Network


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

Hotspot


Dive into the research topics where Qian Zhu is active.

Publication


Featured researches published by Qian Zhu.


OncoImmunology | 2017

Tumor cells PD-L1 expression as a favorable prognosis factor in nasopharyngeal carcinoma patients with pre-existing intratumor-infiltrating lymphocytes

Qian Zhu; Mu Yan Cai; Chang Long Chen; Hao Hu; Huan Xin Lin; Min Li; De Sheng Weng; Jing Jing Zhao; Ling Guo; Jian Chuan Xia

ABSTRACT Programmed death ligand 1 (PD-L1) expression represents a mechanism of immune escape by inhibiting T cell immunity. This study systematically evaluated the expression of PD-L1, spatial distribution of CD3+ immune cells and the relationship of both factors to survival in nasopharyngeal carcinoma (NPC) patients. A total of 209 NPC patients treated between 1991 and 2000 were included. Pairs of TMAs were immunohistochemically stained with PD-L1 and CD3. Survival analysis was evaluated according to PD-L1 status and the spatial distribution of CD3+ immune cells in the primary lesion microenvironment. PD-L1 staining was observed on tumor cells and tumor-infiltrating immune cells (TILs); however, PD-L1-positive immune cells were more common (98/209) than PD-L1-positive tumor cells (68/209). Limited numbers of intra-tumoral CD3+ T cells (median number: 20) were detected. Patients with higher CD3+ T cell infiltration, both intratumorally and peritumorally, had higher PD-L1 expression on tumor cells (both p < 0.001) and immune cells (p = 0.002 and p < 0.001, respectively). Increasing intratumoral CD3 infiltration was correlated with increased overall survival (OS) (p = 0.008) and disease-free survival (DFS) (p = 0.003). Nevertheless, patients with low levels of peritumoral TILs showed superior OS (p = 0.557) and DFS to those with higher levels of peritumoral TILs (p = 0.671). Moreover, type classification based on intratumoral CD3 infiltration and tumor cell PD-L1 expression was an independent prognostic factor for NPC patients. PD-L1 expression on tumor cells is a favorable prognosis factor in NPC patients with pre-existing intratumor-infiltrating lymphocytes.


OncoImmunology | 2018

IL-17 induces antitumor immunity by promoting beneficial neutrophil recruitment and activation in esophageal squamous cell carcinoma

Chang Long Chen; Ying Wang; Chun Yu Huang; Zi Qi Zhou; Jing Jing Zhao; Xiao Fei Zhang; Qiu Zhong Pan; Jiang Xue Wu; De Sheng Weng; Yan Tang; Qian Zhu; Lu Ping Yuan; Jian Chuan Xia

ABSTRACT Interleukin (IL)-17 has been reported to play a controversial role in tumor immunity. Our previous studies showed that infiltration of IL-17-producing cells in esophageal squamous cell carcinoma (ESCC) induced tumor protective immunity by recruiting CD8+T lymphocytes, natural killer (NK) cells, and B lymphocytes into the tumor microenvironment. However, the mechanism of IL-17 regulation of tumor-associated neutrophils remains elusive in ESCC. In this study, we therefore evaluated the accumulation of myeloperoxidase (MPO)+ neutrophils and its association with IL-17-producing cells within ESCC tumor nests. We also investigated the effects of IL-17 on the recruitment and antitumor activity of neutrophils. MPO+ neutrophil infiltration was found to predict a favorable prognosis in ESCC patients and was positively correlated with IL-17+ cell density. IL-17 stimulated ESCC tumor cells to release more of the CXC chemokines CXCL2 and CXCL3, which are involved in neutrophil migration. Furthermore, IL-17 potentiates the direct killing capability of neutrophils by enhancing the production of cytotoxic molecules, including reactive oxygen species (ROS), MPO, TNF-related apoptosis-inducing ligand (TRAIL), and IFN-γ. Experiments in mice suggested that IL-17 alone might not affect tumor progression in the tumor-bearing host, but IL-17 can inhibit tumor growth by promoting beneficial neutrophil infiltration and activation at tumor sites. As emerging evidence indicates that targeting tumor-associated neutrophils is a strategy for antitumor therapy, our findings reveal a positive contribution of IL-17 to the modulation of neutrophil-mediated antitumor immunity in ESCC. This study provides further understanding of the mechanisms that selectively regulate functional activities of neutrophils, which may be critical for developing new tumor immunotherapy.


