Network


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

Hotspot


Dive into the research topics where Yanjun Wen is active.

Publication


Featured researches published by Yanjun Wen.


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.


ACS Nano | 2010

Efficient inhibition of C-26 colon carcinoma by VSVMP gene delivered by biodegradable cationic nanogel derived from polyethyleneimine.

Maling Gou; Ke Men; Juan Zhang; Yuhua Li; Jia Song; Shan Luo; Huashan Shi; Yanjun Wen; Gang Guo; Meijuan Huang; Xia Zhao; Zhiyong Qian; Yuquan Wei

Biodegradable cationic nanoparticles have promising application as a gene delivery system. In this article, heparin-polyethyleneimine (HPEI) nanogels were prepared, and these nanogels were developed as a nonviral gene vector. The transfection efficiency of HPEI nanogels was comparable with that of PEI25K, while the cytotoxicity was lower than that of PEI2K and much lower than that of PEI25K in vitro. These HPEI nanogels also had better blood compatibility than PEI25K. After intravenous administration, HPEI nanogels degraded, and the degradation products were excreted through urine. The plasmid expressing vesicular stomatitis virus matrix protein (pVSVMP) could be efficiently transfected into C-26 colon carcinoma cells by HPEI nanogels in vitro, inhibiting the cell proliferation through apoptosis induction. Intraperitoneal injection of pVSVMP/HPEI complexes efficiently inhibited the abdominal metastases of C-26 colon carcinoma through apoptosis induction (mean tumor weight in mice treated with pVSVMP/HPEI complex = 0.93 g and in control mice = 3.28 g, difference = 2.35 g, 95% confidence interval [CI] = 1.75-2.95 g, P < 0.001) and prolonged the survival of treated mice. Moreover, intravenous application of pVSVMP/HPEI complexes also inhibited the growth of pulmonary metastases of C-26 colon carcinoma through apoptosis induction. The HPEI nanogels delivering pVSVMP have promising application in treating colon carcinoma.


Smart Materials and Structures | 2007

A thermosensitive hydrogel based on biodegradable amphiphilic poly(ethylene glycol)–polycaprolactone–poly(ethylene glycol) block copolymers

Changyang Gong; Zhiyong Qian; CaiBing Liu; Meijuan Huang; YingChun Gu; Yanjun Wen; Bing Kan; Ke Wang; Mei Dai; XingYi Li; Maling Gou; Mingjing Tu; Yuquan Wei

A series of low molecular weight poly(ethylene glycol)–polycaprolactone–poly(ethylene glycol) (PEG–PCL–PEG) biodegradable block copolymers were successfully synthesized using isophorone diisocyanate (IPDI) as the coupling agent, and were characterized using 1H NMR and Fourier transform infrared spectroscopy. The aqueous solutions of the PEG–PCL–PEG copolymers displayed a special thermosensitive gel–sol transition when the concentration was above the corresponding critical gel concentration. Gel–sol phase diagrams were recorded using the test-tube-inversion method; they depended on the hydrophilic/hydrophobic balance in the macromolecular structure, as well as some other factors, including the heating history, volume, and the ageing time of the copolymer aqueous solutions and dissolution temperature of the copolymers. As a result, the gel–sol transition temperature range could be altered, which might be very useful for application in injectable drug delivery systems.


Molecular Cancer Therapeutics | 2008

Human α-defensin-1 inhibits growth of human lung adenocarcinoma xenograft in nude mice

Ning Xu; Yongsheng Wang; Wu-bin Pan; Bo Xiao; Yanjun Wen; Xiancheng Chen; Lijuan Chen; Hongxin Deng; Jia You; Bing Kan; Afu Fu; Dan Li; Xia Zhao; Yuquan Wei

Human α-defensin-1 (HNP1), a small antimicrobial peptide, shows cytotoxicity to tumor cells in vitro and inhibitory activity for pathologic neovascularization in vivo. Here, we did a gene therapy with a plasmid that expresses a secretable form of HNP1 for assaying its antitumor activity. The expression and secretion of HNP1 were determined by reverse transcription-PCR and ELISA in vitro. We found that expression of HNP1 in A549 tumor cells caused significant growth inhibition. This effect is most likely cell autonomous, as a significant amount of recombinant HNP1 protein was found to be accumulated in the cytoplasm by immunohistochemical staining using an anti-HNP1 antibody and the supernatant containing secreted HNP1 failed to produce any noticeable antitumor activity. Flow cytometry and Hoechst 33258 staining showed that the number of apoptotic cells among the A549 cells expressing recombinant HNP1 proteins was significantly greater than that of the nontransfected control cultures, suggesting that this growth-inhibitory activity was due to an apoptotic mechanism triggered by the intracellular HNP1. The antitumor activity of intracellularly expressed HNP1 was also shown in vivo. Decreased microvessel density and increased lymphocyte infiltration were observed in tumor tissue from HNP1-treated mice through histologic analysis. These results indicate that intracellularly expressed HNP1 induces tumor cell apoptosis, which inhibits tumor growth. The antiangiogenesis effect of HNP1 may contribute to its inhibitory activity in vivo, and HNP1 might involve the host immune response to tumor. These findings provide a rationale for developing HNP1-based gene therapy for cancer. [Mol Cancer Ther 2008;7(6):1588–97]


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.


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.


International Journal of Cancer | 2004

Induction of apoptosis and tumor regression by vesicular stomatitis virus in the presence of gemcitabine in lung cancer

Qiu Li; Yuquan Wei; Yanjun Wen; Xia Zhao; Tian L; Li Yang; Yong-Qiu Mao; Bing Kan; Yang Wu; Zhenyu Ding; Hongxin Deng; Jiong Li; Yan Luo; Hong-Li Li; Qiu-Ming He; Jing-Mei Su; Fei Xiao; Chun-hua Zou; Chunhua Fu; Xing-Jiang Xie; Tao Yi; Guang-Hong Tan; Lian Wang; Jing Chen; Jian Liu; Zhen-Nan Gao

Vesicular stomatitis virus (VSV) has been shown to replicate rapidly in vitro and kill selectively a variety of tumor cell lines. The present study was designed to determine whether gemcitabine potentiates the antitumor activity of VSV in vitro and in vivo. A549 human lung adenocarcinoma cells and LLC Lewis lung carcinoma cells were treated with VSV (0.1–10 plaque‐forming units per cell) plus gemcitabine (20 nM to 20 μM). Mice bearing A549 or LLC were treated with VSV (5 × 104 to 1 × 108 plaque‐forming units) daily for 5 days plus gemcitabine (5–125 mg/kg/day) once every 3 days for 4 times. Induction of apoptosis and effects on growth inhibition were assessed. The lung cancer cells treated with VSV plus gemcitabine displayed the apparently increased apoptotic cells compared with treatment with VSV or gemcitabine alone. The combined treatment with VSV plus gemcitabine induced the apparent antitumor activity with complete regression of the established lung cancer in both A549 and LLC lung cancer models and augmented the induction of apoptosis in lung cancer cells in vivo as well. This study suggests that the combined treatment with VSV plus gemcitabine may augment the induction of apoptosis in lung cancer cells in vitro and in vivo, and that the augmented antitumor activity in vivo may result from the increased induction of apoptosis in lung cancer cells. The present findings may be of importance to the further exploration of the potential application of this combined approach in the treatment of lung cancer.

Collaboration


Dive into the Yanjun Wen'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge