1
|
Xu C, Du D, Han Z, Si H, Li W, Li L, Tang B. Separation and Analysis of Rare Tumor Cells in Various Body Fluids Based on Microfluidic Technology for Clinical Applications. Anal Chem 2025; 97:7567-7588. [PMID: 40186540 DOI: 10.1021/acs.analchem.4c06925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Affiliation(s)
- Chang Xu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Dexin Du
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Zhaojun Han
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Haibin Si
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Wei Li
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Lu Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
- Laoshan Laboratory, Qingdao 266237, P. R. China
| |
Collapse
|
2
|
Chong Y, Zhou H, Zhang P, Xue L, Du Q, Chong T, Wang Z. Establishing cM0 (i+) stage criteria in localized renal cell carcinoma based on postoperative circulating tumor cells monitoring. BMC Cancer 2025; 25:436. [PMID: 40069681 PMCID: PMC11895218 DOI: 10.1186/s12885-025-13815-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The diagnostic criteria for cM0 (i+) stage proposed by American Joint Committee on Cancer (AJCC) in renal cell carcinoma (RCC) still remains unclear. The present study aimed to establish and validate the criteria of cM0 (i+) stage based on postoperative circulating tumor cells (CTCs) monitoring in patients with localized renal cell carcinoma (LRCC). MATERIALS AND METHODS This study enrolled 204 patients with LRCC who received partial or radical nephrectomy from January 2015 to November 2021. Cases were randomly divided into test set and validation set. The correlation between clinicopathological features and CTCs counts were analyzed and prognostic variables were determined by Lasso regression. Receiver operating characteristic curve of the prognosis-related CTCs terms were plotted to determine their optimal cut-off value to establish the criteria of cM0 (i+) stage. Its clinical prognostic significance was explored by Kaplan-Meier analysis and Log-rank test. The above analysis was conducted by SPSS26.0 software and R Studio software. P < 0.05 was considered to be statistically significant. RESULTS A total of 204 patients were analyzed in this study.There were no significant differences in variables between the validation and test sets (P>0.05). Total CTCs, mesenchymal CTCs (MCTCs), and CTCs showing a progressive trend were selected as the diagnostic basis for the cM0 (i+) stage through correlation analysis and Lasso regression. The cM0 (i+) stage identified patients meeting the following criteria simultaneously: (1) total CTCs ≥ 6; (2) MCTCs ≥ 1; and (3) a demonstrated trend of progression in either total CTCs or MCTCs. In the validation group, Kaplan-Meier analysis showed that patients with cM0 (i+) stage had significantly shorter progression-free survival than the control group(P<0.05). The results of multivariate Cox regression analysis also showed cM0 (i+) was an independent risk factor for postoperative progression of LRCC patients [12.448 (1.874-82.666) P < 0.05]. Its 1-3 years' prediction discrimination is better than that of UISS score and SSIGN score, which was also verified in the validation set. CONCLUSION The study proposed a diagnostic criterion for M0 (i+) stage in LRCC based on postoperative CTCs monitoring. It was identified as an independent risk factor for postoperative progression and demonstrated potential advantages over the UISS and SSIGN scores in internal validation.
Collapse
Affiliation(s)
- Yue Chong
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
| | - Haibin Zhou
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
- Department of Urology, First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, 710077, China
| | - Peng Zhang
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
| | - Li Xue
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
| | - Qiao Du
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
- Baoji People's Hospital, Baoji, 721001, Shaanxi, China
| | - Tie Chong
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
| | - Zhenlong Wang
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
3
|
Zhang Z, Wang J, Wuethrich A, Trau M. Conventional techniques and emerging nanotechnologies for early detection of cancer metastasis via epithelial-mesenchymal transition monitoring. Natl Sci Rev 2025; 12:nwae452. [PMID: 39926198 PMCID: PMC11804803 DOI: 10.1093/nsr/nwae452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 02/11/2025] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a critical process for cancer to metastasize by promoting invasiveness and dissemination of cancer cells in the body. Understanding and tracking EMT could improve cancer therapy by intervening in metastasis. Current approaches for investigating and detecting the EMT process often utilize traditional molecular biology techniques like immunohistochemistry, mass spectrometry and sequencing. These approaches have provided valuable insights into understanding signaling pathways and identifying biomarkers. Liquid biopsy analysis using advanced nanotechnologies allows the longitudinal tracking of EMT in patients to become feasible. This review article offers a molecular overview of EMT, summarizes current EMT models used in cancer research, and reviews both traditional techniques and emerging nanotechnologies employed in recent EMT studies. Additionally, we discuss the limitations and prospects of applying nanotechnologies in EMT research. By evaluating this rapidly emerging field, we propose strategies to facilitate the clinical translation of nanotechnologies for early detection and monitoring of EMT.
Collapse
Affiliation(s)
- Zhen Zhang
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jing Wang
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou 350007, China
| | - Alain Wuethrich
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Matt Trau
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
4
|
Chen B, Liu J. Advancements in Hydrogel-Based Therapies for Ovarian Cancer: A Review. Cell Biochem Biophys 2025; 83:87-108. [PMID: 39190214 DOI: 10.1007/s12013-024-01483-7] [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] [Accepted: 08/09/2024] [Indexed: 08/28/2024]
Abstract
Ovarian cancer, the most deadly gynecologic malignancy, is often resistant to conventional antitumor therapy due to various factors such as severe side effects, unexpected recurrence, and significant tissue damage. The limitations of current treatments and the resistance of invasive tumor cells contribute to these challenges. Hydrogel therapy has recently emerged as a potential treatment option for ovarian cancer, offering advantages such as controllability, biocompatibility, high drug loading capacity, prolonged drug release, and responsiveness to specific stimuli. Hence, the utilization of biodegradable hydrogels as carriers for chemotherapeutic agents has emerged as a significant concern in the field. Injectable hydrogel-based drug delivery systems, in particular, have demonstrated superior efficacy compared to traditional systemic chemotherapy for cancer treatment. The pliability of hydrogel therapy allows for access to anatomical regions that may be challenging for surgical intervention. This review article examines recent advancements in the application of hydrogels for diagnosing and treating ovarian cancer, while also proposing a novel direction for the use of hydrogel technology in this context. The objective of this article is to offer a novel point of reference and serve as a source of inspiration for the advancement of more precise and individualized cancer therapies.
