/macrophage (CFU-GM) and CFU megakaryocyte (CFU-MK) formed by stem cells in bone marrow was significantly higher after the combined implantation of NIH3T3-IL-3 and NIH3T3-IL-6 cells than after the implantation of NIH3T3-IL-3 alone, NIH3T3-IL-6 alone, or neomycin gene-modified NIH3T3 cells. In hematopoiesis-depressed mice induced by preinjection with 5-fluorouracil at the dose of 150 mg/kg before cell implantation, the platelets, neutrophils, and white blood cells showed accelerated recovery, and the numbers of CFU-GM and CFU-MK formed by bone marrow cells were also markedly higher after the combined implantation of NIH3T3-IL-3 and NIH3T3-IL-6 cells than in control groups. Our data show that combined use of fibroblast-mediated IL-3 and IL-6 gene therapy may be of clinical relevance for the recovery of hematopoietic depression for patients after chemotherapy.


Subject(s)
Fluorouracil/pharmacology , Genetic Therapy , Hematopoiesis , Immunosuppressive Agents/pharmacology , Interleukin-3/pharmacology , Interleukin-6/pharmacology , 3T3 Cells , Animals , Bone Marrow Cells , Granulocytes , Interleukin-3/biosynthesis , Interleukin-3/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Leukocytes , Macrophages , Male , Megakaryocytes , Mice , Mice, Inbred BALB C , Platelet Count
14.
Pharmacology ; 56(4): 187-95, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9566020

ABSTRACT

Esculentoside A, a kind of saponin isolated from the root of the Chinese herb Phytolaca esculenta, is reported to possess potent anti-inflammatory effects in acute and chronic experimental models. In the present study, we investigated the effects of esculentoside A on the production of tumor necrosis factor (TNF), interleukin-1 (IL-1) and interleukin-6 (IL-6) induced by lipopolysaccharide (LPS) in mice. In vitro experiments demonstrated that esculentoside A (0.1-10 mumol/l) significantly reduced the release of TNF from the peritoneal macrophages derived from mice pretreated with thioglycolate. IL-1 and IL-6 secretion was also obviously inhibited in a concentration-dependent manner by esculentoside A from 0.01 to 10 mumol/l. In vivo experiments demonstrated that detectable TNF was observed 0.25 h after injection, was maximal at 0.5 h, and returned to baseline at 4 h. Maximal production of IL-1 and IL-6 were observed to be 1 and 2 h, respectively, after injection of LPS. Pretreatment of mice with 5, 10, or 20 mg/kg esculentoside A once a day for 7 consecutive days dose-dependently decreased the TNF, IL-1 and IL-6 levels in the sera of mice following LPS challenge. TNF, IL-1, and IL-6 are important cytokines involved in the pathogenesis of inflammatory lesions. Inhibition of inflammatory cytokine production may contribute to the anti-inflammatory effects of esculentoside A.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Interleukin-1/metabolism , Interleukin-6/metabolism , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , Shock, Septic/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Division/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Macrophages, Peritoneal/drug effects , Male , Mice , Mice, Inbred ICR , Regression Analysis , Shock, Septic/metabolism , Thymus Gland/drug effects
15.
J Interferon Cytokine Res ; 18(4): 227-33, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9568724

ABSTRACT

Murine fibroblast NIH3T3 cells engineered to secrete interleukin-6 (NIH3T3-IL-6) were implanted intraperitoneally into mice and observed for their hematopoietic effects with or without 5-fluorouracil (5-FU) administration. In normal mice, the platelet and neutrophil counts in peripheral blood increased significantly after implantation of NIH3T3-IL-6 cells, but the total white blood cell numbers showed no obvious elevation. The granulocyte-macrophage colony-forming unit (CFU-GM) and megakaryocyte colony-forming unit (CFU-MK) numbers formed by stem cells in bone marrow and spleen were also found to be significantly increased after implantation of NIH3T3-IL-6 cells when compared with those in mice after implantation of NIH3T3 cells transduced with neomycin gene (NIH3T3-Neo). To observe the protective effects of NIH3T3-IL-6 cells on hematopoietic depression in chemotherapy-treated mice, the mice were preinjected with 5-FU at a dosage of 150 mg/kg before implantation of NIH3T3-IL-6 cells. The platelet and neutrophil counts showed accelerated recovery after implantation of NIH3T3-IL-6 cells. The numbers of CFU-GM and CFU-MK in bone marrow and spleen were also found to be markedly increased in hematopoietic-depressed mice when compared with those in mice implanted with NIH3T3-Neo cells. These data suggest that fibroblast-mediated IL-6 gene therapy, which can augment hematopoiesis in mice with or without chemotherapy, will be of great help in the recovery from hematopoietic depression after chemotherapy.


