1
|
Hassanzadeh-Barforoushi A, Tukova A, Nadalini A, Inglis DW, Chang-Hao Tsao S, Wang Y. Microfluidic-SERS Technologies for CTC: A Perspective on Clinical Translation. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38652011 DOI: 10.1021/acsami.4c01158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Enumeration and phenotypic profiling of circulating tumor cells (CTCs) provide critical information for clinical diagnosis and treatment monitoring in cancer. To achieve this goal, an integrated system is needed to efficiently isolate CTCs from patient samples and sensitively evaluate their phenotypes. Such integration would comprise a high-throughput single-cell processing unit for the isolation and manipulation of CTCs and a sensitive and multiplexed quantitation unit to detect clinically relevant signals from these cells. Surface-enhanced Raman scattering (SERS) has been used as an analytical method for molecular profiling and in vitro cancer diagnosis. More recently, its multiplexing capability and power to create distinct molecular signatures against their targets have garnered attention. Here, we share our insights into the combined power of microfluidics and SERS in realizing CTC isolation, enumeration, and detection from a clinical translation perspective. We highlight the key operational factors in CTC microfluidic processing and SERS detection from patient samples. We further discuss microfluidic-SERS integration and its clinical utility as a paradigm shift in clinical CTC-based cancer diagnosis and prognostication. Finally, we summarize the challenges and attempt to look forward to what lies ahead of us in potentially translating the technique into real clinical applications.
Collapse
Affiliation(s)
- Amin Hassanzadeh-Barforoushi
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Anastasiia Tukova
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Audrey Nadalini
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - David W Inglis
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Simon Chang-Hao Tsao
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| | - Yuling Wang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
2
|
Song Q, Liu H, Wang W, Chen C, Cao Y, Chen B, Cai B, He R. Carboxyl graphene modified PEDOT:PSS organic electrochemical transistor for in situ detection of cancer cell morphology. NANOSCALE 2024; 16:3631-3640. [PMID: 38276969 DOI: 10.1039/d3nr06190f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Circulating tumor cells in human peripheral blood play an important role in cancer metastasis. In addition to the size-based and antibody-based capture and separation of cancer cells, their electrical characterization is important for rare cell detection, which can prove fatal in point-of-care testing. Herein, an organic electrochemical transistor (OECT) biosensor made of solution-gated carboxyl graphene mixed with PEDOT:PSS for the detection of cancer cells in situ is reported. Carboxyl graphene was used in this work to modulate cancer cell morphology, which differs significantly from normal blood cells, to achieve rare cancer cell detection. When the concentration of carboxyl graphene mixed in PEDOT:PSS was increased from 0 to 5 mg mL-1, the cancer cell surface area increased from 218 μm2 to 530 μm2, respectively. A change in cell morphology was also detected by the OECT. Negative charges in the cancer cells induced a positive shift in gate voltage, which was approximately 40 mV for spherical-shaped cells. When the cell surface area increased, transfer curves of transistor revealed a negative shift in gate voltage. Therefore, the sensor can be used for in situ detection of cancer cell morphology during the cell capture process, which can be used to identify whether the captured cells are deformable.
Collapse
Affiliation(s)
- Qingyuan Song
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, China.
| | - Hongni Liu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Weiyi Wang
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, China.
| | - Chaohui Chen
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, China.
| | - Yiping Cao
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, China.
| | - Bolei Chen
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Jianghan University, Wuhan 430056, China.
| | - Bo Cai
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Jianghan University, Wuhan 430056, China.
| | - Rongxiang He
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
3
|
You Q, Liang F, Wu G, Cao F, Liu J, He Z, Wang C, Zhu L, Chen X, Yang Y. The Landscape of Biomimetic Nanovesicles in Brain Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306583. [PMID: 37713652 DOI: 10.1002/adma.202306583] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Brain diseases, such as brain tumors, neurodegenerative diseases, cerebrovascular diseases, and brain injuries, are caused by various pathophysiological changes, which pose a serious health threat. Brain disorders are often difficult to treat due to the presence of the blood-brain barrier (BBB). Biomimetic nanovesicles (BNVs), including endogenous extracellular vesicles (EVs) derived from various cells and artificial nanovesicles, possess the ability to penetrate the BBB and thus can be utilized for drug delivery to the brain. BNVs, especially endogenous EVs, are widely distributed in body fluids and usually carry various disease-related signal molecules such as proteins, RNA, and DNA, and may also be analyzed to understand the etiology and pathogenesis of brain diseases. This review covers the exhaustive classification and characterization of BNVs and pathophysiological roles involved in various brain diseases, and emphatically focuses on nanotechnology-integrated BNVs for brain disease theranostics, including various diagnosis strategies and precise therapeutic regulations (e.g., immunity regulation, disordered protein clearance, anti-neuroinflammation, neuroregeneration, angiogenesis, and the gut-brain axis regulation). The remaining challenges and future perspectives regarding the nanotechnology-integrated BNVs for the diagnosis and treatment of brain diseases are also discussed and outlined.
Collapse
Affiliation(s)
- Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Fuming Liang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing, 400016, China
| | - Gege Wu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Jingyi Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhaohui He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing, 400016, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
4
|
Xie P, Yao X, Chu Z, Yang Y, Li H, Tan S, Tang H, Zhou J, Jin W. Homoporous polydimethylsiloxane membrane microfilter for ultrafast label-free isolation and recognition of circulating tumor cells in peripheral blood. iScience 2023; 26:108246. [PMID: 38026152 PMCID: PMC10665804 DOI: 10.1016/j.isci.2023.108246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/03/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
The detection of circulating tumor cells (CTCs) in peripheral blood is a novel and accurate technique for the early diagnosis of cancers. However, this method is challenging because of the need for high collection efficiency due to the ultralow content and similar size of CTCs compared with other blood cells. To address the aforementioned issue, we proposed a homoporous polydimethylsiloxane (PDMS) membrane and its microfilter device to perform the ultrafast isolation and identification of CTCs directly from peripheral blood without any labeling treatment. The membrane pores can be homogenously controlled at a size of 6.3 μm through the cross-linking time of PDMS during a filtration-coating strategy. Within only 10 s, the designed device achieved a retention rate greater than 70% for pancreatic cancer cells, and it exhibited excellent cell compatibility to support cell proliferation. The isolated CTCs on this membrane can be easily observed and identified using a fluorescence microscope.
Collapse
Affiliation(s)
- Peng Xie
- Department of Hepatopancreatobiliary Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Xiaoyue Yao
- State Key Laboratory of Materials–Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing 211816, P.R. China
| | - Zhenyu Chu
- State Key Laboratory of Materials–Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing 211816, P.R. China
| | - Yang Yang
- Department of Hepatopancreatobiliary Surgery, Zhongda Hospital Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Haifeng Li
- Department of Hepatopancreatobiliary Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Siyuan Tan
- Department of Hepatopancreatobiliary Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Haodong Tang
- Department of Hepatopancreatobiliary Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Jiahua Zhou
- Department of Hepatopancreatobiliary Surgery, Zhongda Hospital Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Wanqin Jin
- State Key Laboratory of Materials–Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing 211816, P.R. China
| |
Collapse
|
5
|
Pedrosa VA, Chen K, George TJ, Fan ZH. Gold Nanoparticle-Based Microfluidic Chips for Capture and Detection of Circulating Tumor Cells. BIOSENSORS 2023; 13:706. [PMID: 37504105 PMCID: PMC10377447 DOI: 10.3390/bios13070706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/24/2023] [Accepted: 07/01/2023] [Indexed: 07/29/2023]
Abstract
Liquid biopsy has progressed to its current use to diagnose and monitor cancer. Despite the recent advances in investigating cancer detection and diagnosis strategies, there is still a room for improvements in capturing CTCs. We developed an efficient CTC detection system by integrating gold nanoparticles with a microfluidic platform, which can achieve CTC capture within 120 min. Here, we report our development of a simple and effective way to isolate CTCs using antibodies attached on gold nanoparticles to the surface of a lateral filter array (LFA) microdevice. Our method was optimized using three pancreatic tumor cell lines, enabling the capture with high efficiency (90% ± 3.2%). The platform was further demonstrated for isolating CTCs from patients with metastatic pancreatic cancer. Our method and platform enables the production of functionalized, patterned surfaces that interact with tumor cells, enhancing the selective capture of CTCs for biological assays.
Collapse
Affiliation(s)
- Valber A Pedrosa
- Institute of Bioscience of Botucatu, Sao Paulo State University-Unesp, Botucatu 18603-560, Brazil
| | - Kangfu Chen
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA
| | - Thomas J George
- Department of Medicine, University of Florida, P.O. Box 100278, Gainesville, FL 32610, USA
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL 32611, USA
| |
Collapse
|
6
|
Qi M, Ruan M, Liang J, Zhang Z, Chen C, Cao Y, He R. Three-Dimensional PLGA Nanofiber-Based Microchip for High-Efficiency Cancer Cell Capture. MATERIALS (BASEL, SWITZERLAND) 2023; 16:3065. [PMID: 37109900 PMCID: PMC10144435 DOI: 10.3390/ma16083065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 06/19/2023]
Abstract
A 3D network capture substrate based on poly(lactic-co-glycolic acid) (PLGA) nanofibers was studied and successfully used for high-efficiency cancer cell capture. The arc-shaped glass micropillars were prepared by chemical wet etching and soft lithography. PLGA nanofibers were coupled with micropillars by electrospinning. Given the size effect of the microcolumn and PLGA nanofibers, a three-dimensional of micro-nanometer spatial network was prepared to form a network cell trapping substrate. After the modification of a specific anti-EpCAM antibody, MCF-7 cancer cells were captured successfully with a capture efficiency of 91%. Compared with the substrate composed of 2D nanofibers or nanoparticles, the developed 3D structure based on microcolumns and nanofibers had a greater contact probability between cells and the capture substrate, leading to a high capture efficiency. Cell capture based on this method can provide technical support for rare cells in peripheral blood detection, such as circulating tumor cells and circulating fetal nucleated red cells.
Collapse
|
7
|
Sun N, Zhang C, Wang J, Yue X, Kim HY, Zhang RY, Liu H, Widjaja J, Tang H, Zhang TX, Ye J, Qian A, Liu C, Wu A, Wang K, Johanis M, Yang P, Liu H, Meng M, Liang L, Pei R, Chai-Ho W, Zhu Y, Tseng HR. Hierarchical integration of DNA nanostructures and NanoGold onto a microchip facilitates covalent chemistry-mediated purification of circulating tumor cells in head and neck squamous cell carcinoma. NANO TODAY 2023; 49:101786. [PMID: 38037608 PMCID: PMC10688595 DOI: 10.1016/j.nantod.2023.101786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
It is well-established that the combined use of nanostructured substrates and immunoaffinity agents can enhance the cell-capture performance of the substrates, thus offering a practical solution to effectively capture circulating tumor cells (CTCs) in peripheral blood. Developing along this strategy, this study first demonstrated a top-down approach for the fabrication of tetrahedral DNA nanostructure (TDN)-NanoGold substrates through the hierarchical integration of three functional constituents at various length-scales: a macroscale glass slide, sub-microscale self-organized NanoGold, and nanoscale self-assembled TDN. The TDN-NanoGold substrates were then assembled with microfluidic chaotic mixers to give TDN-NanoGold Click Chips. In conjunction with the use of copper (Cu)-catalyzed azide-alkyne cycloaddition (CuAAC)-mediated CTC capture and restriction enzyme-triggered CTC release, TDN-NanoGold Click Chips allow for effective enumeration and purification of CTCs with intact cell morphologies and preserved molecular integrity. To evaluate the clinical utility of TDN-NanoGold Click Chips, we used these devices to isolate and purify CTCs from patients with human papillomavirus (HPV)-positive (+) head and neck squamous cell carcinoma (HNSCC). The purified HPV(+) HNSCC CTCs were then subjected to RT-ddPCR testing, allowing for detection of E6/E7 oncogenes, the characteristic molecular signatures of HPV(+) HNSCC. We found that the resulting HPV(+) HNSCC CTC counts and E6/E7 transcript copy numbers are correlated with the treatment responses in the patients, suggesting the potential clinical utility of TDN-NanoGold Click Chips for non-invasive diagnostic applications of HPV(+) HNSCC.