Collapse
Affiliation(s)
- Biqing Chen
- Harbin Medical University, Harbin, Heilongjiang, China.
| | - Jiaqi Liu
- Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Szerenyi D, Jarvas G, Guttman A. Multifaceted Approaches in Epithelial Cell Adhesion Molecule-Mediated Circulating Tumor Cell Isolation. Molecules 2025; 30:976. [PMID: 40076201 PMCID: PMC11901967 DOI: 10.3390/molecules30050976] [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: 11/30/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Circulating tumor cells (CTCs) are pivotal in cancer metastasis and serve as valuable biomarkers for diagnosis, prognosis, and treatment monitoring. Traditional CTC capture methods predominantly utilize the epithelial cell adhesion molecule (EpCAM) as a marker for isolation. However, the heterogeneity of these circulating cells and the epithelial-to-mesenchymal transition process (wherein epithelial cells acquire mesenchymal characteristics) limit the efficacy of EpCAM-based capture techniques. In this paper, we critically review the role of the EpCAM in CTC capture, explore the impact of epithelial-to-mesenchymal transition on EpCAM expression, and discuss alternative biomarkers and strategies to enhance CTC isolation. By evaluating the limitations of EpCAM-mediated capture and the challenges posed by epithelial-to-mesenchymal transition, we aim to provide insights into the development of more comprehensive liquid biopsy approaches for cancer management.
Collapse
Affiliation(s)
- Dora Szerenyi
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, 8200 Veszprem, Hungary;
| | - Gabor Jarvas
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, 8200 Veszprem, Hungary;
- CAPTEC Medical Ltd., 8200 Veszprem, Hungary
| | - Andras Guttman
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, 8200 Veszprem, Hungary;
- CAPTEC Medical Ltd., 8200 Veszprem, Hungary
- Horváth Csaba Memorial Laboratory of Bioseparation Sciences, Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
6
|
Dai Y, He J, Zhou Y, Yu Y, Hui H, Guo L, Yin H. Constructing a highly sensitive duplex immunoassay using AuNPs and AgNPs as nanolabels for investigating the epithelial-mesenchymal transition occurring on circulating tumor cells with lung cancer patients. Biosens Bioelectron 2025; 270:116947. [PMID: 39561553 DOI: 10.1016/j.bios.2024.116947] [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: 09/06/2024] [Revised: 11/01/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
Transformation of epithelial to mesenchymal (EMT) is an important event in the process of tumor initiation, invasion and metastasis. Circulating tumor cells (CTCs) are one kind of important markers in the field of liquid tumor biopsy, whose number and phenotype represent the occurrence and progression of tumors. Therefore, it is our interest to investigate the epithelial mesenchymal transition process occurring on the surface of CTCs. Herein in this work, two proteins of E-cadherin (E-cad) and N-cadherin (N-cad) were selected as representative proteins of EMT process. To achieve simultaneous analysis of E-cad and N-cad on the surface of rare CTCs, we designed a duplex and portable immunosensor using AuNPs and AgNPs as nanolabels to amplify the immunoreaction signals. The dual channel immunosensor not only exhibited good electrochemical responses for recombinant E-cad and N-cad as low as 0.1 ng/mL and 0.05 ng/mL, respectively, but also showed good linear correlations with different numbers of phenotypic CTCs (10-500 cells/10 μL). The above strategy was further employed to inspect the occurrence of EMT on CTCs surface, which displayed a high consistence with other molecular biological characterizations. Finally, this immunoassay was successfully applied to inspect the correlations of numbers, phenotype of CTCs, as well as E-cad and N-cad expressions on these CTCs in bloods of NSCLC patients with disease stage.
Collapse
Affiliation(s)
- Yunuo Dai
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Jie He
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Yun Zhou
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, PR China
| | - Hui Hui
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Lin Guo
- Department of Radiotherapy Central Hospital, Affiliated Xuzhou Clinical College of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, PR China
| | - Haitao Yin
- The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, 269 University Road, Xuzhou, 221002, Jiangsu, PR China.
| |
Collapse
|
7
|
Rzhevskiy AS, Sagitova GR, Karashaeva TA, Morozov AO, Fatyanova AS, Kazantseva VV, Joosse SA, Zvyagin AV, Warkini ME. A comprehensive review and meta-analysis of CTC isolation methods in breast cancer. Crit Rev Oncol Hematol 2025; 206:104579. [PMID: 39615710 DOI: 10.1016/j.critrevonc.2024.104579] [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: 09/17/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/08/2024] Open
Abstract
The application of circulating tumor cells (CTCs) as diagnostic and prognostic markers in oncology is gaining increasing importance in clinical practice. Currently, various methods exist for detecting CTCs in patients' biological fluids. This systematic review aimed to compare the efficacy of different techniques for isolating and detecting CTCs from blood, against the FDA-cleared CellSearch® technology, in breast cancer patients. We performed a systematic literature search using two databases (PubMed and the Cochrane Library) with the following terms: ("Circulating tumor cells" OR CTC) AND "breast cancer", covering the period from 2004 to April 2023. The primary outcome measured was the sensitivity, specificity, and overall accuracy of various CTC enrichment methods in comparison with the CellSearch® System. Secondary outcomes included the prognostic value of CTCs in evaluating response to treatment based on survival rates. Generally, a high level of agreement between CellSearch and other methods was observed in isolating CTCs from patients' blood with both metastatic and early-stage disease. Studies asserting the superiority of new methods over CellSearch frequently used clinically unvalidated cut-off thresholds for their patient cohorts. Additionally, these studies sometimes included different nonoverlapping patient cohorts and lacked a standardized chemotherapy treatment protocol, which could affect the quantitative changes in CTC. It is evident that methods simultaneously composed of physical and immunomagnetic approaches for CTC isolation significantly surpass CellSearch, which relies solely on the expression of specific markers on the CTCs' surface. The count of CTCs has been established as a predictive marker in terms of clinically important parameters namely progression-free survival (PFS) and overall survival (OS). The CTC-count value was significantly correlated with PFS and OS rates.
