1
|
Zhao J, Tian W, Zhang X, Dong S, Shen Y, Gao X, Yang M, Lv J, Hu F, Han J, Zhan Q, An F. The diagnostic value of serum trefoil factor 3 and pepsinogen combination in chronic atrophic gastritis: a retrospective study based on a gastric cancer screening cohort in the community population. Biomarkers 2024; 29:384-392. [PMID: 39234749 DOI: 10.1080/1354750x.2024.2400927] [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: 05/12/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is an important precursor of gastric cancer(GC), and there is currently a lack of reliable non-invasive diagnostic markers. This study aims to find a biomarker for non-invasive screening of CAG in the community. METHODS A total of 540 individuals were enrolled (test set = 385, validation set = 155). ROC curve analysis was used to evaluate the diagnostic significance of serum Trefoil Factor 3 (TFF3) alone or in combination with pepsinogen (PG) for CAG in the test and validation set. Furthermore, the diagnostic value of TFF3 and PG in different Helicobacter pylori (H. pylori) infection states was studied. RESULTS When compared with chronic superficial gastritis (CSG), the expression level of serum TFF3 in the CAG was higher (27 ng/ml vs 19.61, P < 0.001). ROC curve analysis found that the sensitivity, specificity, and area under the curve (AUC) of CAG diagnosis using serum TFF3 alone at the optimal cut-off value of 26.55 ng/ml were 0.529, 0.87, and 0.739, respectively. When TFF3 was combined with The Ratio of PGI to PGII (PGR), the AUC and specificity reached 0.755 and 0.825, respectively. TFF3 individual or combined with PGR had good predictive value, especially in the H. Pylori negative patients. CONCLUSION TFF3 combined with PGR can effectively predict CAG, especially in patients with H. pylori negative.
Collapse
Affiliation(s)
- Jiamin Zhao
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Wenying Tian
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Xiaoxue Zhang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Shengrong Dong
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Yao Shen
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Xiaojuan Gao
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Mei Yang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Jiale Lv
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Feifan Hu
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Jinglue Han
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Qiang Zhan
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Fangmei An
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| |
Collapse
|
2
|
Liu SS, Wan QS, Lv C, Wang JK, Jiang S, Cai D, Liu MS, Wang T, Zhang KH. Integrating trans-omics, cellular experiments and clinical validation to identify ILF2 as a diagnostic serum biomarker and therapeutic target in gastric cancer. BMC Cancer 2024; 24:465. [PMID: 38622522 PMCID: PMC11017608 DOI: 10.1186/s12885-024-12175-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/24/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) lacks serum biomarkers with clinical diagnostic value. Multi-omics analysis is an important approach to discovering cancer biomarkers. This study aimed to identify and validate serum biomarkers for GC diagnosis by cross-analysis of proteomics and transcriptomics datasets. METHODS A cross-omics analysis was performed to identify overlapping differentially expressed genes (DEGs) between our previous aptamer-based GC serum proteomics dataset and the GC tissue RNA-Seq dataset in The Cancer Genome Atlas (TCGA) database, followed by lasso regression and random forest analysis to select key overlapping DEGs as candidate biomarkers for GC. The mRNA levels and diagnostic performance of these candidate biomarkers were analyzed in the original and independent GC datasets to select valuable candidate biomarkers. The valuable candidate biomarkers were subjected to bioinformatics analysis to select those closely associated with the biological behaviors of GC as potential biomarkers. The clinical diagnostic value of the potential biomarkers was validated using serum samples, and their expression levels and functions in GC cells were validated using in vitro cell experiments. RESULTS Four candidate biomarkers (ILF2, PGM2L1, CHD7, and JCHAIN) were selected. Their mRNA levels differed significantly between tumor and normal tissues and showed different diagnostic performances for GC, with areas under the receiver operating characteristic curve (AUROCs) of 0.629-0.950 in the TCGA dataset and 0.736-0.840 in the Gene Expression Omnibus (GEO) dataset. In the bioinformatics analysis, only ILF2 (interleukin enhancer-binding factor 2) gene levels were associated with immune cell infiltration, some checkpoint gene expression, chemotherapy sensitivity, and immunotherapy response. Serum levels of ILF2 were higher in GC patients than in controls, with an AUROC of 0.944 for the diagnosis of GC, and it was also detected in the supernatants of GC cells. Knockdown of ILF2 by siRNA significantly reduced the proliferation and colony formation of GC cells. Overexpression of ILF2 significantly promotes the proliferation and colony formation of gastric cancer cells. CONCLUSIONS Trans-omics analysis of proteomics and transcriptomics is an efficient approach for discovering serum biomarkers, and ILF2 is a potential diagnostic biomarker and therapeutic target of gastric cancer.
Collapse
Affiliation(s)
- Shao-Song Liu
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Qin-Si Wan
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Cong Lv
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Jin-Ke Wang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Song Jiang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Dan Cai
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Mao-Sheng Liu
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Ting Wang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Kun-He Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China.
| |
Collapse
|
3
|
Minegishi K, Dobashi Y, Koyama T, Ishibashi Y, Furuya M, Tsubochi H, Ohmoto Y, Yasuda T, Nomura S. Diagnostic utility of trefoil factor families for the early detection of lung cancer and their correlation with tissue expression. Oncol Lett 2023; 25:139. [PMID: 36909373 PMCID: PMC9996639 DOI: 10.3892/ol.2023.13725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/22/2022] [Indexed: 02/23/2023] Open
Abstract
Trefoil factors (TFFs) are upregulated in numerous types of cancer, including those of the breast, the colon, the lung and the pancreas, suggesting their potential utility as biomarkers for screening. In the present study, the clinical relevance of serum or urinary TFFs as biomarkers were comprehensively evaluated and the correlation with TFF expression levels in lung cancer tissue was examined. Serum and urine were collected from 199 patients with lung cancer and 198 healthy individuals. Concentrations of serum and urinary TFF1, TFF2 and TFF3 were measured using ELISA and the potential of TFF levels to discriminate between cancer and non-cancer samples was evaluated. In 100 of the cancer cases, expression of TFF1-3 was analyzed using immunohistochemical staining of paraffin sections. Furthermore, the relationship between TFF levels and clinicopathological factors among these cancer cases was analyzed using immunohistochemistry of tissue specimens, quantified and statistically analyzed. While serum levels of all TFFs measured using ELISA were significantly higher in patients with lung cancer compared with those in healthy individuals, urinary TFFs were lower. Areas under the curve (AUC) of the receiver operating characteristic curves for serum/urinary TFF1, TFF2 and TFF3 were 0.709/0.594, 0.722/0.501 and 0.663/0.665, respectively. Furthermore, the combination of serum TFF1, TFF2, TFF3 and urinary TFF1 and TFF3 demonstrated the highest AUC (0.826). In the clinicopathological analysis, serum TFF1 was higher in the early pathological T-stage (pTis/1/2) compared with the later stage (pT3/4) and TFF2 was higher in the pN0/1 than the pN2 group. With regards to the histological types, urinary TFF1 was higher in squamous cell carcinoma than adenocarcinoma (AC), but TFF2 tended to be higher in AC. Using immunohistochemical analysis, although TFF1 and TFF3 expression showed positive correlation with serum concentrations, TFF2 was inversely correlated. In conclusion, serum and urinary TFF levels are promising predictive biomarkers, and their measurements provide a useful in vivo and non-invasive diagnostic screening tool. In particular, TFF1 and TFF3 could be surrogate markers of clinicopathological profiles of human lung cancer.
Collapse
Affiliation(s)
- Kentaro Minegishi
- Department of Thoracic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Saitama 330-8500, Japan
| | - Yoh Dobashi
- Department of Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Saitama 330-8500, Japan.,Department of Pathology, School of Medicine, International University of Health and Welfare Hospital, Nasushiobara, Tochigi 329-2763, Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto 602-8566, Japan
| | - Yuko Ishibashi
- Department of Surgery, Breast Surgery, Tokyo Women's Medical University, Adachi Medical Center, Adachi, Tokyo 123-8558, Japan
| | - Miki Furuya
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroyoshi Tsubochi
- Department of Thoracic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Saitama 330-8500, Japan
| | - Yasukazu Ohmoto
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8505, Japan
| | - Tomohiko Yasuda
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Department of Gastrointestinal Surgery, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Chiba 270-1694, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
4
|
Haque E, Esmail A, Muhsen I, Salah H, Abdelrahim M. Recent Trends and Advancements in the Diagnosis and Management of Gastric Cancer. Cancers (Basel) 2022; 14:5615. [PMID: 36428707 PMCID: PMC9688354 DOI: 10.3390/cancers14225615] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Gastric cancer is an enigmatic malignancy that has recently been shown to be increasing in incidence globally. There has been recent progress in emerging technologies for the diagnosis and treatment of the disease. Improvements in non-invasive diagnostic techniques with serological tests and biomarkers have led to decreased use of invasive procedures such as endoscopy. A multidisciplinary approach is used to treat gastric cancer, with recent significant advancements in systemic therapies used in combination with cytotoxic chemotherapies. New therapeutic targets have been identified and clinical trials are taking place to assess their efficacy and safety. In this review, we provide an overview of the current and emerging treatment strategies and diagnostic techniques for gastric cancer.
Collapse
Affiliation(s)
- Emaan Haque
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Abdullah Esmail
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
| | - Ibrahim Muhsen
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haneen Salah
- Department of Pathology, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Maen Abdelrahim
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
- Cockrell Center for Advanced Therapeutic Phase I Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| |
Collapse
|
5
|
Latorre G, Pizarro M, Ford J, Gándara V, Muñoz G, Araya J, Bellolio E, Villaseca MÁ, Fuentes-López E, Cortés P, Rollán A, Bufadel M, Araya R, Vargas J, Espino A, Sharp A, Agüero C, Donoso A, Bresky G, Pedrero P, Rueda C, Calvo A, Odagaki T, Moriyama T, Ishida T, Parra-Blanco A, Camargo M, González R, Corvalán A, Riquelme A. Evaluation of Trefoil Factor 3 as a Non-Invasive Biomarker of Gastric Intestinal Metaplasia and Gastric Cancer in a High-Risk Population. GASTROENTEROLOGIA Y HEPATOLOGIA 2022; 46:411-418. [DOI: 10.1016/j.gastrohep.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/07/2022] [Accepted: 04/01/2022] [Indexed: 01/06/2023]
|
6
|
Yang H, Yang WJ, Hu B. Gastric epithelial histology and precancerous conditions. World J Gastrointest Oncol 2022; 14:396-412. [PMID: 35317321 PMCID: PMC8919001 DOI: 10.4251/wjgo.v14.i2.396] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
The most common histological type of gastric cancer (GC) is gastric adenocarcinoma arising from the gastric epithelium. Less common variants include mesenchymal, lymphoproliferative and neuroendocrine neoplasms. The Lauren scheme classifies GC into intestinal type, diffuse type and mixed type. The WHO classification includes papillary, tubular, mucinous, poorly cohesive and mixed GC. Chronic atrophic gastritis (CAG) and intestinal metaplasia are recommended as common precancerous conditions. No definite precancerous condition of diffuse/poorly/undifferentiated type is recommended. Chronic superficial inflammation and hyperplasia of foveolar cells may be the focus. Presently, the management of early GC and precancerous conditions mainly relies on endoscopy including diagnosis, treatment and surveillance. Management of precancerous conditions promotes the early detection and treatment of early GC, and even prevent the occurrence of GC. In the review, precancerous conditions including CAG, metaplasia, foveolar hyperplasia and gastric hyperplastic polyps derived from the gastric epithelium have been concluded, based on the overview of gastric epithelial histological organization and its renewal.
Collapse
Affiliation(s)
- Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wen-Juan Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
7
|
Ji R, Zhang Z, Zhang J, Wu Z, Li M, Ye Y, Li X, An F, Xu C, Lu L, Fan P, Chen Z, Guo Q, Wang Y, Wang X, Li Y, Zhang J, Guan Q, Hu X, Yang T, Liu J, Qiao L, Zheng Y, Zhou Y. Cohort Profile: A population-based cohort for the study of gastric cancer in northwest area of China (Wuwei Cohort). Int J Epidemiol 2021; 50:1433-1442. [PMID: 34143880 DOI: 10.1093/ije/dyab083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 12/29/2022] Open
Affiliation(s)
- Rui Ji
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhiyi Zhang
- Gansu Wuwei Tumor Hospital, Wuwei, Gansu, China
| | - Jinhua Zhang
- Gansu Second Provincial People's Hospital, Lanzhou, Gansu, China
| | - Zhengqi Wu
- Gansu Wuwei Tumor Hospital, Wuwei, Gansu, China
| | - Min Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Yuwei Ye
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaohua Li
- Wuwei Liangzhou Hospital, Wuwei, Gansu, China
| | - Feng An
- Wuwei People's Hospital, Wuwei, Gansu, China
| | | | - Linzhi Lu
- Gansu Wuwei Tumor Hospital, Wuwei, Gansu, China
| | - Ping Fan
- Gansu Wuwei Tumor Hospital, Wuwei, Gansu, China
| | - Zhaofeng Chen
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Qinghong Guo
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiang Wang
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Youpeng Li
- Minqin People's Hospital, Minqin, Gansu, China
| | - Jun Zhang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Quanlin Guan
- Surgical Oncology Department, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaobin Hu
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Tianlin Yang
- Wuwei City Center for Disease Control and Prevention, Wuwei, Gansu, China
| | - Jiankang Liu
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, Australia
| | - Ya Zheng
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
8
|
Calanzani N, Druce PE, Snudden C, Milley KM, Boscott R, Behiyat D, Saji S, Martinez-Gutierrez J, Oberoi J, Funston G, Messenger M, Emery J, Walter FM. Identifying Novel Biomarkers Ready for Evaluation in Low-Prevalence Populations for the Early Detection of Upper Gastrointestinal Cancers: A Systematic Review. Adv Ther 2021; 38:793-834. [PMID: 33306189 PMCID: PMC7889689 DOI: 10.1007/s12325-020-01571-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Introduction Detecting upper gastrointestinal (GI) cancers in primary care is challenging, as cancer symptoms are common, often non-specific, and most patients presenting with these symptoms will not have cancer. Substantial investment has been made to develop biomarkers for cancer detection, but few have reached routine clinical practice. We aimed to identify novel biomarkers for upper GI cancers which have been sufficiently validated to be ready for evaluation in low-prevalence populations. Methods We systematically searched MEDLINE, Embase, Emcare, and Web of Science for studies published in English from January 2000 to October 2019 (PROSPERO registration CRD42020165005). Reference lists of included studies were assessed. Studies had to report on second measures of diagnostic performance (beyond discovery phase) for biomarkers (single or in panels) used to detect pancreatic, oesophageal, gastric, and biliary tract cancers. We included all designs and excluded studies with less than 50 cases/controls. Data were extracted on types of biomarkers, populations and outcomes. Heterogeneity prevented pooling of outcomes. Results We identified 149 eligible studies, involving 22,264 cancer cases and 49,474 controls. A total of 431 biomarkers were identified (183 microRNAs and other RNAs, 79 autoantibodies and other immunological markers, 119 other proteins, 36 metabolic markers, 6 circulating tumour DNA and 8 other). Over half (n = 231) were reported in pancreatic cancer studies. Only 35 biomarkers had been investigated in at least two studies, with reported outcomes for that individual marker for the same tumour type. Apolipoproteins (apoAII-AT and apoAII-ATQ), and pepsinogens (PGI and PGII) were the most promising biomarkers for pancreatic and gastric cancer, respectively. Conclusion Most novel biomarkers for the early detection of upper GI cancers are still at an early stage of matureness. Further evidence is needed on biomarker performance in low-prevalence populations, in addition to implementation and health economic studies, before extensive adoption into clinical practice can be recommended. Electronic Supplementary Material The online version of this article (10.1007/s12325-020-01571-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Natalia Calanzani
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Paige E Druce
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Victoria, Australia
| | - Claudia Snudden
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Kristi M Milley
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Victoria, Australia
| | - Rachel Boscott
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Dawnya Behiyat
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Smiji Saji
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Javiera Martinez-Gutierrez
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Victoria, Australia
- Department of Family Medicine, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Jasmeen Oberoi
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Victoria, Australia
| | - Garth Funston
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Mike Messenger
- Leeds Centre for Personalised Medicine and Health, University of Leeds, Leeds, UK
| | - Jon Emery
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Victoria, Australia
| | - Fiona M Walter
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Zhang CX, Wu CT, Xiao L, Tang SH. The diagnostic and clinicopathological value of trefoil factor 3 in patients with gastric cancer: a systematic review and meta-analysis. Biomarkers 2021; 26:95-102. [PMID: 33401971 DOI: 10.1080/1354750x.2020.1871411] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To assess the diagnostic value of Trefoil factor 3 (TFF3) and the correlation between TFF3 expression and clinicopathological features in patients with gastric cancer (GC). METHODS PubMed, The Cochrane, EMbase, and Web of Science were retrieved comprehensively to collect relevant literature. The search ended on 31 May 2020. All data were analyzed using PubMed, The Cochrane, EMbase, and Web of Science were retrieved to collect relevant articles. All data from the included studies were subjected to meta-analysis using Stata 12.0 software. RESULTS Seventeen studies involved 4654 subjects were included. For the diagnostic value of TFF3 for GC, the sensitivity, specificity, and AUC were 0.71, 0.80, and 0.80, respectively. For the clinicopathological value of TFF3, tissue TFF3 expression showed a higher risk of lymph node metastasis (OR 2.20, 95% CI 1.75-2.78, p < 0.001) and muscularis propria invasion (≥T2) (1.51, 1.13-2.03, p = 0.006), as well as worse TNM stage (2.26, 1.63-3.12, p < 0.001) and histological type (1.72, 1.34-2.20, p < 0.001), while no apparent relationship was found for serous membrane invasion (T4), venous invasion, and peritoneal metastasis. CONCLUSION TFF3 may be a promising biomarker for GC, and the TFF3 expression is likely to be involved in the invasion, metastasis, and differentiation of GC.
Collapse
Affiliation(s)
- Chen-Xing Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, P. R. China.,The First Clinical Medical College, Jinan University, Guangzhou, P. R. China
| | - Chu-Tian Wu
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, P. R. China.,The First Clinical Medical College, Jinan University, Guangzhou, P. R. China
| | - Lin Xiao
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, P. R. China.,The First Clinical Medical College, Jinan University, Guangzhou, P. R. China
| | - Shao-Hui Tang
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, P. R. China.,The First Clinical Medical College, Jinan University, Guangzhou, P. R. China
| |
Collapse
|
10
|
Matsuzaki J, Tsugawa H, Suzuki H. Precision Medicine Approaches to Prevent Gastric Cancer. Gut Liver 2021; 15:3-12. [PMID: 31893631 PMCID: PMC7817924 DOI: 10.5009/gnl19257] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/18/2019] [Accepted: 11/15/2019] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer remains one of the most common causes of cancer-related death worldwide, although the incidence is declining gradually. The primary risk factor for gastric cancer is Helicobacter pylori infection. The Kyoto global consensus report recommends eradication of H. pylori in all infected patients. However, because it is difficult to stratify the risk of carcinogenesis among patients with a history of H. pylori infection, annual endoscopic surveillance is performed for everyone after eradication. This review summarizes the current approaches used to screen for novel molecules that could assist in the diagnosis of gastric cancer and reduce mortality. Most well-studied molecules are tissue protein biomarkers expressed by the gastric epithelium and associated with metaplasia-dysplasia-carcinoma sequences. Other strategies focus on the origin of cancer stem cell-related markers, such as CD44, and immune reaction-related markers, such as matrix metallopeptidases. Noninvasive methods such as blood-based approaches are more attractive. Serum pepsinogen levels predict the severity of gastric mucosal atrophy before H. pylori eradication, whereas plasma ghrelin levels are associated with atrophy even after eradication. Cell-free DNAs and RNAs are attractive tools for the early detection of cancer. These ideas could lead to the development of more personalized strategies for cancer prevention based on cutting-edge technologies.
Collapse
Affiliation(s)
- Juntaro Matsuzaki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hitoshi Tsugawa
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Hidekazu Suzuki
- Department of Gastroenterology and Hepatology, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
11
|
Takako K, Hoang L, Terinte C, Pesci A, Aviel-Ronen S, Alvarado-Cabrero I, Oliva E, Park KJ, Soslow RA, Stolnicu S. Trefoil Factor 2 (TFF2) as a Surrogate Marker for Endocervical Gastric-type Carcinoma. Int J Gynecol Pathol 2021; 40:65-72. [PMID: 32897966 PMCID: PMC7725933 DOI: 10.1097/pgp.0000000000000680] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Gastric-type carcinoma (GAS) is the most common human papilloma virus-independent endocervical adenocarcinoma (ECA), characterized by an aggressive behavior. Trefoil factor 2 (TFF2) is a mucin-associated peptide expressed in normal gastric but not endocervical glands. This study was carried out to investigate whether TFF2 could be a surrogate marker to separate GAS from other types of ECA. ECAs from 9 international institutions were reviewed for consensus histotype. Of them, expression of TFF2 was immunohistochemically examined compared with that of HIK1083, using whole sections of 50 ECAs (10 GASs and 40 non-GASs) and 179 ECAs (24 GASs and 155 non-GASs) with tissue microarrays (TMAs). TMAs were assessed to simulate assessment of immunohistochemical stains in small biopsies. Both markers were similarly scored, and any cytoplasmic/membranous staining of >5% of tumor cells was considered positive. Of 50 ECAs with whole sections, TFF2 was significantly more frequently expressed in GASs (8/10) compared with non-GASs (5/40) (P<0.01). In 179 ECAs with TMAs, TFF2 was also significantly more frequently expressed in GASs (7/24) compared with non-GASs (4/155) (P<0.01). There was no significant difference in specificity among the 2 markers. Double positivity for TFF2 and HIK1083 in ECAs was highly specific in separating GASs from non-GAS (P<0.01). A significantly smaller percentage of GASs were TFF2 positive in TMAs than in whole sections (P<0.01). Our results suggest that TFF2 is a promising marker, along with HIK1083, to confirm a diagnosis of GAS. This marker may be negative in small biopsies, indicating the necessity of using other exclusionary markers in combination with rigorous morphologic review and extensive sampling in resection specimens.
Collapse
Affiliation(s)
- Kiyokawa Takako
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Lien Hoang
- Department of Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| | - Cristina Terinte
- Department of Pathology, Regional Institute of Oncology, Iasi, Romania
| | - Anna Pesci
- Department of Pathology, Ospedale Sacro Cuore Don Calabria, Negrar, Italy
| | - Sarit Aviel-Ronen
- Department of Pathology, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | | | - Esther Oliva
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Kay J. Park
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert A. Soslow
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Simona Stolnicu
- Department of Pathology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Romania
| |
Collapse
|
12
|
Nieminen AA, Kontto J, Puolakkainen P, Virtamo J, Kokkola A. Comparison of operative link for gastritis assessment, operative link on gastric intestinal metaplasia assessment, and TAIM stagings among men with atrophic gastritis. World J Gastroenterol 2020; 26:3447-3457. [PMID: 32655268 PMCID: PMC7327789 DOI: 10.3748/wjg.v26.i24.3447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/29/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer is the world’s third most lethal malignancy. Most gastric cancers develop through precancerous states of atrophic gastritis and intestinal metaplasia. Two staging systems, operative link for gastritis assessment (OLGA) and operative link on gastric intestinal metaplasia assessment (OLGIM), have been developed to detect high gastric cancer risk. European guidelines recommend surveillance for high-risk OLGA/OLGIM patients (stages III–IV), and for those with advanced stage of atrophic gastritis in the whole stomach mucosa. We hypothesize, that by combining atrophy and intestinal metaplasia into one staging named TAIM, more patients with increased gastric cancer risk could be detected.
AIM To evaluate the clinical value of the OLGA, OLGIM, and novel TAIM stagings as prognostic indicators for gastric cancer.
METHODS In the Helsinki Gastritis Study, 22346 elderly male smokers from southwestern Finland were screened for serum pepsinogen I (PGI). Between the years 1989 and 1993, men with low PGI values (PGI < 25 μg/L), were invited to undergo an oesophagogastroduodenoscopy. In this retrospective cohort study, 1147 men that underwent gastroscopy were followed for gastric cancer for a median of 13.7 years, and a maximum of 27.3 years. We developed a new staging system, TAIM, by combining the topography with the severity of atrophy or intestinal metaplasia in gastric biopsies. In TAIM staging, the gastric cancer risk is classified as low or high.
RESULTS Twenty-eight gastric cancers were diagnosed during the follow-up, and the incidence rate was 1.72 per 1000 patient-years. The cancer risk associated positively with TAIM [Hazard ratio (HR) 2.70, 95%CI: 1.09–6.69, P = 0.03]. The risk increased through OLGIM stages 0-IV (0 vs IV: HR 5.72, 95%CI: 1.03–31.77, P for trend = 0.004), but not through OLGA stages 0–IV (0 vs IV: HR 5.77, 95%CI: 0.67–49.77, P for trend = 0.10). The sensitivities of OLGA and OLGIM stages III–IV were low, 21% and 32%, respectively, whereas that of TAIM high-risk was good, 79%. On the contrary, OLGA and OLGIM had high specificity, 85% and 81%, respectively, but TAIM showed low specificity, 42%. In all three staging systems, the high-risk men had three- to four-times higher gastric cancer risk compared to the general male population of the same age.
CONCLUSION OLGIM and TAIM stagings show prognostic value in assessing gastric cancer risk in elderly male smokers with atrophic gastritis.
Collapse
Affiliation(s)
- Anna A Nieminen
- Department of Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki 00290, Finland
| | - Jukka Kontto
- Department of Public Health Solutions, National Institute for Health and Welfare (THL), Helsinki 00271, Finland
| | - Pauli Puolakkainen
- Department of Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki 00290, Finland
| | - Jarmo Virtamo
- Department of Public Health Solutions, National Institute for Health and Welfare (THL), Helsinki 00271, Finland
| | - Arto Kokkola
- Department of Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki 00290, Finland
| |
Collapse
|
13
|
Xu Y, Miremadi A, Link A, Malfertheiner P, Fitzgerald RC, Bornschein J. Feasibility of combined screening for upper gastrointestinal adenocarcinoma risk by serology and Cytosponge testing: the SUGAR study. J Clin Pathol 2019; 72:825-829. [PMID: 31235543 PMCID: PMC6874497 DOI: 10.1136/jclinpath-2019-205700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/18/2019] [Indexed: 01/11/2023]
Abstract
AIMS Aim was to assess the feasibility of serum markers to identify individuals at risk for gastro-oesophageal adenocarcinoma to reduce the number of individuals requiring invasive assessment by endoscopy. METHODS Blood samples from 56 patients with Barrett's oesophagus and 202 non-Barrett controls who previously took part in a trial assessing the accuracy of the Cytosponge for Barrett's oesophagus were assessed for serum pepsinogen (PG) 1 and 2, gastrin-17, trefoil factor 3 (TFF3) and Helicobacter pylori infection. RESULTS PG1 was pathological (<50 ng/mL) in 26 patients (10.1%), none of whom had Barrett's oesophagus (p<0.001). Smoking and drinking had no influence on these results. Pathological PG1 was associated with stomach pain (p=0.029), disruption of sleep (p=0.027) and disruption of diet by symptoms (p=0.019). Serum TFF3 was not associated with any clinical parameter. CONCLUSIONS Assessment of serum PG1 could be combined with a test for Barrett's oesophagus to identify additional patients requiring endoscopy.
Collapse
Affiliation(s)
- Yiwang Xu
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Ahmad Miremadi
- Histopathology Department, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Alexander Link
- Dept. of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Magdeburg, Germany
| | - Peter Malfertheiner
- Dept. of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Magdeburg, Germany
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Jan Bornschein
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford University Hospitals, Oxford, UK
| |
Collapse
|
14
|
Bang CS, Lee JJ, Baik GH. Prediction of Chronic Atrophic Gastritis and Gastric Neoplasms by Serum Pepsinogen Assay: A Systematic Review and Meta-Analysis of Diagnostic Test Accuracy. J Clin Med 2019; 8:jcm8050657. [PMID: 31083485 PMCID: PMC6572271 DOI: 10.3390/jcm8050657] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 12/16/2022] Open
Abstract
Serum pepsinogen assay (sPGA), which reveals serum pepsinogen (PG) I concentration and the PG I/PG II ratio, is a non-invasive test for predicting chronic atrophic gastritis (CAG) and gastric neoplasms. Although various cut-off values have been suggested, PG I ≤70 ng/mL and a PG I/PG II ratio of ≤3 have been proposed. However, previous meta-analyses reported insufficient systematic reviews and only pooled outcomes, which cannot determine the diagnostic validity of sPGA with a cut-off value of PG I ≤70 ng/mL and/or PG I/PG II ratio ≤3. We searched the core databases (MEDLINE, Cochrane Library, and Embase) from their inception to April 2018. Fourteen and 43 studies were identified and analyzed for the diagnostic performance in CAG and gastric neoplasms, respectively. Values for sensitivity, specificity, diagnostic odds ratio, and area under the curve with a cut-off value of PG I ≤70 ng/mL and PG I/PG II ratio ≤3 to diagnose CAG were 0.59, 0.89, 12, and 0.81, respectively and for diagnosis of gastric cancer (GC) these values were 0.59, 0.73, 4, and 0.7, respectively. Methodological quality and ethnicity of enrolled studies were found to be the reason for the heterogeneity in CAG diagnosis. Considering the high specificity, non-invasiveness, and easily interpretable characteristics, sPGA has potential for screening of CAG or GC.
Collapse
Affiliation(s)
- Chang Seok Bang
- Department of Internal Medicine, Hallym University College of Medicine, Sakju-ro 77, Chuncheon, Gangwon-do 24253, Korea.
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24253, Korea.
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24253, Korea.
| | - Jae Jun Lee
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24253, Korea.
- Department of Anesthesiology and Pain Medicine, Hallym University College of Medicine, Chuncheon 24253, Korea.
| | - Gwang Ho Baik
- Department of Internal Medicine, Hallym University College of Medicine, Sakju-ro 77, Chuncheon, Gangwon-do 24253, Korea.
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24253, Korea.
| |
Collapse
|
15
|
Jahan R, Ganguly K, Smith LM, Atri P, Carmicheal J, Sheinin Y, Rachagani S, Natarajan G, Brand RE, Macha MA, Grandgenett PM, Kaur S, Batra SK. Trefoil factor(s) and CA19.9: A promising panel for early detection of pancreatic cancer. EBioMedicine 2019; 42:375-385. [PMID: 30956167 PMCID: PMC6491718 DOI: 10.1016/j.ebiom.2019.03.056] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/21/2022] Open
Abstract
Background Trefoil factors (TFF1, TFF2, and TFF3) are small secretory molecules that recently have gained significant attention in multiple studies as an integral component of pancreatic cancer (PC) subtype-specific gene signature. Here, we comprehensively investigated the diagnostic potential of all the member of trefoil family, i.e., TFF1, TFF2, and TFF3 in combination with CA19.9 for detection of PC. Methods Trefoil factors (TFFs) gene expression was analyzed in publicly available cancer genome datasets, followed by assessment of their expression in genetically engineered spontaneous mouse model (GEM) of PC (KrasG12D; Pdx1-Cre (KC)) and in human tissue microarray consisting of normal pancreas adjacent to tumor (NAT), precursor lesions (PanIN), and various pathological grades of PC by immunohistochemistry (IHC). Serum TFFs and CA19.9 levels were evaluated via ELISA in comprehensive sample set (n = 362) comprised of independent training and validation sets each containing benign controls (BC), chronic pancreatitis (CP), and various stages of PC. Univariate and multivariate logistic regression and receiver operating characteristic curves (ROC) were used to examine their diagnostic potential both alone and in combination with CA19.9. Findings The publicly available datasets and expression analysis revealed significant increased expression of TFF1, TFF2, and TFF3 in human PanINs and PC tissues. Assessment of KC mouse model also suggested upregulated expression of TFFs in PanIN lesions and early stage of PC. In serum analyses studies, TFF1 and TFF2 were significantly elevated in early stages of PC in comparison to benign and CP control group while significant elevation in TFF3 levels were observed in CP group with no further elevation in its level in early stage PC group. In receiver operating curve (ROC) analyses, combination of TFFs with CA19.9 emerged as promising panel for discriminating early stage of PC (EPC) from BC (AUCTFF1+TFF2+TFF3+CA19.9 = 0.93) as well as CP (AUCTFF1+TFF2+TFF3+CA19.9 = 0.93). Notably, at 90% specificity (desired for blood-based biomarker panel), TFFs combination improved CA19.9 sensitivity by 10% and 25% to differentiate EPC from BC and CP respectively. In an independent blinded validation set, the combination of TFFs and CA19.9 (AUCTFF1+TFF2+TFF3+CA19.9 = 0.82) also improved the overall efficacy of CA19.9 (AUCCA19.9 = 0.66) to differentiate EPC from CP proving unique biomarker capabilities of TFFs to distinguish early stage of this deadly lethal disease. Interpretation In silico, tissue and serum analyses validated significantly increased level of all TFFs in precursor lesions and early stages of PC. The combination of TFFs enhanced sensitivity and specificity of CA19.9 to discriminate early stage of PC from benign control and chronic pancreatitis groups.
Collapse
Affiliation(s)
- Rahat Jahan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Lynette M Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Joseph Carmicheal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Yuri Sheinin
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Gopalakrishnan Natarajan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Randall E Brand
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Department of Otolaryngology-Head & Neck Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Paul M Grandgenett
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
16
|
Genome maps across 26 human populations reveal population-specific patterns of structural variation. Nat Commun 2019; 10:1025. [PMID: 30833565 PMCID: PMC6399254 DOI: 10.1038/s41467-019-08992-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/12/2019] [Indexed: 01/10/2023] Open
Abstract
Large structural variants (SVs) in the human genome are difficult to detect and study by conventional sequencing technologies. With long-range genome analysis platforms, such as optical mapping, one can identify large SVs (>2 kb) across the genome in one experiment. Analyzing optical genome maps of 154 individuals from the 26 populations sequenced in the 1000 Genomes Project, we find that phylogenetic population patterns of large SVs are similar to those of single nucleotide variations in 86% of the human genome, while ~2% of the genome has high structural complexity. We are able to characterize SVs in many intractable regions of the genome, including segmental duplications and subtelomeric, pericentromeric, and acrocentric areas. In addition, we discover ~60 Mb of non-redundant genome content missing in the reference genome sequence assembly. Our results highlight the need for a comprehensive set of alternate haplotypes from different populations to represent SV patterns in the genome. Large structural variants (SV) are understudied in human genetics research because of the difficulty to detect them in the routinely generated short-read sequencing data. Here, the authors generate optical genome maps of 154 individuals from 26 populations that allow comprehensive examination of large SVs.
Collapse
|
17
|
Screening Biomarker as an Alternative to Endoscopy for the Detection of Early Gastric Cancer: The Combination of Serum Trefoil Factor Family 3 and Pepsinogen. Gastroenterol Res Pract 2018; 2018:1024074. [PMID: 29977284 PMCID: PMC5994275 DOI: 10.1155/2018/1024074] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 04/03/2018] [Indexed: 12/12/2022] Open
Abstract
Objective The serum pepsinogen test has limitation in its predictive power as a noninvasive biomarker for gastric cancer screening. We aimed to investigate whether the combination of TFF3 and pepsinogen could be an effective biomarker for the detection of gastric cancer even in the early stages. Methods In total, 281 patients with early gastric cancer (EGC), who underwent endoscopic submucosal dissection in Korea, and 708 healthy individuals from Japan were enrolled in the derivation cohort. The validation cohort included 30 Korean patients with EGC and 30 Korean healthy control blood donors. Serum TFF3 levels were examined using enzyme-linked immunosorbent assay. Results Using a cutoff of 6.73 ng/mL in the derivation cohort, the sensitivity of the combination of tests for EGC detection was superior (87.5%) to that of TFF3 (80.4%) or pepsinogen test alone (39.5%). Similarly, in the validation cohort, the sensitivity of TFF3 plus pepsinogen was higher (90.4%) than that of TFF3 (80.0%) or pepsinogen test alone (33.3%). Conclusion The combination of serum TFF3 and pepsinogen is a more effective noninvasive biomarker for gastric cancer detection compared with pepsinogen or TFF3 alone, even in EGC. This trial is registered with NCT03046745.
Collapse
|
18
|
Urine Trefoil Factors as Prognostic Biomarkers in Chronic Kidney Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3024698. [PMID: 29850501 PMCID: PMC5903307 DOI: 10.1155/2018/3024698] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/01/2018] [Indexed: 12/18/2022]
Abstract
Introduction Trefoil factor family (TFF) peptides are increased in serum and urine in patients with chronic kidney disease (CKD). However, whether the levels of TFF predict the progression of CKD remains to be elucidated. Methods We determined the TFF levels using peptide-specific ELISA in spot urine samples and performed a prospective cohort study. The association between the levels of urine TFFs and other urine biomarkers as well as the renal prognosis was analyzed in 216 CKD patients (mean age: 53.7 years, 47.7% female, 56.9% with chronic glomerulonephritis, and mean eGFR: 58.5 ml/min/1.73 m2). Results The urine TFF1 and TFF3 levels significantly increased with the progression of CKD stages, but not the urine TFF2 levels. The TFF1 and TFF3 peptide levels predicted the progression of CKD ≥ stage 3b by ROC analysis (AUC 0.750 and 0.879, resp.); however, TFF3 alone predicted CKD progression in a multivariate logistic regression analysis (odds ratio 3.854, 95% confidence interval 1.316–11.55). The Kaplan-Meier survival curves demonstrated that patients with a higher TFF1 and TFF3 alone, or in combination with macroalbuminuria, had a significantly worse renal prognosis. Conclusion The data suggested that urine TFF peptides are associated with renal progression and the outcomes in patients with CKD.
Collapse
|
19
|
Pandey V, Zhang M, Chong QY, You M, Raquib AR, Pandey AK, Liu DX, Liu L, Ma L, Jha S, Wu ZS, Zhu T, Lobie PE. Hypomethylation associated enhanced transcription of trefoil factor-3 mediates tamoxifen-stimulated oncogenicity of ER+ endometrial carcinoma cells. Oncotarget 2017; 8:77268-77291. [PMID: 29100386 PMCID: PMC5652779 DOI: 10.18632/oncotarget.20461] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/16/2017] [Indexed: 12/20/2022] Open
Abstract
Tamoxifen (TAM) is widely used as an adjuvant therapy for women with breast cancer (BC). However, TAM possesses partial oestrogenic activity in the uterus and its use has been associated with an increased incidence of endometrial carcinoma (EC). The molecular mechanism for these observations is not well understood. Herein, we demonstrated that forced expression of Trefoil factor 3 (TFF3), in oestrogen receptor-positive (ER+) EC cells significantly increased cell cycle progression, cell survival, anchorage-independent growth, invasiveness and tumour growth in xenograft models. Clinically, elevated TFF3 protein expression was observed in EC compared with normal endometrial tissue, and its increased expression in EC was significantly associated with myometrial invasion. TAM exposure increased expression of TFF3 in ER+ EC cells and its elevated expression resulted in increased oncogenicity and invasiveness. TAM-stimulated expression of TFF3 in EC cells was associated with hypomethylation of the TFF3 promoter sequence and c-JUN/SP1-dependent transcriptional activation. In addition, small interfering (si) RNA-mediated depletion or polyclonal antibody inhibition of TFF3 significantly abrogated oncogenicity and invasiveness in EC cells consequent to TAM induction or forced expression of TFF3. Hence, TAM-stimulated upregulation of TFF3 in EC cells was critical in promoting EC progression associated with TAM treatment. Importantly, inhibition of TFF3 function might be an attractive molecular modality to abrogate the stimulatory effects of TAM on endometrial tissue and to limit the progression of EC.
Collapse
Affiliation(s)
- Vijay Pandey
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Min Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, P.R. China
| | - Qing-Yun Chong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Mingliang You
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Amit K Pandey
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Dong-Xu Liu
- School of Science, Auckland University of Technology, Auckland, New Zealand
| | - Liang Liu
- Department of Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, P.R China.,Department of Radiology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, P.R China
| | - Lan Ma
- Tsinghua Berkeley Shenzhen Institute, Division of Life Sciences & Health, Tsinghua University Graduate School, Shenzhen, P.R China
| | - Sudhakar Jha
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Zheng-Sheng Wu
- Department of Pathology, Anhui Medical University, Hefei, P.R China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, P.R. China
| | - Peter E Lobie
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacology, National University of Singapore, Singapore.,Tsinghua Berkeley Shenzhen Institute, Division of Life Sciences & Health, Tsinghua University Graduate School, Shenzhen, P.R China
| |
Collapse
|
20
|
Diao S, Zheng Q, Gao J, Yao Y, Ren S, Liu Y, Xu Y. Trefoil factor 3 contributes to the malignancy of glioma via regulating HIF-1α. Oncotarget 2017; 8:76770-76782. [PMID: 29100347 PMCID: PMC5652741 DOI: 10.18632/oncotarget.20010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/27/2017] [Indexed: 12/22/2022] Open
Abstract
Trefoil factor 3 (TFF3) plays significant roles in several solid tumors. However, the expression pattern and function of TFF3 in glioblastoma (GBM) have not been reported. Here, we report that expression level of TFF3 significantly elevated in glioma and correlated with the prognosis of glioma patients. Then we found TFF3 promotes proliferation, invasion, and migration and inhibits apoptosis of glioma cells in vitro, and delayed tumor progression in subcutaneous xenograft nude mice, and prolonged the median survival time in orthotopic xenograft mice. Moreover, knockdown of TFF3 reduced the expression of HIF-1α through a hypoxia-independent manner. These findings suggest that targeting TFF3 may offer a novel strategy for therapeutic intervention of malignant gliomas.
Collapse
Affiliation(s)
- Shuo Diao
- Department of Neurosurgery, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medical College, China Medical University, Shenyang, People's Republic of China
| | - Jian Gao
- Center of Laboratory Technology and Experimental Medicine, China Medical University, Shenyang, People's Republic of China
| | - Yiqun Yao
- Department of Neurosurgery, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Siyang Ren
- Department of Neurosurgery, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Yongjian Liu
- Department of Interventional Therapy, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Yinghui Xu
- Department of Neurosurgery, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| |
Collapse
|
21
|
Fujimoto A, Ishikawa Y, Ishii T, Yamada A, Igarashi Y, Ohmoto Y, Kaise M. Differences between gastric signet-ring cell carcinoma and poorly differentiated adenocarcinoma: A comparison of histopathologic features determined by mucin core protein and trefoil factor family peptide immunohistochemistry. Pathol Int 2017; 67:398-403. [PMID: 28691258 DOI: 10.1111/pin.12559] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 06/09/2017] [Indexed: 12/22/2022]
Abstract
We investigated differences between the pathological features of gastric signet-ring cell carcinoma (sig) and poorly differentiated adenocarcinoma (por) by examining the expressions of the trefoil factor family peptides (TFFs) and mucin core proteins (MUCs). Ninety-seven tissues of 97 gastric cancer patients were selected for this study. After gastrectomy, the major histopathologic types were determined to be sig, solid-type poorly differentiated adenocarcinoma (por1), non-solid type poorly differentiated adenocarcinoma (por2), and well-differentiated tubular adenocarcinoma (tub1). We evaluated the prevalence of positive staining for MUCs (MUC5AC and MUC2) and TFFs (TFF1 and TFF3) and assessed the correlation between MUCs and TFFs in each histopathological type. The rate of MUC2 expression significantly differed between sig and por2 (50.0% vs 11.7%, P = 0.011). TFF3 expression in sig significantly differed from TFF3 expression in both por2 (100% vs 17.6%, P < 0.0001) and por1 (100% vs 33.3%, P = 0.0004). MUC5AC and TFF1 expressions were significantly correlated in por1 (r = 0.705, P = 0.002), por2 (r = 0.535, P = 0.0009), and tub1 (r = 0.470, P = 0.0034), while MUC2 and TFF3 expressions were significantly correlated only in sig (r = 0.593, P = 0.040). The expression and correlation patterns of the TFFs and MUCs suggest that the histopathologic features of gastric sig differ from those of por.
Collapse
Affiliation(s)
- Ai Fujimoto
- Department of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan.,Division for Research and Development of Minimally Invasive Treatment, Cancer Center, Keio University, Tokyo, Japan
| | | | | | - Akihiro Yamada
- Department of Gastroenterology, Toranomon Hospital, Tokyo, Japan
| | - Yoshinori Igarashi
- Department of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Yasukazu Ohmoto
- Institute of Biomedical Innovation, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Mitsuru Kaise
- Department of Gastroenterology, Toranomon Hospital, Tokyo, Japan
| |
Collapse
|
22
|
Choi B, Lee HJ, Min J, Choe HN, Choi YS, Son YG, Ahn HS, Suh YS, Goldenring JR, Yang HK. Plasma expression of the intestinal metaplasia markers CDH17 and TFF3 in patients with gastric cancer. Cancer Biomark 2017; 19:231-239. [DOI: 10.3233/cbm-160147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Boram Choi
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Hyuk-Joon Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery,
| | - Jimin Min
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hwi-Nyeong Choe
- Department of Nursing, Seoul National University Hospital, Seoul, Korea
| | | | | | | | | | - James R. Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, and the Nashville VA Medical Center, Nashville, TN, USA
| | - Han-Kwang Yang
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery,
| |
Collapse
|
23
|
Xie H, Guo JH, An WM, Tian ST, Yu HP, Yang XL, Wang HM, Guo Z. Diagnostic value evaluation of trefoil factors family 3 for the early detection of colorectal cancer. World J Gastroenterol 2017; 23:2159-2167. [PMID: 28405143 PMCID: PMC5374127 DOI: 10.3748/wjg.v23.i12.2159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/10/2017] [Accepted: 03/17/2017] [Indexed: 02/06/2023] Open
Abstract
AIM The purpose of this study was to evaluate the diagnostic value of trefoil factor family 3 (TFF3) for the early detection of colorectal cancer (CC).
METHODS Serum TFF3 and carcino-embryonic antigen (CEA) were detected in 527 individuals, including 115 healthy control (HC), 198 colorectal adenoma (CA), and 214 CC individuals in the training group.
RESULTS Serum TFF3 showed no significant correlation with age, gender, or tumor location but showed significant correlation with the tumor stage. Serum TFF3 in the CC group was significantly higher than in the HC or CA group. The AUC values of TFF3 for discriminating between HC and CC and between CA and CC were 0.930 (0.903, 0.958) and 0.834 (0.796, 0.873). A multivariate model combining TFF3 and CEA was built. Compared to TFF3 or CEA alone, the multivariate model showed significant improvement (P < 0.001). For discriminating between HC and CC, HC and early stage CC, HC and advanced stage CC, CA and CC, CA and early stage CC, and CA and advanced stage CC in the training group, the sensitivities were 92.99%, 91.46%, 93.18%, 73.83%, 76.83%, and 81.82%, and the specificities were 91.30%, 91.30%, 93.91%, 88.38%, 77.27%, and 88.38%, respectively. After validation, the sensitivities were 89.39%, 85.71%, 90.79%, 72.73%, 71.43%, and 78.95%, and the specificities were 87.85%, 87.85%, 2.52%, 87.85%, 80.77%, and 87.50%, respectively.
CONCLUSION The multivariate diagnostic model that included TFF3 and CEA showed significant improvement over the conventional biomarker CEA and might provide a potential method for the early detection of CC.
Collapse
|
24
|
Kuo HY, Chang WL, Yeh YC, Tsai YC, Wu CT, Cheng HC, Yang HB, Lu CC, Sheu BS. Serum Level of Trefoil Factor 2 can Predict the Extent of Gastric Spasmolytic Polypeptide-Expressing Metaplasia in the H. pylori-Infected Gastric Cancer Relatives. Helicobacter 2017; 22. [PMID: 27220894 DOI: 10.1111/hel.12320] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Gastric cancer has familial clustering in incidence, and the familial relatives of gastric cancer sufferers are prone to have spasmolytic polypeptide-expressing metaplasia (SPEM), and intestinal metaplasia (IM) after H. pylori infection. This study tested whether serum pepsinogen I/II and trefoil factor family (TFF) proteins can predict SPEM or IM in the H. pylori-infected relatives of patients with gastric cancer. METHODS We prospectively enrolled 119 H. pylori-infected relatives of gastric cancer patients of noncardiac gastric cancer patients, who then received panendoscopy to obtain gastric biopsy to define the presence of corpus gastritis index (CGI), SPEM, and IM. The advanced SPEM in histology was defined by TFF2 immunohistochemistry. Each patient also had checkups of serum TFF2, TFF3, and pepsinogen I/II by enzyme-linked immunosorbent assay (ELISA). RESULTS The 119 H. pylori-infected relatives included 61 with SPEM, and 34 with IM. The presence of either IM or SPEM was not related to the serum TFF2, TFF3, and pepsinogen I/II levels (p > .05). Serum TFF2 levels were higher in relatives with CGI who also had advanced SPEM (p = .032). For relatives without CGI, the elevated serum TFF2 levels correlated with higher H. pylori density and more severe gastritis in antrum (p = .001). CONCLUSION The serum TFF2 level cannot predict SPEM or IM in H. pylori-infected relatives of patients with gastric cancer. For H. pylori-infected relatives with CGI, serum TFF2 levels may predict the advanced severity of SPEM. Elevated serum TFF2 levels may indicate severe H. pylori-related inflammation, at risk of development or progression of SPEM in relatives without CGI.
Collapse
Affiliation(s)
- Hsin-Yu Kuo
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Lun Chang
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Chun Yeh
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ching Tsai
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, Tainan Hospital, Tainan, Taiwan.,Department of Health, Executive Yuan, Tainan, Taiwan
| | - Chung-Tai Wu
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiu-Chi Cheng
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Bai Yang
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pathology, Ton-Yen General Hospital, Hsin-chu, Taiwan
| | - Cheng-Chang Lu
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bor-Shyang Sheu
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
25
|
Yusup A, Huji B, Fang C, Wang F, Dadihan T, Wang HJ, Upur H. Expression of trefoil factors and TWIST1 in colorectal cancer and their correlation with metastatic potential and prognosis. World J Gastroenterol 2017; 23:110-120. [PMID: 28104986 PMCID: PMC5221274 DOI: 10.3748/wjg.v23.i1.110] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/19/2016] [Accepted: 12/02/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To detect the expression of trefoil factors (TFFs) and TWIST1 in colorectal cancer (CRC) and analyze their correlation with metastasis and survival.
METHODS This study examined the expression of TFF1, TFF3 and TWIST1 in a total of 75 tumor samples, 47 matched normal samples (15 cm from the lesion margin), 30 metastatic lymph nodes, and 10 liver metastatic cancer samples from patients with CRC. The relationship was then analyzed between the protein expression and different clinical records. TFF1, TFF3, TWIST1,E-cadherin, vimentin and β-catenin mRNA and protein expression levels were measured in colon cancer cell lines with different metastatic potentials (HIEC, HT29, SW620, and LoVo cells), and the correlation of the expression levels with epithelial-mesenchymal transition (EMT) was discussed.
RESULTS It was found that 66.7% (50/75), 78.7% (59/75) and 54.7% (41/75) of tumor tissue samples exhibited positive staining for TFF1, TFF3 and TWIST1 and so did 27.3% (13/47), 100% (47/47) and 17% (8/47) of adjacent normal colorectal tissues. Compared with adjacent normal tissues, significant differences were found in the expression of all three proteins in different cancerous tissues (P < 0.05). Higher expression of TFF3 and TWIST1 was significantly correlated with lymph node metastasis (P = 0.034, P = 0.000), advanced stage (P = 0.031, P = 0.003), and poorer survival (P = 0.042 for the TFF3 group, P = 0.003 for the TWIST1 group). The expression of TFF3 and TWIST1 in cancer cell lines was higher than that in HIEC (a normal human intestinal epithelial cell line)(P < 0.05), and the expression intensity demonstrated a tendency to rise with increased metastatic potential both at the protein and mRNA levels. However, TFF1 expression demonstrated the opposite tendency. It was also observed that the expression of E-cadherin and β-catenin tended to decrease while that of vimentin, TWIST1 and Snail tended to rise with the increase in metastatic potential.
CONCLUSION The expression of TFF3 and TWIST1 might be associated with the survival of patients with CRC after curative resection and might be pivotal predictors of disease progression. TFF3 may be correlated to the invasiveness of CRC.
Collapse
|
26
|
Li Q, Wang K, Su C, Fang J. Serum Trefoil Factor 3 as a Protein Biomarker for the Diagnosis of Colorectal Cancer. Technol Cancer Res Treat 2016; 16:440-445. [PMID: 27760866 DOI: 10.1177/1533034616674323] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Colorectal cancer is one of the most frequent causes of cancer-related deaths worldwide. Thus, there is a need for timely diagnosis and effective treatment. Our aim in the present study was to detect the serum level of trefoil factor 3 protein and evaluate the diagnostic accuracy of trefoil factor 3 in patients with colorectal cancer. We collected serum samples from 204 participants (127 patients with colorectal cancer, 35 patients with polyps, and 42 healthy controls). The levels of serum trefoil factor 3 and carcinoembryonic antigen expression were measured by enzyme-linked immunosorbent assay. Receiver operating characteristic curves were plotted to calculate the diagnostic accuracy of trefoil factor 3 in patients with colorectal cancer. The serum levels of trefoil factor 3 in patients with colorectal cancer (6.66 ± 2.4 ng/mL; P < .00l) and polyps (3.86 ± 1.3 ng/mL; P < .00l) were significantly increased compared to healthy controls (2.09 ± 1.0 ng/mL). Moreover, the area under the receiver operating characteristic curve for trefoil factor 3 was greater than carcinoembryonic antigen (0.889 vs 0.715). At a cutoff value of 5.591 ng/mL, the diagnostic sensitivity, specificity, and likelihood ratio of serum trefoil factor 3 for colorectal cancer was 74.2%, 94.8%, and 14.25, respectively. Furthermore, the serum trefoil factor 3 levels in early colorectal cancer (TNM stage I, 3.67 ± 1.27 ng/mL) were significantly increased compared to healthy controls ( P < .001); however, there was no significant difference compared to patients with polyps ( P = .576). We observed that the serum trefoil factor 3 levels decreased after surgery (6.66 ± 2.4 vs 4.48 ± 1.80 ng/mL; P < .001). In addition, high serum trefoil factor 3 levels were associated with poor tumor differentiation and clinical TNM stage ( P < .05). In conclusion, serum trefoil factor 3 is a promising biomarker for the diagnosis of colorectal cancer and prognosis of patients.
Collapse
Affiliation(s)
- Qiang Li
- 1 Department of Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Keke Wang
- 2 Department of Emergency, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Chang Su
- 3 Department of Internal Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jieyu Fang
- 4 Department of Anesthesia, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
27
|
Comparison of serum trefoil factor 3 with the pepsinogen test for the screening of diffuse-type gastric cancer. Clin Exp Med 2016; 17:403-410. [PMID: 27154568 DOI: 10.1007/s10238-016-0426-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/27/2016] [Indexed: 02/06/2023]
Abstract
Emerging data show that serum trefoil factor 3 (TFF3) alone and combined with the serum pepsinogen (PG) test can increase the diagnostic yield of gastric cancer. We aimed to evaluate the diagnostic value of serum TFF3 for the screening of gastric cancer in Korean patients, especially for the screening of the diffuse type of gastric cancer, and compared TFF3 to the serum PG test. We enrolled 25 healthy controls and 79 subjects with gastric cancer who underwent endoscopic resection or surgery from June 2006 to June 2015. Data about age, sex, histological type according to the Lauren classification, stage of gastric cancer, and status of H. pylori were collected. Serum levels of PG I and PG II were measured by the latex-enhanced turbidimetric immunoassay, and serum TFF3 levels were measured by enzyme-linked immunosorbent assay. The optimal cutoff value of serum TFF3 was ≥8.9 ng/mL to diagnose gastric cancer, with 73.4 % sensitivity and 92.0 % specificity, which were higher than those of the serum PG I/II ratio, with 69.6 % sensitivity and 68.0 % specificity. The optimal sensitivity and specificity of serum TFF3 for the diagnosis of diffuse-type gastric cancer were 68.0 and 92.0 %, respectively, which were lower than those for the diagnosis of intestinal-type gastric cancer (75.6 and 100 %, respectively). Serum TFF3 is a more stable and useful marker than the serum PG test for the screening of gastric cancer in Korean patients. Serum TFF3 showed good diagnostic power in detecting both intestinal- and diffuse-type gastric cancer although it showed decreased power in diffuse type.
Collapse
|
28
|
Abstract
In North-Eastern Asian countries, where incidence and mortality of gastric cancer remain very high, population-based gastric cancer screenings have been conducted under governmental subsidy in Japan and Korea. Reduction of gastric cancer mortality by the screening was documented in Japan, but the Japanese gastric cancer screening with the X-ray photofluorography is criticized for its high cost and a low uptake rate. Although the Korean program seems to achieve a high-rate of uptake with increasing use of endoscopy, the work load is substantial. In the meantime, more attention in the world turns to primary prevention through eradication of Helicobacter pylori. Indeed, fairly large-scale studies to examine the feasibility of mass-eradication to prevent gastric cancer are underway in China and Taiwan. In the future, gastric cancer screening should incorporate 'screen to treat' of H. pylori infection at younger age followed by endoscopic surveillance for subjects at risk.
Collapse
Affiliation(s)
- Kentaro Sugano
- Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan.
| |
Collapse
|
29
|
Relationship between atrophic gastritis, gastric intestinal metaplasia and Helicobacter pylori on endoscopic screening of upper gastrointestinal tract and a brief review of the literature. Eur Surg 2015. [DOI: 10.1007/s10353-015-0378-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
30
|
Abstract
Gastric cancer is associated with high morbidity and mortality worldwide. To reduce the socioeconomic burden related to gastric cancer, it is very important to identify and manage high risk group for gastric cancer. In this review, we describe the general risk factors for gastric cancer and define high risk group for gastric cancer. We discuss strategies for the effective management of patients for the prevention and early detection of gastric cancer. Atrophic gastritis (AG) and intestinal metaplasia (IM) are the most significant risk factors for gastric cancer. Therefore, the accurate selection of individuals with AG and IM may be a key strategy for the prevention and/or early detection of gastric cancer. Although endoscopic evaluation using enhanced technologies such as narrow band imaging-magnification, the serum pepsinogen test, Helicobacter pylori serology, and trefoil factor 3 have been evaluated, a gold standard method to accurately select individuals with AG and IM has not emerged. In terms of managing patients at high risk of gastric cancer, it remains uncertain whether H. pylori eradication reverses and/or prevents the progression of AG and IM. Although endoscopic surveillance in high risk patients is expected to be beneficial, further prospective studies in large populations are needed to determine the optimal surveillance interval.
Collapse
Affiliation(s)
- Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
31
|
Xiao P, Ling H, Lan G, Liu J, Hu H, Yang R. Trefoil factors: Gastrointestinal-specific proteins associated with gastric cancer. Clin Chim Acta 2015; 450:127-34. [PMID: 26265233 DOI: 10.1016/j.cca.2015.08.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 12/11/2022]
Abstract
Trefoil factor family (TFF), composed of TFF1, TFF2, and TFF3, is a cluster of secreted peptides characterized by trefoil domain (s) and C-terminal dimerization domain. TFF1, a gastric tumor suppressor, is a single trefoil peptide originally detected in breast cancer cell lines but expressed mainly in the stomach; TFF2, a candidate of gastric cancer suppressor with two trefoil domains, is abundant in the stomach and duodenal Brunner's glands; and TFF3 is another single trefoil peptide expressed throughout the intestine which can promote the development of gastric carcinoma. According to multiple studies, TFFs play a regulatory function in the mammals' digestive system, namely in mucosal protection and epithelial cell reconstruction, tumor suppression or promotion, signal transduction and the regulation of proliferation and apoptosis. Action mechanisms of TFFs remain unresolved, but the recent demonstration of a GKN (gastrokine) 2-TFF1 heterodimer implicates structural and functional interplay with gastrokines. This review aims to encapsulate the structural and biological characteristics of TFF.
Collapse
Affiliation(s)
- Ping Xiao
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Hui Ling
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China.
| | - Gang Lan
- Key Laboratory for Atherosclerology of Hunan Province, Cardiovascular Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Jiao Liu
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Haobin Hu
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Ruirui Yang
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| |
Collapse
|
32
|
Kaise M. Advanced endoscopic imaging for early gastric cancer. Best Pract Res Clin Gastroenterol 2015; 29:575-87. [PMID: 26381303 DOI: 10.1016/j.bpg.2015.05.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/22/2015] [Accepted: 05/21/2015] [Indexed: 01/31/2023]
Abstract
Considerable numbers of early gastric cancers can be missed or misdiagnosed with conventional white light imaging endoscopy (WLI), thus advanced endoscopic imaging modalities have been applied to overcome the issue. High definition endoscopy can improve diagnostic accuracy, but still misses 20-25% of early gastric cancer. Magnifying endoscopy combined with narrow band imaging (NBI) allows for very high accuracy, with sensitivity and specificity of over 95%. The algorithm for magnifying endoscopy diagnosis of gastric cancer is composed of 1) presence of demarcation line, and 2) presence of irregular microsurface and/or microvascular pattern. Ultra-high magnification of 400 times with endocytoscopy (ECS) can produce images reflecting structural and cellular atypia. Using high grade ECS atypia as the diagnostic criteria for gastric cancer, ECS achieves a high diagnostic accuracy (86% of sensitivity, 100% of specificity) although approximately 10% of target lesions are not assessable because of poor dye staining.
Collapse
Affiliation(s)
- Mitsuru Kaise
- Department of Gastroenterology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo 105-8470, Japan.
| |
Collapse
|
33
|
Xiao L, Liu YP, Xiao CX, Ren JL, Guleng B. Serum TFF3 may be a pharamcodynamic marker of responses to chemotherapy in gastrointestinal cancers. BMC Clin Pathol 2014; 14:26. [PMID: 25031551 PMCID: PMC4099389 DOI: 10.1186/1472-6890-14-26] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 06/10/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND As a secreted protein, serum trefoil factor 3 (TFF3) has been reported to be a biomarker of several malignancies. We further investigated whether TFF3 can be applied as a biomarker for and predictor of responses to chemotherapy in gastrointestinal cancer. METHODS Serum and urine samples were collected from 90 patients with gastric cancer, 128 patients with colorectal cancer and 91 healthy individuals. Serum and urine TFF3 levels were measured using an ELISA. RESULTS Serum and urine TFF3 levels were significantly higher in the patients with gastric and colorectal cancer compared with the healthy individuals (P < 0.05). Higher serum levels of TFF3 were significantly correlated with distant metastasis and an advanced stage in the two types of cancer (P < 0.05). Age and the number of lymph node metastases were significantly correlated with serum TFF3 levels in colorectal cancer, and decreased serum TFF3 levels were significantly correlated with responses to chemotherapy in both the gastric and the colorectal cancer partial response (PR) groups. A combination of serum and urine data did not significantly improve the detection of either cancer, although urine levels have shown a significant negative relationship with the glomerular filtration rate (GFR). CONCLUSIONS Our data indicate that TFF3 may be an effective biomarker of tumor stage and the presence of distant metastasis, and may be a pharmacodynamic marker of response to chemotherapy in gastrointestinal cancer.
Collapse
Affiliation(s)
- Li Xiao
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen 361004, Fujian Province, China
| | - Yun-Peng Liu
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen 361004, Fujian Province, China
| | - Chuan-Xing Xiao
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen 361004, Fujian Province, China
| | - Jian-Lin Ren
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen 361004, Fujian Province, China
| | - Bayasi Guleng
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen 361004, Fujian Province, China.,Faculty of Clinical Medicine, Medical College of Xiamen University, Xiangan South Road, Xiangan District, Xiamen 361102, Fujian Province, China
| |
Collapse
|
34
|
Huang Z, Zhang X, Lu H, Wu L, Wang D, Zhang Q, Ding H. Serum trefoil factor 3 is a promising non-invasive biomarker for gastric cancer screening: a monocentric cohort study in China. BMC Gastroenterol 2014; 14:74. [PMID: 24720760 PMCID: PMC4012276 DOI: 10.1186/1471-230x-14-74] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/24/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The search for better non-invasive biomarkers for gastric cancer remains ongoing. We investigated the predictive power of serum trefoil factor (TFF) levels as biomarkers for gastric cancer in comparison with the pepsinogen (PG) test. METHODS Patients with gastric cancer, chronic atrophic gastritis (CAG) or chronic non-atrophic gastritis (CNAG), and healthy people were recruited. Serum concentrations of TFFs, PG I, and PG II, as well as the presence of antibodies against Helicobacter pylori, were measured by enzyme-linked immunosorbent assays (ELISA). Receiver operating characteristics (ROC) were used to compare the predictive powers of the selected factors. RESULTS The serum concentrations of TFF1, TFF2, and TFF3 in the control groups were significantly lower than those in the gastric cancer group with the exception of TFF2 which was elevated in CAG. The area under the ROC curve for TFF3 was greater than that for the PG I/II ratio (0.81 vs 0.78). TFF3 also had a significantly higher predictive power for distinguishing gastric cancer than the PG test (odds ratio: 10.33 vs 2.57). Moreover, combining the serum TFF3 and PG tests for gastric cancer had better predictive power than either alone. CONCLUSIONS Serum TFF3 may be a better predictor of gastric cancer than the PG test, while the combined testing of serum PG and TFF3 could further improve the efficacy of gastric cancer screening.
Collapse
Affiliation(s)
- Zhigang Huang
- Department of Gastroenterology, Lihuili Hospital of Ningbo Medical Center, 57# Xingning Road, Ningbo 315000, China.
| | | | | | | | | | | | | |
Collapse
|
35
|
Zheng Q, Gao J, Li H, Guo W, Mao Q, Gao E, Zhu YQ. Trefoil factor 3 peptide regulates migration via a Twist-dependent pathway in gastric cell. Biochem Biophys Res Commun 2013; 438:6-12. [PMID: 23845905 DOI: 10.1016/j.bbrc.2013.06.115] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 06/29/2013] [Indexed: 12/22/2022]
Abstract
Trefoil factor 3 (TFF3) is a member of the TFF-domain peptide family and essential in regulating cell migration and maintaining mucosal integrity in gastrointestinal tract. However, the role of TFF3 and its downstream regulating mechanisms in cancer cell migration remain unclear. We previously reported that TFF3 prolonged the up-regulation of Twist protein to modulate IL-8 secretion in intestinal epithelial cells. In this study, we investigated the role of Twist protein in TFF3-induced migration of SGC7901 cells. While Twist was activated by TFF3, siRNA-mediated knockdown of Twist abolished TFF3-induced cell migration. Furthermore, the migration related marker CK-8 as well as ZO-1 and MMP-9 was also regulated by TFF3 via a Twist-dependent mechanism. Our study suggests that Twist, as an important potential downstream effector, plays a key role in TFF3-modulated metastasis in gastric cancer and can be a promising therapeutic target against intestinal-type gastric cancer.
Collapse
Affiliation(s)
- Qianqian Zheng
- Division of Cell Pathobiology, Key Laboratory of Medical Cell Biology, Ministry of Education, Department of Cell Biology, College of Basic Medical Science, China Medical University, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Influence of Helicobacter pylori status and eradication on the serum levels of trefoil factors and pepsinogen test: serum trefoil factor 3 is a stable biomarker. Gastric Cancer 2013; 16:329-37. [PMID: 22907485 DOI: 10.1007/s10120-012-0185-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 07/31/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIM Emerging data indicate that serum trefoil factors (TFFs), especially TFF3, could be potential biomarkers for gastric cancer risk. We aimed to evaluate the influence of Helicobacter pylori (H. pylori) status and eradication on serum TFFs and the pepsinogen test. METHODS Healthy individuals who underwent a thorough medical checkup were enrolled in study 1, and gastric ulcer patients who undertook H. pylori eradication therapy were enrolled in studies 2 and 3. Serum levels of the TFFs (TFF1, TFF2 and TFF3), H. pylori antibody and pepsinogen test were examined in all studies. In study 3, TFF expressions in biopsy samples of the gastric mucosa were additionally examined before and 2 months after eradication. RESULTS In 1,260 healthy individuals enrolled in study 1, serum TFF1 and TFF2 levels were markedly different between H. pylori antibody-positive and -negative participants (P < 0.0001). Differences in serum TFF3 levels between H. pylori antibody-positive (5.85 ± 3.93 ng/ml) and -negative subjects (5.27 ± 2.38 ng/ml) were statistically significant (P = 0.002) but small in absolute value. In 178 gastric ulcer patients enrolled in study 2, serum TFF1, TFF2 and positive rates of the pepsinogen test significantly decreased 2 months after H. pylori eradication therapy (P < 0.001). In contrast, serum TFF3 levels and positive rates of high TFF3 levels (≥7 ng/ml) did not significantly change with H. pylori-eradication until 5 years after eradication. In 18 gastric ulcer patients (study 3), TFF1 and TFF2 were mainly expressed in the foveolar epithelium, and TFF2 was additionally expressed in the pyloric glands. These expressions significantly decreased with H. pylori eradication. TFF3s were scarcely expressed in the gastric mucosa except in goblet cells of intestinal metaplasia, which did not change with H. pylori eradication. CONCLUSION In serum TFFs and pepsinogen tests, only serum TFF3s were not significantly affected by H. pylori eradication, suggesting that serum TFF3 could be a stable biomarker of gastric cancer risk even after H.pylori eradication in contrast with the pepsinogen test.
Collapse
|
37
|
Samson MH. Quantitative measurements of trefoil factor family peptides: possibilities and pitfalls. Scandinavian Journal of Clinical and Laboratory Investigation 2013; 73:193-202. [PMID: 23391285 DOI: 10.3109/00365513.2013.765962] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The trefoil factor family (TFF) peptides TFF1, TFF2, and TFF3 are produced and secreted by mucous membranes throughout the body. Their importance for the protection and repair of epithelial surfaces is well established, and the three peptides are present in various amounts in mucosal secretions as well as in the circulation. They have been linked to both inflammatory diseases and to various types of cancer, and serum concentrations of TFF3 show a more than 47-fold increase during pregnancy. Several both commercial and in-house immunoassays exist, but a number of methodological issues remain unresolved. This review describes methodological challenges in the measurement of the peptides in humans, and summarizes current knowledge concerning the occurrence and possible significance of the peptides in human health and disease.
Collapse
|
38
|
Im S, Yoo C, Jung JH, Choi HJ, Yoo J, Kang CS. Reduced expression of TFF1 and increased expression of TFF3 in gastric cancer: correlation with clinicopathological parameters and prognosis. Int J Med Sci 2013; 10:133-40. [PMID: 23329884 PMCID: PMC3547210 DOI: 10.7150/ijms.5500] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 12/26/2012] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES The trefoil factor family (TFF) is composed of three thermostable, and protease-resistant proteins, named TFF1, TFF2 and TFF3, and plays a role in gastrointestinal mucosal defence and repair. Recently, TFFs have been found to be related to the development of various types of cancer. This study assessed the relationship between the expression of TFF1 and TFF3 and the clinicopathological parameters in gastric carcinoma (GC). MATERIALS AND METHODS The expression of TFF1 and TFF3 was analyzed by immunohistochemistry in 292 GCs and 20 normal gastric tissues. RESULTS All normal gastric tissues expressed TFF1, but 53.8% of GCs showed reduced TFF1 expression. However, TFF3 was not detected in normal gastric tissues and 44.2% of GCs showed a high level of expression. Highly expressed TFF3 was significantly correlated with lymph node metastasis, lymphatic invasion, vein invasion, and advanced stage. The overall survival was shorter in patients with high expression of TFF3 than in those with low expression of TFF3 in 292 GCs and in 125 early GCs (EGCs). Moreover, in patients with EGCs, high expression of TFF3, associated with reduced expression of TFF1, was determined as an independent poor prognostic marker. CONCLUSIONS Reduced expression of TFF1 and increased expression of TFF3 may play a role in the carcinogenesis of gastric cancer. Furthermore, high expression of TFF3 with reduced expression of TFF1 may be a marker of poor prognosis for patients with EGC.
Collapse
Affiliation(s)
- Soyoung Im
- Department of Hospital Pathology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Republic of Korea
| | | | | | | | | | | |
Collapse
|