1
|
Reduzzi C, Nicolo' E, Singhal S, Venetis K, Ortega-Franco A, de Miguel-Perez D, Dipasquale A, Gouda MA, Saldanha EF, Kasi PM, Jantus-Lewintre E, Fusco N, Malapelle U, Gandara DR, Rolfo C, Serrano MJ, Cristofanilli M. Unveiling the impact of circulating tumor cells: Two decades of discovery and clinical advancements in solid tumors. Crit Rev Oncol Hematol 2024; 203:104483. [PMID: 39159706 DOI: 10.1016/j.critrevonc.2024.104483] [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: 05/21/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024] Open
Abstract
Circulating tumor cells (CTCs) enumeration and molecular profiling hold promise in revolutionizing the management of solid tumors. Their understanding has evolved significantly over the past two decades, encompassing pivotal biological discoveries and clinical studies across various malignancies. While for some tumor types, such as breast, prostate, and colorectal cancer, CTCs are ready to enter clinical practice, for others, additional research is required. CTCs serve as versatile biomarkers, offering insights into tumor biology, metastatic progression, and treatment response. This review summarizes the latest advancements in CTC research and highlights future directions of investigation. Special attention is given to concurrent evaluations of CTCs and other circulating biomarkers, particularly circulating tumor DNA. Multi-analyte assessment holds the potential to unlock the full clinical capabilities of liquid biopsy. In conclusion, CTCs represent a transformative biomarker in precision oncology, offering extraordinary opportunities to translate scientific discoveries into tangible improvements in patient care.
Collapse
Affiliation(s)
- Carolina Reduzzi
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY 10021, USA.
| | - Eleonora Nicolo'
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY 10021, USA.
| | - Surbhi Singhal
- Division of Hematology and Oncology, Department of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Konstantinos Venetis
- Division of Pathology, IEO European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Ana Ortega-Franco
- Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, United Kingdom
| | - Diego de Miguel-Perez
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Angelo Dipasquale
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Erick F Saldanha
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, ON, Canada
| | - Pashtoon M Kasi
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY 10021, USA
| | - Eloisa Jantus-Lewintre
- Department of Biotechnology, Universitat Politècnica de València, Unidad Mixta TRIAL (Fundación para la Investigación del Hospital General Universitario de Valencia y Centro de Investigación Príncipe Felipe) and CIBERONC, Valencia, Spain
| | - Nicola Fusco
- Division of Pathology, IEO European Institute of Oncology IRCCS, Milan 20141, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan 20121, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Napoli 80131, Italy
| | - David R Gandara
- Division of Hematology and Oncology, Department of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Maria Jose Serrano
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain; Pathological Anatomy Unit, Molecular Pathology Laboratory,Virgen de las Nieves. University Hospital, Av. Dr. Olóriz 16, Granada 18012, Spain
| | - Massimo Cristofanilli
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY 10021, USA
| |
Collapse
|
2
|
Zhu JW, Gong LZ, Wang QW. Serum tumor markers (carcinoembryonic antigen, carbohydrate antigen 19-9, carbohydrate antigen 72-4, carbohydrate antigen 24-2, ferritin) and gastric cancer prognosis correlation. World J Gastrointest Surg 2024; 16:2808-2814. [PMID: 39351575 PMCID: PMC11438793 DOI: 10.4240/wjgs.v16.i9.2808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/02/2024] [Accepted: 07/22/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Gastric cancer is a kind of malignant tumor which is prevalent all over the world. Although some progress has been made in the treatment of gastric cancer, its prognosis is still not optimistic, so it is of great significance to find reliable prognostic indicators to guide the treatment and management of patients with gastric cancer. AIM To explore the relationship between serum levels of five biomarkers [carcinoembryonic antigen (CEA), carbohydrate antigen (CA) 19-9, CA72-4, CA24-2, and ferritin] and prognosis in patients with gastric cancer. METHODS This study included 200 patients with gastric adenocarcinoma, and conducted an in-depth analysis of their baseline characteristics, relationship between tumor markers and staging, and prognosis. The study found that CA19-9 has a significant correlation with tumor stage, the average levels of CA24-2, CEA, CA72-4 and ferritin were slightly increased disregarding the stage of tumor. Survival analysis showed that increases in CEA, CA19-9, CA24-2, and ferritin were all associated with shortened overall survival of patients. Further multivariate analysis revealed that elevated serum CA72-4 levels were an independent adverse prognostic factor. RESULTS This study reveals that there is a significant correlation between the expression levels of serum tumor markers CEA, CA19-9, CA72-4, CA24-2 and ferritin in patients with gastric cancer and prognosis, and can be used as important indicators for prognostic evaluation of gastric cancer. In particular, markers that appear abnormally elevated initially may help identify gastric cancer patients with poor prognosis. CONCLUSION Serum CEA and CA19-9 play an important role in the prognosis assessment of gastric cancer, and are effective tools to guide clinical practice and optimize individualized treatment strategies for gastric cancer patients.
