Qi Jing Wang
Sun Yat-sen University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Qi Jing Wang.
Journal of Immunotherapy | 2008
De Sheng Weng; Jun Zhou; Qi Ming Zhou; Ming Zhao; Qi Jing Wang; Li Xi Huang; Yong Qiang Li; Shi Ping Chen; Pei Hong Wu; Jian Chuan Xia
The recurrence of hepatocellular carcinoma (HCC) after minimally invasive therapy is frequent. Adoptive immunotherapy is thought to be an effective method to lower recurrence and metastasis rates of malignant tumors. Therefore, 85 HCC patients after transcatheter arterial chemoembolization and radiofrequency ablation therapy were randomized to immunotherapy group and no adjuvant therapy group. Autologous cytokine-induced killer (CIK) cells were transfused via hepatic artery to the patients. The alteration of levels of lymphocyte subsets in peripheral blood of patients was examined by flow cytometry. All patients were screened by computed tomography every 2 months to observe the tumor recurrent conditions. After CIK cell infusions, the percentages of CD3+, CD4+, CD56+, CD3+CD56+ cells, and CD4+/CD8+ ratio increased from 68.6±11.0%, 31.1±9.0%, 15.6±7.9%, 5.2±3.1%, and 1.1±0.5 to 70.7±10.1%, 33.5±8.0%, 18.4±9.4%, 5.9±2.8%, and 1.3±0.7, respectively (P<0.05); whereas the percentage of CD8+ cells decreased from 31.1±7.8% to 28.6±8.3% (P<0.05). The 1-year and 18-month recurrence rates of the study group were 8.9% and 15.6%, compared with 30.0% and 40.0% of the control group (both P value <0.05). The data suggest that CIK cell transfusion is an effective treatment. It can boost the immunologic function in HCC patients and plays an important role in reducing the recurrence rate of HCC.
PLOS ONE | 2012
Dan Dan Wang; Yi Bing Chen; Ke Pan; Wei Wang; Shi Ping Chen; Ju gao Chen; Jing Jing Zhao; Lin Lv; Qiu Zhong Pan; Yong Qiang Li; Qi Jing Wang; Li Xi Huang; Miao La Ke; Jia He; Jian Chuan Xia
Background The ARID1A gene encodes adenine-thymine (AT)-rich interactive domain-containing protein 1A, which participates in chromatin remodeling. ARID1A has been showed to function as a tumor suppressor in various cancer types. In the current study, we investigated the expression and prognosis value of ARID1A in primary gastric cancer. Meanwhile, the biological role of ARID1A was further investigated using cell model in vitro. Methodology/Principal Findings To investigate the role of ARID1A gene in primary gastric cancer pathogenesis, real-time quantitative PCR and western blotting were used to examine the ARID1A expression in paired cancerous and noncancerous tissues. Results revealed decreased ARID1A mRNA (P = 0.0029) and protein (P = 0.0015) expression in most tumor-bearing tissues compared with the matched adjacent non-tumor tissues, and in gastric cancer cell lines. To further investigate the clinicopathological and prognostic roles of ARID1A expression, we performed immunohistochemical analyses of the 224 paraffin-embedded gastric cancer tissue blocks. Data revealed that the loss of ARID1A expression was significantly correlated with T stage (P = 0.001) and grade (P = 0.006). Consistent with these results, we found that loss of ARID1A expression was significantly correlated with poor survival in gastric cancer patients (P = 0.003). Cox regression analyses showed that ARID1A expression was an independent predictor of overall survival (P = 0.029). Furthermore, the functions of ARID1A in the proliferation and colony formation of gastric cell lines were analyzed by transfecting cells with full-length ARID1A expression vector or siRNA targeting ARID1A. Restoring ARID1A expression in gastric cancer cells significantly inhibited cell proliferation and colony formation. Silencing ARID1A expression in gastric epithelial cell line significantly enhanced cell growth rate. Conclusions/Significance Our data suggest that ARID1A may play an important role in gastric cancer and may serve as a valuable prognostic marker and potential target for gene therapy in the treatment of gastric cancer.