Collapse
Affiliation(s)
- Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou 215123, China
| | - Ceng Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jing Wang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xinmin Yue
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Hyo Yong Kim
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ryan Y. Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hongtao Liu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong 250014, China
| | - Josephine Widjaja
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hubert Tang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tiffany X. Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jinglei Ye
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Audrey Qian
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chensong Liu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alex Wu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Katharina Wang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael Johanis
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peng Yang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Honggang Liu
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Meng Meng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, Guangdong Province, China
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou 215123, China
| | - Wanxing Chai-Ho
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
8
|
Mao Y, Zhang Y, Yu Y, Zhu N, Zhou X, Li G, Yi Q, Wu Y. Self-assembled supramolecular immunomagnetic nanoparticles through π-π stacking strategy for the enrichment of circulating tumor cells. Regen Biomater 2023; 10:rbad016. [PMID: 37020751 PMCID: PMC10070042 DOI: 10.1093/rb/rbad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Owing to their high-specific binding toward targets as well as fast and convenient separation operations, immunomagnetic beads (IMBs) are widely used in the capture and detection of circulating tumor cells (CTCs). To construct the IMBs, surface modifications are generally performed to functionalize the magnetic cores (e.g. Fe3O4 nanoparticles), and the employed surface modification strategies normally influence the structure and functions of the prepared IMBs in return. Different from the existing work, we proposed the use of supramolecular layer-by-layer (LBL) self-assembly strategy to construct the IMBs. In general, owing to the π-π stacking interactions, the polydopamine, graphene oxide and 'molecular glue' γ-oxo-1-pyrenebutyric acid were self-assembled on Fe3O4 nanoparticles sequentially, thereby accomplishing the integration of different functional components onto magnetic cores to prepare the self-assembled supramolecular immunomagnetic beads (ASIMBs). The ASIMBs showed high sensitivity, specificity and good biocompatibility to the model CTCs and low nonspecific adsorption to the negative cells (∼93% for MCF-7 cells and 17% for Jurkat cells). Meanwhile, ASIMBs possessed a remarkable potential to screen the rare MCF-7 cells out of large amounts of interfering Jurkat cells with the capture efficiency of 75-100% or out of mouse whole blood with the capture efficiency of 20-90%. The captured cells can be further recultured directly without any more treatment, which showed huge applicability of the ASIMBs for in vitro detection in clinical practices.
Collapse
Affiliation(s)
- Yanchao Mao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Yujia Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Yue Yu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Nanhang Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Xiaoxi Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Guohao Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Qiangying Yi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| |
Collapse
|
9
|
Li J, Dong C, Gan H, Gu X, Zhang J, Zhu Y, Xiong J, Song C, Wang L. Nondestructive separation/enrichment and rolling circle amplification-powered sensitive SERS enumeration of circulating tumor cells via aptamer recognition. Biosens Bioelectron 2023; 231:115273. [PMID: 37054599 DOI: 10.1016/j.bios.2023.115273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Nondestructive separation/enrichment and reliable detection of extremely rare circulating tumor cells (CTCs) in peripheral blood are of considerable importance in tumor precision diagnosis and treatment, yet this remains a big challenge. Herein, a novel strategy for nondestructive separation/enrichment and ultra-sensitive surface-enhanced Raman scattering (SERS)-based enumeration of CTCs is proposed via aptamer recognition and rolling circle amplification (RCA). In this work the magnetic beads modified with "Aptamer (Apt)-Primer" (AP) probes were utilized to specifically capture CTCs, and then after magnetic separation/enrichment, the RCA-powered SERS counting and benzonase nuclease cleavage-assisted nondestructive release of CTCs were realized, respectively. The AP was assembled by hybridizing the EpCAM-specific aptamer with a primer, and the optimal AP contains 4 mismatched bases. The RCA enhanced SERS signal nearly 4.5-fold, and the SERS strategy has good specificity, uniformity and reproducibility. The proposed SERS detection possesses a good linear relationship with the concentration of MCF-7 cells spiked in PBS with the limit of detection (LOD) of 2 cells/mL, which shows good potential practicality for detecting CTCs in blood with recoveries ranging from 100.56% to 116.78%. Besides, the released CTCs remained good cellular activity with the normal proliferation after re-culture for 48 h and normal growth for at least three generations. The proposed strategy of nondestructive separation/enrichment and SERS-based sensitive enumeration is promising for reliable analysis of EpCAM-positive CTCs in blood, which is expected to provide a powerful tool for analysis of extremely rare circulating tumor cells in complex peripheral blood for liquid biopsy.
Collapse
|
10
|
Isolation, Detection and Analysis of Circulating Tumour Cells: A Nanotechnological Bioscope. Pharmaceutics 2023; 15:pharmaceutics15010280. [PMID: 36678908 PMCID: PMC9864919 DOI: 10.3390/pharmaceutics15010280] [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: 11/09/2022] [Revised: 12/17/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Cancer is one of the dreaded diseases to which a sizeable proportion of the population succumbs every year. Despite the tremendous growth of the health sector, spanning diagnostics to treatment, early diagnosis is still in its infancy. In this regard, circulating tumour cells (CTCs) have of late grabbed the attention of researchers in the detection of metastasis and there has been a huge surge in the surrounding research activities. Acting as a biomarker, CTCs prove beneficial in a variety of aspects. Nanomaterial-based strategies have been devised to have a tremendous impact on the early and rapid examination of tumor cells. This review provides a panoramic overview of the different nanotechnological methodologies employed along with the pharmaceutical purview of cancer. Initiating from fundamentals, the recent nanotechnological developments toward the detection, isolation, and analysis of CTCs are comprehensively delineated. The review also includes state-of-the-art implementations of nanotechnological advances in the enumeration of CTCs, along with future challenges and recommendations thereof.
Collapse
|
11
|
Prasher P, Sharma M, Singh SK, Gulati M, Chellappan DK, Zacconi F, De Rubis G, Gupta G, Sharifi-Rad J, Cho WC, Dua K. Luteolin: a flavonoid with a multifaceted anticancer potential. Cancer Cell Int 2022; 22:386. [PMID: 36482329 PMCID: PMC9730645 DOI: 10.1186/s12935-022-02808-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Therapeutic effect of phytochemicals has been emphasized in the traditional medicine owing to the presence of bioactive molecules, such as polyphenols. Luteolin is a flavone belonging to the flavonoid class of polyphenolic phytochemicals with healing effect on hypertension, inflammatory disorders, and cancer due to its action as pro-oxidants and antioxidants. The anticancer profile of luteolin is of interest due to the toxic effect of contemporary chemotherapy paradigm, leading to the pressing need for the development and identification of physiologically benevolent anticancer agents and molecules. Luteolin exerts anticancer activity by downregulation of key regulatory pathways associated with oncogenesis, in addition to the induction of oxidative stress, cell cycle arrest, upregulation of apoptotic genes, and inhibition of cell proliferation and angiogenesis in cancer cells. In this review, we discuss about the anticancer profile of luteolin.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun, 248007 India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, 248007 India
| | - Sachin Kumar Singh
- School of Pharmacy and Pharmaceutical Science, Lovely Professional University, Phagwara, India
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007 Australia
| | - Monica Gulati
- School of Pharmacy and Pharmaceutical Science, Lovely Professional University, Phagwara, India
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007 Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Flavia Zacconi
- Departamento de Quimica Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuna Mackenna 4860, Macul, 7820436 Santiago, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, 7820436 Santiago, Chile
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007 Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong China
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007 Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| |
Collapse
|
12
|
Zhao K, Zhao P, Dong J, Wei Y, Chen B, Wang Y, Pan X, Wang J. Implementation of an Integrated Dielectrophoretic and Magnetophoretic Microfluidic Chip for CTC Isolation. BIOSENSORS 2022; 12:bios12090757. [PMID: 36140142 PMCID: PMC9496341 DOI: 10.3390/bios12090757] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022]
Abstract
Identification of circulating tumor cells (CTCs) from a majority of various cell pools has been an appealing topic for diagnostic purposes. This study numerically demonstrates the isolation of CTCs from blood cells by the combination of dielectrophoresis and magnetophoresis in a microfluidic chip. Taking advantage of the label-free property, the separation of red blood cells, platelets, T cells, HT-29, and MDA-231 was conducted in the microchannel. By using the ferromagnet structure with double segments and a relatively shorter distance in between, a strong gradient of the magnetic field, i.e., sufficiently large MAP forces acting on the cells, can be generated, leading to a high separation resolution. In order to generate strong DEP forces, the non-uniform electric field gradient is induced by applying the electric voltage through the microchannel across a pair of asymmetric orifices, i.e., a small orifice and a large orifice on the opposite wall of the channel sides. The distribution of the gradient of the magnetic field near the edge of ferromagnet segments, the gradient of the non-uniform electric field in the vicinity of the asymmetric orifices, and the flow field were investigated. In this numerical simulation, the effects of the ferromagnet structure on the magnetic field, the flow rate, as well as the strength of the electric field on their combined magnetophoretic and dielectrophoretic behaviors and trajectories are systemically studied. The simulation results demonstrate the potential of both property- and size-based cell isolation in the microfluidic device by implementing magnetophoresis and dielectrophoresis.
Collapse
Affiliation(s)
- Kai Zhao
- Liaoning Key Laboratory of Marine Sensing and Intelligent Detection, Dalian Maritime University, Dalian 116026, China
- Department of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Penglu Zhao
- Liaoning Key Laboratory of Marine Sensing and Intelligent Detection, Dalian Maritime University, Dalian 116026, China
- Department of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Jianhong Dong
- Liaoning Key Laboratory of Marine Sensing and Intelligent Detection, Dalian Maritime University, Dalian 116026, China
- Department of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Yunman Wei
- Liaoning Key Laboratory of Marine Sensing and Intelligent Detection, Dalian Maritime University, Dalian 116026, China
- Department of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Bin Chen
- Liaoning Key Laboratory of Marine Sensing and Intelligent Detection, Dalian Maritime University, Dalian 116026, China
- Department of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Yanjuan Wang
- Liaoning Key Laboratory of Marine Sensing and Intelligent Detection, Dalian Maritime University, Dalian 116026, China
- Software Institute, Dalian Jiaotong University, Dalian 116028, China
| | - Xinxiang Pan
- Liaoning Key Laboratory of Marine Sensing and Intelligent Detection, Dalian Maritime University, Dalian 116026, China
- Department of Maritime, Guangdong Ocean University, Zhanjiang 524000, China
| | - Junsheng Wang
- Liaoning Key Laboratory of Marine Sensing and Intelligent Detection, Dalian Maritime University, Dalian 116026, China
- Department of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
- Correspondence:
| |
Collapse
|
13
|
Tian R, Li X, Zhang H, Ma L, Zhang H, Wang Z. Ulex Europaeus Agglutinin-I-Based Magnetic Isolation for the Efficient and Specific Capture of SW480 Circulating Colorectal Tumor Cells. ACS OMEGA 2022; 7:30405-30411. [PMID: 36061664 PMCID: PMC9435041 DOI: 10.1021/acsomega.2c03702] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
The efficient and specific capture of circulating tumor cells (CTCs) from patients' peripheral blood is of significant value in precise cancer diagnosis and cancer therapy. As fine targeting molecules, lectins can recognize cancer cells specifically due to the abnormal glycosylation of molecules on the cancer cell membrane and the specific binding of lectin with glycoconjugates. Herein, a Ulex europaeus agglutinin-I (UEA-I)-based magnetic isolation strategy was developed to efficiently and specifically capture α-1,2-fucose overexpression CTCs from colorectal cancer (CRC) patients' peripheral blood. Using UEA-I-modified Fe3O4 magnetic beads (termed MB-UEA-I), up to 94 and 89% of target cells (i.e., SW480 CRC cells) were captured from the cell spiking complete cell culture medium and whole blood, respectively. More than 90% of captured cells show good viability and proliferation ability without detaching from MB-UEA-I. In combination with three-color immunocytochemistry (ICC) identification, MB-UEA-I has been successfully used to capture CTCs from CRC patients' peripheral blood. The experimental results indicate a correlation between CTC characterization and tumor metastasis. Specifically, MB-UEA-I can be applied to screen early CRC by capturing CTCs when served as a liquid biopsy. The presented work offers a new insight into developing cost-effective lectin-functionalized methods for biomedical applications.
Collapse
Affiliation(s)
- Rongrong Tian
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- University
of Science and Technology of China, Jinzhai Road, Baohe District, Hefei, Anhui 230026, P. R. China
| | - Xiaodong Li
- Department
of Radiology, The First Hospital of Jilin
University, Changchun, Jilin 130021, P. R. China
| | - Hua Zhang
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Lina Ma
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Huimao Zhang
- Department
of Radiology, The First Hospital of Jilin
University, Changchun, Jilin 130021, P. R. China
| | - Zhenxin Wang
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- University
of Science and Technology of China, Jinzhai Road, Baohe District, Hefei, Anhui 230026, P. R. China
| |
Collapse
|
14
|
Ju S, Chen C, Zhang J, Xu L, Zhang X, Li Z, Chen Y, Zhou J, Ji F, Wang L. Detection of circulating tumor cells: opportunities and challenges. Biomark Res 2022; 10:58. [PMID: 35962400 PMCID: PMC9375360 DOI: 10.1186/s40364-022-00403-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Circulating tumor cells (CTCs) are cells that shed from a primary tumor and travel through the bloodstream. Studying the functional and molecular characteristics of CTCs may provide in-depth knowledge regarding highly lethal tumor diseases. Researchers are working to design devices and develop analytical methods that can capture and detect CTCs in whole blood from cancer patients with improved sensitivity and specificity. Techniques using whole blood samples utilize physical prosperity, immunoaffinity or a combination of the above methods and positive and negative enrichment during separation. Further analysis of CTCs is helpful in cancer monitoring, efficacy evaluation and designing of targeted cancer treatment methods. Although many advances have been achieved in the detection and molecular characterization of CTCs, several challenges still exist that limit the current use of this burgeoning diagnostic approach. In this review, a brief summary of the biological characterization of CTCs is presented. We focus on the current existing CTC detection methods and the potential clinical implications and challenges of CTCs. We also put forward our own views regarding the future development direction of CTCs.
Collapse
Affiliation(s)
- Siwei Ju
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Cong Chen
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Jiahang Zhang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Lin Xu
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Xun Zhang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Zhaoqing Li
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Yongxia Chen
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Jichun Zhou
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Feiyang Ji
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China.
| | - Linbo Wang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China.
| |
Collapse
|
15
|
Chang TK, Tung PC, Lee MJ, Lee W. A liquid-crystal aptasensing platform for label-free detection of a single circulating tumor cell. Biosens Bioelectron 2022; 216:114607. [PMID: 35969962 DOI: 10.1016/j.bios.2022.114607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022]
Abstract
Circulating tumor cells (CTCs), which are shed from a primary site into the bloodstream and lead to distal metastases, are pivotal as a prognostic marker for evaluating the treatment response of cancer patients. One of the major challenges of detecting CTCs is their scarcity in blood. We report herein a label-free liquid crystal (LC) cytosensor by adopting an aptamer against epithelial cell adhesion molecule (EpCAM) to capture EpCAM-positive cancer cells. The optical and dielectric signals transduced from the interaction between LC and different numbers of captured breast cancer cells were investigated. A limit of detection (LOD) of 5 CTCs was resulted from the optical biosensing approach relying on texture observation and image analysis of the optical signal in polarizing micrographs. The LOD was further lowered to a single CTC in the dielectric approach by studying the real- and imaginary-part dielectric constants of LC at 1 kHz and 30 Hz as well as the relaxation frequency. The LC-based EpCAM-specific dielectric cytosensor was successfully applied to single-cell CTC detection in cancer cell-spiked human serum and whole blood. This platform demonstrates the potential of LC-based biosensing technologies in cellular-level detection and quantitation, which is crucial to the early diagnosis of cancer metastasis and progression.
Collapse
Affiliation(s)
- Tsung-Keng Chang
- College of Photonics, National Yang Ming Chiao Tung University, Guiren Dist, Tainan, 711010, Taiwan; National Laboratory Animal Center, National Applied Research Laboratories, Taipei, 115202, Taiwan
| | - Pei-Chi Tung
- Department of Bioscience Technology, Chang Jung Christian University, Guiren Dist, Tainan, 711301, Taiwan
| | - Mon-Juan Lee
- Department of Bioscience Technology, Chang Jung Christian University, Guiren Dist, Tainan, 711301, Taiwan; Department of Medical Science Industries, Chang Jung Christian University, Guiren Dist, Tainan, 711301, Taiwan.
| | - Wei Lee
- College of Photonics, National Yang Ming Chiao Tung University, Guiren Dist, Tainan, 711010, Taiwan; Institute of Imaging and Biomedical Photonics, College of Photonics, National Yang Ming Chiao Tung University, Guiren Dist, Tainan, 711010, Taiwan.
| |
Collapse
|
16
|
Liu X, Chen W, Zhao D, Liu X, Wang Y, Chen Y, Ma X. Enzyme-Powered Hollow Nanorobots for Active Microsampling Enabled by Thermoresponsive Polymer Gating. ACS NANO 2022; 16:10354-10363. [PMID: 35816232 DOI: 10.1021/acsnano.2c00401] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Achieving molecular sample capture at micro/nanoscales while integrating functions of controllable loading and real-time monitoring of cargo molecules is of great significance in the development of intelligent micro/nanorobots. Herein, we prepare a temperature-responsive microsampling nanorobot by encapsulating metal (Au) nanodots inside hollow mesoporous silica nanoparticles and grafting a temperature-responsive polymer, poly(N-isopropylacrylamide), on their external surface. The molecular gate of nanochannels accessing the internal hollow reservoir can be switched between "open" and "closed" states by regulating the temperature, allowing on-demand loading and releasing of small molecules. The internally embedded surface-enhanced Raman scattering hotspots of gold nanodots can serve as sensing probes for real-time detection of the molecular cargo load inside the hollow nanorobots. Furthermore, we demonstrate temperature-dependent self-propulsion behavior of the nanorobots driven by enzymatic reactions. The active motion behavior can favorably regulate the loading efficiency of molecular cargos. In addition, by further introducing the magnetic component Ni, the nanorobots can accomplish effective transportation of cargo molecules by magnetic guidance under real-time Raman monitoring. The current strategy is expected to provide a manipulable nanorobot platform for precise biomedical sampling, which holds promising potential for disease diagnosis or controlled drug delivery in precision medicine.
Collapse
Affiliation(s)
- Xiaojia Liu
- Sauvage Laboratory for Smart Materials, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| | - Wenjun Chen
- Sauvage Laboratory for Smart Materials, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| | - Dongfang Zhao
- Sauvage Laboratory for Smart Materials, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| | - Xiaoxia Liu
- Sauvage Laboratory for Smart Materials, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| | - Yong Wang
- Sauvage Laboratory for Smart Materials, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| | - Yuduo Chen
- Sauvage Laboratory for Smart Materials, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| | - Xing Ma
- Sauvage Laboratory for Smart Materials, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| |
Collapse
|
17
|
Kwizera EA, Ou W, Lee S, Stewart S, Shamul JG, Xu J, Tait N, Tkaczuk KHR, He X. Greatly Enhanced CTC Culture Enabled by Capturing CTC Heterogeneity Using a PEGylated PDMS-Titanium-Gold Electromicrofluidic Device with Glutathione-Controlled Gentle Cell Release. ACS NANO 2022; 16:11374-11391. [PMID: 35797466 PMCID: PMC9649890 DOI: 10.1021/acsnano.2c05195] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The circulating tumor cells (CTCs, the root cause of cancer metastasis and poor cancer prognosis) are very difficult to culture for scale-up in vitro, which has hampered their use in cancer research/prognosis and patient-specific therapeutic development. Herein, we report a robust electromicrofluidic chip for not only efficient capture of heterogeneous (EpCAM+ and CD44+) CTCs with high purity but also glutathione-controlled gentle release of the CTCs with high efficiency and viability. This is enabled by coating the polydimethylsiloxane (PDMS) surface in the device with a 10 nm gold layer through a 4 nm titanium coupling layer, for convenient PEGylation and linkage of capture antibodies via the thiol-gold chemistry. Surprisingly, the percentage of EpCAM+ mammary CTCs can be as low as ∼35% (∼70% on average), showing that the commonly used approach of capturing CTCs with EpCAM alone may miss many EpCAM- CTCs. Furthermore, the CD44+ CTCs can be cultured to form 3D spheroids efficiently for scale-up. In contrast, the CTCs captured with EpCAM alone are poor in proliferation in vitro, consistent with the literature. By capture of the CTC heterogeneity, the percentage of stage IV patients whose CTCs can be successfully cultured/scaled up is improved from 12.5% to 68.8%. These findings demonstrate that the common practice of CTC capture with EpCAM alone misses the CTC heterogeneity including the critical CD44+ CTCs. This study may be valuable to the procurement and scale-up of heterogeneous CTCs, to facilitate the understanding of cancer metastasis and the development of cancer metastasis-targeted personalized cancer therapies conveniently via the minimally invasive liquid/blood biopsy.