Collapse
Affiliation(s)
- Jie-Wen Zhu
- Department of Clinical Laboratory, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi Tumor Molecular Diagnosis and Treatment Key Laboratory, Huangshi 435000, Hubei Province, China
| | - Ling-Zhen Gong
- Department of Clinical Laboratory, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi Tumor Molecular Diagnosis and Treatment Key Laboratory, Huangshi 435000, Hubei Province, China
| | - Qian-Wen Wang
- Department of Clinical Laboratory, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi Tumor Molecular Diagnosis and Treatment Key Laboratory, Huangshi 435000, Hubei Province, China
| |
Collapse
|
3
|
Qi X, Liu M, Xu K, Tan F, Gao P, Yao Z, Zhang N, Yang H, Zhang C, Xing J, Cui M, Su X. Risk factors and clinical significance of lower perigastric lymph node metastases in Siewert type II and III esophagogastric junction adenocarcinoma: a retrospective cohort study. Surg Endosc 2024; 38:3828-3837. [PMID: 38822144 PMCID: PMC11219428 DOI: 10.1007/s00464-024-10875-y] [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: 02/26/2024] [Accepted: 04/20/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND No consensus has been concluded with regarding to the scope of lymph node (LN) dissection for Siewert type II and III adenocarcinoma of the esophagogastric junction (AEG). This study aimed to explore risk factors for lower perigastric LN (LPLN) metastases (including no. 4d, 5, 6, and 12a LN stations) and analyze the indications for LPLN dissection. METHODS In total, 302 consecutive patients with Siewert type II and III AEG who underwent total gastrectomy (TG) were enrolled. The logistic regression model was used to perform uni- and multivariate analyses of risk factors for LPLN metastases. Kaplan-Meier curves were used for survival analysis, and log-rank tests were used for group comparisons. Basing on the guidelines of Japanese Gastric Cancer Association, the LN metastases (LNM) as well as the efficiency index (EI) of each LN station was further evaluated. RESULTS The independent risk factors for LPLN metastases in patients with Siewert type II and III AEG were distance from the esophagogastric junction (EGJ) to the distal end of the tumor (> 4.0 cm), preoperative carcinoembryonic antigen (CEA) ( +), pT4 stage, and HER-2 ( +). LPLN metastases was an independent risk factor for overall survival following TG. The LNM and EI of LPLN were 8.6% and 2.31%, respectively. The LNM of LPLN > 10% under the stratification of the distance from the EGJ to the distal end of the tumor (> 4.0 cm), pT4, preoperative CEA ( +), and HER-2 ( +) exhibited EI values of 3.55%, 2.09%, 2.51%, and 3.64%, respectively. CONCLUSIONS LPLN metastases was a malignant factor for the prognosis of patients with Siewert type II and III AEG. For patients with preoperative CEA ( +), pT4 stage, HER-2 ( +), and the distance from the EGJ to the distal end of the tumor (> 4.0 cm), TG with LPLN dissection is prioritized for clinical recommendation.