Journal of Cancer | 2017

Overexpression of SMOC2 attenuates the tumorigenicity of hepatocellular carcinoma cells and is associated with a positive postoperative prognosis in human hepatocellular carcinoma

Xu Qiong Huang; Zi Qi Zhou; Xiao Fei Zhang; Chang Long Chen; Yan Tang; Qian Zhu; Jian Hua Zhang; Jian Chuan Xia

Secreted modular calcium binding protein-2 (SMOC2), a recently identified matricellular protein that belongs to the SPARC protein family, has been reported to be downregulated in various cancers. The purpose of this study was to investigate the clinical significance and biological function of SMOC2 in human hepatocellular carcinoma. Real-time quantitative PCR and western blotting analyses revealed that SMOC2 mRNA and protein levels were significantly downregulated in human HCC tissues compared to the matched adjacent normal tissues. Clinicopathological analysis indicated that SMOC2 expression was significantly associated with tumor size, number of tumors, tumor-node-metastasis (TNM) stage and distant metastasis. Kaplan-Meier survival analysis showed that high tumor SMOC2 expression was associated with improved overall survival and disease-free survival in patients with HCC. Functional analyses (cell proliferation and colony formation assays, cell migration and invasion assays, cell cycle and apoptosis assays) demonstrated that stable overexpression of SMOC2 using a lentiviral vector significantly inhibited cell proliferation, colony formation, migration and invasion, and induced G0/G1 phase arrest in HCC cells in vitro. In addition, experiments with a mouse model revealed the suppressed effect of SMOC2 on HCC tumorigenicity and metastases in vivo. These results suggest that SMOC2 functions as a tumor suppressor during the development of HCC and may represent an effective prognostic factor and novel therapeutic target for HCC.


Molecular Carcinogenesis | 2018

HUS1 checkpoint clamp component (HUS1) is a potential tumor suppressor in primary hepatocellular carcinoma

Zi-Qi Zhou; Jingjing Zhao; Chang-Long Chen; Yuan Liu; Jian-Xiong Zeng; Zheng-Rong Wu; Yan Tang; Qian Zhu; Desheng Weng; Jian-Chuan Xia

The HUS1 checkpoint clamp component (HUS1), which is a member of an evolutionarily conserved, genotoxin‐activated checkpoint complex (Rad9‐Rad1‐Hus1 [9‐1‐1] complex), is involved in cell cycle arrest and DNA repair in response to DNA damage. We conducted this study to investigate the biological significances of HUS1 expression in hepatocellular carcinoma (HCC) development. The mRNA and protein expression levels of HUS1 were determined using Real‐time PCR and Western blot, respectively. One hundered and twenty four paraffin sections from HCC tissues were analyzed by immunohistochemistry to assess the association between HUS1 expression and clinicopathological characteristics of patients. The Kaplan‐Meier method was performed to calculate the OS and RFS curves. Cell proliferation and colony formation assays, cell migration and invasion assays and cell cycle assays were used to determine the suppressor role of HUS1 in vitro. A mouse model was used to determine the effect of HUS1 on tumorigenesis. The expression of HUS1 was significantly decreased in HCC cell lines and tissues, and low HUS1 expression was associated with poor prognosis of HCC patients. Upregulation of HUS1 expression inhibited the cell proliferation, colony formation, migration, and invasion, as well as arrested cell cycle at G0/G1 in HCC cells in vitro. Moreover, sufficient HUS1 expression inhibited the tumor growth in nude mice. Our study revealed for the first time that HUS1 is a potential tumor suppressor that might produce an antitumor effect in human HCC. Furthermore, HUS1 may serve as a prognostic indicator and could be used for therapeutic application in HCC patients.


