1
|
Mansor M, Yang C, Chong KL, Jamrus MA, Liu K, Yu M, Ahmad MR, Ren X. Label-Free and Rapid Microfluidic Design Rules for Circulating Tumor Cell Enrichment and Isolation: A Review and Simulation Analysis. ACS OMEGA 2025; 10:6306-6322. [PMID: 40028152 PMCID: PMC11866005 DOI: 10.1021/acsomega.4c08606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 03/05/2025]
Abstract
Enriching and isolating circulating tumor cells (CTCs) have attracted significant interest due to their important role in early cancer diagnosis and prognosis, allowing for minimally invasive approaches and providing vital information about metastasis at the cellular level. This review comprehensively summarizes the recent developments in microfluidic devices for CTC enrichment and isolation. The advantages and limitations of several microfluidic devices are discussed, and the design specifications of microfluidic devices for CTC enrichment are highlighted. We also developed a set of methodologies and design rules of label-free microfluidics such as spiral, deterministic lateral displacement (DLD) and dielectrophoresis (DEP) to allow researchers to design and develop microfluidic devices systematically and effectively, promoting rapid research on design, fabrication, and experimentation.
Collapse
Affiliation(s)
- Muhammad
Asraf Mansor
- Department
of Control and Mechatronics Engineering, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Chun Yang
- School
of Microelectronics, Tianjin University, Tianjin 300072, China
| | - Kar Lok Chong
- Department
of Control and Mechatronics Engineering, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Muhammad Asyraf Jamrus
- Department
of Control and Mechatronics Engineering, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Kewei Liu
- Sino-German
College of Intelligent Manufacturing, Shenzhen
Technology University, Shenzhen 518118, China
| | - Miao Yu
- Department
of Research and Development, Stedical Scientific, Carlsbad, California 92010, United States
| | - Mohd Ridzuan Ahmad
- Department
of Control and Mechatronics Engineering, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Xiang Ren
- School
of Microelectronics, Tianjin University, Tianjin 300072, China
| |
Collapse
|
2
|
Fu SW, Tang C, Tan X, Srivastava S. Liquid biopsy for early cancer detection: technological revolutions and clinical dilemma. Expert Rev Mol Diagn 2024; 24:937-955. [PMID: 39360748 DOI: 10.1080/14737159.2024.2408744] [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: 05/08/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Liquid biopsy is an innovative advancement in oncology, offering a noninvasive method for early cancer detection and monitoring by analyzing circulating tumor cells, DNA, RNA, and other biomarkers in bodily fluids. This technique has the potential to revolutionize precision oncology by providing real-time analysis of tumor dynamics, enabling early detection, monitoring treatment responses, and tailoring personalized therapies based on the molecular profiles of individual patients. AREAS COVERED In this review, the authors discuss current methodologies, technological challenges, and clinical applications of liquid biopsy. This includes advancements in detecting minimal residual disease, tracking tumor evolution, and combining liquid biopsy with other diagnostic modalities for precision oncology. Key areas explored are the sensitivity, specificity, and integration of multi-omics, AI, ML, and LLM technologies. EXPERT OPINION Liquid biopsy holds great potential to revolutionize cancer care through early detection and personalized treatment strategies. However, its success depends on overcoming technological and clinical hurdles, such as ensuring high sensitivity and specificity, interpreting results amidst tumor heterogeneity, and making tests accessible and affordable. Continued innovation and collaboration are crucial to fully realize the potential of liquid biopsy in improving early cancer detection, treatment, and monitoring.
Collapse
Affiliation(s)
- Sidney W Fu
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Cong Tang
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Xiaohui Tan
- Division of LS Research, LSBioscience, LLC, Frederick, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
3
|
Qiao Z, Teng X, Liu A, Yang W. Novel Isolating Approaches to Circulating Tumor Cell Enrichment Based on Microfluidics: A Review. MICROMACHINES 2024; 15:706. [PMID: 38930676 PMCID: PMC11206030 DOI: 10.3390/mi15060706] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Circulating tumor cells (CTCs), derived from the primary tumor and carrying genetic information, contribute significantly to the process of tumor metastasis. The analysis and detection of CTCs can be used to assess the prognosis and treatment response in patients with tumors, as well as to help study the metastatic mechanisms of tumors and the development of new drugs. Since CTCs are very rare in the blood, it is a challenging problem to enrich CTCs efficiently. In this paper, we provide a comprehensive overview of microfluidics-based enrichment devices for CTCs in recent years. We explore in detail the methods of enrichment based on the physical or biological properties of CTCs; among them, physical properties cover factors such as size, density, and dielectric properties, while biological properties are mainly related to tumor-specific markers on the surface of CTCs. In addition, we provide an in-depth description of the methods for enrichment of single CTCs and illustrate the importance of single CTCs for performing tumor analyses. Future research will focus on aspects such as improving the separation efficiency, reducing costs, and increasing the detection sensitivity and accuracy.
Collapse
Affiliation(s)
- Zezheng Qiao
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China; (Z.Q.); (X.T.)
| | - Xiangyu Teng
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China; (Z.Q.); (X.T.)
| | - Anqin Liu
- School of Mechanical and Electrical Engineering, Yantai Institute of Technology, Yantai 264005, China
| | - Wenguang Yang
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China; (Z.Q.); (X.T.)
| |
Collapse
|
4
|
Temilola DO, Adeola HA, Grobbelaar J, Chetty M. Liquid Biopsy in Head and Neck Cancer: Its Present State and Future Role in Africa. Cells 2023; 12:2663. [PMID: 37998398 PMCID: PMC10670726 DOI: 10.3390/cells12222663] [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: 10/10/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023] Open
Abstract
The rising mortality and morbidity rate of head and neck cancer (HNC) in Africa has been attributed to factors such as the poor state of health infrastructures, genetics, and late presentation resulting in the delayed diagnosis of these tumors. If well harnessed, emerging molecular and omics diagnostic technologies such as liquid biopsy can potentially play a major role in optimizing the management of HNC in Africa. However, to successfully apply liquid biopsy technology in the management of HNC in Africa, factors such as genetic, socioeconomic, environmental, and cultural acceptability of the technology must be given due consideration. This review outlines the role of circulating molecules such as tumor cells, tumor DNA, tumor RNA, proteins, and exosomes, in liquid biopsy technology for the management of HNC with a focus on studies conducted in Africa. The present state and the potential opportunities for the future use of liquid biopsy technology in the effective management of HNC in resource-limited settings such as Africa is further discussed.
Collapse
Affiliation(s)
- Dada Oluwaseyi Temilola
- Department of Craniofacial Biology, Faculty of Dentistry, University of the Western Cape, Tygerberg Hospital, Cape Town 7505, South Africa;
| | - Henry Ademola Adeola
- Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, University of the Western Cape, Tygerberg Hospital, Cape Town 7505, South Africa;
- Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town 7925, South Africa
| | - Johan Grobbelaar
- Division of Otorhinolaryngology, Department of Surgical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town 7505, South Africa;
| | - Manogari Chetty
- Department of Craniofacial Biology, Faculty of Dentistry, University of the Western Cape, Tygerberg Hospital, Cape Town 7505, South Africa;
| |
Collapse
|
5
|
Lawrence R, Watters M, Davies CR, Pantel K, Lu YJ. Circulating tumour cells for early detection of clinically relevant cancer. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00781-y. [PMID: 37268719 DOI: 10.1038/s41571-023-00781-y] [Citation(s) in RCA: 136] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 06/04/2023]
Abstract
Given that cancer mortality is usually a result of late diagnosis, efforts in the field of early detection are paramount to reducing cancer-related deaths and improving patient outcomes. Increasing evidence indicates that metastasis is an early event in patients with aggressive cancers, often occurring even before primary lesions are clinically detectable. Metastases are usually formed from cancer cells that spread to distant non-malignant tissues via the blood circulation, termed circulating tumour cells (CTCs). CTCs have been detected in patients with early stage cancers and, owing to their association with metastasis, might indicate the presence of aggressive disease, thus providing a possible means to expedite diagnosis and treatment initiation for such patients while avoiding overdiagnosis and overtreatment of those with slow-growing, indolent tumours. The utility of CTCs as an early diagnostic tool has been investigated, although further improvements in the efficiency of CTC detection are required. In this Perspective, we discuss the clinical significance of early haematogenous dissemination of cancer cells, the potential of CTCs to facilitate early detection of clinically relevant cancers, and the technological advances that might improve CTC capture and, thus, diagnostic performance in this setting.
Collapse
Affiliation(s)
- Rachel Lawrence
- Centre for Biomarkers and Therapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Melissa Watters
- Barts and London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Caitlin R Davies
- Centre for Biomarkers and Therapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Klaus Pantel
- Department of Tumour Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Yong-Jie Lu
- Centre for Biomarkers and Therapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
6
|
Pirone D, Montella A, Sirico DG, Mugnano M, Villone MM, Bianco V, Miccio L, Porcelli AM, Kurelac I, Capasso M, Iolascon A, Maffettone PL, Memmolo P, Ferraro P. Label-free liquid biopsy through the identification of tumor cells by machine learning-powered tomographic phase imaging flow cytometry. Sci Rep 2023; 13:6042. [PMID: 37055398 PMCID: PMC10101968 DOI: 10.1038/s41598-023-32110-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/21/2023] [Indexed: 04/15/2023] Open
Abstract
Image-based identification of circulating tumor cells in microfluidic cytometry condition is one of the most challenging perspectives in the Liquid Biopsy scenario. Here we show a machine learning-powered tomographic phase imaging flow cytometry system capable to provide high-throughput 3D phase-contrast tomograms of each single cell. In fact, we show that discrimination of tumor cells against white blood cells is potentially achievable with the aid of artificial intelligence in a label-free flow-cyto-tomography method. We propose a hierarchical machine learning decision-maker, working on a set of features calculated from the 3D tomograms of the cells' refractive index. We prove that 3D morphological features are adequately distinctive to identify tumor cells versus the white blood cell background in the first stage and, moreover, in recognizing the tumor type at the second decision step. Proof-of-concept experiments are shown, in which two different tumor cell lines, namely neuroblastoma cancer cells and ovarian cancer cells, are used against monocytes. The reported results allow claiming the identification of tumor cells with a success rate higher than 97% and with an accuracy over 97% in discriminating between the two cancer cell types, thus opening in a near future the route to a new Liquid Biopsy tool for detecting and classifying circulating tumor cells in blood by stain-free method.