Collapse
Affiliation(s)
- Xinyu Qi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Maoxing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Kai Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Fei Tan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Pin Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Zhendan Yao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Nan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Hong Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Chenghai Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Jiadi Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China.
| | - Ming Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, People's Republic of China
| | - Xiangqian Su
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
4
|
Wang X, Wang L, Lin H, Zhu Y, Huang D, Lai M, Xi X, Huang J, Zhang W, Zhong T. Research progress of CTC, ctDNA, and EVs in cancer liquid biopsy. Front Oncol 2024; 14:1303335. [PMID: 38333685 PMCID: PMC10850354 DOI: 10.3389/fonc.2024.1303335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024] Open
Abstract
Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and extracellular vehicles (EVs) have received significant attention in recent times as emerging biomarkers and subjects of transformational studies. The three main branches of liquid biopsy have evolved from the three primary tumor liquid biopsy detection targets-CTC, ctDNA, and EVs-each with distinct benefits. CTCs are derived from circulating cancer cells from the original tumor or metastases and may display global features of the tumor. ctDNA has been extensively analyzed and has been used to aid in the diagnosis, treatment, and prognosis of neoplastic diseases. EVs contain tumor-derived material such as DNA, RNA, proteins, lipids, sugar structures, and metabolites. The three provide different detection contents but have strong complementarity to a certain extent. Even though they have already been employed in several clinical trials, the clinical utility of three biomarkers is still being studied, with promising initial findings. This review thoroughly overviews established and emerging technologies for the isolation, characterization, and content detection of CTC, ctDNA, and EVs. Also discussed were the most recent developments in the study of potential liquid biopsy biomarkers for cancer diagnosis, therapeutic monitoring, and prognosis prediction. These included CTC, ctDNA, and EVs. Finally, the potential and challenges of employing liquid biopsy based on CTC, ctDNA, and EVs for precision medicine were evaluated.
Collapse
Affiliation(s)
- Xiaoling Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Lijuan Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Haihong Lin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Yifan Zhu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Mi Lai
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xuxiang Xi
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Junyun Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Wenjuan Zhang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| |
Collapse
|
5
|
Zhang X, Wu YY, Qin YY, Lin FQ. The combined detection of hematological indicators is used for the differential diagnosis of colorectal cancer and benign-colorectal lesions. Cancer Biomark 2024; 39:223-230. [PMID: 38217586 PMCID: PMC11091605 DOI: 10.3233/cbm-230157] [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: 05/26/2023] [Accepted: 11/16/2023] [Indexed: 01/15/2024]
Abstract
OBJECTIVE This article aims to investigate the clinical value of hemoglobin/red cell distribution width ratio (Hb/RDW), C-reactive protein/albumin ratio (CAR) and plateletcrit (PCT) combined with carcinoembryonic antigen (CEA) in colorectal cancer (CRC) auxiliary diagnosis. METHODS We retrospectively analyzed in 718 subjects (212 with CRC, 209 with benign colorectal lesions (BCL), 111 with other cancers, and 186 healthy controls). RESULTS The CAR, PCT, and CEA in the CRC group were higher than those in the BCL, other cancers, and the healthy control group. However, Hb/RDW in the CRC group was lower than the other three groups. Moreover, there were significant differences in Hb/RDW and CEA among different T-N-M stages (all P< 0.05). Multivariate logistic regression showed that low level of Hb/RDW and high level of CAR, CEA, PCT were risk factors for CRC, and are correlated with CRC stage. Additionally, the area under the receiver operating characteristic curve (AUC) of Hb/RDW+CEA (AUC: 0.735), CAR+CEA (AUC: 0.748), PCT+CEA (AUC: 0.807) was larger than that of Hb/RDW (AUC: 0.503), CAR (AUC: 0.614), or PCT (AUC: 0.713) alone (all P< 0.001) in distinguishing CRC from BCL. CONCLUSIONS Hb/RDW, CAR, PCT, and CEA are independent risk factors for CRC. Hb/RDW, CAR, and PCT combined with CEA have significant value for auxiliary differential diagnosis of CRC and BCL.