Journal of Cancer | 2018

Weekly versus triweekly cisplatin plus intensity-modulated radiotherapy in locally advanced nasopharyngeal carcinoma: A propensity score analysis with a large cohort

Qian Zhu; Hao Hu; Lin-Quan Tang; Rui You; Jingjing Zhao; Desheng Weng; Qiu-Zhong Pan; Chang-Long Chen; Zi-Qi Zhou; Yan Tang; Jian-Chuan Xia

Purpose: To directly compare the efficacy and acute toxicities of intensity-modulated radiotherapy (IMRT) concurrent with weekly cisplatin (40 mg/m2) to high-dose concurrent cisplatin (100 mg/m2) at three-week intervals. Materials and Methods: A total of 3,799 patients diagnosed with locally advanced nasopharyngeal carcinoma (NPC) at Sun Yat-Sen University Cancer Center between January 2010 and December 2013 were retrospectively reviewed. Propensity score analysis was conducted to balance the baseline characteristics between the groups, which allowed us to draw reliable conclusions. The efficacy and safety profiles were then assessed in the well-balanced large cohort. Results: The risk of distant metastasis was lower among the patients treated with weekly concurrent cisplatin than among those treated with the triweekly regimen (hazard ratio [HR], 0.45; P = .028). However, the disease-free survival, loco-regional relapse-free survival and overall survival rates were similar. The weekly group showed significantly higher rates of grade 3-4 thrombocytopenia, but lower rates of grade 3-4 mucositis, nausea and vomiting than the triweekly group. Conclusion: IMRT concurrent with a weekly cisplatin regimen was associated with significantly improved distant metastasis-free survival in locally advanced NPC. Differences in the selected acute toxicities between the weekly and triweekly concurrent cisplatin regimens were noted.


Journal of Cancer | 2018

The prognosis of neck residue nasopharyngeal carcinoma (NPC) patients: results from a case-cohort study

Sai Lan Liu; Lin Quan Tang; Qiu Yan Chen; Huan Xin Lin; Qi Yang; Qian Zhu; Wen Wen; Shan Shan Guo; Li Ting Liu; Yang Li; Hao Jun Xie; Qing Nan Tang; Xue Song Sun; Yu Jing Liang; Xiao Yun Li; Jin Jie Yan; chao lin; Xiao Wen Lan; Hai Qiang Mai; Ling Guo

Background: To assess the prognosis of neck residue nasopharyngeal carcinoma (NPC) patients and the efficacy of neck dissection in the treatment of these patients. Methods: We recruited 68 neck residue NPC patients. For each neck residue patient we had three matched NPC patients without neck residue as controls (n = 204). The primary endpoint was progression-free survival (PFS). The Cox proportional hazards model was used to calculate hazard ratios (HRs) with 95% confidence intervals (CIs), and multivariable analysis was used to test the independent statistical significance of NPC patients. Results: Compared to controls, the neck residue patients showed significantly lower 3-year PFS (46.7% vs. 87.6%; P < 0.001). Multivariable analysis showed that neck residue was an independent prognostic factor for PFS. Conclusions: NPC patients who had pathologically proven neck residue are associated with poor prognosis. Management with neck dissection alone seems not to be sufficient for these patients.


Journal of Cancer | 2018

Clinical Effect of Adjuvant Cytokine-Induced Killer Cells Immunotherapy in Patients with Stage II-IVB Nasopharyngeal Carcinoma after Chemoradiotherapy: A propensity score analysis

Jingjing Zhao; Shu Zhou; Chang-Long Chen; Hong-Xia Zhang; Zi-Qi Zhou; Zheng-Rong Wu; Yuan Liu; Qiu-Zhong Pan; Qian Zhu; Yan Tang; Jian-Chuan Xia; Desheng Weng

