Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Tian L is active.

Publication


Featured researches published by Tian L.


Nature Medicine | 2000

Immunotherapy of tumors with xenogeneic endothelial cells as a vaccine.

Yu Quan Wei; Qing Ru Wang; Xia Zhao; Li Yang; Tian L; You Lu; Bin Kang; Chong Jiu Lu; Mei Juan Huang; Yan Yan Lou; Fei Xiao; Qiu Ming He; Jing Mei Shu; Xing Jiang Xie; Yun Qiu Mao; Shong Lei; Feng Luo; Li Qun Zhou; Chong En Liu; Hao Zhou; Yu Jiang; Feng Peng; Liang Ping Yuan; Qiu Li; Yang Wu; Liu J

The breaking of immune tolerance against autologous angiogenic endothelial cells should be a useful approach for cancer therapy. Here we show that immunotherapy of tumors using fixed xenogeneic whole endothelial cells as a vaccine was effective in affording protection from tumor growth, inducing regression of established tumors and prolonging survival of tumor-bearing mice. Furthermore, autoreactive immunity targeting to microvessels in solid tumors was induced and was probably responsible for the anti-tumor activity. These observations may provide a new vaccine strategy for cancer therapy through the induction of an autoimmune response against the tumor endothelium in a cross-reaction.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Immunogene therapy of tumors with vaccine based on Xenopus homologous vascular endothelial growth factor as a model antigen

Yu Quan Wei; Mei Juan Huang; Li Yang; Xia Zhao; Tian L; You Lu; Jing Mei Shu; Chong Jiu Lu; Ting Niu; Bin Kang; Yun Qiu Mao; Fen Liu; Wen Yj; Shong Lei; Feng Luo; Li Qun Zhou; Feng Peng; Yu Jiang; Liu J; Hao Zhou; Qing Ru Wang; Qiu Ming He; Fei Xiao; Yan Yan Lou; Xing Jiang Xie; Qiu Li; Yang Wu; Zhen Yu Ding; Bin Hu; Min Hu

Overcoming immune tolerance of the growth factors associated with tumor growth should be a useful approach to cancer therapy by active immunity. We used vascular endothelial growth factor (VEGF) as a model antigen to explore the feasibility of the immunogene tumor therapy with a vaccine based on a single xenogeneic homologous gene, targeting the growth factors associated with angiogenesis. To test this concept, we constructed a plasmid DNA encoding Xenopus homologous VEGF (XVEGF-p) and control vectors. We found that immunogene tumor therapy with a vaccine based on XVEGF was effective at both protective and therapeutic antitumor immunity in several tumor models in mice. VEGF-specific autoantibodies in sera of mice immunized with XVEGF-p could be found in Western blotting analysis and ELISA assay. The purified immunoglobulins were effective at the inhibition of VEGF-mediated endothelial cell proliferation in vitro, and at antitumor activity and the inhibition of angiogenesis by adoptive transfer in vivo. The elevation of VEGF in the sera of the tumor-bearing mice could be abrogated with XVEGF-p immunization. The antitumor activity and production of VEGF-specific autoantibodies, significantly elevated IgG1 and IgG2b, could be abrogated by the depletion of CD4+ T lymphocytes. The observations may provide a vaccine strategy for cancer therapy through the induction of autoimmunity against the growth factors associated with tumor growth in a cross reaction with single xenogeneic homologous gene and may be of importance in the further exploration of the applications of other xenogeneic homologous genes identified in human and other animal genome sequence projects in cancer therapy.