Collapse
Affiliation(s)
- Daniele Pirone
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Annalaura Montella
- CEINGE Advanced Biotechnologies, Naples, Italy
- DMMBM, Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Daniele G Sirico
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Martina Mugnano
- Department of Chemical, Materials and Production Engineering, DICMaPI, University of Naples "Federico II", Piazzale Tecchio 80, 80125, Naples, Italy
| | - Massimiliano M Villone
- Department of Chemical, Materials and Production Engineering, DICMaPI, University of Naples "Federico II", Piazzale Tecchio 80, 80125, Naples, Italy
| | - Vittorio Bianco
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Lisa Miccio
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Anna Maria Porcelli
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
- Interdepartmental Centre for Industrial Research 'Scienze Della Vita e Tecnologie per La Salute', University of Bologna, Bologna, Italy
- Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Ivana Kurelac
- Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
- DIMEC, Department of Medical and Surgical Sciences, Centro di Studio e Ricerca Sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, 40138, Bologna, Italy
| | - Mario Capasso
- CEINGE Advanced Biotechnologies, Naples, Italy
- DMMBM, Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Achille Iolascon
- CEINGE Advanced Biotechnologies, Naples, Italy
- DMMBM, Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Pier Luca Maffettone
- Department of Chemical, Materials and Production Engineering, DICMaPI, University of Naples "Federico II", Piazzale Tecchio 80, 80125, Naples, Italy
| | - Pasquale Memmolo
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy.
| | - Pietro Ferraro
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy.
| |
Collapse
|
7
|
Hasanzadeh Kafshgari M, Hayden O. Advances in analytical microfluidic workflows for differential cancer diagnosis. NANO SELECT 2023. [DOI: 10.1002/nano.202200158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Morteza Hasanzadeh Kafshgari
- Heinz‐Nixdorf‐Chair of Biomedical Electronics Campus Klinikum München rechts der Isar TranslaTUM Technical University of Munich Munich Germany
| | - Oliver Hayden
- Heinz‐Nixdorf‐Chair of Biomedical Electronics Campus Klinikum München rechts der Isar TranslaTUM Technical University of Munich Munich Germany
| |
Collapse
|
8
|
Comparative application of microfluidic systems in circulating tumor cells and extracellular vesicles isolation; a review. Biomed Microdevices 2022; 25:4. [PMID: 36574057 DOI: 10.1007/s10544-022-00644-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 12/28/2022]
Abstract
Cancer is a prevalent cause of mortality globally, where early diagnosis leads to a reduced death rate. Many researchers' common strategies are based on personalized diagnostic methods with rapid response and high accuracy. This technology was developed by applying liquid biopsy instead of tissue biopsies in the case of tumor cell analysis that facilitates point-of-care testing for cancer diagnosis and treatment. In recent years, significant progress in microfluidic technology led to the successful isolation, analysis, and monitoring of cancer biomarkers in body liquid biopsy with merits like high sensitivity and flexibility, low sample usage, cost effective, and the ability of automation. The most critical and informative markers in body liquid refer to circulating tumor cells (CTCs) and extracellular vesicles derived from tumors (EVs) that carry various biomarkers in their structure (DNAs, proteins, and RNAs) as compared to ctDNA. The released ctDNA has a low half-life and decreased sensitivity due to large amounts of nucleic acid in serum. This review intends to highlight different cancer screening tests with a particular focus on the details regarding the only FDA-approved and awaiting technologies for FDA clearance to isolate CTCs and EVs based on microfluidics systems.
Collapse
|
9
|
Macaraniag C, Luan Q, Zhou J, Papautsky I. Microfluidic techniques for isolation, formation, and characterization of circulating tumor cells and clusters. APL Bioeng 2022; 6:031501. [PMID: 35856010 PMCID: PMC9288269 DOI: 10.1063/5.0093806] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cell (CTC) clusters that are shed from the primary tumor into the bloodstream are associated with a poor prognosis, elevated metastatic potential, higher proliferation rate, and distinct molecular features compared to single CTCs. Studying CTC clusters may give us information on the differences in the genetic profiles, somatic mutations, and epigenetic changes in circulating cells compared to the primary tumor and metastatic sites. Microfluidic systems offer the means of studying CTC clusters through the ability to efficiently isolate these rare cells from the whole blood of patients in a liquid biopsy. Microfluidics can also be used to develop in vitro models of CTC clusters and make possible their characterization and analysis. Ultimately, microfluidic systems can offer the means to gather insight on the complexities of the metastatic process, the biology of cancer, and the potential for developing novel or personalized therapies. In this review, we aim to discuss the advantages and challenges of the existing microfluidic systems for working with CTC clusters. We hope that an improved understanding of the role microfluidics can play in isolation, formation, and characterization of CTC clusters, which can lead to increased sophistication of microfluidic platforms in cancer research.
Collapse
Affiliation(s)
- Celine Macaraniag
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Qiyue Luan
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Jian Zhou
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
10
|
ALCAM: A Novel Surface Marker on EpCAMlow Circulating Tumor Cells. Biomedicines 2022; 10:biomedicines10081983. [PMID: 36009530 PMCID: PMC9405826 DOI: 10.3390/biomedicines10081983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Current strategies in circulating tumor cell (CTC) isolation in pancreatic cancer heavily rely on the EpCAM and cytokeratin cell status. EpCAM is generally not considered a good marker given its transitory change during Epithelial to Mesenchymal Transition (EMT) or reverse EMT. There is a need to identify other surface markers to capture the complete repertoire of PDAC CTCs. The primary objective of the study is to characterize alternate surface biomarkers to EpCAM on CTCs that express low or negligible levels of surface EpCAM in pancreatic cancer patients. Methods: Flow cytometry and surface mass spectrometry were used to identify proteins expressed on the surface of PDAC CTCs in culture. CTCs were grown under conditions of attachment and in co-culture with naïve neutrophils. Putative biomarkers were then validated in GEMMs and patient samples. Results: Surface proteomic profiling of CTCs identified several novel protein biomarkers. ALCAM was identified as a novel robust marker in GEMM models and in patient samples. Conclusions: We identified several novel surface biomarkers on CTCs expressed under differing conditions of culture. ALCAM was validated and identified as a novel alternate surface marker on EpCAMlow CTCs.
Collapse
|
11
|
Rahmanian M, Sartipzadeh Hematabad O, Askari E, Shokati F, Bakhshi A, Moghadam S, Olfatbakhsh A, Al Sadat Hashemi E, Khorsand Ahmadi M, Morteza Naghib S, Sinha N, Tel J, Eslami Amirabadi H, den Toonder JMJ, Majidzadeh-A K. A micropillar array-based microfluidic chip for label-free separation of circulating tumor cells: The best micropillar geometry? J Adv Res 2022; 47:105-121. [PMID: 35964874 PMCID: PMC10173300 DOI: 10.1016/j.jare.2022.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/22/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022] Open
Abstract
INTRODUCTION The information derived from the number and characteristics of circulating tumor cells (CTCs), is crucial to ensure appropriate cancer treatment monitoring. Currently, diverse microfluidic platforms have been developed for isolating CTCs from blood, but it remains a challenge to develop a low-cost, practical, and efficient strategy. OBJECTIVES This study aimed to isolate CTCs from the blood of cancer patients via introducing a new and efficient micropillar array-based microfluidic chip (MPA-Chip), as well as providing prognostic information and monitoring the treatment efficacy in cancer patients. METHODS We fabricated a microfluidic chip (MPA-Chip) containing arrays of micropillars with different geometries (lozenge, rectangle, circle, and triangle). We conducted numerical simulations to compare velocity and pressure profiles inside the micropillar arrays. Also, we experimentally evaluated the capture efficiency and purity of the geometries using breast and prostate cancer cell lines as well as a blood sample. Moreover, the device's performance was validated on 12 patients with breast cancer (BC) in different states. RESULTS The lozenge geometry was selected as the most effective and optimized micropillar design for CTCs isolation, providing high capture efficiency (>85 %), purity (>90 %), and viability (97 %). Furthermore, the lozenge MPA-chip was successfully validated by the detection of CTCs from 12 breast cancer (BC) patients, with non-metastatic (median number of 6 CTCs) and metastatic (median number of 25 CTCs) diseases, showing different prognoses. Also, increasing the chemotherapy period resulted in a decrease in the number of captured CTCs from 23 to 7 for the metastatic patient. The MPA-Chip size was only 0.25 cm2 and the throughput of a single chip was 0.5 ml/h, which can be increased by multiple MPA-Chips in parallel. CONCLUSION The lozenge MPA-Chip presented a novel micropillar geometry for on-chip CTC isolation, detection, and staining, and in the future, the possibilities can be extended to the culture of the CTCs.
Collapse
Affiliation(s)
- Mehdi Rahmanian
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran; Microsystems Research Section, Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Omid Sartipzadeh Hematabad
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Esfandyar Askari
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Farhad Shokati
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Atin Bakhshi
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Shiva Moghadam
- Breast Diseases Group, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Asiie Olfatbakhsh
- Breast Diseases Group, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Esmat Al Sadat Hashemi
- Breast Diseases Group, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Khorsand Ahmadi
- Microsystems Research Section, Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Nidhi Sinha
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jurjen Tel
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Hossein Eslami Amirabadi
- Microsystems Research Section, Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; AZAR Innovations, Utrecht, the Netherlands
| | - Jaap M J den Toonder
- Microsystems Research Section, Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
12
|
Han J, Lu C, Shen M, Sun X, Mo X, Yang G. Fast, Reusable, Cell Uniformly Distributed Membrane Filtration Device for Separation of Circulating Tumor Cells. ACS OMEGA 2022; 7:20761-20767. [PMID: 35755342 PMCID: PMC9219081 DOI: 10.1021/acsomega.2c01153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
Isolation of circulating tumor cells (CTCs) is of great significance for the diagnosis, prognosis, and treatment of metastatic cancer. Among CTC capture methods independent of antibodies, membrane filtration-based methods have the advantages of simplicity, rapidity, and high throughput but usually have problems such as clogging, high pressure drop, and impaired cell viability. In this study, we designed and tested a reusable device that used horizontal rotor and fluid-assisted separation to capture CTCs by centrifugal membrane filtration, achieving simple, fast, highly efficient, and viable cell capture on traditional centrifuge. The average capture efficiency was 95.8% for different types of cancer cells with >90% survival, and the removal of white blood cells can reach 99.72% under four times cleaning of the membrane after filtration. A further clinic demo was performed using the device to detect residual leukemic cells in patients; the results showed a 10-fold enrichment of the leukemic cells in peripheral blood samples. Taken together, the simple, robust, and efficient CTC capture device may have the potential for clinic routine detection and analysis of circulating tumor cells.