Subject(s)
Fluorouracil/therapeutic use , Genetic Therapy , Hematopoiesis/drug effects , Interleukin-6/genetics , 3T3 Cells , Animals , Bone Marrow Cells/drug effects , Cell Transplantation , Drug Evaluation, Preclinical , Female , Gamma Rays , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Male , Megakaryocytes/drug effects , Mice , Mice, Inbred BALB C , Platelet Count/drug effects , Spleen/cytology , Spleen/drug effects
16.
Transplantation ; 65(3): 325-31, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9484747

ABSTRACT

BACKGROUND: Both fibroblast-mediated cytokine gene therapy and bone marrow transplantation (BMT) have proven to be efficient protocols for the recovery of bone marrow depression. In this report, the effects of fibroblast-mediated interleukin (IL)-6 gene therapy, in combination with BMT, on the recovery of irradiation-induced bone marrow depression were investigated. METHODS: NIH3T3 fibroblast cells engineered to secrete IL-6 (NIH3T3-IL-6) or NIH3T3 cells transduced with the neomycin gene (NIH3T3-Neo), in combination with 10(7), 10(6), or 10(5) syngeneic bone marrow cells, were implanted into irradiated mice. RESULTS: The platelets and white blood cells in the peripheral blood of the irradiated mice increased greatly 12 days after implantation of NIH3T3-IL-6 cells and BMT, the white blood cell counts were restored to a normal level 32 days after the combined therapy, and the platelet number was obviously higher than that in mice implanted with NIH3T3-Neo and BMT. Twenty and 25 days after the combined therapy, the mice showed accelerated recovery of colony-forming unit (CFU)-granulocyte/macrophages and CFU-megakaryocytes when compared with the mice implanted with NIH3T3-Neo cells and BMT. Ten days after lethal irradiation with gamma rays, the spleens formed more CFU-spleen in mice implanted with NIH3T3-IL-6 cells and BMT than in mice injected with phosphate-buffered saline or NIH3T3-Neo cells. Combined therapy with NIH3T3-IL-6 cell implantation and BMT delayed the survival period of the hematopoietic-depressed mice significantly when compared with therapy with NIH3T3-Neo cell implantation and BMT. CONCLUSIONS: These data demonstrated that the combined therapy of fibroblast-mediated IL-6 gene therapy and BMT could significantly promote the recovery of irradiation-induced hematopoietic depression.


Subject(s)
Bone Marrow Transplantation , Bone Marrow/radiation effects , Genetic Therapy , Interleukin-6/genetics , 3T3 Cells/transplantation , Animals , Cobalt Radioisotopes , Combined Modality Therapy , Female , Fibroblasts/transplantation , Gamma Rays , Humans , Interleukin-6/biosynthesis , Male , Mice , Mice, Inbred BALB C , Survival Rate , Transfection , Transplantation, Isogeneic
17.
J Cancer Res Clin Oncol ; 124(12): 683-9, 1998.
Article in English | MEDLINE | ID: mdl-9879829

ABSTRACT

The antitumor effect of the combined transfer of a suicide gene and a cytokine gene was evaluated in the present study. Adenoviruses expressing Escherichia coli cytosine deaminase (AdCD) and adenoviruses expressing murine interleukin-2 (AdIL-2) were utilized for the treatment of established tumors. The mice were inoculated s.c. with FBL-3 erythroleukemia cells and 3 days later received an intratumoral injection of AdCD in the presence or absence of AdIL-2 followed by intraperitoneal 5-fluorocytosine (5-FC) administration. The results demonstrated that tumor-bearing mice treated with AdCD/5-FC in combination with AdIL-2 showed more potent inhibition of tumor growth and survived much longer than did mice treated with AdCD/5-FC, AdIL-2, adenovirus expressing beta-galactosidase/5-FC or phosphate-buffered saline. The tumor mass showed obvious necrosis and inflammatory cell infiltration, and more CD4+ and CD8+ T cells infiltrating the tumor after combined therapy. The splenic natural killer and cytotoxic T lymphocyte activities increased significantly in the mice after combined therapy with AdCD/5-FC/AdIL-2. Our results demonstrate that therapy combining a suicide gene and IL-2 gene can inhibit the growth of established tumors in mice significantly and induce antitumor immunity of the host efficiently.


Subject(s)
Antimetabolites/therapeutic use , Flucytosine/therapeutic use , Gene Transfer Techniques , Genetic Therapy , Interleukin-2/genetics , Neoplasms, Experimental/therapy , Nucleoside Deaminases/genetics , Adenoviridae/genetics , Animals , Cytosine Deaminase , Escherichia coli/enzymology , Female , Interleukin-2/biosynthesis , Male , Mice , Mice, Inbred C57BL , Nucleoside Deaminases/biosynthesis , T-Lymphocytes, Cytotoxic , Tumor Cells, Cultured
18.
Gene Ther ; 5(8): 1130-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-10326037

