Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Shannon Chuai.
Clinical Cancer Research | 2017
A. Li; Jin-Ji Yang; Zhang X; Zhou Zhang; Jian Su; Lan-Ying Gou; Yu Bai; Zhenfan Yang; Han Han-Zhang; Wen-Zhao Zhong; Shannon Chuai; Qi Zhang; Zhi Xie; Hong-Fei Gao; Hua-Jun Chen; Zhen Wang; Zheng Wang; Xue-Ning Yang; Bin-Chao Wang; Bin Gan; Zhi-Hong Chen; Ben-Yuan Jiang; Si-Pei Wu; Si-Yang Liu; Chong-Rui Xu; Yi-Long Wu
Purpose: MET amplification, responsible for 20% of acquired resistance to EGFR tyrosine kinase inhibitor (TKI) in patients with advanced non–small cell lung cancer (NSCLC), presents an attractive target. Numerous studies have conferred susceptibility of MET mutations and focal amplification to targeted MET-TKIs. However, the mechanism underlying MET-TKIs–induced resistance remains elusive. Experimental Design: We conducted a cohort of 12 patients with advanced NSCLC who developed resistance to a combinatorial therapy consisting of gefitinib and a type I MET-TKI. We performed capture-based targeted ultra-deep sequencing on serial tumor biopsies and plasmas ctDNA samples to detect and quantify genetic alterations. Results: We identified 2 newly acquired MET mutations, Y1248H and D1246N, in 2 patients and further confirmed their resistance against type I MET-TKIs in silco, in vitro, and in vivo. Interestingly, NIH3T3 cells harboring either mutation exhibited responses to type II MET-TKIs, suggesting sequential use of MET-TKIs may offer a more durable response. In addition, we also discovered that EGFR amplification may act as an alternative MET-TKI resistance mechanism. Conclusions: Our study provides insight into the diversity of mechanisms underlying MET-TKI–induced resistance and highlights the potential of sequential use of MET-TKIs. Clin Cancer Res; 23(16); 4929–37. ©2017 AACR.
Journal of Thoracic Oncology | 2017
Wenbin Li; Jing Zhang; Lei Guo; Shannon Chuai; Ling Shan; Jianming Ying
Introduction: The purpose of this study was to explore the complicated rearrangement mechanisms underlying cases with atypical and negative anaplastic lymphoma receptor tyrosine kinase gene (ALK) fluorescence hybridization (FISH) and positive immunohistochemistry (IHC) results and to stress the importance of combinational assay of these two methods in current pathological diagnosis. Methods: A total of 3128 NSCLCs were screened for ALK fusions through both FISH analysis and IHC assays with Ventana‐D5F3 antibody. Fourteen cases with atypical and negative FISH results with the current criteria and positive IHC results were analyzed with targeted next‐generation sequencing (NGS). Results: Of the 3128 cases tested, 228 (7.3%) and 214 (6.8%) were ALK positive by IHC and FISH, respectively. Fourteen cases with negative and atypical FISH results all demonstrated IHC positivity. Of 2991 cases, eight (0.27%) with negative FISH results demonstrated echinoderm microtubule associated protein like 4 gene (EML4)‐ALK fusions revealed by targeted NGS, and the relative abundance of fusion ranged from 0.9% to 46.8%. Three of 2991 cases (0.1%) did not exhibit any type of ALK fusions. In addition, two patients showed an isolated 5′ side signal and targeted NGS revealed two novel ALK partner genes, baculoviral IAP repeat containing 6 gene (BIRC6) and phosphatidylinositol binding clathrin assembly protein gene (PICALM). One patient showed an isolated and attenuated 3′ red signal and demonstrated a novel translocation partner with CCAAT/enhancer binding protein zeta gene (CEBPZ). Of all the patients, four received crizotinib treatment and demonstrated partial responses at the end of follow‐up. Conclusions: Our study showed that patients with negative and atypical ALK FISH patterns may have positive results for IHC testing and harbor the translocation partners of EML4 or other genes. Therefore, additional testing with NGS should be conducted to explore the molecular mechanisms underlying the complicated gene rearrangement events.