Collapse
Affiliation(s)
- Jintao Han
- State
Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Chunyang Lu
- State
Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Mengzhu Shen
- Beijing
Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking
University People’s Hospital, Beijing 100044, China
| | - Xiaoyi Sun
- State
Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Xiaodong Mo
- Beijing
Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking
University People’s Hospital, Beijing 100044, China
| | - Gen Yang
- State
Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
- Wenzhou
Institute, University of Chinese Academy
of Sciences, Wenzhou 352001, China
| |
Collapse
|
13
|
Cai S, Ma Z, Ge Z, Yang W. Recent advances in optically induced di-electrophoresis and its biomedical applications. Biomed Microdevices 2022; 24:22. [PMID: 35689721 DOI: 10.1007/s10544-022-00620-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 11/02/2022]
Abstract
The development of the micro/nano science and technology has promoted the evolvement of human civilization tremendously. The advancement of the micro/nano science and technology highly depends on the progress of the micro/nano manipulation techniques, and the micro/nano-scaled manipulation level is the critical sign of the micro/nano science and technology. This review, aimed at the demand and the challenge of the micro/nano material and biomedical fields and related to the scientific issues and implementation techniques of the optically induced di-electrophoresis (ODEP). We explained its working principle, manipulating method, and influencing factors of ODEP force to a certain extent. A number of application fields based-ODEP technology and specific applications so far are summarized and reviewed. Finally, some perspectives are provided on current development trends, future research directions, and challenges of ODEP.
Collapse
Affiliation(s)
- Shuxiang Cai
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai, 264005, China
| | - Zheng Ma
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai, 264005, China
| | - Zhixing Ge
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Wenguang Yang
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai, 264005, China.
| |
Collapse
|
14
|
Descamps L, Le Roy D, Deman AL. Microfluidic-Based Technologies for CTC Isolation: A Review of 10 Years of Intense Efforts towards Liquid Biopsy. Int J Mol Sci 2022; 23:ijms23041981. [PMID: 35216097 PMCID: PMC8875744 DOI: 10.3390/ijms23041981] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/01/2023] Open
Abstract
The selection of circulating tumor cells (CTCs) directly from blood as a real-time liquid biopsy has received increasing attention over the past ten years, and further analysis of these cells may greatly aid in both research and clinical applications. CTC analysis could advance understandings of metastatic cascade, tumor evolution, and patient heterogeneity, as well as drug resistance. Until now, the rarity and heterogeneity of CTCs have been technical challenges to their wider use in clinical studies, but microfluidic-based isolation technologies have emerged as promising tools to address these limitations. This review provides a detailed overview of latest and leading microfluidic devices implemented for CTC isolation. In particular, this study details must-have device performances and highlights the tradeoff between recovery and purity. Finally, the review gives a report of CTC potential clinical applications that can be conducted after CTC isolation. Widespread microfluidic devices, which aim to support liquid-biopsy-based applications, will represent a paradigm shift for cancer clinical care in the near future.
Collapse
Affiliation(s)
- Lucie Descamps
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INSA Lyon, Ecole Centrale de Lyon, CPE Lyon, INL, UMR5270, 69622 Villeurbanne, France;
| | - Damien Le Roy
- Institut Lumière Matière ILM-UMR 5306, CNRS, Université Lyon 1, 69622 Villeurbanne, France;
| | - Anne-Laure Deman
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INSA Lyon, Ecole Centrale de Lyon, CPE Lyon, INL, UMR5270, 69622 Villeurbanne, France;
- Correspondence:
| |
Collapse
|
15
|
Yang Y, Pang W, Zhang H, Cui W, Jin K, Sun C, Wang Y, Zhang L, Ren X, Duan X. Manipulation of single cells via a Stereo Acoustic Streaming Tunnel (SteAST). MICROSYSTEMS & NANOENGINEERING 2022; 8:88. [PMID: 35935274 PMCID: PMC9352906 DOI: 10.1038/s41378-022-00424-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 05/19/2023]
Abstract
At the single-cell level, cellular parameters, gene expression and cellular function are assayed on an individual but not population-average basis. Essential to observing and analyzing the heterogeneity and behavior of these cells/clusters is the ability to prepare and manipulate individuals. Here, we demonstrate a versatile microsystem, a stereo acoustic streaming tunnel, which is triggered by ultrahigh-frequency bulk acoustic waves and highly confined by a microchannel. We thoroughly analyze the generation and features of stereo acoustic streaming to develop a virtual tunnel for observation, pretreatment and analysis of cells for different single-cell applications. 3D reconstruction, dissociation of clusters, selective trapping/release, in situ analysis and pairing of single cells with barcode gel beads were demonstrated. To further verify the reliability and robustness of this technology in complex biosamples, the separation of circulating tumor cells from undiluted blood based on properties of both physics and immunity was achieved. With the rich selection of handling modes, the platform has the potential to be a full-process microsystem, from pretreatment to analysis, and used in numerous fields, such as in vitro diagnosis, high-throughput single-cell sequencing and drug development.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin, 300072 China
| | - Wei Pang
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin, 300072 China
| | - Hongxiang Zhang
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin, 300072 China
| | - Weiwei Cui
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin, 300072 China
| | - Ke Jin
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin, 300072 China
| | - Chongling Sun
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin, 300072 China
| | - Yanyan Wang
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin, 300072 China
| | - Lin Zhang
- Tianjin Medical University Cancer Institute & Hospital, Tianjin Medical University, Tianjin, 300072 China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute & Hospital, Tianjin Medical University, Tianjin, 300072 China
| | - Xuexin Duan
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin, 300072 China
| |
Collapse
|
16
|
Shimmyo N, Furuhata M, Yamada M, Utoh R, Seki M. Process simplification and structure design of parallelized microslit isolator for physical property-based capture of tumor cells. Analyst 2022; 147:1622-1630. [DOI: 10.1039/d2an00052k] [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 facile, scalable, and highly efficient approach to physically capturing CTCs from blood samples has been developed using a microfluidic isolator with parallelized microslit channels.
Collapse
Affiliation(s)
- Natsumi Shimmyo
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, 263-8522, Japan
| | - Makoto Furuhata
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, 263-8522, Japan
| | - Masumi Yamada
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, 263-8522, Japan
| | - Rie Utoh
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, 263-8522, Japan
| | - Minoru Seki
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, 263-8522, Japan
| |
Collapse
|
17
|
He S, Wei J, Ding L, Yang X, Wu Y. State-of-the-arts techniques and current evolving approaches in the separation and detection of circulating tumor cell. Talanta 2021; 239:123024. [PMID: 34952370 DOI: 10.1016/j.talanta.2021.123024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/01/2023]
Abstract
Circulating tumor cells (CTCs) are cancer cells that shed from the primary tumor and then enter the circulatory system, a small part of which may evolve into metastatic cancer under appropriate microenvironment conditions. The detection of CTCs is a truly noninvasive, dynamic monitor for disease changes, which has considerable clinical implications in the selection of targeted drugs. However, their inherent rarity and heterogeneity pose significant challenges to their isolation and detection. Even the "gold standard", CellSearch™, suffers from high expenses, low capture efficiency, and the consumption of time. With the advancement of CTCs analysis technologies in recent years, the yield and efficiency of CTCs enrichment have gradually been improved, as well as detection sensitivity. In this review, the isolation and detection strategies of CTCs have been completely described and the potential directions for future research and development have also been highlighted through analyzing the challenges faced by current strategies.
Collapse
Affiliation(s)
- Sitian He
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| | - Jinlan Wei
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaonan Yang
- School of Information Engineering, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
18
|
Rushton AJ, Nteliopoulos G, Shaw JA, Coombes RC. A Review of Circulating Tumour Cell Enrichment Technologies. Cancers (Basel) 2021; 13:cancers13050970. [PMID: 33652649 PMCID: PMC7956528 DOI: 10.3390/cancers13050970] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Circulating tumour cells (CTCs) are cancer cells shed into the bloodstream from tumours and their analysis can provide important insights into cancer detection and monitoring, with the potential to direct personalised therapies for the patient. These CTCs are rare in the blood, which makes their detection and enrichment challenging and to date, only one technology (the CellSearch) has gained FDA approval for determining the prognosis of patients with advanced breast, prostate and colorectal cancers. Here, we review the wide range of enrichment technologies available to isolate CTCs from other blood components and highlight the important characteristics that new technologies should possess for routine clinical use. Abstract Circulating tumour cells (CTCs) are the precursor cells for the formation of metastatic disease. With a simple blood draw, liquid biopsies enable the non-invasive sampling of CTCs from the blood, which have the potential to provide important insights into cancer detection and monitoring. Since gaining FDA approval in 2004, the CellSearch system has been used to determine the prognosis of patients with metastatic breast, prostate and colorectal cancers. This utilises the cell surface marker Epithelial Cell Adhesion Molecule (EpCAM), to enrich CTCs, and many other technologies have adopted this approach. More recently, the role of mesenchymal-like CTCs in metastasis formation has come to light. It has been suggested that these cells are more aggressive metastatic precursors than their epithelial counterparts; however, mesenchymal CTCs remain undetected by EpCAM-based enrichment methods. This has prompted the development of a variety of ‘label free’ enrichment technologies, which exploit the unique physical properties of CTCs (such as size and deformability) compared to other blood components. Here, we review a wide range of both immunocapture and label free CTC enrichment technologies, summarising the most significant advantages and disadvantages of each. We also highlight the important characteristics that technologies should possess for routine clinical use, since future developments could have important clinical implications, with the potential to direct personalised therapies for patients with cancer.
