1
|
Yin P, Peng Z, Wang Q, Duan Y, Hu B, Lin Q. A rapid dual-mode SERS/FL cytosensor assisted via DNA Walker-based plasmonic nanostructures. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 327:125416. [PMID: 39550818 DOI: 10.1016/j.saa.2024.125416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/10/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
Various surface-enhanced Raman scattering (SERS) biosensors offer powerful tools for the ultrasensitive detection of circulating tumor cells (CTCs) and tumor diagnosis. Despite their efficacy, the swift and precise preparation of SERS plasmonic nanostructures poses an ongoing challenge. In this study, we introduce DNA-assisted plasmonic nanostructures capable of producing dual signals and facilitating DNA Walker signal amplification, resulting in the development of a SERS/Fluorescent (FL) dual-mode cytosensor for CTCs detection. Firstly, Au@Ag nanoparticle multimers (Au@AgNMs) featuring interparticle nano-gaps were synthesized through DNA self-assembly and in-situ deposition, which provided plasmonic nanostructures. Hence, the nano-gap distance among Au@AgNMs was meticulously regulated after optimization to achieve both SERS enhancement and fluorescence quenching. Subsequently, the aptamer (Apt) of MUC1 recognized CTCs specifically for strand displacement reaction (SDR) and further triggered the DNA Walker reaction for signal amplification. The limit of detection (LOD) of proposed cytosensor can be obtained as low as 5 cells/mL in SERS mode and 21 cells/mL in FL mode. Hence, SERS mode confers highly precise information, while FL mode allow for rapid quantitative analysis. This dual-mode cytosensor based on plasmonic nanostructures facilitates the early detection and precise treatment of cancer or infectious diseases.
Collapse
Affiliation(s)
- Pengkun Yin
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhengying Peng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610061, China
| | - Qihui Wang
- College of Chemistry and Life Science, Sichuan Provincial Key Laboratory for Structural Optimization and Application of Functional Molecules, Chengdu Normal University, Chengdu 611130, China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu 610064, China
| | - Bin Hu
- Department of Thoracic Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China.
| | - Qingyu Lin
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
2
|
Wu P, He X, Fan J, Tai Y, Zheng D, Yao Y, Sun S, Luo Y, Chen J, Hu WW, Ying B, Luo F, Niu Q, Sun X, Li Y. Electrochemical cytosensors for non-invasive liquid biopsy: Detection procedures and technologies for circulating tumor cells. Biosens Bioelectron 2025; 267:116818. [PMID: 39353368 DOI: 10.1016/j.bios.2024.116818] [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: 06/04/2024] [Revised: 09/08/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Each year, millions of new cancer cases and cancer-related deaths underscore the urgent need for effective, affordable screening methods. Circulating tumor cells (CTCs), which derived from tumors and shedding into bloodstream, are considered promising biomarkers for liquid biopsy due to their unique biological significance and the substantial volume of supporting research. Among many advanced CTCs detection methods, electrochemical sensing is rapidly developing due to their high selectivity, high sensitivity, low cost, and rapid detection capability, well meeting the growing demand for non-invasive liquid biopsy. This review focuses on the entire procedure of detecting CTCs using electrochemical cytosensors, starting from sample preparation, detailing bio-recognition elements for capturing CTCs, highlighting design strategies of cytosensor, and discussing the prospects and challenges of electrochemical cytosensor applications.
Collapse
Affiliation(s)
- Peilin Wu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Xun He
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
| | - Jiwen Fan
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Yunze Tai
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Dongdong Zheng
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Yongchao Yao
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Shengjun Sun
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Yao Luo
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Jie Chen
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Wenchuang Walter Hu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Binwu Ying
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Fengming Luo
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qian Niu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China.
| | - Xuping Sun
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China; College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Yi Li
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
3
|
Jiang M, Yang J, Liu L, Chattopadhyay AN, Rotello VM. Rapid Identification of Cell Types and Phenotypic States Using a One-Polymer Multichannel Nanosensor Fabricated via Flash Nanoprecipitation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:68928-68935. [PMID: 39651848 DOI: 10.1021/acsami.4c15474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Cell state transitions are fundamental in biology, determining how cells respond to environmental stimuli and adapt to diseases and treatments. Cell surface-based sensing of geno/phenotypes is a versatile approach for distinguishing different cell types and states. Array-based biosensors can provide a highly sensitive platform for distinguishing cells based on the differential interactions of each sensing element with cell surface components. In this work, a highly modular polymer-based supramolecular multichannel sensor array (FNP sensor) was fabricated by encapsulating a hydrophobic dye (pyrene) into the monolayer of a positively charged fluorescent polymer through flash nanoprecipitation (FNP). We utilized this one-polymer sensor array to discriminate among cell types commonly found in tumors: 4T1 cancer cells, NIH/3T3 fibroblast cells, and RAW 264.7 macrophage cells. The sensor also successfully characterized varying ratios of NIH/3T3 cancer-associated fibroblasts (CAFs) and RAW 264.7 tumor-associated macrophages (TAMs). This single polymer-based sensor array provides effective discrimination and high reproducibility, providing a high-throughput tool for diagnostic screening of cell types and states associated with cancer progression.
Collapse
Affiliation(s)
- Mingdi Jiang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Junwhee Yang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Liang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Aritra Nath Chattopadhyay
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
4
|
Khandelwal D, Bhattacharya A, Kumari V, Gupta SS, Ranjan KR, Mishra V. Leveraging nanomaterials for ultrasensitive biosensors in early cancer detection: a review. J Mater Chem B 2024. [PMID: 39635753 DOI: 10.1039/d4tb02107j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Cancer remains a major global health challenge with a high mortality rate, as evidenced by the rise in new cases every year. Conventional diagnostic methods like PET scans, MRIs, and biopsies, despite being widely used, suffer from significant drawbacks such as high radiation exposure, difficulty in distinguishing malignant from benign tumors, and invasiveness. Early detection, which is crucial for improving treatment outcomes and survival rates, is hindered by the asymptomatic nature of early-stage cancer and the limitations of current diagnostic tools. Cancer biomarkers, detectable in body fluids, offer valuable diagnostic information, and recent advances in nanotechnology have led to the development of highly sensitive nano-biosensors. This review explores recent advancements (2022-2024) in the field of ultrasensitive nano-biosensors, emphasizing the strategic integration of nanomaterials to enhance sensitivity and accuracy in cancer biomarker detection. It highlights how precise nanomaterial positioning in sensor components like electrodes and bioreceptors enables early cancer diagnosis at low biomarker concentrations. These innovations underscore the transformative potential of nanomaterials in revolutionizing early cancer diagnostics, improving patient care, and enhancing survival outcomes.
Collapse
Affiliation(s)
- Drishti Khandelwal
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, UP-201313, India.
| | - Aheli Bhattacharya
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University Uttar Pradesh, Noida, UP-201313, India.
| | - Vanshika Kumari
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University Uttar Pradesh, Noida, UP-201313, India.
| | | | - Kumar Rakesh Ranjan
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University Uttar Pradesh, Noida, UP-201313, India.
| | - Vivek Mishra
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, UP-201313, India.
| |
Collapse
|
5
|
Ren D, Wei H, Li N, Fu W, Huang Z, Yang L, Mu S. Colorimetric detection of circulating tumor cells in breast cancer based on ladder-branch hybridization chain reaction and DFs/AuNCs nanozyme. Talanta 2024; 274:125921. [PMID: 38552481 DOI: 10.1016/j.talanta.2024.125921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 05/04/2024]
Abstract
Breast cancer is the most common malignant tumor in women, which accounts for 6.9% of all cancer-related deaths. Early diagnosis is crucial for making the best clinical decision and improving the prognosis of patients. Circulating tumor cells (CTCs) have been regarded as significant tumor biomarkers. Herein, we designed a colorimetric biosensor for breast cancer CTCs quantification based on ladder-branch hybridization chain reaction (HCR) and DNA flowers/gold nanoclusters (DFs/AuNCs) nanozyme. With the assistance of complementary DNA labeled on magnetic beads (MBs), the cleavage products of RNA-cleaving DNAzymes (RCDs) could be rapidly captured, subsequently triggering ladder-branch HCR. In addition, the DFs/AuNCs nanozyme was applied for colorimetric analysis, which further improved the sensitivity for the detection of target CTCs. Benefiting from specific RCDs, ladder-branch HCR and DFs/AuNCs, we achieved a superior detection limit of 3 cells/mL as well as a broad linear range of 10 cells/mL to 104 cells/mL. Conclusively, this colorimetric biosensor achieved sensitively and selectively detection of breast cancer CTCs without the participation of enzymes at room temperature, which might provide new insight into the early detection of breast cancer.
Collapse
Affiliation(s)
- Dongxia Ren
- Department of Transfusion Medicine, Tangdu Hospital, Xi'an, 710032, China
| | - Hua Wei
- Department of Transfusion Medicine, Tangdu Hospital, Xi'an, 710032, China
| | - Na Li
- Department of Transfusion Medicine, Tangdu Hospital, Xi'an, 710032, China
| | - Wenda Fu
- Department of Transfusion Medicine, Tangdu Hospital, Xi'an, 710032, China
| | - Zhijun Huang
- Guilin University of Electronic Science and Technology, Guilin, 541004, China
| | - Longfei Yang
- Department of Transfusion Medicine, Tangdu Hospital, Xi'an, 710032, China.
| | - Shijie Mu
- Department of Transfusion Medicine, Tangdu Hospital, Xi'an, 710032, China.
| |
Collapse
|
6
|
Zhang C, Mo H, Li M, Wang S, Dou X, Zhang X. The effects of postoperative targeted immunotherapy on peripheral blood cytokines and immune cell profile in lung cancer patients. Front Oncol 2024; 14:1342624. [PMID: 38903721 PMCID: PMC11188683 DOI: 10.3389/fonc.2024.1342624] [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: 02/08/2024] [Accepted: 05/02/2024] [Indexed: 06/22/2024] Open
Abstract
Objective Cytokines and cell subsets are important components of the tumor microenvironment. Previous research has revealed that there are differences in cytokines and cell subsets in the peripheral blood of lung cancer (LCA) patients before and after eradication. The purpose of this study is to explore the monitoring value of cytokines and cellular subpopulations as biomarkers in post-immunotherapy monitoring of patients with LCA after surgery. Methods We conducted a case-control study using double-antibody sandwich magnetic microsphere flow cytometry with immunofluorescence technology and fluorescent monoclonal antibody multiparameter flow cytometry to detect differences in peripheral blood cytokines and cell subsets between LCA patients after immunotherapy and healthy controls. Results Our research results show that there are differences in the levels of IL-4, IL-6, IL-10, IL-17, IFN-γ, TNF-α in the peripheral blood of LCA patients (n=70) after immunotherapy compared to the healthy controls (n=55) (P<0.05), and there are differences in 10 cell subgroups including DP T Cells, AT cells, and NLR in the peripheral blood compared to the healthy controls (n=35) (P<0.05). Further analysis revealed significant differences in the detection data of IL-6, IL-10, IFN-γ, CD56dim NK cells, Total B cells, Total NE cells, CD15+M cells, and NLR between LCA deceased patients (n=25) and LCA surviving patients (n=27) during the same period (P<0.05). The continuous monitoring of cytokines and cell subsets is far more valuable than a single-time test, as abnormal fluctuations in the data of cytokines and cell subsets are often associated with poor prognosis. In addition, IL-6 and NLR showed the strongest discriminative ability between postoperative immunotherapy-treated LCA patients and healthy controls, with AUC values of 0.840 and 0.822, respectively. There was a significant association between IFN-γ and distant metastasis in LCA (P<0.05), as well as between CD56dim NK cells and lymph node infiltration (P<0.05). Conclusion This research results support peripheral blood cytokines and cell subsets as biomarkers for monitoring the postoperative immune status and predicting the prognosis of LCA patients after immunotherapy. The continuous monitoring of cytokines and cell subsets is far more valuable than a single-time detection.
Collapse
Affiliation(s)
- Chuang Zhang
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Medical Laboratory of the Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Hongmei Mo
- Medical Laboratory of the Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Min Li
- Department of Laboratory Medicine, Clinical Medical College of Yangzhou University, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Shuaiyan Wang
- Medical Laboratory of the Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaowen Dou
- Medical Laboratory of the Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiuming Zhang
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Medical Laboratory of the Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
7
|
Wang Z, Chen X, Qiu X, Chen Y, Wang T, Lv L, Guo X, Yang F, Tang M, Gu W, Luo Y. High-Fidelity Sensitive Tracing Circulating Tumor Cell Telomerase Activity. Anal Chem 2024; 96:5527-5536. [PMID: 38483815 DOI: 10.1021/acs.analchem.3c05749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Dynamic tracing of intracellular telomerase activity plays a crucial role in cancer cell recognition and correspondingly in earlier cancer diagnosis and personalized precision therapy. However, due to the complexity of the required reaction system and insufficient loading of reaction components into cells, achieving a high-fidelity determination of telomerase activity is still a challenge. Herein, an Aptamer-Liposome mediated Telomerase activated poly-Molecular beacon Arborescent Nanoassembly(ALTMAN) approach was described for direct high-fidelity visualization of telomerase activity. Briefly, intracellular telomerase activates molecular beacons, causing their hairpin structures to unfold and produce fluorescent signals. Furthermore, multiple molecular beacons can self-assemble, forming arborescent nanostructures and leading to exponential amplification of fluorescent signals. Integrating the enzyme-free isothermal signal amplification successfully increased the sensitivity and reduced interference by leveraging the skillful design of the molecular beacon and the extension of the telomerase-activated TTAGGG repeat sequence. The proposed approach enabled ultrasensitive visualization of activated telomerase exclusively with a prominent detection limit of 2 cells·μL-1 and realized real-time imaging of telomerase activity in living cancer cells including blood samples from breast cancer patients and urine samples from bladder cancer patients. This approach opens an avenue for establishing a telomerase activity determination and in situ monitoring technique that can facilitate both telomerase fundamental biological studies and cancer diagnostics.
Collapse
Affiliation(s)
- Zining Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
| | - Xiaohui Chen
- Department of Clinical Laboratory, Fuling Hospital, Chongqing University, Chongqing 408099, P.R. China
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing University, Chongqing 400044, P.R. China
| | - Xiaopei Qiu
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
| | - Yi Chen
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
| | - Tian Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
| | - Linxi Lv
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
| | - Xinlin Guo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
| | - Fei Yang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
| | - Miao Tang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
| | - Wei Gu
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing University, Chongqing 400044, P.R. China
| |
Collapse
|
8
|
Zhao L, Fang Y, Chen X, Meng Y, Wang F, Li C. Carbon dot-based fluorescent probe for early diagnosis of pheochromocytoma through identification of circulating tumor cells. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 310:123921. [PMID: 38271847 DOI: 10.1016/j.saa.2024.123921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/19/2023] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
Pheochromocytoma (PCC), as a rare neuroendocrine tumor, is often missed or misdiagnosed because of its atypical clinical manifestations. To realize the early accurate diagnosis of PCC, we have selected circulating tumor cells (CTCs) with more complete biological information as biomarkers and developed a simple and novel fluorescence cytosensor. Octreotide-2,2',2'',2'''- (1,4,7,10 -tetraazacyclododecane-1,4,7,10-tetrayl) tetraacetic acid (DOTA) modified magnetic Fe3O4 and signal amplification CDs@SiO2 nanospheres are prepared to capture and detect PCC-CTCs from peripheral blood via binding to the somatostatin receptor SSTR2 overexpressed on the surface of PCC cells. During the detection process, the target cells were separated and enriched by magnetic capture probes (Fe3O4-DOTA), and then signal probes (CDs@SiO2-DOTA) could also specifically bound to target cells to form the sandwich-like structure for fluorescence signal output. The proposed fluorescence cytosensor has revealed good sensitivity and selectivity for quantitative analysis of PCC-CTCs in the concentration of 5-1000 cells mL-1 with a LOD of 2 cells mL-1. More importantly, designed fluorescence cytosensor has shown good reliability and stability in complex serum samples. This strategy provides a new way for detection of PCC-CTCs.