Collapse
Affiliation(s)
- Alexey S Rzhevskiy
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia; Faculty of Computer Science, National Research University Higher School of Economics, Moscow 101000, Russia
| | - Guzel R Sagitova
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Tamilla A Karashaeva
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Andrey O Morozov
- Institute for Urology and Reproductive Health, Sechenov University, Moscow 119991, Russia
| | - Anastasia S Fatyanova
- Department of Oncology, Radiotherapy and Reconstructive Surgery, Institution of Clinical Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Vlada V Kazantseva
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany; Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Andrei V Zvyagin
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW 2109, Australia; Research Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354340, Russia.
| | - Majid Ebrahimi Warkini
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow 119991, Russia; School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
8
|
Cao L, Duan Q, Zhu Z, Xu X, Liu J, Li B. Liquid biopsy technologies: innovations and future directions in breast cancer biomarker detection. Biomed Microdevices 2025; 27:4. [PMID: 39849252 DOI: 10.1007/s10544-025-00734-5] [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] [Accepted: 01/07/2025] [Indexed: 01/25/2025]
Abstract
Globally, breast cancer is the most frequent type of cancer, and its early diagnosis and screening can significantly improve the probability of survival and quality of life of those affected. Liquid biopsy-based targets such as circulating tumor cells, circulating tumor DNA, and exosomes have been instrumental in the early discovery of cancer, and have been found to be effective in stage therapy, recurrence monitoring, and drug selection. Biosensors based on these target related biomarkers convert the tested substances into quantifiable signals such as electrical and optical signals through signal transduction, which has the advantages of high sensitivity, simple operation, and low invasiveness. This review provides an overview of the latest progress of liquid biopsy biomarkers in the diagnosis, prognosis and treatment of breast cancer, compares the application and advantages of different biosensors based on these biomarkers in the diagnosis of breast cancer, and analyzes the limitations and solutions of biosensor based methods.
Collapse
Affiliation(s)
- Linhong Cao
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Qingli Duan
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Zixin Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Xuejing Xu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China.
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China.
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China.
| | - Baolin Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China.
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China.
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China.
| |
Collapse
|
9
|
Wei X, Xiong H, Zhou Y, Chen X, Yang W. Tracking epithelial-mesenchymal transition in breast cancer cells based on a multiplex electrochemical immunosensor. Biosens Bioelectron 2024; 258:116372. [PMID: 38735081 DOI: 10.1016/j.bios.2024.116372] [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: 01/31/2024] [Revised: 04/15/2024] [Accepted: 04/29/2024] [Indexed: 05/14/2024]
Abstract
Epithelial-mesenchymal transition (EMT) promotes tumor cell infiltration and metastasis. Tracking the progression of EMT could potentially indicate early cancer metastasis. A key characteristic of EMT is the dynamic alteration in the molecular levels of E-cadherin and N-cadherin. Traditional assays have limited sensitivity and multiplexing capabilities, relying heavily on cell lysis. Here, we developed a multiplex electrochemical biosensor to concurrently track the upregulation of N-cadherin expression and reduction of E-cadherin in breast cancer cells undergoing EMT. Small-sized gold nanoparticles (Au NPs) tagged with redox probes (thionin or amino ferrocene) and bound to two types of antibodies were used as distinguishable signal tags. These tags specifically recognized E-cadherin and N-cadherin proteins on the tumor cell surface without cross-reactivity. The diphenylalanine dipeptide (FF)/chitosan (CS)/Au NPs (FF-CS@Au) composites with high surface area and good biocompatibility were used as the sensing platforms for efficiently fixing cells and recording the dynamic changes in electrochemical signals of surface proteins. The electrochemical immunosensor allowed for simultaneous monitoring of E- and N-cadherins on breast cancer cell surfaces in a single run, enabling tracking of the EMT dynamic process for up to 60 h. Furthermore, the electrochemical detection results are consistent with Western blot analysis, confirming the reliability of the methodology. This present work provides an effective, rapid, and low-cost approach for tracking the EMT process, as well as valuable insights into early tumor metastasis.