Journal of Cancer Research and Clinical Oncology | 2002

Induction of apoptosis by norcantharidin in human colorectal carcinoma cell lines: involvement of the CD95 receptor/ligand

Feng Peng; Yuquan Wei; Tian L; Ling-lin Yang; Xia Zhao; You Lu; Yun Qiu Mao; Bing Kan; Song Lei; G.-S. Wang; Yu Jiang; Qing Ru Wang; Feng Luo; Li-qun Zou; Liu J

Abstract.Purpose: Cantharidin, a natural toxin, is the active substance of mylabris and has antitumor effects in man. Norcantharidin, the demethylated analogue of cantharidin, has been used in the treatment of patients with primary hepatoma and those with leukopenia in China. The present study was designed to investigate whether norcantharidin exerts cytotoxic activity against colorectal cancer cells by inducing apoptosis and to examine the possible mechanism in the phenomenon. Methods: Inhibition of proliferation of norcantharidin on Colo205, HT-29, and SW480 colorectal cancer cells was determined by the trypan blue dye exclusion test. Apoptosis of norcantharidin-treated cells was determined by morphological analysis, agarose gel DNA electrophoresis, and quantitated by flow cytometry after staining with propidium iodide. Cell cycle and the cell surface expression of the CD95/CD95 ligand were evaluated by flow cytometry. Caspase 8-like protease and protein phosphatase 1 and 2A activities were also analyzed. Results: Treatment with norcantharidin of colorectal cancer cells not only inhibited cell proliferation, but also induced apoptosis. Norcantharidin induced apoptosis mainly in two phases: rapid apoptosis in S-phase cells and delayed apoptosis in G2/M arrested cells. Treatment with norcantharidin resulted in an upregulation of the CD95 receptor and CD95 ligand on the cell surface. Furthermore, stimulation with anti-CD95 monoclonal antibody (mAb) resulted in further induction of apoptosis after treatment with norcantharidin. In addition, the apoptosis-inducing effect of norcantharidin was almost completely inhibited by anti-CD95 ligand mAb. Norcantharidin-treated cells showed the activation of caspase 8. Both zVAD-FMK (a broad range caspase inhibitor) and IETD-FMK (a caspase-8 inhibitor) showed apparent inhibition of the apoptosis-inducing effect. Norcantharidin did not show an inhibitory effect on protein phosphatase. Conclusions: These results suggest that norcantharidin triggers apoptosis in colorectal cancer cell lines via the activation of the CD95 receptor/ligand system, and that this agent may be useful for developing new therapeutic regimens for the treatment of colorectal carcinoma.


Gene Therapy | 2002

A gene therapy for cancer based on the angiogenesis inhibitor, vasostatin

Fei Xiao; Yuquan Wei; Li Yang; Xia Zhao; Tian L; Zhenyu Ding; Yuan S; Yan Yan Lou; Fen Liu; Yanjun Wen; Jiong Li; Hongxin Deng; Bin Kang; Yun Qiu Mao; Song Lei; Qiu-Ming He; Jing-Mei Su; You Lu; Ting Niu; Hou J; Meijuan Huang

The growth and persistence of solid tumors and their metastasis are angiogenesis-dependent. Vasostatin, the N-terminal domain of calreticulin inclusive of amino acids 1–180, is a potent angiogenesis inhibitor. To investigate whether intramuscular administration of vasostatin gene has the antitumor activity in mouse tumor models, we constructed a plasmid DNA encoding vasostatin and a control vector. Production and secretion of vasostatin protein by COS cells transfected with the plasmid DNA encoding vasostatin (pSecTag2B-vaso) were confirmed by Western blot analysis and ELISA. Conditioned medium from vasostatin-transfected COS cells apparently inhibited human umbilical vein endothelial cell (HUVEC) and mouse endothelial cell (SVEC4-10) proliferation, compared with conditioned medium from the COS cells transfected with control vector or non-transfected cells. Treatment with pSecTag2B-vaso twice weekly for 4 weeks resulted in the inhibition of tumor growth and the prolongation of the survival of tumor-bearing mice. The sustained high level of vasostatin protein in serum could be identified in ELISA. Angiogenesis was apparently inhibited in tumor by immunohistochemical analysis. Angiogenesis was also inhibited in the chicken embryo CAM assay and mouse corneal micropocket assay. The increased apoptotic cells were found within the tumor tissues from the mice treated with plasmid DNA encoding vasostatin. Taken together, the data in the present study indicate that the cancer gene therapy by the intramuscular delivery of plasmid DNA encoding vasostatin, is effective in the inhibition of the systemic angiogenesis and tumor growth in murine models. The present findings also provide further evidence of the anti-tumor effects of the vasostatin, and may be of importance for the further exploration of the application of this molecule in the treatment of cancer.