Collapse
Affiliation(s)
- Xuan Zhang
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yang-Yang Wu
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yuan-Yuan Qin
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fa-Quan Lin
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
6
|
Jin T, Liang PP, Chen ZH, He FJ, Li ZD, Chen ZW, Hu JK, Yang K. Association between circulating tumor cells in the peripheral blood and the prognosis of gastric cancer patients: a meta-analysis. Ther Adv Med Oncol 2023; 15:17588359231183678. [PMID: 37435560 PMCID: PMC10331349 DOI: 10.1177/17588359231183678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/31/2023] [Indexed: 07/13/2023] Open
Abstract
Background Research on the correlation between circulating tumor cells (CTCs) and gastric cancer (GC) has increased rapidly in recent years. However, whether CTCs are associated with GC patient prognosis is highly controversial. Objective This study aims to evaluate the value of CTCs to predict the prognosis of GC patients. Design A meta-analysis. Data Sources and Methods We searched the PubMed, Embase, and Cochrane Library databases for studies that reported the prognostic value of CTCs in GC patients before October 2022. The association between CTCs and overall survival (OS) and disease-free survival (DFS)/recurrence-free survival (RFS) and progression-free survival (PFS) of GC patients was assessed. Subgroup analyses were stratified by sampling times (pre-treatment and post-treatment), detection targets, detection method, treatment method, tumor stage, region, and HR (Hazard Ratio) extraction methods. Sensitivity analysis was performed by removing individual studies to assess the stability of the results. Publication bias was evaluated using funnel plots, Egger's test, and Begg's test. Results We initially screened 2000 studies, of which 28 were available for further analysis, involving 2383 GC patients. The pooled analysis concluded that the detection of CTCs was associated with poor OS (HR = 1.933, 95% CI 1.657-2.256, p < 0.001), DFS/RFS (HR = 3.228, 95% CI 2.475-4.211, p < 0.001), and PFS (HR = 3.272, 95% CI 1.970-5.435, p < 0.001). Furthermore, the subgroup analysis stratified by tumor stage (p < 0.01), HR extraction methods (p < 0.001), detection targets (p < 0.001), detection method (p < 0.001), sampling times (p < 0.001), and treatment method (p < 0.001) all showed that CTC detection was associated with poor OS and DFS/RFS for GC patients. Furthermore, the study showed that CTCs were associated with the poor DFS/RFS of GC when CTCs were detected for patients from Asian or No-Asian regions (p < 0.05). In addition, higher CTCs predicted poorer OS for GC patients who are from Asian regions (p < 0.001), but without statistical difference for GC patients from No-Asian regions (p = 0.490). Conclusion CTC detection in peripheral blood was associated with poor OS, DFS/RFS, and PFS in patients with GC.
Collapse
Affiliation(s)
- Tao Jin
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pan-Ping Liang
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ze-Hua Chen
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng-Jun He
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ze-Dong Li
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zheng-Wen Chen
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian-Kun Hu
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kun Yang
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
7
|
Akashi T, Okumura T, Terabayashi K, Yoshino Y, Tanaka H, Yamazaki T, Numata Y, Fukuda T, Manabe T, Baba H, Miwa T, Watanabe T, Hirano K, Igarashi T, Sekine S, Hashimoto I, Shibuya K, Hojo S, Yoshioka I, Matsui K, Yamada A, Sasaki T, Fujii T. The use of an artificial intelligence algorithm for circulating tumor cell detection in patients with esophageal cancer. Oncol Lett 2023; 26:320. [PMID: 37332339 PMCID: PMC10272959 DOI: 10.3892/ol.2023.13906] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/25/2023] [Indexed: 06/20/2023] Open
Abstract
Despite recent advances in multidisciplinary treatments of esophageal squamous cell carcinoma (ESCC), patients frequently suffer from distant metastasis after surgery. For numerous types of cancer, circulating tumor cells (CTCs) are considered predictors of distant metastasis, therapeutic response and prognosis. However, as more markers of cytopathological heterogeneity are discovered, the overall detection process for the expression of these markers in CTCs becomes increasingly complex and time consuming. In the present study, the use of a convolutional neural network (CNN)-based artificial intelligence (AI) for CTC detection was assessed using KYSE ESCC cell lines and blood samples from patients with ESCC. The AI algorithm distinguished KYSE cells from peripheral blood-derived mononuclear cells (PBMCs) from healthy volunteers, accompanied with epithelial cell adhesion molecule (EpCAM) and nuclear DAPI staining, with an accuracy of >99.8% when the AI was trained on the same KYSE cell line. In addition, AI trained on KYSE520 distinguished KYSE30 from PBMCs with an accuracy of 99.8%, despite the marked differences in EpCAM expression between the two KYSE cell lines. The average accuracy of distinguishing KYSE cells from PBMCs for the AI and four researchers was 100 and 91.8%, respectively (P=0.011). The average time to complete cell classification for 100 images by the AI and researchers was 0.74 and 630.4 sec, respectively (P=0.012). The average number of EpCAM-positive/DAPI-positive cells detected in blood samples by the AI was 44.5 over 10 patients with ESCC and 2.4 over 5 healthy volunteers (P=0.019). These results indicated that the CNN-based image processing algorithm for CTC detection provides a higher accuracy and shorter analysis time compared to humans, suggesting its applicability for clinical use in patients with ESCC. Moreover, the finding that AI accurately identified even EpCAM-negative KYSEs suggested that the AI algorithm may distinguish CTCs based on as yet unknown features, independent of known marker expression.