Collapse
Affiliation(s)
- Xue Wei
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, PR China
| | - Hanzhi Xiong
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, PR China
| | - Yunfan Zhou
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, PR China
| | - Xu Chen
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, PR China.
| | - Wensheng Yang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, PR China
| |
Collapse
|
10
|
Wang X, Du Y, Jing W, Cao C, Wu X, Yang K, Zhu L. Fluorescent identification of immunomagnetically captured CTCs using triplex-aptamer-targeted dendritic SiO 2@Fe 3O 4 nanocomposite. Mikrochim Acta 2024; 191:424. [PMID: 38922365 DOI: 10.1007/s00604-024-06504-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: 05/09/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
The enumeration of circulating tumor cells (CTCs) in peripheral blood plays a crucial role in the early diagnosis, recurrence monitoring, and prognosis assessment of cancer patients. There is a compelling need to develop an efficient technique for the capture and identification of these rare CTCs. However, the exclusive reliance on a single criterion, such as the epithelial cell adhesion molecule (EpCAM) antibody or aptamer, for the specific recognition of epithelial CTCs is not universally suitable for clinical applications, as it usually falls short in identifying EpCAM-negative CTCs. To address this limitation, we propose a straightforward and cost-effective method involving triplex fluorescently labelled aptamers (FAM-EpCAM, Cy5-PTK7, and Texas Red-CSV) to modify Fe3O4-loaded dendritic SiO2 nanocomposite (dmSiO2@Fe3O4/Apt). This multi-recognition-based strategy not only enhanced the efficiency in capturing heterogeneous CTCs, but also facilitated the rapid and accurate identification of CTCs. The capture efficiency of heterogenous CTCs reached up to 93.33%, with a detection limit as low as 5 cells/mL. Notably, the developed dmSiO2@Fe3O4/Apt nanoprobe enabled the swift identification of captured cells in just 30 min, relying solely on the fluorescently modified aptamers, which reduced the identification time by approximately 90% compared with the conventional immunocytochemistry (ICC) technique. Finally, these nanoprobe characteristics were validated using blood samples from patients with various types of cancers.
Collapse
Affiliation(s)
- Xinwen Wang
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Yu Du
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Weijun Jing
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, P. R. China
| | - Changchun Cao
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Xiaoli Wu
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Kangqun Yang
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Liang Zhu
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China.
| |
Collapse
|
11
|
Wiegmans AP, Ivanova E, Naei VY, Monkman J, Fletcher J, Mullally W, Warkiani ME, O'Byrne K, Kulasinghe A. Poor patient outcome correlates with active engulfment of cytokeratin positive CTCs within cancer-associated monocyte population in lung cancer. Clin Exp Metastasis 2024; 41:219-228. [PMID: 38416302 PMCID: PMC11213738 DOI: 10.1007/s10585-024-10270-w] [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: 10/15/2023] [Accepted: 01/18/2024] [Indexed: 02/29/2024]
Abstract
High rates of mortality in non-small cell lung cancer lung cancer is due to inherent and acquired resistance to systemic therapies and subsequent metastatic burden. Metastasis is supported by suppression of the immune system at secondary organs and within the circulation. Modulation of the immune system is now being exploited as a therapeutic target with immune checkpoint inhibitors. The tracking of therapeutic efficacy in a real-time can be achieved with liquid biopsy, and evaluation of circulating tumour cells and the associated immune cells. A stable liquid biopsy biomarker for non-small cell lung cancer lung cancer has yet to be approved for clinical use. We performed a cross-sectional single-site study, and collected liquid biopsies from patients diagnosed with early, locally advanced, or metastatic lung cancer, undergoing surgery, or systemic therapy (chemotherapy/checkpoint inhibitors). Evaluation of overall circulating tumour cell counts, or cluster counts did not correlate with patient outcome. Interestingly, the numbers of Pan cytokeratin positive circulating tumour cells engulfed by tumour associated monocytes correlated strongly with patient outcome independent of circulating tumour cell counts and the use of checkpoint inhibitors. We suggest that Pan cytokeratin staining within monocytes is an important indicator of tumour-associated inflammation post-therapy and an effective biomarker with strong prognostic capability for patient outcome.
Collapse
Affiliation(s)
- A P Wiegmans
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology, Woolloongabba, QLD, 4102, Australia
- Princess Alexandra Hospital, Oncology, Woolloongabba, QLD, 4102, Australia
| | - E Ivanova
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology, Woolloongabba, QLD, 4102, Australia
| | - V Y Naei
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
- Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - J Monkman
- Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - J Fletcher
- Princess Alexandra Hospital, Oncology, Woolloongabba, QLD, 4102, Australia
| | - W Mullally
- Princess Alexandra Hospital, Oncology, Woolloongabba, QLD, 4102, Australia
| | - M E Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| | - K O'Byrne
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology, Woolloongabba, QLD, 4102, Australia
- Princess Alexandra Hospital, Oncology, Woolloongabba, QLD, 4102, Australia
| | - A Kulasinghe
- Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
12
|
Li M, Shi J, Zhang Y, Cui S, Zhang L, Shen Q. ECL cytosensor for sensitive and label-free detection of circulating tumor cells based on hierarchical flower-like gold microstructures. Anal Chim Acta 2024; 1303:342505. [PMID: 38609273 DOI: 10.1016/j.aca.2024.342505] [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: 02/02/2024] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024]
Abstract
The development of sensitive and efficient cell sensing strategies to detect circulating tumor cells (CTCs) in peripheral blood is crucial for the early diagnosis and prognostic assessment of cancer clinical treatment. Herein, an array of hierarchical flower-like gold microstructures (HFGMs) with anisotropic nanotips was synthesized by a simple electrodeposition method and used as a capture substrate to construct an ECL cytosensor based on the specific recognition of target cells by aptamers. The complex topography of the HFGMs array not only catalyzed the enhancement of ECL signals, but also induced the cells to generate more filopodia, improving the capture efficiency and shortening the capture time. The effect of topographic roughness on cell growth and adhesion propensity was also investigated, while the cell capture efficiency was proposed to be an important indicator affecting the accuracy of the ECL cytosensor. In addition, the capture of cells on the electrode surface increased the steric hindrance, which caused ECL signal changes in the Ru(bpy)32+ and TPrA system, realizing the quantitative detection of MCF-7 cells. The detection range of the sensor was from 102 to 106 cells mL-1 and the detection limit was 18 cells mL-1. The proposed detection method avoids the process of separation, labeling and counting, which has great potential for sensitive detection in clinical applications.
