Network


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

Hotspot


Dive into the research topics where Huajun Jin is active.

Publication


Featured researches published by Huajun Jin.


International Journal of Biological Sciences | 2016

New Strategies for the Treatment of Solid Tumors with CAR-T Cells

Hao Zhang; Zhen-long Ye; Zhen-gang Yuan; Zheng-qiang Luo; Huajun Jin; Qijun Qian

Recent years, we have witnessed significant progresses in both basic and clinical studies regarding novel therapeutic strategies with genetically engineered T cells. Modification with chimeric antigen receptors (CARs) endows T cells with tumor specific cytotoxicity and thus induce anti-tumor immunity against malignancies. However, targeting solid tumors is more challenging than targeting B-cell malignancies with CAR-T cells because of the histopathological structure features, specific antigens shortage and strong immunosuppressive environment of solid tumors. Meanwhile, the on-target/off-tumor toxicity caused by relative expression of target on normal tissues is another issue that should be reckoned. Optimization of the design of CAR vectors, exploration of new targets, addition of safe switches and combination with other treatments bring new vitality to the CAR-T cell based immunotherapy against solid tumors. In this review, we focus on the major obstacles limiting the application of CAR-T cell therapy toward solid tumors and summarize the measures to refine this new cancer therapeutic modality.


Journal of Cancer | 2015

Argonaute 2: A Novel Rising Star in Cancer Research

Zhen-long Ye; Huajun Jin; Qijun Qian

AGO2 (Argonaute 2, EIF2C2) is the only member in AGO family with catalytic activity and of extreme importance during small RNAs guided gene silencing processes. The structural investigations have provided insights into details and functional mechanisms of the four major domains within AGO2. As a multifunction player, AGO2 has been revealed involved in tumorgenesis through miRNAs-dependent or independent ways. And nowadays, AGO2 has also been more importantly found ectopically over-expressed in carcinomas and closely associated with aspects of cancers in means of interacting with well-known tumor factors. Here, we provide a review on structural insights, functional mechanisms, novel roles and relationship with carcinomas of AGO2.


Experimental hematology & oncology | 2015

The combination of dendritic cells-cytotoxic T lymphocytes/cytokine-induced killer (DC-CTL/CIK) therapy exerts immune and clinical responses in patients with malignant tumors.

Ying Wang; Zenghui Xu; Fuping Zhou; Yan Sun; Jingbo Chen; Linfang Li; Huajun Jin; Qijun Qian

BackgroundThe clinical trials using immunotherapy have been performed for the treatment of variety of malignant tumors. However, large-scale meta-analysis of combined DC-CTL/CIK therapy on immune and clinical response in patients has not been well studied yet. The purpose of this study is to investigate the role of DC-CTL/CIK therapy and evaluate the changes of immune indicators and tumor serological markers bothxa0at anxa0individual level and at a system level, which is an important basis for immunotherapy as well as prognosis estimation.MethodsThree cohorts were designed to estimate therapeutic effects on patients with malignant tumors. Tumor serological markers were detected pre- and post-treatment by immunoradiometric methods using commercially available diagnostic kits. Lymphocyte subsets were identified by flow cytometry. The quality of life was assessed by EORTC QLQ-C30 questionnaire.ResultsIn this study, we found out that Tregs was significantly reduced after transfusion of DC-CTL/CIK cells companied by decreasing serological tumor markers including AFP, CA199 and CA242 in primary liver cancer and CA724 in gastric cancer. A system-level analysis showed that lower percentages of Tregs were detected in patients with long-lasting courses of immunotherapy. Strikingly, a tumor progression indicator, myeloid-derived suppressor cells (MDSC), was dramatically decreased in patients after DC-CTL/CIK treatment. These results suggested that DC-CTL/CIK therapy improves immune functions and the quality of life post-treatment versus pre-therapy, indicating that DC-CTL/CIK therapy might block the deterioration of invasive cancers in these patients.ConclusionsThis study demonstrated that DC-CTL/CIK therapy could reduce Tregs, MDSCs, and several crucial serological tumor markers in particular tumors, and improve the function of T cells immune systems and the quality of life in patients with malignant tumor.