Collapse
Affiliation(s)
- Takahisa Akashi
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Tomoyuki Okumura
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Kenji Terabayashi
- Department of Mechanical and Intellectual Systems Engineering, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Yuki Yoshino
- Department of Mechanical and Intellectual Systems Engineering, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Haruyoshi Tanaka
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Takeyoshi Yamazaki
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Yoshihisa Numata
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Takuma Fukuda
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Takahiro Manabe
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Hayato Baba
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Takeshi Miwa
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Toru Watanabe
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Katsuhisa Hirano
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Takamichi Igarashi
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Shinichi Sekine
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Isaya Hashimoto
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Kazuto Shibuya
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Shozo Hojo
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Isaku Yoshioka
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Koshi Matsui
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| | - Akane Yamada
- Department of Mechanical and Intellectual Systems Engineering, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Tohru Sasaki
- Department of Mechanical and Intellectual Systems Engineering, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
8
|
Asawa S, Nüesch M, Gvozdenovic A, Aceto N. Circulating tumour cells in gastrointestinal cancers: food for thought? Br J Cancer 2023; 128:1981-1990. [PMID: 36932192 DOI: 10.1038/s41416-023-02228-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/17/2023] [Accepted: 03/02/2023] [Indexed: 03/19/2023] Open
Abstract
Gastrointestinal (GI) cancers account for 35% of cancer-related deaths, predominantly due to their ability to spread and generate drug-tolerant metastases. Arising from different locations in the GI system, the majority of metastatic GI malignancies colonise the liver and the lungs. In this context, circulating tumour cells (CTCs) are playing a critical role in the formation of new metastases, and their presence in the blood of patients has been correlated with a poor outcome. In addition to their prognostic utility, prospective targeting of CTCs may represent a novel, yet ambitious strategy in the fight against metastasis. A better understanding of CTC biology, mechanistic underpinnings and weaknesses may facilitate the development of previously underappreciated anti-metastasis approaches. Here, along with related clinical studies, we outline a selection of the literature describing biological features of CTCs with an impact on their metastasis forming ability in different GI cancers.
Collapse
Affiliation(s)
- Simran Asawa
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Manuel Nüesch
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland.
| |
Collapse
|
9
|
Ding L, Liang M, Li C, Ji X, Zhang J, Xie W, Reis RL, Li FR, Gu S, Wang Y. Design Strategies of Tumor-Targeted Delivery Systems Based on 2D Nanomaterials. SMALL METHODS 2022; 6:e2200853. [PMID: 36161304 DOI: 10.1002/smtd.202200853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/11/2022] [Indexed: 06/16/2023]
Abstract
Conventional chemotherapy and radiotherapy are nonselective and nonspecific for cell killing, causing serious side effects and threatening the lives of patients. It is of great significance to develop more accurate tumor-targeting therapeutic strategies. Nanotechnology is in a leading position to provide new treatment options for cancer, and it has great potential for selective targeted therapy and controlled drug release. 2D nanomaterials (2D NMs) have broad application prospects in the field of tumor-targeted delivery systems due to their special structure-based functions and excellent optical, electrical, and thermal properties. This review emphasizes the design strategies of tumor-targeted delivery systems based on 2D NMs from three aspects: passive targeting, active targeting, and tumor-microenvironment targeting, in order to promote the rational application of 2D NMs in clinical practice.
