Network


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

Hotspot


Dive into the research topics where Tianxiao Xu is active.

Publication


Featured researches published by Tianxiao Xu.


Clinical Cancer Research | 2017

Frequent Genetic Aberrations in the CDK4 Pathway in Acral Melanoma Indicate the Potential for CDK4/6 Inhibitors in Targeted Therapy

Yan Kong; Xinan Sheng; Xiaowen Wu; Junya Yan; Meng Ma; Jiayi Yu; Lu Si; Zhihong Chi; Chuanliang Cui; Jie Dai; Yiqian Li; Huan Yu; Tianxiao Xu; Huan Tang; Bixia Tang; Lili Mao; Bin Lian; Xuan Wang; Xieqiao Yan; Siming Li; Jun Guo

Purpose: Effective therapies for the majority of metastatic acral melanoma patients have not been established. Thus, we investigated genetic aberrations of CDK4 pathway in acral melanoma and evaluated the efficacy of CDK4/6 inhibitors in targeted therapy of acral melanoma. Experimental Design: A total of 514 primary acral melanoma samples were examined for the copy number variations (CNV) of CDK4 pathway-related genes, including Cdk4, Ccnd1, and P16INK4a, by QuantiGenePlex DNA Assay. The sensitivity of established acral melanoma cell lines and patient-derived xenograft (PDX) containing typical CDK4 aberrations to CDK4/6 inhibitors was evaluated. Results: Among the 514 samples, 203 cases, 137 cases, and 310 cases, respectively, showed Cdk4 gain (39.5%), Ccnd1 gain (26.7%), and P16INK4a loss (60.3%). The overall frequency of acral melanomas that contain at least one aberration in Cdk4, Ccnd1, and P16INK4a was 82.7%. The median overall survival time for acral melanoma patients with concurrent Cdk4 gain with P16INK4a loss was significantly shorter than that for patients without such aberrations (P = 0.005). The pan-CDK inhibitor AT7519 and selective CDK4/6 inhibitor PD0332991 could inhibit the cell viability of acral melanoma cells and the tumor growth of PDX with Cdk4 gain plus Ccnd1 gain, Cdk4 gain plus P16INK4a loss, and Ccnd1 gain plus P16INK4a loss. Conclusions: Genetic aberration of CDK4 pathway is a frequent event in acral melanoma. Acral melanoma cell lines and PDX containing CDK4 pathway aberrations are sensitive to CDK4/6 inhibitors. Our study provides evidence for the testing of CDK4/6 inhibitors in acral melanoma patients. Clin Cancer Res; 23(22); 6946–57. ©2017 AACR.


Translational Oncology | 2017

Systemic Immune-Inflammation Index and Circulating T-Cell Immune Index Predict Outcomes in High-Risk Acral Melanoma Patients Treated with High-Dose Interferon

Jiayi Yu; Xiaowen Wu; Huan Yu; Siming Li; Lili Mao; Zhihong Chi; Lu Si; Xinan Sheng; Chuanliang Cui; Jie Dai; Meng Ma; Huan Tang; Tianxiao Xu; Junya Yan; Yan Kong; Jun Guo

High-dose interferon alfa-2b (IFN-α-2b) improves the survival of patients with high-risk melanoma. We aimed to identify baseline peripheral blood biomarkers to predict the outcome of acral melanoma patients treated with IFN-α-2b. Pretreatment baseline parameters and clinical data were assessed in 226 patients with acral melanoma. Relapse-free survival (RFS) and overall survival (OS) were assessed using the Kaplan-Meier method, and multivariate Cox regression analyses were applied after adjusting for stage, lactate dehydrogenase (LDH), and ulceration. Univariate analysis showed that neutrophil-to-lymphocyte ratio ≥2.35, platelet-to-lymphocyte ratio ≥129, systemic immune-inflammation index (SII) ≥615 × 109/l, and elevated LDH were significantly associated with poor RFS and OS. The SII is calculated as follows: platelet count × neutrophil count/lymphocyte count. On multivariate analysis, the SII was associated with RFS [hazard ratio (HR)=1.661, 95% confidence interval (CI): 1.066-2.586, P=.025] and OS (HR=2.071, 95% CI: 1.204-3.564, P=.009). Additionally, we developed a novel circulating T-cell immune index (CTII) calculated as follows: cytotoxic T lymphocytes/(CD4+ regulatory T cells × CD8+ regulatory T cells). On univariate analysis, the CTII was associated with OS (HR=1.73, 95% CI: 1.01-2.94, P=.044). The SII and CTII might serve as prognostic indicators in acral melanoma patients treated with IFN-α-2b. The indexes are easily obtainable via routine tests in clinical practice.