Collapse
Affiliation(s)
- Liping Zhao
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, Jiangsu, PR China
| | - Yiwei Fang
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, Jiangsu, PR China
| | - Xinhe Chen
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, Jiangsu, PR China
| | - Yang Meng
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, Jiangsu, PR China
| | - Fei Wang
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, Jiangsu, PR China; Cell and Biomolecule Recognition Research Center, School of Science, China Pharmaceutical University, Nanjing 211198, Jiangsu, PR China.
| | - Caolong Li
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, Jiangsu, PR China; Cell and Biomolecule Recognition Research Center, School of Science, China Pharmaceutical University, Nanjing 211198, Jiangsu, PR China.
| |
Collapse
|
9
|
Bounoua N, Cetinkaya A, Piskin E, Kaya SI, Ozkan SA. The sensor applications for prostate and lung cancer biomarkers in terms of electrochemical analysis. Anal Bioanal Chem 2024; 416:2277-2300. [PMID: 38279011 DOI: 10.1007/s00216-024-05134-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/24/2023] [Accepted: 01/09/2024] [Indexed: 01/28/2024]
Abstract
Prostate and lung cancers are the most common types of cancer and affect a large part of the population around the world, causing deaths. Therefore, the rapid identification of cancer can profoundly impact reducing cancer-related death rates and protecting human lives. Significant resources have been dedicated to investigating new methods for early disease detection. Cancer biomarkers encompass various biochemical entities, including nucleic acids, proteins, sugars, small metabolites, cytogenetic and cytokinetic parameters, and whole tumor cells in bodily fluids. These tools can be utilized for various purposes, such as risk assessment, diagnosis, prognosis, treatment efficacy, toxicity evaluation, and predicting a return. Due to these versatile and critical purposes, there are widespread studies on the development of new, sensitive, and selective approaches for the determination of cancer biomarkers. This review illustrates the significant lung and prostate cancer biomarkers and their determination utilizing electrochemical sensors, which have the advantage of improved sensitivity, low cost, and simple analysis. Additionally, approaches such as improving sensitivity with nanomaterials and ensuring selectivity with MIPs are used to increase the performance of the sensor. This review aims to overview the most recent electrochemical biosensor applications for determining vital biomarkers of prostate and lung cancers in terms of nanobiosensors and molecularly imprinted polymer (MIP)-based biosensors.
Collapse
Affiliation(s)
- Nadia Bounoua
- Department of Exact Sciences, Laboratory of the Innovation Sponsorship and the Emerging Institution for Graduates of Higher Education of Sustainable Development and Dealing with Emerging Conditions, Normal Higher School of Bechar, Bechar, Algeria
- Laboratory of Chemical and Environmental Science (LCSE), 8000, Bechar, Algeria
| | - Ahmet Cetinkaya
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
- Graduate School of Health Sciences, Ankara University, Ankara, Turkey
| | - Ensar Piskin
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
- Graduate School of Health Sciences, Ankara University, Ankara, Turkey
| | - S Irem Kaya
- Department of Analytical Chemistry, Gulhane Faculty of Pharmacy, University of Health Sciences, Ankara, Turkey.
| | - Sibel A Ozkan
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey.
| |
Collapse
|
10
|
Jiang W, Wu J, Lin X, Chen Z, Lin L, Yang J. Enumeration and Molecular Characterization of Circulating Tumor Cell Using an Epithelial Cell Adhesion Molecule/Vimentin/Epidermal Growth Factor Receptor Joint Capture System in Lung Cancer. Clin Med Insights Oncol 2024; 18:11795549241231568. [PMID: 38525298 PMCID: PMC10960340 DOI: 10.1177/11795549241231568] [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: 08/22/2023] [Accepted: 01/20/2024] [Indexed: 03/26/2024] Open
Abstract
Background Detection rate and isolation yield of circulating tumor cells (CTCs) are low in lung cancer with approaches due to CTC invasiveness and heterogeneity. In this study, on the basis of the epithelial cell adhesion molecule (EpCAM) phenotype, markers of vimentin and epidermal growth factor receptor (EGFR) phenotype were added to jointly construct a precise and efficient CTC capture system for capture of lung cancer CTCs. Methods A CTC capture system combined with EpCAM lipid magnetic bead (Ep-LMB)/vimentin lipid magnetic bead (Vi-LMB)/EGFR lipid magnetic bead (EG-LMB) was constructed, and its performance was tested. The amount of CTC captured in the blood of patients with lung cancer was detected by immunofluorescence identification and analyzed for clinical relevance. Results The constructed CTC capture system has low cytotoxicity. The capture efficiency of lung cancer cells in phosphate belanced solution (PBS) system was 95.48%. The capture efficiency in the blood simulation system is 94.55%. The average number of CTCs in the blood of patients with lung cancer was 9.73/2 mL. The quantity distribution of CTCs is significantly correlated with tumor staging and metastasis. The area under the curve (AUC) of CTCs for the diagnosis of lung cancer was 0.9994 (95% CI = 0.9981-1.000, P < .0001). The cutoff value was 4.5/2 mL. The sensitivity was 99.39%, and the specificity was 96.88%. Conclusion The EpCAM/vimentin/EGFR combined capture system has feasibility and high sensitivity in the detection of lung cancer CTC typing, which can be used as an auxiliary diagnostic indicator for lung cancer and is expected to promote the clinical application of CTCs.
Collapse
Affiliation(s)
- Wentan Jiang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jingyang Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xianbin Lin
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhiyao Chen
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Liangan Lin
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jiansheng Yang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
11
|
Jiang X, Zhang X, Guo C, Ou L. Antifouling modification for high-performance isolation of circulating tumor cells. Talanta 2024; 266:125048. [PMID: 37579675 DOI: 10.1016/j.talanta.2023.125048] [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: 05/05/2023] [Revised: 07/22/2023] [Accepted: 08/05/2023] [Indexed: 08/16/2023]
Abstract
Circulating tumor cells (CTCs), which shed from solid tumor tissue into blood circulatory system, have attracted wide attention as a biomarker in the early diagnosis and prognosis of cancer. Given their potential significance in clinics, many platforms have been developed to separate CTCs. However, the high-performance isolation of CTCs remains significant challenges including achieving the sensitivity and specificity necessary due to their extreme rarity and severe biofouling in blood, such as billions of background cells and various proteins. With the advancement of CTCs detection technologies in recent years, the highly efficient and highly specific detection platforms for CTCs have gradually been developed, resulting in improving CTC capture efficiency, purity and sensitivity. In this review, we systematically describe the current strategies with surface modifications by utilizing the antifouling property of polymer, peptide, protein and cell membrane for high-performance enrichment of CTCs. To wrap up, we discuss the substantial challenges facing by current technologies and the potential directions for future research and development.
Collapse
Affiliation(s)
- Xinbang Jiang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Xiangyun Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Chen Guo
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Lailiang Ou
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
12
|
Stefanes NM, Cunha-Silva ME, de Oliveira Silva L, Walter LO, Santos-Silva MC, Gartia MR. Circulating biomarkers for diagnosis and response to therapies in cancer patients. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024. [DOI: 10.1016/bs.ircmb.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Hirasawa T, Tachi K, Ishikawa T, Miyashita M, Ito K, Ishihara M. Photoacoustic microscopy for real-time monitoring of near-infrared optical absorbers inside biological tissue. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:S11527. [PMID: 38464883 PMCID: PMC10924425 DOI: 10.1117/1.jbo.29.s1.s11527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/12/2024]
Abstract
Significance We developed a high-speed optical-resolution photoacoustic microscopy (OR-PAM) system using a high-repetition-rate supercontinuum (SC) light source and a two-axes Galvano scanner. The OR-PAM system enabled real-time imaging of optical absorbers inside biological tissues with excellent excitation wavelength tunability. Aim In the near-infrared (NIR) wavelength range, high-speed OR-PAM faces limitations due to the lack of wavelength-tunable light sources. Our study aimed to enable high-speed OR-PAM imaging of various optical absorbers, including NIR contrast agents, and validate the performance of high-speed OR-PAM in the detection of circulating tumor cells (CTCs). Approach A high-repetition nanosecond pulsed SC light source was used for OR-PAM. The excitation wavelength was adjusted by bandpass filtering of broadband light pulses produced by an SC light source. Phantom and in vivo experiments were performed to detect tumor cells stained with an NIR contrast agent within flowing blood samples. Results The newly developed high-speed OR-PAM successfully detected stained cells both in the phantom and in vivo. The phantom experiment confirmed the correlation between the tumor cell detection rate and tumor cell concentration in the blood sample. Conclusions The high-speed OR-PAM effectively detected stained tumor cells. Combining high-speed OR-PAM with molecular probes that stain tumor cells in vivo enables in vivo CTC detection.
