1
|
Hood MC, Gardner K, Li W, Tan J. Interplay of size, deformability, and device layout on cell transport in microfluidics. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2024; 36:425106. [PMID: 38975704 DOI: 10.1088/1361-648x/ad5ff3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
Microfluidics have been widely used for cell sorting and capture. In this work, numerical simulations of cell transport in microfluidic devices were studied considering cell sizes, deformability, and five different device designs. Among these five designs, deterministic lateral displacement device (DLD) and hyperuniform device (HU) performed better in promoting cell-micropost collision due to the continuously shifted micropost positions as compared with regular grid, staggered, and hexagonal layout designs. However, the grid and the hexagonal layouts showed best in differentiating cells by their size dependent velocity due to the size exclusion effect for cell transport in clear and straight paths in the flow direction. A systematic study of the velocity differentiation under different dimensionless groups was performed showing that the velocity difference is dominated by the micropost separation distance perpendicular to the direction of flow. Microfluidic experiments also confirmed the velocity differentiation results. The study can provide guiding principles for microfluidic design.
Collapse
Affiliation(s)
- Michael C Hood
- Department of Mechanical Engineering, Northern Illinois University, DeKalb, IL 60115, United States of America
| | - Karl Gardner
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Wei Li
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Jifu Tan
- Department of Mechanical Engineering, Northern Illinois University, DeKalb, IL 60115, United States of America
| |
Collapse
|
2
|
Wei YJ, Wei X, Zhang X, Wu CX, Cai JY, Chen ML, Wang JH. A hydrodynamic-based dual-function microfluidic chip for high throughput discriminating tumor cells. Talanta 2024; 273:125884. [PMID: 38508128 DOI: 10.1016/j.talanta.2024.125884] [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: 10/25/2023] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
A hydrodynamic-based microfluidic chip consisted of two function units that could not only separate tumor cells (TCs) from whole blood but also remove residual blood cells was designed. The separation of TCs was achieved by a straight contraction-expansion array (CEA) microchannel on the front end of the chip. The addition of contractive structure brought a micro-vortex like Dean vortex that promoted cell focusing in the channel, while when cells entered the dilated region, the wall-induced lift force generated by the channel wall gave cells a push away from the wall. As the wall-induced lift force is proportional to the third power of the cell diameter, TCs with larger diameter will have a larger lateral migration under the wall-induced lift force, realizing the separation of TCs from blood sample. Fluorescent particles with diameters of 19.3 μm and 4.5 μm were used to simulate TCs and red blood cells, respectively, to verify the separation capacity of the proposed CEA microchannel for particles with different diameter. And a separation efficiency 98.7% for 19.3 μm particles and a removal rate 96.2% for 4.5 μm particles was observed at sample flow rate of 10 μL min-1 and sheath flow rate of 190 μL min-1. In addition, a separation efficiency about 96.1% for MCF-7 cells (stained with DiI) and removal rates of 96.2% for red blood cells (RBCs) and 98.7% for white blood cells (WBCs) were also obtained under the same condition. However, on account of the large number of blood cells in the blood, there will be a large number of blood cells remained in the isolated TCs, so a purification unit based on hydrodynamic filtration (HDF) was added after the separation microchannel. The purification channel is a size-dictated cell filter that can remove residual blood cells but retain TCs, thus achieving the purification of TCs. Combined the CEA microchannel and the purifier, the microchip facilitates sorting of MCF-7 cells from whole blood with a separation rate about 95.3% and a removal rate over 99.99% for blood cells at a sample flow rate of 10 μL min-1, sheath flow rate of 190 μL min-1 and washing flow rate of 63 μL min-1.
Collapse
Affiliation(s)
- Yu-Jia Wei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Xing Wei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Xuan Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Cheng-Xing Wu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Ji-Ying Cai
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Ming-Li Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China.
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China.
| |
Collapse
|
3
|
Uddin MR, Chen X. Enhancing cell separation in a hybrid spiral dielectrophoretic microchannel: Numerical insights and optimal operating conditions. Biotechnol Prog 2024; 40:e3437. [PMID: 38289677 DOI: 10.1002/btpr.3437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/27/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024]
Abstract
Reliable separation of circulating tumor cells from blood cells is crucial for early cancer diagnosis and prognosis. Many conventional microfluidic platforms take advantage of the size difference between particles for their separation, which renders them impractical for sorting overlapping-sized cells. To address this concern, a hybrid inertial-dielectrophoretic microfluidic chip is proposed in this work for continuous and single-stage separation of lung cancer cell line A549 cells from white blood cells of overlapping size. The working mechanism of the proposed spiral microchannel embedded with planar interdigitated electrodes is validated against the experimental results. A numerical investigation is carried out over a range of flow conditions and electric field intensity to determine the separation efficiency and migration characteristics of the cell mixture. The results demonstrate the unique capability of the proposed microchannel to achieve high-throughput separation of cells at low applied voltages in both vertical and lateral directions. A significant lateral separation distance between the CTCs and the WBCs has been achieved, which allows for high-resolution and effective separation of cells. The separation resolution can be controlled by adjusting the strength of the applied electric field. Furthermore, the results demonstrate that the lateral separation distance is maximum at a voltage termed the critical voltage, which increases with the increase in the flow rate. The proposed microchannel and the developed technique can provide valuable insight into the development of a tunable and robust medical device for effective and high-throughput separation of cancer cells from the WBCs.
Collapse
Affiliation(s)
- Mohammed Raihan Uddin
- Department of Mechanical and Aerospace Engineering, Ohio State University, Columbus, Ohio, USA
| | - Xiaolin Chen
- School of Engineering and Computer Science, Washington State University, Vancouver, Washington, USA
| |
Collapse
|
4
|
Yan S, Liu Y, Nguyen NT, Zhang J. Magnetophoresis-Enhanced Elasto-Inertial Migration of Microparticles and Cells in Microfluidics. Anal Chem 2024; 96:3925-3932. [PMID: 38346322 DOI: 10.1021/acs.analchem.3c05803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Microfluidic particle and cell manipulation techniques possess many potentials for biomedicine and healthcare. Many techniques have been developed based on active (e.g., electrical, magnetic, acoustic, and thermal) force fields and passive hydrodynamic forces (e.g., inertial and elastic lift forces). However, techniques based on a single active or passive manipulating physics cannot always meet the demands, and combining multiple physics becomes a promising strategy to promote technique flexibility and versatility. In this work, we explored the physical coupling of magnetophoresis with the elastic and inertial (i.e., elasto-inertial) lift forces for the manipulation of microparticles. Particle lateral migration was studied in a coflowing configuration of viscoelastic ferrofluid/water (sample/sheath). The particles were suspended in the viscoelastic ferrofluid and confined near the channel sidewall by a sheath flow. The coordination of magnetophoresis and elasto-inertial lift forces promoted the cross-stream migration of particles. Besides, we investigated the effect of the flow rate ratio and total flow rate on the migration of particles. Furthermore, we also investigated the effects of fluid elasticity in sample and sheath flows on particle migration using different combinations of sample and sheath flows, including Newtonian ferrofluid/water, Newtonian ferrofluid/viscoelastic fluid, and viscoelastic ferrofluid/viscoelastic coflows. Experimental results demonstrated and ascertained the promoted particle lateral migration in the PEO-based ferrofluid/water coflow. Finally, we demonstrate the proof-of-concept application of the physical coupling strategy for cell cross-stream migration and solution exchange. We envisage that this novel multiphysical coupling scheme has great potential for the flexible and versatile manipulation of microparticles and cells.
Collapse
Affiliation(s)
- Sheng Yan
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yong Liu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen 518060, China
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
| | - Jun Zhang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
- School of Engineering and Built Environment, Griffith University, Nathan, Queensland 4111, Australia
| |
Collapse
|
5
|
Uddin MR, Sarowar MT, Chen X. Separation of CTCs from WBCs using DEP-assisted inertial manipulation: A numerical study. Electrophoresis 2023; 44:1781-1794. [PMID: 37753944 DOI: 10.1002/elps.202300090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/09/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023]
Abstract
Isolation and detection of circulating tumor cells (CTCs) hold significant importance for the early diagnosis of cancer and the assessment of therapeutic strategies. However, the scarcity of CTCs among peripheral blood cells presents a major challenge to their detection. Additionally, a similar size range between CTCs and white blood cells (WBCs) makes conventional microfluidic platforms inadequate for the isolation of CTCs. To overcome these challenges, in this study, a novel inertial-dielectrophoretic microfluidic channel for size-independent, single-stage separation of CTCs from WBCs has been presented. The proposed device utilizes a spiral microchannel embedded with interdigitated electrodes. A numerical model is developed and validated to investigate the influence of various parameters related to the channel design, fluid flow, and electrode configuration. It was found that optimal separation of CTCs could be obtained at a relatively low voltage, termed the critical voltage. Furthermore, at the critical voltage of 7.5 V, the hybrid microchannel is demonstrated to be capable of separating CTCs from different WBC subtypes including granulocytes, monocytes, T-, and B-lymphocytes. The unique capabilities of the hybrid spiral microchannel allow for this size-independent isolation of CTCs from a mixture of WBCs. Overall, the proposed technique can be readily utilized for continuous and high-throughput separation of cancer cells.
Collapse
Affiliation(s)
- Mohammed Raihan Uddin
- School of Engineering and Computer Science, Washington State University, Vancouver, Washington, USA
| | - Md Tanbir Sarowar
- School of Engineering and Computer Science, Washington State University, Vancouver, Washington, USA
| | - Xiaolin Chen
- School of Engineering and Computer Science, Washington State University, Vancouver, Washington, USA
| |
Collapse
|
6
|
Gupta A, Burada PS. Separation of interacting active particles in an asymmetric channel. Phys Rev E 2023; 108:034605. [PMID: 37849171 DOI: 10.1103/physreve.108.034605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/25/2023] [Indexed: 10/19/2023]
Abstract
We study the diffusive behavior of interacting active particles (self-propelled) with mass m in an asymmetric channel. The particles are subjected to an external oscillatory force along the length of the channel. In this setup, particles may exhibit rectification. In the absence of interaction, the mean velocity 〈v〉 of the particles shows a maximum at moderate m values. It means that particles of moderate m have higher velocities than the others. However, by incorporating short-range interaction between the particles, 〈v〉 exhibits an additional peak at lower m values, indicating that particles of lower and moderate m can be separated simultaneously from the rest. Furthermore, by tuning the interaction strength, the self-propelled velocity, and the parameters of the oscillatory force, one can selectively separate the particles of lower m, moderate m, or both. Empirical relations for estimating the optimal mass as a function of these parameters are proposed. These findings are beneficial for separating the particles of selective m from the rest of the particles.