Collapse
Affiliation(s)
- Amelia J. Rushton
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
- Correspondence:
| | - Georgios Nteliopoulos
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
| | - Jacqueline A. Shaw
- Leicester Cancer Research Centre, University of Leicester, Leicester LE2 7LX, UK;
| | - R. Charles Coombes
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
| |
Collapse
|
19
|
Lu C, Xu J, Han J, Li X, Xue N, Li J, Wu W, Sun X, Wang Y, Ouyang Q, Yang G, Luo C. A novel microfluidic device integrating focus-separation speed reduction design and trap arrays for high-throughput capture of circulating tumor cells. LAB ON A CHIP 2020; 20:4094-4105. [PMID: 33089845 DOI: 10.1039/d0lc00631a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Isolation and analysis of circulating tumor cells (CTCs) from peripheral blood provides a potential way to detect and characterize cancer. Existing technologies to separate or capture CTCs from whole blood still have issues with sample throughput, separation efficiency or stable efficiency at different flow rates. Here, we proposed a new concept to capture rare CTCs from blood by integrating a triangular prism array-based capture apparatus with streamline-based focus-separation speed reduction design. The focus-separation design could focus and maintain CTCs, while removing a considerable proportion of liquid (98.9%) containing other blood cells to the outlet, therefore, a high CTC capture efficiency could be achieved in the trap arrays with a high initial flow rate. It is worth mentioning that the new design works well over a wide range of flow rates, so it does not require the stability of the flow rate. The results showed that this novel integrated chip can achieve a sample throughput from 5 to 40 mL h-1 with a stable and high CTC capture efficiency (up to 94.8%) and high purity (up to 4 log white blood cells/WBC depletion). The clinical experiment showed that CTCs including CTC clusters were detected in 11/11 (100.0%) patients (mean = 31 CTCs mL-1, median = 25 CTCs mL-1). In summary, our chip enriches and captures CTCs based on physical properties, and it is simple, cheap, fast, and efficient and has low requirements on flow rate, which is very suitable for large-scale application of CTC testing in clinics.
Collapse
Affiliation(s)
- Chunyang Lu
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China.
| | - Jian Xu
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, China.
| | - Jintao Han
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China.
| | - Xiao Li
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ningtao Xue
- Jining No. 2 People's Hospital, Jining 272049, China
| | - Jinsong Li
- Jining No. 2 People's Hospital, Jining 272049, China
| | - Wenhua Wu
- Jining No. 2 People's Hospital, Jining 272049, China
| | - Xinlei Sun
- Jining Tumor Hospital, Jining 272007, China
| | - Yugang Wang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China.
| | - Qi Ouyang
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, China. and Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Gen Yang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China.
| | - Chunxiong Luo
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, China. and Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
20
|
Electrochemical Detection and Point-of-Care Testing for Circulating Tumor Cells: Current Techniques and Future Potentials. SENSORS 2020; 20:s20216073. [PMID: 33114569 PMCID: PMC7663783 DOI: 10.3390/s20216073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/18/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022]
Abstract
Circulating tumor cells (CTCs) are tumor cells that escaped from the primary tumor or the metastasis into the blood and they play a major role in the initiation of metastasis and tumor recurrence. Thus, it is widely accepted that CTC is the main target of liquid biopsy. In the past few decades, the separation of CTC based on the electrochemical method has attracted widespread attention due to its convenience, rapidness, low cost, high sensitivity, and no need for complex instruments and equipment. At present, CTC detection is not widely used in the clinic due to various reasons. Point-of-care CTC detection provides us with a possibility, which is sensitive, fast, cheap, and easy to operate. More importantly, the testing instrument is small and portable, and the testing does not require specialized laboratories and specialized clinical examiners. In this review, we summarized the latest developments in the electrochemical-based CTC detection and point-of-care CTC detection, and discussed the challenges and possible trends.
Collapse
|
21
|
Miccio L, Cimmino F, Kurelac I, Villone MM, Bianco V, Memmolo P, Merola F, Mugnano M, Capasso M, Iolascon A, Maffettone PL, Ferraro P. Perspectives on liquid biopsy for label‐free detection of “circulating tumor cells” through intelligent lab‐on‐chips. VIEW 2020. [DOI: 10.1002/viw.20200034] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Lisa Miccio
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | | | - Ivana Kurelac
- Dipartimento di Scienze Mediche e Chirurgiche Università di Bologna Bologna Italy
- Centro di Ricerca Biomedica Applicata (CRBA) Università di Bologna Bologna Italy
| | - Massimiliano M. Villone
- Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale Università degli Studi di Napoli “Federico II” Napoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Vittorio Bianco
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Pasquale Memmolo
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Francesco Merola
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Martina Mugnano
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Mario Capasso
- CEINGE Biotecnologie Avanzate Naples Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche Università degli Studi di Napoli Federico II Naples Italy
| | - Achille Iolascon
- CEINGE Biotecnologie Avanzate Naples Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche Università degli Studi di Napoli Federico II Naples Italy
| | - Pier Luca Maffettone
- Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale Università degli Studi di Napoli “Federico II” Napoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Pietro Ferraro
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| |
Collapse
|
22
|
Li T, Liu Y, Zhang W, Lin L, Zhang J, Xiong Y, Nie L, Liu X, Li H, Wang W. A rapid liquid biopsy of lung cancer by separation and detection of exfoliated tumor cells from bronchoalveolar lavage fluid with a dual-layer "PERFECT" filter system. Theranostics 2020; 10:6517-6529. [PMID: 32483467 PMCID: PMC7255025 DOI: 10.7150/thno.44274] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 04/19/2020] [Indexed: 12/24/2022] Open
Abstract
Separation and detection of exfoliated tumor cells (ETCs) from bronchoalveolar lavage fluid (BALF), namely the liquid biopsy of BALF, has been proved to be a valuable tool for the diagnosis of lung cancer. Herein, we established a rapid liquid biopsy of BALF based on a dual-layer PERFECT (precise, efficient, rapid, flexible, easy-to-operate, controllable and thin) filter system for the first time. Methods: The dual-layer PERFECT filter system consists of an upper-layer filter with large micropores (feature size of 49.4 ± 0.5 μm) and a lower-layer filter with small micropores (9.1 ± 0.1 μm). The upper-layer filter contributes to the isolation of cell clusters and removal of mucus from BALF samples, meanwhile the lower-layer one targets for the separation of single ETCs. First, separation of 10000 spiked A549s (cultured lung cancer cells) from 10 mL clinical BALF samples (n=3) were performed to investigate the performance of the proposed system in rare cell separation. Furthermore, separation and detection of ETCs and ETC clusters from clinical BALF samples were performed with this system to test its efficacy and compared with the routine cytocentrifuge. The clinical BALF samples were collected from 33 lung cancer-suspected patients with visible lesions under bronchoscope. The final histopathological results showed that 20 samples were from lung cancer positive patients while the other 13 cases were from lung cancer negative patients. Results: The recovery rate of spiked A549 cells from clinical BALF samples with the developed system (89.8 ± 5.2%) is significantly higher than that with the cytocentrifuge (13.6 ± 7.8%). In the preliminary clinical trial, although 33 clinical BALF samples with volume ranging from 6 mL to 18 mL showed greatly varied turbidity, filtrations could be finished within 3 min for 54.6% of samples (18/33), and 10 min at most for the rest. The dual-layer PERFECT filter system is proved to have a much higher sensitivity (80.0%, 95% CI: 55.7%-93.4%) than the routine cytocentrifuge (45.0%, 95% CI: 23.8%-68.0%), p=0.016 (McNemar test, two-tail). Moreover, the sensitivity of this platform is neither interfered by the variations of turbidity of the BALF samples, nor associated with the types of lung cancer. Conclusions: The easy and rapid processing of BALF samples with varying volume and turbidity, competitive sensitivity and good versatility for different lung cancer types will make the established dual-layer PERFECT filter system a promising approach for the liquid biopsy of BALF. The high-performance BALF-based liquid biopsy will improve the cytopathological identification and diagnosis of lung cancer.
Collapse
Affiliation(s)
- Tingyu Li
- Institute of Microelectronics, Peking University, Beijing, 100871, China
| | - Yaoping Liu
- Institute of Microelectronics, Peking University, Beijing, 100871, China
- Antimicrobial Resistance (AMR) and Critical Analytics for Manufacturing Personalized-Medicine (CAMP) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore, 138602, Singapore
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Lianjun Lin
- Department of Geriatrics, Peking University First Hospital, Beijing, 100034, China
| | - Jixin Zhang
- Department of Pathology, Peking University First Hospital, Beijing, 100034, China
| | - Yan Xiong
- Department of Pathology, Peking University First Hospital, Beijing, 100034, China
| | - Ligong Nie
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Xinmin Liu
- Department of Geriatrics, Peking University First Hospital, Beijing, 100034, China
| | - Haichao Li
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Wei Wang
- Institute of Microelectronics, Peking University, Beijing, 100871, China
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Beijing, 100871, China
- Frontiers Science Center for Nano-optoelectronics, Peking University, Beijing, 100871, China
| |
Collapse
|
23
|
Sabirova A, Pisig F, Rayapuram N, Hirt H, Nunes SP. Nanofabrication of Isoporous Membranes for Cell Fractionation. Sci Rep 2020; 10:6138. [PMID: 32273573 PMCID: PMC7145805 DOI: 10.1038/s41598-020-62937-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/15/2020] [Indexed: 12/14/2022] Open
Abstract
Cell fractionations and other biological separations frequently require several steps. They could be much more effectively done by filtration, if isoporous membranes would be available with high pore density, and sharp pore size distribution in the micro- and nanoscale. We propose a combination of two scalable methods, photolithography and dry reactive ion etching, to fabricate a series of polyester membranes with isopores of size 0.7 to 50 μm and high pore density with a demonstrated total area of 38.5 cm2. The membranes have pore sizes in the micro- and submicro-range, and pore density 10-fold higher than track-etched analogues, which are the only commercially available isoporous polymeric films. Permeances of 220,000 L m−2 h−1bar−1 were measured with pore size 787 nm. The method does not require organic solvents and can be applied to many homopolymeric materials. The pore reduction from 2 to 0.7 μm was obtained by adding a step of chemical vapor deposition. The isoporous system was successfully demonstrated for the organelle fractionation of Arabidopsis homogenates and could be potentially extended to other biological fractionations.