Collapse
Affiliation(s)
- Meixing Li
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| | - Jiawei Shi
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Yuhang Zhang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Shuaihui Cui
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Lei Zhang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Qingming Shen
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| |
Collapse
|
13
|
Chen K, Wang Z. A Micropillar Array Based Microfluidic Device for Rare Cell Detection and Single-Cell Proteomics. Methods Protoc 2023; 6:80. [PMID: 37736963 PMCID: PMC10514859 DOI: 10.3390/mps6050080] [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: 08/02/2023] [Revised: 08/25/2023] [Accepted: 08/31/2023] [Indexed: 09/23/2023] Open
Abstract
Advancements in single-cell-related technologies have opened new possibilities for analyzing rare cells, such as circulating tumor cells (CTCs) and rare immune cells. Among these techniques, single-cell proteomics, particularly single-cell mass spectrometric analysis (scMS), has gained significant attention due to its ability to directly measure transcripts without the need for specific reagents. However, the success of single-cell proteomics relies heavily on efficient sample preparation, as protein loss in low-concentration samples can profoundly impact the analysis. To address this challenge, an effective handling system for rare cells is essential for single-cell proteomic analysis. Herein, we propose a microfluidics-based method that offers highly efficient isolation, detection, and collection of rare cells (e.g., CTCs). The detailed fabrication process of the micropillar array-based microfluidic device is presented, along with its application for CTC isolation, identification, and collection for subsequent proteomic analysis.
Collapse
Affiliation(s)
- Kangfu Chen
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA;
- Chan Zuckerberg Biohub Chicago, Chicago, IL 60607, USA
| | - Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA;
- Chan Zuckerberg Biohub Chicago, Chicago, IL 60607, USA
| |
Collapse
|
14
|
Gu Q, Wu J, Xu H, Cao H, Zhang J, Jing C, Wang Z, Du M, Ma R, Feng J. CLASRP oncogene as a novel target for colorectal cancer. Funct Integr Genomics 2023; 23:290. [PMID: 37658940 PMCID: PMC10474993 DOI: 10.1007/s10142-023-01208-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 09/05/2023]
Abstract
Clk4-associated serine/arginine-rich protein (CLASRP), an alternative splicing regulator, may be involved in the development and progression of cancer by regulating the activity of the CDC-like kinase (Clk) family. This study explored the biological function of CLASRP in colorectal cancer (CRC). The expression of CLASRP, which is associated with clinicopathological features, was analysed in CRC tissues and paired noncancer tissues by RT-PCR. The roles of CLASRP were investigated in CRC cells transfected with plasmids or shRNA through proliferation, migration and invasion assays in vitro and a xenograft model in vivo. Apoptosis was analysed using CLASRP-overexpressing CRC cells by western blotting. Clk inhibitors were used to perform functional research on CLASRP in CLASRP-overexpressing CRC cells. CLASRP was significantly upregulated in CRC cell lines, while high CLASRP expression was correlated with metastasis in CRC patients. Functionally, overexpression of CLASRP significantly promoted the proliferation, migration and invasion of CRC cells in vitro and tumour growth in vivo. Mechanistically, the proliferation, migration and invasion of CLASRP-overexpressing CRC cells were inhibited by Clk inhibitors, accompanied by low expression of CLASRP at the gene and protein levels. Clk inhibitors induced apoptosis of CLASRP-overexpressing CRC cells, resulting in direct blockade of cell growth. The expression levels of cleaved caspase 3 and cleaved caspase 8 were increased in CLASRP-overexpressing CRC cells treated with Clk inhibitors. CLASRP might serve as a promotional oncogene in CRC cells and be suppressed by Clk inhibitors through activation of caspase pathways.
Collapse
Affiliation(s)
- Quan Gu
- Nanjing Medical University Affiliated Cancer Hospital and Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 BaiZiTing Road, Nanjing, Jiangsu, 210000, People's Republic of China
| | - Jianzhong Wu
- Nanjing Medical University Affiliated Cancer Hospital and Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 BaiZiTing Road, Nanjing, Jiangsu, 210000, People's Republic of China
| | - Heng Xu
- Jiangsu Provincial Institute of Materia Medica, Nanjing Tech University, Nanjing, Jiangsu, China
| | - Haixia Cao
- Nanjing Medical University Affiliated Cancer Hospital and Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 BaiZiTing Road, Nanjing, Jiangsu, 210000, People's Republic of China
| | - Junying Zhang
- Nanjing Medical University Affiliated Cancer Hospital and Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 BaiZiTing Road, Nanjing, Jiangsu, 210000, People's Republic of China
| | - Changwen Jing
- Nanjing Medical University Affiliated Cancer Hospital and Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 BaiZiTing Road, Nanjing, Jiangsu, 210000, People's Republic of China
| | - Zhuo Wang
- Nanjing Medical University Affiliated Cancer Hospital and Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 BaiZiTing Road, Nanjing, Jiangsu, 210000, People's Republic of China
| | - Mengjie Du
- Jiangsu Provincial Institute of Materia Medica, Nanjing Tech University, Nanjing, Jiangsu, China
| | - Rong Ma
- Nanjing Medical University Affiliated Cancer Hospital and Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 BaiZiTing Road, Nanjing, Jiangsu, 210000, People's Republic of China.
| | - Jifeng Feng
- Nanjing Medical University Affiliated Cancer Hospital and Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 BaiZiTing Road, Nanjing, Jiangsu, 210000, People's Republic of China.