Collapse
Affiliation(s)
- Ankit Gupta
- Department of Physics, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - P S Burada
- Department of Physics, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
7
|
Cha H, Dai Y, Hansen HHWB, Ouyang L, Chen X, Kang X, An H, Ta HT, Nguyen NT, Zhang J. Asymmetrical Obstacles Enable Unilateral Inertial Focusing and Separation in Sinusoidal Microchannel. CYBORG AND BIONIC SYSTEMS 2023; 4:0036. [PMID: 37342212 PMCID: PMC10278993 DOI: 10.34133/cbsystems.0036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/25/2023] [Indexed: 06/22/2023] Open
Abstract
Inertial microfluidics uses the intrinsic fluid inertia in confined channels to manipulate the particles and cells in a simple, high-throughput, and precise manner. Inertial focusing in a straight channel results in several equilibrium positions within the cross sections. Introducing channel curvature and adjusting the cross-sectional aspect ratio and shape can modify inertial focusing positions and can reduce the number of equilibrium positions. In this work, we introduce an innovative way to adjust the inertial focusing and reduce equilibrium positions by embedding asymmetrical obstacle microstructures. We demonstrated that asymmetrical concave obstacles could break the symmetry of original inertial focusing positions, resulting in unilateral focusing. In addition, we characterized the influence of obstacle size and 3 asymmetrical obstacle patterns on unilateral inertial focusing. Finally, we applied differential unilateral focusing on the separation of 10- and 15-μm particles and isolation of brain cancer cells (U87MG) from white blood cells (WBCs), respectively. The results indicated an excellent cancer cell recovery of 96.4% and WBC rejection ratio of 98.81%. After single processing, the purity of the cancer cells was dramatically enhanced from 1.01% to 90.13%, with an 89.24-fold enrichment. We believe that embedding asymmetric concave micro-obstacles is a new strategy to achieve unilateral inertial focusing and separation in curved channels.
Collapse
Affiliation(s)
- Haotian Cha
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
| | - Yuchen Dai
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
| | - Helena H. W. B. Hansen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
| | - Lingxi Ouyang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
| | - Xiangxun Chen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
| | - Xiaoyue Kang
- School of Engineering, University of Tasmania, Churchill Avenue, Tasmania 7005, Australia
| | - Hongjie An
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
| | - Hang Thu Ta
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
- Bioscience Discipline, School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
| | - Jun Zhang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia
| |
Collapse
|
8
|
Halawa T, Baeesa S, Fadul MM, Badahdah AA, Enani M, Fathaddin AA, Kawass D, Alkhotani A, Bahakeem B, Kurdi M. The Role of Liquid Biopsy in the Diagnosis and Prognosis of WHO Grade 4 Astrocytoma. Cureus 2023; 15:e41221. [PMID: 37525780 PMCID: PMC10387356 DOI: 10.7759/cureus.41221] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/02/2023] Open
Abstract
Liquid biopsy, as a non-invasive diagnostic tool, has recently gained significant attention in the field of oncology. It involves the analysis of various biomarkers present in bodily fluids, such as blood or cerebrospinal fluid, to provide information about the underlying cancer. In the case of WHO grade 4 astrocytomas, liquid biopsy has the potential to significantly impact the diagnosis and prognosis of this aggressive malignant brain tumor. By detecting specific genetic mutations, such as IDH1 or EGFR, and monitoring levels of circulating tumor DNA, liquid biopsy can aid in the early detection and monitoring of disease progression. This innovative approach is gradually being acknowledged as a less invasive and cost-effective procedure for cancer diagnosis and management to improve patient outcomes and quality of life. Various kinds of biomarkers circulating in cerebrospinal fluid (CSF), such as circulating tumor cells (CTC) and different types of nucleic acids like cell-free DNA (cfDNA), cell-free RNA (ctRNA), and microRNAs (miRNA), have been identified. These biomarkers, which require dependable detection methods, are comparatively simple to obtain and allow for repeated measurements, making them significantly superior for disease monitoring. This review aims to compare the latest liquid biopsy analysis tools for both CSF and plasma in the central nervous system.
Collapse
Affiliation(s)
- Taher Halawa
- Department of Pediatrics, Faculty of Medicine King Abdulaziz University, Rabigh, SAU
| | - Saleh Baeesa
- Department of Neuroscience, King Faisal Specialist Hospital and Research Centre, Jeddah, SAU
| | - Motaz M Fadul
- Department of Pathology, Faculty of Medicine King Abdulaziz University, Rabigh, SAU
| | - Adnan A Badahdah
- Department of Internal Medicine, University of Jeddah, Jeddah, SAU
| | - Maryam Enani
- Department of Surgery, King Abdulaziz University Hospital, Jeddah, SAU
| | - Amany A Fathaddin
- Department of Pathology, College of Medicine, King Saud University, Riyadh, SAU
- Department of Pathology, King Saud University Medical City, Riyadh, SAU
| | - Dania Kawass
- Department of Family Medicine, Faculty of Medicine King Abdulaziz University, Jeddah, SAU
| | - Alaa Alkhotani
- Department of Pathology, Umm Al-Qura University, Makkah, SAU
| | - Basem Bahakeem
- Department of Internal Medicine, Umm Al-Qura University, Makkah, SAU
| | - Maher Kurdi
- Department of Pathology, Faculty of Medicine King Abdulaziz University, Rabigh, SAU
| |
Collapse
|
9
|
Derakhshan R, Bozorgzadeh A, Ramiar A. Numerical investigation of ternary particle separation in a microchannel with a wall-mounted obstacle using dielectrophoresis. J Chromatogr A 2023; 1702:464079. [PMID: 37263054 DOI: 10.1016/j.chroma.2023.464079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023]
Abstract
In recent years, microfluidic-based particle/cell manipulation techniques have catalyzed significant advances in several fields of science. As an efficient, precise, and label-free particle/cell manipulation technique, dielectrophoresis (DEP) has recently attracted widespread attention. This paper presents the design and investigation of a straight sheathless 3D microchannel with a wall-mounted trapezoidal obstacle for continuous-flow separation of three different populations of polystyrene (PS) particles (5, 10 and 20 µm) using DEP. An OpenFOAM code is developed to simulate and investigate the movement of particles in the microchannel. Then, the code is validated by performing various experimental tests using a microdevice previously fabricated in our lab. By comparing the numerical simulation results with the experimental tests, it can be claimed that the newly developed solver is highly accurate, and its results agree well with experimental tests. Next, the effect of various operational and geometrical parameters such as obstacle height, applied voltage, electrode pairs angle, and flow rate on the efficient focusing and separation of particles are numerically investigated. The results showed that efficient particle separation could only be achieved for obstacle heights of more than 350 µm. Furthermore, the appropriate voltage range for efficient particle separation is increased by decreasing the electrode angle as well as increasing the flow rate. Moreover, the results showed that by employing the appropriate channel design and operational conditions, at a maximum applied voltage of 10V, a sample flow rate of 2.5μL/min could be processed. The proposed design can be beneficial for integrating with lab-on-a-chip and clinical diagnosis applications due to advantages, such as simple design, no need for sheath flow, the simultaneous ternary separation of particles, and providing precise particle separation.
Collapse
Affiliation(s)
- Reza Derakhshan
- Mechanical Engineering Department, Microfluidics and MEMS lab, Babol Noshirvani University of Technology, Babol, Mazandaran, Iran.
| | - Ali Bozorgzadeh
- Mechanical Engineering Department, Microfluidics and MEMS lab, Babol Noshirvani University of Technology, Babol, Mazandaran, Iran.
| | - Abas Ramiar
- Mechanical Engineering Department, Microfluidics and MEMS lab, Babol Noshirvani University of Technology, Babol, Mazandaran, Iran.
| |
Collapse
|
10
|
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: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
|
11
|
Lv B, Cai J. Simulation and analysis of geometric parameters based on Taguchi method in Y-Y microfluidic device for circulating tumor cell separation by alternating current dielectrophoresis. J Chromatogr A 2023; 1693:463894. [PMID: 36854211 DOI: 10.1016/j.chroma.2023.463894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023]
Abstract
Microfluidic technology has shown a remarkable ability to separate circulating tumor cells (CTC) in microfluidic devices. It can be used more effectively by further understanding the effect of geometric parameters on its separation performance. In this paper, the separation performance of a Y-Y microfluidic device was examined by varying its geometry parameters. In the device, the alternating current dielectrophoresis (AC DEP) method was used to separate CTC. 16 device models with various geometric parameters were created based on the Taguchi method. The geometric parameters included main channel length L, main channel width W, interelectrode angle α, and buffer inlet channel angle β. The electric field, flow field, and cell trajectory in the device were all numerically simulated to analyze the effect of geometric parameters. Signal-to-noise ratio (SNR) was used to determine the order of effect degree and optimal combination of geometric parameters. The results demonstrated that raising the flow velocity in the buffer inlet could enhance the separation purity. The separation purity was affected by the geometric parameters in the order of W> α> L> β. β had the weakest impact on the separation purity and accounted for 7.81%, while W had the most remarkable impact and accounted for 50.48%. It is found that the set of L = 1080 µm, W = 110 µm, α= 60°, and β= 60° is the optimal combination of geometric parameters. A fitting regression equation is found to describe well the effect of these parameters on separation purity. The results may provide a guide for designing microfluidic devices for separating CTC.
Collapse
Affiliation(s)
- Binghui Lv
- School of Nuclear Science and Engineering, North China Electric Power University, Beijing 102206, China
| | - Jun Cai
- School of Nuclear Science and Engineering, North China Electric Power University, Beijing 102206, China.
| |
Collapse
|
12
|
David P, Mittelstädt A, Kouhestani D, Anthuber A, Kahlert C, Sohn K, Weber GF. Current Applications of Liquid Biopsy in Gastrointestinal Cancer Disease-From Early Cancer Detection to Individualized Cancer Treatment. Cancers (Basel) 2023; 15:cancers15071924. [PMID: 37046585 PMCID: PMC10093361 DOI: 10.3390/cancers15071924] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Worldwide, gastrointestinal (GI) cancers account for a significant amount of cancer-related mortality. Tests that allow an early diagnosis could lead to an improvement in patient survival. Liquid biopsies (LBs) due to their non-invasive nature as well as low risk are the current focus of cancer research and could be a promising tool for early cancer detection. LB involves the sampling of any biological fluid (e.g., blood, urine, saliva) to enrich and analyze the tumor's biological material. LBs can detect tumor-associated components such as circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and circulating tumor cells (CTCs). These components can reflect the status of the disease and can facilitate clinical decisions. LBs offer a unique and new way to assess cancers at all stages of treatment, from cancer screenings to prognosis to management of multidisciplinary therapies. In this review, we will provide insights into the current status of the various types of LBs enabling early detection and monitoring of GI cancers and their use in in vitro diagnostics.