Collapse
Affiliation(s)
- Ainur Sabirova
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE) Division, Advanced Membranes and Porous Materials Center, 23955-6900, Thuwal, Saudi Arabia
| | - Florencio Pisig
- King Abdullah University of Science and Technology (KAUST), Nanofabrication Core Laboratory, 23955-6900, Thuwal, Saudi Arabia
| | - Naganand Rayapuram
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE) Division, Center for Desert Agriculture, 23955-6900, Thuwal, Saudi Arabia
| | - Heribert Hirt
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE) Division, Center for Desert Agriculture, 23955-6900, Thuwal, Saudi Arabia
| | - Suzana P Nunes
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE) Division, Advanced Membranes and Porous Materials Center, 23955-6900, Thuwal, Saudi Arabia.
| |
Collapse
|
24
|
Chen C, Xu D, Bai S, Yu Z, Zhu Y, Xing X, Chen H. Dynamic screening and printing of single cells using a microfluidic chip with dual microvalves. LAB ON A CHIP 2020; 20:1227-1237. [PMID: 32100799 DOI: 10.1039/d0lc00040j] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Inoculation of single cells into separate culture chambers is one of the key requirements in single-cell analysis. This paper reports an innovative microfluidic chip integrating two pneumatic microvalves to screen and print single cells onto a well plate. The upper and lower size limits of cells can be dynamically controlled by regulating the deformation of two adjacent microvalves. Numerical simulations were employed to systematically study the influence of membrane dimensions and pressure on the deflection of a valve. A mathematical model was then modified to predict the size of cells captured by a microvalve at various pressures. The membrane deflection was further studied using confocal imaging. The critical pressure trapping beads of various sizes was experimentally determined. These experiments validated the accuracy of both numerical simulations and the mathematical model. Furthermore, single beads and endothelial cells with the desired size range were screened using dual valves and printed onto well plates with 100% efficiency. Viability studies suggested that the screening process had no significant impact on cells. This device enables dynamic regulation of both the lower and the upper size limits of cells for printing. It has significant application potential in inoculating cells with desired sizes for various fields such as clonal expansion, monoclonality development and single-cell genomic studies.
Collapse
Affiliation(s)
- Chang Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China.
| | | | | | | | | | | | | |
Collapse
|
25
|
Cheng Y, Shen J, Yuan L, Yang Y, Shen X, Qian H, Yu L, Li R, Lv X, Yan T, Li Y, Wang L, Liu B. A novel device to capture circulating tumor cells: Quantification and molecular analysis in lung cancer patients. J Biomater Appl 2020; 35:49-58. [PMID: 32223499 DOI: 10.1177/0885328220914408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Here, we describe a novel microfilter device to capture circulating tumor cells in an efficient and low-cost manner. Then, we validated the safety and clinical utility of the novel microfilter device. We next performed mutation analysis from circulating tumor cells collected from lung cancer patients using this new device. Our results indicate that this microfilter system can be used to investigate the genome landscape of circulating tumor cells collected from lung cancer patients. Further, our results highlight a proof-of-concept demonstration indicating that circulating tumor cell can be used for mutation profiling during tumor evolution, therapy prediction, and monitoring, with immediate clinical applicability.
Collapse
Affiliation(s)
- Yuxin Cheng
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Jie Shen
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Ling Yuan
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Yan Yang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xiaoyan Shen
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Hanqing Qian
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Rutian Li
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xin Lv
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Tingting Yan
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Yan Li
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lifeng Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| |
Collapse
|
26
|
Lin Z, Luo G, Du W, Kong T, Liu C, Liu Z. Recent Advances in Microfluidic Platforms Applied in Cancer Metastasis: Circulating Tumor Cells' (CTCs) Isolation and Tumor-On-A-Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1903899. [PMID: 31747120 DOI: 10.1002/smll.201903899] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/13/2019] [Indexed: 05/03/2023]
Abstract
Cancer remains the leading cause of death worldwide despite the enormous efforts that are made in the development of cancer biology and anticancer therapeutic treatment. Furthermore, recent studies in oncology have focused on the complex cancer metastatic process as metastatic disease contributes to more than 90% of tumor-related death. In the metastatic process, isolation and analysis of circulating tumor cells (CTCs) play a vital role in diagnosis and prognosis of cancer patients at an early stage. To obtain relevant information on cancer metastasis and progression from CTCs, reliable approaches are required for CTC detection and isolation. Additionally, experimental platforms mimicking the tumor microenvironment in vitro give a better understanding of the metastatic microenvironment and antimetastatic drugs' screening. With the advancement of microfabrication and rapid prototyping, microfluidic techniques are now increasingly being exploited to study cancer metastasis as they allow precise control of fluids in small volume and rapid sample processing at relatively low cost and with high sensitivity. Recent advancements in microfluidic platforms utilized in various methods for CTCs' isolation and tumor models recapitulating the metastatic microenvironment (tumor-on-a-chip) are comprehensively reviewed. Future perspectives on microfluidics for cancer metastasis are proposed.
Collapse
Affiliation(s)
- Zhengjie Lin
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Guanyi Luo
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Weixiang Du
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Tiantian Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Changkun Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Zhou Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
27
|
Suzuki T, Kaji N, Yasaki H, Yasui T, Baba Y. Mechanical Low-Pass Filtering of Cells for Detection of Circulating Tumor Cells in Whole Blood. Anal Chem 2020; 92:2483-2491. [PMID: 31922717 DOI: 10.1021/acs.analchem.9b03939] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The detection of circulating tumor cells (CTCs) from liquid biopsies using microfluidic devices is attracting a considerable amount of attention as a new, less-invasive cancer diagnostic and prognostic method. One of the drawbacks of the existing antibody-based detection systems is the false negatives for epithelial cell adhesion molecule detection of CTCs. Here we report a mechanical low-pass filtering technique based on a microfluidic constriction and electrical current sensing system for the novel CTC detection in whole blood without any specific antigen-antibody interaction or biochemical modification of the cell surface. The mechanical response of model cells of CTCs, such as HeLa, A549, and MDA-MB-231 cells, clearly demonstrated different behaviors from that of Jurkat cells, a human T-lymphocyte cell line, when they passed through the 6-μm wide constriction channel. A 6-μm wide constriction channel was determined as the optimum size to identify CTCs in whole blood with an accuracy greater than 95% in tens of milliseconds. The mechanical filtering of cells at a single cell level was achieved from whole blood without any pretreatment (e.g., dilution of lysing) and prelabeling (e.g., fluorophores or antibodies).
Collapse
Affiliation(s)
| | - Noritada Kaji
- JST, PRESTO , 4-1-8 Honcho , Kawaguchi , Saitama 332-0012 , Japan.,Department of Applied Chemistry, Graduate School of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka 819-0395 , Japan
| | | | - Takao Yasui
- JST, PRESTO , 4-1-8 Honcho , Kawaguchi , Saitama 332-0012 , Japan
| | - Yoshinobu Baba
- Health Research Institute , National Institute of Advanced Industrial Science and Technology (AIST) , Hayashi-cho 2217-14 , Takamatsu 761-0395 , Japan.,School of Pharmacy, College of Pharmacy , Kaohsiung Medical University , 100, Shih-Chuan First Road , Kaohsiung , 807 , Taiwan , R.O.C
| |
Collapse
|
28
|
Zhu S, Jiang F, Han Y, Xiang N, Ni Z. Microfluidics for label-free sorting of rare circulating tumor cells. Analyst 2020; 145:7103-7124. [DOI: 10.1039/d0an01148g] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A review discussing the working principles and performances of label-free CTC sorting methods.
Collapse
Affiliation(s)
- Shu Zhu
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Fengtao Jiang
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Yu Han
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Nan Xiang
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Zhonghua Ni
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| |
Collapse
|
29
|
Abstract
Circulating tumor cells (CTCs) are responsible for the metastatic spread of cancer and therefore are extremely valuable not only for basic research on cancer metastasis but also as potential biomarkers in diagnosing and managing cancer in the clinic. While relatively non-invasive access to the blood tissue presents an opportunity, CTCs are mixed with approximately billion-times more-populated blood cells in circulation. Therefore, the accuracy of technologies for reliable enrichment of the rare CTC population from blood samples is critical to the success of downstream analyses. The focus of this chapter is to provide the reader an overview of significant advances made in the development of diverse CTC enrichment technologies by presenting the strengths of individual techniques in addition to specific challenges remaining to be addressed.
Collapse
|
30
|
Wu X, Bai Z, Wang L, Cui G, Yang M, Yang Q, Ma B, Song Q, Tian D, Ceyssens F, Puers R, Kraft M, Zhao W, Wen L. Magnetic Cell Centrifuge Platform Performance Study with Different Microsieve Pore Geometries. SENSORS (BASEL, SWITZERLAND) 2019; 20:E48. [PMID: 31861791 PMCID: PMC6983067 DOI: 10.3390/s20010048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/04/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022]
Abstract
The detection and analysis of circulating tumor cells (CTCs) plays a crucial role in clinical practice. However, the heterogeneity and rarity of CTCs make their capture and separation from peripheral blood very difficult while maintaining their structural integrity and viability. We previously demonstrated the effectiveness of the Magnetic Cell Centrifuge Platform (MCCP), which combined the magnetic-labeling cell separation mechanism with the size-based method. In this paper, a comparison of the effectiveness of different microsieve pore geometries toward MCCP is demonstrated to improve the yield of the target cell capture. Firstly, models of a trapped cell with rectangular and circular pore geometries are presented to compare the contact force using finite element numerical simulations. The device performance is then evaluated with both constant pressure and constant flow rate experimental conditions. In addition, the efficient isolation of magnetically labeled Hela cells with red fluorescent proteins (target cells) from Hela cells with green fluorescent protein (background cells) is validated. The experimental results show that the circular sieves yield 97% purity of the target cells from the sample with a throughput of up to 2 μL/s and 66-fold sample enrichment. This finding will pave the way for the design of a higher efficient MCCP systems.