| |
Collapse
|
15
|
Wang J, Meng X, Yu M, Li X, Chen Z, Wang R, Fang J. A novel microfluidic system for enrichment of functional circulating tumor cells in cancer patient blood samples by combining cell size and invasiveness. Biosens Bioelectron 2023; 227:115159. [PMID: 36841114 DOI: 10.1016/j.bios.2023.115159] [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: 11/01/2022] [Revised: 01/29/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
A highly invasive subpopulation of circulating tumor cells (CTCs) may constitute seeds for metastases, which are therefore considered functional CTCs. However, there are few effective strategies to detect CTCs based on invasive phenotypes. Herein, we focused on functional CTCs with high invasiveness and designed an integrated microfluidic system to differentiate the invasive potential of CTCs for more accurate metastasis prediction. By combining size-based enrichment and invasiveness-based analysis, the system managed to continuously remove most hemocytes by 8 μm gaps and analyze the invasiveness of the enriched CTCs by Matrigel loading. In addition to a device, a single pump and a Petri dish were included to provide an FBS gradient for driving cell invasion and maintain a long-term cell culture. The system successfully identified functional CTCs derived from different types of cancer patients, including colorectal, kidney and bladder cancer patients, using whole blood without any sample pretreatment process. Within 28 cases of colorectal cancer patients, functional CTCs were detected in 61.54% of patients with metastases, along with stronger invasiveness evaluated by migration/invasion distance than those from patients without metastases (P < 0.05). Furthermore, one bladder cancer patient was diagnosed with recurrence six months after detection, indicating the excellent value for cancer metastases prediction. In addition, great phenotypic heterogeneity of CTCs was also observed at the single-cell level, including invasion, proliferation and dormancy, which provided an effective strategy for metastasis prediction based on CTC function as a single cell.
Collapse
Affiliation(s)
- Jie Wang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Xianmeng Meng
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Min Yu
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Xin Li
- Department of Anesthesiology, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, PR China
| | - Zhezhou Chen
- Department of Emergency Laboratory, The First People's Hospital of Shenyang, No. 67 Qingquan Road, Dadong District, Shenyang, Liaoning Province, 110041, PR China
| | - Rui Wang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Jin Fang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China.
| |
Collapse
|
16
|
Balamurugan K, Poria DK, Sehareen SW, Krishnamurthy S, Tang W, McKennett L, Padmanaban V, Czarra K, Ewald AJ, Ueno NT, Ambs S, Sharan S, Sterneck E. Stabilization of E-cadherin adhesions by COX-2/GSK3β signaling is a targetable pathway in metastatic breast cancer. JCI Insight 2023; 8:156057. [PMID: 36757813 PMCID: PMC10070121 DOI: 10.1172/jci.insight.156057] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/07/2023] [Indexed: 02/10/2023] Open
Abstract
Metastatic progression of epithelial cancers can be associated with epithelial-mesenchymal transition (EMT) including transcriptional inhibition of E-cadherin (CDH1) expression. Recently, EM plasticity (EMP) and E-cadherin-mediated, cluster-based metastasis and treatment resistance have become more appreciated. However, the mechanisms that maintain E-cadherin expression in this context are less understood. Through studies of inflammatory breast cancer (IBC) and a 3D tumor cell "emboli" culture paradigm, we discovered that cyclooxygenase 2 (COX-2; PTGS2), a target gene of C/EBPδ (CEBPD), or its metabolite prostaglandin E2 (PGE2) promotes protein stability of E-cadherin, β-catenin, and p120 catenin through inhibition of GSK3β. The COX-2 inhibitor celecoxib downregulated E-cadherin complex proteins and caused cell death. Coexpression of E-cadherin and COX-2 was seen in breast cancer tissues from patients with poor outcome and, along with inhibitory GSK3β phosphorylation, in patient-derived xenografts (PDX) including triple negative breast cancer (TNBC).Celecoxib alone decreased E-cadherin protein expression within xenograft tumors, though CDH1 mRNA levels increased, and reduced circulating tumor cell (CTC) clusters. In combination with paclitaxel, celecoxib attenuated or regressed lung metastases. This study has uncovered a mechanism by which metastatic breast cancer cells can maintain E-cadherin-mediated cell-to-cell adhesions and cell survival, suggesting that some patients with COX-2+/E-cadherin+ breast cancer may benefit from targeting of the PGE2 signaling pathway.
Collapse
Affiliation(s)
- Kuppusamy Balamurugan
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Dipak K Poria
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Saadiya W Sehareen
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Savitri Krishnamurthy
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Tang
- Laboratory of Human Carcinogenesis, CCR, NCI, Bethesda, Maryland, USA
| | - Lois McKennett
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Veena Padmanaban
- Departments of Cell Biology and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kelli Czarra
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Andrew J Ewald
- Departments of Cell Biology and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Naoto T Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, CCR, NCI, Bethesda, Maryland, USA
| | - Shikha Sharan
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Esta Sterneck
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| |
Collapse
|
17
|
Niu ZS, Wang WH, Niu XJ. Recent progress in molecular mechanisms of postoperative recurrence and metastasis of hepatocellular carcinoma. World J Gastroenterol 2022; 28:6433-6477. [PMID: 36569275 PMCID: PMC9782839 DOI: 10.3748/wjg.v28.i46.6433] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/31/2022] [Accepted: 11/21/2022] [Indexed: 12/08/2022] Open
Abstract
Hepatectomy is currently considered the most effective option for treating patients with early and intermediate hepatocellular carcinoma (HCC). Unfortunately, the postoperative prognosis of patients with HCC remains unsatisfactory, predominantly because of high postoperative metastasis and recurrence rates. Therefore, research on the molecular mechanisms of postoperative HCC metastasis and recurrence will help develop effective intervention measures to prevent or delay HCC metastasis and recurrence and to improve the long-term survival of HCC patients. Herein, we review the latest research progress on the molecular mechanisms underlying postoperative HCC metastasis and recurrence to lay a foundation for improving the understanding of HCC metastasis and recurrence and for developing more precise prevention and intervention strategies.