Collapse
Affiliation(s)
- Paul David
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anke Mittelstädt
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Dina Kouhestani
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Anthuber
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christoph Kahlert
- Department of Surgery, Carl Gustav Carus University Hospital, 01307 Dresden, Germany
| | - Kai Sohn
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
| | - Georg F Weber
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
13
|
Akbarnataj K, Maleki S, Rezaeian M, Haki M, Shamloo A. Novel size-based design of spiral microfluidic devices with elliptic configurations and trapezoidal cross-section for ultra-fast isolation of circulating tumor cells. Talanta 2023; 254:124125. [PMID: 36462283 DOI: 10.1016/j.talanta.2022.124125] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/12/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Investigation and analysis of circulating tumor cells (CTCs) have been valuable resources for detecting and diagnosing cancer in its early stages. Recently, enumeration and separation of CTCs via microfluidic devices have attracted significant attention due to their low cost and easy setup. In this study, novel microfluidic devices based on size-dependent cell-sorting with a trapezoidal cross-section and elliptic spiral configurations were proposed to reach label-free, ultra-fast CTCs enrichment. Firstly, the possibility and quality of separation in the devices were evaluated via a numerical simulation. Subsequently, these devices were fabricated to investigate the effects of the altering curvature and the trapezoidal cross-section on the isolation of CTCs from the peripheral blood sample at varying flow rates ranging from 0.5 mL/min to 3.5 mL/min. The experimental results indicated that the flow rate of 2.5 mL/min provided the optimal separation efficiency in the proposed devices, which was in fine agreement with the numerical analysis results. In this experiment, the purity values of CTCs between 88% and 90% were achieved, which is an indicator of the high capability of the proposed devices for the isolation and enrichment of CTCs. This strategy is hoped to overcome the limitations of classical affinity-based CTC separation approaches in the future.
Collapse
Affiliation(s)
- Kazem Akbarnataj
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, 11365-11155, Iran; Department of Mechanical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | - Sasan Maleki
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, 11365-11155, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, 11155-9161, Iran
| | - Masoud Rezaeian
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, 11365-11155, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, 11155-9161, Iran
| | - Mohammad Haki
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, 11365-11155, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, 11155-9161, Iran
| | - Amir Shamloo
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, 11365-11155, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, 11155-9161, Iran.
| |
Collapse
|
14
|
Qiu H, Wang H, Yang X, Huo F. High performance isolation of circulating tumor cells by acoustofluidic chip coupled with ultrasonic concentrated energy transducer. Colloids Surf B Biointerfaces 2023; 222:113138. [PMID: 36638753 DOI: 10.1016/j.colsurfb.2023.113138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/02/2023] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
The isolation of circulating tumor cells (CTCs) from whole blood is a challenging task. Although various studies on the separation of CTCs by acoustofluidic devices have been reported, difficulties still persist, such as the complicated equipment, high cost, and difficult operation. Those problems should be resolved urgently. Herein, we developed an acoustofluidic chip separation system coupled with an ultrasonic concentrated energy transducer (UCET) system for efficient separation of CTCs. In the separation system, the acoustically sensitive particles were pre-focused by inertial forces of the PDMS chip channel structure. Then, the particles with different sizes were separated by acoustic radiation forces (ARF). In this study, the circulating tumor cells was simulated (CTCs-like particles) by aminated mesoporous acoustically sensitive particles (MSN@AM) encapsulated carboxylate polystyrene microspheres (PS-COOH). Subsequently, efficient CTCs-like particles separation was achieved by the acoustofluidic chip coupling system. This study effectively separated polystyrene microspheres carrying acoustically sensitive particles (MSN@AM@PS-COOH). However, the MSNs agglomerates and PS microspheres without acoustically sensitive particles did not show phenomenon of separation. This method allows to efficiently separate 2 µm MSNs agglomerates,8.0-8.9 µm PS microspheres and 10-10.5 µm MSN@AM@PS-COOH particles. It is demonstrated that the CTCs-like particles show more sensitive response, longer moving distance, and more obvious separation effect at the condition of the low frequency traveling wave sound field (20 kHz from UCET). This system can maintain the same separation with reduced amount of reagents used for cancer detection. It may provide a reliable basis for sorting out CTCs efficiently from the whole blood of cancer patients.
Collapse
Affiliation(s)
- Hui Qiu
- Analytical Testing Center, Institute of Micro&Nano Intelligent Sensing, Neijiang Normal University, Neijiang 641100, PR China; School of Mechanical Engineering, Chengdu University, Chengdu 610106 Sichuan, PR China
| | - Haoyu Wang
- Analytical Testing Center, Institute of Micro&Nano Intelligent Sensing, Neijiang Normal University, Neijiang 641100, PR China; School of Mechanical Engineering, Chengdu University, Chengdu 610106 Sichuan, PR China
| | - Xiupei Yang
- Chemical Synthesis and Pollution Control Key Laboratory of Sichuan Province, China West Normal University, Nanchong 637000, PR China
| | - Feng Huo
- Analytical Testing Center, Institute of Micro&Nano Intelligent Sensing, Neijiang Normal University, Neijiang 641100, PR China; School of Mechanical Engineering, Chengdu University, Chengdu 610106 Sichuan, PR China.
| |
Collapse
|
15
|
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
|
16
|
Linh ND, Huyen NTT, Dang NH, Piro B, Thi Thu V. Electrochemical interface based on polydopamine and gold nanoparticles/reduced graphene oxide for impedimetric detection of lung cancer cells †. RSC Adv 2023; 13:10082-10089. [PMID: 37006357 PMCID: PMC10052696 DOI: 10.1039/d3ra00793f] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
The use of non-invasive approaches for monitoring therapy processes in cancer patients at late stages is truly needed. In this work, we aim to develop an electrochemical interface based on polydopamine combined with gold nanoparticles and reduced graphene oxide for impedimetric detection of lung cancer cells. Gold nanoparticles (around 75 nm) were dispersed onto reduced graphene oxide material pre-electrodeposited onto disposable fluorine doped tin oxide electrodes. The coordination between gold and carbonaceous material has somehow improved the mechanical stability of this electrochemical interface. Polydopamine was later introduced onto modified electrodes via self-polymerization of dopamine in an alkaline solution. The result has demonstrated the good adhesion and biocompatibility of polydopamine towards A-549 lung cancer cells. The presence of the two conductive materials (gold nanoparticles and reduced graphene oxide) has led to a six-times decrease in charge transfer resistance of polydopamine film. Finally, the as-prepared electrochemical interface was employed for impedimetric detection of A-549 cells. The detection limit was estimated to be only 2 cells per mL. These findings have proved the possibilities to use advanced electrochemical interfaces for point-of-care applications. Non-invasive approaches for monitoring therapy processes in cancer patients at late stages is truly needed.![]()
Collapse
Affiliation(s)
- Nguyen Dieu Linh
- University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST)18 Hoang Quoc Viet, Cau GiayHanoiVietnam
| | - Nguyen Thi Trang Huyen
- University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST)18 Hoang Quoc Viet, Cau GiayHanoiVietnam
| | - Nguyen Hai Dang
- University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST)18 Hoang Quoc Viet, Cau GiayHanoiVietnam
| | - Benoit Piro
- Université Paris Cité, ITODYS, CNRSUMR 7086, 15 Rue J.-A. de BaïfParisF-75013 France
| | - Vu Thi Thu
- University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST)18 Hoang Quoc Viet, Cau GiayHanoiVietnam
| |
Collapse
|
17
|
Reversible capture and release of circulating tumor cells on a three‐dimensional conductive interface to improve cell purity for gene mutation analysis. VIEW 2022. [DOI: 10.1002/viw.20220054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
18
|
Razavi Bazaz S, Mihandust A, Salomon R, Joushani HAN, Li W, A Amiri H, Mirakhorli F, Zhand S, Shrestha J, Miansari M, Thierry B, Jin D, Ebrahimi Warkiani M. Zigzag microchannel for rigid inertial separation and enrichment (Z-RISE) of cells and particles. LAB ON A CHIP 2022; 22:4093-4109. [PMID: 36102894 DOI: 10.1039/d2lc00290f] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Separation and enrichment of target cells prior to downstream analyses is an essential pre-treatment step in many biomedical and clinical assays. Separation techniques utilizing simple, cost-effective, and user-friendly devices are highly desirable, both in the lab and at the point of need. Passive microfluidic approaches, especially inertial microfluidics, fit this brief perfectly and are highly desired. Using an optimized additive manufacturing technique, we developed a zigzag microchannel for rigid inertial separation and enrichment, hereafter referred to as Z-RISE. We empirically showed that the Z-RISE device outperforms equivalent devices based on curvilinear (sinusoidal), asymmetric curvilinear, zigzag with round corners, or square-wave formats and modelled this behavior to gain a better understanding of the physics underpinning the improved focusing and separation performance. The comparison between rigid and soft zigzag microchannels reveals that channel rigidity significantly affects and enhances the focusing performance of the microchannel. Compared to other serpentine microchannels, zigzag microfluidics demonstrates superior separation and purity efficiency due to the sudden channel cross-section expansion at the corners. Within Z-RISE, particles are aligned in either double-side or single-line focusing positions. The transition of particles from a double-focusing line to a single focusing line introduced a new phenomenon referred to as the plus focusing position. We experimentally demonstrated that Z-RISE could enrich leukocytes and their subtypes from diluted and RBC lysed blood while depleting dead cells, debris, and RBCs. Z-RISE was also shown to yield outstanding particle or cell concentration with a concentration efficiency of more than 99.99%. Our data support the great potential of Z-RISE for applications that involve particle and cell manipulations and pave the way for commercialization perspective in the near future.
Collapse
Affiliation(s)
- Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia.