Collapse
Affiliation(s)
- Takeshi Hirasawa
- National Defense Medical College, Department of Medical Engineering, Tokorozawa, Japan
| | - Kazuyoshi Tachi
- National Defense Medical College, Department of Medical Engineering, Tokorozawa, Japan
- National Defense Medical College, Department of Urology, Tokorozawa, Japan
| | - Tomohiro Ishikawa
- National Defense Medical College, Department of Medical Engineering, Tokorozawa, Japan
| | - Manami Miyashita
- National Defense Medical College, Department of Medical Engineering, Tokorozawa, Japan
| | - Keiichi Ito
- National Defense Medical College, Department of Urology, Tokorozawa, Japan
| | - Miya Ishihara
- National Defense Medical College, Department of Medical Engineering, Tokorozawa, Japan
| |
Collapse
|
14
|
Nie C, Shaw I, Chen C. Application of microfluidic technology based on surface-enhanced Raman scattering in cancer biomarker detection: A review. J Pharm Anal 2023; 13:1429-1451. [PMID: 38223444 PMCID: PMC10785256 DOI: 10.1016/j.jpha.2023.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 01/16/2024] Open
Abstract
With the continuous discovery and research of predictive cancer-related biomarkers, liquid biopsy shows great potential in cancer diagnosis. Surface-enhanced Raman scattering (SERS) and microfluidic technology have received much attention among the various cancer biomarker detection methods. The former has ultrahigh detection sensitivity and can provide a unique fingerprint. In contrast, the latter has the characteristics of miniaturization and integration, which can realize accurate control of the detection samples and high-throughput detection through design. Both have the potential for point-of-care testing (POCT), and their combination (lab-on-a-chip SERS (LoC-SERS)) shows good compatibility. In this paper, the basic situation of circulating proteins, circulating tumor cells, exosomes, circulating tumor DNA (ctDNA), and microRNA (miRNA) in the diagnosis of various cancers is reviewed, and the detection research of these biomarkers by the LoC-SERS platform in recent years is described in detail. At the same time, the challenges and future development of the platform are discussed at the end of the review. Summarizing the current technology is expected to provide a reference for scholars engaged in related work and interested in this field.
Collapse
Affiliation(s)
- Changhong Nie
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Ibrahim Shaw
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Chuanpin Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| |
Collapse
|
15
|
Wang M, Liu H, Fan K. Signal Amplification Strategy Design in Nanozyme-Based Biosensors for Highly Sensitive Detection of Trace Biomarkers. SMALL METHODS 2023; 7:e2301049. [PMID: 37817364 DOI: 10.1002/smtd.202301049] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/12/2023] [Indexed: 10/12/2023]
Abstract
Nanozymes show great promise in enhancing disease biomarker sensing by leveraging their physicochemical properties and enzymatic activities. These qualities facilitate signal amplification and matrix effects reduction, thus boosting biomarker sensing performance. In this review, recent studies from the last five years, concentrating on disease biomarker detection improvement through nanozyme-based biosensing are examined. This enhancement primarily involves the modulations of the size, morphology, doping, modification, electromagnetic mechanisms, electron conduction efficiency, and surface plasmon resonance effects of nanozymes for increased sensitivity. In addition, a comprehensive description of the synthesis and tuning strategies employed for nanozymes has been provided. This includes a detailed elucidation of their catalytic mechanisms in alignment with the fundamental principles of enhanced sensing technology, accompanied by the presentation of quantitatively analyzed results. Moreover, the diverse applications of nanozymes in strip sensing, colorimetric sensing, electrochemical sensing, and surface-enhanced Raman scattering have been outlined. Additionally, the limitations, challenges, and corresponding recommendations concerning the application of nanozymes in biosensing have been summarized. Furthermore, insights have been offered into the future development and outlook of nanozymes for biosensing. This review aims to serve not only as a reference for enhancing the sensitivity of nanozyme-based biosensors but also as a catalyst for exploring nanozyme properties and their broader applications in biosensing.
Collapse
Affiliation(s)
- Mengting Wang
- Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Hongxing Liu
- Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
16
|
Paglia EB, Baldin EKK, Freitas GP, Santiago TSA, Neto JBMR, Silva JVL, Carvalho HF, Beppu MM. Circulating Tumor Cells Adhesion: Application in Biosensors. BIOSENSORS 2023; 13:882. [PMID: 37754116 PMCID: PMC10526177 DOI: 10.3390/bios13090882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/23/2023] [Accepted: 09/01/2023] [Indexed: 09/28/2023]
Abstract
The early and non-invasive diagnosis of tumor diseases has been widely investigated by the scientific community focusing on the development of sensors/biomarkers that act as a way of recognizing the adhesion of circulating tumor cells (CTCs). As a challenge in this area, strategies for CTCs capture and enrichment currently require improvements in the sensors/biomarker's selectivity. This can be achieved by understanding the biological recognition factors for different cancer cell lines and also by understanding the interaction between surface parameters and the affinity between macromolecules and the cell surface. To overcome some of these concerns, electrochemical sensors have been used as precise, fast-response, and low-cost transduction platforms for application in cytosensors. Additionally, distinct materials, geometries, and technologies have been investigated to improve the sensitivity and specificity properties of the support electrode that will transform biochemical events into electrical signals. This review identifies novel approaches regarding the application of different specific biomarkers (CD44, Integrins, and EpCAm) for capturing CTCs. These biomarkers can be applied in electrochemical biosensors as a cytodetection strategy for diagnosis of cancerous diseases.
Collapse
Affiliation(s)
- Eduarda B. Paglia
- School of Chemical Engineering, Department of Process and Product Development, University of Campinas, Campinas 13083-852, Brazil; (E.B.P.); (E.K.K.B.); (G.P.F.); (T.S.A.S.)
| | - Estela K. K. Baldin
- School of Chemical Engineering, Department of Process and Product Development, University of Campinas, Campinas 13083-852, Brazil; (E.B.P.); (E.K.K.B.); (G.P.F.); (T.S.A.S.)
- Renato Archer Information Technology Center, Campinas 13069-901, Brazil;
| | - Gabriela P. Freitas
- School of Chemical Engineering, Department of Process and Product Development, University of Campinas, Campinas 13083-852, Brazil; (E.B.P.); (E.K.K.B.); (G.P.F.); (T.S.A.S.)
- Renato Archer Information Technology Center, Campinas 13069-901, Brazil;
| | - Thalyta S. A. Santiago
- School of Chemical Engineering, Department of Process and Product Development, University of Campinas, Campinas 13083-852, Brazil; (E.B.P.); (E.K.K.B.); (G.P.F.); (T.S.A.S.)
| | - João B. M. R. Neto
- Technology Center, Federal University of Alagoas, Maceió 57072-900, Brazil;
| | - Jorge V. L. Silva
- Renato Archer Information Technology Center, Campinas 13069-901, Brazil;
| | - Hernandes F. Carvalho
- Institute of Biology, Department of Structural and Functional Biology, University of Campinas, Campinas 13083-864, Brazil;
| | - Marisa M. Beppu
- School of Chemical Engineering, Department of Process and Product Development, University of Campinas, Campinas 13083-852, Brazil; (E.B.P.); (E.K.K.B.); (G.P.F.); (T.S.A.S.)
| |
Collapse
|
17
|
Wang J, Ben-David R, Mehrazin R, Yang W, Tewari AK, Kyprianou N. Novel signatures of prostate cancer progression and therapeutic resistance. Expert Opin Ther Targets 2023; 27:1195-1206. [PMID: 38108262 DOI: 10.1080/14728222.2023.2293757] [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: 08/31/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION The extensive heterogeneity of prostate cancer (PCa) and multilayered complexity of progression to castration-resistant prostate cancer (CRPC) have contributed to the challenges of accurately monitoring advanced disease. Profiling of the tumor microenvironment with large-scale transcriptomic studies have identified gene signatures that predict biochemical recurrence, lymph node invasion, metastases, and development of therapeutic resistance through critical determinants driving CRPC. AREAS COVERED This review encompasses understanding of the role of different molecular determinants of PCa progression to lethal disease including the phenotypic dynamic of cell plasticity, EMT-MET interconversion, and signaling-pathways driving PCa cells to advance and metastasize. The value of liquid biopsies encompassing circulating tumor cells and extracellular vesicles to detect disease progression and emergence of therapeutic resistance in patients progressing to lethal disease is discussed. Relevant literature was added from PubMed portal. EXPERT OPINION Despite progress in the tumor-targeted therapeutics and biomarker discovery, distant metastasis and therapeutic resistance remain the major cause of mortality in patients with advanced CRPC. No single signature can encompass the tremendous phenotypic and genomic heterogeneity of PCa, but rather multi-threaded omics-derived and phenotypic markers tailored and validated into a multimodal signature.