Journal of Thoracic Oncology | 2017
Xiaowei Mao; Zhou Zhang; Xiaoxuan Zheng; Fangfang Xie; Feidie Duan; Liyan Jiang; Shannon Chuai; Han Han-Zhang; Baohui Han; Jiayuan Sun
Introduction Circulating tumor DNA (ctDNA), which represents an unbiased way to assess tumor genetic profile noninvasively, facilitates studying intratumor heterogeneity. Although intratumor heterogeneity has been elucidated substantially in a few cancer types, including NSCLC, how it influences the ability of tumor cells harboring different genetic abnormalities in releasing their DNA remains elusive. We designed a capture‐based panel targeting NSCLC to detect and quantify genetic alterations from plasma samples by using deep sequencing. By applying the panel to paired biopsy and plasma samples, we imputed and compared the ctDNA‐releasing efficiency in subclones harboring distinct genetic variants. Methods We collected 40 pairs of matched biopsy and plasma samples from patients with advanced lung cancer and applied capture‐based sequencing using our LungPlasma panel, which consists of critical exons and introns of 168 genes. We derived a normalized relative allelic fraction score (NRAFS) to reflect ctDNA‐releasing efficiency. Results By using mutations detected in biopsy samples as a reference, we achieved 87.2% by‐variant sensitivity, including for single‐nucleotide variants, insertions or deletions, and gene fusions. Furthermore, the by‐variant sensitivity for the seven most critical and actionable genes was 96.2%. The average NRAFS for subclones carrying mutations from seven actionable genes was 0.877; in contrast, the average NRAFS for other mutations was 0.658. Mutations from four genes involved in cell cycle pathways had a particularly low NRAFS (0.480) compared with the other two groups (p = 0.07). Conclusions We have demonstrated that subclones carrying driver mutations are more prone to release DNA. We have also demonstrated the quantitative ability of capture‐based sequencing, paving its way for routine utilization in clinical settings.
Annals of Oncology | 2018
Y. Li; Ben-Yuan Jiang; J.-. Yang; Zhang X; Zhou Zhang; J Y Ye; Wen-Zhao Zhong; Hai-Yan Tu; Hua-Jun Chen; Wang Z; Chong-Rui Xu; B. Wang; H J Du; Shannon Chuai; Han Han-Zhang; Jian Su; Xue-Ning Yang; Wei-Bang Guo; Hong-Hong Yan; Y. Liu; Li-Xu Yan; Huang B; Ming-Ying Zheng; Yi-Long Wu
Background Leptomeningeal metastases (LM) are more frequent in non-small-cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutations. Due to limited access to leptomeningeal lesions, the purpose of this study was to explore the potential role of cerebrospinal fluid (CSF) as a source of liquid biopsy in patients with LM. Patients and methods Primary tumor, CSF, and plasma in NSCLC with LM were tested by next-generation sequencing. In total, 45 patients with suspected LM underwent lumbar puncture, and those with EGFR mutations diagnosed with LM were enrolled. Results A total of 28 patients were enrolled in this cohort; CSF and plasma were available in 26 patients, respectively. Driver genes were detected in 100% (26/26), 84.6% (22/26), and 73.1% (19/26) of samples comprising CSF cell-free DNA (cfDNA), CSF precipitates, and plasma, respectively; 92.3% (24/26) of patients had much higher allele fractions in CSF cfDNA than the other two media. Unique genetic profiles were captured in CSF cfDNA compared with those in plasma and primary tissue. Multiple copy number variations (CNVs) were mainly identified in CSF cfDNA, and MET copy number gain identified in 47.8% (11/23) of patients was the most frequent one, while other CNVs included ERBB2, KRAS, ALK, and MYC. Moreover, loss of heterozygosity (LOH) of TP53 was identified in 73.1% (19/26) CSF cfDNA, which was much higher than that in plasma (2/26, 7.7%; P < 0.001). There was a trend towards a higher frequency of concomitant resistance mutations in patients with TP53 LOH than those without (70.6% versus 33.3%; P = 0.162). EGFR T790M was identified in CSF cfDNA of 30.4% (7/23) of patients who experienced TKI progression. Conclusion CSF cfDNA could reveal the unique genetic profiles of LM and should be considered as the most representative liquid biopsy medium for LM in EGFR-mutant NSCLC.
Lung Cancer | 2018
Yan Li; Tong-Tong Zhang; Jing Zhang; Wenbin Li; Pei Yuan; Puyuan Xing; Zhou Zhang; Shannon Chuai; J. Li; Jianming Ying
INTRODUCTION Anaplastic lymphoma kinase (ALK) rearrangements are present in approximately 5% of non-small-cell lung cancers (NSCLCs). NSCLCs with ALK-rearrangement can be effectively treated with crizotinib. However, magnitude and duration of responses are found to be heterogeneous. This study explored the clinical efficacy of crizotinib in different ALK variants. METHODS Among 96 ALK-rearrangement patients treated with crizotinib, 60 patients were identified with tumor specimens that could be evaluated by next-generation sequencing (NGS). We retrospectively evaluated the efficacy of crizotinib in different ALK variants. RESULTS The median Progression-free survival (PFS) of the 96 ALK-rearrangement patients was 14.17 months. Among the 60 patients with NGS results, the most frequent variants were variant 3a/b (33.33%), variant 1 (23.33%) and variant 2 (15.00%). The percentage of rare EML4-ALK variants and non EML4-ALK variants were 10.00% and 18.33%. Survival analysis showed that patients with variant 2 appeared to have longer PFS than others (P = .021); also, patients with TP53 mutation seemed to have an unfavorable PFS than those with TP53 wild-type with a borderline p value (P = .068). After adjusting for other baseline characteristics, EML4-ALK variant 2 was identified as an important factor for a better PFS of crizotinib. We also found that patients with variant 3a/b had shorter duration of response to crizotinib; however, no significant difference of PFS was observed between the PFS of variant3a/b and non-v3 EML4-ALK variants. CONCLUSIONS Our results indicate prolonged PFS in patients with EML4-ALK variant 2.
Lung Cancer | 2018
Panwen Tian; Ye Wang; Wei-Ya Wang; Yalun Li; Ke Wang; Xiaowei Cheng; Yuan Tang; Han Han-Zhang; Junyi Ye; Shannon Chuai; Weimin Li
INTRODUCTION De novo T790 M mutation in EGFR has been reported in various studies. However, its genetic characteristics and association with EGFR tyrosine kinase inhibitors (TKIs) treatment response remain poorly studied. METHODS We retrospectively screened 1228 consecutive non-small cell lung cancer (NSCLC) patients and identified 29 de novo T790 M carriers. Capture-based targeted deep sequencing was conducted on 21 eligible samples as well as a 20-sample cohort with acquired T790 M mutation after EGFR-TKIs treatment. We characterized and compared their mutational profiles using a panel consisting of 168 lung cancer-related genes. RESULTS De novo T790 M mutation was found in 5.8% of the TKI-naive patients harboring EGFR activating mutations. Among the de novo T790 M samples, T790 M was significantly more likely to coexist with L858R than with 19del (76.2% vs. 23.8%) compared to the acquired T790 M cohort (30.0% vs. 70.0%) (p = 0.003). These two groups harbored different concurrent gene mutations as well. Notably, the ratio of allele frequency (AF) of the T790 M mutation to the EGFR activating mutation in each patient, defined as the T790 M relative AF (RAF), differed significantly between the de novo and acquired T790 M cohorts (86.1% vs. 22.3%, p < 0.0001). Among the 10 patients with de novo T790 M who received the 1st-generation EGFR-TKIs treatment, interestingly, the only one who achieved partial response (PR) had the lowest T790 M RAF of 19.7%. The other 9 patients with an average T790 M RAF of 85.9% (±22.6%) achieved stable disease or progressive disease as the best response. One patient, treated with osimertinib after erlotinib failure, achieved PR and the therapeutic response sustained for more than 14.5 months. CONCLUSION The molecular characteristics of de novo T790 M carriers differ distinctly from acquired T790 M carriers. The RAF of EGFR T790 M mutation may serve as a predictive biomarker for treatment response to EGFR-TKIs. Osimertinib is potentially an effective drug for the treatment of NSCLC with de novo T790 M.
Journal of Thoracic Oncology | 2018
M. Zheng; Y. Li; Ben-Yuan Jiang; Hai-Yan Tu; Zhang X; Junyi Ye; W. Tang; Shannon Chuai; Y. Wu
Journal of Thoracic Oncology | 2018
Y. Li; Ben-Yuan Jiang; M. Zheng; Hai-Yan Tu; J.-. Yang; Zhang X; Junyi Ye; Qinghua Zhou; W. Zhong; C. Zhang; Shannon Chuai; Y. Wu
Journal of Thoracic Oncology | 2018
Qinghua Zhou; Y. Zhang; Zhu Chen; Chong-Rui Xu; J.-. Yang; Zhang X; Xiao-Yan Bai; Hong-Hong Yan; Zhi Xie; Wuxia Li; Shannon Chuai; Junyi Ye; H. Zhang-Han; Zhou Zhang; Y. Wu
Journal of Thoracic Oncology | 2018
Y. Zhang; Qinghua Zhou; Zhu Chen; Chong-Rui Xu; J.-. Yang; Zhang X; Xiao-Yan Bai; Hong-Hong Yan; Wuxia Li; Shannon Chuai; Junyi Ye; H. Zhang-Han; Zhou Zhang; Y. Wu