Journal of Hematology & Oncology | 2018

Anti-GD2/4-1BB chimeric antigen receptor T cell therapy for the treatment of Chinese melanoma patients

Jiayi Yu; Xiaowen Wu; Junya Yan; Huan Yu; Longwen Xu; Zhihong Chi; Xinan Sheng; Lu Si; Chuanliang Cui; Jie Dai; Meng Ma; Tianxiao Xu; Yan Kong; Jun Guo

BackgroundChimeric antigen receptor (CAR)-engineered T cells have demonstrated promising clinical efficacy in patients with B cell lymphoma. However, the application of CAR-T cell therapy in the treatment of other solid tumors has been limited. We incorporated 4-1BB into the anti-GD2 CAR-T cells to test their cytotoxicity in melanoma in vitro and in vivo. Moreover, we reported the expression of ganglioside GD2 in non-Caucasian melanoma populations for the first time, thus providing a basis for future clinical research.MethodsThis study included tumor samples from 288 melanoma patients at the Peking University Cancer Hospital & Institute. Clinical data were collected. Immunohistochemical assays using antibodies against ganglioside GD2 were performed on formalin-fixed, paraffin-embedded specimens. The ability of ganglioside GD2 CAR-T cells to kill ganglioside GD2+ melanoma cells was evaluated in vitro and in a patient-derived xenograft (PDX) model.ResultsAmong the 288 samples, 49.3% of cases (142/288) demonstrated positive staining with ganglioside GD2. The median survival time in patients exhibiting ganglioside GD2 expression was significantly shorter than that in patients without ganglioside GD2 expression (31 vs. 47.1xa0months, Pu2009<u20090.001). In the present study, CAR was constructed using a GD2-specific scFv (14.G2a), T cell receptor CD3ζ chain, and the CD137 (4-1BB) costimulatory motif. In addition, the GD2.BBζ CAR-T cells demonstrated specific lysis of ganglioside GD2-expressing melanoma cells in vitro. In two PDX models, mice that received intravenous or local intratumor injections of GD2.BBζ CAR-T cells experienced rapid tumor regression.ConclusionsThese data demonstrate that the rate of GD2 expression in Chinese patients is 49.3%. GD2.BBζ CAR-T cells can both efficiently lyse melanoma in a GD2-specific manner and release Th1 cytokines in an antigen-dependent manner in vitro and in vivo. Anti-GD2/4-1BB CAR-T cells represent a clinically appealing treatment strategy for Chinese melanoma patients exhibiting GD2 expression and provide a basis for future studies of the clinical application of immunotherapy for melanoma.


Oncology Letters | 2017

Mutations in BRAF codons 594 and 596 predict good prognosis in melanoma

Xiaowen Wu; Junya Yan; Jie Dai; Meng Ma; Huan Tang; Jiayi Yu; Tianxiao Xu; Huan Yu; Lu Si; Zhihong Chi; Xinan Sheng; Chuanliang Cui; Yan Kong; Jun Guo

B-Raf proto-oncogene serine/threonine kinase (BRAF) V600E is the most common kinase-activating mutation and is associated with poor prognosis in melanoma. However, the clinical significance of kinase-impairing mutations remains unclear. The present study aimed to analyze kinase-impairing mutations in BRAF codons 594 and 596 in non-Caucasian patients with melanoma and to investigate their possible clinical significance. To detect hotspot mutations, exon 15 of the BRAF gene was amplified using polymerase chain reaction in samples from 1,554 patients with melanoma. Among these patients, a total of 912 valid follow-up data were obtained. These patients were divided into three groups according to their BRAF activation status: BRAF wild-type (n=752), BRAF V600E (n=147); and BRAF D594/G596 (n=13). Then the correlation between BRAF activation status, and the clinicopathological features and overall survival (OS) of the patients were analyzed. The prevalence of BRAF mutations in non-Caucasian patients with melanoma was 24.3% (377/1554). Three patients carried two mutations simultaneously. The overall mutation frequencies of kinase-activating mutations, kinase-impairing mutations, and mutations with unknown effects were 93.4 (355/380), 3.4 (13/380), and 3.2% (12/380), respectively. BRAF V600E was identified to be associated with a poor prognosis. Patients with BRAF mutations in codons 594 and 596 had a longer OS time compared with those with a BRAF V600E mutation [median OS, 45 vs. 25 months; HR, 0.45 (95% confidence interval, 0.31–0.97); P=0.043]. To the best of our knowledge, this is the first study to examine a large number of samples from non-Caucasian patients with melanoma and report the characteristics of BRAF mutations according to mutant kinase activity. Melanoma arising from a mutation in BRAF codon 594 or 596 can be differentiated from BRAF V600E-induced melanoma, and mutations in these codons may be good prognostic factors for melanoma. The results of the present study are thus of significance for the development of accurate personalized medicine to treat melanoma.


Journal of Cancer | 2018

Increased AURKA Gene Copy Number Correlates with Poor Prognosis and Predicts the Efficacy of High-dose Interferon Therapy in Acral Melanoma

Junya Yan; Jiayi Yu; Xiaowen Wu; Tianxiao Xu; Huan Yu; Jie Dai; Meng Ma; Huan Tang; Longwen Xu; Zhihong Chi; Lu Si; Xinan Sheng; Chuanliang Cui; Yan Kong; Jun Guo

Background: AURKA kinase is an essential serine/threonine kinase for mitosis and chromosome stability. The aberrant amplification and overexpression of AURKA are commonly observed in various types of cancer, including cutaneous melanoma. However, the status and the clinical significance of AURKA copy number (CN) in acral melanoma (AM) have not been fully elucidated. Methods: Four hundred and seventy-two AM samples were included in the study. AURKA CN was examined using the QuantiGenePlex DNA Assay. We analysed the relationship of AURKA CN to clinicopathological characteristics and survival of patients with AM. Results: In this study, AURKA copy gain (set as more than 2.0 copies) was detected in 24.6% (116/472) of the samples. We did not observe any obvious correlation between clinicopathological characteristics and AURKA copy gain of the patients. However, patients with AURKA copy gain had a significantly shorter overall survival time (OS) and progression-free survival time (PFS) than those with normal AURKA CN (OS: P = 0.022; PFS: P < 0.001). Furthermore, multivariate Cox regression analysis showed that AURKA copy gain was an independent poor prognostic factor for patients with AM undergoing adjuvant interferon therapy. Conclusions: This study suggested that AURKA copy gain is an adverse prognostic factor for AM. Furthermore, AURKA copy gain may be a useful biomarker to predict the outcome of interferon therapy in patients with AM.


European Journal of Cancer | 2018

Analysis of NRAS gain in 657 patients with melanoma and evaluation of its sensitivity to a MEK inhibitor

Junya Yan; Xiaowen Wu; Jiayi Yu; Huan Yu; Tianxiao Xu; Kevin M. Brown; Xue Bai; Jie Dai; Meng Ma; Huan Tang; Lu Si; Zhihong Chi; Xinan Sheng; Chuanliang Cui; Yan Kong; Jun Guo

BACKGROUNDnNeuroblastoma rat-sarcoma (NRAS) mutations have been described in Chinese patients with melanoma. However, the status and the clinical significance of NRAS gain have not been investigated on a large scale.nnnMETHODSnA total of 657 melanoma samples were included in the study. NRAS copy number was examined using the QuantiGene Plex DNA assay. The sensitivities of cell lines and patient-derived xenograft (PDX) models containing NRAS gain to a MAP/ERK kinase (MEK) inhibitor (binimetinib) were also evaluated.nnnRESULTSnThe overall incidence of NRAS gain was 14.0% (92 of 657). Incidence of NRAS gain in acral, mucosal, chronic sun-induced damage (CSD) and non-CSD melanomas was 12.2%, 15.8%, 9.5% and 19.4%, respectively. NRAS gain was mutually exclusive to NRAS mutations (Pxa0=xa00.036). The median survival time for melanoma patients with NRAS gain was significantly shorter than that for patients with normal NRAS copy number (Pxa0=xa00.006). For patients containing NRAS gain, the median survival time for higher copy number (>4 copies) was significantly shorter than those with lower copy number (2-4 copies; Pxa0=xa00.002). The MEK inhibitor (binimetinib) inhibited the proliferation of melanoma cells and the tumour growth of PDX models with NRAS gain.nnnCONCLUSIONSnNRAS gain is frequent in patients with melanoma and may predict a poor prognosis of melanoma. The melanoma cells and PDX models containing NRAS gain are sensitive to MEK inhibitor (binimetinib), indicating that NRAS gain might be a new therapeutic target for melanoma.


Translational cancer research | 2018

cMET-N375S germline mutation is associated with poor prognosis of melanoma in Chinese patients

Tianxiao Xu; Sifan Yu; Meng Ma; Jie Dai; Zhihong Chi; Lu Si; Xinan Sheng; Chuanliang Cui; Yan Kong; Jun Guo

Background: cMET has been known to play an essential role in malignant melanoma tumor progression. Recent studies have shown that a germline missense variant in exon 2 of the cMET , N375S (rs33917957 A>G) occurred at a much higher frequency in East Asian patients with lung cancer. However, the status of this mutation in melanoma is unclear. In this study, we examined the mutation frequency of cMET-N375S in 181 melanoma samples and analyzed its clinicopathological significance. Methods: Tissue samples (n=181) were analyzed for cMET-N375S, BRAF, NRAS , and CKIT mutation in genomic DNA by polymerase chain reaction amplification and Sanger sequencing. The levels of p-cMET protein were determined by immunohistochemistry. Clinical data of patients were also collected. Results: We identified a highly frequent variant (N375S) of cMET in 13.26% (24/181) of the patients with melanoma. This mutation was relatively more frequent in acral (22.8%) and mucosal (12.7%) melanomas (the two most common melanoma subtypes in Asian, but not in Caucasians) than in non-chronic sun induced damage (non-CSD; 5.9%) and CSD (8.3%) melanomas. Among the 24 cases with cMET-N375S mutations, six cases were found to harbor the BRAF mutation, four cases harbored NRAS mutations, and two cases harbored CKIT mutations. And there was no correlation between this mutation and p-cMET levels. Kaplan- Meier analysis showed significant differences in overall survival (OS) and disease-free survival (DFS) between the N375S mutation group and non-N375S mutation group (P=0.039; P=0.030). The Cox proportional hazards model revealed that cMET-N375S mutation was an independent adverse prognostic factor for OS [P=0.003, hazard ratio (HR) =3.577]. Conclusions: Our findings suggested that the cMET-N375S mutation was an adverse prognostic factor for melanoma in a Chinese cohort.


Translational cancer research | 2018

Methylation of O6-methylguanine DNA methyltransferase promoter is a predictive biomarker in Chinese melanoma patients treated with alkylating agents

Jiayi Yu; Huan Yu; Junya Yan; Xiaowen Wu; Lu Yang; Jie Dai; Meng Ma; Huan Tang; Tianxiao Xu; Zhihong Chi; Lu Si; Xinan Sheng; Chuangliang Cui; Jun Guo; Yan Kong

Background: The O6-methylguanine-DNA methyltransferase gene ( MGMT ) promoter methylation status can be used to predict the prognosis of patients with various cancers following treatment with alkylating agents. Moreover, MGMT promoter methylation often coexists with TP53 gene mutation. However, MGMT has not been identified as a biomarker of melanoma. Therefore, this study systematically analyzed the prognostic role of MGMT and the correlation between MGMT methylation and TP53 mutation in non-Caucasian patients with melanoma. Methods: This study involved tumor samples and clinical data collected from 205 melanoma patients treated with alkylating agents at Peking University Cancer Hospital & Institute. The MGMT promoter methylation and TP53 mutation status were analyzed respectively using methylation-specific polymerase chain reaction and polymerase chain reaction followed by Sanger sequencing. Additionally, MGMT protein expression in tumor samples was assessed via immunohistochemistry. n Results: MGMT promoter methylation was detected in 97 (47%) of the 205 tumor samples, and was significantly associated with a loss of MGMT protein expression (P=0.021). MGMT promoter methylation was also significantly associated with the presence of TP53 mutation (P=0.004). Regarding prognosis, patients without MGMT promoter methylation exhibited worse overall survival outcomes, compared to those with methylation (hazard ratio: 1.443; 95% confidence interval: 0.731–2.342; P=0.015). Conclusions: MGMT promoter methylation appears to coexist frequently with TP53 mutation. Patients harboring MGMT promoter methylation within tumors may benefit from therapy with alkylating agents.