As an adjuvant immunotherapy, cytokine-induced killer cells (CIKs) infusion has been demonstrated to exert potent effectiveness in several types of cancer patients who received curative treatment. However, controversy exists regarding whether nasopharyngeal carcinoma (NPC) patients can benefit from additional treatment after radical radiotherapy or chemoradiotherapy to improve their distant control and survival. In this retrospective study, we aimed to evaluate the efficacy of adjuvant CIK cells therapy in NPC patients with stage II-IVB after curative treatment. From January 1, 2005 to December 31, 2012, 85 pairs of NPC patients matching by propensity score matching (PSM) method to balance prognostic factors were included in this study: 85 cases underwent radical treatment, 85 cases received radical treatment and sequential CIKs infusion. We found that disease-free survival (DFS) and overall survival (OS) were significantly better in the CIK group than that in the control group (P = 0.009, P < 0.001, respectively). Adjuvant CIK cells immunotherapy was showed to be an independent prognostic factor for survival of the patients in further multivariate analysis. In subgroup analyses, the DFS and OS of patients with T3/4, III and IV A-B TNM (tumor-node-metastasis) stages were significantly enhanced in CIK group compared to control group. Nevertheless, both NPC patients with high and low EBV DNA benefited from adjuvant CIK cells immunotherapy. In conclusion, CIKs infusion is an effective adjuvant immunotherapy for enhancing the prognosis of NPC patients who have received the standard treatment, particularly for those with more aggressive tumor (T3/4) or advanced TNM stage.


OncoImmunology | 2017

Safety and activity of PD-1 blockade-activated DC-CIK cells in patients with advanced solid tumors

Chang Long Chen; Qiu Zhong Pan; De Sheng Weng; Chuan Miao Xie; Jing Jing Zhao; Min Shan Chen; Rui Qing Peng; Dan Dan Li; Ying Wang; Yan Tang; Qi Jing Wang; Zhi Ling Zhang; Xiao Fei Zhang; Li Juan Jiang; Zi Qi Zhou; Qian Zhu; Jia He; Yuan Liu; Fang Jian Zhou; Jian Chuan Xia

ABSTRACT Cytokine-induced killer (CIK) cells that are stimulated using mature dendritic cells (DCs), referred to as (DC-CIK cells) exhibit superior anti-tumor potency. Anti-programmed death-1 (PD-1) antibodies reinvigorate T cell-mediated antitumor immunity. This phase I study aimed to assess the safety and clinical activity of immunotherapy with PD-1 blockade (pembrolizumab)-activated autologous DC-CIK cells in patients with advanced solid tumors. Patients with selected types of advanced solid tumors received a single intravenous infusion of activated autologous DC-CIK cells weekly for the first month and every 2 weeks thereafter. The primary end points were safety and adverse event (AE) profiles. Antitumor responses, overall survival (OS), progression-free survival (PFS) and cytolytic activity were secondary end points. Treatment-related AEs occurred in 20/31 patients. Grade 3 or 4 toxicities, including fever and chills, were observed in two patients. All treatment-related AEs were reversible or controllable. The cytotoxicity of DC-CIK cells induced up-regulation of PD-L1 expression on autologous tumor cells. When activated using pembrolizumab ex vivo, DC-CIK cells exerted superior antitumor properties and elevated IFN-γ secretion. Objective responses (complete or partial responses) were observed in 7 of the 31patients.These responses were durable, with 6 of 7 responses lasting more than 5 months. The overall disease control rate in the patients was 64.5%. At the time of this report, the median OS and PFS were 270 and 162 days, respectively. In conclusions, treatment with pembrolizumab-activated autologous DC-CIK cells was safe and exerted encouraging antitumor activity in advanced solid tumors. A larger phase II trial is warranted.


Applied Mathematical Modelling | 2009

Third order approximation to capillary gravity short crested waves with uniform currents

Y.J. Jian; Qian Zhu; Jing Zhang; Y.F. Wang


BMC Cancer | 2017

Pooled safety analyses of ALK-TKI inhibitor in ALK-positive NSCLC

Qian Zhu; Hao Hu; De Sheng Weng; Xiao Fei Zhang; Chang Long Chen; Zi Qi Zhou; Yan Tang; Jian Chuan Xia

Collaboration


Dive into the Qian Zhu's collaboration.

Top Co-Authors

Avatar

Yan Tang

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zi Qi Zhou

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hao Hu

Nanchang University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge