1
|
Ding P, Wu J, Wu H, Ma W, Li T, Yang P, Guo H, Tian Y, Yang J, Er L, Gu R, Zhang L, Meng N, Li X, Guo Z, Meng L, Zhao Q. Preoperative liquid biopsy transcriptomic panel for risk assessment of lymph node metastasis in T1 gastric cancer. J Exp Clin Cancer Res 2025; 44:43. [PMID: 39915770 PMCID: PMC11804050 DOI: 10.1186/s13046-025-03305-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND The increasing incidence of early-stage T1 gastric cancer (GC) underscores the need for accurate preoperative risk stratification of lymph node metastasis (LNM). Current pathological assessments often misclassify patients, leading to unnecessary radical surgeries. METHODS Through analysis of transcriptomic data from public databases and T1 GC tissues, we identified a 4-mRNA panel (SDS, TESMIN, NEB, and GRB14). We developed and validated a Risk Stratification Assessment (RSA) model combining this panel with clinical features using surgical specimens (training cohort: n = 218; validation cohort: n = 186), gastroscopic biopsies (n = 122), and liquid biopsies (training cohort: n = 147; validation cohort: n = 168). RESULTS The RSA model demonstrated excellent predictive accuracy for LNM in surgical specimens (training AUC = 0.890, validation AUC = 0.878), gastroscopic biopsies (AUC = 0.928), and liquid biopsies (training AUC = 0.873, validation AUC = 0.852). This model significantly reduced overtreatment rates from 83.9 to 44.1% in tissue specimens and from 84.4 to 56.0% in liquid biopsies. The 4-mRNA panel showed specificity for T1 GC compared to other gastrointestinal cancers (P < 0.001). CONCLUSIONS We developed and validated a novel liquid biopsy-based RSA model that accurately predicts LNM in T1 GC patients. This non-invasive approach could significantly reduce unnecessary surgical interventions and optimize treatment strategies for high-risk T1 GC patients.
Collapse
Affiliation(s)
- Ping'an Ding
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Jiaxiang Wu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Haotian Wu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Wenqian Ma
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
- Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Tongkun Li
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Peigang Yang
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Honghai Guo
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Yuan Tian
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Jiaxuan Yang
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Limian Er
- Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Renjun Gu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430065, China
| | - Ning Meng
- Department of General Surgery, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, 050050, China
| | - Xiaolong Li
- Department of General Surgery, Baoding Central Hospital, Baoding, Hebei, 071030, China
| | - Zhenjiang Guo
- General Surgery Department, Hengshui People's Hospital, Hengshui, Hebei, 053099, China
| | - Lingjiao Meng
- Research Center, Tumor Research Institute of the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| | - Qun Zhao
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China.
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China.
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China.
| |
Collapse
|
2
|
Marcisz-Grzanka K, Kotowicz B, Nowak A, Winiarek M, Fuksiewicz M, Kowalska M, Tysarowski A, Olesinski T, Palucki J, Sulkowska U, Kolasinska-Cwikla A, Wyrwicz LS. Interleukin-6 as a Predictive Factor of Pathological Response to FLOT Regimen Systemic Treatment in Locally Advanced Gastroesophageal Junction or Gastric Cancer Patients. Cancers (Basel) 2024; 16:757. [PMID: 38398148 PMCID: PMC10887209 DOI: 10.3390/cancers16040757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Perioperative treatment is a gold standard in locally advanced gastric cancer or GEJ cancer in the Western population. Unfortunately, the response rate after neoadjuvant chemotherapy (NAC) remains limited. Moreover, there are currently no biomarkers enabling an individual prediction of therapeutic efficacy. The aim of this study was the identification of serum biomarkers of early response to NAC. METHODS We conducted this prospective study in the MSCNRIO in Warsaw, Poland. A total of 71 patients and 15 healthy volunteers gave informed consent. Complete blood count, carcinoembryonic antigen (CEA), carcinoma antigen 125 (CA125), carcinoma antigen 19.9 (CA19.9), and fibrinogen (F) were measured at baseline and before every cycle. Circulating tumour cells (CTCs) and interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8), and interleukin-10 (IL-10) were measured in a pilot group of 40 patients at baseline and before cycle two (C2) and cycle three (C3). RESULTS Of all the measured parameters, only the IL-6 serum level was statistically significant. The IL-6 level before C2 of chemotherapy was significantly decreased in the complete pathological response (pCR) vs. the non-pCR group (3.71 pg/mL vs. 7.63 pg/mL, p = 0.004). In all patients with an IL-6 level below 5.0 pg/mL in C2, tumour regression TRG1a/1b according to the Becker classification and ypN0 were detected in postoperative histopathological specimens. The IL-6 level before C1 of chemotherapy was significantly elevated in ypN+ vs. ypN0 (7.69 pg/mL vs. 2.89 pg/mL, p = 0.022). CONCLUSIONS The trial showed that an elevated level of IL-6 prior to treatment and C2 might be a predictor of pathological response to NAC.
Collapse
Affiliation(s)
- Katarzyna Marcisz-Grzanka
- Department of Clinical Oncology and Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), Wawelska 15, 02-034 Warsaw, Poland; (M.W.); (A.K.-C.)
| | - Beata Kotowicz
- Cancer Biomarker and Cytokines Laboratory Unit, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), W.K. Roentgena 5, 02-781 Warsaw, Poland; (B.K.); (M.F.); (M.K.)
| | - Aleksandra Nowak
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), W.K. Roentgena 5, 02-781 Warsaw, Poland; (A.N.); (A.T.)
| | - Mariola Winiarek
- Department of Clinical Oncology and Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), Wawelska 15, 02-034 Warsaw, Poland; (M.W.); (A.K.-C.)
| | - Malgorzata Fuksiewicz
- Cancer Biomarker and Cytokines Laboratory Unit, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), W.K. Roentgena 5, 02-781 Warsaw, Poland; (B.K.); (M.F.); (M.K.)
| | - Maria Kowalska
- Cancer Biomarker and Cytokines Laboratory Unit, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), W.K. Roentgena 5, 02-781 Warsaw, Poland; (B.K.); (M.F.); (M.K.)
| | - Andrzej Tysarowski
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), W.K. Roentgena 5, 02-781 Warsaw, Poland; (A.N.); (A.T.)
| | - Tomasz Olesinski
- Department of Oncological Surgery and Neuroendocrine Tumors, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), W.K. Roentgena 5, 02-781 Warsaw, Poland;
| | - Jakub Palucki
- Department of Radiology, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), W.K. Roentgena 5, 02-781 Warsaw, Poland;
| | - Urszula Sulkowska
- National Cancer Registry, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), Wawelska 15B, 02-034 Warsaw, Poland;
| | - Agnieszka Kolasinska-Cwikla
- Department of Clinical Oncology and Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), Wawelska 15, 02-034 Warsaw, Poland; (M.W.); (A.K.-C.)
| | - Lucjan Stanislaw Wyrwicz
- Department of Clinical Oncology and Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), Wawelska 15, 02-034 Warsaw, Poland; (M.W.); (A.K.-C.)
| |
Collapse
|
3
|
Jin T, Liang PP, Chen ZH, He FJ, Li ZD, Chen ZW, Hu JK, Yang K. Association between circulating tumor cells in the peripheral blood and the prognosis of gastric cancer patients: a meta-analysis. Ther Adv Med Oncol 2023; 15:17588359231183678. [PMID: 37435560 PMCID: PMC10331349 DOI: 10.1177/17588359231183678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/31/2023] [Indexed: 07/13/2023] Open
Abstract
Background Research on the correlation between circulating tumor cells (CTCs) and gastric cancer (GC) has increased rapidly in recent years. However, whether CTCs are associated with GC patient prognosis is highly controversial. Objective This study aims to evaluate the value of CTCs to predict the prognosis of GC patients. Design A meta-analysis. Data Sources and Methods We searched the PubMed, Embase, and Cochrane Library databases for studies that reported the prognostic value of CTCs in GC patients before October 2022. The association between CTCs and overall survival (OS) and disease-free survival (DFS)/recurrence-free survival (RFS) and progression-free survival (PFS) of GC patients was assessed. Subgroup analyses were stratified by sampling times (pre-treatment and post-treatment), detection targets, detection method, treatment method, tumor stage, region, and HR (Hazard Ratio) extraction methods. Sensitivity analysis was performed by removing individual studies to assess the stability of the results. Publication bias was evaluated using funnel plots, Egger's test, and Begg's test. Results We initially screened 2000 studies, of which 28 were available for further analysis, involving 2383 GC patients. The pooled analysis concluded that the detection of CTCs was associated with poor OS (HR = 1.933, 95% CI 1.657-2.256, p < 0.001), DFS/RFS (HR = 3.228, 95% CI 2.475-4.211, p < 0.001), and PFS (HR = 3.272, 95% CI 1.970-5.435, p < 0.001). Furthermore, the subgroup analysis stratified by tumor stage (p < 0.01), HR extraction methods (p < 0.001), detection targets (p < 0.001), detection method (p < 0.001), sampling times (p < 0.001), and treatment method (p < 0.001) all showed that CTC detection was associated with poor OS and DFS/RFS for GC patients. Furthermore, the study showed that CTCs were associated with the poor DFS/RFS of GC when CTCs were detected for patients from Asian or No-Asian regions (p < 0.05). In addition, higher CTCs predicted poorer OS for GC patients who are from Asian regions (p < 0.001), but without statistical difference for GC patients from No-Asian regions (p = 0.490). Conclusion CTC detection in peripheral blood was associated with poor OS, DFS/RFS, and PFS in patients with GC.
Collapse
Affiliation(s)
- Tao Jin
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pan-Ping Liang
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ze-Hua Chen
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng-Jun He
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ze-Dong Li
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zheng-Wen Chen
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian-Kun Hu
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kun Yang
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
4
|
Liu S, Lin D, Lai Y, Hou L, Lin T, Zhao S. Gas-Mediated Immunoassay for the Carcinoembryonic Antigen at Atmospheric Pressure with Smartphone Coupling with the Fluorescence Quenching Length of Perovskite Capillary. Anal Chem 2022; 94:18074-18082. [PMID: 36516357 DOI: 10.1021/acs.analchem.2c04622] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
By combining the photothermal properties of the 3,3',5,5'-tetramethylbenzidine oxidation product (TMBox) with the sensitive quenching of perovskite fluorescence by ammonia gas, a gas-mediated immunoassay at atmospheric pressure was constructed, which took the fluorescence quenching length of perovskite fluorescent capillary as the signal output. First, a CsPbBr3 perovskite with surface modification of 3-aminopropyl triethoxysilane was synthesized by thermal injection and decorated to the capillary wall by glutaraldehyde cross-linking. In the presence of H2O2 and the tumor marker carcinoembryonic antigen (CEA), TMB was oxidized to TMBox by the horseradish peroxidase (HRP)-labeled CEA antibody. The photothermal effect of TMBox at 808 nm laser irradiation increases the concentration of ammonia gas, and the prepared fluorescent capillary can respond sensitively to ammonia gas. The fluorescence quenching length can be observed by the naked eye for a semiquantitative evaluation of CEA concentration. At the same time, we developed a mobile APP for the first time to measure the fluorescence quenching length. In the range of 0-20 ng mL-1, the quenching length increased linearly with the increase in CEA concentration, and the detection limit was 0.078 ng mL-1. This method has been successfully used for the detection of CEA in human serum with a recovery of 95.8%-106.5%.
Collapse
Affiliation(s)
- Shendong Liu
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, People's Republic of China
| | - Danxuan Lin
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, People's Republic of China
| | - Yunping Lai
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, People's Republic of China
| | - Li Hou
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, People's Republic of China
| | - Tianran Lin
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, People's Republic of China
| | - Shulin Zhao
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, People's Republic of China
| |
Collapse
|
5
|
Tade RS, Patil PO. Fabrication of Poly-l-lysine-Functionalized Graphene Quantum Dots for the Label-Free Fluorescent-Based Detection of Carcinoembryonic Antigen. ACS Biomater Sci Eng 2021; 8:470-483. [PMID: 34967597 DOI: 10.1021/acsbiomaterials.1c01087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The diagnosis of tumor biomarkers is an attentive approach for the early detection and treatment of cancer. However, a cost-effective, simple, rapid, selective, and sensitive method is a basic prerequisite for diagnostic research. Herein, we present a novel fluorescence-based label-free sensing strategy for the sensitive and selective detection of carcinoembryonic antigen (CEA) using poly-l-lysine (PLL)-functionalized graphene quantum dots (GQDs). The GQDs were synthesized using a greener method by employing carbonized peanut shell (PNS) waste as a carbon source, and functionalization was accomplished using PLL (PLL-GQDs). The fluorescence stability of the PLL-GQDs was tested in a variety of solvent systems and pH solutions. When compared to nonfunctionalized GQDs (PNS-GQDs), prepared PLL-GQDs demonstrated increased fluorescence lifetime, high quantum yield, excellent photostability, biocompatibility, and greater cellular uptake. The PLL-GQDs with abundant surface amine and carboxylic groups showed selective interactions with an activated CEA antibody (CEA-Ab), resulting in the quenching of fluorescence signals. Because of the strong bioaffinity of CEA to the CEA-Ab, the antibody was unwrapped, resulting in the formation of an antibody-antigen complex and the recovery of fluorescence. As a result of this relationship, a turn "on-off-on" sensing mechanism with a strong response to CEA concentration (0.01 ng mL-1 to 100 μg mL-1) and a detection limit of 1.19 pg mL-1 was demonstrated. Furthermore, the fabricated CEA immunosensor (CEA-Ab@PLL-GQDs) performed admirably in real sample analysis, with an average recovery of 98.32%. The cellular uptake performance of PLL-GQDs was also demonstrated in the A427 cell lines, exhibiting a greater cellular uptake potential than PNS-GQDs. The cellular bioimaging study demonstrates that PLL-GQDs can be used for additional therapeutic and biological applications.
Collapse
Affiliation(s)
- Rahul Shankar Tade
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur 425405, Maharashtra, India
| | - Pravin Onkar Patil
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur 425405, Maharashtra, India
| |
Collapse
|
6
|
Gao Y, Xi H, Wei B, Cui J, Zhang K, Li H, Cai A, Shen W, Li J, Rosell R, Chao J, Chen T, Klempner S, Qiao Z, Chen L. Association Between Liquid Biopsy and Prognosis of Gastric Cancer Patients: A Systematic Review and Meta-Analysis. Front Oncol 2019; 9:1222. [PMID: 31850190 PMCID: PMC6901923 DOI: 10.3389/fonc.2019.01222] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 10/25/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Reports regarding liquid biopsy and gastric cancer (GC) have emerged rapidly in recent decades, yet their prognostic value still remains controversial. This study was aimed to assess the impact of liquid biopsy, including circulating tumor cells (CTCs) and cell-free nucleic acids, on GC patients' prognosis. Methods: PubMed, Medline, EMBASE, and ClinicalTrial.gov databases were searched for studies that report GC patient survival data stratified by CTC/circulating tumor DNA (ctDNA)/circulating miRNAs' status. The hazard ratios (HRs) and their 95% confidence intervals (CIs) for patients' overall survival (OS) and disease-free survival (DFS)/progression-free survival (PFS) were recorded or calculated depending on circulating target status. Results: We initially identified 4,221 studies, from which 43 were eligible for further analysis, comprising 3,814 GC patients. Pooled analyses showed that detection of certain CTCs, ctDNA, and circulating miRNA was associated with poorer OS (CTCs: HR = 1.84, 95%CI 1.50–2.26, p < 0.001; ctDNA: HR = 1.78, 95%CI 1.36–2.34, p < 0.001; circulating miRNA: HR = 1.74, 95%CI 1.13–2.69, p < 0.001) and DFS/PFS (CTCs: HR = 3.39, 95%CI 2.21–5.20, p < 0.001; ctDNA: HR = 2.38, 95%CI 1.31–4.32, p = 0.004; circulating miRNA: HR = 3.30, 95%CI 2.39–4.55, p < 0.001) of GC patients, regardless of disease stage and time point at which sample is taken (at baseline or post-treatment). Conclusions: The presence of CTCs and/or cellular components identifies a group of GC with poorer prognosis. Among circulating markers, CTCs demonstrated a stronger and more stable predictive value for late-stage disease and among Mongolian populations with GC. Less data are available for ctDNA and miRNA; however, their presence may also reflect aggressive biology and warrants further prospective study.
Collapse
Affiliation(s)
- Yunhe Gao
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China.,General Surgery Institute, Chinese PLA General Hospital, Beijing, China
| | - Hongqing Xi
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Bo Wei
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China.,General Surgery Institute, Chinese PLA General Hospital, Beijing, China
| | - Jianxin Cui
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Kecheng Zhang
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Hua Li
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Aizhen Cai
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China.,General Surgery Institute, Chinese PLA General Hospital, Beijing, China
| | - Weishen Shen
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China.,Nanjing General Hospital of Nanjing Military Command, Nanjing, China
| | - Jiyang Li
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China.,General Surgery Institute, Chinese PLA General Hospital, Beijing, China
| | - Rafael Rosell
- Catalan Institute of Oncology, Germans Trias i Pujol Health Science Institute and Hospital, Barcelona, Spain
| | - Joseph Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Tianhui Chen
- Department of Cancer Prevention, Institute of Cancer and Basic Medicine (ICBM), Zhejiang Provincial Office for Cancer Prevention and Control, Cancer Hospital of the University of CAS, Chinese Academy of Sciences (CAS), Hangzhou, China
| | - Samuel Klempner
- The Angeles Clinic and Research Institute, Los Angeles, CA, United States.,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Zhi Qiao
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Lin Chen
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China.,General Surgery Institute, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Ye X, Li G, Han C, Han Q, Shang L, Su H, Han B, Gong Y, Lu G, Peng T. Circulating tumor cells as a potential biomarker for postoperative clinical outcome in HBV-related hepatocellular carcinoma. Cancer Manag Res 2018; 10:5639-5647. [PMID: 30532586 PMCID: PMC6245351 DOI: 10.2147/cmar.s175489] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND This study aimed to determine if the number of circulating tumor cells (CTCs) and changes in their numbers affected tumor recurrence and metastasis after surgical resection in patients with hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). METHODS The primary endpoints were overall survival (OS) and progression-free survival (PFS). A total of 42 patients with HCC were selected from the First Affiliated Hospital of Guangxi Medical College from 2014 to 2017. CTCs were counted 1 day prior to and 30 days after surgical excision of HCC using the CanPatrol™ system. RESULTS Numbers of CTCs (> 2 CTCs and > 5 CTCs per 5 ml peripheral blood) were significantly associated with Edmondson stage in HBV-related HCC prior to surgery (P = 0.004 and 0.014, respectively). However there were no significant associations between other tested clinicopathological factors and CTC counts. Postoperative CTC counts (> 2 and > 5) and pre/postoperative change in CTC counts were significantly associated with PFS (P = 0.02, 0.009, and 0.001, respectively), but not with OS. Receiver operating characteristic curve analysis showed that pre/postoperative changes in the CTC count were a better predictor of performance than absolute count. The postoperative CTC count was also significantly associated with positive TP53 expression (P < 0.05). CONCLUSION These results demonstrate that postoperative CTC counts (> 2 and > 5) and changes in CTC counts may be independent prognostic indicators for PFS in patients with HBV-related HCC, with the change in number of CTCs showing better predictive performance.
Collapse
Affiliation(s)
- Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Guanghui Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Quanfa Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Liming Shang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Bowen Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Yizhen Gong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Guodong Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi Province, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China, ,Correspondence: Tao Peng, Department of Hepatobiliary Surgery, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning 530021, Guangxi Province, China, Tel +86 771 535 6528, Fax +86 771 535 0031, Email
| |
Collapse
|
8
|
Li TT, Liu H, Yu J, Shi GY, Zhao LY, Li GX. Prognostic and predictive blood biomarkers in gastric cancer and the potential application of circulating tumor cells. World J Gastroenterol 2018; 24:2236-2246. [PMID: 29881233 PMCID: PMC5989238 DOI: 10.3748/wjg.v24.i21.2236] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/27/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC), with its high incidence and mortality rates, is a highly fatal cancer that is common in East Asia particularly in China. Its recurrence and metastasis are the main causes of its poor prognosis. Circulating tumor cells (CTCs) or other blood biomarkers that are released into the circulating blood stream by tumors are thought to play a crucial role in the recurrence and metastasis of gastric cancer. Therefore, the detection of CTCs and other blood biomarkers has an important clinical significance; in fact, they can help predict the prognosis, assess the staging, monitor the therapeutic effects and determine the drug susceptibility. Recent research has identified many blood biomarkers in GC, such as various serum proteins, autoantibodies against tumor associated antigens, and cell-free DNAs. The analysis of CTCs and circulating cell-free tumor DNA (ctDNA) in the peripheral blood of patients with gastric cancer is called as liquid biopsy. These blood biomarkers provide the disease status for individuals and have clinical meaning. In this review, we focus on the recent scientific advances regarding CTCs and other blood biomarkers, and discuss their origins and clinical meaning.
Collapse
Affiliation(s)
- Ting-Ting Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Guang-Yao Shi
- Division of Cardiology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
| | - Li-Ying Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Guo-Xin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
9
|
Yang C, Zou K, Yuan Z, Guo T, Xiong B. Prognostic value of circulating tumor cells detected with the CellSearch System in patients with gastric cancer: evidence from a meta-analysis. Onco Targets Ther 2018. [PMID: 29520152 PMCID: PMC5833773 DOI: 10.2147/ott.s154114] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Circulating tumor cells (CTCs) have been proposed as a marker for predicting the prognosis of cancer. However, the prognostic value of CTCs detected with the CellSearch System in patients with gastric cancer (GC) remains controversial. We performed a meta-analysis of available studies to investigate this topic. Methods Two authors systematically searched the studies independently in PubMed, Science Citation Index, Cochrane Database, Embase, and the references in relevant studies (up to September 2017) using keywords. Our meta-analysis was performed in Stata software, version 12.0 (2011; Stata Corp, College Station, TX, USA), with the risk ratio (RR), hazard ratio (HR), and 95% CI as the effect measures. Subgroup analyses and meta-regression were also conducted. Results Seven studies (including eight sets of data) containing 579 patients with GC from four countries were included in this meta-analysis. The pooled results showed CTC-positive status detected by the CellSearch System was significantly associated with poor overall survival (HR =2.09, 95% CI [1.71, 2.55], P<0.001, I2=31.5%) and progression-free survival (HR =2.11, 95% CI [1.25, 3.57], P=0.005, I2=75.6%) of patients with GC, regardless of sampling time. The disease control rate of CTC-positive group was lower than that of CTC-negative group for both baseline and intra-therapy, although no statistical difference existed at both sampling time points (baseline: 69.5% versus 81.8%, RR=0.79, 95% CI [0.54, 1.16], P=0.23, I2=68.0%; intra-therapy: 50.0% versus 85.9%, RR=0.24, 95% CI [0.02, 3.13], P=0.28, I2=87.4%). Conclusion Our meta-analysis demonstrated that CTCs detected with the CellSearch System from the peripheral blood had significant prognostic value and might predict poor response to chemotherapy for patients with GC.
Collapse
Affiliation(s)
- Chaogang Yang
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University.,Hubei Key Laboratory of Tumor Biological Behaviors.,Hubei Cancer Clinical Study Center
| | - Kun Zou
- Department of Oncology, Central Hospital of Wuhan, Wuhan, Hubei, People's Republic of China
| | - Zewei Yuan
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University.,Hubei Key Laboratory of Tumor Biological Behaviors.,Hubei Cancer Clinical Study Center
| | - Tangxi Guo
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University.,Hubei Key Laboratory of Tumor Biological Behaviors.,Hubei Cancer Clinical Study Center
| | - Bin Xiong
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University.,Hubei Key Laboratory of Tumor Biological Behaviors.,Hubei Cancer Clinical Study Center
| |
Collapse
|
10
|
Arigami T, Uenosono Y, Yanagita S, Okubo K, Kijima T, Matsushita D, Amatatsu M, Kurahara H, Maemura K, Natsugoe S. Clinical significance of circulating tumor cells in blood from patients with gastric cancer. Ann Gastroenterol Surg 2017; 1:60-68. [PMID: 29863113 PMCID: PMC5881297 DOI: 10.1002/ags3.12005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 02/27/2017] [Indexed: 12/19/2022] Open
Abstract
Circulating tumor cells (CTC) have been focused on as a target for detecting occult tumors, predicting therapeutic responses and prognoses, and monitoring postoperative recurrence in the clinical management of patients with various malignancies, including gastric cancer. Recent advances in molecular diagnostic tools have contributed to high sensitivity and specificity for the detection of CTC. A conspicuous disparity exists in the incidence of CTC among studies. However, a close relationship has been reported between positivity for CTC and well‐known prognostic clinicopathological factors including depth of tumor invasion, lymph node metastasis, stage, and lymphatic and venous invasion in patients with gastric cancer. According to most studies published on the clinical impact of CTC, the presence of CTC negatively affects the prognosis of patients with gastric cancer. Moreover, the study of CTC based on a meta‐analysis demonstrated their importance as a poor prognostic indicator. In clinical management, pre‐ and post‐therapeutic monitoring of CTC using liquid biopsy may be useful for early detection of subclinical patients or disease recurrence, prediction of tumor progression, and administrative control of adjuvant chemotherapy. Although their functional properties remain unclear, molecular profiling of CTC may contribute to the development of personalized treatment that effectively inhibits tumor progression in patients with advanced gastric cancer. We herein review the clinical significance of CTC as a promising blood marker and therapeutic target in patients with gastric cancer.
Collapse
Affiliation(s)
- Takaaki Arigami
- Department of Digestive Surgery Breast and Thyroid Surgery Field of Oncology Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan.,Molecular Frontier Surgery Course of Advanced Therapeutics Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Yoshikazu Uenosono
- Molecular Frontier Surgery Course of Advanced Therapeutics Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Shigehiro Yanagita
- Department of Digestive Surgery Breast and Thyroid Surgery Field of Oncology Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Keishi Okubo
- Department of Digestive Surgery Breast and Thyroid Surgery Field of Oncology Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Takashi Kijima
- Department of Digestive Surgery Breast and Thyroid Surgery Field of Oncology Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Daisuke Matsushita
- Department of Digestive Surgery Breast and Thyroid Surgery Field of Oncology Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Masahiko Amatatsu
- Department of Digestive Surgery Breast and Thyroid Surgery Field of Oncology Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Hiroshi Kurahara
- Department of Digestive Surgery Breast and Thyroid Surgery Field of Oncology Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Kosei Maemura
- Department of Digestive Surgery Breast and Thyroid Surgery Field of Oncology Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Shoji Natsugoe
- Department of Digestive Surgery Breast and Thyroid Surgery Field of Oncology Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan.,Molecular Frontier Surgery Course of Advanced Therapeutics Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| |
Collapse
|
11
|
Circulating tumour cells predict survival in gastric cancer patients: a meta-analysis. Contemp Oncol (Pozn) 2016; 19:451-7. [PMID: 26843841 PMCID: PMC4731451 DOI: 10.5114/wo.2015.56651] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 05/29/2014] [Indexed: 12/11/2022] Open
Abstract
AIM OF THE STUDY The prognostic value of the detection of circulating tumour cells (CTCs) in gastric cancer has been studied intensely in recent years. However, the application of different technologies led to inconsistent results between the studies. Here, we performed a meta-analysis of published studies to summarise the evidence. MATERIAL AND METHODS Medline and ISI Web of Knowledge were searched up to March 2013 using "circulating tumor cells" and "gastric cancer" as search terms. Hazard ratio (HR) with 95% confidence intervals (CIs) for prognostic outcomes and clinical characteristics were extracted from each study. Pooled hazard ratios (HR) and odds ratios (OR) were calculated using random or fixed-effects models. RESULTS Twelve studies enrolling 774 patients were included. The combined HR estimate for overall survival (OS), disease-free survival (DFS), and progression-free survival (PFS) were 1.41 (95% CI: 1.28-1.62), 2.99 (95% CI: 2.01-4.45) and 1.64 (95% CI: 1.02-2.62), respectively. Subgroup analysis concerning detection methods and sampling time showed that results of RT-PCR for the OS group and RT-PCR for the DFS group suggest a prognostic significance of CTC detection (pooled HR [95% CI]: 1.45 [1.28-1.65], I(2) = 38%, p = 0.13; 2.99 [2.01-4.45], I(2) = 0%, p = 0.32). In addition, results of the baseline CTC detection group also indicated a significant prognostic value to predict OS and DFS (pooled HR [95% CI]: 1.47 [1.19-1.82], I(2) = 38%, p = 0.14; 2.99 [2.01-4.45], I(2) = 0%, p = 0.32). We simultaneously found that the detection of CTCs correlated with pathological stage (pooled OR [95% CI]: 2.95 [1.65-5.28], I(2) = 56%, p = 0.03), lymph node status (pooled OR [95% CI]: 2.26 [1.50-3.41], I(2) = 37%, p = 0.09), the depth of invasion (pooled OR [95% CI]: 3.21 [1.38-7.43], I(2) = 72%, p = 0.002), and distant metastasis (pooled OR [95% CI]: 2.68 [1.25-5.73], I(2) = 43%, p = 0.15). CONCLUSIONS Detection of CTCs is associated with poorer prognosis in gastric cancer patients.
Collapse
|
12
|
Court CM, Ankeny JS, Sho S, Tomlinson JS. Circulating Tumor Cells in Gastrointestinal Cancer: Current Practices and Future Directions. Cancer Treat Res 2016; 168:345-376. [PMID: 29206383 DOI: 10.1007/978-3-319-34244-3_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
GI cancers are the leading cause of cancer-related death worldwide primarily due to a combination of late presentation and aggressive biology. The lack of adequate biomarkers for screening, diagnosis, staging, and prognosis confounds clinical decision-making and delays potentially effective therapies. Circulating tumor cells (CTCs) are a new biomarker with particular promise in GI cancers, potentially offering clinicians and researchers real-time access to tumor tissue in a reliable, safe, and cost-effective manner. Preliminary studies have investigated the potential clinical utility of CTCs for all GI cancer types with promising results. Furthermore, advances in single cell analytics have been successfully applied to CTCs, allowing for exciting new clinical and research applications. In this chapter, we will review the current state of CTC research in GI cancers as well as the potential future applications that are currently being developed.
Collapse
|
13
|
Han Y, Ye J, Dong Y, Xu Z, DU Q. Expression and significance of annexin A2 in patients with gastric adenocarcinoma and the association with E-cadherin. Exp Ther Med 2015; 10:549-554. [PMID: 26622352 DOI: 10.3892/etm.2015.2565] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 05/15/2015] [Indexed: 12/12/2022] Open
Abstract
Annexin A2 is a calcium-dependent phospholipid-binding protein, involved in invasion, angiogenesis and migration in cancer cells. The aims of the present study were to evaluate the expression levels of annexin A2 and E-cadherin in gastric adenocarcinoma (GAC), and to investigate the association between the expression of annexin A2 and that of E-cadherin and Ki67, in addition to various clinicopathological factors. This study included 126 patients that were histopathologically diagnosed with GAC. Tissue samples were acquired by surgical resection, and annexin A2 mRNA expression levels were determined using reverse transcription-quantitative polymerase chain reaction. Annexin A2, E-cadherin and Ki67 protein expression levels were detected using western blot analysis and/or immunohistochemical staining. The expression of annexin A2 mRNA and protein was significantly upregulated in the GAC tissues. Annexin A2 expression was detected in 52/126 cases (41.3%) of gastric cancer (GC), and correlations were identified between annexin A2 expression and Tumor, Node, Metastasis (TNM) stage (P=0.002), lymph node metastasis (P=0.016) and distal metastasis (P=0.005). The positive expression rates of E-cadherin and Ki67 in the tumor tissue of patients with GAC were 27.8% (35/126) and 56.2% (71/126), respectively. A negative correlation was observed between the expression of annexin A2 and E-cadherin (P<0.001). No significant association was detected between the expression levels of annexin A2 and Ki67 (P=0.801). In conclusion, upregulated annexin A2 expression was associated with lymph node metastasis, distal metastasis, advanced TNM stage and E-cadherin expression in patients with GAC. The association between the expression of annexin A2 and that of E-cadherin may indicate an underlying mechanism by which annexin A2 contributes to the metastasis in GC, and thus annexin A2 may represent a potential target for the treatment of GAC.
Collapse
Affiliation(s)
- Yuehua Han
- Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jun Ye
- Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Ying Dong
- Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhipeng Xu
- Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Qin DU
- Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
14
|
Wang S, Zheng G, Cheng B, Chen F, Wang Z, Chen Y, Wang Y, Xiong B. Circulating tumor cells (CTCs) detected by RT-PCR and its prognostic role in gastric cancer: a meta-analysis of published literature. PLoS One 2014; 9:e99259. [PMID: 24901848 PMCID: PMC4047117 DOI: 10.1371/journal.pone.0099259] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 05/12/2014] [Indexed: 12/27/2022] Open
Abstract
Objective The prognostic significance of circulating tumor cells (CTCs) is controversial in gastric cancer (GC). We performed a meta-analysis of available studies to assess its prognostic value detected by RT-PCR for patients diagnosed with GC. Methods EMBase, PubMed, Ovid, Web of Science, Cochrane library and Google Scholar database search was conducted on all studies reporting the outcomes of interest. The studies were set up according to the inclusion/exclusion criteria. Meta-analysis was performed by using a random-effects model; hazard ratio (HR), risk ratio (RR) and their 95% confidence intervals (95% CIs) were set as effect measures. The information about trial design, results from the data was independently extracted. Heterogeneity of the studies was tested for each pooled analysis. Results Nineteen studies published matched the selection criteria and were included in this meta-analysis. CTCs positivity was significantly associated with poor relapse free survival (RFS) (HR 2.42, 95% CI: [1.94–3.02]; P<0.001) and poor overall survival (OS) (HR 2.42, 95% CI: [1.94–3.02]; P<0.001). CTCs positivity were also significantly associated with regional lymph nodes (RLNs) metastasis (RR 1.42, 95% CI: [1.20–1.68]; p<0.0001), depth of infiltration (RR 1.51, 95% CI: [1.27–1.79]; p<0.0001), vascular invasion (RR = 1.43, 95% CI: [1.18–1.74], p = 0.0002) and TNM stage(I,II versus III) (RR 0.63, 95% CI [0.48–0.84]; p = 0.001). Conclusion Preoperative CTCs positivity indicates poor prognosis in patients with gastric cancer, and associated with poor clinicopathological prognostic factors.
Collapse
Affiliation(s)
- Shuyi Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, P. R. China
| | - Gang Zheng
- Department of General Surgery, The 5 Hospital of Wuhan, Wuhan, Hubei, P. R. China
| | - Boran Cheng
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, P. R. China
| | - Fangfang Chen
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, P. R. China
| | - Zhenmeng Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, P. R. China
| | - Yuanyuan Chen
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, P. R. China
| | - You Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, P. R. China
| | - Bin Xiong
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, P. R. China
- * E-mail:
| |
Collapse
|
15
|
Tsujiura M, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Otsuji E. Liquid biopsy of gastric cancer patients: Circulating tumor cells and cell-free nucleic acids. World J Gastroenterol 2014; 20:3265-3286. [PMID: 24696609 PMCID: PMC3964398 DOI: 10.3748/wjg.v20.i12.3265] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/27/2013] [Accepted: 02/20/2014] [Indexed: 02/06/2023] Open
Abstract
To improve the clinical outcomes of cancer patients, early detection and accurate monitoring of diseases are necessary. Numerous genetic and epigenetic alterations contribute to oncogenesis and cancer progression, and analyses of these changes have been increasingly utilized for diagnostic, prognostic and therapeutic purposes in malignant diseases including gastric cancer (GC). Surgical and/or biopsy specimens are generally used to understand the tumor-associated alterations; however, those approaches cannot always be performed because of their invasive characteristics and may fail to reflect current tumor dynamics and drug sensitivities, which may change during the therapeutic process. Therefore, the importance of developing a non-invasive biomarker with the ability to monitor real-time tumor dynamics should be emphasized. This concept, so called “liquid biopsy”, would provide an ideal therapeutic strategy for an individual cancer patient and would facilitate the development of “tailor-made” cancer management programs. In the blood of cancer patients, the presence and potent utilities of circulating tumor cells (CTCs) and cell-free nucleic acids (cfNAs) such as DNA, mRNA and microRNA have been recognized, and their clinical relevance is attracting considerable attention. In this review, we discuss recent developments in this research field as well as the relevance and future perspectives of CTCs and cfNAs in cancer patients, especially focusing on GC.
Collapse
|
16
|
Blom A, Bhatia S, Pietromonaco S, Koehler K, Iyer JG, Nagase K, Paulson K, Sabath DE, Nghiem P. Clinical utility of a circulating tumor cell assay in Merkel cell carcinoma. J Am Acad Dermatol 2013; 70:449-55. [PMID: 24388423 DOI: 10.1016/j.jaad.2013.10.051] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 10/07/2013] [Accepted: 10/28/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND Quantitation of circulating tumor cells (CTCs) has utility in managing breast, colon, and prostate carcinomas. OBJECTIVE We sought to determine whether a commercially available CTC assay provides prognostic information in Merkel cell carcinoma (MCC), insight into treatment responses, or both. METHODS We analyzed CTCs in 52 specimens from 34 patients with MCC. RESULTS The presence of CTCs correlated with extent of disease at blood draw (P = .004). Among 15 patients with regional nodal disease, CTC-negative patients had 80% disease-specific survival at 2 years after the test, versus 29% for CTC-positive patients (P = .015). Among the entire cohort, those without CTCs had 72% MCC-specific survival whereas CTC-positive patients had 25% survival (n = 34, median follow-up 19 months, P = .0003). Fifty seven percent of patients with MCC had a cytokeratin "dot" visible in 20% or more of CTCs, a feature that was absent among CTCs from other carcinomas (0 of 13 cases). LIMITATIONS CTC assay was performed at variable times after diagnosis and heterogeneity in extent of disease affects interpretability of the data. CONCLUSION CTC detection in MCC is feasible and appears to add prognostic information, particularly in patients with regional nodal disease. It may also assist clinical management in certain situations, including differentiating metastatic MCC cells from those of other carcinomas.
Collapse
Affiliation(s)
- Astrid Blom
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Shailender Bhatia
- Department of Medicine/Oncology, University of Washington, Seattle, Washington
| | | | - Karen Koehler
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Jayasri G Iyer
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Kotaro Nagase
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Kelly Paulson
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Daniel E Sabath
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Paul Nghiem
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington.
| |
Collapse
|
17
|
Matsumura N, Zembutsu H, Yamaguchi K, Sasaki K, Tsuruma T, Nishidate T, Denno R, Hirata K. Identification of novel molecular markers for detection of gastric cancer cells in the peripheral blood circulation using genome-wide microarray analysis. Exp Ther Med 2011; 2:705-713. [PMID: 22977563 DOI: 10.3892/etm.2011.252] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 04/05/2011] [Indexed: 12/20/2022] Open
Abstract
Although metastasis or relapse is a leading cause of death for patients with gastric cancer, the hematogenous spread of cancer cells remains undetected at the time of initial therapy. The development of novel diagnostic molecular marker(s) to detect circulating gastric cancer cells is an issue of great clinical importance. We obtained peripheral blood samples from 10 patients with gastric cancer who underwent laparotomy and 4 healthy volunteers. Microarray analysis consisting of 30,000 genes or ESTs was carried out using eight gastric cancer tissues and normal gastric mucosae. We selected 53 genes up-regulated in gastric cancer compared to normal gastric mucosae from our microarray data set, and, among these, identified five candidate marker genes (TSPAN8, EPCAM, MMP12, MMP7 and REG3A) which were not expressed in peripheral blood mononuclear cells (PBMCs) from 4 healthy volunteers. We further carried out semi-quantitative nested reverse transcription-polymerase chain reaction (RT-PCR) for HRH1, EGFR, CK20 and CEA in addition to the five newly identified genes using PBMCs of patients with gastric cancer, and found that expression of one or more genes out of the nine was detected in 80% of the patients with gastric cancer. Moreover, the numbers of genes expressed in PBMCs were ≤2 and ≥2 in all vascular invasion-negative cases and in 5 of 6 positive cases, respectively, showing significant differences between the two groups (P=0.041). Nested RT-PCR analysis for the set of nine marker genes using PBMCs may provide the potential for detection of circulating gastric cancer cells prior to metastasis formation in other organs.
Collapse
Affiliation(s)
- Nobuyuki Matsumura
- First Department of Surgery, Sapporo Medical University, School of Medicine, Hokkaido 060-0061, Japan
| | | | | | | | | | | | | | | |
Collapse
|