Acta Pharmacologica Sinica | 2015

Argonaute 2 promotes angiogenesis via the PTEN/VEGF signaling pathway in human hepatocellular carcinoma

Zhen-long Ye; Yao Huang; Linfang Li; Hai-li Zhu; Haixia Gao; Hui Liu; Saiqun Lv; Zenghui Xu; Luo-ning Zheng; Tao Liu; Jing-lei Zhang; Huajun Jin; Qijun Qian

Aim:Argonaute2 (AGO2) protein is the active part of RNA-induced silencing complex, cleaving the target mRNA strand complementary to their bound siRNA. An increasing number of miRNAs has been identified as essential to angiogenesis of hepatocellular carcinoma (HCC). In this study we investigated how AGO2 affected HCC angiogenesis.Methods:Human HCC cell lines HepG2, Hep3B, Huh7, SMMC-7721, Bel-7404, MHCC97-H and LM-3, and human umbilical vein endothelial cells (HUVEC) were tested. The expression of AGO2 in HCC cells was knocked down with siRNA and restored using recombinant adenovirus expressing Ago2. The levels of relevant mRNAs and proteins were examined using RT-PCR, Western blot and EILSA. Nude mice were implanted with Huh7 or SMMC-7721 cells, and tumor volumes were measured. After the mice were euthanized, the xenograft tumors were used for immunohistological analysis.Results:In 6 HCC cell lines, AGO2 protein expression was significantly correlated with VEGF expression (r=+0.79), and with VEGF secretion (r=+0.852). Knockdown of Ago2 in Huh7 cells and SMMC-7721 cells substantially decreased VEGF expression, whereas the restoration of AGO2 reversed both VEGF expression and secretion. Furthermore, knockdown of Ago2 significantly up-regulated the expression of PTEN (a tumor suppressor involved in the inhibition of HCC angiogenesis), and vice versa. Moreover, the specific PTEN inhibitor bisperoxovanadate (7, 14, 28 nmol/L) dose-dependently restored the expression of VEGF and the capacity of HCC cells to induce HUVECs to form capillary tubule structures. In the xenograft nude mice, knockdown of Ago2 markedly suppressed the tumor growth and decreased PTEN expression and CD31-positive microvascular in the xenograft tumors.Conclusion:A direct relationship exists between the miRNA processing machinery AGO2 and HCC angiogenesis that is mediated by the AGO2/PTEN/VEGF signaling pathway. The results suggest the high value of Ago2 knockdown in anti-angiogenesis therapy for HCC.


Expert Opinion on Therapeutic Patents | 2009

Humanized anti-EphB4 antibodies for the treatment of carcinomas and vasculogenesis-related diseases

Zenghui Xu; Huajun Jin; Qijun Qian

The invention provides human, humanized or chimeric versions of anti-EphB4 mouse monoclonal antibodies that bind to the human EphB4 receptor tyrosine kinase. The described anti-EphB4 antibodies are derived from two murine mAbs #47 and #131 through framework shuffling and include those of the IgGl, IgG2, IgG3 or IgG4 human isotype. The patent further relates to pharmaceutical compositions, immunotherapeutic compositions and methods using therapeutic antibodies that bind to the human EphB4 antigen and that may induce phosphorylation and degradation of EphB4 and mediate antigen-dependent cell-mediated-cytotoxicity, complement-dependent cell-mediated cytotoxicity and/or apoptosis for the treatment of human malignancies and vasculogenesis-related disorders and diseases.


Tumor Biology | 2017

Mesothelin-targeting chimeric antigen receptor–modified T cells by piggyBac transposon system suppress the growth of bile duct carcinoma:

Jie-Ying Xu; Zhen-long Ye; Duqing Jiang; Jiangchuan He; Yong-Mei Ding; Linfang Li; Saiqun Lv; Ying Wang; Huajun Jin; Qijun Qian