Collapse
Affiliation(s)
- Zhao-Shan Niu
- Laboratory of Micromorphology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Wen-Hong Wang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Xiao-Jun Niu
- Department of Internal Medicine, Qingdao Shibei District People's Hospital, Qingdao 266033, Shandong Province, China
| |
Collapse
|
18
|
Cohen EN, Jayachandran G, Moore RG, Cristofanilli M, Lang JE, Khoury JD, Press MF, Kim KK, Khazan N, Zhang Q, Zhang Y, Kaur P, Guzman R, Miller MC, Reuben JM, Ueno NT. A Multi-Center Clinical Study to Harvest and Characterize Circulating Tumor Cells from Patients with Metastatic Breast Cancer Using the Parsortix ® PC1 System. Cancers (Basel) 2022; 14:5238. [PMID: 36358657 PMCID: PMC9656921 DOI: 10.3390/cancers14215238] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 08/22/2023] Open
Abstract
Circulating tumor cells (CTCs) captured from the blood of cancer patients may serve as a surrogate source of tumor material that can be obtained via a venipuncture (also known as a liquid biopsy) and used to better understand tumor characteristics. However, the only FDA-cleared CTC assay has been limited to the enumeration of surface marker-defined cells and not further characterization of the CTCs. In this study, we tested the ability of a semi-automated device capable of capturing and harvesting CTCs from peripheral blood based on cell size and deformability, agnostic of cell-surface markers (the Parsortix® PC1 System), to yield CTCs for evaluation by downstream techniques commonly available in clinical laboratories. The data generated from this study were used to support a De Novo request (DEN200062) for the classification of this device, which the FDA recently granted. As part of a multicenter clinical trial, peripheral blood samples from 216 patients with metastatic breast cancer (MBC) and 205 healthy volunteers were subjected to CTC enrichment. A board-certified pathologist enumerated the CTCs from each participant by cytologic evaluation of Wright-Giemsa-stained slides. As proof of principle, cells harvested from a concurrent parallel sample provided by each participant were evaluated using one of three additional evaluation techniques: molecular profiling by qRT-PCR, RNA sequencing, or cytogenetic analysis of HER2 amplification by FISH. The study demonstrated that the Parsortix® PC1 System can effectively capture and harvest CTCs from the peripheral blood of MBC patients and that the harvested cells can be evaluated using orthogonal methodologies such as gene expression and/or Fluorescence In Situ Hybridization (FISH).
Collapse
Affiliation(s)
- Evan N. Cohen
- Department of Hematopathology Research, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gitanjali Jayachandran
- Department of Hematopathology Research, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Richard G. Moore
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14620, USA
| | - Massimo Cristofanilli
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Julie E. Lang
- USC Breast Cancer Program, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Joseph D. Khoury
- Department of Pathology, Breast Cancer Analysis Laboratory, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Michael F. Press
- Department of Pathology, Breast Cancer Analysis Laboratory, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Kyu Kwang Kim
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14620, USA
| | - Negar Khazan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14620, USA
| | - Qiang Zhang
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Youbin Zhang
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pushpinder Kaur
- USC Breast Cancer Program, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Roberta Guzman
- Department of Pathology, Breast Cancer Analysis Laboratory, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Michael C. Miller
- ANGLE Clinical Studies, ANGLE Europe Limited, Guildford, Surrey GU2 7AF, UK
| | - James M. Reuben
- Department of Hematopathology Research, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoto T. Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
19
|
Wang J, Koo KM, Trau M. Tetraplex Immunophenotyping of Cell Surface Proteomes via Synthesized Plasmonic Nanotags and Portable Raman Spectroscopy. Anal Chem 2022; 94:14906-14916. [PMID: 36256869 DOI: 10.1021/acs.analchem.2c02262] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Multiplex immunophenotyping of cell surface proteomes is useful for cell characterization as well as providing valuable information on a patient's physiological or pathological state. Current methods for multiplex immunophenotyping of cell surface proteomes still have associated technical pitfalls in terms of limited multiplexing capability, challenging result interpretation, and large equipment footprint limited to use in a laboratory setting. Herein, we presented a portable surface-enhanced Raman spectroscopy (SERS) assay for multiplex cell surface immunophenotyping. We synthesized and functionalized customizable SERS nanotags for cell labeling and subsequent signal measurement using a portable Raman spectrometer. We extensively evaluated and validated the analytical assay performance of the portable SERS immunophenotyping assay in two different cellular models (red blood cells and breast cancer cells). In terms of analytical specificity, the cell surface immunophenotyping of both red blood cells and breast cancer cells correlated well with flow cytometry. The portable SERS immunophenotyping assay also has comparable analytical repeatability to flow cytometry, with coefficient of variation values of 21.89-23.33% and 6.88-17.32% for detecting red blood cells and breast cancer cells, respectively. The analytical detection limits were 77 cells/mL for red blood cells and 1-17 cells/mL for breast cancer cells. As an alternative to flow cytometry, the portable SERS immunophenotyping assay demonstrated excellent analytical assay performance and possessed advantages such as quick sample-to-result turnaround time, multiplexing capability, and small equipment footprint.