Collapse
Affiliation(s)
- Jason Wang
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Reuben Ben-David
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Reza Mehrazin
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wei Yang
- Department of Pathology, Stony Brook University, New York, NY, USA
| | - Ashutosh K Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology & Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
18
|
Aydın EB, Aydın M, Sezgintürk MK. A novel electrochemical impedance immunosensor for the quantification of CYFRA 21-1 in human serum. Mikrochim Acta 2023; 190:235. [PMID: 37219635 DOI: 10.1007/s00604-023-05813-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023]
Abstract
A sensitive, simple, and reliable immunosensor was constructed to detect the lowest alteration of a fragment of cytokeratin subunit 19 (CYFRA 21-1), a protein lung carcinoma biomarker. The proposed immunosensor was manufactured with a carbon black C45/polythiophene polymer-containing amino terminal groups (C45-PTNH2) conductive nanocomposite, resulting in an excellent, biocompatible, low-cost, and electrically conductive electrode surface. Anti-CYFRA 21-1 biorecognition molecules were attached to the electrode thanks to the amino terminal groups of the used PTNH2 polymer with a relatively simple procedure. All electrode surfaces after modifications were characterized by electrochemical, chemical, and microscopic techniques. Electrochemical impedance spectroscopy (EIS) was also utilized for the evaluation of the analytical feature of the immunosensor. The charge transfer resistance of the immunosensor signal was correlated with the CYFRA 21-1 concentration in the concentration range 0.03 to 90 pg/mL. The limit of detection (LOD) and the limit of quantification (LOQ) of the suggested system were 4.7 fg/mL and 14.1 fg/mL, respectively. The proposed biosensor had favorable repeatability and reproducibility, long storage stability, excellent selectivity, and low cost. Furthermore, it was applied to determine CYFRA 21-1 in commercial serum samples, and satisfactory recovery results (98.63-106.18%) were obtained. Thus, this immunosensor can be offered for clinical purposes as a rapid, stable, low-cost, selective, reproducible, and reusable tool.
Collapse
Affiliation(s)
- Elif Burcu Aydın
- Scientific and Technological Research Center, Tekirdağ Namık Kemal University, Tekirdağ, Turkey.
| | - Muhammet Aydın
- Scientific and Technological Research Center, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Mustafa Kemal Sezgintürk
- Bioengineering Department, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
19
|
Smejkal J, Aubrecht P, Semerádtová A, Štofik M, Liegertová M, Malý J. Immunocapturing rare cells from blood: A simple and robust microsystem approach. Biosens Bioelectron 2023; 227:115155. [PMID: 36821992 DOI: 10.1016/j.bios.2023.115155] [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: 12/13/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/21/2023]
Abstract
Cell immunocapture microsystems are a fast-emerging field with several potential medical diagnostic applications. Isolation and quantification of circulating rare cells (CRCs) show great importance in the early stages of disease diagnostics and prognostics. Here, we present a simple and robust stop-flow microsystem (fabricated by a combination of glass microblasting and 3D printing) based on a planar antibody-coated surface that is effective in the immunocapture of the model as well as naturally occurring rare cells. A chip with a planar immunocapture channel working in the so-called stop-flow dynamic regime was designed to enable monitoring the efficiency of the cell capture by fluorescence microscopy. Up to 90% immunocapture efficiency of MCF-7 cells spiked into whole blood on CD326 antibody-coated planar surfaces was achieved. We discuss the role of the planar surface modifications, the influence of the set stop-flow dynamic conditions, and medium complexity on the efficiency of cell immunocapture. The presented results could be further employed in the design of microsystems for cell-size-independent isolation and identification of rare cells from blood.
Collapse
Affiliation(s)
- Jiří Smejkal
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, 400 96, Ústí nad Labem, Czech Republic.
| | - Petr Aubrecht
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, 400 96, Ústí nad Labem, Czech Republic
| | - Alena Semerádtová
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, 400 96, Ústí nad Labem, Czech Republic
| | - Marcel Štofik
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, 400 96, Ústí nad Labem, Czech Republic
| | - Michaela Liegertová
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, 400 96, Ústí nad Labem, Czech Republic
| | - Jan Malý
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, 400 96, Ústí nad Labem, Czech Republic
| |
Collapse
|
20
|
Huang Y, Li X, Hou J, Luo Z, Yang G, Zhou S. Conductive Nanofibers-Enhanced Microfluidic Device for the Efficient Capture and Electrical Stimulation-Triggered Rapid Release of Circulating Tumor Cells. BIOSENSORS 2023; 13:bios13050497. [PMID: 37232858 DOI: 10.3390/bios13050497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/18/2023] [Accepted: 04/22/2023] [Indexed: 05/27/2023]
Abstract
The effective detection and release of circulating tumor cells (CTCs) are of great significance for cancer diagnosis and monitoring. The microfluidic technique has proved to be a promising method for CTCs isolation and subsequent analysis. However, complex micro-geometries or nanostructures were often constructed and functionalized to improve the capture efficiency, which limited the scale-up for high-throughput production and larger-scale clinical applications. Thus, we designed a simple conductive nanofiber chip (CNF-Chip)-embedded microfluidic device with a herringbone microchannel to achieve the efficient and specific capture and electrical stimulation-triggered rapid release of CTCs. Here, the most used epithelial cell adhesion molecule (EpCAM) was selected as the representative biomarker, and the EpCAM-positive cancer cells were mainly studied. Under the effects of the nanointerface formed by the nanofibers with a rough surface and the herringbone-based high-throughput microfluidic mixing, the local topographic interaction between target cells and nanofibrous substrate in the microfluidic was synergistically enhanced, and the capture efficiency for CTCs was further improved (more than 85%). After capture, the sensitive and rapid release of CTCs (release efficiency above 97%) could be conveniently achieved through the cleavage of the gold-sulfur bond by applying a low voltage (-1.2 V). The device was successfully used for the effective isolation of CTCs in clinical blood samples from cancer patients, indicating the great potential of this CNF-Chip-embedded microfluidic device in clinical applications.
Collapse
Affiliation(s)
- Yisha Huang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xilin Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jianwen Hou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Zhouying Luo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Guang Yang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
21
|
Sadeghi M, Sadeghi S, Naghib SM, Garshasbi HR. A Comprehensive Review on Electrochemical Nano Biosensors for Precise Detection of Blood-Based Oncomarkers in Breast Cancer. BIOSENSORS 2023; 13:bios13040481. [PMID: 37185556 PMCID: PMC10136762 DOI: 10.3390/bios13040481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023]
Abstract
Breast cancer (BC), one of the most common and life-threatening cancers, has the highest incidence rate among women. Early diagnosis of BC oncomarkers is considered the most effective strategy for detecting and treating BC. Finding the type and stage of BC in women as soon as possible is one of the greatest ways to stop its incidence and negative effects on medical treatment. The development of biosensors for early, sensitive, and selective detection of oncomarkers has recently attracted much attention. An electrochemical nano biosensor (EN) is a very suitable option for a powerful tool for cancer diagnosis. This comprehensive review provides information about the prevalence and pathobiology of BC, recent advances in clinically available BC oncomarkers, and the most common electrochemical nano biosensors for point-of-care (POC) detection of various BC oncomarkers using nanomaterial-based signal amplification techniques.
Collapse
Affiliation(s)
- Mahdi Sadeghi
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center (BCRC), Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Somayeh Sadeghi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - Hamid Reza Garshasbi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| |
Collapse
|
22
|
Vajhadin F, Mazloum-Ardakani M, Hemati M, Moshtaghioun SM. Facile preparation of a cost-effective platform based on ZnFe 2O 4 nanomaterials for electrochemical cell detection. Sci Rep 2023; 13:4962. [PMID: 36973342 PMCID: PMC10042879 DOI: 10.1038/s41598-023-31377-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
Circulating tumor cells (CTCs) are important tumor markers that indicate early metastasis, tumor recurrence, and treatment efficacy. To identify and separate these cells from the blood, new nanomaterials need to be developed. The present study explored the potential application of ZnFe2O4 magnetic nanoparticles in capturing CTCs with cell surface markers. Folic acid was coupled to L-cysteine-capped ZnFe2O4 nanoparticles (ZC) to provide binding sites on ZnFe2O4 nanoparticles for the recognition of folate bioreceptors, which are highly expressed in MCF-7 breast cancer cells. The cytotoxicity of ZnFe2O4 nanoparticles and ZC against MCF-7 was analyzed with the MTT assay. After 24 h of incubation, there were IC50 values of 702.6 and 805.5 µg/mL for ZnFe2O4 and ZC, respectively. However, after 48 h of incubation, IC50 values of ZnFe2O4 and ZC were reduced to 267.3 and 389.7 µg/mL, respectively. The cell quantification was conducted with magnetically collected cells placed on a glassy carbon electrode, and the differential pulse voltammetry (DPV) responses were analyzed. This cost-effective ZnFe2O4-based biosensing platform allowed cancer cell detection with a limit of detection of 3 cells/mL, ranging from 25 to 104 cells/mL. In future, these functionalized zinc ferrites may be used in electrochemical cell detection and targeted cancer therapy.