Chimeric antigen receptor modified T cell–based immunotherapy is revolutionizing the field of cancer treatment. However, its potential in treating bile duct carcinoma has not been fully explored. Herein, we developed the second-generation mesothelin-targeting chimeric antigen receptor–modified T cells with the 4-1BB co-stimulatory module by the piggyBac transposon system. Mesothelin-targeting chimeric antigen receptor was expressed by 66.0% of mesothelin-targeting chimeric antigen receptor–modified T cells post electrophoretic transfection and stimulation with K562-meso cells; the expressions of activation markers were tested by flow cytometry assay and showed greater activation of mesothelin-targeting chimeric antigen receptor–modified T cells than control T cells (CD107α: 71.9% vs 48.6%; CD27: 92.1% vs 61.8%; CD137: 55.5% vs 8.4%; CD28: 98.0% vs 82.1%; CD134: 37.5% vs 10.4%). Furthermore, mesothelin-targeting chimeric antigen receptor–modified T cells exerted cytotoxicity toward mesothelin-expressing EH-CA1b and EH-CA1a cells in an effector-to-target ratio-dependent manner, while leaving mesothelin-negative GSC-SD and EH-GB1 cells and normal liver L02 cells almost unharmed. Mesothelin-targeting chimeric antigen receptor–modified T cells secreted cytokines at higher levels when co-cultured with mesothelin-positive EH-CA1a and EH-CA1b cells than with mesothelin-negative GSC-SD and EH-GB1 cells. Enhanced cytotoxicity and cytokine secretion of mesothelin-targeting chimeric antigen receptor–modified T cells compared to control T cells were also observed when co-cultured with 293-meso cells (interferon γ: 85.1%u2009±u20091.47% vs 8.3%u2009±u20092.50%, pu2009=u20090.000; tumor necrosis factor α: 90.9%u2009±u20094.67% vs 18.5%u2009±u20093.62%, pu2009=u20090.0004; interleukin 2: 60.8%u2009±u20092.00% vs 15.6%u2009±u20092.06%, pu2009=u20090.002; interleukin 6: 6.4%u2009±u20092.95% vs 1.7%u2009±u20090.63%, pu2009=u20090.055). In addition, mesothelin-targeting chimeric antigen receptor–modified T cells showed greater inhibitory and proliferative capability than control T cells within EH-CA1a cell xenografts. This study shows the potential of mesothelin-targeting chimeric antigen receptor–modified T cells in treating bile duct carcinoma.


Journal of Cancer | 2018

Cancer vaccine: learning lessons from immune checkpoint inhibitors

Zhen-long Ye; Qiming Qian; Huajun Jin; Qijun Qian

Cancer vaccines have been exclusively studied all through the past decades, and have made exceptional achievements in cancer treatment. Few cancer vaccines have been approved by the US Food and Drug Administration (FDA), for instance, Provenge, which was approved for the treatment of prostate carcinoma in 2012. Moreover, more recently, T-VEC got approval for the treatment of melanoma. While, the overall therapeutic effects of cancer vaccines have been taken into consideration as below expectations, low antigenicity of targeting antigen and tumor heterogeneity are the two key limiting barriers encountered by the cancer vaccines. Nonetheless, recent developments in cancer immune-therapies together with associated technologies, for instance the unparalleled achievements bagged by immune checkpoint inhibitor based therapies and neo-antigen identification tools, envisage potential improvements in cancer vaccines in respect to the treatments of malignancies. This review brings forth measures for the purpose of refining therapeutic cancer vaccines by learning lessons from the success of PD-1 inhibitor based immune-therapies.


Journal of Cancer | 2018

Immunotherapy of patient with hepatocellular carcinoma using cytotoxic T lymphocytes ex vivo activated with tumor antigen-pulsed dendritic cells

Ying Wang; Xijing Yang; Yi Yu; Zenghui Xu; Yan Sun; Hui Liu; Jingbo Cheng; Min Liu; Bibo Sha; Linfang Li; Na Ding; Zhong Li; Huajun Jin; Qijun Qian

Purpose The aim of this study was to evaluate the clinical response of immunotherapy with dendritic cell-cytotoxic T lymphocytes (DC-CTLs) in patients with hepatocellular carcinoma (HCC). Method Sixty-eight patients with a confirmed diagnosis of HCC and who received follow-up until December 2015 were enrolled. We measured immune phenotypes of DCs and activated T cells using flow cytometry and clinical indexes using an electrochemiluminescence method. Results DCs exhibited up-regulation of the maturation markers CD83, CD80, CD11c, and CD86 on day8. Levels of IFN-γ and TNF-α were higher in the DCs pulsed with tumor-associated antigens (TAAs) than in DCs with a non-proliferative recombinant adenovirus. The percentage of regulatory T cells (Tregs) decreased in patients after DC-CTLs therapy. In addition, serum levels of AFP, AFP-L3, ALT, and CA19-9 were significantly reduced in these patients. Quality of life was improved, especially on physical functioning scales. Median overall survival (OS) and progression-free survival (PFS) were 8.2 months and 4.3 months, respectively, for the control group and 12.8 months and 9 months, respectively, for the DC-CTL group. Patients treated with DC-CTLs therapy showed a statistically significant PFS and OS curve (OS: p=0.016; PFS: p<0.0001). In addition, no serious adverse reactions were observed. Conclusion This study indicated that Tregs, as well as serum levels of AFP, AFP-L3, ALT, and CA19-9, which were correlated with a poor prognosis, decreased after DC-CTL treatments. The OS, PFS and the quality of life of HCC patients partially improved.