Collapse
Affiliation(s)
- Elyahb A Kwizera
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Sojeong Lee
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Samantha Stewart
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Jiangsheng Xu
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Nancy Tait
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| | - Katherine H R Tkaczuk
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| |
Collapse
|
18
|
Bioprobes-regulated precision biosensing of exosomes: From the nanovesicle surface to the inside. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214538] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
19
|
Carvalho Â, Guimarães-Teixeira C, Constâncio V, Fernandes M, Macedo-Silva C, Henrique R, Monteiro FJ, Jerónimo C. One sample fits all: a microfluidic-assisted methodology for label-free isolation of CTCs with downstream methylation analysis of cfDNA in lung cancer. Biomater Sci 2022; 10:3296-3308. [PMID: 35583893 DOI: 10.1039/d2bm00044j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Lung cancer (LC) is a major cause of mortality. Late diagnosis, associated with limitations in tissue biopsies for adequate tumor characterization contribute to limited survival of lung cancer patients. Liquid biopsies have been introduced to improve tumor characetrization through the analysis of biomarkers, including circulating tumour cells (CTCs) and cell-free DNA (cfDNA). Considering their availability in blood, several enrichment strategies have been developed to augment circulating biomarkers for improving diagnostic, prognostic and treament efficacy assessment; often, however, only one biomarker is tested. In this work we developed and implemented a microfluidic chip for label-free enrichment of CTCs with a methodology for subsequent cfDNA analysis from the same cryopreserved sample. CTCs were successfully isolated in 38 of 42 LC patients with the microfluidic chip. CTCs frequency was significantly higher in LC patients with advanced disease. A cut-off of 1 CTC per mL was established for diagnosis (sensitivity = 76.19%, specificity = 100%) and in patients with late stage lung cancer, the presence of ≥5 CTCs per mL was significantly associated with shorter overall survival. MIR129-2me and ADCY4me panel of cfDNA methylation performed well for LC detection, whereas MIR129-2me combined with HOXA11me allowed for patient risk stratification. Analysis of combinations of biomarkers enabled the definition of panels for LC diagnosis and prognosis. Overall, this study demonstrates that multimodal analysis of tumour biomarkers via microfluidic devices may significantly improve LC characterization in cryopreserved samples, constituting a reliable source for continuous disease monitoring.
Collapse
Affiliation(s)
- Ângela Carvalho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal. .,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Catarina Guimarães-Teixeira
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Vera Constâncio
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Mariana Fernandes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal. .,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Catarina Macedo-Silva
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Rui Henrique
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Fernando Jorge Monteiro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal. .,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.,Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e Materiais, Universidade do Porto, Rua Dr Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Carmen Jerónimo
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| |
Collapse
|
20
|
Zhang Y, Li Y, Tan Z. A review of enrichment methods for circulating tumor cells: from single modality to hybrid modality. Analyst 2021; 146:7048-7069. [PMID: 34709247 DOI: 10.1039/d1an01422f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Circulating tumor cell (CTC) analysis as a liquid biopsy can be used for early diagnosis of cancer, evaluating cancer progression, and assessing treatment efficacy. The enrichment of CTCs from patient blood is important for CTC analysis due to the extreme rarity of CTCs. This paper updates recent advances in CTC enrichment methods. We first review single-modality methods, including biophysical and biochemical methods. Hybrid-modality methods, combining at least two single-modality methods, are gaining increasing popularity for their improved performance. Then this paper reviews hybrid-modality methods, which are categorized into integrated and sequenced hybrid-modality methods. The state of the art indicates that the CTC capture efficiencies of integrated hybrid-modality methods can reach 85% or higher by taking advantage of the superimposed and enhanced capture effects from multiple single-modality methods. Moreover, a hybrid method integrating biophysical with biochemical methods is characterized by both high processing rate and high specificity.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, Canada N2L 3G1.
| | - Yifu Li
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, Canada N2L 3G1.
| | - Zhongchao Tan
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, Canada N2L 3G1.
| |
Collapse
|
21
|
Zhong H, Yuan C, He J, Yu Y, Jin Y, Huang Y, Zhao R. Engineering Peptide-Functionalized Biomimetic Nanointerfaces for Synergetic Capture of Circulating Tumor Cells in an EpCAM-Independent Manner. Anal Chem 2021; 93:9778-9787. [PMID: 34228920 DOI: 10.1021/acs.analchem.1c01254] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Broad-spectrum detection and long-term monitoring of circulating tumor cells (CTCs) remain challenging due to the extreme rarity, heterogeneity, and dynamic nature of CTCs. Herein, a dual-affinity nanostructured platform was developed for capturing different subpopulations of CTCs and monitoring CTCs during treatment. Stepwise assembly of fibrous scaffolds, a ligand-exchangeable spacer, and a lysosomal protein transmembrane 4 β (LAPTM4B)-targeting peptide creates biomimetic, stimuli-responsive, and multivalent-binding nanointerfaces, which enable harvest of CTCs directly from whole blood with high yield, purity, and viability. The stable overexpression of the target LAPTM4B protein in CTCs and the enhanced peptide-protein binding facilitate the capture of rare CTCs in patients at an early stage, detection of both epithelial-positive and nonepithelial CTCs, and tracking of therapeutic responses. The reversible release of CTCs allows downstream molecular analysis and identification of specific liver cancer genes. The consistency of the information with clinical diagnosis presents the prospect of this platform for early diagnosis, metastasis prediction, and prognosis assessment.
Collapse
Affiliation(s)
- Huifei Zhong
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunwang Yuan
- Center of Interventional Oncology and Liver Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Jiayuan He
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Yu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yulong Jin
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Enkhbat M, Liu Y, Kim J, Xu Y, Yin Z, Liu T, Deng C, Zou C, Xie X, Li X, Wang P. Expansion of Rare Cancer Cells into Tumoroids for Therapeutic Regimen and Cancer Therapy. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Myagmartsend Enkhbat
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation Shenzhen Institute of Advanced Technology Chinese Academy of Sciences Shenzhen Guangdong 518055 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Yung‐Chiang Liu
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation Shenzhen Institute of Advanced Technology Chinese Academy of Sciences Shenzhen Guangdong 518055 China
| | - Jua Kim
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation Shenzhen Institute of Advanced Technology Chinese Academy of Sciences Shenzhen Guangdong 518055 China
| | - Yanshan Xu
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation Shenzhen Institute of Advanced Technology Chinese Academy of Sciences Shenzhen Guangdong 518055 China
| | - Zongyi Yin
- Department of Hepatobiliary Surgery General Hospital of Shenzhen University Guangdong 518055 China
| | - Tzu‐Ming Liu
- Cancer Center, Faculty of Health Sciences University of Macau Macao 999078 China
| | - Chu‐Xia Deng
- Cancer Center, Faculty of Health Sciences University of Macau Macao 999078 China
| | - Chang Zou
- The First Affiliated Hospital of Southern University Shenzhen People's Hospital Shenzhen Guangdong 518020 China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies School of Electronics and Information Technology Sun Yat‐sen University Guangzhou 510275 China
| | - Xiaowu Li
- Department of Hepatobiliary Surgery General Hospital of Shenzhen University Guangdong 518055 China
| | - Peng‐Yuan Wang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation Shenzhen Institute of Advanced Technology Chinese Academy of Sciences Shenzhen Guangdong 518055 China
- Department of Chemistry and Biotechnology Swinburne University of Technology Victoria 3122 Australia
| |
Collapse
|
23
|
Hassan S, Blick T, Thompson EW, Williams ED. Diversity of Epithelial-Mesenchymal Phenotypes in Circulating Tumour Cells from Prostate Cancer Patient-Derived Xenograft Models. Cancers (Basel) 2021; 13:cancers13112750. [PMID: 34206049 PMCID: PMC8198708 DOI: 10.3390/cancers13112750] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/29/2021] [Accepted: 04/12/2021] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Spread of prostate cancer to other parts of the body is responsible for the majority of deaths. Tumour cell epithelial mesenchymal plasticity (EMP) increases their metastatic potential and facilitates their survival in the blood as circulating tumour cells (CTCs). The aim of this study was to molecularly characterise CTCs in a panel of prostate cancer patient-derived xenografts using genes associated with epithelial and mesenchymal phenotypes, and to compare the EMP status of CTCs with their matched primary tumours. The study highlights high heterogeneity in CTC enumeration and EMP gene expression between tumour-bearing mice and within individual blood samples, and therefore caution should be taken when interpreting pooled CTC analyses. Critically, tumour cells were present in the epithelial-mesenchymal hybrid state in the circulation. The study also demonstrates that there is high variation in CTC size, which would introduce sample bias to size-based CTC isolation techniques. Abstract Metastasis is the leading cause of cancer-related deaths worldwide. The epithelial-mesenchymal plasticity (EMP) status of primary tumours has relevance to metastatic potential and therapy resistance. Circulating tumour cells (CTCs) provide a window into the metastatic process, and molecular characterisation of CTCs in comparison to their primary tumours could lead to a better understanding of the mechanisms involved in the metastatic cascade. In this study, paired blood and tumour samples were collected from four prostate cancer patient-derived xenograft (PDX) models (BM18, LuCaP70, LuCaP96, LuCaP105) and assessed using an EMP-focused, 42 gene human-specific, nested quantitative RT-PCR assay. CTC burden varied amongst the various xenograft models with LuCaP96 having the highest number of CTCs per mouse (mean: 704; median: 31) followed by BM18 (mean: 101; median: 21), LuCaP70 (mean: 73; median: 16) and LuCaP105 (mean: 57; median: 6). A significant relationship was observed between tumour size and CTC number (p = 0.0058). Decreased levels of kallikrein-related peptidase 3 (KLK3) mRNA (which encodes prostate-specific antigen; PSA) were observed in CTC samples from all four models compared to their primary tumours. Both epithelial- and mesenchymal-associated genes were commonly expressed at higher levels in CTCs compared to the bulk primary tumour, although some common EMT-associated genes (CDH1, VIM, EGFR, EPCAM) remained unchanged. Immunofluorescence co-staining for pan-cytokeratin (KRT) and vimentin (VIM) indicated variable proportions of CTCs across the full EMP axis, even in the same model. EMP hybrids predominated in the BM18 and LuCaP96 models, but were not detected in the LuCaP105 model, and variable numbers of KRT+ and human VIM+ cells were observed in each model. SERPINE1, which encodes plasminogen activator inhibitor-1 (PAI-1), was enriched at the RNA level in CTCs compared to primary tumours and was the most commonly expressed mesenchymal gene in the CTCs. Co-staining for SERPINE1 and KRT revealed SERPINE1+ cells in 7/11 samples, six of which had SERPINE+KRT+ CTCs. Cell size variation was observed in CTCs. The majority of samples (8/11) contained larger CTCs ranging from 15.3 to 37.8 µm, whilst smaller cells (10.7 ± 4.1 µm, similar in size to peripheral blood mononuclear cells (PBMCs)) were identified in 6 of 11 samples. CTC clusters were also identified in 9/11 samples, containing 2–100 CTCs per cluster. Where CTC heterogeneity was observed in the clusters, epithelial-like cells (KRT+VIM−) were located on the periphery of the cluster, forming a layer around hybrid (KRT+VIM+) or mesenchymal-like (KRT−VIM+) cells. The CTC heterogeneity observed in these models emphasises the complexity in CTC isolation and classification and supports the increasingly recognised importance of the epithelial-mesenchymal hybrid state in cancer progression and metastasis.
Collapse
Affiliation(s)
- Sara Hassan
- Faculty of Health and Institute of Health & Biomedical Innovation (IHBI), School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane City, QLD 4000, Australia; (S.H.); (T.B.)
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Tony Blick
- Faculty of Health and Institute of Health & Biomedical Innovation (IHBI), School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane City, QLD 4000, Australia; (S.H.); (T.B.)
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Erik W. Thompson
- Faculty of Health and Institute of Health & Biomedical Innovation (IHBI), School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane City, QLD 4000, Australia; (S.H.); (T.B.)
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- Correspondence: (E.W.T.); (E.D.W.)
| | - Elizabeth D. Williams
- Faculty of Health and Institute of Health & Biomedical Innovation (IHBI), School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane City, QLD 4000, Australia; (S.H.); (T.B.)