Collapse
Affiliation(s)
- Fereshteh Vajhadin
- Department of Chemistry, Faculty of Science, Yazd University, Yazd, 8915818411, Iran
| | | | - Mahdie Hemati
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | |
Collapse
|
23
|
Asci Erkocyigit B, Ozufuklar O, Yardim A, Guler Celik E, Timur S. Biomarker Detection in Early Diagnosis of Cancer: Recent Achievements in Point-of-Care Devices Based on Paper Microfluidics. BIOSENSORS 2023; 13:387. [PMID: 36979600 PMCID: PMC10046104 DOI: 10.3390/bios13030387] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
Microfluidics is very crucial in lab-on-a-chip systems for carrying out operations in a large-scale laboratory environment on a single chip. Microfluidic systems are miniaturized devices in which the fluid behavior and control can be manipulated on a small platform, with surface forces on the platform being greater than volumetric forces depending on the test method used. In recent years, paper-based microfluidic analytical devices (μPADs) have been developed to be used in point-of-care (POC) technologies. μPADs have numerous advantages, including ease of use, low cost, capillary action liquid transfer without the need for power, the ability to store reagents in active form in the fiber network, and the capability to perform multiple tests using various measurement techniques. These benefits are critical in the advancement of paper-based microfluidics in the fields of disease diagnosis, drug application, and environment and food safety. Cancer is one of the most critical diseases for early detection all around the world. Detecting cancer-specific biomarkers provides significant data for both early diagnosis and controlling the disease progression. μPADs for cancer biomarker detection hold great promise for improving cure rates, quality of life, and minimizing treatment costs. Although various types of bioanalytical platforms are available for the detection of cancer biomarkers, there are limited studies and critical reviews on paper-based microfluidic platforms in the literature. Hence, this article aims to draw attention to these gaps in the literature as well as the features that future platforms should have.
Collapse
Affiliation(s)
- Bilge Asci Erkocyigit
- Department of Biotechnology, Institute of Natural Sciences, Ege University, Izmir 35100, Turkey
| | - Ozge Ozufuklar
- Department of Biotechnology, Institute of Natural Sciences, Ege University, Izmir 35100, Turkey
| | - Aysenur Yardim
- Department of Bioengineering, Institute of Natural Sciences, Ege University, Izmir 35100, Turkey
| | - Emine Guler Celik
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir 35100, Turkey
| | - Suna Timur
- Department of Biochemistry, Faculty of Science, Ege University, Izmir 35100, Turkey
- Central Research Test and Analysis Laboratory Application, Research Center, Ege University, Izmir 35100, Turkey
| |
Collapse
|
24
|
Lin J, Zhang D, Yu J, Pan T, Wu X, Chen T, Gao C, Chen C, Wang X, Wu A. Amorphous Nitrogen-Doped Carbon Nanocages with Excellent SERS Sensitivity and Stability for Accurate Identification of Tumor Cells. Anal Chem 2023; 95:4671-4681. [PMID: 36735867 DOI: 10.1021/acs.analchem.2c05272] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The surface-enhanced Raman scattering (SERS) bioprobe's strategy for identifying tumor cells always depended on the intensity difference of the Raman signal compared with that of normal cells. Hence, exploring novel SERS nanostructure with excellent spectra stability, a high enhancement factor (EF), and good biocompatibility is a primary premise for boosting SERS signal reliability and accuracy of tumor cells. Here, high SERS EF (5.52 × 106) is acquired by developing novel amorphous nitrogen-doped carbon (NDC) nanocages (NCs), whose EF value was in a leading position among carbon-based SERS substrates. In addition, a uniform SERS signal was obtained on NDC NCs due to homogeneous morphology and size. The delocalized carbon-conjugated systems of graphitic-N, pyrrole-N, and pyridine-N with lone pair electrons increase the electronic density of states and reduce the electron localization function of NDC NCs, thereby promoting the charge transfer process. The electron-donor platform of the NDC NCs facilitates the thermodynamic process of charge transfer, resulting in multimode vibrational coupling in the surface complexes, which greatly amplifies the molecular polarizability. Importantly, the good biocompatibility and signal stability endow these NDC NC SERS bioprobes unique superiority in distinguishing tumor cells, and quantitative recognition of two triple-negative breast cancer cells based on SERS detection mode has been successfully realized.
Collapse
Affiliation(s)
- Jie Lin
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo315201, People's Republic of China.,Advanced Energy Science and Technology Guangdong Laboratory, Huizhou516000, People's Republic of China
| | - Dinghu Zhang
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo315201, People's Republic of China
| | - Jian Yu
- School of Chemistry, Beihang University, Beijing100191, China
| | - Ting Pan
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo315201, People's Republic of China
| | - Xiaoxia Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo315201, People's Republic of China
| | - Tianxiang Chen
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo315201, People's Republic of China.,Advanced Energy Science and Technology Guangdong Laboratory, Huizhou516000, People's Republic of China
| | - Changyong Gao
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo315201, People's Republic of China.,Advanced Energy Science and Technology Guangdong Laboratory, Huizhou516000, People's Republic of China
| | - Chao Chen
- School of Materials Science and Engineering, Nanyang Technological University, Singapore639798, Singapore
| | - Xiaotian Wang
- School of Chemistry, Beihang University, Beijing100191, China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo315201, People's Republic of China.,Advanced Energy Science and Technology Guangdong Laboratory, Huizhou516000, People's Republic of China
| |
Collapse
|
25
|
Kling A, Dirscherl L, Dittrich PS. Laser-assisted protein micropatterning in a thermoplastic device for multiplexed prostate cancer biomarker detection. LAB ON A CHIP 2023; 23:534-541. [PMID: 36642981 PMCID: PMC9890490 DOI: 10.1039/d2lc00840h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
Immunoassays are frequently used for analysis of protein biomarkers. The specificity of antibodies enables parallel analysis of several target proteins, at the same time. However, the implementation of such multiplexed assays into cost-efficient and mass-producible thermoplastic microfluidic platforms remains difficult due to the lack of suitable immobilization strategies for different capture antibodies. Here, we introduce and characterize a method to functionalize the surfaces of microfluidic devices manufactured in the thermoplastic material cyclic olefin copolymer (COC) by a rapid prototyping process. A laser-induced immobilization process enables the surface patterning of anchor biomolecules at a spatial resolution of 5 μm. We employ the method for the analysis of prostate cancer associated biomarkers by competitive immunoassays in a microchannel with a total volume of 320 nL, and successfully detected the proteins PSA, CRP, CEA and IGF-1 at clinically relevant concentrations. Finally, we also demonstrate the simultaneous analysis of three markers spiked into undiluted human plasma. In conclusion, this method opens the way to transfer multiplexed immunoassays into mass-producible microfluidic platforms that are suitable for point of care applications.
Collapse
Affiliation(s)
- André Kling
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland.
| | - Lorin Dirscherl
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland.
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland.
| |
Collapse
|
26
|
Pourmadadi M, Moammeri A, Shamsabadipour A, Moghaddam YF, Rahdar A, Pandey S. Application of Various Optical and Electrochemical Nanobiosensors for Detecting Cancer Antigen 125 (CA-125): A Review. BIOSENSORS 2023; 13:99. [PMID: 36671934 PMCID: PMC9856029 DOI: 10.3390/bios13010099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/23/2022] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
Nowadays, diagnosing early-stage cancers can be vital for saving patients and dramatically decreases mortality rates. Therefore, specificity and sensitivity in the detection of cancer antigens should be elaborately ensured. Some early-stage cancers can be diagnosed via detecting the cancer antigen CA-125, such as ovarian cancer, and required treatments can be applied more efficiently. Thus, detection of CA-125 by employing various optical or electrochemical biosensors is a preliminary and crucial step to treating cancers. In this review, a diverse range of optical and electrochemical means of detecting CA-125 are reviewed. Furthermore, an applicable comparison of their performance and sensitivity is provided, several commercial detection kits are investigated, and their applications are compared and discussed to determine whether they are applicable and accurate enough.