OncoTargets and Therapy | 2018

TERT copy gain predicts the outcome of high-dose interferon α-2b therapy in acral melanoma

Sifan Yu; Tianxiao Xu; Jie Dai; Meng Ma; Huan Tang; Zhihong Chi; Lu Si; Chuanliang Cui; Xinan Sheng; Yan Kong; Jun Guo

Background Asian populations are more likely to develop acral melanoma (AM) than Caucasians. The high-dose interferon (HD-IFN) α-2b regimen is the main adjuvant treatment for AM. TERT encodes the catalytic subunit of telomerase reverse transcriptase, which plays an important role in melanoma. Frequent TERT mutation and increased TERT gene expression have been described in AM. Our study aimed to investigate the status and the clinical significance of TERT copy number in a large cohort of patients with AM and to analyze the relationship between TERT copy number gain and the efficiency of HD-IFN. Patients and methods A total of 573 melanoma samples were retrospectively collected and analyzed for TERT copy number via Sanger sequencing. Clinical data of patients were also collected. Results TERT copy gain (copy number >2) was detected in 257 of the 573 patients with AM (44.9%). Of the 573 patients, 81 (14.1%) had a high copy gain (copy number >4). Patients with ulceration showed a significantly higher copy gain rate of TERT compared to the patients without ulceration (P=0.028). Patients with a tumor thicker than 4 mm also had a higher copy number rate of TERT than those with <4 mm (P=0.048). Our results showed that the overall survival (OS) was not significantly different between patients with and without TERT copy gain (P=0.890). However, among the 278 patients who received an HD-IFN regimen, Kaplan–Meier survival analysis demonstrated a significant correlation between TERT copy gain and relapse-free survival (RFS) (P=0.008). In addition, multivariate Cox regression assays validated TERT copy gain to be an independent prognostic factor of RFS for patients with AM undergoing HD-IFN therapy (hazard ratio =1.50; P=0.019). Conclusion The copy number status of TERT might be a predictor for HD-IFN efficacy, but it is not a prognostic factor of OS in patients with AM.


Journal of Cancer Research and Clinical Oncology | 2018

Analysis of TSC1 mutation spectrum in mucosal melanoma

Meng Ma; Jie Dai; Tianxiao Xu; Sifan Yu; Huan Yu; Huan Tang; Junya Yan; Xiaowen Wu; Jiayi Yu; Zhihong Chi; Lu Si; Chuanliang Cui; Xinan Sheng; Yan Kong; Jun Guo

PurposeMucosal melanoma is a relatively rare subtype of melanoma for which no clearly established therapeutic strategy exists. The genes of the mTOR signalling pathway have drawn great attention as key targets for cancer treatment, including melanoma. In this study, we aimed to investigate the mutation status of the upstream mTOR regulator TSC1 and evaluated its correlation with the clinicopathological features of mucosal melanoma.MethodsWe collected 91 mucosal melanoma samples for detecting TSC1 mutations. All the coding exons of TSC1 were amplified by PCR and subjected to Sanger sequencing. Expression level of TSC1 encoding protein (hamartin) was detected by immunohistochemistry. The activation of mTOR pathway was determined by evaluating the phosphorylation status of S6RP and 4E-BP1.ResultsThe overall mutation frequency of TSC1 was found to be 17.6% (16/91 patients). TSC1 mutations were more inclined to occur in advanced mucosal melanoma (stages III and IV). In the 16 patients with TSC1 mutations, 14 different mutations were detected, affecting 11 different exons. TSC1 mutations were correlated with upregulation of S6RP phosphorylation but were unrelated to 4E-BP1 phosphorylation or hamartin expression. Mucosal melanoma patients with TSC1 mutations had a worse outcome than patients without TSC1 mutations (24.0 versus 34.0xa0months, Pu2009=u20090.007).ConclusionsOur findings suggest that TSC1 mutations are frequent in mucosal melanoma. TSC1 mutations can activate the mTOR pathway through phospho-S6RP and might be a poor prognostic predictor of mucosal melanoma. Our data implicate the potential significance of TSC1 mutations for effective and specific drug therapy for mucosal melanoma.

Collaboration


Dive into the Tianxiao Xu'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