ABSTRACT

Antitumor effects of combined transfer of suicide and cytokine genes were investigated in this study. Adenovirus harboring E. coli cytosine deaminase gene (AdCD) and adenovirus harboring murine granulocyte-macrophage colony-stimulating factor gene (AdGMCSF) were used simultaneously for in vivo gene transfer in melanoma-bearing mice. Growth inhibition of established tumors and prolongation of survival period were observed more significantly in tumor-bearing mice after transfection with AdGMCSF and AdCD followed by continuous injection of prodrug 5-fluorocytosine (5FC) when compared with mice treated with control adenovirus AdlacZ/5FC, AdCD/5FC or AdGMCSF alone (P < 0.01). After combined therapy the expression of MHC-I (H-2Db) and B7-1 molecules on freshly isolated tumor cells increased greatly and more dendritic cells and CD8+ T cells infiltrated into the tumor mass. The activity of specific cytotoxic T lymphocytes was also found to be induced more significantly after the combined therapy. Further experiments showed that apoptosis of tumor cells and induction of antitumor immune response might be involved in the mechanisms of the tumor cell killing by the combined therapy. Our results demonstrated that combined transfer of the GM-CSF and CD suicide genes, being able to inhibit the growth of melanoma synergistically and induce specific antitumor immune response efficiently, thus addressing the drawbacks of suicide gene therapy or cytokine gene therapy which were proved to be not satisfactory when used alone, might be of therapeutic potential for gene therapy of cancer.


Subject(s)
Antimetabolites/therapeutic use , Flucytosine/therapeutic use , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Melanoma, Experimental/therapy , Nucleoside Deaminases/genetics , Prodrugs/therapeutic use , Adenoviridae , Animals , Apoptosis , Combined Modality Therapy , Cytosine Deaminase , Cytotoxicity Tests, Immunologic , Escherichia coli/enzymology , Female , Flow Cytometry , Genetic Vectors , Male , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Transfection/methods
19.
Cancer Gene Ther ; 4(2): 139-44, 1997.
Article in English | MEDLINE | ID: mdl-9080123

ABSTRACT

A recombinant vaccinia virus containing and expressing the gene for murine granulocyte-macrophage colony-stimulating factor (VVGM-CSF) was constructed and tested for its antitumor activity. A murine tumor model was established by injecting 10(5) B16F10 melanoma cells into the right rear leg of C57BL/6 mice. Three days after B16F10 inoculation, VVGM-CSF or a thymidine kinase gene-deficient vaccinia virus (VVTK) were injected intratumorly twice weekly for 3 weeks. The results showed that VVGM-CSF treatment significantly inhibited the growth of subcutaneous tumor and delayed the survival period of tumor-bearing mice. Splenic lymphokine-activated killer cell, natural killer cell, and cytotoxic T lymphocyte activities were not found to be altered after VVGM-CSF or VVTK therapy. Cytotoxic and phagocytic activity of peritoneal macrophages were found to be greatly elevated in mice treated with VVGM-CSF. Nitric oxide released from the macrophages was also increased. Considering these data, we may speculate that continuous secretion of GM-CSF and activation of macrophages may contribute to the antitumor effects of VVGM-CSF injected intratumorally.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Melanoma/pathology , Melanoma/therapy , Vaccinia/genetics , Animals , Cell Death/drug effects , Cell Death/genetics , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/metabolism , Injections, Intralesional , Macrophages/drug effects , Macrophages/metabolism , Male , Melanoma/genetics , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Phagocytosis/drug effects , Phagocytosis/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Survival Rate , Tumor Cells, Cultured
20.
J Cancer Res Clin Oncol ; 122(12): 716-22, 1996.
Article in English | MEDLINE | ID: mdl-8954168

ABSTRACT

Vaccinia melanoma oncolysate (VMO) prepared with recombinant vaccinia virus encoding the gene of murine granulocyte/macrophage-colony-stimulating factor (GM-CSF) was tested for its therapeutic effect on melanoma pulmonary metastasis. The murine pulmonary metastasis model was established by injecting 2 x 10(5) B16F10 melanoma cells into the tail vein of a C57BL/6 mouse. Intraperitoneal injection of VMO was performed in tumor-bearing mice 3 and 10 days after B16F10 cell inoculation. The results showed that treatment with VMO prepared with GM-CSF-gene-encoded vaccinia virus (GM-CSFVMO) significantly decreased the number of murine pulmonary metastases and prolonged the survival of the tumor-bearing mice. Lymphocytes isolated from fresh blood and spleen of GM-CSFVMO-treated mice showed higher cytolytic activity against B16F10 melanoma cells when compared with lymphocytes from the mice of other treatment groups. Natural killer activity remained unchanged in the GM-CSFVMO-treated group. Cytotoxic activities of peritoneal macrophages were found to be greatly elevated in mice treated with GM-CSFVMO. Further study illustrated that the increased tumor necrosis factor and nitric oxide release from macrophages may contribute to their cytotoxic effects. These results suggest that the tumor oncolysate vaccine prepared with GM-CSF-gene-encoded vaccinia virus has a potent therapeutic effect on tumor metastasis through the efficient induction of antitumor immunity of the host, mainly through the cytotoxic effects of cytotoxic T lymphocytes and macrophages.


Subject(s)
Cancer Vaccines/pharmacology , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Immunotherapy, Active/methods , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/secondary , Melanoma, Experimental/therapy , Vaccinia virus/genetics , Animals , Cancer Vaccines/genetics , Chlorocebus aethiops , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Macrophage Activation/immunology , Macrophages, Peritoneal/immunology , Male , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/metabolism , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...