Collapse
Affiliation(s)
- Mehrab Pourmadadi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 11155-4563, Iran
| | - Ali Moammeri
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 11155-4563, Iran
| | - Amin Shamsabadipour
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 11155-4563, Iran
| | | | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | - Sadanand Pandey
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
27
|
Hu X, Tan W, Cheng S, Xian Y, Zhang C. Nucleic acid and nanomaterial-assisted signal-amplified strategies in fluorescent analysis of circulating tumor cells and small extracellular vesicles. Anal Bioanal Chem 2023:10.1007/s00216-022-04509-2. [PMID: 36599923 DOI: 10.1007/s00216-022-04509-2] [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: 11/13/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023]
Abstract
As two main types of liquid biopsy markers, both circulating tumor cells (CTCs) and small extracellular vesicles (sEVs) play important roles in the diagnosis and prognosis of cancers. CTCs are malignant cells that detach from the original tumor tissue and enter the circulation of body fluids. sEVs are nanoscale vesicles secreted by normal cells or pathological cells. However, CTCs and sEVs in body fluids are scarce, leading to great difficulties in the accurate analysis of related diseases. For the sensitive detection of CTCs and sEVs in body fluids, various types of nucleic acid and nanomaterial-assisted signal amplification strategies have been developed. In this review, we summarize the recent advances in fluorescent detection of CTCs and sEVs in liquid biopsy based on nucleic acid and nanomaterial-assisted signal amplification strategies. We also discuss their advantages, challenges, and future prospects.
Collapse
Affiliation(s)
- Xinyu Hu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Wenqiao Tan
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Shasha Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yuezhong Xian
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Cuiling Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
28
|
Accurate diagnosis of prostate cancer with CRISPR-based nucleic acid test strip by simultaneously identifying PCA3 and KLK3 genes. Biosens Bioelectron 2023; 220:114854. [DOI: 10.1016/j.bios.2022.114854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
|
29
|
Chen L, Yang J, Xu G, Wu Y. Potential Value and Application of Liquid Biopsy in Tumor, Neurodegeneration, and Muscle Degenerative Diseases. Methods Mol Biol 2023; 2695:317-335. [PMID: 37450129 DOI: 10.1007/978-1-0716-3346-5_22] [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] [Indexed: 07/18/2023]
Abstract
Liquid biopsy provides a promising alternative for the detection of disease-specific markers due to its superior noninvasive and original tissue representativeness. Liquid biopsies have a wide range of health and disease applications involving components ranging from circulating cells to acellular nucleic acid molecules and other metabolites. Here, we review the different components of liquid biopsy and investigate the most advanced noninvasive methods for detecting these components as well as their existing problems and trends. In particular, we emphasize the importance of analyzing liquid biopsy data from extracellular vesicles and small nucleic acids in neurological and muscle degeneration, with the aim of using this technique to enhance personalized healthcare. Although previous reviews have focused on cancer, this review mainly emphasizes the potential application of extracellular vesicles and microRNAs in liquid biopsy in neurodegeneration and muscle degeneration.
Collapse
Affiliation(s)
- Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei, People's Republic of China
| | - Jun Yang
- Jianghan University Library, Wuhan, Hubei, People's Republic of China
| | - Guodong Xu
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei, People's Republic of China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
30
|
Kim H, Zhbanov A, Yang S. Microfluidic Systems for Blood and Blood Cell Characterization. BIOSENSORS 2022; 13:13. [PMID: 36671848 PMCID: PMC9856090 DOI: 10.3390/bios13010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
A laboratory blood test is vital for assessing a patient's health and disease status. Advances in microfluidic technology have opened the door for on-chip blood analysis. Currently, microfluidic devices can reproduce myriad routine laboratory blood tests. Considerable progress has been made in microfluidic cytometry, blood cell separation, and characterization. Along with the usual clinical parameters, microfluidics makes it possible to determine the physical properties of blood and blood cells. We review recent advances in microfluidic systems for measuring the physical properties and biophysical characteristics of blood and blood cells. Added emphasis is placed on multifunctional platforms that combine several microfluidic technologies for effective cell characterization. The combination of hydrodynamic, optical, electromagnetic, and/or acoustic methods in a microfluidic device facilitates the precise determination of various physical properties of blood and blood cells. We analyzed the physical quantities that are measured by microfluidic devices and the parameters that are determined through these measurements. We discuss unexplored problems and present our perspectives on the long-term challenges and trends associated with the application of microfluidics in clinical laboratories. We expect the characterization of the physical properties of blood and blood cells in a microfluidic environment to be considered a standard blood test in the future.
Collapse
Affiliation(s)
- Hojin Kim
- Department of Mechatronics Engineering, Dongseo University, Busan 47011, Republic of Korea
| | - Alexander Zhbanov
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Sung Yang
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
31
|
Singh S, Podder PS, Russo M, Henry C, Cinti S. Tailored point-of-care biosensors for liquid biopsy in the field of oncology. LAB ON A CHIP 2022; 23:44-61. [PMID: 36321747 DOI: 10.1039/d2lc00666a] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
In the field of cancer detection, technologies to analyze tumors using biomarkers circulating in fluids such as blood have developed rapidly based on liquid biopsy. A proactive approach to early cancer detection can lead to more effective treatments with minimal side effects and better long-term patient survival. However, early detection of cancer is hindered by the existing limitations of conventional cancer diagnostic methods. To enable early diagnosis and regular monitoring and improve automation, the development of integrated point-of-care (POC) and biosensors is needed. This is expected to fundamentally change the diagnosis, management, and monitoring of response to treatment of cancer. POC-based techniques will provide a way to avoid complications that occur after invasive tissue biopsy, such as bleeding, infection, and pain. The aim of this study is to provide a comprehensive view of biosensors and their clinical relevance in oncology for the detection of biomarkers with liquid biopsies of proteins, miRNA, ctDNA, exosomes, and cancer cells. The preceding discussion also illustrates the changing landscape of liquid biopsy-based cancer diagnosis through nanomaterials, machine learning, artificial intelligence, wearable devices, and sensors, many of which apply POC design principles. With the advent of sensitive, selective, and timely detection of cancer, we see the field of POC technology for cancer detection and treatment undergoing a positive paradigm shift in the foreseeable future.
Collapse
Affiliation(s)
- Sima Singh
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy.
| | - Pritam Saha Podder
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Matt Russo
- Department of Chemistry, Colorado State University, Fort Collins, CO, 80523-1872, USA
| | - Charles Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO, 80523-1872, USA
| | - Stefano Cinti
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy.
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Napoli Federico II, 80055 Naples, Italy
| |
Collapse
|
32
|
Arjun AM, Krishna PH, Nath AR, Rasheed PA. A review on advances in the development of electrochemical sensors for the detection of anesthetic drugs. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:4040-4052. [PMID: 36173296 DOI: 10.1039/d2ay01290a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Surgeries are a crucial medical intervention that has saved countless lives from time immemorial. To reduce pain during surgeries patients are administered with anesthetic drugs, which cause loss of sensation and thus reduce the pain involved. However, anesthetists control the effects of the drug by depending on pharmacokinetic calculations, which may vary from patient to patient, thus leading to a reduction in the quality of anesthetic care and adverse effects. To avoid these adverse effects, it is highly necessary to implement a real time monitoring of plasma drug concentration, which will adjust the drug infusion and maintain the levels of drug within therapeutic levels. To implement such a system, it is highly essential to analyze current advances in electrochemical sensor systems for different types of anesthetic drugs like opioids, intravenous anesthetics, and neuromuscular blockers. This review focuses on the present strategy of electrochemical sensors implemented for the detection of anesthetic drugs and it helps towards developing a real time drug dispensing system with respect to the plasma concentration of the drug. This analysis will contribute towards establishing highly effective real time drug dispensing systems like the total intravenous anesthesia technique and patient-controlled analgesia. Such systems will lead to better usage of anesthetic drugs and improve the quality of anesthetic care thus making surgeries safer and more painless.
Collapse
Affiliation(s)
- Ajith Mohan Arjun
- Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, Kerala, India-678 557.
| | - Prasannakumari H Krishna
- Department of Anaesthesiology, Regional Cancer Center, Medical College Campus, Post Bag No. 2417, Thiruvananthapuram, India 695011
| | - Anish R Nath
- DST Unit on Nanoscience and Thematic Unit of Excellence, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India-600036
| | - P Abdul Rasheed
- Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, Kerala, India-678 557.