Collapse
Affiliation(s)
- Lin Ding
- School of Pharmaceutical Sciences and The First Affiliated Hospital, Hainan Medical University, Haikou, 570228, P. R. China
- The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, 518055, China
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, Guangdong, 518055, China
- Guangdong Engineering Technology Research Centerof Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Minli Liang
- The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, 518055, China
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, Guangdong, 518055, China
- Guangdong Engineering Technology Research Centerof Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Chenchen Li
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Xinting Ji
- School of Pharmaceutical Sciences and The First Affiliated Hospital, Hainan Medical University, Haikou, 570228, P. R. China
| | - Junfeng Zhang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Weifen Xie
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials Biodegradables and Biomimetics, University of Minho, Guimarães, 4805-017, Portugal
| | - Fu-Rong Li
- The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, 518055, China
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, Guangdong, 518055, China
- Guangdong Engineering Technology Research Centerof Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Shuo Gu
- School of Pharmaceutical Sciences and The First Affiliated Hospital, Hainan Medical University, Haikou, 570228, P. R. China
| | - Yanli Wang
- School of Pharmaceutical Sciences and The First Affiliated Hospital, Hainan Medical University, Haikou, 570228, P. R. China
| |
Collapse
|
10
|
Zhang Z, Wu H, Chong W, Shang L, Jing C, Li L. Liquid biopsy in gastric cancer: predictive and prognostic biomarkers. Cell Death Dis 2022; 13:903. [PMID: 36302755 PMCID: PMC9613678 DOI: 10.1038/s41419-022-05350-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022]
Abstract
Gastric cancer (GC) is a high-incidence cancer worldwide. Most patients are diagnosed at an advanced stage, by which time they have limited treatment options and poor prognosis. Early diagnosis and precise treatment are important. In the past few years, emerging research has been conducted on the use of non-invasive liquid biopsy, with its advantages of minimal invasiveness and repeated sampling, to monitor tumor occurrence and recurrence in real time and to evaluate prognosis and treatment response. Many studies have demonstrated the potential of liquid biopsy in GC, and the detection of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), circulating free DNA (cfDNA), and exosomes has achieved gratifying results. In this review, we summarize evolving technologies for and information regarding liquid biopsy, the most recently discovered GC liquid biopsy biomarkers, and ongoing clinical trials and discuss the challenges and application prospects of liquid biopsy in GC.
Collapse
Affiliation(s)
- Zihao Zhang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Hao Wu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, 250021, China
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, 250021, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, 250021, China.
| | - Changqing Jing
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, 250021, China.
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, 250021, China.
| |
Collapse
|
11
|
Jiang T, Mei L, Yang X, Sun T, Wang Z, Ji Y. Biomarkers of gastric cancer: current advancement. Heliyon 2022; 8:e10899. [PMID: 36247151 PMCID: PMC9561735 DOI: 10.1016/j.heliyon.2022.e10899] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/22/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
Gastric cancer (GC) is one of the most prevalent malignant types worldwide, especially in East Asia. Due to its frequently advanced stage at diagnosis, the mortality from GC is high and the prognosis is still unsatisfactory. Thus, early detection using effective screening approaches is vital to decrease the morbidity and mortality of GC. Interestingly, biomarkers can be used for diagnosis, prediction of sensitivity to treatment, and prognosis in GC. The potential biomarkers detectable in liquid biopsies such as circulating tumor cells (CTCs), long non-coding RNAs (lncRNAs), cell-free DNA (cfDNA), microRNAs, and exosomes reveal numerous information regarding the early prediction and the outcomes for GC patients. Additionally, using the novel serum biomarkers has opened up new opportunities for diagnosing and monitoring patients with GC. This review mainly summarizes the novel progress and approaches in GC biomarkers, which could be potentially used for early diagnosis and therapy monitoring. Meanwhile, we also discussed the advantages, disadvantages, and future perspectives of GC biomarkers.