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Asma Mihandust
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia.
| | - Robert Salomon
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia
| | | | - Wenyan Li
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia
| | - Hoseyn A Amiri
- Micro+Nanosystems & Applied Biophysics Laboratory, Department of Mechanical Engineering, Babol Noshirvani University of Technology, P.O. Box 484, Babol 47148-71167, Iran
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Isar 11, Babol 47138-18983, Iran
| | - Fateme Mirakhorli
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia.
| | - Sareh Zhand
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia.
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia.
| | - Morteza Miansari
- Micro+Nanosystems & Applied Biophysics Laboratory, Department of Mechanical Engineering, Babol Noshirvani University of Technology, P.O. Box 484, Babol 47148-71167, Iran
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Isar 11, Babol 47138-18983, Iran
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Adelaide, SA, 5095 Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, Victoria, 3052 Australia
| | - Dayong Jin
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia.
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
19
|
Bakhshi MS, Rizwan M, Khan GJ, Duan H, Zhai K. Design of a novel integrated microfluidic chip for continuous separation of circulating tumor cells from peripheral blood cells. Sci Rep 2022; 12:17016. [PMID: 36220844 PMCID: PMC9554048 DOI: 10.1038/s41598-022-20886-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/20/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer is one of the foremost causes of death globally. Late-stage presentation, inaccessible diagnosis, and treatment are common challenges in developed countries. Detection, enumeration of Circulating Tumor Cells (CTC) as early as possible can reportedly lead to more effective treatment. The isolation of CTC at an early stage is challenging due to the low probability of its presence in peripheral blood. In this study, we propose a novel two-stage, label-free, rapid, and continuous CTC separation device based on hydrodynamic inertial focusing and dielectrophoretic separation. The dominance and differential of wall-induced inertial lift force and Dean drag force inside a curved microfluidic channel results in size-based separation of Red Blood Cells (RBC) and platelets (size between 2-4 µm) from CTC and leukocytes (9-12.2 µm). A numerical model was used to investigate the mechanism of hydrodynamic inertial focusing in a curvilinear microchannel. Simulations were done with the RBCs, platelets, CTCs, and leukocytes (four major subtypes) to select the optimized value of the parameters in the proposed design. In first stage, the focusing behavior of microscale cells was studied to sort leukocytes and CTCs from RBCs, and platelets while viable CTCs were separated from leukocytes based on their inherent electrical properties using dielectrophoresis in the second stage. The proposed design of the device was evaluated for CTC separation efficiency using numerical simulations. This study considered the influence of critical factors like aspect ratio, dielectrophoretic force, channel size, flow rate, separation efficiency, and shape on cell separation. Results show that the proposed device yields viable CTC with 99.5% isolation efficiency with a throughput of 12.2 ml/h.
Collapse
Affiliation(s)
- Maliha Saleem Bakhshi
- grid.444938.60000 0004 0609 0078Mechatronics and Control Engineering Department, University of Engineering and Technology, Lahore, Pakistan
| | - Mohsin Rizwan
- grid.444938.60000 0004 0609 0078Mechatronics and Control Engineering Department, University of Engineering and Technology, Lahore, Pakistan
| | - Ghulam Jilany Khan
- grid.444936.80000 0004 0608 9608Department of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Hong Duan
- grid.263761.70000 0001 0198 0694School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000 China
| | - Kefeng Zhai
- grid.263761.70000 0001 0198 0694School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000 China ,grid.459584.10000 0001 2196 0260Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Guangxi Normal University), Guilin, 541004 People’s Republic of China
| |
Collapse
|
20
|
Bayareh M. Active cell capturing for organ-on-a-chip systems: a review. BIOMED ENG-BIOMED TE 2022; 67:443-459. [PMID: 36062551 DOI: 10.1515/bmt-2022-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/25/2022] [Indexed: 11/15/2022]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that has been proposed as a new powerful cell-based tool to imitate the pathophysiological environment of human organs. For most OOC systems, a pivotal step is to culture cells in microfluidic devices. In active cell capturing techniques, external actuators, such as electrokinetic, magnetic, acoustic, and optical forces, or a combination of these forces, can be applied to trap cells after ejecting cell suspension into the microchannel inlet. This review paper distinguishes the characteristics of biomaterials and evaluates microfluidic technology. Besides, various types of OOC and their fabrication techniques are reported and various active cell capture microstructures are analyzed. Furthermore, their constraints, challenges, and future perspectives are provided.
Collapse
Affiliation(s)
- Morteza Bayareh
- Department of Mechanical Engineering, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
21
|
Sierra-Agudelo J, Rodriguez-Trujillo R, Samitier J. Microfluidics for the Isolation and Detection of Circulating Tumor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:389-412. [PMID: 35761001 DOI: 10.1007/978-3-031-04039-9_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nowadays, liquid biopsy represents one of the most promising techniques for early diagnosis, monitoring, and therapy screening of cancer. This novel methodology includes, among other techniques, the isolation, capture, and analysis of circulating tumor cells (CTCs). Nonetheless, the identification of CTC from whole blood is challenging due to their extremely low concentration (1-100 per ml of whole blood), and traditional methods result insufficient in terms of purity, recovery, throughput and/or viability of the processed sample. In this context, the development of microfluidic devices for detecting and isolating CTCs offers a wide range of new opportunities due to their excellent properties for cell manipulation and the advantages to integrate and bring different laboratory processes into the microscale improving the sensitivity, portability, reducing cost and time. This chapter explores current and recent microfluidic approaches that have been developed for the analysis and detection of CTCs, which involve cell capture methods based on affinity binding and label-free methods and detection based on electrical, chemical, and optical sensors. All the exposed technologies seek to overcome the limitations of commercial systems for the analysis and isolation of CTCs, as well as to provide extended analysis that will allow the development of novel and more efficient diagnostic tools.
Collapse
Affiliation(s)
- Jessica Sierra-Agudelo
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Romen Rodriguez-Trujillo
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. .,Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain.
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| |
Collapse
|
22
|
Zhu Z, Li S, Wu D, Ren H, Ni C, Wang C, Xiang N, Ni Z. High-throughput and label-free enrichment of malignant tumor cells and clusters from pleural and peritoneal effusions using inertial microfluidics. LAB ON A CHIP 2022; 22:2097-2106. [PMID: 35441644 DOI: 10.1039/d2lc00082b] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Accurate and rapid diagnosis of malignant pleural and peritoneal effusions is critical due to potential association with advanced disease stages or progression. Traditional cytodiagnosis suffers from low efficiency and has difficulties in finding malignant tumor cells (MTCs) from a mass of exfoliated cells. Hence, a polymer microfluidic chip with a slanted spiral channel was employed for high-throughput and label-free enrichment of MTCs and MTC clusters from clinical malignant pleural and peritoneal effusions. The slanted spiral channel with trapezoidal cross-sections was fabricated by assembling two patterned polymer films of different thicknesses within one flow channel layer. After systematically exploring the effects of the particle size, effusion concentration, and flow rate on separation performance of the device, we realized the enrichment of MTCs from abundant blood cells in 2-fold diluted effusions. The results indicated that approximately 85% of the spiked tumor cells (A549 and MCF-7 cell lines) were recovered with high purities of over 37% at a high throughput of 2000 μL min-1. In clinical applications, we successfully enriched 24-2691 MTCs per mL from the diluted malignant pleural and peritoneal effusions collected from four types of cancer patients (n = 22). More importantly, the MTC clusters were further purified from single MTCs using a higher flow rate of 3000 μL min-1. Finally, we performed the rapid drug sensitivity test by coupling the microfluidic enrichment with CCK-8 assay. Our approach may serve as valuable assistance to accelerate cancer diagnosis and guide the selection of treatment medications.
Collapse
Affiliation(s)
- Zhixian Zhu
- School of Mechanical Engineering, and, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Shuang Li
- Department of Oncology, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| | - Dan Wu
- Department of Oncology, Jiangyin People's Hospital, Jiangyin, 214400, China
| | - Hui Ren
- School of Mechanical Engineering, and, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Chen Ni
- School of Mechanical Engineering, and, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Cailian Wang
- Department of Oncology, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| | - Nan Xiang
- School of Mechanical Engineering, and, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Zhonghua Ni
- School of Mechanical Engineering, and, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
23
|
Zhang H, Lu M, Xiong Z, Yang J, Tan M, Huang L, Zhu X, Lu Z, Liang Z, Liu H. Rapid trapping and tagging of microparticles in controlled flow by in situ digital projection lithography. LAB ON A CHIP 2022; 22:1951-1961. [PMID: 35377378 DOI: 10.1039/d2lc00186a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Real-time and fast trapping and tagging of microfeatures, such as microparticles and cells, are of great significance for biomedical research. In this work, we propose a novel in situ digital projection lithography technology that integrates real-time, in situ generation of digital masks for particle processing and fluid control into conventional DMD-based projection lithography. With the help of image recognition technology, we rapidly resolve the information of the microparticle profile or channel location, combining the selection of existing masks of different shapes, thus enabling in situ generation of user-customized micro-trap arrays and microfilter arrays for particle trapping and tagging. The success in trapping and filtering single particles, particle arrays, and cells has indicated the promising prospects of this novel technology for broad applications in microfluidics, single-cell analysis, and early-stage disease diagnostics.
Collapse
Affiliation(s)
- Han Zhang
- Center for Advanced Optoelectronic Functional Materials Research, and, Key Laboratory for UV Emitting Materials and Technology of Ministry of Education, National Demonstration Center for Experimental Physics Education, Northeast Normal University, 5268 Renmin Street, Changchun 130024, China.
| | - Meiying Lu
- Center for Advanced Optoelectronic Functional Materials Research, and, Key Laboratory for UV Emitting Materials and Technology of Ministry of Education, National Demonstration Center for Experimental Physics Education, Northeast Normal University, 5268 Renmin Street, Changchun 130024, China.
| | - Zheng Xiong
- Department of Biomedical Engineering and Chemical Engineering, Syracuse University, Syracuse, New York 13244, USA
| | - Jing Yang
- Key Laboratory of Molecular Epigenetics Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
| | - Mingyue Tan
- Center for Advanced Optoelectronic Functional Materials Research, and, Key Laboratory for UV Emitting Materials and Technology of Ministry of Education, National Demonstration Center for Experimental Physics Education, Northeast Normal University, 5268 Renmin Street, Changchun 130024, China.
| | - Long Huang
- Center for Advanced Optoelectronic Functional Materials Research, and, Key Laboratory for UV Emitting Materials and Technology of Ministry of Education, National Demonstration Center for Experimental Physics Education, Northeast Normal University, 5268 Renmin Street, Changchun 130024, China.