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- Australian Prostate Cancer Research Centre—Queensland (APCRC-Q), Brisbane, QLD 4102, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD 4102, Australia
- Correspondence: (E.W.T.); (E.D.W.)
| |
Collapse
|
24
|
Yu L, Tang P, Nie C, Hou Y, Haag R. Well-Defined Nanostructured Biointerfaces: Strengthened Cellular Interaction for Circulating Tumor Cells Isolation. Adv Healthc Mater 2021; 10:e2002202. [PMID: 33943037 PMCID: PMC11468763 DOI: 10.1002/adhm.202002202] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/27/2021] [Indexed: 12/11/2022]
Abstract
The topographic features at the cell-material biointerface are critical for cellular sensing of the extracellular environment (ECM) and have gradually been recognized as key factors that regulate cell adhesion behavior. Herein, a well-defined nanostructured biointerface is fabricated via a new generation of mussel-inspired polymer coating to mimic the native ECM structures. Upon the bioinert background presence and biospecific ligands conjugation, the affinity of cancer cells to the resulting biofunctional surfaces, which integrate topographic features and biochemical cues, is greatly strengthened. Both the conjugated bioligand density, filopodia formation, and focal adhesion expression are significantly enhanced by the surficial nano-features with an optimized size-scale. Thus, this nanostructured biointerface exhibits high capture efficiency for circulating tumor cells (CTCs) with high sensitivity, high biospecificity, and high purity. Benefiting from the unique bioligands conjugation chemistry herein, the captured cancer cells can be responsively detached from the biointerfaces without damage for downstream analysis. The present biofunctional nanostructured interfaces offer a good solution to address current challenges to efficiently isolate rare CTCs from blood samples for earlier cancer diagnosis.
Collapse
Affiliation(s)
- Leixiao Yu
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 3Berlin14195Germany
| | - Peng Tang
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 3Berlin14195Germany
| | - Chuanxiong Nie
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 3Berlin14195Germany
| | - Yong Hou
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 3Berlin14195Germany
| | - Rainer Haag
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 3Berlin14195Germany
| |
Collapse
|
25
|
Shen S, Yi Z, Li X, Xie S, Jin M, Zhou G, Yan Z, Shui L. Flow-Field-Assisted Dielectrophoretic Microchips for High-Efficiency Sheathless Particle/Cell Separation with Dual Mode. Anal Chem 2021; 93:7606-7615. [PMID: 34003009 DOI: 10.1021/acs.analchem.1c00018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Prefocusing of cell mixtures through sheath flow is a common technique used for continuous and high-efficiency dielectrophoretic (DEP) cell separation. However, it usually limits the separation flow velocity and requires a complex multichannel fluid control system that hinders the integration of a DEP separator with other microfluidic functionalities for comprehensive biomedical applications. Here, we propose and develop a high-efficiency, sheathless particle/cell separation method without prefocusing based on flow-field-assisted DEP by combining the effects of AC electric field (E-field) and flow field (F-field). A hollow lemon-shaped electrode array is designed to generate a long-range E-field gradient in the microchannel, which can effectively induce lateral displacements of particles/cells in a continuous flow. A series of arc-shaped protrusion structures is designed along the microchannel to form a F-field, which can effectively guide the particles/cells toward the targeted E-field region without prefocusing. By tuning the E-field, two distinct modes can be realized and switched in one single device, including the sheathless separation (ShLS) and the adjustable particle mixing ratio (AMR) modes. In the ShLS mode, we have achieved the continuous separation of breast cancer cells from erythrocytes with a recovery rate of 95.5% and the separation of polystyrene particles from yeast cells with a purity of 97.1% at flow velocities over 2.59 mm/s in a 2 cm channel under optimized conditions. The AMR mode provides a strategy for controlling the mixing ratio of different particles/cells as a well-defined pretreatment method for biomedical research studies. The proposed microchip is easy to use and displays high versatility for biological and medical applications.
Collapse
Affiliation(s)
- Shitao Shen
- International Joint Laboratory of Optofluidic Technology and System, National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics and School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China
| | - Zichuan Yi
- International Joint Laboratory of Optofluidic Technology and System, National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics and School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China.,College of Electron and Information, University of Electronic Science and Technology of China, Zhongshan Institute, Zhongshan 528402, China
| | - Xing Li
- International Joint Laboratory of Optofluidic Technology and System, National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics and School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China
| | - Shuting Xie
- International Joint Laboratory of Optofluidic Technology and System, National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics and School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China
| | - Mingliang Jin
- International Joint Laboratory of Optofluidic Technology and System, National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics and School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China
| | - Guofu Zhou
- International Joint Laboratory of Optofluidic Technology and System, National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics and School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China
| | - Zhibin Yan
- International Joint Laboratory of Optofluidic Technology and System, National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics and School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China
| | - Lingling Shui
- International Joint Laboratory of Optofluidic Technology and System, National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics and School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China
| |
Collapse
|
26
|
Kang K, Zhou X, Zhang Y, Zhu N, Li G, Yi Q, Wu Y. Cell-Released Magnetic Vesicles Capturing Metabolic Labeled Rare Circulating Tumor Cells Based on Bioorthogonal Chemistry. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007796. [PMID: 33749110 DOI: 10.1002/smll.202007796] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/24/2021] [Indexed: 06/12/2023]
Abstract
Capture of circulating tumor cells (CTCs) with high efficiency and high purity holds great value for potential clinical applications. Besides the existing problems of contamination from blood cells and plasma proteins, unknown/down-regulated expression of targeting markers (e.g., antigen, receptor, etc.) of CTCs have questioned the reliability and general applicability of current CTCs capture methodologies based on immune/aptamer-affinity. Herein, a cell-engineered strategy is designed to break down such barriers by employing the cell metabolism as the leading force to solve key problems. Generally, through an extracellular vesicle generation way, the cell-released magnetic vesicles inherited parent cellular membrane characteristics are produced, and then functionalized with dibenzoazacyclooctyne to target and isolate the metabolic labeled rare CTCs. This strategy offers good reliability and broader possibilities to capture different types of tumor cells, as proven by the capture efficiency above 84% and 82% for A549 and HepG2 cell lines as well as an extremely low detection limitation of 5 cells. Moreover, it enabled high purity enrichment of CTCs from 1 mL blood samples of tumor-bearing mice, only ≈5-757 white blood cells are non-specific caught, ignoring the potential phenotypic fluctuation associated with the cancer progression.
Collapse
Affiliation(s)
- Ke Kang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xiaoxi Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yujia Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Nanhang Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Guohao Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qiangying Yi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
27
|
Guo S, Huang H, Zeng W, Jiang Z, Wang X, Huang W, Wang X. Facile cell patterning induced by combined surface topography and chemistry on polydopamine-defined nanosubstrates. NANOTECHNOLOGY 2021; 32:145303. [PMID: 33361576 DOI: 10.1088/1361-6528/abd6d2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cell patterning holds significant implications for cell-based analysis and high-throughput screening. The challenge and key factor for formation of cell patterns is to precisely modulate the interaction between cells and substrate surfaces. Many nanosubstrates have been developed to control cell adhesion and patterning, however, requirements of complicated fabrication procedures, harsh reaction conditions, and delicate manipulation are not routinely feasible. Here, we developed a hierarchical polydimethylsiloxane nanosubstrate (HPNS) coated with mussel-inspired polydopamine (PDA) micropatterns for effective cell patterning, depending on both surface topography and chemistry. HPNSs obtained by facile template-assisted replication brought enhanced topographic interaction between cells and substrates, but they were innately hydrophobic and cell-repellent. The hydrophobic nanosubstrates were converted to be hydrophilic after PDA coatings formed via spontaneous self-polymerization, which greatly facilitated cell adhesion. As such, without resorting to any external forces or physical constraints, cells selectively adhered and spread on spatially defined PDA regions with high efficiency, and well-defined cell microarrays could be formed within 20 min. Therefore, this easy-to-fabricate nanosubstrate with no complex chemical modification will afford a facile yet effective platform for rapid cell patterning.
Collapse
Affiliation(s)
- Shan Guo
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Haiyan Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Weiwu Zeng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Zhuoran Jiang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Xin Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Weihua Huang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Xinghuan Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| |
Collapse
|
28
|
Hu X, Zang X, Lv Y. Detection of circulating tumor cells: Advances and critical concerns. Oncol Lett 2021; 21:422. [PMID: 33850563 PMCID: PMC8025150 DOI: 10.3892/ol.2021.12683] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Metastasis is the main cause of cancer-related death and the major challenge in cancer treatment. Cancer cells in circulation are termed circulating tumor cells (CTCs). Primary tumor metastasis is likely due to CTCs released into the bloodstream. These CTCs extravasate and form fatal metastases in different organs. Analyses of CTCs are clarifying the biological understanding of metastatic cancers. These data are also helpful to monitor disease progression and to inform the development of personalized cancer treatment-based liquid biopsy. However, CTCs are a rare cell population with 1-10 CTCs per ml and are difficult to isolate from blood. Numerous approaches to detect CTCs have been developed based on the physical and biological properties of the cells. The present review summarizes the progress made in detecting CTCs.
Collapse
Affiliation(s)
- Xiuxiu Hu
- School of Medical Technology, Jiangsu College of Nursing, Huai'an, Jiangsu 22300, P.R. China
| | - Xiaojuan Zang
- Department of Ultrasonography, Huai'an Maternity and Child Health Care Hospital, Huai'an, Jiangsu 223002, P.R. China
| | - Yanguan Lv
- Clinical Medical Laboratory, Huai'an Maternity and Child Health Care Hospital, Huai'an, Jiangsu 223002, P.R. China
| |
Collapse
|
29
|
Wu TH, Wu CH, Huang CJ, Chang YC. Anticlogging Hemofiltration Device for Mass Collection of Circulating Tumor Cells by Ligand-Free Size Selection. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:3399-3409. [PMID: 33689353 DOI: 10.1021/acs.langmuir.0c03613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A new hemofiltration system was developed to continuously capture circulating tumor cells (CTCs) from a large volume of whole blood using a column that was packed with antifouling zwitterionized silica microspheres. The silica microspheres were modified with sulfobetaine silane (SBSi) to inhibit fouling, resist clogging, and give a high surface wettability and prolonged operation time. Packed microspheres with different diameters formed size-controllable interstitial pores that effectively captured CTCs by ligand-free size selection. For optimized performance of the hemofiltration system, operational factors, including the size of microspheres, flow rate, and cross-sectional area of the column, were considered with respect to the removal rate for colorectal cancer cells and the retention rate for white blood cells and red blood cells. The captured CTCs were collected from the column by density sedimentation. A large quantity of colorectal cancer cells was spiked into sheep blood, and the sample was circulated for 5 h with a total operational volume of 2 L followed by collection and culture in vitro. The results showed that the proposed hemofiltration device selectively removed abundant CTCs from in vitro circulatory blood. The viable cells were harvested for amplification and potential applications for precision medicine.
Collapse
Affiliation(s)
- Tzu-Hsien Wu
- Department of Biomedical Sciences and Engineering, National Central University, Jhong-Li, Taoyuan 320, Taiwan
| | - Cheng-Han Wu
- Department of Biomedical Sciences and Engineering, National Central University, Jhong-Li, Taoyuan 320, Taiwan
| | - Chun-Jen Huang
- Chemical & Materials Engineering Department, National Central University, Jhong-Li, Taoyuan 320, Taiwan
- R&D Center for Membrane Technology, Chung Yuan Christian University, 200 Chung Pei Road, Chung-Li City 32023, Taiwan
- NCU-DSM Research Center, National Central University, Jhong-Li, Taoyuan 320, Taiwan
| | - Ying-Chih Chang
- Genomics Research Center, Academia Sinica, 128, Sec 2, Academic Road, Nankang, Taipei 115, Taiwan
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
30
|
Ding P, Wang Z, Wu Z, Zhu W, Liu L, Sun N, Pei R. Aptamer-based nanostructured interfaces for the detection and release of circulating tumor cells. J Mater Chem B 2021; 8:3408-3422. [PMID: 32022083 DOI: 10.1039/c9tb02457c] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Analysis of circulating tumor cells (CTCs) can provide significant clinical information for tumors, which has proven to be helpful for cancer diagnosis, prognosis monitoring, treatment efficacy, and personalized therapy. However, CTCs are an extremely rare cell population, which challenges the isolation of CTCs from patient blood. Over the last few decades, many strategies for CTC detection have been developed based on the physical and biological properties of CTCs. Among them, nanostructured interfaces have been widely applied as CTC detection platforms to overcome the current limitations associated with CTC capture. Furthermore, aptamers have attracted significant attention in the detection of CTCs due to their advantages, including good affinity, low cost, easy modification, excellent stability, and low immunogenicity. In addition, effective and nondestructive release of CTCs can be achieved by aptamer-mediated methods that are used under mild conditions. Herein, we review some progress in the detection and release of CTCs through aptamer-functionalized nanostructured interfaces.
Collapse
Affiliation(s)
- Pi Ding
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | | | | | | | | | | | | |
Collapse
|
31
|
Wang J, Zhang R, Ji X, Wang P, Ding C. SERS and fluorescence detection of circulating tumor cells (CTCs) with specific capture-release mode based on multifunctional gold nanomaterials and dual-selective recognition. Anal Chim Acta 2021; 1141:206-213. [PMID: 33248653 DOI: 10.1016/j.aca.2020.10.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 10/16/2020] [Indexed: 01/05/2023]
Abstract
Herein, a dual-selective recognition and multi-enhanced surface-enhanced Raman scattering (SERS)-fluorescence dual mode detection platform is designed for the detection of circulating tumor cells (CTCs). The gold nanoflowers (AuNFs) substrate was synthesized and the CTCs were captured on the surface area of AuNFs/ITO substrate by aptamers modified. At the same time, the novel nanoprobe was designed, anti-EpCAM (AE) and trigger DNA were modified onto the surface of gold nanostars (AuNSs) through a PEG linker. The novel nanoprobe identified CTCs through the specific recognition reaction between AE and the cell epithelial adhesion molecule of the CTCs. The dual-recognition cellular mechanism of the aptamers and AE improves selectivity. Then, the complementary sequence (CS) hybridize with aptamers to release the captured CTCs into the culture medium. The number of CTCs released was detected by SERS and fluorescence. The limit of SERS detection was 5 cells/mL with a linear relationship from 5 to 200 cells/mL. The limit of fluorescence detection was 10 cells/mL with a linear relationship from 10 to 200 cells/mL. Thus, the developed CTCs detection platform demonstrates promising applications for clinical diagnosis.
Collapse
Affiliation(s)
- Jing Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Ruiyuan Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Xiaoting Ji
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Peipei Wang
- Qingdao Central Hospital, Qingdao, 266042, China
| | - Caifeng Ding
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China.
| |
Collapse
|
32
|
Jiang W, Han L, Yang L, Xu T, He J, Peng R, Liu Z, Zhang C, Yu X, Jia L. Natural Fish Trap-Like Nanocage for Label-Free Capture of Circulating Tumor Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002259. [PMID: 33240774 PMCID: PMC7675191 DOI: 10.1002/advs.202002259] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/04/2020] [Indexed: 06/11/2023]
Abstract
Nanomaterials have achieved several breakthroughs in the capture of circulating tumor cells (CTCs) over the past decades. However, artificial fabrication of label-free nanomaterials used for high-efficiency CTC capture is still a challenge. Through billions of years of evolution and natural selection, various complicated and precise hierarchical structures are developed. Here, a novel fish trap-like "nanocage" structure derived from the natural Chrysanthemum pollen is reported and a nanocage-featured film for the label-free capture of CTCs and CTC clusters is constructed. The nanocage-featured film effectively captures 92% rare cancer cells with a broad spectrum of cancer types, due to the synergistic effect of nanocage-CTC filopodia matching, high contact area, and strong adhesion force between the cancer cells and the nanocage. Furthermore, the nanocage-featured film successfully detects CTCs and CTC clusters in 2 or 4 mL blood taken from 21 cancer patients (stages I-IV) suffering from various types of cancers. This novel, abundant, and economical fish trap-like "nanocage" may provide new perspectives for the application of natural nanomaterials in clinical CTC capture and analysis.
Collapse
Affiliation(s)
- Wenning Jiang
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Lulu Han
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Liwei Yang
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Ting Xu
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Jiabei He
- Department of OncologyThe Dalian Municipal Central Hospital Affiliated of Dalian Medical UniversityDalian116033P. R. China
| | - Ruilian Peng
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Ziyu Liu
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Chong Zhang
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Xiaomin Yu
- Department of OncologyThe Dalian Municipal Central Hospital Affiliated of Dalian Medical UniversityDalian116033P. R. China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| |
Collapse
|
33
|
Park JE, Oh N, Nam H, Park JH, Kim S, Jeon JS, Yang M. Efficient Capture and Raman Analysis of Circulating Tumor Cells by Nano-Undulated AgNPs-rGO Composite SERS Substrates. SENSORS (BASEL, SWITZERLAND) 2020; 20:E5089. [PMID: 32906807 PMCID: PMC7570931 DOI: 10.3390/s20185089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/02/2020] [Accepted: 09/05/2020] [Indexed: 12/12/2022]
Abstract
The analysis of circulating tumor cells (CTCs) in the peripheral blood of cancer patients is critical in clinical research for further investigation of tumor progression and metastasis. In this study, we present a novel surface-enhanced Raman scattering (SERS) substrate for the efficient capture and characterization of cancer cells using silver nanoparticles-reduced graphene oxide (AgNPs-rGO) composites. A pulsed laser reduction of silver nanowire-graphene oxide (AgNW-GO) mixture films induces hot-spot formations among AgNPs and artificial biointerfaces consisting of rGOs. We also use in situ electric field-assisted fabrication methods to enhance the roughness of the SERS substrate. The AgNW-GO mixture films, well suited for the proposed process due to its inherent electrophoretic motion, is adjusted between indium tin oxide (ITO) transparent electrodes and the nano-undulated surface is generated by applying direct-current (DC) electric fields during the laser process. As a result, MCF7 breast cancer cells are efficiently captured on the AgNPs-rGO substrates, about four times higher than the AgNWs-GO films, and the captured living cells are successfully analyzed by SERS spectroscopy. Our newly designed bifunctional substrate can be applied as an effective system for the capture and characterization of CTCs.
Collapse
Affiliation(s)
- Jong-Eun Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
| | - Nuri Oh
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (N.O.); (J.-H.P.)
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hyeono Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (N.O.); (J.-H.P.)
| | - Sanha Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
| | - Jessie S. Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
| | - Minyang Yang
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
- Department of Mechanical Engineering, State University of New York Korea, Incheon 21985, Korea
| |
Collapse
|
34
|
Tian C, Xu X, Wang Y, Li D, Lu H, Yang Z. Development and Clinical Prospects of Techniques to Separate Circulating Tumor Cells from Peripheral Blood. Cancer Manag Res 2020; 12:7263-7275. [PMID: 32884342 PMCID: PMC7434565 DOI: 10.2147/cmar.s248380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Detection of circulating tumor cells (CTC) is an important liquid biopsy technique that has advanced considerably in recent years. To further advance the development of technology for curing cancer, several CTC technologies have been proposed by various research groups. Despite their potential role in early cancer diagnosis and prognosis, CTC methods are currently used for research purposes only, and very few methods have been accepted for clinical applications because of difficulties, including CTC heterogeneity, CTC separation from the blood, and a lack of thorough clinical validation. Although current CTC technologies have not been truly implemented, they possess high potential as future clinical diagnostic techniques for individualized cancer. Here, we review current developments in CTC separation technology. We also explore new CTC detection methods based on telomerase and nanomaterials, such as in vivo flow cytometry. In addition, we discuss the difficulties that must be overcome before CTC can be applied in clinical settings.
Collapse
Affiliation(s)
- Cheng Tian
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Xinhua Xu
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Yuke Wang
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Dailong Li
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Haiyan Lu
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Ziwei Yang
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| |
Collapse
|
35
|
Han L, Peng R, Jiang W, Xu T, Zhang C, Chen K, Zhang Y, Song H, Jia L. Coordination-driven reversible surfaces with site-specifically immobilized nanobody for dynamic cancer cell capture and release. J Mater Chem B 2020; 8:7511-7520. [PMID: 32677632 DOI: 10.1039/d0tb00574f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Selective isolation of circulating tumor cells (CTCs) from blood provides a non-invasive avenue for the diagnosis, prognosis and personalized treatment for patients with cancer. The specific capture of CTCs is conventionally based on the immunoaffinity recognition between antibody and receptor on cell membranes. However, using a traditional antibody for high-efficiency isolation of CTCs remains a challenge due to the limited loading capacity of the large antibodies on material surfaces. Herein, using a small-sized nanobody (Nb), we developed a widely applicable strategy to construct reversible site-specifically immobilized Nb surfaces for the capture and release of epidermoid cancer cell line A431 cells. Coordination interaction between the histidine tag (His-tag) of the nanobody (Nb) and Ni2+ ions that chelated to the NTA-modified poly(2-hydroxyethyl methacrylate) (PHEMA) brushes was used to achieve site-specific immobilization of EGFR Nb (PHEMA-aEGFR surfaces). The high-density immobilized nanobody possessing maximized activity resulted in the high-efficiency capture of 81% rare A431 cells within just 30 min, showing a higher capture yield and shorter capture time compared with that achieved by the conventional antibody immobilized on the flat surface. Additionally, the PHEMA-aEGFR surfaces exhibited low capture limit (1 cell mL-1), cytocompatibility for captured cells, as well as negligible non-specific adhesion of PBMCs. With a one-step treatment using imidazole for competitive coordination, 86% of the captured cells were effectively released. This multifunctional and dynamic site-specifically immobilized nanobody strategy paves a new path in the development of materials and instruments for the high-efficiency capture and release of rare cells at a low cost.