Clinical Cancer Research | 2014
Ke Pan; Xun Xing Guan; Yong Qiang Li; Jing Jing Zhao; Jian Jun Li; Hui Juan Qiu; De Sheng Weng; Qi Jing Wang; Qing Liu; Li Xi Huang; Jia He; Shi Ping Chen; Miao La Ke; Yi Xin Zeng; Jian Chuan Xia
Purpose: Triple-negative breast cancer (TNBC) is a high risk form of this disease, even after surgery, due to the absence of targets for hormone treatment and anti–Her-2 therapy. Chemotherapy is the main therapeutic strategy for such patients with breast cancer, although the outcome is often unsatisfactory. Thus, the development of combination adjuvant therapies is essential for improved prognosis in patients with TNBC. In this study, we investigated the efficacy of a sequential combination of cytokine-induced killer cell (CIK) infusion and chemotherapy for patients with post-mastectomy TNBC. Experimental Design: From 2008 to 2012, 90 patients with post-mastectomy TNBC were included in this retrospective study: 45 cases received chemotherapy alone or with sequential radiotherapy; a further 45 cases received chemotherapy with/without radiotherapy and sequential CIK infusion. Results: Survival analysis showed significantly higher disease-free survival (DFS) and overall survival (OS) rates in the CIK treatment group compared with the control group (P = 0.0382, P = 0.0046, respectively; log-rank test). Multivariate survival analysis showed that CIK adjuvant treatment was an independent prognostic factor for OS of patients with TNBC. In subgroup analyses, CIK adjuvant treatment significantly increased the DFS rate of patients with pathologic grade 3, and significantly increased the OS rate of patients in N1, N2, N3, IIB, III TNM (tumor–node–metastasis) stages, and with pathologic grade 3. Conclusions: These data indicate that adjuvant CIK treatment combined with chemotherapy is an effective therapeutic strategy to prevent disease recurrence and prolong survival of patients with TNBC, particularly those with lymph node metastasis, advanced TNM stage, and poor pathologic grade. Clin Cancer Res; 20(11); 3003–11. ©2014 AACR.
Scientific Reports | 2015
Jing Jing Zhao; Qiu Zhong Pan; Ke Pan; De Sheng Weng; Qi Jing Wang; Jian Jun Li; Lin Lv; Dan Dan Wang; Hai Xia Zheng; Shan Shan Jiang; Xiao Fei Zhang; Jian Chuan Xia
The biological role of interleukin-37 (IL-37) in cancer is large unknown. Through immunohistochemical detection using 163 primary hepatocellular carcinoma (HCC) clinical specimens, we found the expression of IL-37 was decreased in tumor tissues, and the expression level was negatively correlated with tumor size. High expression of IL-37 in HCC tumor tissues was associated with better overall survival (OS) and disease-free survival (DFS). IL-37 expression in tumor tissues was positively associated with the density of tumor-infiltrating CD57+ natural killer (NK) cells, but not with the CD3+ and CD8+ T cells. Consistently, in vitro chemotaxis analysis showed that IL-37- overexpressing HCC cells could recruit more NK cells. The in vivo mouse model experiments also revealed that overexpression IL-37 in HCC cells significantly delayed tumor growth and recruited more NK cells into tumors tissues. Our finding suggested that IL-37 might play an important role for the prognosis of HCC patients via regulating innate immune-action.
Neoplasma | 2012
Lin X; Xu Dong Zhang; Qi Jing Wang; Li J; Zhang P; Zhao M; Li X
P-glycoprotein (P-gp)-mediated multidrug resistance (MDR) is the major clinical impediment to chemotherapy of breast cancers. Down-regulation of PI3K/Akt pathway has been described as related to reversal of MDR in cancer cells. Here, we investigated the reversal effect on MDR phenotype of perifosine, a new Akt inhibitor, in breast cancer cell lines. In this study, MCF-7/ADM cells and MCF-7 cells were treated with different concentrations of perifosine. Our results suggested that perifosine reversed MDR partially by downregulation of P-gp expression and inhibition of PI3K/Akt/NF-κB pathway in the MCF-7/ADM cell line. The novel Akt inhibitor perifosine may be a promising new drug due to its ability to reverse MDR in human breast cancer cells.