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
| | - Zifeng Lu
- Center for Advanced Optoelectronic Functional Materials Research, and, Key Laboratory for UV Emitting Materials and Technology of Ministry of Education, National Demonstration Center for Experimental Physics Education, Northeast Normal University, 5268 Renmin Street, Changchun 130024, China.
| | - Zhongzhu Liang
- Center for Advanced Optoelectronic Functional Materials Research, and, Key Laboratory for UV Emitting Materials and Technology of Ministry of Education, National Demonstration Center for Experimental Physics Education, Northeast Normal University, 5268 Renmin Street, Changchun 130024, China.
| | - Hua Liu
- Center for Advanced Optoelectronic Functional Materials Research, and, Key Laboratory for UV Emitting Materials and Technology of Ministry of Education, National Demonstration Center for Experimental Physics Education, Northeast Normal University, 5268 Renmin Street, Changchun 130024, China.
| |
Collapse
|
24
|
Sung HW, Choi SE, Chu CH, Ouyang M, Kalyan S, Scott N, Hur SC. Sensitizing drug-resistant cancer cells from blood using microfluidic electroporator. PLoS One 2022; 17:e0264907. [PMID: 35259174 PMCID: PMC8903260 DOI: 10.1371/journal.pone.0264907] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Direct assessment of patient samples holds unprecedented potential in the treatment of cancer. Circulating tumor cells (CTCs) in liquid biopsies are a rapidly evolving source of primary cells in the clinic and are ideal candidates for functional assays to uncover real-time tumor information in real-time. However, a lack of routines allowing direct and active interrogation of CTCs directly from liquid biopsy samples represents a bottleneck for the translational use of liquid biopsies in clinical settings. To address this, we present a workflow for using a microfluidic vortex-assisted electroporation system designed for the functional assessment of CTCs purified from blood. Validation of this approach was assessed through drug response assays on wild-type (HCC827 wt) and gefitinib-resistant (HCC827 GR6) non-small cell lung cancer (NSCLC) cells. HCC827 cells trapped within microscale vortices were electroporated to sequentially deliver drug agents into the cytosol. Electroporation conditions facilitating multi-agent delivery were characterized for both cell lines using an automatic single-cell image fluorescence intensity algorithm. HCC827 GR6 cells spiked into the blood to emulate drug-resistant CTCs were able to be collected with high purity, demonstrating the ability of the device to minimize background cell impact for downstream sensitive cell assays. Using our proposed workflow, drug agent combinations to restore gefitinib sensitivity reflected the anticipated cytotoxic response. Taken together, these results represent a microfluidics multi-drug screening panel workflow that can enable functional interrogation of patient CTCs in situ, thereby accelerating the clinical standardization of liquid biopsies.
Collapse
Affiliation(s)
- Hyun Woo Sung
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sung-Eun Choi
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Chris H. Chu
- Department of Internal Medicine, Virginia Mason Medical Center, Seattle, Washington, United States of America
| | - Mengxing Ouyang
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
| | - Srivathsan Kalyan
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Nathan Scott
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Soojung Claire Hur
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland, United States of America
- Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
25
|
A microfluidic device for label-free separation sensitivity enhancement of circulating tumor cells of various and similar size. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2021.118192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
26
|
Xiang N, Ni Z. High-throughput concentration of rare malignant tumor cells from large-volume effusions by multistage inertial microfluidics. LAB ON A CHIP 2022; 22:757-767. [PMID: 35050294 DOI: 10.1039/d1lc00944c] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
On-chip concentration of rare malignant tumor cells (MTCs) in malignant pleural effusions (MPEs) with a large volume is challenging. Previous microfluidic concentrators suffer from a low concentration factor (CF) and a limited processing throughput. This study describes a low-cost multiplexed microfluidic concentrator that can enable high-throughput (up to 16 mL min-1) and high CF (over 40-fold for single run) concentration of rare cells from large-volume biofluids (up to hundreds of milliliters). The multiplexed device was fabricated using inexpensive polymer-film materials using a quick non-clean-room process within 30 min. The multiplexing and flow distribution approaches applied in the device achieved high-throughput processing. By adopting serial cascading, an ultrahigh CF of approximately 1400 was achieved. Moreover, the microfluidic concentrator was successfully applied for the concentration and purification of rare MTCs within MPEs collected from patients with advanced metastatic lung and breast cancers. The provision of concentrated samples with low background cells could improve the sensitivity of cytology and thus reduce the time required for cytological examination. This novel concentrator offers the distinct advantages of a remarkable CF, high throughput, low device cost, and label-free processing and can therefore be readily integrated with other on-chip cell sorters to enhance the identification of MPEs.
Collapse
Affiliation(s)
- Nan Xiang
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Zhonghua Ni
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
27
|
Xu K, Jiao X, Wang P, Chen C, Chen C. Isolation of circulating tumor cells based on magnetophoresis. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2022. [DOI: 10.1016/j.cjac.2022.100058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
28
|
Cha H, Fallahi H, Dai Y, Yuan D, An H, Nguyen NT, Zhang J. Multiphysics microfluidics for cell manipulation and separation: a review. LAB ON A CHIP 2022; 22:423-444. [PMID: 35048916 DOI: 10.1039/d1lc00869b] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Multiphysics microfluidics, which combines multiple functional physical processes in a microfluidics platform, is an emerging research area that has attracted increasing interest for diverse biomedical applications. Multiphysics microfluidics is expected to overcome the limitations of individual physical phenomena through combining their advantages. Furthermore, multiphysics microfluidics is superior for cell manipulation due to its high precision, better sensitivity, real-time tunability, and multi-target sorting capabilities. These exciting features motivate us to review this state-of-the-art field and reassess the feasibility of coupling multiple physical processes. To confine the scope of this paper, we mainly focus on five common forces in microfluidics: inertial lift, elastic, dielectrophoresis (DEP), magnetophoresis (MP), and acoustic forces. This review first explains the working mechanisms of single physical phenomena. Next, we classify multiphysics techniques in terms of cascaded connections and physical coupling, and we elaborate on combinations of designs and working mechanisms in systems reported in the literature to date. Finally, we discuss the possibility of combining multiple physical processes and associated design schemes and propose several promising future directions.
Collapse
Affiliation(s)
- Haotian Cha
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Hedieh Fallahi
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Yuchen Dai
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Dan Yuan
- Centre for Regional and Rural Futures, Deakin University, Geelong, Victoria 3216, Australia
| | - Hongjie An
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Jun Zhang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| |
Collapse
|
29
|
Zhang Y, Li Y, Tan Z. A review of enrichment methods for circulating tumor cells: from single modality to hybrid modality. Analyst 2021; 146:7048-7069. [PMID: 34709247 DOI: 10.1039/d1an01422f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Circulating tumor cell (CTC) analysis as a liquid biopsy can be used for early diagnosis of cancer, evaluating cancer progression, and assessing treatment efficacy. The enrichment of CTCs from patient blood is important for CTC analysis due to the extreme rarity of CTCs. This paper updates recent advances in CTC enrichment methods. We first review single-modality methods, including biophysical and biochemical methods. Hybrid-modality methods, combining at least two single-modality methods, are gaining increasing popularity for their improved performance. Then this paper reviews hybrid-modality methods, which are categorized into integrated and sequenced hybrid-modality methods. The state of the art indicates that the CTC capture efficiencies of integrated hybrid-modality methods can reach 85% or higher by taking advantage of the superimposed and enhanced capture effects from multiple single-modality methods. Moreover, a hybrid method integrating biophysical with biochemical methods is characterized by both high processing rate and high specificity.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, Canada N2L 3G1.
| | - Yifu Li
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, Canada N2L 3G1.
| | - Zhongchao Tan
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, Canada N2L 3G1.
| |
Collapse
|
30
|
Khatri N, Burada PS. Mass separation in an asymmetric channel. Phys Rev E 2021; 104:044109. [PMID: 34781428 DOI: 10.1103/physreve.104.044109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/23/2021] [Indexed: 11/07/2022]
Abstract
We present a mechanism to sort out particles of different masses in an asymmetric channel, where the entropic barriers arise naturally and control the diffusion of these particles. When particles are subjected to an oscillatory force, with the scaled amplitude a and frequency ω, the mean particle velocity exhibits a bell-shaped behavior as a function of the particle mass, indicating that particles with an optimal mass m_{op} drift faster than other particles. By tuning a and ω, we get an empirical relation to estimate m_{op}∼(aω^{2})^{-0.4}. An additional static bias, applied in the opposite direction of the rectified velocity, would push the particles of lighter mass to move in its direction while the others drift opposite to it. This study is useful to design lab-on-a-chip devices for separating particles of different masses.
Collapse
Affiliation(s)
- Narender Khatri
- Department of Physics, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - P S Burada
- Department of Physics, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
31
|
Pang L, Ding J, Liu XX, Kou Z, Guo L, Xu X, Fan SK. Microfluidics-Based Single-Cell Research for Intercellular Interaction. Front Cell Dev Biol 2021; 9:680307. [PMID: 34458252 PMCID: PMC8397490 DOI: 10.3389/fcell.2021.680307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/20/2021] [Indexed: 12/21/2022] Open
Abstract
Intercellular interaction between cell-cell and cell-ECM is critical to numerous biology and medical studies, such as stem cell differentiation, immunotherapy and tissue engineering. Traditional methods employed for delving into intercellular interaction are limited by expensive equipment and sophisticated procedures. Microfluidics technique is considered as one of the powerful measures capable of precisely capturing and manipulating cells and achieving low reagent consumption and high throughput with decidedly integrated functional components. Over the past few years, microfluidics-based systems for intercellular interaction study at a single-cell level have become frequently adopted. This review focuses on microfluidic single-cell studies for intercellular interaction in a 2D or 3D environment with a variety of cell manipulating techniques and applications. The challenges to be overcome are highlighted.
Collapse
Affiliation(s)
- Long Pang
- School of Basic Medical Science, The Shaanxi Key Laboratory of Brain Disorders, Xi’an Medical University, Xi’an, China
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an Medical University, Xi’an, China
| | - Jing Ding
- Department of Mechanical and Nuclear Engineering, Kansas State University, Manhattan, KS, United States
| | - Xi-Xian Liu
- Key Laboratory of Thermo-Fluid Science and Engineering of MOE, School of Energy and Power Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Zhixuan Kou
- School of Basic Medical Science, The Shaanxi Key Laboratory of Brain Disorders, Xi’an Medical University, Xi’an, China
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an Medical University, Xi’an, China
| | - Lulu Guo
- School of Basic Medical Science, The Shaanxi Key Laboratory of Brain Disorders, Xi’an Medical University, Xi’an, China
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an Medical University, Xi’an, China
| | - Xi Xu
- School of Basic Medical Science, The Shaanxi Key Laboratory of Brain Disorders, Xi’an Medical University, Xi’an, China
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an Medical University, Xi’an, China
| | - Shih-Kang Fan
- Department of Mechanical and Nuclear Engineering, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
32
|
A Novel Approach for Tuning of Fluidic Resistance in Deterministic Lateral Displacement Array for Enhanced Separation of Circulating Tumor Cells. Cognit Comput 2021. [DOI: 10.1007/s12559-021-09904-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
33
|
Bai X, Song B, Chen Z, Zhang W, Chen D, Dai Y, Liang S, Zhang D, Zhao Z, Feng L. Postoperative evaluation of tumours based on label-free acoustic separation of circulating tumour cells by microstreaming. LAB ON A CHIP 2021; 21:2721-2729. [PMID: 34165474 DOI: 10.1039/d1lc00165e] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Metastatic tumour recurrence caused by circulating tumour cells (CTCs) after surgery is responsible for more than 90% of tumour-related deaths. A postoperative evaluation system based on the long-term dynamic detection of CTCs helps in guiding the postoperative treatment of tumours in real time and preventing metastases and recurrence of tumours after treatment. In this study, a simple, rapid, and low-cost postoperative evaluation system was established based on the number of CTCs captured by a label-free acoustic separation device from whole blood samples of mice, of which breast tumours were surgically removed, and tumour metastasis was successfully predicted. First, an acoustofluidic device with a custom-designed bottom microcavity array was fabricated to induce highly localised acoustic microstreaming by applying acoustic vibration. Second, experiments of capturing 'defined' cells (artificially mixed individual 4T1 cancer cells into normal blood) based on optimal acoustic streaming were performed. The separation device exhibited a high capture efficiency (>96%). Further applications of capturing the 'true' CTCs derived from postoperative mice were successfully developed to predict tumour prognosis based on the number of captured CTCs. Finally, the prediction was verified through long-term observation of mice with excised tumours. The acoustofluidic device can efficiently capture CTCs and precisely predict tumour metastasis in a low-cost and non-invasive manner. This will help clinicians monitor patients that underwent surgical resection of tumours over a long period of time and facilitate optimal treatment strategies in a timely manner.
Collapse
Affiliation(s)
- Xue Bai
- School of Mechanical Engineering & Automation, Beihang University, Xueyuan Road No. 37, Haidian District, Beijing 100191, China.
| | - Bin Song
- School of Mechanical Engineering & Automation, Beihang University, Xueyuan Road No. 37, Haidian District, Beijing 100191, China.
| | - Ziteng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Wei Zhang
- School of Mechanical Engineering & Automation, Beihang University, Xueyuan Road No. 37, Haidian District, Beijing 100191, China.
| | - Dixiao Chen
- School of Mechanical Engineering & Automation, Beihang University, Xueyuan Road No. 37, Haidian District, Beijing 100191, China.
| | - Yuguo Dai
- School of Mechanical Engineering & Automation, Beihang University, Xueyuan Road No. 37, Haidian District, Beijing 100191, China.
| | - Shuzhang Liang
- School of Mechanical Engineering & Automation, Beihang University, Xueyuan Road No. 37, Haidian District, Beijing 100191, China.
| | - Deyuan Zhang
- School of Mechanical Engineering & Automation, Beihang University, Xueyuan Road No. 37, Haidian District, Beijing 100191, China. and Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
| | - Zhijun Zhao
- Clinical Laboratory Center, General Hospital of Ningxia Medical University, Yinchuan 750001, China and Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan 750001, China
| | - Lin Feng
- School of Mechanical Engineering & Automation, Beihang University, Xueyuan Road No. 37, Haidian District, Beijing 100191, China. and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| |
Collapse
|
34
|
Hymel SJ, Fujioka H, Khismatullin DB. Modeling of Deformable Cell Separation in a Microchannel with Sequenced Pillars. ADVANCED THEORY AND SIMULATIONS 2021. [DOI: 10.1002/adts.202100086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Scott J. Hymel
- Department of Biomedical Engineering Tulane University New Orleans LA 70118 USA
| | - Hideki Fujioka
- Center for Computational Science Tulane University New Orleans LA 70118 USA
| | - Damir B. Khismatullin
- Department of Biomedical Engineering Tulane University New Orleans LA 70118 USA
- Center for Computational Science Tulane University New Orleans LA 70118 USA
| |
Collapse
|
35
|
Müller Bark J, Kulasinghe A, Hartel G, Leo P, Warkiani ME, Jeffree RL, Chua B, Day BW, Punyadeera C. Isolation of Circulating Tumour Cells in Patients With Glioblastoma Using Spiral Microfluidic Technology - A Pilot Study. Front Oncol 2021; 11:681130. [PMID: 34150645 PMCID: PMC8210776 DOI: 10.3389/fonc.2021.681130] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive type of tumour arising from the central nervous system. GBM remains an incurable disease despite advancement in therapies, with overall survival of approximately 15 months. Recent literature has highlighted that GBM releases tumoural content which crosses the blood-brain barrier (BBB) and is detected in patients’ blood, such as circulating tumour cells (CTCs). CTCs carry tumour information and have shown promise as prognostic and predictive biomarkers in different cancer types. Currently, there is limited data for the clinical utility of CTCs in GBM. Here, we report the use of spiral microfluidic technology to isolate CTCs from whole blood of newly diagnosed GBM patients before and after surgery, followed by characterization for GFAP, cell-surface vimentin protein expression and EGFR amplification. CTCs were found in 13 out of 20 patients (9/20 before surgery and 11/19 after surgery). Patients with CTC counts equal to 0 after surgery had a significantly longer recurrence-free survival (p=0.0370). This is the first investigation using the spiral microfluidics technology for the enrichment of CTCs from GBM patients and these results support the use of this technology to better understand the clinical value of CTCs in the management of GBM in future studies.
Collapse
Affiliation(s)
- Juliana Müller Bark
- Saliva and Liquid Biopsy Translational Laboratory, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia
| | - Arutha Kulasinghe
- Saliva and Liquid Biopsy Translational Laboratory, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia
| | - Gunter Hartel
- Department of Statistics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Paul Leo
- Translational Research Institute, Brisbane, QLD, Australia.,Translational Genomics Group, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Majid Ebrahimi Warkiani
- The School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Rosalind L Jeffree
- Department of Statistics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Kenneth G. Jamieson Department of Neurosurgery, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.,Cell and Molecular Biology Department, Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer MRI, Brisbane, QLD, Australia
| | - Benjamin Chua
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Cancer Care Services, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Bryan W Day
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Cell and Molecular Biology Department, Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer MRI, Brisbane, QLD, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational Laboratory, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
36
|
Biomimetic recognition strategy for efficient capture and release of circulating tumor cells. Mikrochim Acta 2021; 188:220. [PMID: 34076759 DOI: 10.1007/s00604-021-04856-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
Efficient capture and release of circulating tumor cells play an important role in cancer diagnosis, but the limited affinity of monovalent adhesion molecules in existing capture technologies leads to low capture efficiency, and the captured cells are difficult to be separated. Inspired by the phenomenon that the long tentacles of jellyfish contain multiple adhesion domains and can effectively capture moving food, we have constructed a biomimetic recognition strategy to capture and release tumor cells. In details, gold-coated magnetic nanomaterials (Au@Fe3O4 NPs) were first prepared and characterized by scanning electron microscopy, UV-vis absorption spectra, and Zeta potential. Then, the DNA primers modified on Au@Fe3O4 nanoparticles can be extended to form many radialized DNA products by rolling circle amplification. These long DNA products resemble jellyfish tentacles and contain multivalent aptamers that can be extended into three dimensions to increase the accessibility of target cells, resulting in efficient, simple, rapid, and specific cells capture. The capture efficiencies are no less than 92% in PBS buffer and 77% in blood. Subsequently, DNase I was selected to degrade biomimetic tentacles to release the captured tumor cells with high viability. This release strategy can not only improve cell viability, but also reduce a tedious release process and unnecessary costs. We believe that the proposed method can be expanded for the capture and release of various tumor cells and will inspire the development of circulating tumor cells analysis. A biomimetic recognition strategy for capture and release of circulating tumor cells has been developed. This method modified specific P1 DNA primers on Au@Fe3O4 NPs to form many radialized DNA products by rolling circle amplification. These products can efficiently capture CTCs since it contains multiple aptamers with a multivalent binding capacity. This make it a promising tool to capture and release of other tumor cells, and will inspire the development of CTC analysis.
Collapse
|
37
|
Carvalho Â, Ferreira G, Seixas D, Guimarães-Teixeira C, Henrique R, Monteiro FJ, Jerónimo C. Emerging Lab-on-a-Chip Approaches for Liquid Biopsy in Lung Cancer: Status in CTCs and ctDNA Research and Clinical Validation. Cancers (Basel) 2021; 13:cancers13092101. [PMID: 33925308 PMCID: PMC8123575 DOI: 10.3390/cancers13092101] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/16/2021] [Accepted: 04/25/2021] [Indexed: 01/31/2023] Open
Abstract
Simple Summary Lung cancer (LCa) remains the leading cause of cancer-related mortality worldwide, with late diagnosis and limited therapeutic approaches still constraining patient’s outcome. In recent years, liquid biopsies have significantly improved the disease characterization and brought new insights into LCa diagnosis and management. The integration of microfluidic devices in liquid biopsies have shown promising results regarding circulating biomarkers isolation and analysis and these tools are expected to establish automatized and standardized results for liquid biopsies in the near future. Herein, we review the status of lab-on-a-chip approaches for liquid biopsies in LCa and highlight their current applications for circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) research and clinical validation studies. Abstract Despite the intensive efforts dedicated to cancer diagnosis and treatment, lung cancer (LCa) remains the leading cause of cancer-related mortality, worldwide. The poor survival rate among lung cancer patients commonly results from diagnosis at late-stage, limitations in characterizing tumor heterogeneity and the lack of non-invasive tools for detection of residual disease and early recurrence. Henceforth, research on liquid biopsies has been increasingly devoted to overcoming these major limitations and improving management of LCa patients. Liquid biopsy is an emerging field that has evolved significantly in recent years due its minimally invasive nature and potential to assess various disease biomarkers. Several strategies for characterization of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) have been developed. With the aim of standardizing diagnostic and follow-up practices, microfluidic devices have been introduced to improve biomarkers isolation efficiency and specificity. Nonetheless, implementation of lab-on-a-chip platforms in clinical practice may face some challenges, considering its recent application to liquid biopsies. In this review, recent advances and strategies for the use of liquid biopsies in LCa management are discussed, focusing on high-throughput microfluidic devices applied for CTCs and ctDNA isolation and detection, current clinical validation studies and potential clinical utility.