Collapse
Affiliation(s)
- Tiantian Jiang
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Lin Mei
- Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Xiao Yang
- Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Tingkai Sun
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Zhidong Wang
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Yuanyuan Ji
- Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| |
Collapse
|
12
|
Ju S, Chen C, Zhang J, Xu L, Zhang X, Li Z, Chen Y, Zhou J, Ji F, Wang L. Detection of circulating tumor cells: opportunities and challenges. Biomark Res 2022; 10:58. [PMID: 35962400 PMCID: PMC9375360 DOI: 10.1186/s40364-022-00403-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Circulating tumor cells (CTCs) are cells that shed from a primary tumor and travel through the bloodstream. Studying the functional and molecular characteristics of CTCs may provide in-depth knowledge regarding highly lethal tumor diseases. Researchers are working to design devices and develop analytical methods that can capture and detect CTCs in whole blood from cancer patients with improved sensitivity and specificity. Techniques using whole blood samples utilize physical prosperity, immunoaffinity or a combination of the above methods and positive and negative enrichment during separation. Further analysis of CTCs is helpful in cancer monitoring, efficacy evaluation and designing of targeted cancer treatment methods. Although many advances have been achieved in the detection and molecular characterization of CTCs, several challenges still exist that limit the current use of this burgeoning diagnostic approach. In this review, a brief summary of the biological characterization of CTCs is presented. We focus on the current existing CTC detection methods and the potential clinical implications and challenges of CTCs. We also put forward our own views regarding the future development direction of CTCs.
Collapse
Affiliation(s)
- Siwei Ju
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Cong Chen
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Jiahang Zhang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Lin Xu
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Xun Zhang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Zhaoqing Li
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Yongxia Chen
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Jichun Zhou
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Feiyang Ji
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China.
| | - Linbo Wang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China.
| |
Collapse
|
13
|
Chen K, Chen Z, Ou M, Wang J, Huang X, Wu Y, Zhong W, Yang J, Huang J, Huang M, Pan D. Clinical significance of circulating tumor cells in predicating the outcomes of patients with colorectal cancer. Clinics (Sao Paulo) 2022; 77:100070. [PMID: 36087570 PMCID: PMC9464896 DOI: 10.1016/j.clinsp.2022.100070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/04/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Relapse and metastasis of patients with Colorectal Cancer (CRC) is the major obstacle to the long-term life of patients. Its mechanisms remain defined. METHODS A total of 48 CRC patients were enrolled and 68 samples were obtained from the peripheral blood of patients before or after treatments in this study. Twenty non-cancer patients were also detected as a negative control. Circulating Tumor Cells (CTCs), including Epithelial CTCs (eCTCs), Mesenchymal (MCTCs), and epithelial/mesenchymal mixed phenotypes (mixed CTCs), were identified by CanPatrolTM CTC enrichment and RNA in situ hybridization. The relationship between CTCs number and Progression-Free Survival (PFS) or Overall Survival (OS) was evaluated. RESULTS Thirty-four of 48 patients (70.8%) were found to have positive CTCs. Total CTCs and MCTCs in the post-treatment had a significant correlation PFS and OS. When total CTCs or MCTCs in 5 mL blood of patients were more than 6 CTCs or 5 MCTCs, PFS of the patients was significantly shorter (p < 0.05) than that in patients with less than 6 CTCs or 5 MCTCs. The patients with > 5 CTCs count changes were found to exhibit poor PFS and OS rates (p < 0.05). CONCLUSION Total CTCs and MCTCs number detection in patients with colorectal cancer was very useful biomarker for predicting the prognosis of patients. Higher CTCs or MCTCs had poorer PFS and OS rates.