Journal of Immunology | 2003

Immunogene Therapy of Tumors with Vaccine Based on Xenogeneic Epidermal Growth Factor Receptor

You Lu; Yuquan Wei; Tian L; Xia Zhao; Li Yang; Bin Hu; Bin Kan; Yanjun Wen; Feng Liu; Hongxin Deng; Jiong Li; Yong-Qiu Mao; Song Lei; Meijuan Huang; Feng Peng; Yu Jiang; Hao Zhou; Li-qun Zhou; Feng Luo

The breaking of immune tolerance against self epidermal growth factor receptor (EGFr) should be a useful approach for the treatment of receptor-positive tumors with active immunization. To test this concept, we constructed a plasmid DNA encoding extracellular domain of xenogeneic (human) EGFr (hEe-p) or corresponding control mouse EGFr (mEe-p) and empty vector (c-p). Mice immunized with hEe-p showed both protective and therapeutic antitumor activity against EGFr-positive tumor. Sera isolated from the hEe-p-immunized mice exhibited positive staining for EGFr-positive tumor cells in flow cytometric analysis and recognized a single 170-kDa band in Western blot analysis. Ig subclasses responded to rEGFr proteins were elevated in IgG1, Ig2a, and Ig2b. There was the deposition of IgG on the tumor cells. Adoptive transfer of the purified Igs showed the antitumor activity. The increased killing activity of CTL against EGFr-positive tumor cells could be blocked by anti-CD8 or anti-MHC class I mAb. In vivo depletion of CD4+ T lymphocytes could completely abrogate the antitumor activity, whereas the depletion of CD8+ cells showed partial abrogation. The adoptive transfer of CD4-depleted (CD8+) or CD8-depleted (CD4+) T lymphocytes isolated from mice immunized with hEe-p vaccine showed the antitumor activity. In addition, the increase in level of both IFN-γ and IL-4 was found. Taken together, these findings may provide a new vaccine strategy for the treatment of EGFr-positive tumors through the induction of the autoimmune response against EGFr in a cross-reaction between the xenogeneic homologous and self EGFr.


Clinical Cancer Research | 2005

Improved Therapeutic Effectiveness by Combining Recombinant CXC Chemokine Ligand 10 with Cisplatin in Solid Tumors

Gang Li; Tian L; Jian-mei Hou; Zhenyu Ding; Qiu-Ming He; Ping Feng; Yanjun Wen; Fei Xiao; Bing Yao; Ru Zhang; Feng Peng; Yu Jiang; Feng Luo; Xia Zhao; Lin Zhang; Qiao Zhou; Yuquan Wei