Collapse
Affiliation(s)
- Jing Wang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350007, P. R. China.,Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kevin M Koo
- XING Applied Research & Assay Development (XARAD) Division, XING Technologies Pty Ltd, Sinnamon Park, QLD 4073, Australia.,The University of Queensland Centre for Clinical Research (UQCCR), Herston, QLD 4029, Australia
| | - Matt Trau
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
20
|
Hashemi M, Moosavi MS, Abed HM, Dehghani M, Aalipour M, Heydari EA, Behroozaghdam M, Entezari M, Salimimoghadam S, Gunduz ES, Taheriazam A, Mirzaei S, Samarghandian S. Long non-coding RNA (lncRNA) H19 in human cancer: From proliferation and metastasis to therapy. Pharmacol Res 2022; 184:106418. [PMID: 36038043 DOI: 10.1016/j.phrs.2022.106418] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 02/07/2023]
Abstract
Initiation and development of cancer depend on multiple factors that mutations in genes and epigenetic level can be considered as important drivers. Epigenetic factors include a large family of members and understanding their function in cancer has been a hot topic. LncRNAs are RNA molecules with no capacity in synthesis of proteins, and they have regulatory functions in cells. LncRNAs are localized in nucleus and cytoplasm, and their abnormal expression is related to development of tumor. This manuscript emphasizes on the role of lncRNA H19 in various cancers and its association with tumor hallmarks. The function of lncRNA H19 in most tumors is oncogenic and therefore, tumor cells increase its expression for promoting their progression. LncRNA H19 contributes to enhancing growth and cell cycle of cancers and by EMT induction, it is able to elevate metastasis rate. Silencing H19 induces apoptotic cell death and disrupts progression of tumors. LncRNA H19 triggers chemo- and radio-resistance in cancer cells. miRNAs are dually upregulated/down-regulated by lncRNA H19 in increasing tumor progression. Anti-cancer agents reduce lncRNA H19 in impairing tumor progression and increasing therapy sensitivity. A number of downstream targets and molecular pathways for lncRNA H19 have been detected in cancers including miRNAs, RUNX1, STAT3, β-catenin, Akt2 and FOXM1. Clinical studies have revealed potential of lncRNA H19 as biomarker and its association with poor prognosis. LncRNA H19 can be transferred to cancer cells via exosomes in enhancing their progression.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Marzieh Sadat Moosavi
- Department of Biochemistry, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Hedyeh Maghareh Abed
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Dehghani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Masoumeh Aalipour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Ali Heydari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mitra Behroozaghdam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Emine Selda Gunduz
- Vocational School of Health Services, Department of First and Emergency Aid, Akdeniz University, Antalya, Turkey.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
21
|
Novel Circulating Tumour Cell-Related Risk Model Indicates Prognosis and Immune Infiltration in Lung Adenocarcinoma. J Immunol Res 2022; 2022:6521290. [PMID: 35677538 PMCID: PMC9168189 DOI: 10.1155/2022/6521290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/03/2022] [Indexed: 01/07/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most common histological subtype of lung cancer (LC) and one of the leading causes of cancer-related death worldwide. LUAD has a low survival rate owing to tumour invasion and metastasis. Circulating tumour cells (CTCs) are precursors of distant metastasis, which are considered to adopt the characteristics of cancer stem cells (CSCs). Therefore, analysing the risk factors of LUAD from the perspective of CTCs may provide novel insights into the metastatic mechanisms and may help to develop diagnostic and therapeutic strategies. Methods A total of 447 patients from TCGA dataset were included in the training cohort, and 460 patients from the GEO dataset were included in the validation cohort. A CTC-related-gene risk model was constructed using LASSO penalty–Cox analysis, and its predictive value was further verified. Functional enrichment analysis was performed on differentially expressed genes (DEGs), followed by immune correlation analysis based on the results. In addition, western blot, CCK-8 and colony formation assays were used to validate the biological function of RAB26 in LUAD. Results A novel in-silico CTC-related-gene risk model, named the CTCR model, was constructed, which successfully divided patients into the high- and low-risk groups. The prognosis of the high-risk group was worse than that of the low-risk group. ROC analysis revealed that the risk model outperformed traditional clinical markers in predicting the prognosis of patients with LUAD. Further study demonstrated that the identified DEGs were significantly enriched in immune-related pathways. The immune score of the low-risk group was higher than that of the high-risk group. In addition, RAB26 was found to promote the proliferation of LUAD. Conclusion A prognostic risk model based on CTC-related genes was successfully constructed, and the relationship between DEGs and tumour immunity was analysed. In addition, RAB26 was found to promote the proliferation of LUAD cells.
Collapse
|
22
|
Folic Acid-Modified Fluorescent-Magnetic Nanoparticles for Efficient Isolation and Identification of Circulating Tumor Cells in Ovarian Cancer. BIOSENSORS 2022; 12:bios12030184. [PMID: 35323454 PMCID: PMC8946694 DOI: 10.3390/bios12030184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/16/2022]
Abstract
Ovarian cancer (OC) is a lethal disease occurring in women worldwide. Due to the lack of obvious clinical symptoms and sensitivity biomarkers, OC patients are often diagnosed in advanced stages and suffer a poor prognosis. Circulating tumor cells (CTCs), released from tumor sites into the peripheral blood, have been recognized as promising biomarkers in cancer prognosis, treatment monitoring, and metastasis diagnosis. However, the number of CTCs in peripheral blood is low, and it is a technical challenge to isolate, enrich, and identify CTCs from the blood samples of patients. This work develops a simple, effective, and inexpensive strategy to capture and identify CTCs from OC blood samples using the folic acid (FA) and antifouling-hydrogel-modified fluorescent-magnetic nanoparticles. The hydrogel showed a good antifouling property against peripheral blood mononuclear cells (PBMCs). The FA was coupled to the hydrogel surface as the targeting molecule for the CTC isolation, held a good capture efficiency for SK-OV-3 cells (95.58%), and successfully isolated 2–12 CTCs from 10 OC patients’ blood samples. The FA-modified fluorescent-magnetic nanoparticles were successfully used for the capture and direct identification of CTCs from the blood samples of OC patients.
Collapse
|