Collapse
Affiliation(s)
- Kehe Chen
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zhenxiang Chen
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Mei Ou
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Junping Wang
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiao Huang
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yingying Wu
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Wenhe Zhong
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jiao Yang
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jinging Huang
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Min Huang
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Deng Pan
- Department of Medical Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
| |
Collapse
|
14
|
Lin D, Shen L, Luo M, Zhang K, Li J, Yang Q, Zhu F, Zhou D, Zheng S, Chen Y, Zhou J. Circulating tumor cells: biology and clinical significance. Signal Transduct Target Ther 2021; 6:404. [PMID: 34803167 PMCID: PMC8606574 DOI: 10.1038/s41392-021-00817-8] [Citation(s) in RCA: 347] [Impact Index Per Article: 115.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023] Open
Abstract
Circulating tumor cells (CTCs) are tumor cells that have sloughed off the primary tumor and extravasate into and circulate in the blood. Understanding of the metastatic cascade of CTCs has tremendous potential for the identification of targets against cancer metastasis. Detecting these very rare CTCs among the massive blood cells is challenging. However, emerging technologies for CTCs detection have profoundly contributed to deepening investigation into the biology of CTCs and have facilitated their clinical application. Current technologies for the detection of CTCs are summarized herein, together with their advantages and disadvantages. The detection of CTCs is usually dependent on molecular markers, with the epithelial cell adhesion molecule being the most widely used, although molecular markers vary between different types of cancer. Properties associated with epithelial-to-mesenchymal transition and stemness have been identified in CTCs, indicating their increased metastatic capacity. Only a small proportion of CTCs can survive and eventually initiate metastases, suggesting that an interaction and modulation between CTCs and the hostile blood microenvironment is essential for CTC metastasis. Single-cell sequencing of CTCs has been extensively investigated, and has enabled researchers to reveal the genome and transcriptome of CTCs. Herein, we also review the clinical applications of CTCs, especially for monitoring response to cancer treatment and in evaluating prognosis. Hence, CTCs have and will continue to contribute to providing significant insights into metastatic processes and will open new avenues for useful clinical applications.
Collapse
Affiliation(s)
- Danfeng Lin
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lesang Shen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Luo
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinfan Li
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Yang
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fangfang Zhu
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhou
- Department of Surgery, Traditional Chinese Medical Hospital of Zhuji, Shaoxing, China
| | - Shu Zheng
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiding Chen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jiaojiao Zhou
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
15
|
Bonvini P, Rossi E, Zin A, Manicone M, Vidotto R, Facchinetti A, Tombolan L, Affinita MC, Santoro L, Zamarchi R, Bisogno G. Case Report: Circulating Tumor Cells as a Response Biomarker in ALK-Positive Metastatic Inflammatory Myofibroblastic Tumor. Front Pediatr 2021; 9:652583. [PMID: 33996693 PMCID: PMC8116882 DOI: 10.3389/fped.2021.652583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory myofibroblastic tumors (IMTs) are locally aggressive malignancies occurring at various sites. Surgery is the mainstay of treatment and prognosis is generally good. For children with unresectable or metastatic tumors, however, outcome is particularly severe, limited also by the lack of predictive biomarkers of therapy efficacy and disease progression. Blood represents a minimally invasive source of cancer biomarkers for real-time assessment of tumor growth, particularly when it involves the analysis of circulating tumor cells (CTC). As CTCs potentially represent disseminated disease, their detection in the blood correlates with the presence of metastatic lesions and may reflect tumor response to treatment. Herein, we present a case report of a 19-year-old boy with an ALK-positive IMT of the bladder, proximal osteolytic and multiple bilateral lung lesions, who received ALK inhibitor entrectinib postoperatively and underwent longitudinal CTC analysis during treatment. Antitumor activity of entrectinib was demonstrated and was accompanied by regression of lung lesions, elimination of CTCs from the blood and no development of relapses afterwards. Therapy continued without any clinical sign of progression and 24 months since the initiation of treatment the patient remains symptom-free and disease-free.
Collapse
Affiliation(s)
- Paolo Bonvini
- Institute of Pediatric Research, Fondazione Città della Speranza, Padova, Italy
| | - Elisabetta Rossi
- Department of Surgery, Oncology and Gastroenterology, Oncology Section, University of Padova, Padova, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Angelica Zin
- Institute of Pediatric Research, Fondazione Città della Speranza, Padova, Italy
| | | | | | - Antonella Facchinetti
- Department of Surgery, Oncology and Gastroenterology, Oncology Section, University of Padova, Padova, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Lucia Tombolan
- Hematology and Oncology Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Maria Carmen Affinita
- Hematology and Oncology Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Luisa Santoro
- Department of Medicine, Padova University Hospital, Padova, Italy
| | - Rita Zamarchi
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Gianni Bisogno
- Hematology and Oncology Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
| |
Collapse
|