Purpose: CXC chemokine ligand 10 (CXCL10) is a potent inhibitor of angiogenesis. We wonder whether the combination of CXCL10 with cisplatin would improve the therapeutic antitumor efficacy. Experiment Design: We evaluated the antitumor activity of the combination therapy in the immunocompetent C57BL/6 and BALB/c mice bearing LL/2 Lewis lung cancer and CT26 colon adenocarcinoma, respectively. Mice were treated with either CXCL10 s.c. at 25 μg per kg per day once daily for 30 days, cisplatin cycled twice (5 mg/kg i.p. on days 14 and 21 after the initiation of CXCL10), or both agents together. Tumor volume and survival time were observed. Antiangiogenesis of CXCL10 in vivo were determined by alginate capsule models and CD31 immunohistochemistry. Histologic analysis and assessment of apoptotic cells were also conducted in tumor tissues. Results: CXCL10 + cisplatin reduced tumor growth in LL/2 and CT26 tumor model, respectively, more effectively, although cisplatin or CXCL10 individually resulted in suppression of tumor growth and improved survival time of tumor-bearing mice. CXCL10 successfully inhibited angiogenesis as assessed by alginate model and CD31 (P < 0.05). Histologic analysis of tumors exhibited that CXCL10 in combination with cisplatin led to the increased rate of apoptosis, tumor necrosis, and elevated lymphocyte infiltration. Conclusions: Our data suggest that the combination of CXCL10, a well-tolerated angiogenesis inhibitor, with cisplatin can enhance the antitumor activity. The present findings may be of importance to the further exploration of the potential application of this combined approach in the treatment of lung and colon carcinoma.


European Journal of Immunology | 2004

Active immunotherapy of tumors with a recombinant xenogeneic endoglin as a model antigen

Guang-Hong Tan; Yuquan Wei; Tian L; Xia Zhao; Li Yang; Jiong Li; Qiu Ming He; Yang Wu; Yanjun Wen; Tao Yi; Zhenyu Ding; Bin Kan; Yong-Qiu Mao; Hongxin Deng; Hong-Li Li; Chun-Hua Zhou; Chunhua Fu; Fei Xiao; Xiao-wei Zhang

Angiogenesis play a critical role in tumor growth and metastasis. Increasing evidence suggests that endoglin is a powerful marker of angiogenesis in solid malignancies. Thus, breaking of immune tolerance of self‐endoglin‐associated angiogenesis is an attractive approach to cancer therapy. To test this concept, we recombined the extracellular domains of porcine endoglin, and used it as a xenogeneic vaccine. We found that immunotherapy with porcine endoglin was effective at both protective and therapeutic anti‐tumor immunity in several mouse tumor models. Autoantibodies against mouseendoglin were identified by Western blot and ELISA. IgG1 and IgG2b were substantially increased. Anti‐endoglin antibody‐producing B cells were detectable by ELISPOT assay. There was endothelial deposition of immunoglobulins within tumors. The anti‐tumor activity was also induced by the adoptive transfer of the purified immunoglobulins. Angiogenesis was apparently inhibited within the tumor tissues and on the alginate beads. The increased apoptotic cells were found within the tumor tissues from the mice treated with porcine endoglin. The anti‐tumor activity and production of autoantibodies againstmouse endoglin could be abrogated by depletion of CD4+ T lymphocytes. Remarkably, no marked toxicity was found in the immunized mice. These observations may provide an alternative rationalstrategy for active cancer immunotherapy.


International Journal of Cancer | 2004

Combination of low‐dose cisplatin and recombinant xenogeneic endoglin as a vaccine induces synergistic antitumor activities

Guang-Hong Tan; Tian L; Yuquan Wei; Xia Zhao; Jiong Li; Yang Wu; Yanjun Wen; Tao Yi; Zhenyu Ding; Bin Kan; Yong-Qiu Mao; Hongxin Deng; Hong-Li Li; Chun-hua Zou; Chunhua Fu