Collapse
Affiliation(s)
- Lulu Han
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kanioura A, Constantoudis V, Petrou P, Kletsas D, Tserepi A, Gogolides E, Chatzichristidi M, Kakabakos S. Oxygen plasma micro-nanostructured PMMA plates and microfluidics for increased adhesion and proliferation of cancer versus normal cells: The role of surface roughness and disorder. MICRO AND NANO ENGINEERING 2020. [DOI: 10.1016/j.mne.2020.100060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
37
|
Khizar S, Ben Halima H, Ahmad NM, Zine N, Errachid A, Elaissari A. Magnetic nanoparticles in microfluidic and sensing: From transport to detection. Electrophoresis 2020; 41:1206-1224. [DOI: 10.1002/elps.201900377] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Sumera Khizar
- Université de Lyon LAGEP, UMR‐5007, CNRS, Université Lyon 1, 5007 43 Bd 11 Novembre 1918 Villeurbanne F‐69622 France
- Polymer Research Lab School of Chemical and Materials Engineering (SCME) National University of Sciences and Technology (NUST) H‐12 Sector Islamabad 44000 Pakistan
| | - Hamdi Ben Halima
- Université de Lyon Institut des Science Analytiques UMR 5280, CNRS Université Lyon 1 ENS Lyon-5, rue de la Doua Villeurbanne F‐69100 France
| | - Nasir M. Ahmad
- Polymer Research Lab School of Chemical and Materials Engineering (SCME) National University of Sciences and Technology (NUST) H‐12 Sector Islamabad 44000 Pakistan
| | - Nadia Zine
- Université de Lyon Institut des Science Analytiques UMR 5280, CNRS Université Lyon 1 ENS Lyon-5, rue de la Doua Villeurbanne F‐69100 France
| | - Abdelhamid Errachid
- Université de Lyon Institut des Science Analytiques UMR 5280, CNRS Université Lyon 1 ENS Lyon-5, rue de la Doua Villeurbanne F‐69100 France
| | - Abdelhamid Elaissari
- Université de Lyon LAGEP, UMR‐5007, CNRS, Université Lyon 1, 5007 43 Bd 11 Novembre 1918 Villeurbanne F‐69622 France
| |
Collapse
|
38
|
Feng J, Mo J, Zhang A, Liu D, Zhou L, Hang T, Yang C, Wu Q, Xia D, Wen R, Yang J, Feng Y, Huang Y, Hu N, He G, Xie X. Antibody-free isolation and regulation of adherent cancer cells via hybrid branched microtube-sandwiched hydrodynamic system. NANOSCALE 2020; 12:5103-5113. [PMID: 32068774 DOI: 10.1039/d0nr00153h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The detection of circulating tumor cells (CTCs) has achieved promising progress for early diagnosis and disease analysis. Microfluidic chip techniques have recently promoted the technologies of CTC sorting and analysis, yet seldom can the microfluidic chips for CTC enrichment via antibody-free capture provide in situ regulation of both extracellular and intracellular activity, which would be advantageous for cell-based pharmaceutical therapeutics and screening. Herein, we have demonstrated a hybrid TiO2/ZnO branched microtube array (HBMTA)-sandwiched hydrodynamic device that integrates the multiple functions of selective enrichment of adherent tumor cells in an antibody-free manner and in situ delivery to the extracellular and intracellular spaces of the enriched tumor cells. More than 90% cancer cells were enriched on the device due to their preferential adhesion with the nano-branches of HBMTA, while more than 91% blood cells were eliminated from the device by constant hydrodynamic fluid shearing. For in situ regulation, temporally and spatially controlled extracellular delivery to the enriched tumor cells could be precisely achieved through the hollow structures of the HBMTA. In addition, reagents (e.g. propidium iodide) could be delivered into the intracellular spaces of enriched tumor cells by coupling an electric field to nondestructively perforate the cell membrane. Our study not only offers a promising and facile strategy for antibody-free isolation of tumor cells, but also provides unique opportunities to facilitate cancer research, including antitumor drug screening and personalized therapeutics.
Collapse
Affiliation(s)
- Jianming Feng
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Jingshan Mo
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Aihua Zhang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Di Liu
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637, USA
| | - Lingfei Zhou
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Tian Hang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Cheng Yang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Qianni Wu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Dehua Xia
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Rui Wen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Jiang Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yuping Feng
- School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Yan Huang
- School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology; The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510006, China.
| |
Collapse
|
39
|
Moussa HI, Chan WY, Logan M, Aucoin MG, Tsui TY. Limitation in Controlling the Morphology of Mammalian Vero Cells Induced by Cell Division on Asymmetric Tungsten-Silicon Oxide Nanocomposite. MATERIALS 2020; 13:ma13020335. [PMID: 31940759 PMCID: PMC7013836 DOI: 10.3390/ma13020335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 12/30/2022]
Abstract
Engineered nanomaterials are often used in tissue engineering applications to influence and manipulate the behavior of cells. Recently, a number of tungsten-silicon oxide nanocomposite devices containing equal width (symmetric) tungsten and silicon oxide parallel line comb structures were developed and used by our group. The devices induced over 90% of seeded cells (Vero) to align within ±20° of the axes of 10 µm wide tungsten lines. Furthermore, a mathematical model was successfully developed to predict this alignment behavior and forecast the minimum width of isolated tungsten lines required to induce such behavior. However, the mechanism by which the widths of the symmetrical tungsten and silicon oxide lines induce the alignment behavior is still unknown. Furthermore, the model was never tested on more complex asymmetrical structures. Herewith, experiments were conducted with mammalian cells on complex asymmetrical structures with unequal tungsten and silicon oxide line widths. Results showed that the model could be extended to more complex pattern structures. In addition, cell morphology on the patterned structures reset during cell division because of mitotic rounding, which reduced the population of cells that elongated and aligned on the tungsten lines. Ultimately, we concluded that it was impossible to achieve a 100% alignment with cells having unsynchronized cell cycles because cell rounding during mitosis took precedence over cell alignment; in other words, internal chemical cues had a stronger role in cell morphology than external cues.
Collapse
Affiliation(s)
- Hassan I. Moussa
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (H.I.M.); (W.Y.C.); (M.L.); (M.G.A.)
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Wing Y. Chan
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (H.I.M.); (W.Y.C.); (M.L.); (M.G.A.)
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Megan Logan
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (H.I.M.); (W.Y.C.); (M.L.); (M.G.A.)
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Marc G. Aucoin
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (H.I.M.); (W.Y.C.); (M.L.); (M.G.A.)
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Ting Y. Tsui
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (H.I.M.); (W.Y.C.); (M.L.); (M.G.A.)
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Correspondence: ; Tel.: +1-519-888-4567 (ext. 38404)
| |
Collapse
|
40
|
Dong J, Chen JF, Smalley M, Zhao M, Ke Z, Zhu Y, Tseng HR. Nanostructured Substrates for Detection and Characterization of Circulating Rare Cells: From Materials Research to Clinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903663. [PMID: 31566837 PMCID: PMC6946854 DOI: 10.1002/adma.201903663] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/02/2019] [Indexed: 05/03/2023]
Abstract
Circulating rare cells in the blood are of great significance for both materials research and clinical applications. For example, circulating tumor cells (CTCs) have been demonstrated as useful biomarkers for "liquid biopsy" of the tumor. Circulating fetal nucleated cells (CFNCs) have shown potential in noninvasive prenatal diagnostics. However, it is technically challenging to detect and isolate circulating rare cells due to their extremely low abundance compared to hematologic cells. Nanostructured substrates offer a unique solution to address these challenges by providing local topographic interactions to strengthen cell adhesion and large surface areas for grafting capture agents, resulting in improved cell capture efficiency, purity, sensitivity, and reproducibility. In addition, rare-cell retrieval strategies, including stimulus-responsiveness and additive reagent-triggered release on different nanostructured substrates, allow for on-demand retrieval of the captured CTCs/CFNCs with high cell viability and molecular integrity. Several nanostructured substrate-enabled CTC/CFNC assays are observed maturing from enumeration and subclassification to molecular analyses. These can one day become powerful tools in disease diagnosis, prognostic prediction, and dynamic monitoring of therapeutic response-paving the way for personalized medical care.
Collapse
Affiliation(s)
- Jiantong Dong
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jie-Fu Chen
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
41
|
Lei Y, Ye H, Xiang S, Huang Y, Zhu C, Zhang W, Chen Y, Cao Y. Pipette-like action of a reusable and NIR light-responsive film for the aspiration and removal of viable cancer cells. NEW J CHEM 2020. [DOI: 10.1039/c9nj05449a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A reusable and NIR light-responsive composite membrane is developed to capture/release viable cancer cells.
Collapse
Affiliation(s)
- Yang Lei
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education
- Jianghan University
- Wuhan 430056
- China
| | - Haixia Ye
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education
- Jianghan University
- Wuhan 430056
- China
| | - Siqi Xiang
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education
- Jianghan University
- Wuhan 430056
- China
| | - Yuan Huang
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education
- Jianghan University
- Wuhan 430056
- China
| | - Chao Zhu
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education
- Jianghan University
- Wuhan 430056
- China
| | - Weiying Zhang
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education
- Jianghan University
- Wuhan 430056
- China
| | - Yong Chen
- Département de Chimie
- Ecole Normale Supérieure
- F-75231 Paris Cedex 05
- France
| | - Yiping Cao
- Institute for Interdisciplinary Research & Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education
- Jianghan University
- Wuhan 430056
- China
| |
Collapse
|
42
|
Carbon-nanotube reinforcement of DNA-silica nanocomposites yields programmable and cell-instructive biocoatings. Nat Commun 2019; 10:5522. [PMID: 31797918 PMCID: PMC6892801 DOI: 10.1038/s41467-019-13381-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 11/04/2019] [Indexed: 12/22/2022] Open
Abstract
Biomedical applications require substrata that allow for the grafting, colonization and control of eukaryotic cells. Currently available materials are often limited by insufficient possibilities for the integration of biological functions and means for tuning the mechanical properties. We report on tailorable nanocomposite materials in which silica nanoparticles are interwoven with carbon nanotubes by DNA polymerization. The modular, well controllable and scalable synthesis yields materials whose composition can be gradually adjusted to produce synergistic, non-linear mechanical stiffness and viscosity properties. The materials were exploited as substrata that outperform conventional culture surfaces in the ability to control cellular adhesion, proliferation and transmigration through the hydrogel matrix. The composite materials also enable the construction of layered cell architectures, the expansion of embryonic stem cells by simplified cultivation methods and the on-demand release of uniformly sized stem cell spheroids.
Collapse
|
43
|
Kanioura A, Petrou P, Kletsas D, Tserepi A, Chatzichristidi M, Gogolides E, Kakabakos S. Three-dimensional (3D) hierarchical oxygen plasma micro/nanostructured polymeric substrates for selective enrichment of cancer cells from mixtures with normal ones. Colloids Surf B Biointerfaces 2019; 187:110675. [PMID: 31810566 DOI: 10.1016/j.colsurfb.2019.110675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 10/16/2019] [Accepted: 11/25/2019] [Indexed: 12/30/2022]
Abstract
The enrichment of cancer cell population when in mixtures with normal ones is of great importance for cancer diagnosis. In this work, poly(methyl methacrylate) films have been processed applying different oxygen plasma conditions to fabricate surfaces with structure height ranging from 22 to more than 2000 nm. The surfaces were then evaluated with respect to adhesion and proliferation of both normal and cancer human cells. In particular, normal skin and lung fibroblasts, and four different cancer cell lines, A431 (skin cancer), HT1080 (fibrosarcoma), A549 (lung cancer), and PC3 (prostate cancer), have been employed. It was found that adhesion and proliferation of cancer cells was favored when cultured onto the hierarchical micro/nanostructured surfaces as compared to untreated ones with the maximum values obtained for substrates treated at -100 V for 3 min. On the other hand, although the adhesion of normal fibroblasts was not influenced by the micro/nanostructured surfaces, their morphology and proliferation was significantly impaired, especially after 3-day culture on these surfaces. The reduced proliferation rate of adherent fibroblasts was linked to reduced focal points formation, as it was verified through vinculin staining, and not to apoptosis. The micro/nanostructured surfaces prepared with plasma treatment at -100 V for 3 min (hierarchical topography with mean height of ∼800 nm) were selected as substrates for normal and cancer cell co-culture experiments. It was found that 25-80 times enrichment of cancer over the normal cells was achieved on the nanostructured surfaces after 3-day culture, while it was 5-8 times lower on the untreated ones. It should be noticed that this is the first time such high enrichment ratios are achieved without implementing surfaces modified with binding molecules specific for cancer cells. Thus, the nanostructured surfaces hold a strong promise as culture substrates for separation and enrichment of cancer cells from mixtures with normal ones that should find application in cancer diagnostics.
Collapse
Affiliation(s)
- Anastasia Kanioura
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | - Panagiota Petrou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | - Dimitris Kletsas
- Institute of Biosciences and Applications, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | - Angeliki Tserepi
- Institute of Nanoscience & Nanotechnology, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | | | - Evangelos Gogolides
- Institute of Nanoscience & Nanotechnology, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | - Sotirios Kakabakos
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece.
| |
Collapse
|
44
|
Li W, Wang H, Zhao Z, Gao H, Liu C, Zhu L, Wang C, Yang Y. Emerging Nanotechnologies for Liquid Biopsy: The Detection of Circulating Tumor Cells and Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805344. [PMID: 30589111 DOI: 10.1002/adma.201805344] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/29/2018] [Indexed: 05/18/2023]
Abstract
Liquid biopsy enables noninvasive and dynamic analysis of molecular or cellular biomarkers, and therefore holds great potential for the diagnosis, prognosis, monitoring of disease progress and treatment efficacy, understanding of disease mechanisms, and identification of therapeutic targets for drug development. In this review, the recent progress in nanomaterials, nanostructures, nanodevices, and nanosensors for liquid biopsy is summarized, with a focus on the detection and molecular characterization of circulating tumor cells (CTCs) and extracellular vesicles (EVs). The developments and advances of nanomaterials and nanostructures in enhancing the sensitivity, specificity, and purity for the detection of CTCs and EVs are discussed. Sensing techniques for signal transduction and amplification as well as visualization strategies are also discussed. New technologies for the reversible release of the isolated CTCs and EVs and for single-CTC/EV analysis are summarized. Emerging microfluidic platforms for the integral on-chip isolation, detection, and molecular analysis are also included. The opportunities, challenges, and prospects of these innovative materials and technologies, especially with regard to their feasibility in clinical applications, are discussed. The applications of nanotechnology-based liquid biopsy will bring new insight into the clinical practice in monitoring and treatment of tumor and other significant diseases.
Collapse
Affiliation(s)
- Wenzhe Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Huayi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zijian Zhao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Houqian Gao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Changliang Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
45
|
Integration of Hierarchical Micro-/Nanostructures in a Microfluidic Chip for Efficient and Selective Isolation of Rare Tumor Cells. MICROMACHINES 2019; 10:mi10100698. [PMID: 31615080 PMCID: PMC6843196 DOI: 10.3390/mi10100698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/31/2022]
Abstract
Circulating tumor cells (CTCs) are important clinical markers for both cancer early diagnosis and prognosis. Various techniques have been developed in the past decade to isolate and quantify these cells from the blood while microfluidic technology attracts significant attention due to better controlled microenvironment. When combined with advanced nanotechnologies, CTC isolation performance in microfluidic devices can be further improved. In this article, by extending the wavy-herringbone concept developed earlier in our team, we prepared a hierarchical microfluidic chip by introducing a uniform coating of nanoparticles with anti-epithelial cell adhesion molecule (EpCAM) on wavy microgrooves. This hierarchical structured platform not only maintains the capture purity of the wavy-herringbone structure but improves the capture efficiency thanks to the larger surface area to volume ratio brought by nanoparticles. Our results demonstrated a capture efficiency of almost 100% at a low shear rate of 60/s. Even at a higher shear rate of 400/s, the hierarchical micro/nanostructures demonstrated an enhancement of up to ~3-fold for capture efficiency (i.e., 70%) and ~1.5-fold for capture purity (i.e., 68%), compared to wavy-herringbone structures without nanoparticle coating. With these promising results, this hierarchical structured platform represents a technological advancement for CTC isolation and cancer care.
Collapse
|
46
|
Li W, Li R, Huang B, Wang Z, Sun Y, Wei X, Heng C, Liu W, Yu M, Guo SS, Zhao XZ. TiO 2 nanopillar arrays coated with gelatin film for efficient capture and undamaged release of circulating tumor cells. NANOTECHNOLOGY 2019; 30:335101. [PMID: 30965310 DOI: 10.1088/1361-6528/ab176c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Circulating tumor cells (CTCs) are important for the detection and treatment of cancer. Nevertheless, a low density of circulating tumor cells makes the capture and release of CTCs an obstacle. In this work, TiO2 nanopillar arrays coated with gelatin film were synthesized for efficient capture and undamaged release of circulating tumor cells. The scanning electron microscope and atomic force microscope images demonstrate that the substrate has a certain roughness. The interaction between the cell membrane and the nanostructure substrate contributes to the efficient capture of CTC (capture efficiency up to 94.98%). The gelatin layer has excellent biocompatibility and can be rapidly digested by matrix metalloproteinase (MMP9), which realizes the non-destructive release of CTCs (0.1 mg ml-1, 5 min, nearly 100% release efficiency, activity 100%). Therefore, by our strategy, the CTCs can be efficiently captured and released undamaged, which is important for subsequent analysis.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yue WQ, Tan Z, Li XP, Liu FF, Wang C. Micro/nanofluidic technologies for efficient isolation and detection of circulating tumor cells. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
48
|
Masigol M, Fattahi N, Barua N, Lokitz BS, Retterer ST, Platt TG, Hansen RR. Identification of Critical Surface Parameters Driving Lectin-Mediated Capture of Bacteria from Solution. Biomacromolecules 2019; 20:2852-2863. [PMID: 31150217 DOI: 10.1021/acs.biomac.9b00609] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Lectin-functional interfaces are useful for isolation of bacteria from solution because they are low-cost and allow nondestructive, reversible capture. This study provides a systematic investigation of physical and chemical surface parameters that influence bacteria capture over lectin-functionalized polymer interfaces and then applies these findings to construct surfaces with significantly enhanced bacteria capture. The designer block copolymer poly(glycidyl methacrylate)- block-poly(vinyldimethyl azlactone) was used as a lectin attachment layer, and lectin coupling into the polymer film through azlactone-lectin coupling reactions was first characterized. Here, experimental parameters including polymer areal chain density, lectin molecular weight, and lectin coupling buffer were systematically varied to identify parameters driving highest azlactone conversions and corresponding lectin surface densities. To introduce physical nanostructures into the attachment layer, nanopillar arrays (NPAs) of varied heights (300 and 2100 nm) were then used to provide an underlying surface template for the functional polymer layer. Capture of Escherichia coli on lectin-polymer surfaces coated over both flat and NPA surfaces was then investigated. For flat polymer interfaces, bacteria were detected on the surface after incubation at a solution concentration of 103 cfu/mL, and a corresponding detection limit of 1.7 × 103 cfu/mL was quantified. This detection limit was 1 order of magnitude lower than control lectin surfaces functionalized with standard, carbodiimide coupling chemistry. NPA surfaces containing 300 nm tall pillars further improved the detection limit to 2.1 × 102 cfu/mL, but also reduced the viability of captured cells. Finally, to investigate the impact of cell surface parameters on capture, we used Agrobacterium tumefaciens cells genetically modified to allow manipulation of exopolysaccharide adhesin production levels. Statistical analysis of surface capture levels revealed that lectin surface density was the primary factor driving capture, as opposed to exopolysaccharide adhesin expression. These findings emphasize the critical importance of the synthetic interface and the development of surfaces that combine high lectin densities with tailored physical features to drive high levels of capture. These insights will aid in design of biofunctional interfaces with physicochemical surface properties favorable for capture and isolation of bacteria cells from solutions.
Collapse
|
49
|
Ding L, Wu Y, Liu W, Liu L, Yu F, Yu S, Tian Y, Feng J, He L. Magnetic-assisted self-assembled aptamer/protein hybrid probes for efficient capture and rapid detection of cancer cells in whole blood. Talanta 2019; 205:120129. [PMID: 31450438 DOI: 10.1016/j.talanta.2019.120129] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/26/2019] [Accepted: 07/08/2019] [Indexed: 12/17/2022]
Abstract
Self-assembly of building blocks for constructing multifunctional materials has opened prospects for sensing applications in the biomedical fields. In particular, the combination of aptamer with DNA assembly-based nanotechnology has greatly improved the performance of cancer cell detection. Nevertheless, the cancer cell detection strategies of integrating aptamer with protein are relatively sparse. So we have developed a self-assembled aptamer method to realize the efficient capture and rapid detection of cancer cells by ingeniously combining aptamer modified magnetic nanoparticles as capture nanoprobes with self-assembled aptamer/protein hybrid probes (SAPPs) as signal amplification probes. By merely mixing the component materials together simultaneously, the SAPPs, integrating aptamer for cancer cell recognition with protein for amplifying signal, were fabricated by DNA-governed one-step assembly. In addition, the SAPPs-based method exhibits efficient capture, rapid (about 45 min) and specific CCRF-CEM detection performance, with limits of detection down to 75 cells/mL in buffer and 200 cells/mL in whole blood.
Collapse
Affiliation(s)
- Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Wei Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lie Liu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Fei Yu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Songcheng Yu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongmei Tian
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiaodi Feng
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, 450001, China
| | - Leiliang He
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
50
|
Iliescu FS, Poenar DP, Yu F, Ni M, Chan KH, Cima I, Taylor HK, Cima I, Iliescu C. Recent advances in microfluidic methods in cancer liquid biopsy. BIOMICROFLUIDICS 2019; 13:041503. [PMID: 31431816 PMCID: PMC6697033 DOI: 10.1063/1.5087690] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/24/2019] [Indexed: 05/04/2023]
Abstract
Early cancer detection, its monitoring, and therapeutical prediction are highly valuable, though extremely challenging targets in oncology. Significant progress has been made recently, resulting in a group of devices and techniques that are now capable of successfully detecting, interpreting, and monitoring cancer biomarkers in body fluids. Precise information about malignancies can be obtained from liquid biopsies by isolating and analyzing circulating tumor cells (CTCs) or nucleic acids, tumor-derived vesicles or proteins, and metabolites. The current work provides a general overview of the latest on-chip technological developments for cancer liquid biopsy. Current challenges for their translation and their application in various clinical settings are discussed. Microfluidic solutions for each set of biomarkers are compared, and a global overview of the major trends and ongoing research challenges is given. A detailed analysis of the microfluidic isolation of CTCs with recent efforts that aimed at increasing purity and capture efficiency is provided as well. Although CTCs have been the focus of a vast microfluidic research effort as the key element for obtaining relevant information, important clinical insights can also be achieved from alternative biomarkers, such as classical protein biomarkers, exosomes, or circulating-free nucleic acids. Finally, while most work has been devoted to the analysis of blood-based biomarkers, we highlight the less explored potential of urine as an ideal source of molecular cancer biomarkers for point-of-care lab-on-chip devices.
Collapse
Affiliation(s)
- Florina S. Iliescu
- School of Applied Science, Republic Polytechnic, Singapore 738964, Singapore
| | - Daniel P. Poenar
- VALENS-Centre for Bio Devices and Signal Analysis, School of EEE, Nanyang Technological University, Singapore 639798, Singapore
| | - Fang Yu
- Singapore Institute of Manufacturing Technology, A*STAR, Singapore 138634, Singapore
| | - Ming Ni
- School of Biological Sciences and Engineering, Yachay Technological University, San Miguel de Urcuquí 100105, Ecuador
| | - Kiat Hwa Chan
- Division of Science, Yale-NUS College, Singapore 138527, Singapore
| | | | - Hayden K. Taylor
- Department of Mechanical Engineering, University of California, Berkeley, California 94720, USA
| | - Igor Cima
- DKFZ-Division of Translational Oncology/Neurooncology, German Cancer Consortium (DKTK), Heidelberg and University Hospital Essen, Essen 45147, Germany
| | | |
Collapse
|