Collapse
Affiliation(s)
- Xinyu Wu
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Zhongyang Bai
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Lin Wang
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Guangchao Cui
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Mengzheng Yang
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Qing Yang
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Bo Ma
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
| | - Qinglin Song
- Qingdao Goertek Microelectronics Research Institute Co., Ltd., Qingdao 266104, China
| | - Dewen Tian
- Qingdao Goertek Microelectronics Research Institute Co., Ltd., Qingdao 266104, China
| | - Frederik Ceyssens
- ESAT-MICAS, KU Leuven, Kasteelpark Arenberg 10, B-3001 Leuven, Belgium
| | - Robert Puers
- ESAT-MICAS, KU Leuven, Kasteelpark Arenberg 10, B-3001 Leuven, Belgium
| | - Michael Kraft
- ESAT-MICAS, KU Leuven, Kasteelpark Arenberg 10, B-3001 Leuven, Belgium
| | - Weisheng Zhao
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Lianggong Wen
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| |
Collapse
|
31
|
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: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
32
|
Insights on CTC Biology and Clinical Impact Emerging from Advances in Capture Technology. Cells 2019; 8:cells8060553. [PMID: 31174404 PMCID: PMC6627072 DOI: 10.3390/cells8060553] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 01/01/2023] Open
Abstract
Circulating tumor cells (CTCs) and circulating tumor microemboli (CTM) have been shown to correlate negatively with patient survival. Actual CTC counts before and after treatment can be used to aid in the prognosis of patient outcomes. The presence of circulating tumor materials (CTMat) can advertise the presence of metastasis before clinical presentation, enabling the early detection of relapse. Importantly, emerging evidence is indicating that cancer treatments can actually increase the incidence of CTCs and metastasis in pre-clinical models. Subsequently, the study of CTCs, their biology and function are of vital importance. Emerging technologies for the capture of CTC/CTMs and CTMat are elucidating vitally important biological and functional information that can lead to important alterations in how therapies are administered. This paves the way for the development of a "liquid biopsy" where treatment decisions can be informed by information gleaned from tumor cells and tumor cell debris in the blood.
Collapse
|
33
|
Qin L, Zhou W, Zhang S, Cheng B, Wang S, Li S, Yang Y, Wang S, Liu K, Zhang N. Highly Efficient Isolation of Circulating Tumor Cells Using a Simple Wedge-Shaped Microfluidic Device. IEEE Trans Biomed Eng 2019; 66:1536-1541. [DOI: 10.1109/tbme.2018.2875361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
34
|
Ribeiro-Samy S, Oliveira MI, Pereira-Veiga T, Muinelo-Romay L, Carvalho S, Gaspar J, Freitas PP, López-López R, Costa C, Diéguez L. Fast and efficient microfluidic cell filter for isolation of circulating tumor cells from unprocessed whole blood of colorectal cancer patients. Sci Rep 2019; 9:8032. [PMID: 31142796 PMCID: PMC6541613 DOI: 10.1038/s41598-019-44401-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
Liquid biopsy offers unique opportunities for low invasive diagnosis, real-time patient monitoring and treatment selection. The phenotypic and molecular profile of circulating tumor cells (CTCs) can provide key information about the biology of tumor cells, contributing to personalized therapy. CTC isolation is still challenging, mainly due to their heterogeneity and rarity. To overcome this limitation, a microfluidic chip for label-free isolation of CTCs from peripheral blood was developed. This device, the CROSS chip, captures CTCs based on their size and deformability with an efficiency of 70%. Using 2 chips, 7.5 ml of whole blood are processed in 47 minutes with high purity, as compared to similar technologies and assessed by in situ immunofluorescence. The CROSS chip performance was compared to the CellSearch system in a set of metastatic colorectal cancer patients, resulting in higher capture of DAPI+/CK+/CD45- CTCs in all individuals tested. Importantly, CTC enumeration by CROSS chip enabled stratification of patients with different prognosis. Lastly, cells isolated in the CROSS chip were lysed and further subjected to molecular characterization by droplet digital PCR, which revealed a mutation in the APC gene for most patient samples analyzed, confirming their colorectal origin and the versatility of the technology for downstream applications.
Collapse
Affiliation(s)
- Silvina Ribeiro-Samy
- Department of Life Sciences, International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga s/n, 4715-330, Braga, Portugal
| | - Marta I Oliveira
- Department of Life Sciences, International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga s/n, 4715-330, Braga, Portugal
| | - Thais Pereira-Veiga
- Roche-CHUS Joint Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706, Santiago de Compostela, Spain
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Sandra Carvalho
- Department of Life Sciences, International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga s/n, 4715-330, Braga, Portugal
| | - João Gaspar
- Department of Micro and Nanofabrication, International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga s/n, 4715-330, Braga, Portugal
| | - Paulo P Freitas
- Department of Nanoelectronics Engineering, International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga s/n, 4715-330, Braga, Portugal
| | - Rafael López-López
- Roche-CHUS Joint Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706, Santiago de Compostela, Spain
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Clotilde Costa
- Roche-CHUS Joint Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain.
| | - Lorena Diéguez
- Department of Life Sciences, International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga s/n, 4715-330, Braga, Portugal.
| |
Collapse
|
35
|
Ruzycka M, Cimpan MR, Rios-Mondragon I, Grudzinski IP. Microfluidics for studying metastatic patterns of lung cancer. J Nanobiotechnology 2019; 17:71. [PMID: 31133019 PMCID: PMC6537392 DOI: 10.1186/s12951-019-0492-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/04/2019] [Indexed: 01/09/2023] Open
Abstract
The incidence of lung cancer continues to rise worldwide. Because the aggressive metastasis of lung cancer cells is the major drawback of successful therapies, the crucial challenge of modern nanomedicine is to develop diagnostic tools to map the molecular mechanisms of metastasis in lung cancer patients. In recent years, microfluidic platforms have been given much attention as tools for novel point-of-care diagnostic, an important aspect being the reconstruction of the body organs and tissues mimicking the in vivo conditions in one simple microdevice. Herein, we present the first comprehensive overview of the microfluidic systems used as innovative tools in the studies of lung cancer metastasis including single cancer cell analysis, endothelial transmigration, distant niches migration and finally neoangiogenesis. The application of the microfluidic systems to study the intercellular crosstalk between lung cancer cells and surrounding tumor microenvironment and the connection with multiple molecular signals coming from the external cellular matrix are discussed. We also focus on recent breakthrough technologies regarding lab-on-chip devices that serve as tools for detecting circulating lung cancer cells. The superiority of microfluidic systems over traditional in vitro cell-based assays with regard to modern nanosafety studies and new cancer drug design and discovery is also addressed. Finally, the current progress and future challenges regarding printable and paper-based microfluidic devices for personalized nanomedicine are summarized.
Collapse
Affiliation(s)
- Monika Ruzycka
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland
| | - Mihaela R Cimpan
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ivan Rios-Mondragon
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ireneusz P Grudzinski
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland.
| |
Collapse
|
36
|
Bankó P, Lee SY, Nagygyörgy V, Zrínyi M, Chae CH, Cho DH, Telekes A. Technologies for circulating tumor cell separation from whole blood. J Hematol Oncol 2019; 12:48. [PMID: 31088479 PMCID: PMC6518774 DOI: 10.1186/s13045-019-0735-4] [Citation(s) in RCA: 221] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022] Open
Abstract
The importance of early cancer diagnosis and improved cancer therapy has been clear for years and has initiated worldwide research towards new possibilities in the care strategy of patients with cancer using technological innovations. One of the key research fields involves the separation and detection of circulating tumor cells (CTC) because of their suggested important role in early cancer diagnosis and prognosis, namely, providing easy access by a liquid biopsy from blood to identify metastatic cells before clinically detectable metastasis occurs and to study the molecular and genetic profile of these metastatic cells. Provided the opportunity to further progress the development of technology for treating cancer, several CTC technologies have been proposed in recent years by various research groups and companies. Despite their potential role in cancer healthcare, CTC methods are currently mainly used for research purposes, and only a few methods have been accepted for clinical application because of the difficulties caused by CTC heterogeneity, CTC separation from the blood, and a lack of thorough clinical validation. Therefore, the standardization and clinical application of various developed CTC technologies remain important subsequent necessary steps. Because of their suggested future clinical benefits, we focus on describing technologies using whole blood samples without any pretreatment and discuss their advantages, use, and significance. Technologies using whole blood samples utilize size-based, immunoaffinity-based, and density-based methods or combinations of these methods as well as positive and negative enrichment during separation. Although current CTC technologies have not been truly implemented yet, they possess high potential as future clinical diagnostic techniques for the individualized therapy of patients with cancer. Thus, a detailed discussion of the clinical suitability of these new advanced technologies could help prepare clinicians for the future and can be a foundation for technologies that would be used to eliminate CTCs in vivo.
Collapse
Affiliation(s)
- Petra Bankó
- Department of Biochemical Engineering, Budapest University of Technology and Economics, Budapest, Hungary
| | - Sun Young Lee
- Department of Radiation Oncology, Chonbuk National University Hospital, Jeonju, Republic of Korea
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical, Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | | | - Miklós Zrínyi
- Laboratory of Nanochemistry, Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Chang Hoon Chae
- Laboratory of Nanochemistry, Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Dong Hyu Cho
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical, Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
- Department of Obstetrics and Gynecology, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - András Telekes
- Department of Oncology, St. Lazarus Hospital, Salgótarján, Hungary
| |
Collapse
|
37
|
Abstract
Circulating tumor cells (CTCs) play a central role in tumor dissemination and metastases, which are ultimately responsible for most cancer deaths. Technologies that allow for identification and enumeration of rare CTC from cancer patients' blood have already established CTC as an important clinical biomarker for cancer diagnosis and prognosis. Indeed, current efforts to robustly characterize CTC as well as the associated cells of the tumor microenvironment such as circulating cancer associated fibroblasts (cCAF), are poised to unmask key insights into the metastatic process. Ultimately, the clinical utility of CTC will be fully realized once CTC can be reliably cultured and proliferated as a biospecimen for precision management of cancer patients, and for discovery of novel therapeutics. In this review, we highlight the latest CTC capture and analyses technologies, and discuss in vitro strategies for culturing and propagating CTC.