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala, India-678 557
| |
Collapse
|
33
|
A CRISPR-Cas12a-powered magnetic relaxation switching biosensor for the sensitive detection of Salmonella. Biosens Bioelectron 2022; 213:114437. [PMID: 35696867 DOI: 10.1016/j.bios.2022.114437] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/10/2022] [Accepted: 05/27/2022] [Indexed: 11/23/2022]
Abstract
Magnetic relaxation switching (MRS) biosensors are attractive in the field of food safety owing to their simplicity and high signal-to-noise ratio. But they are less in sensitivity and stability caused by the insufficient crosslinking or non-specific binding of magnetic nanoparticles (MNPs) with targets. To address this problem, the CRISPR-Cas12a system was introduced into an MRS biosensor for the first time, to precisely control the binding of two types of MNPs with sizes of 130 nm (MNP130) and 30 nm (MNP30), for the sensitive detection of Salmonella. Delicately, the biosensor was designed based on the different magnetic properties of the two sizes of MNPs. The target Salmonella activated the collateral cleavage activity of the CRISPR-Cas12a system, which inhibited the binding of the two sizes of MNPs, resulting in an increase of unbound MNP30. After separating MNP130-MNP30 complexes and MNP130 from MNP30, the free MNP30 left in solution acted as transverse relaxation time (T2) signal reporters for Salmonella detection. Under optimized conditions, the CRISPR-MRS biosensor presented a limit of detection of 1.3 × 102 CFU mL-1 for Salmonella, which is lower than most MRS biosensor analogues. It also showed satisfactory specificity and performed well in spiked chicken meat samples. This biosensing strategy not only extends the reach of the CRISPR-Cas12a system in biosensors but also offers an alternative for pathogen detection with satisfactory sensitivity.
Collapse
|
34
|
Dathathri E, Isebia KT, Abali F, Lolkema MP, Martens JWM, Terstappen LWMM, Bansal R. Liquid Biopsy Based Circulating Biomarkers in Metastatic Prostate Cancer. Front Oncol 2022; 12:863472. [PMID: 35669415 PMCID: PMC9165750 DOI: 10.3389/fonc.2022.863472] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer is the most dominant male malignancy worldwide. The clinical presentation of prostate cancer ranges from localized indolent to rapidly progressing lethal metastatic disease. Despite a decline in death rate over the past years, with the advent of early diagnosis and new treatment options, challenges remain towards the management of metastatic prostate cancer, particularly metastatic castration sensitive prostate cancer (mCSPC) and castration resistant prostate cancer (mCRPC). Current treatments involve a combination of chemotherapy with androgen deprivation therapy and/or androgen receptor signalling inhibitors. However, treatment outcomes are heterogeneous due to significant tumor heterogeneity indicating a need for better prognostic biomarkers to identify patients with poor outcomes. Liquid biopsy has opened a plethora of opportunities from early diagnosis to (personalized) therapeutic disease interventions. In this review, we first provide recent insights about (metastatic) prostate cancer and its current treatment landscape. We highlight recent studies involving various circulating biomarkers such as circulating tumor cells, genetic markers, circulating nucleic acids, extracellular vesicles, tumor-educated platelets, and the secretome from (circulating) tumor cells and tumor microenvironment in metastatic prostate cancer. The comprehensive array of biomarkers can provide a powerful approach to understanding the spectrum of prostate cancer disease and guide in developing improved and personalized treatments for patients.
Collapse
Affiliation(s)
- Eshwari Dathathri
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| | - Khrystany T. Isebia
- Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Department of Medical Oncology, Rotterdam, Netherlands
| | - Fikri Abali
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| | - Martijn P. Lolkema
- Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Department of Medical Oncology, Rotterdam, Netherlands
| | - John W. M. Martens
- Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Department of Medical Oncology, Rotterdam, Netherlands
| | - Leon W. M. M. Terstappen
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| | - Ruchi Bansal
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| |
Collapse
|
35
|
Khoshroo A, Fattahi A, Hosseinzadeh L. Development of paper-based aptasensor for circulating tumor cells detection in the breast cancer. J Electroanal Chem (Lausanne) 2022. [DOI: 10.1016/j.jelechem.2022.116182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
He S, Yu S, Wei J, Ding L, Yang X, Wu Y. New horizons in the identification of circulating tumor cells (CTCs): An emerging paradigm shift in cytosensors. Biosens Bioelectron 2022; 203:114043. [PMID: 35121449 DOI: 10.1016/j.bios.2022.114043] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/02/2022] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
Abstract
Circulating tumor cells (CTCs) are cancer cells that are shed from a primary tumor into the bloodstream and function as seeds for cancer metastasis at distant locations. Enrichment and identification methods of CTCs in the blood of patients plays an important role in diagnostic assessments and personalized treatments of cancer. However, the current traditional identification methods not only impact the viability of cells, but also cannot determine the type of cancer cells when the disease is unknown. Hence, new methods to identify CTCs are urgently needed. In this context, many advanced and safe technologies have emerged to distinguish between cancer cells and blood cells, and to distinguish specific types of cancer cells. In this review, at first we have briefly discussed recent advances in technologies related to the enrichment of CTCs, which lay a good foundation for the identification of CTCs. Next, we have summarized state-of-the-art technologies to confirm whether a given cell is indeed a tumor cell and determine the type of tumor cell. Finally, the challenges for application and potential directions of the current identification methods in clinical analysis of CTCs have been discussed.
Collapse
Affiliation(s)
- Sitian He
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Songcheng Yu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Jinlan Wei
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaonan Yang
- Institute of Intelligent Sensing, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
37
|
Haince JF, Joubert P, Bach H, Ahmed Bux R, Tappia PS, Ramjiawan B. Metabolomic Fingerprinting for the Detection of Early-Stage Lung Cancer: From the Genome to the Metabolome. Int J Mol Sci 2022; 23:ijms23031215. [PMID: 35163138 PMCID: PMC8835988 DOI: 10.3390/ijms23031215] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
The five-year survival rate of lung cancer patients is very low, mainly because most newly diagnosed patients present with locally advanced or metastatic disease. Therefore, early diagnosis is key to the successful treatment and management of lung cancer. Unfortunately, early detection methods of lung cancer are not ideal. In this brief review, we described early detection methods such as chest X-rays followed by bronchoscopy, sputum analysis followed by cytological analysis, and low-dose computed tomography (LDCT). In addition, we discussed the potential of metabolomic fingerprinting, compared to that of other biomarkers, including molecular targets, as a low-cost, high-throughput blood-based test that is both feasible and affordable for early-stage lung cancer screening of at-risk populations. Accordingly, we proposed a paradigm shift to metabolomics as an alternative to molecular and proteomic-based markers in lung cancer screening, which will enable blood-based routine testing and be accessible to those patients at the highest risk for lung cancer.
Collapse
Affiliation(s)
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Pathology, Laval University, Quebec, QC G1V 4G5, Canada;
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC V6H 3Z6, Canada;
| | - Rashid Ahmed Bux
- BioMark Diagnostics Inc., Richmond, BC V6X 2W8, Canada; (J.-F.H.); (R.A.B.)
| | - Paramjit S. Tappia
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Correspondence: ; Tel.: +1-204-258-1230
| | - Bram Ramjiawan
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| |
Collapse
|
38
|
Lapchuk AS, Gorbov IV, Prygun AV, Balagura IV, Morozov YM. Combined small and large magnetic nanoparticle extraction and concentration from biofluids for non-toxic detection of biomarkers. SENSORS & DIAGNOSTICS 2022; 1:829-840. [PMID: 35923776 PMCID: PMC9280443 DOI: 10.1039/d2sd00078d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/08/2022] [Indexed: 11/21/2022]
Abstract
Sketch of the concentration of small and large magnetic nanoparticles in biofluid to the area of biomarker detection for enhancing the method sensitivity.
Collapse
Affiliation(s)
- Anatoliy S. Lapchuk
- Department of Optical Engineering, Institute for Information Recording of NAS of Ukraine, 03113 Kyiv, Ukraine
| | - Ivan V. Gorbov
- Department of Optical Engineering, Institute for Information Recording of NAS of Ukraine, 03113 Kyiv, Ukraine
| | - Alexander V. Prygun
- Department of Optical Engineering, Institute for Information Recording of NAS of Ukraine, 03113 Kyiv, Ukraine
| | - Iryna V. Balagura
- Department of Optical Engineering, Institute for Information Recording of NAS of Ukraine, 03113 Kyiv, Ukraine
| | - Yevhenii M. Morozov
- Department of Optical Engineering, Institute for Information Recording of NAS of Ukraine, 03113 Kyiv, Ukraine
- Biosensor Technologies, AIT-Austrian Institute of Technology, 3430 Tulln, Austria
| |
Collapse
|