Journal of Translational Medicine | 2014
Shan Shan Jiang; De Sheng Weng; Qi Jing Wang; Ke Pan; Yao Jun Zhang; Yong Qiang Li; Jian Jun Li; Jing Jing Zhao; Jia He; Lin Lv; Qiu Zhong Pan; Jian Chuan Xia
BackgroundGalectin-3, a member of the beta-galactoside-binding lectin family, is a multifunctional protein with various biological functions, including the proliferation and differentiation of tumor cells, angiogenesis, cancer progression, and metastasis. We aimed to clarify if expression of galectin-3 is related to the clinicopathological characteristics and prognosis of hepatocellular carcinoma (HCC) patients, and to explore the possible mechanisms of galectin-3 in hepatocellular carcinoma.MethodsFirst, we investigated galectin-3 mRNA and protein expression by using RT-PCR and Western blotting. Second, tissues from 165 HCC patients were used to evaluate clinicopathological characteristics and prognosis through immunohistochemical analyses. Furthermore, the functions of galectin-3 were analyzed with respect to the proliferation, cell cycle,apoptosis, migration, and invasion of HCC cell lines. Finally, we analyzed galectin-3 expression and micro-vessel density (MVD) by immunohistochemistry (IHC) to find its correlation with angiogenesis in Hepatocellular Carcinoma. Flow cytometer was used to explore apoptosis and Western-blot was used to detect the pathway proteins of apoptosis.ResultsGalectin-3 showed high expression at the mRNA and protein levels in HCC cancer tissues and cell lines. Clinicopathological analyses revealed that increased expression of galectin-3 in tumors was closely associated with a poor prognosis. Galectin-3 knockdown by siRNA significantly inhibited cell growth, migration, and invasion, and induced apoptosis in HCC cells in vitro, whereas galectin-3 overexpression promoted cell growth, migration, and invasion. Correlation analysis of galectin-3 expression and micro-vessel density (MVD) showed that galectin-3 expression in tumor cells stimulates angiogenesis. The observed regulation of cell apoptosis was accompanied by the galectin-3-mediated modulation of caspase3 signaling pathways in HCC cells.ConclusionsThese data suggest that galectin-3 plays an important part in HCC progression and may serve as a prognostic factor for HCC.
Cancer Biology & Therapy | 2010
Jian Cong Sun; Ke Pan; Min Shan Chen; Qi Jing Wang; Hui Wang; Hai Qing Ma; Yong Qiang Li; Xiao Ting Liang; Jian Jun Li; Jing Jing Zhao; Yi Bing Chen; Xiong Hao Pang; Wang Li Liu; Yun Cao; Xin Yuan Guan; Qi Zhou Lian; Jian Chuan Xia
Immunotherapy, especially using dendritic cells (DCs)-based vaccine, appears promising in the treatment of hepatocellular carcinoma (HCC) following surgery. However, the therapeutic efficacy of current DC vaccines loaded with HCC antigen is limited in clinical practice. One important reason might be that the DC vaccines for the treatment of HCC were not aimed at targeting the hepatocellular carcinoma cancer stem cells (HCCCSCs). Therefore, establishing an immunotherapy to kill HCC stem cells could be a novel therapeutic strategy. In this study, we have developed an immunotherapy to target CD133+ HCC cells in the treatment of HCC. This study had three main findings; (1), CD133+HCC cells RNA loaded DCs could induce special CD8+ cytotoxic T lymphocytes (CD133+Huh7-CTLs) response against CD133+ Huh7 cells in vitro. (2), Huh7 cells-induced tumor growth in vivo was effectively inhibited by CD133+Huh7-CTLs. (3), the great inhibition potential of CD133+Huh7-CTLs to Huh7-induced tumor growth might not be only associated with anti-tumor cytokines such as IFN-γ, but also to CD133+Huh7-DCs induced specific CTLs. This study shows an experimental proof that CD133+HCC cells RNA loaded DC vaccine has potential in treating HCC and may provide a new therapy for clinical post operative adjuvant therapy in future.
PLOS ONE | 2013
Xiao bo Yang; Jing Jing Zhao; Chun Yu Huang; Qi Jing Wang; Ke Pan; Dan Dan Wang; Qiu Zhong Pan; Shan Shan Jiang; Lin Lv; Xiang Gao; Huang wei Chen; Jia yin Yao; Min Zhi; Jian Chuan Xia
Background FOXO3a, a member of the forkhead class ‘O’ (FOXO) transcription factor family, controls a wide spectrum of biological processes, such as DNA damage repair, apoptosis, and cell cycle regulation. FOXO3a has been shown to be a tumor suppressor in various cancers. This study investigated the expression of FOXO3a in primary gastric adenocarcinomas and its prognostic value for primary gastric adenocarcinoma patients. Methods Real-time quantitative RT-PCR (qRT-PCR), western blotting, and immunohistochemical staining were used to detect FOXO3a expression in primary gastric cancerous surgical specimens and adjacent non-tumorous tissues. Results Our data showed that the expression of FOXO3a mRNA (p = 0.03) and protein (p = 0.019) was lower in cancerous tissues compared with their adjacent non-tumorous tissues. In addition, the chi-square test revealed that low FOXO3a expression was significantly correlated with larger tumor size (p = 0.007), poor histopathological classification (p = 0.029), depth of invasion (p = 0.049), local lymph node metastasis (p = 0.013), distant metastasis (p = 0.013) and AJCC staging (p<0.001). Kaplan-Meier survival analysis demonstrated that low expression of FOXO3a was significantly correlated with a poor prognosis for gastric cancer patients (p<0.001). The multivariate analysis showed that FOXO3a expression was an independent prognostic factor of the overall survival rate of patients with primary gastric adenocarcinoma. Conclusion Our study suggested that decreased FOXO3a expression may play an important role in the progression of gastric cancer. FOXO3a could be a valuable prognostic marker as well as a potential molecular therapy target for gastric cancer patients.