Collapse
Affiliation(s)
- Ashutosh Agarwal
- Assistant Professor, Department of Biomedical Engineering, Department of Pathology & Laboratory Medicine, University of Miami
| | - Marija Balic
- Associate Professor, Division of Oncology, Department of Internal Medicine, Research Unit Circulating Tumor Cells and Cancer Stem Cells, Medical University of Graz, Austria
| | - Dorraya El-Ashry
- Associate Professor, Department of Laboratory Medicine and Pathology, University of Minnesota
| | - Richard J. Cote
- Professor and Joseph R. Coulter Jr. Chair, Department of Pathology & Laboratory Medicine, Director, John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami Miller School of Medicine
| |
Collapse
|
38
|
Cho H, Kim J, Song H, Sohn KY, Jeon M, Han KH. Microfluidic technologies for circulating tumor cell isolation. Analyst 2019; 143:2936-2970. [PMID: 29796523 DOI: 10.1039/c7an01979c] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metastasis is the main cause of tumor-related death, and the dispersal of tumor cells through the circulatory system is a critical step in the metastatic process. Early detection and analysis of circulating tumor cells (CTCs) is therefore important for early diagnosis, prognosis, and effective treatment of cancer, enabling favorable clinical outcomes in cancer patients. Accurate and reliable methods for isolating and detecting CTCs are necessary to obtain this clinical information. Over the past two decades, microfluidic technologies have demonstrated great potential for isolating and detecting CTCs from blood. The present paper reviews current advanced microfluidic technologies for isolating CTCs based on various biological and physical principles, and discusses their fundamental advantages and drawbacks for subsequent cellular and molecular assays. Owing to significant genetic heterogeneity among CTCs, microfluidic technologies for isolating individual CTCs have recently been developed. We discuss these single-cell isolation methods, as well as approaches to overcoming the limitations of current microfluidic CTC isolation technologies. Finally, we provide an overview of future innovative microfluidic platforms.
Collapse
Affiliation(s)
- Hyungseok Cho
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae 621-749, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
39
|
Rostami P, Kashaninejad N, Moshksayan K, Saidi MS, Firoozabadi B, Nguyen NT. Novel approaches in cancer management with circulating tumor cell clusters. JOURNAL OF SCIENCE: ADVANCED MATERIALS AND DEVICES 2019; 4:1-18. [DOI: 10.1016/j.jsamd.2019.01.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
40
|
Choi MK, Kim GH, I H, Park SJ, Lee MW, Lee BE, Park DY, Cho YK. Circulating tumor cells detected using fluid-assisted separation technique in esophageal squamous cell carcinoma. J Gastroenterol Hepatol 2019; 34:552-560. [PMID: 30426559 DOI: 10.1111/jgh.14543] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIM Esophageal squamous cell carcinoma (ESCC) is one of the aggressive gastrointestinal tract cancers. Detection of circulating tumor cells (CTCs) in peripheral blood from patients with various malignancies has been reported to have diagnostic, prognostic, and therapeutic implications. We aimed to evaluate CTCs in patients with ESCC and assess the clinical significance of CTCs in the early diagnosis of ESCC. METHODS Peripheral blood samples for CTCs analyses were prospectively obtained from 73 patients with ESCC prior to treatment between March 2015 and June 2018. CTCs were detected using a centrifugal microfluidic system with a new fluid-assisted separation technique. Blood samples from 31 healthy volunteers were used as controls. RESULTS After creating a receiver operating characteristic curve to determine the optimal CTC threshold to differentiate patients with ESCC from healthy controls, sensitivity and specificity were most optimized at a CTC threshold of two per 7.5 mL of blood. Among 66 subjects with ≥ 2 CTCs per 7.5 mL of blood, 63 (95.5%) had ESCC. Among 38 subjects with < 2 CTCs per 7.5 mL of blood, 28 (73.7%) were healthy controls. When using this threshold, the sensitivity and specificity for differentiating patients with ESCC from healthy controls were 86.3% and 90.3%, respectively. CTC count was associated with tumor-node-metastasis stage, especially lymph node metastasis, but there was no correlation with any other relevant clinicopathologic variable. CONCLUSIONS Our results suggest that CTCs detected using fluid-assisted separation technique could be helpful for early diagnosis of ESCC. Further large-scale prospective studies are warranted to validate our findings.
Collapse
Affiliation(s)
- Mun Ki Choi
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Busan, Korea
| | - Gwang Ha Kim
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Busan, Korea
| | - Hoseok I
- Department of Chest Surgery, Pusan National University School of Medicine, Busan, Korea
| | - Su Jin Park
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Busan, Korea
| | - Moon Won Lee
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Busan, Korea
| | - Bong Eun Lee
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Busan, Korea
| | - Do Youn Park
- Department of Pathology, Pusan National University School of Medicine, Busan, Korea
| | - Yoon-Kyoung Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS) and Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Korea
| |
Collapse
|
41
|
S Iliescu F, Sim WJ, Heidari H, P Poenar D, Miao J, Taylor HK, Iliescu C. Highlighting the uniqueness in dielectrophoretic enrichment of circulating tumor cells. Electrophoresis 2019; 40:1457-1477. [PMID: 30676660 DOI: 10.1002/elps.201800446] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/19/2019] [Accepted: 01/20/2019] [Indexed: 12/14/2022]
Abstract
Circulating tumor cells (CTCs) play an essential role in the metastasis of tumors, and thus can serve as a valuable prognostic factor for malignant diseases. As a result, the ability to isolate and characterize CTCs is essential. This review underlines the potential of dielectrophoresis for CTCs enrichment. It begins by summarizing the key performance parameters and challenges of CTCs isolation using microfluidics. The two main categories of CTCs enrichment-affinity-based and label-free methods-are analysed, emphasising the advantages and disadvantages of each as well as their clinical potential. While the main argument in favour of affinity-based methods is the strong specificity of CTCs isolation, the major advantage of the label-free technologies is in preserving the integrity of the cellular membrane, an essential requirement for downstream characterization. Moving forward, we try to answer the main question: "What makes dielectrophoresis a method of choice in CTCs isolation?" The uniqueness of dielectrophoretic CTCs enrichment resides in coupling the specificity of the isolation process with the conservation of the membrane surface. The specificity of the dielectrophoretic method stems from the differences in the dielectric properties between CTCs and other cells in the blood: the capacitances of the malignantly transformed cellular membranes of CTCs differ from those of other cells. Examples of dielectrophoretic devices are described and their performance evaluated. Critical requirements for using dielectrophoresis to isolate CTCs are highlighted. Finally, we consider that DEP has the potential of becoming a cytometric method for large-scale sorting and characterization of cells.
Collapse
Affiliation(s)
| | - Wen Jing Sim
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore
| | - Hossein Heidari
- Department of Mechanical Engineering, University of California, Berkeley, CA, USA
| | - Daniel P Poenar
- VALENS-Centre for Bio Devices and Signal Analysis, Nanyang Technological University, Singapore
| | - Jianmin Miao
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore
| | - Hayden K Taylor
- Department of Mechanical Engineering, University of California, Berkeley, CA, USA
| | - Ciprian Iliescu
- Biomedical Institute for Global Health Research & Technology (BIGHEART), National University of Singapore, Singapore
| |
Collapse
|
42
|
Song Y, Shi Y, Huang M, Wang W, Wang Y, Cheng J, Lei Z, Zhu Z, Yang C. Bioinspired Engineering of a Multivalent Aptamer-Functionalized Nanointerface to Enhance the Capture and Release of Circulating Tumor Cells. Angew Chem Int Ed Engl 2019; 58:2236-2240. [PMID: 30548959 DOI: 10.1002/anie.201809337] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/07/2018] [Indexed: 12/24/2022]
Abstract
Circulating tumor cell (CTC)-enrichment by using aptamers has a number of advantages, but the issue of compromised binding affinities and stabilities in real samples hinders its wide applications. Inspired by the high efficiency of the prey mechanism of the octopus, we engineered a deterministic lateral displacement (DLD)-patterned microfluidic chip modified with multivalent aptamer-functionalized nanospheres (AP-Octopus-Chip) to enhance capture efficiency. The multivalent aptamer-antigen binding efficiency improves 100-fold and the capture efficiency is enhanced more than 300 % compared with a monovalent aptamer-modified chip. Moreover, the captured cancer cells can be released through a thiol exchange reaction with up to 80 % efficiency and 96 % viability, which is fully compatible with downstream mutation detection and CTC culture. Using the chip, we were able to find CTCs in all cancer samples analyzed.
Collapse
Affiliation(s)
- Yanling Song
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yuanzhi Shi
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Mengjiao Huang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Wei Wang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yang Wang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Jie Cheng
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Zhichao Lei
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Zhi Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Chaoyong Yang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.,The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
43
|
Xia Y, Wan Y, Hao S, Nisic M, Harouaka RA, Chen Y, Zou X, Zheng SY. Nucleus of Circulating Tumor Cell Determines Its Translocation Through Biomimetic Microconstrictions and Its Physical Enrichment by Microfiltration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802899. [PMID: 30286282 DOI: 10.1002/smll.201802899] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/24/2018] [Indexed: 06/08/2023]
Abstract
The mechanism of cells passing through microconstrictions, such as capillaries and endothelial junctions, influences metastasis of circulating tumor cells (CTCs) in vivo, as well as size-based enrichment of CTCs in vitro. However, very few studies observe such translocation of microconstrictions in real time, and thus the inherent biophysical mechanism is poorly understood. In this study, a multiplexed microfluidic device is fabricated for real-time tracking of cell translocation under physiological pressure and recording deformation of the whole cell and nucleus, respectively. It is found that the deformability and size of the nucleus instead of the whole cell dominate cellular translocation through microconstrictions under a normal physiological pressure range. More specifically, cells with a large and stiff nucleus are prone to be blocked by relatively small constrictions. The same phenomenon is also observed in the size-based enrichment of CTCs from peripheral blood of metastatic cancer patients. These findings are different from a popular viewpoint that the size and deformability of a whole cell mainly determine cell translation through microconstrictions, and thus may elucidate interactions between CTCs and capillaries from a new perspective and guide the rational design of size-based microfilters for rare cell enrichment.
Collapse
Affiliation(s)
- Yiqiu Xia
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yuan Wan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Sijie Hao
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Merisa Nisic
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Ramdane A Harouaka
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yizhu Chen
- Department of Electrical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Xin Zou
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Si-Yang Zheng
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Electrical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Penn State Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, USA
- Penn State Cancer Institute, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
44
|
Liu F, Wang S, Lu Z, Sun Y, Yang C, Zhou Q, Hong S, Wang S, Xiong B, Liu K, Zhang N. A simple pyramid-shaped microchamber towards highly efficient isolation of circulating tumor cells from breast cancer patients. Biomed Microdevices 2018; 20:83. [PMID: 30221311 DOI: 10.1007/s10544-018-0326-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Isolation and detection of circulating tumor cells (CTCs) has showed a great clinical impact for tumor diagnosis and treatment monitoring. Despite significant progresses of the existing technologies, feasible and cost-effective CTC isolation techniques are more desirable. In this study, a novel method was developed for highly efficient isolation of CTCs from breast cancer patients based on biophysical properties using a pyramid-shaped microchamber. Through optimization tests, the outlet height of 6 μm and the flow rate of 200 μL/min were chosen as the optimal conditions. The capture efficiencies of more than 85% were achieved for cancer cell lines (SKBR3, BGC823, PC3, and H1975) spiked in DMEM and healthy blood samples without clogging issue. In clinic assay, the platform identified CTCs in 13 of 20 breast cancer patients (65%) with an average of 4.25 ± 4.96 CTCs/2 mL, whereas only one cell was recognized as CTC in 1 of 15 healthy blood samples. The statistical analyses results demonstrated that both CTC positive rate and CTC counts were positive correlated with TNM stage (p < 0.001; p = 0.02, respectively). This microfluidic platform successfully demonstrated the clinical feasibility of CTC isolation and would hold great potential of clinical application in predicting and monitoring the prognosis of cancer patients.
Collapse
Affiliation(s)
- Feng Liu
- College of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan, Hubei, 430200, People's Republic of China
| | - Shuibing Wang
- College of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan, Hubei, 430200, People's Republic of China
| | - Zhigang Lu
- College of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan, Hubei, 430200, People's Republic of China
| | - Yumei Sun
- College of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan, Hubei, 430200, People's Republic of China
| | - Chaogang Yang
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
| | - Qiongwei Zhou
- College of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan, Hubei, 430200, People's Republic of China
| | - Shaoli Hong
- College of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan, Hubei, 430200, People's Republic of China
| | - Shengxiang Wang
- College of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan, Hubei, 430200, People's Republic of China
| | - Bin Xiong
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
| | - Kan Liu
- College of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan, Hubei, 430200, People's Republic of China. .,School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, People's Republic of China.
| | - Nangang Zhang
- College of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan, Hubei, 430200, People's Republic of China.
| |
Collapse
|
45
|
Qiu X, Lombardo JA, Westerhof TM, Pennell M, Ng A, Alshetaiwi H, Luna BM, Nelson EL, Kessenbrock K, Hui EE, Haun JB. Microfluidic filter device with nylon mesh membranes efficiently dissociates cell aggregates and digested tissue into single cells. LAB ON A CHIP 2018; 18:2776-2786. [PMID: 30090895 PMCID: PMC6171522 DOI: 10.1039/c8lc00507a] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Tissues are increasingly being analyzed at the single cell level in order to characterize cellular diversity and identify rare cell types. Single cell analysis efforts are greatly limited, however, by the need to first break down tissues into single cell suspensions. Current dissociation methods are inefficient, leaving a significant portion of the tissue as aggregates that are filtered away or left to confound results. Here, we present a simple and inexpensive microfluidic device that simultaneously filters large tissue fragments and dissociates smaller aggregates into single cells, thereby improving single cell yield and purity. The device incorporates two nylon mesh membranes with well-defined, micron-sized pores that operate on aggregates of different size scales. We also designed the device so that the first filtration could be performed under tangential flow to minimize clogging. Using cancer cell lines, we demonstrated that aggregates were effectively dissociated using high flow rates and pore sizes that were smaller than a single cell. However, pore sizes that were less than half the cell size caused significant damage. We then improved results by passing the sample through two filter devices in series, with single cell yield and purity predominantly determined by the pore size of the second membrane. Next, we optimized performance using minced and digested murine kidney tissue samples, and determined that the combination of 50 and 15 μm membranes was optimal. Finally, we integrated these two membranes into a single filter device and performed validation experiments using minced and digested murine kidney, liver, and mammary tumor tissue samples. The dual membrane microfluidic filter device increased single cell numbers by at least 3-fold for each tissue type, and in some cases by more than 10-fold. These results were obtained in minutes without affecting cell viability, and additional filtering would not be required prior to downstream applications. In future work, we will create complete tissue analysis platforms by integrating the dual membrane microfluidic filter device with additional upstream tissue processing technologies, as well as downstream operations such as cell sorting and detection.
Collapse
Affiliation(s)
- Xiaolong Qiu
- Department of Biomedical Engineering, University of California Irvine, 3107 Natural Sciences II, Irvine, CA 92697, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Khetani S, Mohammadi M, Nezhad AS. Filter-based isolation, enrichment, and characterization of circulating tumor cells. Biotechnol Bioeng 2018; 115:2504-2529. [DOI: 10.1002/bit.26787] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 01/12/2023]
Affiliation(s)
- Sultan Khetani
- Department of Mechanical and Manufacturing Engineering, BioMEMS and Bioinspired Microfluidic Laboratory; University of Calgary; Calgary Canada
- Center for BioEngineering Research and Education, University of Calgary; Calgary Canada
| | - Mehdi Mohammadi
- Department of Mechanical and Manufacturing Engineering, BioMEMS and Bioinspired Microfluidic Laboratory; University of Calgary; Calgary Canada
- Center for BioEngineering Research and Education, University of Calgary; Calgary Canada
- Department of Biological Sciences; University of Calgary; Calgary Canada
| | - Amir Sanati Nezhad
- Department of Mechanical and Manufacturing Engineering, BioMEMS and Bioinspired Microfluidic Laboratory; University of Calgary; Calgary Canada
- Center for BioEngineering Research and Education, University of Calgary; Calgary Canada
| |
Collapse
|
47
|
Separable Bilayer Microfiltration Device for Label-Free Enrichment of Viable Circulating Tumor Cells. Methods Mol Biol 2018; 1634:81-91. [PMID: 28819842 DOI: 10.1007/978-1-4939-7144-2_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Analysis of rare circulating tumor cells enriched from metastatic cancer patients yields critical information on disease progression, therapy response, and the mechanism of cancer metastasis. Here we describe in detail a label-free enrichment process of circulating tumor cells based on its unique physical properties (size and deformability). Viable circulating tumor cells can be successfully enriched and analyzed, or easily released for further characterization due to the novel separable two-layer design.
Collapse
|
48
|
Jiang Y, Yu Z, Huang X, Chen R, Chen W, Zeng Y, Xu C, Min H, Zheng N, Cheng X. A multilayer lateral-flow microfluidic device for particle separation. MICROFLUIDICS AND NANOFLUIDICS 2018; 22:40. [DOI: 10.1007/s10404-018-2053-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 02/28/2018] [Indexed: 01/03/2025]
|
49
|
Jiang W, Wang H, Cui Y, Lei Y, Wang Y, Xu D, Jiang N, Chen Y, Sun Y, Zhang Y, Cao J, Ke Z. Polymer nanofiber-based microchips for EGFR mutation analysis of circulating tumor cells in lung adenocarcinoma. Int J Nanomedicine 2018; 13:1633-1642. [PMID: 29588590 PMCID: PMC5862013 DOI: 10.2147/ijn.s157154] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Circulating tumor cells (CTCs) detection, an approach considered to be “liquid biopsy”, is a potential alternative method in clinical use for early diagnosis of solid tumor progression. Methods In this study, we developed a poly (lactic-co-glycolic acid) (PLGA) – nanofiber (PN)-NanoVelcro chip as an efficient device for simple and rapid capture of CTCs from peripheral blood. We evaluated the device performance by assessing the capture efficiency and purity. Single CTC was isolated via laser microdissection system for subsequent genetic analysis, with an aim to find the concordance of epidermal growth factor receptor (EGFR) mutations between tumor tissue and CTCs. Results PN-NanoVelcro chip exhibits great performance in capture efficiency and high purity. The genetic analysis results showed that most EGFR mutation in tumor tissue could also be detected in CTCs. Conclusion Compared to computed tomography image results, CTC detection can be implemented throughout the course of diseases and provides an accurate and earlier diagnosis of tumor progression, which make it possible for patients to acquire suitable and timely treatment.
Collapse
Affiliation(s)
- Wenting Jiang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Han Wang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yongmei Cui
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yiyan Lei
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuefeng Wang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Di Xu
- Department of Thoracic Surgery, The Central Hospital of Wuhan, Jiang'an District, Wuhan, Hubei, China
| | - Neng Jiang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yangshan Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu Sun
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yang Zhang
- Biomedical Engineering, The University of Texas at El Paso, El Paso, TX, USA
| | - Jessica Cao
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
50
|
Luo YT, Cheng J, Feng X, He SJ, Wang YW, Huang Q. The viable circulating tumor cells with cancer stem cells feature, where is the way out? J Exp Clin Cancer Res 2018; 37:38. [PMID: 29482576 PMCID: PMC5828305 DOI: 10.1186/s13046-018-0685-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/19/2018] [Indexed: 02/07/2023] Open
Abstract
With cancer stem cells (CSCs) became the research hotspot, emerging studies attempt to reveal the functions of these special subsets in tumorigenesis. Although various approaches have been used in CSCs researches, only a few could really reflect or simulate the microenvironment in vivo. At present, CSCs theories are still difficult to apply for clinical remedy because CSCs subpopulations are always hard to identify and trace. Thus an ideal approach for clinicians and researchers is urgently needed. Circulating tumor cells (CTCs), as the method of noninvasive-liquid biopsy, could be detected in the peripheral blood (PB) from many tumors and even could be treated as procurators for CSCs deeper researches from patient-derived sample. However, CTCs, as a diagnostic marker, also raise much controversy over theirs clinical value. Mechanisms causing CTCs to shed from the tumor have not been fully characterized, thus it is unclear whether CTCs represent the entire makeup of cancer cells in the tumor or only a subset. The heterogeneity of CTCs also caused different clinical outcomes. To overcome these unsolved problems, recently, CTC researches are not just depend on enumerations, whereas those CTC subsets that could expand in vitro may play a pivotal role in the metastatic cascade. Here, we retrospect the CTC developmental history and discourse upon the enrichment of viable CTCs in functional assays, probe the further avenue at the crossroad.
Collapse
Affiliation(s)
- Y T Luo
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - J Cheng
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - X Feng
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - S J He
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Y W Wang
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Q Huang
- Molecular Diagnostic Laboratory of Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China.
| |
Collapse
|