Journal of Cancer | 2017

Activation or suppression of the immune response mediators in biliary tract cancer (BTC) patients: a systematic review and meta-analysis

Ying Wang; Min Ding; Qian Zhang; Jinghan Wang; Xijing Yang; Fuping Zhou; Linfang Li; Zhengang Yuan; Huajun Jin; Qijun Qian

Background: Infiltration of immune cells and immune microenvironment determine the proliferative activity of the tumor and metastasis. The aim of this study was to analyze the influence of activation or suppression of the immune response mediators on the prognosis of biliary tract cancer (BTC). Methods: We searched Pubmed, Web of Science, Embase and The Cochrane Library for relevant literatures until June 2016. The quality of studies was assessed by QUADAS-2 and NOS tools. Forest and funnel plots and all statistical analyses were generated by using Review Manager 5.3. The bias of included studies was estimated by Eggers test using Meta R package. Results: A total of 2339 patients from 12 studies were finally enrolled in this meta-analysis. Patients with high expression of immune active factors, intraepithelial tumor-infiltrating CD4+ , CD8+, and Foxp3+ T lymphocytes, MHC I, NKG2D, showed a better overall survival (OS) than those with low expression (HR=0.52, 95% CI=0.41-0.67, P<0.00001). On the contrary, the high expression of immune suppressive factors (CD66b+ neutrophils, Neutrophil-lymphocyte ratio, Intratumoral IL-17+ cells and PD-1+/CD8+ TILs) was significantly associated with poor OS (HR=1.79, 95% CI=1.44-2.22, P<0.00001). A further analysis of therapies targeting tumor microenvironment modulation showed that the median progression free survival (PFS) for BTC patients who received adjuvant immunotherapy was longer than those who received surgery or chemotherapy alone, and the estimated pooled mean difference demonstrated a highly significant improvement (MD =2.33; 95% CI: 0.63-4.02, P=0.007). The total effect of PFS and OS was statistically longer in experimental group, compared to patients in control groups, respectively (PFS: RR=1.25; 95% CI: 1.08-1.46, P=0.004; OS: RR=1.16; 95% CI: 1.07-1.27, P=0.0006). In subgroup meta-analysis of studies on 6-, 12- and 18-month PFS and OS, it showed that adjuvant immunotherapy could improve the 6-month PFS (RR=1.23; 95% CI: 1.05-1.44, P=0.009), and 6-month OS (RR=1.17; 95% CI: 1.06-1.30, P=0.002). Conclusions: So given the above issue, our meta-analysis confirmed that the level of immune mediators could be a predicative factor for prognosis of BTC patients, and immunotherapy regimens by modulating the tumor microenvironment was superior for enhancing median PFS, 6-month PFS and OS.


Cell Death and Disease | 2018

Antitumor activity of EGFR-specific CAR T cells against non-small-cell lung cancer cells in vitro and in mice

He Li; Yao Huang; Duqing Jiang; Lianzhen Cui; Zhou He; Chao Wang; Zhiwei Zhang; Hai-li Zhu; Yong-Mei Ding; Linfang Li; Qiang Li; Huajun Jin; Qijun Qian

Effective control of non-small-cell lung cancer (NSCLC) remains clinically challenging, especially during advanced stages of the disease. This study developed an adoptive T-cell treatment through expression of a chimeric antigen receptor (CAR) to target human epidermal growth factor receptor (EGFR) in NSCLC. We optimized the non-viral piggyBac transposon system to engineer human T cells for the expression of EGFR-CAR, consisting of EGFR scFv, transmembrane domain, and intracellular 4-1BB-CD3ζ signaling domains. The modified CAR T cells exhibited expansion capability and anticancer efficacy in a time- and antigen-dependent manner in vitro as well as regression of EGFR-positive human lung cancer xenografts in vivo. EGFR-CAR T therapy is a promising strategy to improve the efficacy and potency of the adoptive immunotherapy in NSCLC. Moreover, EGFR-CAR T therapy could become a clinical application for NSCLC patients in the future.

Collaboration


Dive into the Huajun Jin's collaboration.

Top Co-Authors

Avatar

Qijun Qian

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Linfang Li

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Saiqun Lv

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Ying Wang

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zenghui Xu

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hai-li Zhu

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hui Liu

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yao Huang

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Fuping Zhou

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jiangchuan He

Zhejiang Sci-Tech University

View shared research outputs
Researchain Logo
Decentralizing Knowledge