OncoImmunology | 2016
Chang Long Chen; Qiu Zhong Pan; Jing Jing Zhao; Ying Wang; Yong Qiang Li; Qi Jing Wang; Ke Pan; De Sheng Weng; Shan Shan Jiang; Yan Tang; Xiao Fei Zhang; Hong Xia Zhang; Zi Qi Zhou; Yi Xin Zeng; Jian Chuan Xia
ABSTRACT Cytokine-induced killer (CIK) cell immunotherapy represents an effective treatment strategy for treating hepatocellular carcinoma (HCC). However, the therapeutic benefits of CIK cell treatment can be influenced by differences in complex immune microenvironment between patients. Herein, we investigated the relationship between PD-L1 expression and survival benefits of CIK cell immunotherapy in HCC patients. This retrospective study included 448 HCC patients: 217 cases underwent hepatectomy alone; 231 cases received hepatectomy and post-operative CIK cell transfusion. Immunohistochemistry was used to measure PD-L1 expression in tumor tissue sections from all patients. Meanwhile, flow cytometry was performed to explore the relationship between PD-L1 expression and localized inflammatory response in HCC microenvironment. We found a significantly improved prognosis in CIK treatment group compared with surgery alone group. In the CIK treatment group, higher PD-L1 expression was observed in patients who exhibited long-term survival benefit. Survival analysis showed patients with ≥5% PD-L1 expression had better overall survival (OS) and recurrence-free survival (RFS) than patients with 1–5% or <1% PD-L1 expression, particularly in the subgroup with high hepatitis B viral load. By contrast, PD-L1 expression did not show direct impact on the survival of patients in surgery alone group. Additionally, PD-L1 expression was found to be highly associated with hepatitis B viral load and the proportion of tumor-infiltrating lymphocytes in HCC patients. In conclusions, our study indicates that PD-L1 expression may reflect the presence of endogenous host immune response to tumor and serve as a biomarker for predicting survival benefits from adjuvant CIK cell immunotherapy in HCC patients.
Stem Cells | 2015
Qiu Zhong Pan; Ke Pan; Qi Jing Wang; De Sheng Weng; Jing Jing Zhao; Hai Xia Zheng; Xiao Fei Zhang; Shan Shan Jiang; Lin Lv; Yan Tang; Yong Qiang Li; Jia He; Qing Liu; Chang Long Chen; Hong Xia Zhang; Jian Chuan Xia
Cancer stem‐like cells/cancer‐initiating cells (CSCs/CICs) are considered to represent a small population of cancer cells that is resistant to conventional cancer treatments and responsible for tumor recurrence and metastasis. The aim of this study was to establish CSC/CIC‐targeting immunotherapy. In this study, we found that Annexin A3 (ANXA3) was preferentially expressed in CSCs/CICs derived from hepatocellular carcinoma (HCC) cells compared to non‐CSCs/CICs. In HCC samples, high levels of ANXA3 correlated with expansion of CD133+ tumor cells representing CSCs/CICs in HCC; the combination of high levels of ANXA3 and CD133 was associated with progression of HCC. Overexpression of ANXA3 increased the proportion of CD133+ cells, enhancing their tumorigenicity. On the contrary, knockdown of ANXA3 decreased CD133+ cells and inhibited tumorigenicity. The mechanistic study revealed that ANXA3‐mediated maintenance of HCC CSCs/CICs activity was likely involved with the HIF1A/Notch pathway. Using ANXA3 as a target, ANXA3‐transfected dendritic cells could induce more functionally active T cells and these effector T cells could superiorly kill CD133+ HCC CSCs/CICs in vitro and in vivo. Taken together, our findings suggest that ANXA3 plays a role in HCC CSC/CIC maintenance, and that ANXA3 may represent a potential CSC/CIC‐specific therapeutic target for improving the treatment of HCC. Stem Cells 2015;33:354–366