Collapse
Affiliation(s)
- Ângela Carvalho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (G.F.); (D.S.); (F.J.M.)
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.G.-T.); (R.H.); (C.J.)
- Correspondence: ; Tel.: +351-226-074-900
| | - Gabriela Ferreira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (G.F.); (D.S.); (F.J.M.)
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.G.-T.); (R.H.); (C.J.)
| | - Duarte Seixas
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (G.F.); (D.S.); (F.J.M.)
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.G.-T.); (R.H.); (C.J.)
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Catarina Guimarães-Teixeira
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.G.-T.); (R.H.); (C.J.)
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Rui Henrique
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.G.-T.); (R.H.); (C.J.)
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Fernando J. Monteiro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (G.F.); (D.S.); (F.J.M.)
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.G.-T.); (R.H.); (C.J.)
- Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e Materiais, Universidade do Porto, Rua Dr Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Carmen Jerónimo
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.G.-T.); (R.H.); (C.J.)
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| |
Collapse
|
38
|
Zhang W, Li N, Lin L, Li H, Lin JM. Metabolism-Based Capture and Analysis of Circulating Tumor Cells in an Open Space. Anal Chem 2021; 93:6955-6960. [PMID: 33900729 DOI: 10.1021/acs.analchem.0c05155] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The level of circulating tumor cells (CTCs) in blood is a predictor of metastatic cancer progress, serving as an important biomarker for cancer diagnosis, prognosis, and therapy. Currently, there are mainly two conventional strategies to distinguish CTCs, including biological property-based affinity capture and physical property-based label-free isolation. Although great progress has been made in this field, the ability to distinguish CTCs still needs to be improved further due to the cell heterogeneity. Herein, a metabolism-based isolation approach was applied to identify tumor cells according to the "Warburg effect", and a bifunctional open-space platform with fluid walls was developed for real-time monitoring and in situ capture/analysis of tumor cells. A drop-on-demand inkjet printing technique was introduced to create a single cell-containing droplet array with high throughput and high encapsulation rate, and the homogeneous crystalline matrix spots ejected from the inkjet also provided high-quality and reproducible lipid profiling. This platform could combine both microscopic image and mass data, and it has been proven to be capable of isolating and identifying CTCs in complex blood samples, making it a promising tool for evaluating the efficacy of therapy and monitoring the disease progression.
Collapse
Affiliation(s)
- Weifei Zhang
- Division of Chemistry and Analytical Science, National Institute of Metrology, Beijing 100029, China.,Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Nan Li
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Ling Lin
- Department of Bioengineering, Beijing Technology and Business University, Beijing 100048, China
| | - Hongmei Li
- Division of Chemistry and Analytical Science, National Institute of Metrology, Beijing 100029, China
| | - Jin-Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
39
|
Zhu Z, Wu D, Li S, Han Y, Xiang N, Wang C, Ni Z. A polymer-film inertial microfluidic sorter fabricated by jigsaw puzzle method for precise size-based cell separation. Anal Chim Acta 2021; 1143:306-314. [PMID: 33384126 DOI: 10.1016/j.aca.2020.11.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/25/2020] [Accepted: 11/02/2020] [Indexed: 12/31/2022]
Abstract
A polymer-film inertial microfluidic jigsaw (PIMJ) sorter with trapezoidal spiral channels using the jigsaw puzzle method was proposed to realize precise and high-throughput rare cell separation. The PIMJ sorter was fabricated by assembling laser-patterned polymer-film layers of different thicknesses. After illustrating the conceptual design and fabrication process, the effects of the cross-sectional dimension, particle size, and operational flow rate on particle focusing were systematically explored under a broad flow rate range. Then, the separation performances of the PIMJ sorter were characterized using the binary particle mixture and the blood samples spiked with four types of tumor cells. The results indicated that the complete separation of the binary particles with a minimum size difference of 2 μm was successfully realized at a high throughput up to 3000 μL/min. A high recovery ratio of 90.57%-94.14% and a high purity of 48.67%-79.05% were achieved for the separation of rare tumor cells from white blood cells (WBCs). Finally, the PIMJ sorter was successfully employed for separating rare circulating tumor cells (CTCs) from the metastatic breast and lung cancer patients with a capture ratio of 7-226 CTCs per 5 mL sample. The results proved the high sensitivity and high reliability of the PIMJ sorter. The PIMJ sorter is expected to be a potential device for precise CTC separation towards the clinical applications.
Collapse
Affiliation(s)
- Zhixian Zhu
- School of Mechanical Engineering, And Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China
| | - Dan Wu
- Nanjing Medical University, Nanjing, 210011, Jiangsu, China; Department of Oncology, Jiangyin People's Hospital, Jiangyin, 214400, China
| | - Shuang Li
- Department of Oncology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Yu Han
- School of Mechanical Engineering, And Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China
| | - Nan Xiang
- School of Mechanical Engineering, And Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Cailian Wang
- Nanjing Medical University, Nanjing, 210011, Jiangsu, China; Department of Oncology, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| | - Zhonghua Ni
- School of Mechanical Engineering, And Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
40
|
Dudaie M, Nissim N, Barnea I, Gerling T, Duschl C, Kirschbaum M, Shaked NT. Label-free discrimination and selection of cancer cells from blood during flow using holography-induced dielectrophoresis. JOURNAL OF BIOPHOTONICS 2020; 13:e202000151. [PMID: 32700785 DOI: 10.1002/jbio.202000151] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/02/2020] [Accepted: 07/21/2020] [Indexed: 05/26/2023]
Abstract
We present a method for label-free imaging and sorting of cancer cells in blood, which is based on a dielectrophoretic microfluidic chip and label-free interferometric phase microscopy. The chip used for imaging has been embedded with dielectrophoretic electrodes, and therefore it can be used to sort the cells based on the decisions obtained during the cell flow by the label-free quantitative imaging method. Hence, we obtained a real-time, automatic, label-free imaging flow cytometry with the ability to sort the cells during flow. To validate our model, we combined into the label-free imaging interferometer a fluorescence imaging channel that indicated the correctness of the label-free sorting. We have achieved above 98% classification success and 69% sorting accuracy at flow rates of 4 to 7 μL hr-1 . In the future, this method is expected to help in label-free sorting of circulating tumor cells in blood following an initial state-of-the-art cell enrichment.
Collapse
Affiliation(s)
- Matan Dudaie
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Noga Nissim
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Itay Barnea
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Tobias Gerling
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses, Potsdam, Germany
| | - Claus Duschl
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses, Potsdam, Germany
| | - Michael Kirschbaum
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses, Potsdam, Germany
| | - Natan T Shaked
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
41
|
|
42
|
Derakhshan R, Ramiar A, Ghasemi A. Numerical investigation into continuous separation of particles and cells in a two-component fluid flow using dielectrophoresis. J Mol Liq 2020. [DOI: 10.1016/j.molliq.2020.113211] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
43
|
Nasiri R, Shamloo A, Ahadian S, Amirifar L, Akbari J, Goudie MJ, Lee K, Ashammakhi N, Dokmeci MR, Di Carlo D, Khademhosseini A. Microfluidic-Based Approaches in Targeted Cell/Particle Separation Based on Physical Properties: Fundamentals and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2000171. [PMID: 32529791 DOI: 10.1002/smll.202000171] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/15/2020] [Indexed: 06/11/2023]
Abstract
Cell separation is a key step in many biomedical research areas including biotechnology, cancer research, regenerative medicine, and drug discovery. While conventional cell sorting approaches have led to high-efficiency sorting by exploiting the cell's specific properties, microfluidics has shown great promise in cell separation by exploiting different physical principles and using different properties of the cells. In particular, label-free cell separation techniques are highly recommended to minimize cell damage and avoid costly and labor-intensive steps of labeling molecular signatures of cells. In general, microfluidic-based cell sorting approaches can separate cells using "intrinsic" (e.g., fluid dynamic forces) versus "extrinsic" external forces (e.g., magnetic, electric field, etc.) and by using different properties of cells including size, density, deformability, shape, as well as electrical, magnetic, and compressibility/acoustic properties to select target cells from a heterogeneous cell population. In this work, principles and applications of the most commonly used label-free microfluidic-based cell separation methods are described. In particular, applications of microfluidic methods for the separation of circulating tumor cells, blood cells, immune cells, stem cells, and other biological cells are summarized. Computational approaches complementing such microfluidic methods are also explained. Finally, challenges and perspectives to further develop microfluidic-based cell separation methods are discussed.
Collapse
Affiliation(s)
- Rohollah Nasiri
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, 11365-11155, Iran
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, 11365-11155, Iran
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA, 90024, USA
| | - Leyla Amirifar
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, 11365-11155, Iran
| | - Javad Akbari
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, 11365-11155, Iran
| | - Marcus J Goudie
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - KangJu Lee
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Mehmet R Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA, 90024, USA
- Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA, 90024, USA
- Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
44
|
Akpe V, Kim TH, Brown CL, Cock IE. Circulating tumour cells: a broad perspective. J R Soc Interface 2020; 17:20200065. [PMCID: PMC7423436 DOI: 10.1098/rsif.2020.0065] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/09/2020] [Indexed: 08/13/2023] Open
Abstract
Circulating tumour cells (CTCs) have recently been identified as valuable biomarkers for diagnostic and prognostic evaluations, as well for monitoring therapeutic responses to treatments. CTCs are rare cells which may be present as one CTC surrounded by approximately 1 million white blood cells and 1 billion red blood cells per millilitre of peripheral blood. Despite the various challenges in CTC detection, considerable progress in detection methods have been documented in recent times, particularly for methodologies incorporating nanomaterial-based platforms and/or integrated microfluidics. Herein, we summarize the importance of CTCs as biological markers for tumour detection, highlight their mechanism of cellular invasion and discuss the various challenges associated with CTC research, including vulnerability, heterogeneity, phenotypicity and size differences. In addition, we describe nanomaterial agents used for electrochemistry and surface plasmon resonance applications, which have recently been used to selectively capture cancer cells and amplify signals for CTC detection. The intrinsic properties of nanomaterials have also recently been exploited to achieve photothermal destruction of cancer cells. This review describes recent advancements and future perspectives in the CTC field.
Collapse
Affiliation(s)
- Victor Akpe
- School of Environment and Science, Griffith University, Nathan Campus, Queensland 4111, Australia
- Environmental Futures Research Institute, Griffith University, Nathan Campus, Queensland 4111, Australia
| | - Tak H. Kim
- School of Environment and Science, Griffith University, Nathan Campus, Queensland 4111, Australia
- Environmental Futures Research Institute, Griffith University, Nathan Campus, Queensland 4111, Australia
| | - Christopher L. Brown
- School of Environment and Science, Griffith University, Nathan Campus, Queensland 4111, Australia
- Environmental Futures Research Institute, Griffith University, Nathan Campus, Queensland 4111, Australia
| | - Ian E. Cock
- School of Environment and Science, Griffith University, Nathan Campus, Queensland 4111, Australia
- Environmental Futures Research Institute, Griffith University, Nathan Campus, Queensland 4111, Australia
| |
Collapse
|
45
|
Tottori N, Nisisako T. Particle/cell separation using sheath-free deterministic lateral displacement arrays with inertially focused single straight input. LAB ON A CHIP 2020; 20:1999-2008. [PMID: 32373868 DOI: 10.1039/d0lc00354a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
This paper proposes microfluidic particle separation by sheath-free deterministic lateral displacement (DLD) with inertial focusing in a single straight input channel. Unlike conventional DLD devices for size-based particle separation, in which sheath streams are used to focus the particles before the solution containing them reaches the DLD arrays, the proposed method uses inertial focusing to align the particles along the middle or the sidewalls of the straight rectangular input channel. The two-stage model of inertial focusing is applied to reduce the length of the side-focusing channel. The proposed method is demonstrated by using it to separate fluorescent polymer particles of diameters 13 and 7 μm, in the process of which the effect of the particle focusing regime on the separation performance is also investigated. Through middle focusing, the method is further used to separate MCF-7 cells (a model of circulating tumor cells (CTCs)) and blood cells, with ∼99.0% capture efficiency achieved.
Collapse
Affiliation(s)
- Naotomo Tottori
- Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.
| | - Takasi Nisisako
- Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.
| |
Collapse
|
46
|
Luo L, He Y. Magnetically driven microfluidics for isolation of circulating tumor cells. Cancer Med 2020; 9:4207-4231. [PMID: 32325536 PMCID: PMC7300401 DOI: 10.1002/cam4.3077] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Circulating tumor cells (CTCs) largely contribute to cancer metastasis and show potential prognostic significance in cancer isolation and detection. Miniaturization has progressed significantly in the last decade which in turn enabled the development of several microfluidic systems. The microfluidic systems offer a controlled microenvironment for studies of fundamental cell biology, resulting in the rapid development of microfluidic isolation of CTCs. Due to the inherent ability of magnets to provide forces at a distance, the technology of CTCs isolation based on the magnetophoresis mechanism has become a routine methodology. This historical review aims to introduce two principles of magnetic isolation and recent techniques, facilitating research in this field and providing alternatives for researchers in their study of magnetic isolation. Researchers intend to promote effective CTC isolation and analysis as well as active development of next-generation cancer treatment. The first part of this review summarizes the primary principles based on positive and negative magnetophoretic isolation and describes the metrics for isolation performance. The second part presents a detailed overview of the factors that affect the performance of CTC magnetic isolation, including the magnetic field sources, functionalized magnetic nanoparticles, magnetic fluids, and magnetically driven microfluidic systems.
Collapse
Affiliation(s)
- Laan Luo
- School of Chemical EngineeringKunming University of Science and TechnologyKunmingChina
| | - Yongqing He
- School of Chemical EngineeringKunming University of Science and TechnologyKunmingChina
- Chongqing Key Laboratory of Micro‐Nano System and Intelligent SensingChongqing Technology and Business UniversityChongqingChina
| |
Collapse
|
47
|
Chelakkot C, Ryu J, Kim MY, Kim JS, Kim D, Hwang J, Park SH, Ko SB, Park JW, Jung MY, Kim RN, Song K, Kim YJ, Choi YL, Lee HS, Shin YK. An Immune-Magnetophoretic Device for the Selective and Precise Enrichment of Circulating Tumor Cells from Whole Blood. MICROMACHINES 2020; 11:mi11060560. [PMID: 32486306 PMCID: PMC7345362 DOI: 10.3390/mi11060560] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
Abstract
Here, we validated the clinical utility of our previously developed microfluidic device, GenoCTC, which is based on bottom magnetophoresis, for the isolation of circulating tumor cells (CTCs) from patient whole blood. GenoCTC allowed 90% purity, 77% separation rate, and 80% recovery of circulating tumor cells at a 90 μL/min flow rate when tested on blood spiked with epithelial cell adhesion molecule (EpCAM)-positive Michigan Cancer Foundation-7 (MCF7) cells. Clinical studies were performed using blood samples from non-small cell lung cancer (NSCLC) patients. Varying numbers (2 to 114) of CTCs were found in each NSCLC patient, and serial assessment of CTCs showed that the CTC count correlated with the clinical progression of the disease. The applicability of GenoCTC to different cell surface biomarkers was also validated in a cholangiocarcinoma patient using anti-EPCAM, anti-vimentin, or anti-tyrosine protein kinase MET (c-MET) antibodies. After EPCAM-, vimentin-, or c-MET-positive cells were isolated, CTCs were identified and enumerated by immunocytochemistry using anti-cytokeratin 18 (CK18) and anti-CD45 antibodies. Furthermore, we checked the protein expression of PDL1 and c-MET in CTCs. A study in a cholangiocarcinoma patient showed that the number of CTCs varied depending on the biomarker used, indicating the importance of using multiple biomarkers for CTC isolation and enumeration.
Collapse
Affiliation(s)
- Chaithanya Chelakkot
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Jiyeon Ryu
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Mi Young Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Korea; (M.Y.K.); (J.-S.K.)
| | - Jin-Soo Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Korea; (M.Y.K.); (J.-S.K.)
| | - Dohyeong Kim
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Juhyun Hwang
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Sung Hoon Park
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Seok Bum Ko
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Jeong Won Park
- IT Convergence Technology Research Laboratory, Electronic and Telecommunications Research Institute, Daejon 34129, Korea; (J.W.P.); (M.Y.J.)
| | - Moon Youn Jung
- IT Convergence Technology Research Laboratory, Electronic and Telecommunications Research Institute, Daejon 34129, Korea; (J.W.P.); (M.Y.J.)
| | - Ryong Nam Kim
- Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Korea;
| | - Kyoung Song
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul 08394, Korea; (K.S.); (Y.J.K.)
| | - Yu Jin Kim
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul 08394, Korea; (K.S.); (Y.J.K.)
| | - Yoon-La Choi
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 08394, Korea;
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hun Seok Lee
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
- Correspondence: (H.S.L.); (Y.K.S.)
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Seoul 08826, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea
- The Center for Anti-Cancer Companion Diagnostics, Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Korea
- Correspondence: (H.S.L.); (Y.K.S.)
| |
Collapse
|
48
|
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: 7.3] [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
|
49
|
Yang C, Chen F, Wang S, Xiong B. Circulating Tumor Cells in Gastrointestinal Cancers: Current Status and Future Perspectives. Front Oncol 2019; 9:1427. [PMID: 31921680 PMCID: PMC6923205 DOI: 10.3389/fonc.2019.01427] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/29/2019] [Indexed: 12/24/2022] Open
Abstract
Circulating tumor cells (CTCs), which are now defined as the "break away" cancer cells that derive from primary- or metastatic-tumor sites and present in the bloodstream, are considered to be the precursors of metastases. Considering the key role of CTCs in cancer progression, researchers are committed to analyze them in the past decades and many technologies have been proposed for achieving CTCs isolation and characterization with highly sensitivity and specificity until now. On this basis, clinicians gradually realize the clinical values of CTCs' detection through various clinical studies. As a "liquid biopsy," CTCs' detection and measurement can supply important information for predicting patient's survival, monitoring of response/resistance, assessment of minimal residual disease, evaluating distant metastasis, and sometimes, customizing therapy choices. Nowadays, eliminating CTCs of the blood circulation has been regarded as a promising method to prevent tumor metastasis. However, research on CTCs still faces many challenges. Herein, we present an overview to discuss the current concept of CTCs, summarize the available techniques for CTCs detection, and provide an update on the clinical significance of CTCs in gastrointestinal malignancies, especially focus on gastric and colorectal cancer.
Collapse
Affiliation(s)
- Chaogang Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Fangfang Chen
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China.,Department of Breast and Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuyi Wang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| |
Collapse
|
50
|
Nanduri LK, Hissa B, Weitz J, Schölch S, Bork U. The prognostic role of circulating tumor cells in colorectal cancer. Expert Rev Anticancer Ther 2019; 19:1077-1088. [PMID: 31778322 DOI: 10.1080/14737140.2019.1699065] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Metastasis is the main cause of cancer-associated death in colorectal cancer (CRC). The presence of circulating tumor cells (CTC) in the blood is associated with an increased risk of recurrence and poor prognosis. The clinical significance of CTCs as a novel biomarker has been extensively studied in the last decade. It has been shown that CTC detection applies to early cancer detection. The presence of CTCs is associated with metastatic spread and poor survival and is also useful as a marker for therapy response.Areas covered: We summarize the role of CTC in CRC, their clinical significance, current methods for CTC detection and challenges as well as future perspectives of CTC research.Expert commentary: The clinical significance of CTC in CRC patients is well established. Although insightful, the available marker-based approaches hampered our understanding of the CTCs and their biology, as such approaches do not take into account the heterogeneity of these cell populations. New technologies should expand the marker-based detection to multi biomarker-based approaches together with recent technological advances in microfluidics for single cell enrichment and analysis.
Collapse
Affiliation(s)
- Lahiri Kanth Nanduri
- Department of Gastrointestinal-, Thoracic- and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Hissa
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jürgen Weitz
- Department of Gastrointestinal-, Thoracic- and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Schölch
- Department of Gastrointestinal-, Thoracic- and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ulrich Bork
- Department of Gastrointestinal-, Thoracic- and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|