Angiogenesis is critical to the growth and metastasis of solid tumors, and acquired drug resistance is one of the major hindrances to chemotherapy. Thus, we sought a rational strategy using the combination of antiangiogenic biotherapy and chemotherapy for cancer therapy. We explored the efficacy of a strategy combining low‐dose cisplatin and a recombinant xenogeneic endoglin as a protein vaccine, which we previously demonstrated to have effective antiangiogenic effects in several mouse models. We found that both low‐dosage cisplatin and xenogeneic endoglin vaccine individually resulted in effective suppression of tumor growth in 2 tumor models via inhibition of tumor angiogenesis. Remarkably, the combination therapy resulted in not only significant antiangiogenic effects but also additional promotion of tumor cell apoptosis and inhibition of tumor cell proliferation, without any ensuing increase in host toxicity during the course of treatment, which lasted for 6 months. In addition, the combination demonstrated a synergistic relationship, which was shown in all of the synergistic indexes, i.e., tumor volume, angiogenesis, apoptosis and proliferation. Both antibodies and antibody‐producing B cells against mouse self‐endoglin were observed in all mice immunized by the xenogeneic endoglin vaccine (alone and combination), which suggested that low‐dose cisplatin did not suppress the host immune response but potentiated the antitumor activity of the xenogeneic endoglin vaccine. These observations may provide the basis for an effective alternative strategy for cancer therapy in the near future.


Gene Therapy | 2006

Combination of MIG (CXCL9) chemokine gene therapy with low-dose cisplatin improves therapeutic efficacy against murine carcinoma

Zhang R; Tian L; Chen Lj; Fei Xiao; Hou Jm; Xia Zhao; Li G; Yao B; Wen Yj; Jiong Li; Zhang L; Chen Xc; Luo F; Peng F; Jiang Y; Yuquan Wei

MIG (monokine induced by interferon-γ) is a CXC chemokine ligand (CXCL9) that can potently inhibit angiogenesis, and displays thymus-dependent antitumor effects. The effectiveness of a treatment combining gene therapy with plasmid-borne MIG (pORF-MIG) and low-dose cisplatin chemotherapy was determined using colon carcinoma (CT26) and Lewis lung carcinoma (LL/2c) murine models. The program was carried out via intramuscular delivery of pORF-MIG at 100 μg/mouse twice a week for 4 weeks, and/or intraperitoneal delivery of cisplatin at 0.6 mg/kg/mouse every 3 days for 48 days. Tumor volume and survival time were evaluated after treatment. CD31 immunohistochemical staining in tumor tissues and alginate capsule models in vivo was used to evaluate angiogenesis. Induction of apoptosis and cytotoxic T-lymphocyte (CTL) activity were also assessed. The combination of pORF-MIG and low-dose cisplatin produced significant antitumor activity, with complete tumor regression in 4/10 of CT26 colon carcinomas and 3/10 of LL/2c lung carcinomas, low vascularity, in alginate capsules, apparently degraded tumor microvessel density, and increased induction of apoptotic and CTL activities compared with either treatment alone. This study suggests that the combination of pORF-MIG plus cisplatin augments the inhibition of angiogenesis and the induction of apoptosis or CTL activity, all of which enhance antitumor activity. These findings may prove useful in further explorations of the application of combinatorial approaches to the treatment of solid tumors.


Anti-Cancer Drugs | 2005

Synergistic anti-tumor effect of recombinant human endostatin adenovirus combined with gemcitabine.

Yang Wu; Li Yang; Bing Hu; Ji-Yan Liu; Jing-Mei Su; Yan Luo; Zhenyu Ding; Ting Niu; Qiu Li; Xing-Jiang Xie; Yanjun Wen; Tian L; Bing Kan; Yong-Qiu Mao; Yuquan Wei

Endostatin is an important endogenous inhibitor of neovascularization, which has been widely used in anti-angiogenesis therapy for cancer. To fully explore the potential of endostatin, we evaluated the anti-tumor efficacy of the combination of recombinant human endostatin adenovirus and low-dose gemcitabine in nude mice. We injected recombinant human endostatin adenovirus intratumorally plus a low dose of gemcitabine i.p. routinely. The combination treatment produced no side-effects, and resulted in marked suppression in tumor formation and growth of established human lung carcinoma xenografts in nude mice, with decreased microvessel density and increased apoptosis percentage. Our data support the idea of synergistic anti-tumor properties of endostatin plus low-dose chemotherapy against human lung cancer in vivo.

Collaboration


Dive into the Tian L's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liu J

Sichuan University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge