1
|
Salomon R, Razavi Bazaz S, Mutafopulos K, Gallego-Ortega D, Warkiani M, Weitz D, Jin D. Challenges in blood fractionation for cancer liquid biopsy: how can microfluidics assist? LAB ON A CHIP 2025. [PMID: 39775440 DOI: 10.1039/d4lc00563e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Liquid biopsy provides a minimally invasive approach to characterise the molecular and phenotypic characteristics of a patient's individual tumour by detecting evidence of cancerous change in readily available body fluids, usually the blood. When applied at multiple points during the disease journey, it can be used to monitor a patient's response to treatment and to personalise clinical management based on changes in disease burden and molecular findings. Traditional liquid biopsy approaches such as quantitative PCR, have tended to look at only a few biomarkers, and are aimed at early detection of disease or disease relapse using predefined markers. With advances in the next generation sequencing (NGS) and single-cell genomics, simultaneous analysis of both circulating tumour DNA (ctDNA) and circulating tumour cells (CTCs) is now a real possibility. To realise this, however, we need to overcome issues with current blood collection and fractionation processes. These include overcoming the need to add a preservative to the collection tube or the need to rapidly send blood tubes to a centralised processing lab with the infrastructure required to fractionate and process the blood samples. This review focuses on outlining the current state of liquid biopsy and how microfluidic blood fractionation tools can be used in cancer liquid biopsy. We describe microfluidic devices that can separate plasma for ctDNA analysis, and devices that are important in isolating the cellular component(s) in liquid biopsy, i.e., individual CTCs and CTC clusters. To facilitate a better understanding of these devices, we propose a new categorisation system based on how these devices operate. The three categories being 1) solid Interaction devices, 2) fluid Interaction devices and 3) external force/active devices. Finally, we conclude that whilst some assays and some cancers are well suited to current microfluidic techniques, new tools are necessary to support broader, clinically relevant multiomic workflows in cancer liquid biopsy.
Collapse
Affiliation(s)
- Robert Salomon
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, Australia.
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
| | - Sajad Razavi Bazaz
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, Australia.
| | - Kirk Mutafopulos
- Department of Physics, Harvard University, Cambridge, MA, 02138, USA
| | - David Gallego-Ortega
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - Majid Warkiani
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - David Weitz
- Department of Physics, Harvard University, Cambridge, MA, 02138, USA
| | - Dayong Jin
- Institute for Biomedical Materials and Devices (IBMD)/Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007 Australia
| |
Collapse
|
2
|
Clark MJ, Moser HJ, Anand RK. Dielectrophoretic capture and electrochemical enzyme-linked immunosorbent assay of single melanoma cells at an array of interlocked spiral bipolar electrodes. ChemElectroChem 2024; 11:e202400182. [PMID: 39483376 PMCID: PMC11526340 DOI: 10.1002/celc.202400182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Indexed: 11/03/2024]
Abstract
Analysis of single cancer cells is critical to obtain accurate patient diagnosis and prognosis. In this work, we report the selective dielectrophoretic capture and electrochemical analysis of single melanoma cells at an array of interlocked spiral bipolar electrodes (iBPEs). Following dielectrophoretic capture, individual melanoma cells are hydrodynamically transferred into picoliter-scale chambers for subsequent analysis. The interlocked spiral end of the iBPE (the sensing pole) is utilized to read out an electrochemical enzyme-linked immunosorbent assay (eELISA), which quantifies the expression of a cell surface antigen, melanoma cell adhesion marker (MCAM). The opposite pole of each BPE is located in a fluidically isolated compartment containing reagents for electrogenerated chemiluminescence (ECL), such that luminescence reports iBPE current. In a preliminary device design, the ECL intensity was insufficient to detect MCAM expression on single cells. To achieve single-cell analysis, we decreased the gap size between the interlocked spirals tenfold (5.0 μm to 0.5 μm), thereby creating a more sensitive biosensor by enhanced redox cycling of the product of eELISA. This work is significant because it allows for the selective isolation and sensitive analysis of individual melanoma cells in a device amenable to point-of-care (POC) application by combining dielectrophoresis (DEP) with interdigitated bipolar electrodes (IDBPEs).
Collapse
Affiliation(s)
- Morgan J Clark
- Department of Chemistry, Iowa State University, 1605 Gilman Hall, 2415 Osborn Drive, Ames, IA 50011-1021
| | - Hanna J Moser
- Department of Chemistry, Iowa State University, 1605 Gilman Hall, 2415 Osborn Drive, Ames, IA 50011-1021
| | - Robbyn K Anand
- Department of Chemistry, Iowa State University, 1605 Gilman Hall, 2415 Osborn Drive, Ames, IA 50011-1021
| |
Collapse
|
3
|
Gu H, Chen Y, Lüders A, Bertrand T, Hanedan E, Nielaba P, Bechinger C, Nelson BJ. Scalable high-throughput microfluidic separation of magnetic microparticles. DEVICE 2024; 2:100403. [PMID: 39081390 PMCID: PMC11285115 DOI: 10.1016/j.device.2024.100403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/05/2024] [Accepted: 05/01/2024] [Indexed: 08/02/2024]
Abstract
Surface-engineered magnetic microparticles are used in chemical and biomedical engineering due to their ease of synthesis, high surface-to-volume ratio, selective binding, and magnetic separation. To separate them from fluid suspensions, existing methods rely on the magnetic force introduced by the local magnetic field gradient. However, this strategy has poor scalability because the magnetic field gradient decreases rapidly as one moves away from the magnets. Here, we present a scalable high-throughput magnetic separation strategy using a rotating permanent magnet and two-dimensional arrays of micromagnets. Under a dynamic magnetic field, nickel micromagnets allow the surrounding magnetic microparticles to self-assemble into large clusters and effectively propel themselves through the flow. The collective speed of the microparticle swarm reaches about two orders of magnitude higher than the gradient-based separation method over a wide range of operating frequencies and distances from a rotating magnet.
Collapse
Affiliation(s)
- Hongri Gu
- Department of Physics, University of Konstanz, Konstanz 78464, Germany
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich CH-8092, Switzerland
| | - Yonglin Chen
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich CH-8092, Switzerland
| | - Anton Lüders
- Department of Physics, University of Konstanz, Konstanz 78464, Germany
| | - Thibaud Bertrand
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich CH-8092, Switzerland
| | - Emre Hanedan
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich CH-8092, Switzerland
| | - Peter Nielaba
- Department of Physics, University of Konstanz, Konstanz 78464, Germany
| | - Clemens Bechinger
- Department of Physics, University of Konstanz, Konstanz 78464, Germany
| | - Bradley J. Nelson
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich CH-8092, Switzerland
| |
Collapse
|
4
|
Sun W, You X, Zhao X, Zhang X, Yang C, Zhang F, Yu J, Yang K, Wang J, Xu F, Chang Y, Qu B, Zhao X, He Y, Wang Q, Chen J, Qing G. Precise Capture and Dynamic Release of Circulating Liver Cancer Cells with Dual-Histidine-Based Cell Imprinted Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402379. [PMID: 38655900 DOI: 10.1002/adma.202402379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/22/2024] [Indexed: 04/26/2024]
Abstract
Circulating tumor cells (CTCs) detection presents significant advantages in diagnosing liver cancer due to its noninvasiveness, real-time monitoring, and dynamic tracking. However, the clinical application of CTCs-based diagnosis is largely limited by the challenges of capturing low-abundance CTCs within a complex blood environment while ensuring them alive. Here, an ultrastrong ligand, l-histidine-l-histidine (HH), specifically targeting sialylated glycans on the surface of CTCs, is designed. Furthermore, HH is integrated into a cell-imprinted polymer, constructing a hydrogel with precise CTCs imprinting, high elasticity, satisfactory blood compatibility, and robust anti-interference capacities. These features endow the hydrogel with excellent capture efficiency (>95%) for CTCs in peripheral blood, as well as the ability to release CTCs controllably and alive. Clinical tests substantiate the accurate differentiation between liver cancer, cirrhosis, and healthy groups using this method. The remarkable diagnostic accuracy (94%), lossless release of CTCs, material reversibility, and cost-effectiveness ($6.68 per sample) make the HH-based hydrogel a potentially revolutionary technology for liver cancer diagnosis and single-cell analysis.
Collapse
Affiliation(s)
- Wenjing Sun
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, P. R. China
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
| | - Xin You
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, 116023, P. R. China
| | - Xinjia Zhao
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
| | - Xiaoyu Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
| | - Chunhui Yang
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, 116023, P. R. China
| | - Fusheng Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
- College of Chemistry and Chemical Engineering, Wuhan Textile University, Wuhan, 430200, P. R. China
| | - Jiaqi Yu
- College of Chemistry and Chemical Engineering, Wuhan Textile University, Wuhan, 430200, P. R. China
| | - Kaiguang Yang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
| | - Jixia Wang
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, P. R. China
| | - Fangfang Xu
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, P. R. China
| | - Yongxin Chang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
| | - Boxin Qu
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, 116023, P. R. China
| | - Xinmiao Zhao
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, P. R. China
| | - Yuxuan He
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, P. R. China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, 116023, P. R. China
| | - Jinghua Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| | - Guangyan Qing
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
- College of Chemistry and Chemical Engineering, Wuhan Textile University, Wuhan, 430200, P. R. China
| |
Collapse
|
5
|
Amontree J, Chen K, Varillas J, Fan ZH. A Capillary-Force-Driven, Single-Cell Transfer Method for Studying Rare Cells. Bioengineering (Basel) 2024; 11:542. [PMID: 38927778 PMCID: PMC11200440 DOI: 10.3390/bioengineering11060542] [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: 03/01/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
The characterization of individual cells within heterogeneous populations (e.g., rare tumor cells in healthy blood cells) has a great impact on biomedical research. To investigate the properties of these specific cells, such as genetic biomarkers and/or phenotypic characteristics, methods are often developed for isolating rare cells among a large number of background cells before studying their genetic makeup and others. Prior to using real-world samples, these methods are often evaluated and validated by spiking cells of interest (e.g., tumor cells) into a sample matrix (e.g., healthy blood) as model samples. However, spiking tumor cells at extremely low concentrations is challenging in a standard laboratory setting. People often circumvent the problem by diluting a solution of high-concentration cells, but the concentration becomes inaccurate after series dilution due to the fact that a cell suspension solution can be inhomogeneous, especially when the cell concentration is very low. We report on an alternative method for low-cost, accurate, and reproducible low-concentration cell spiking without the use of external pumping systems. By inducing a capillary force from sudden pressure drops, a small portion of the cellular membrane was aspirated into the reservoir tip, allowing for non-destructive single-cell transfer. We investigated the surface membrane tensions induced by cellular aspiration and studied a range of tip/tumor cell diameter combinations, ensuring that our method does not affect cell viability. In addition, we performed single-cell capture and transfer control experiments using human acute lymphoblastic leukemia cells (CCRF-CEM) to develop calibrated data for the general production of low-concentration samples. Finally, we performed affinity-based tumor cell isolation using this method to generate accurate concentrations ranging from 1 to 15 cells/mL.
Collapse
Affiliation(s)
- Jacob Amontree
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA;
| | - Kangfu Chen
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA;
- Department of Biomedical Engineering, Northwestern University, Chicago, IL 60611, USA
| | - Jose Varillas
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA;
| | - Z. Hugh Fan
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA;
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA;
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
6
|
Wang Z, Wang H, Lin S, Angers S, Sargent EH, Kelley SO. Phenotypic targeting using magnetic nanoparticles for rapid characterization of cellular proliferation regulators. SCIENCE ADVANCES 2024; 10:eadj1468. [PMID: 38718125 PMCID: PMC11078187 DOI: 10.1126/sciadv.adj1468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 04/03/2024] [Indexed: 05/12/2024]
Abstract
Genome-wide CRISPR screens have provided a systematic way to identify essential genetic regulators of a phenotype of interest with single-cell resolution. However, most screens use live/dead readout of viability to identify factors of interest. Here, we describe an approach that converts cell proliferation into the degree of magnetization, enabling downstream microfluidic magnetic sorting to be performed. We performed a head-to-head comparison and verified that the magnetic workflow can identify the same hits from a traditional screen while reducing the screening period from 4 weeks to 1 week. Taking advantage of parallelization and performance, we screened multiple mesenchymal cancer cell lines for their dependency on cell proliferation. We found and validated pan- and cell-specific potential therapeutic targets. The method presented provides a nanoparticle-enabled approach means to increase the breadth of data collected in CRISPR screens, enabling the rapid discovery of drug targets for treatment.
Collapse
Affiliation(s)
- Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
| | - Hansen Wang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
| | - Sichun Lin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto M5S 3E1, Canada
| | - Stephane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto M5S 3E1, Canada
| | - Edward H. Sargent
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada
- Department of Chemistry, Weinberg College of Arts and Science, Northwestern University, Evanston, IL 60208, USA
- Department of Electrical and Computer Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
| | - Shana O. Kelley
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada
- Department of Chemistry, Weinberg College of Arts and Science, Northwestern University, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Chan Zuckerberg Biohub Chicago, Chicago, IL 60607, USA
| |
Collapse
|
7
|
Wang M, Jin L, Hang-Mei Leung P, Wang-Ngai Chow F, Zhao X, Chen H, Pan W, Liu H, Li S. Advancements in magnetic nanoparticle-based biosensors for point-of-care testing. Front Bioeng Biotechnol 2024; 12:1393789. [PMID: 38725992 PMCID: PMC11079239 DOI: 10.3389/fbioe.2024.1393789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
The significance of point-of-care testing (POCT) in early clinical diagnosis and personalized patient care is increasingly recognized as a crucial tool in reducing disease outbreaks and improving patient survival rates. Within the realm of POCT, biosensors utilizing magnetic nanoparticles (MNPs) have emerged as a subject of substantial interest. This review aims to provide a comprehensive evaluation of the current landscape of POCT, emphasizing its growing significance within clinical practice. Subsequently, the current status of the combination of MNPs in the Biological detection has been presented. Furthermore, it delves into the specific domain of MNP-based biosensors, assessing their potential impact on POCT. By combining existing research and spotlighting pivotal discoveries, this review enhances our comprehension of the advancements and promising prospects offered by MNP-based biosensors in the context of POCT. It seeks to facilitate informed decision-making among healthcare professionals and researchers while also promoting further exploration in this promising field of study.
Collapse
Affiliation(s)
- Miaomiao Wang
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China
| | - Lian Jin
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China
| | - Polly Hang-Mei Leung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Franklin Wang-Ngai Chow
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiaoni Zhao
- Guangzhou Wanfu Biotechnology Company, Guangzhou, China
| | - Hui Chen
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China
| | - Wenjing Pan
- Hengyang Medical School, University of South China, Hengyang, China
| | - Hongna Liu
- Hengyang Medical School, University of South China, Hengyang, China
| | - Song Li
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China
- Hengyang Medical School, University of South China, Hengyang, China
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Healthcare Hospital, Changsha, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, China
| |
Collapse
|
8
|
Huang J, Zhang D, Zu Y, Zhang L. Procalcitonin Detection Using Immunomagnetic Beads-Mediated Surface-Enhanced Raman Spectroscopy. BIOSENSORS 2024; 14:164. [PMID: 38667157 PMCID: PMC11048292 DOI: 10.3390/bios14040164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024]
Abstract
The early detection of procalcitonin (PCT) is crucial for diagnosing bacterial infections due to its high sensitivity and specificity. While colloidal gold colorimetric and immune-chemiluminescence methods are commonly employed in clinical detection, the former lacks sensitivity, and the latter faces challenges with a brief luminescence process and an elevated background. Here, we introduce a novel approach for the quantitative analysis of PCT using surface-enhanced Raman spectroscopy (SERS), leveraging the enhanced properties of metal nanoparticles. Simultaneously, we employed a magnetic nanoparticle coating and surface biofunctionalization modification to immobilize PCT-trapping antibodies, creating the required immune substrates. The resulting magnetic nanoparticles and antibody complexes, acting as carriers and recognition units, exhibited superparamagnetism and the specific recognition of biomarkers. Then, this complex efficiently underwent magnetic separation with an applied magnetic field, streamlining the cumbersome steps of traditional ELISA and significantly reducing the detection time. In conclusion, the exploration of immunomagnetic bead detection technology based on surface-enhanced Raman spectroscopy holds crucial practical significance for the sensitive detection of PCT.
Collapse
Affiliation(s)
- Jiayue Huang
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China;
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative In-novation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Dagan Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yan Zu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Lexiang Zhang
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China;
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| |
Collapse
|
9
|
Labib M, Wang Z, Kim Y, Lin S, Abdrabou A, Yousefi H, Lo PY, Angers S, Sargent EH, Kelley SO. Identification of druggable regulators of cell secretion via a kinome-wide screen and high-throughput immunomagnetic cell sorting. Nat Biomed Eng 2024; 8:263-277. [PMID: 38012306 DOI: 10.1038/s41551-023-01135-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/16/2023] [Indexed: 11/29/2023]
Abstract
The identification of genetic regulators of cell secretions is challenging because it requires the sorting of a large number of cells according to their secretion patterns. Here we report the development and applicability of a high-throughput microfluidic method for the analysis of the secretion levels of large populations of immune cells. The method is linked with a kinome-wide loss-of-function CRISPR screen, immunomagnetically sorting the cells according to their secretion levels, and the sequencing of their genomes to identify key genetic modifiers of cell secretion. We used the method, which we validated against flow cytometry for cytokines secreted from primary mouse CD4+ (cluster of differentiation 4-positive) T cells, to discover a subgroup of highly co-expressed kinase-coding genes that regulate interferon-gamma secretion by these cells. We validated the function of the kinases identified using RNA interference, CRISPR knockouts and kinase inhibitors and confirmed the druggability of selected kinases via the administration of a kinase inhibitor in an animal model of colitis. The technique may facilitate the discovery of regulatory mechanisms for immune-cell activation and of therapeutic targets for autoimmune diseases.
Collapse
Affiliation(s)
- Mahmoud Labib
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth, UK
- Department of Chemistry, Northwestern University, Evanston, IL, USA
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - Yunhye Kim
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Sichun Lin
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Abdalla Abdrabou
- Robert H. Laurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Hanie Yousefi
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Pei-Ying Lo
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - Stéphane Angers
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - Edward H Sargent
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
- Department of Electrical & Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Shana O Kelley
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada.
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA.
- Robert H. Laurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada.
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA.
| |
Collapse
|
10
|
Zhang Y, Zhang F, Song Y, Shen X, Bu F, Su D, Luo C, Ge L, Deng S, Wu Z, Zhang Z, Duan P, Li N, Min L, Zhang S, Wang S. Interfacial Polymerization Produced Magnetic Particles with Nano-Filopodia for Highly Accurate Liquid Biopsy in the PSA Gray Zone. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303821. [PMID: 37643459 DOI: 10.1002/adma.202303821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Magnetic particles are leading separation materials for biological purification and detection. Existing magnetic particles, which almost rely on molecule-level interactions, however, often encounter bottlenecks in highly efficient cell-level separation due to the underestimate of surface structure effects. Here, immune cell-inspired magnetic particles with nano-filopodia (NFMPs) produced by interfacial polymerization for highly efficient capture of circulating tumor cells (CTCs) and further accurate clinical diagnosis of prostate cancer are reported . The unprecedented construction of nano-filopodia on polymer-based magnetic particles is achieved by introducing electrostatic interactions in emulsion interfacial polymerization. Due to the unique nano-filopodia, the NFMPs allow remarkably enhanced CTCs capture efficiency (86.5% ± 2.8%) compared with smooth magnetic particles (SMPs, 35.7% ± 5.7%). Under the assistance of machine learning by combining with prostate-specific antigen (PSA) and free to total PSA (F/T-PSA), the NFMPs strategy demonstrates high sensitivity (100%), high specificity (93.3%), and a high area under the curve (AUC) value (98.1%) for clinical diagnosis of prostate cancer in the PSA gray zone. The NFMPs are anticipated as an efficient platform for CTCs-based liquid biopsy toward early cancer diagnosis and prognosis evaluation.
Collapse
Affiliation(s)
- Yue Zhang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Fan Zhang
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Yongyang Song
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xinyi Shen
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Fanqin Bu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Dandan Su
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Chen Luo
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Liyuan Ge
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Shaohui Deng
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Zonglong Wu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Zhanyi Zhang
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Peichen Duan
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Nan Li
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Shudong Zhang
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P. R. China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
11
|
Chen K, Duong BTV, Ahmed SU, Dhavarasa P, Wang Z, Labib M, Flynn C, Xu J, Zhang YY, Wang H, Yang X, Das J, Zargartalebi H, Ma Y, Kelley SO. A magneto-activated nanoscale cytometry platform for molecular profiling of small extracellular vesicles. Nat Commun 2023; 14:5576. [PMID: 37696888 PMCID: PMC10495366 DOI: 10.1038/s41467-023-41285-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023] Open
Abstract
Exosomal PD-L1 (exoPD-L1) has recently received significant attention as a biomarker predicting immunotherapeutic responses involving the PD1/PD-L1 pathway. However, current technologies for exosomal analysis rely primarily on bulk measurements that do not consider the heterogeneity found within exosomal subpopulations. Here, we present a nanoscale cytometry platform NanoEPIC, enabling phenotypic sorting and exoPD-L1 profiling from blood plasma. We highlight the efficacy of NanoEPIC in monitoring anti-PD-1 immunotherapy through the interrogation of exoPD-L1. NanoEPIC generates signature exoPD-L1 patterns in responders and non-responders. In mice treated with PD1-targeted immunotherapy, exoPD-L1 is correlated with tumor growth, PD-L1 burden in tumors, and the immune suppression of CD8+ tumor-infiltrating lymphocytes. Small extracellular vesicles (sEVs) with different PD-L1 expression levels display distinctive inhibitory effects on CD8 + T cells. NanoEPIC offers robust, high-throughput profiling of exosomal markers, enabling sEV subpopulation analysis. This platform holds the potential for enhanced cancer screening, personalized treatment, and therapeutic response monitoring.
Collapse
Affiliation(s)
- Kangfu Chen
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Bill T V Duong
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Sharif U Ahmed
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | | | - Zongjie Wang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Mahmoud Labib
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, Northwestern University, Evanston, IL, USA
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth, UK
| | - Connor Flynn
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Jingya Xu
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Yi Y Zhang
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Hansen Wang
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Xiaolong Yang
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Jagotamoy Das
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Hossein Zargartalebi
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Yuan Ma
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Shana O Kelley
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.
- Department of Chemistry, University of Toronto, Toronto, ON, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA.
| |
Collapse
|
12
|
Wang Z, Ahmed S, Labib M, Wang H, Wu L, Bavaghar-Zaeimi F, Shokri N, Blanco S, Karim S, Czarnecka-Kujawa K, Sargent EH, McGray AJR, de Perrot M, Kelley SO. Isolation of tumour-reactive lymphocytes from peripheral blood via microfluidic immunomagnetic cell sorting. Nat Biomed Eng 2023; 7:1188-1203. [PMID: 37037966 DOI: 10.1038/s41551-023-01023-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 03/11/2023] [Indexed: 04/12/2023]
Abstract
The clinical use of tumour-infiltrating lymphocytes for the treatment of solid tumours is hindered by the need to obtain large and fresh tumour fractions, which is often not feasible in patients with unresectable tumours or recurrent metastases. Here we show that circulating tumour-reactive lymphocytes (cTRLs) can be isolated from peripheral blood at high yield and purity via microfluidic immunomagnetic cell sorting, allowing for comprehensive downstream analyses of these rare cells. We observed that CD103 is strongly expressed by the isolated cTRLs, and that in mice with subcutaneous tumours, tumour-infiltrating lymphocytes isolated from the tumours and rapidly expanded CD8+CD103+ cTRLs isolated from blood are comparably potent and respond similarly to immune checkpoint blockade. We also show that CD8+CD103+ cTRLs isolated from the peripheral blood of patients and co-cultured with tumour cells dissociated from their resected tumours resulted in the enrichment of interferon-γ-secreting cell populations with T-cell-receptor clonotypes substantially overlapping those of the patients' tumour-infiltrating lymphocytes. Therapeutically potent cTRLs isolated from peripheral blood may advance the clinical development of adoptive cell therapies.
Collapse
Affiliation(s)
- Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Sharif Ahmed
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - Mahmoud Labib
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, IL, USA
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth, UK
| | - Hansen Wang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Fatemeh Bavaghar-Zaeimi
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Nastaran Shokri
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Soraly Blanco
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Saraf Karim
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Kasia Czarnecka-Kujawa
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Edward H Sargent
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| | - A J Robert McGray
- Department of Immunology, Division of Translational Immuno-Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shana O Kelley
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, IL, USA.
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Department of Biochemistry, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Wang Z. Assessing Tumorigenicity in Stem Cell-Derived Therapeutic Products: A Critical Step in Safeguarding Regenerative Medicine. Bioengineering (Basel) 2023; 10:857. [PMID: 37508884 PMCID: PMC10376867 DOI: 10.3390/bioengineering10070857] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/08/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Stem cells hold promise in regenerative medicine due to their ability to proliferate and differentiate into various cell types. However, their self-renewal and multipotency also raise concerns about their tumorigenicity during and post-therapy. Indeed, multiple studies have reported the presence of stem cell-derived tumors in animal models and clinical administrations. Therefore, the assessment of tumorigenicity is crucial in evaluating the safety of stem cell-derived therapeutic products. Ideally, the assessment needs to be performed rapidly, sensitively, cost-effectively, and scalable. This article reviews various approaches for assessing tumorigenicity, including animal models, soft agar culture, PCR, flow cytometry, and microfluidics. Each method has its advantages and limitations. The selection of the assay depends on the specific needs of the study and the stage of development of the stem cell-derived therapeutic product. Combining multiple assays may provide a more comprehensive evaluation of tumorigenicity. Future developments should focus on the optimization and standardization of microfluidics-based methods, as well as the integration of multiple assays into a single platform for efficient and comprehensive evaluation of tumorigenicity.
Collapse
Affiliation(s)
- Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Chan Zuckerberg Biohub Chicago, Chicago, IL 60607, USA
| |
Collapse
|
14
|
Chang D, Wang Z, Flynn CD, Mahmud A, Labib M, Wang H, Geraili A, Li X, Zhang J, Sargent EH, Kelley SO. A high-dimensional microfluidic approach for selection of aptamers with programmable binding affinities. Nat Chem 2023; 15:773-780. [PMID: 37277648 DOI: 10.1038/s41557-023-01207-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 04/17/2023] [Indexed: 06/07/2023]
Abstract
Aptamers are being applied as affinity reagents in analytical applications owing to their high stability, compact size and amenability to chemical modification. Generating aptamers with different binding affinities is desirable, but systematic evolution of ligands by exponential enrichment (SELEX), the standard for aptamer generation, is unable to quantitatively produce aptamers with desired binding affinities and requires multiple rounds of selection to eliminate false-positive hits. Here we introduce Pro-SELEX, an approach for the rapid discovery of aptamers with precisely defined binding affinities that combines efficient particle display, high-performance microfluidic sorting and high-content bioinformatics. Using the Pro-SELEX workflow, we were able to investigate the binding performance of individual aptamer candidates under different selective pressures in a single round of selection. Using human myeloperoxidase as a target, we demonstrate that aptamers with dissociation constants spanning a 20-fold range of affinities can be identified within one round of Pro-SELEX.
Collapse
Affiliation(s)
- Dingran Chang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - Connor D Flynn
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, IL, USA
| | - Alam Mahmud
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Mahmoud Labib
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, IL, USA
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth, UK
| | - Hansen Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Armin Geraili
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Xiangling Li
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Jiaqi Zhang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Edward H Sargent
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, IL, USA
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Electrical and Computer Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA
| | - Shana O Kelley
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Department of Biochemistry, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA.
| |
Collapse
|
15
|
Qian L, Lin X, Gao X, Khan RU, Liao JY, Du S, Ge J, Zeng S, Yao SQ. The Dawn of a New Era: Targeting the "Undruggables" with Antibody-Based Therapeutics. Chem Rev 2023. [PMID: 37186942 DOI: 10.1021/acs.chemrev.2c00915] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The high selectivity and affinity of antibodies toward their antigens have made them a highly valuable tool in disease therapy, diagnosis, and basic research. A plethora of chemical and genetic approaches have been devised to make antibodies accessible to more "undruggable" targets and equipped with new functions of illustrating or regulating biological processes more precisely. In this Review, in addition to introducing how naked antibodies and various antibody conjugates (such as antibody-drug conjugates, antibody-oligonucleotide conjugates, antibody-enzyme conjugates, etc.) work in therapeutic applications, special attention has been paid to how chemistry tools have helped to optimize the therapeutic outcome (i.e., with enhanced efficacy and reduced side effects) or facilitate the multifunctionalization of antibodies, with a focus on emerging fields such as targeted protein degradation, real-time live-cell imaging, catalytic labeling or decaging with spatiotemporal control as well as the engagement of antibodies inside cells. With advances in modern chemistry and biotechnology, well-designed antibodies and their derivatives via size miniaturization or multifunctionalization together with efficient delivery systems have emerged, which have gradually improved our understanding of important biological processes and paved the way to pursue novel targets for potential treatments of various diseases.
Collapse
Affiliation(s)
- Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuefen Lin
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xue Gao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shubo Du
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544
| |
Collapse
|
16
|
Xiang Y, Zhang H, Lu H, Wei B, Su C, Qin X, Fang M, Li X, Yang F. Bioorthogonal Microbubbles with Antifouling Nanofilm for Instant and Suspended Enrichment of Circulating Tumor Cells. ACS NANO 2023; 17:9633-9646. [PMID: 37144647 DOI: 10.1021/acsnano.3c03194] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Integrating clinical rare cell enrichment, culture, and single-cell phenotypic profiling is currently hampered by the lack of competent technologies, which typically suffer from weak cell-interface collision affinity, strong nonspecific adsorption, and the potential uptake. Here, we report cells-on-a-bubble, a bioinspired, self-powered bioorthogonal microbubble (click bubble) that leverages a clickable antifouling nanointerface and a DNA-assembled sucker-like polyvalent cell surface, to enable instant and suspended isolation of circulating tumor cells (CTCs) within minutes. Using this biomimetic engineering strategy, click bubbles achieve a capture efficiency of up to 98%, improved by 20% at 15 times faster over their monovalent counterparts. Further, the buoyancy-activated bubble facilitates self-separation, 3D suspension culture, and in situ phenotyping of the captured single cancer cells. By using a multiantibody design, this fast, affordable micromotor-like click bubble enables suspended enrichment of CTCs in a cohort (n = 42) across three cancer types and treatment response evaluation, signifying its great potential to enable single-cell analysis and 3D organoid culture.
Collapse
Affiliation(s)
- Yuanhang Xiang
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Hui Zhang
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Hao Lu
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Binqi Wei
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Cuiyun Su
- Department of Respiratory Oncology, Department of Clinical Laboratory, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xiaojie Qin
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Min Fang
- Department of Respiratory Oncology, Department of Clinical Laboratory, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xinchun Li
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Fan Yang
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
17
|
Cheng Y, Zhang S, Qin L, Zhao J, Song H, Yuan Y, Sun J, Tian F, Liu C. Poly(ethylene oxide) Concentration Gradient-Based Microfluidic Isolation of Circulating Tumor Cells. Anal Chem 2023; 95:3468-3475. [PMID: 36725367 DOI: 10.1021/acs.analchem.2c05257] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Circulating tumor cells (CTCs) have emerged as promising circulating biomarkers for non-invasive cancer diagnosis and management. Isolation and detection of CTCs in clinical samples are challenging due to the extreme rarity and high heterogeneity of CTCs. Here, we describe a poly(ethylene oxide) (PEO) concentration gradient-based microfluidic method for rapid, label-free, highly efficient isolation of CTCs directly from whole blood samples. Stable concentration gradients of PEO were formed within the microchannel by co-injecting the side fluid (blood sample spiked with 0.025% PEO) and center fluid (0.075% PEO solution). The competition between the elastic lift force and the inertial lift force enabled size-based separation of large CTCs and small blood cells based on their distinct migration patterns. The microfluidic device could process 1 mL of blood sample in 30 min, with a separation efficiency of >90% and an enrichment ratio of >700 for tumor cells. The isolated CTCs from blood samples were enumerated by immunofluorescence staining, allowing for discrimination of breast cancer patients from healthy donors with an accuracy of 84.2%. The concentration gradient-based microfluidic separation provides a powerful tool for label-free isolation of CTCs for a wide range of clinical applications.
Collapse
Affiliation(s)
- Yangchang Cheng
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shaohua Zhang
- Department of Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Lili Qin
- Department of Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Junxiang Zhao
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hua Song
- Department of Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Yang Yuan
- Department of Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Jiashu Sun
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fei Tian
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chao Liu
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Liu Y, Vieira RMS, Mao L. Simultaneous and Multimodal Antigen-Binding Profiling and Isolation of Rare Cells via Quantitative Ferrohydrodynamic Cell Separation. ACS NANO 2023; 17:94-110. [PMID: 36541668 DOI: 10.1021/acsnano.2c04542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Simultaneous cell profiling and isolation based on cellular antigen-binding capacity plays an important role in understanding and treating diseases. However, fluorescence-activated cell sorting (FACS) and magnetic-activated cell sorting (MACS) are not able to meet this need, due to their requirement for a large quantity of target cells and the limitation stemming from bimodal separation. Here we report a microfluidic method, termed quantitative ferrohydrodynamic cell separation (qFCS), that achieved multimodal rare cell sorting and simultaneous antigen profiling at a ∼30,000 cell min-1 throughput with a 96.49% recovery rate and a 98.72% purity of recovered cells. qFCS profiles and sorts cells via cellular magnetic content of the magnetically labeled cells, which correlates to cellular antigen-binding capacity. By integrating cellular magnetophoresis and diamagnetophoresis in biocompatible ferrofluids, we demonstrate that the resulting qFCS device can accurately profile and isolate rare cells even when present at ∼1:50,000 target to background cells frequency. We show that the qFCS device could accurately profile and isolate T lymphocytes based on a low-expression CD154 antigen and allow on-device analysis of cells after processing. This method could address the need for simultaneous and multimodal rare cell isolation and profiling in disease diagnostics, prognostics, and treatment.
Collapse
Affiliation(s)
- Yang Liu
- Department of Chemistry, The University of Georgia, Athens, Georgia30602, United States
| | - Rafaela Maggioni Simoes Vieira
- J. Crayton Pruitt Family Department of Biomedical Engineering, The University of Florida, Gainesville, Florida32611, United States
| | - Leidong Mao
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, Georgia30602, United States
| |
Collapse
|
19
|
Abdrabou A, Duong BTV, Chen K, Atwal RS, Labib M, Lin S, Angers S, Kelley SO. nuPRISM: Microfluidic Genome-Wide Phenotypic Screening Platform for Cellular Nuclei. ACS CENTRAL SCIENCE 2022; 8:1618-1626. [PMID: 36589880 PMCID: PMC9801500 DOI: 10.1021/acscentsci.2c00836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Indexed: 06/17/2023]
Abstract
Genome-wide loss-of-function screens are critical tools to identify novel genetic regulators of intracellular proteins. However, studying the changes in the organelle-specific expression profile of intracellular proteins can be challenging due to protein localization differences across the whole cell, hindering context-dependent protein expression and activity analyses. Here, we describe nuPRISM, a microfluidics chip specifically designed for large-scale isolated nuclei sorting. The new device enables rapid genome-wide loss-of-function phenotypic CRISPR-Cas9 screens directed at intranuclear targets. We deployed this technology to identify novel genetic regulators of β-catenin nuclear accumulation, a phenotypic hallmark of APC-mutated colorectal cancer. nuPRISM expands our ability to capture aberrant nuclear morphological and functional traits associated with distinctive signal transduction and subcellular localization-driven functional processes with substantial resolution and high throughput.
Collapse
Affiliation(s)
- Abdalla
M. Abdrabou
- Department
of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Bill T. V. Duong
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Kangfu Chen
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Randy Singh Atwal
- Department
of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Mahmoud Labib
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60611, United States
| | - Sichun Lin
- Terrence
Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Stephane Angers
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
- Department
of Biochemistry, Faculty of Medicine, University
of Toronto, Toronto, Ontario M5S 1A8, Canada
- Terrence
Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Shana O. Kelley
- Department
of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60611, United States
- Department
of Biomedical Engineering, Northwestern
University, Evanston, Illinois 60611, United States
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| |
Collapse
|
20
|
Farheen J, Hosmane NS, Zhao R, Zhao Q, Iqbal MZ, Kong X. Nanomaterial-assisted CRISPR gene-engineering - A hallmark for triple-negative breast cancer therapeutics advancement. Mater Today Bio 2022; 16:100450. [PMID: 36267139 PMCID: PMC9576993 DOI: 10.1016/j.mtbio.2022.100450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/16/2022] [Accepted: 10/02/2022] [Indexed: 11/05/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most violent class of tumor and accounts for 20-24% of total breast carcinoma, in which frequently rare mutation occurs in high frequency. The poor prognosis, recurrence, and metastasis in the brain, heart, liver and lungs decline the lifespan of patients by about 21 months, emphasizing the need for advanced treatment. Recently, the adaptive immunity mechanism of archaea and bacteria, called clustered regularly interspaced short palindromic repeats (CRISPR) combined with nanotechnology, has been utilized as a potent gene manipulating tool with an extensive clinical application in cancer genomics due to its easeful usage and cost-effectiveness. However, CRISPR/Cas are arguably the efficient technology that can be made efficient via organic material-assisted approaches. Despite the efficacy of the CRISPR/Cas@nano complex, problems regarding successful delivery, biodegradability, and toxicity remain to render its medical implications. Therefore, this review is different in focus from past reviews by (i) detailing all possible genetic mechanisms of TNBC occurrence; (ii) available treatments and gene therapies for TNBC; (iii) overview of the delivery system and utilization of CRISPR-nano complex in TNBC, and (iv) recent advances and related toxicity of CRISPR-nano complex towards clinical trials for TNBC.
Collapse
Affiliation(s)
- Jabeen Farheen
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Zhejiang-Mauritius Joint Research Centre for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Narayan S. Hosmane
- Department of Chemistry & Biochemistry, Northern Illinois University, DeKalb, IL, 60115, USA
| | - Ruibo Zhao
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Zhejiang-Mauritius Joint Research Centre for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Department of Materials, Imperial College London, London, SW7 2AZ, UK
| | - Qingwei Zhao
- Research Center for Clinical Pharmacy & Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - M. Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Zhejiang-Mauritius Joint Research Centre for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Zhejiang-Mauritius Joint Research Centre for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| |
Collapse
|
21
|
Descamps L, Garcia J, Barthelemy D, Laurenceau E, Payen L, Le Roy D, Deman AL. MagPure chip: an immunomagnetic-based microfluidic device for high purification of circulating tumor cells from liquid biopsies. LAB ON A CHIP 2022; 22:4151-4166. [PMID: 36148526 DOI: 10.1039/d2lc00443g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The isolation of circulating tumor cells (CTCs) directly from blood, as a liquid biopsy, could lead to a paradigm shift in cancer clinical care by providing an earlier diagnosis, a more accurate prognosis, and personalized treatment. Nevertheless, CTC-specific challenges, including their rarity and heterogeneity, have hampered the wider use of CTCs in clinical studies. Microfluidic-based isolation technologies have emerged as promising tools to circumvent these limitations but still fail to meet the constraints of high purity and short processing time required to ensure compatibility with clinical follow-up. In this study, we developed an immunomagnetic-based microfluidic device, the MagPure chip, to achieve the negative selection of CTCs through the depletion of white blood cells (WBCs) and provide highly purified samples for subsequent analysis. We demonstrate that the MagPure chip depletes all magnetically labeled WBCs (85% of WBCs were successfully labeled) and ensures a CTC recovery rate of 81%. In addition, we show its compatibility with conventional biological studies, including 2D and 3D cell culture, as well as phenotypic and genotypic analyses. Finally, we successfully implemented a two-step separation workflow for whole blood processing by combining a size-based pre-enrichment system (ClearCell FX1®) with the MagPure chip as a subsequent purification step. The total workflow led to high throughput (7.5 mL blood in less than 4 h) and high purity (947 WBCs per mL remaining, 99.99% depletion rate), thus enabling us to quantify CTC heterogeneity in size and tumor marker expression level. This tumor-marker-free liquid biopsy workflow could be used in a clinical context to assess phenotype aggressiveness and the prognosis rate.
Collapse
Affiliation(s)
- Lucie Descamps
- Institut des Nanotechnologies de Lyon, INL UMR5270, Université Claude Bernard Lyon 1, Villeurbanne, France.
| | - Jessica Garcia
- Laboratoire de Biochimie et Biologie Moléculaire, CICLY UR3738, Groupe Hospitalier Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - David Barthelemy
- Laboratoire de Biochimie et Biologie Moléculaire, CICLY UR3738, Groupe Hospitalier Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Emmanuelle Laurenceau
- Institut des Nanotechnologies de Lyon, INL UMR5270, Ecole Centrale de Lyon, Ecully, France
| | - Léa Payen
- Laboratoire de Biochimie et Biologie Moléculaire, CICLY UR3738, Groupe Hospitalier Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Damien Le Roy
- Institut Lumière Matière, ILM UMR5306, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Anne-Laure Deman
- Institut des Nanotechnologies de Lyon, INL UMR5270, Université Claude Bernard Lyon 1, Villeurbanne, France.
| |
Collapse
|
22
|
Xia F, Ma Y, Chen K, Duong B, Ahmed S, Atwal R, Philpott D, Ketela T, Pantea J, Lin S, Angers S, Kelley SO. Genome-wide in vivo screen of circulating tumor cells identifies SLIT2 as a regulator of metastasis. SCIENCE ADVANCES 2022; 8:eabo7792. [PMID: 36054348 PMCID: PMC10848953 DOI: 10.1126/sciadv.abo7792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Circulating tumor cells (CTCs) break free from primary tumors and travel through the circulation system to seed metastatic tumors, which are the major cause of death from cancer. The identification of the major genetic factors that enhance production and persistence of CTCs in the bloodstream at a whole genome level would enable more comprehensive molecular mechanisms of metastasis to be elucidated and the identification of novel therapeutic targets, but this remains a challenging task due to the heterogeneity and extreme rarity of CTCs. Here, we describe an in vivo genome-wide CRISPR knockout screen using CTCs directly isolated from a mouse xenograft. This screen elucidated SLIT2-a gene encoding a secreted protein acting as a cellular migration cue-as the most significantly represented gene knockout in the CTC population. SLIT2 knockout cells are highly metastatic with hypermigratory and mesenchymal phenotype, resulting in enhanced cancer progression in xenograft models.
Collapse
Affiliation(s)
- Fan Xia
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
| | - Yuan Ma
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
- Key Laboratory of Tribology, Tsinghua University, Beijing 100084, P.R. China
| | - Kangfu Chen
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
| | - Bill Duong
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Sharif Ahmed
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
| | - Randy Atwal
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - David Philpott
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Troy Ketela
- Princess Margret Genomics Centre, University Health Network, Toronto, Ontario, Canada
| | - Jennifer Pantea
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
| | - Sichun Lin
- Donnelly Centre for Cellular & Biomolecular Research, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Stephane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
- Donnelly Centre for Cellular & Biomolecular Research, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, ON, Canada
| | - Shana O. Kelley
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
- Department of Chemistry, Northwestern University, Evanston, IL, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| |
Collapse
|
23
|
Li S, Ferrer-Ruiz A, Dai J, Ramos-Soriano J, Du X, Zhu M, Zhang W, Wang Y, Herranz MÁ, Jing L, Zhang Z, Li H, Xia F, Martín N. A pH-independent electrochemical aptamer-based biosensor supports quantitative, real-time measurement in vivo. Chem Sci 2022; 13:8813-8820. [PMID: 35975161 PMCID: PMC9350589 DOI: 10.1039/d2sc02021a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/23/2022] [Indexed: 12/25/2022] Open
Abstract
The development of biosensors capable of achieving accurate and precise molecular measurements in the living body in pH-variable biological environments (e.g. subcellular organelles, biological fluids and organs) plays a significant role in personalized medicine. Because they recapitulate the conformation-linked signaling mechanisms, electrochemical aptamer-based (E-AB) sensors are good candidates to fill this role. However, this class of sensors suffers from a lack of a stable and pH-independent redox reporter to support their utility under pH-variable conditions. Here, in response, we demonstrate the efficiency of an electron donor π-extended tetrathiafulvalene (exTTF) as an excellent candidate (due to its good electrochemical stability and no proton participation in its redox reaction) of pH-independent redox reporters. Its use has allowed improvement of E-AB sensing performance in biological fluids under different pH conditions, achieving high-frequency, real-time molecular measurements in biological samples both in vitro and in the bladders of living rats.
Collapse
Affiliation(s)
- Shaoguang Li
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Andrés Ferrer-Ruiz
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid 28040 Madrid Spain
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430074 China
| | - Javier Ramos-Soriano
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid 28040 Madrid Spain
| | - Xuewei Du
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Man Zhu
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Wanxue Zhang
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Yuanyuan Wang
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - M Ángeles Herranz
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid 28040 Madrid Spain
| | - Le Jing
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Zishuo Zhang
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Hui Li
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Fan Xia
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Nazario Martín
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid 28040 Madrid Spain
- IMDEA-Nanoscience C/Faraday, 9, Campus de Cantoblanco 28049 Madrid Spain
| |
Collapse
|
24
|
Varillas JI, Chen K, Dopico P, Zhang J, George TJ, Fan ZH. Comparison of sample preparation methods for rare cell isolation in microfluidic devices. CAN J CHEM 2022; 100:512-519. [PMID: 36338875 PMCID: PMC9635407 DOI: 10.1139/cjc-2021-0229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2024]
Abstract
The analysis of circulating tumor cells (CTCs) is important for cancer diagnosis and prognosis. Microfluidics has been employed for CTC analysis due to its scaling advantages and high performance. However, pre-analytical methods for CTC sample preparation are often combined with microfluidic platforms because a large sample volume is required to detect extremely rare CTCs. Among pre-analytical methods, Ficoll-Paque™, OncoQuick™, and RosetteSep™ are commonly used to separate cells of interest. To compare their performance, we spiked L3.6pl pancreatic cancer cells into healthy blood samples and then employed each technique to prepare blood samples, followed by using a microfluidic platform to capture and detect L3.6pl cells. We found these three methods have similar performance, though the slight edge of RosetteSep™ over Ficoll-Paque™ is statistically significant. We also studied the effects of the tumor cell concentrations on the performance of the frequently used Ficoll-Paque™ method. Furthermore, we examined the repeatability and variability of each pre-analytical technique and the microfluidics-enabled detection. This study will provide researchers and clinicians with comparative data that can influence the choice of sample preparation method, help estimate CTC loss in each pre-analytical method, and correlate the results of clinical studies that employ different techniques.
Collapse
Affiliation(s)
- Jose I Varillas
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL 32611, USA
| | - Kangfu Chen
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA
| | - Pablo Dopico
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA
| | - Jinling Zhang
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA
| | - Thomas J George
- Department of Medicine, University of Florida, P.O. Box 100278, Gainesville, FL 32610, USA
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL 32611, USA; Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL 32611, USA
| |
Collapse
|
25
|
Wang Z, Wang H, Lin S, Ahmed S, Angers S, Sargent EH, Kelley SO. Nanoparticle Amplification Labeling for High-Performance Magnetic Cell Sorting. NANO LETTERS 2022; 22:4774-4783. [PMID: 35639489 DOI: 10.1021/acs.nanolett.2c01018] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Magnetic cell sorting is an enabling tool for the isolation of specific cellular subpopulations for downstream applications and requires the cells to be labeled by a sufficient number of magnetic nanoparticles to leverage magnetophoresis for efficient separation. This requirement makes it challenging to target weakly expressed biomarkers. Here, we developed a new approach that selectively and efficiently amplifies the magnetic labeling on cells through sequentially connected antibodies and nanoparticles delivered to the surface or interior of the cell. Using this approach, we achieved amplification up to 100-fold for surface and intracellular markers. We also demonstrated the utility of this assay for enabling high-performance magnetic cell sorting when it is applied to the analysis of rare tumor cells for cancer diagnosis and the purification of transfected CAR T cells for immunotherapy. The data presented demonstrate a useful tool for the stratification of rare cell subpopulations.
Collapse
Affiliation(s)
- Zongjie Wang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, Canada
| | - Hansen Wang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
| | - Sichun Lin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto M5S 3E1, Canada
| | - Sharif Ahmed
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
| | - Stephane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto M5S 3E1, Canada
| | - Edward H Sargent
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada
| | - Shana O Kelley
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, Canada
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto M5S 1A8, Canada
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
26
|
Wang L, Abdulla A, Wang A, Warden AR, Ahmad KZ, Xin Y, Ding X. Sickle-like Inertial Microfluidic System for Online Rare Cell Separation and Tandem Label-Free Quantitative Proteomics (Orcs-Proteomics). Anal Chem 2022; 94:6026-6035. [PMID: 35380437 DOI: 10.1021/acs.analchem.2c00679] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Label-free proteomics with trace clinical samples provides a wealth of actionable insights for personalized medicine. Clinically acquired primary cells, such as circulating tumor cells (CTCs), are usually with low abundance that is prohibitive for conventional label-free proteomics analysis. Here, we present a sickle-like inertial microfluidic system for online rare cell separation and tandem label-free proteomics (namely, Orcs-proteomics). Orcs-proteomics adopts a buffer system with 0.1% N-dodecyl β-d-maltoside (DDM), 1 mM Tris (2-carboxyethyl) phosphine (TCEP), and 2 mM 2-chloroacetamide (CAA) for cell lysis and reductive alkylation. We demonstrate the application of Orcs-proteomics with 293T cells and manage to identify 913, 1563, 2271, and 2770 protein groups with 4, 13, 68, and 119 cells, respectively. We then spike MCF7 cells with white blood cells (WBCs) to simulate the patient's blood sample. Orcs-proteomics identifies more than 2000 protein groups with an average of 61 MCF7 cells. We further recruit two advanced breast cancer patients and collect 5 and 7 CTCs from each patient through minimally invasive blood drawing. Orcs-proteomics manages to identify 973 and 1135 protein groups for each patient. Therefore, Orcs-proteomics empowers rare cells simultaneously to be separated and counted for proteomics and provides technical support for personalized treatment decision making with rare primary patient samples.
Collapse
Affiliation(s)
- Liping Wang
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Aynur Abdulla
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Aiting Wang
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Antony R Warden
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Khan Zara Ahmad
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yufang Xin
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xianting Ding
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
27
|
Descamps L, Le Roy D, Deman AL. Microfluidic-Based Technologies for CTC Isolation: A Review of 10 Years of Intense Efforts towards Liquid Biopsy. Int J Mol Sci 2022; 23:ijms23041981. [PMID: 35216097 PMCID: PMC8875744 DOI: 10.3390/ijms23041981] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/01/2023] Open
Abstract
The selection of circulating tumor cells (CTCs) directly from blood as a real-time liquid biopsy has received increasing attention over the past ten years, and further analysis of these cells may greatly aid in both research and clinical applications. CTC analysis could advance understandings of metastatic cascade, tumor evolution, and patient heterogeneity, as well as drug resistance. Until now, the rarity and heterogeneity of CTCs have been technical challenges to their wider use in clinical studies, but microfluidic-based isolation technologies have emerged as promising tools to address these limitations. This review provides a detailed overview of latest and leading microfluidic devices implemented for CTC isolation. In particular, this study details must-have device performances and highlights the tradeoff between recovery and purity. Finally, the review gives a report of CTC potential clinical applications that can be conducted after CTC isolation. Widespread microfluidic devices, which aim to support liquid-biopsy-based applications, will represent a paradigm shift for cancer clinical care in the near future.
Collapse
Affiliation(s)
- Lucie Descamps
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INSA Lyon, Ecole Centrale de Lyon, CPE Lyon, INL, UMR5270, 69622 Villeurbanne, France;
| | - Damien Le Roy
- Institut Lumière Matière ILM-UMR 5306, CNRS, Université Lyon 1, 69622 Villeurbanne, France;
| | - Anne-Laure Deman
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INSA Lyon, Ecole Centrale de Lyon, CPE Lyon, INL, UMR5270, 69622 Villeurbanne, France;
- Correspondence:
| |
Collapse
|
28
|
Wang Z, Ahmed S, Labib M, Wang H, Hu X, Wei J, Yao Y, Moffat J, Sargent EH, Kelley SO. Efficient recovery of potent tumour-infiltrating lymphocytes through quantitative immunomagnetic cell sorting. Nat Biomed Eng 2022; 6:108-117. [PMID: 35087171 DOI: 10.1038/s41551-021-00820-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022]
Abstract
Adoptive cell therapies require the recovery and expansion of highly potent tumour-infiltrating lymphocytes (TILs). However, TILs in tumours are rare and difficult to isolate efficiently, which hinders the optimization of therapeutic potency and dose. Here we show that a configurable microfluidic device can efficiently recover potent TILs from solid tumours by leveraging specific expression levels of target cell-surface markers. The device, which is sandwiched by permanent magnets, balances magnetic forces and fluidic drag forces to sort cells labelled with magnetic nanoparticles conjugated with antibodies for the target markers. Compared with conventional cell sorting, immunomagnetic cell sorting recovered up to 30-fold higher numbers of TILs, and the higher levels and diversity of the recovered TILs accelerated TIL expansion and enhanced their therapeutic potency. Immunomagnetic cell sorting also allowed us to identify and isolate potent TIL subpopulations, in particular TILs with moderate levels of CD39 (a marker of T-cell reactivity to tumours and T-cell exhaustion), which we found are tumour-specific, self-renewable and essential for the long-term success of adoptive cell therapies.
Collapse
Affiliation(s)
- Zongjie Wang
- The Edward S. Rogers Sr Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada.,Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Sharif Ahmed
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Mahmoud Labib
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Hansen Wang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Xiyue Hu
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Jiarun Wei
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Yuxi Yao
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Jason Moffat
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Edward H Sargent
- The Edward S. Rogers Sr Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Shana O Kelley
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. .,Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. .,Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada. .,Department of Chemistry, Northwestern University, Evanston, IL, USA. .,Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
29
|
The Origins and the Current Applications of Microfluidics-Based Magnetic Cell Separation Technologies. MAGNETOCHEMISTRY 2022. [DOI: 10.3390/magnetochemistry8010010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The magnetic separation of cells based on certain traits has a wide range of applications in microbiology, immunology, oncology, and hematology. Compared to bulk separation, performing magnetophoresis at micro scale presents advantages such as precise control of the environment, larger magnetic gradients in miniaturized dimensions, operational simplicity, system portability, high-throughput analysis, and lower costs. Since the first integration of magnetophoresis and microfluidics, many different approaches have been proposed to magnetically separate cells from suspensions at the micro scale. This review paper aims to provide an overview of the origins of microfluidic devices for magnetic cell separation and the recent technologies and applications grouped by the targeted cell types. For each application, exemplary experimental methods and results are discussed.
Collapse
|
30
|
Abdulla A, Zhang T, Li S, Guo W, Warden AR, Xin Y, Maboyi N, Lou J, Xie H, Ding X. Integrated microfluidic single-cell immunoblotting chip enables high-throughput isolation, enrichment and direct protein analysis of circulating tumor cells. MICROSYSTEMS & NANOENGINEERING 2022; 8:13. [PMID: 35136652 PMCID: PMC8807661 DOI: 10.1038/s41378-021-00342-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/07/2021] [Accepted: 11/11/2021] [Indexed: 05/14/2023]
Abstract
Effective capture and analysis of a single circulating tumor cell (CTC) is instrumental for early diagnosis and personalized therapy of tumors. However, due to their extremely low abundance and susceptibility to interference from other cells, high-throughput isolation, enrichment, and single-cell-level functional protein analysis of CTCs within one integrated system remains a major challenge. Herein, we present an integrated multifunctional microfluidic system for highly efficient and label-free CTC isolation, CTC enrichment, and single-cell immunoblotting (ieSCI). The ieSCI-chip is a multilayer microfluidic system that combines an inertia force-based cell sorter with a membrane filter for label-free CTC separation and enrichment and a thin layer of a photoactive polyacrylamide gel with microwell arrays at the bottom of the chamber for single-cell immunoblotting. The ieSCI-chip successfully identified a subgroup of apoptosis-negative (Bax-negative) cells, which traditional bulk analysis did not detect, from cisplatin-treated cells. Furthermore, we demonstrated the clinical application of the ieSCI-chip with blood samples from breast cancer patients for personalized CTC epithelial-to-mesenchymal transition (EMT) analysis. The expression level of a tumor cell marker (EpCAM) can be directly determined in isolated CTCs at the single-cell level, and the therapeutic response to anticancer drugs can be simultaneously monitored. Therefore, the ieSCI-chip provides a promising clinical translational tool for clinical drug response monitoring and personalized regimen development.
Collapse
Grants
- National Natural Science Foundation of China (National Science Foundation of China)
- Project by National Innovation Special Zone, Project 2017SHZDZX01, 17DZ2203400, and 18430760500 by Shanghai Municipal Science and Technology, Project G20180101 by Shanghai Agriculture Applied Technology Development Program, Project ZXWF082101 by Shanghai Municipal Education Commission, Project 2017ZX10203205-006-002 by National Key Research and Development Program of China, Project 19X190020154, ZH2018ZDA01, YG2016QN24 and YG2016MS60 by Shanghai Jiao Tong University Biomedical Interdisciplinary Program, Project ZH2018QNA54 and ZH2018QNA49 by the Medical-Engineering Cross Foundation of Shanghai Jiao Tong University, Project 2019CXJQ03 by Innovation Group Project of Shanghai Municipal Health Comission, Project 19MC1910800 by Shanghai Clinical Medical Research Center, Project SD0820016 by the third batch of industrialization project of Innovation Incubation Fund of Nantong and Shanghai Jiao Tong University, Project SL2020MS026 by the Oceanic Interdisciplinary Program of Shanghai Jiao Tong University, Project Agri-X20200101 by Shanghai Jiao Tong University, SJTU Global Strategic Partnership Fund (2020 SJTU-HUJI).
Collapse
Affiliation(s)
- Aynur Abdulla
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Ting Zhang
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Shanhe Li
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Wenke Guo
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Antony R. Warden
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Yufang Xin
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Nokuzola Maboyi
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Jiatao Lou
- Shanghai General Hospital, Shanghai Jiao Tong University, No.85 Wujing Road, Shanghai, 200080 China
| | - Haiyang Xie
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Xianting Ding
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| |
Collapse
|
31
|
Zhen W, An S, Wang S, Hu W, Li Y, Jiang X, Li J. Precise Subcellular Organelle Targeting for Boosting Endogenous-Stimuli-Mediated Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2101572. [PMID: 34611949 DOI: 10.1002/adma.202101572] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/15/2021] [Indexed: 06/13/2023]
Abstract
Though numerous external-stimuli-triggered tumor therapies, including phototherapy, radiotherapy, and sonodynamic therapy have made great progress in cancer therapy, the low penetration depth of the laser, safety concerns of radiation, the therapeutic resistance, and the spatio-temporal constraints of the specific equipment restrict their convenient clinical applications. What is more, the inherent physiological barriers of the tumor microenvironment (TME), including hypoxia, heterogeneity, and high expression of antioxidant molecules also restrict the efficiency of tumor therapy. As a result, the development of nanoplatforms responsive to endogenous stimuli (such as glucose, acidic pH, cellular redox events, and etc.) has attracted great attention for starvation therapy, ion therapy, prodrug-mediated chemotherapy, or enzyme-catalyzed therapy. In addition, nanomedicines can be modified by some targeted units for precisely locating in subcellular organelles and boosting the destroying of tumor tissue, decreasing the dosage of nanoagents, reducing side effects, and enhancing the therapeutic efficiency. Herein, the properties of the TME, the advantages of endogenous stimuli, and the principles of subcellular-organelle-targeted strategies will be emphasized. Some necessary considerations for the exploitation of precision medicine and clinical translation of multifunctional nanomedicines in the future are also pointed out.
Collapse
Affiliation(s)
- Wenyao Zhen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Shangjie An
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Shuqi Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Wenxue Hu
- Shenyang University of Chemical Technology, Shenyang, Liaoning, 110142, China
| | - Yujie Li
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Xiue Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Jinghong Li
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
32
|
Nakagawa Y, Ohnuki S, Kondo N, Itto-Nakama K, Ghanegolmohammadi F, Isozaki A, Ohya Y, Goda K. Are droplets really suitable for single-cell analysis? A case study on yeast in droplets. LAB ON A CHIP 2021; 21:3793-3803. [PMID: 34581379 DOI: 10.1039/d1lc00469g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Single-cell analysis has become one of the main cornerstones of biotechnology, inspiring the advent of various microfluidic compartments for cell cultivation such as microwells, microtrappers, microcapillaries, and droplets. A fundamental assumption for using such microfluidic compartments is that unintended stress or harm to cells derived from the microenvironments is insignificant, which is a crucial condition for carrying out unbiased single-cell studies. Despite the significance of this assumption, simple viability or growth tests have overwhelmingly been the assay of choice for evaluating culture conditions while empirical studies on the sub-lethal effect on cellular functions have been insufficient in many cases. In this work, we assessed the effect of culturing cells in droplets on the cellular function using yeast morphology as an indicator. Quantitative morphological analysis using CalMorph, an image-analysis program, demonstrated that cells cultured in flasks, large droplets, and small droplets significantly differed morphologically. From these differences, we identified that the cell cycle was delayed in droplets during the G1 phase and during the process of bud growth likely due to the checkpoint mechanism and impaired mitochondrial function, respectively. Furthermore, comparing small and large droplets, cells cultured in large droplets were morphologically more similar to those cultured in a flask, highlighting the advantage of increasing the droplet size. These results highlight a potential source of bias in cell analysis using droplets and reinforce the significance of assessing culture conditions of microfluidic cultivation methods for specific study cases.
Collapse
Affiliation(s)
- Yuta Nakagawa
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Shinsuke Ohnuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Naoko Kondo
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Kaori Itto-Nakama
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Farzan Ghanegolmohammadi
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Akihiro Isozaki
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8654, Japan.
| | - Keisuke Goda
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, 420 Westwood Plaza, California 90095, USA
- Institute of Technological Sciences, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
33
|
Zhang B, Wang C, Du Y, Paxton R, He X. A 'smart' aptamer-functionalized continuous label-free cell catch-transport-release system. J Mater Chem B 2021; 9:7196-7204. [PMID: 34291267 DOI: 10.1039/d1tb00739d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Label-free cell sorting devices are of great significance for biomedical research and clinical therapeutics. However, current platforms for label-free cell sorting cannot achieve continuity and selectivity simultaneously, resulting in complex steps and limited reliability. Here, an immunoaffinity-based cell catch-transport-release thermo-chemo-mechanical coupling hydrogel (iCatch) device is reported. It contains a temperature-responsive hydrogel that can generate spatial movement synergically with the reversible binding of affinity handle modified. The functionalized hydrogel is embedded inside a biphasic microfluidic platform to enable cell transportation between the flows. The cell sorting capability and biocompatibility of the iCatch device were validated with CCRF-CEM cells as a proof-of-concept, and CCRF-CEM-specific aptamers with thermo-responsive affinity as well as a hydrogel with temperature-dependent volume were employed accordingly. A cell catching efficiency of ∼40% and a recovery rate of ∼70% were achieved. The iCatch device provides a high-throughput (∼900 cells mm-1 s-1) platform for cell sorting and is ultimately valuable for downstream biomedical applications.
Collapse
Affiliation(s)
- Bozhen Zhang
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Canran Wang
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Yingjie Du
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Rebecca Paxton
- University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA
| | - Ximin He
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
34
|
Sinton D, Kelley SO. AbCellera's success is unprecedented: what have we learned? LAB ON A CHIP 2021; 21:2330-2332. [PMID: 34095928 DOI: 10.1039/d1lc00155h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The search for antibody therapeutic candidates is a timely and important challenge well-suited to lab on a chip approaches. Vancouver-based AbCellera Biologics Inc. developed a microfluidic antibody screening platform, ancillary technologies, and a service-based drug discovery business model that has proved a tremendous success. We take the opportunity here to reflect on what enabled this success. We consider the common lab on a chip motivations that were part of their success, and those that were not.
Collapse
Affiliation(s)
- David Sinton
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, Ontario M5S3G8, Canada.
| | - Shana O Kelley
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Labib M, Kelley SO. Circulating tumor cell profiling for precision oncology. Mol Oncol 2021; 15:1622-1646. [PMID: 33448107 PMCID: PMC8169448 DOI: 10.1002/1878-0261.12901] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/19/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Analysis of circulating tumor cells (CTCs) collected from patient's blood offers a broad range of opportunities in the field of precision oncology. With new advances in profiling technology, it is now possible to demonstrate an association between the molecular profiles of CTCs and tumor response to therapy. In this Review, we discuss mechanisms of tumor resistance to therapy and their link to phenotypic and genotypic properties of CTCs. We summarize key technologies used to isolate and analyze CTCs and discuss recent clinical studies that examined CTCs for genomic and proteomic predictors of responsiveness to therapy. We also point out current limitations that still hamper the implementation of CTCs into clinical practice. We finally reflect on how these shortcomings can be addressed with the likely contribution of multiparametric approaches and advanced data analytics.
Collapse
Affiliation(s)
- Mahmoud Labib
- Department of Pharmaceutical SciencesUniversity of TorontoCanada
| | - Shana O. Kelley
- Department of Pharmaceutical SciencesUniversity of TorontoCanada
- Institute for Biomaterials and Biomedical EngineeringUniversity of TorontoCanada
- Department of BiochemistryUniversity of TorontoCanada
- Department of ChemistryUniversity of TorontoCanada
| |
Collapse
|
36
|
Szlachcic WJ, Ziojla N, Kizewska DK, Kempa M, Borowiak M. Endocrine Pancreas Development and Dysfunction Through the Lens of Single-Cell RNA-Sequencing. Front Cell Dev Biol 2021; 9:629212. [PMID: 33996792 PMCID: PMC8116659 DOI: 10.3389/fcell.2021.629212] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/06/2021] [Indexed: 12/16/2022] Open
Abstract
A chronic inability to maintain blood glucose homeostasis leads to diabetes, which can damage multiple organs. The pancreatic islets regulate blood glucose levels through the coordinated action of islet cell-secreted hormones, with the insulin released by β-cells playing a crucial role in this process. Diabetes is caused by insufficient insulin secretion due to β-cell loss, or a pancreatic dysfunction. The restoration of a functional β-cell mass might, therefore, offer a cure. To this end, major efforts are underway to generate human β-cells de novo, in vitro, or in vivo. The efficient generation of functional β-cells requires a comprehensive knowledge of pancreas development, including the mechanisms driving cell fate decisions or endocrine cell maturation. Rapid progress in single-cell RNA sequencing (scRNA-Seq) technologies has brought a new dimension to pancreas development research. These methods can capture the transcriptomes of thousands of individual cells, including rare cell types, subtypes, and transient states. With such massive datasets, it is possible to infer the developmental trajectories of cell transitions and gene regulatory pathways. Here, we summarize recent advances in our understanding of endocrine pancreas development and function from scRNA-Seq studies on developing and adult pancreas and human endocrine differentiation models. We also discuss recent scRNA-Seq findings for the pathological pancreas in diabetes, and their implications for better treatment.
Collapse
Affiliation(s)
- Wojciech J. Szlachcic
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Natalia Ziojla
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Dorota K. Kizewska
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Marcelina Kempa
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Malgorzata Borowiak
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
37
|
Jiang R, Agrawal S, Aghaamoo M, Parajuli R, Agrawal A, Lee AP. Rapid isolation of circulating cancer associated fibroblasts by acoustic microstreaming for assessing metastatic propensity of breast cancer patients. LAB ON A CHIP 2021; 21:875-887. [PMID: 33351008 DOI: 10.1039/d0lc00969e] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
We demonstrate a label free and high-throughput microbubble-based acoustic microstreaming technique to isolate rare circulating cells such as circulating cancer associated fibroblasts (cCAFs) in addition to circulating tumor cells (CTCs) and immune cells (i.e. leukocytes) from clinically diagnosed patients with a capture efficiency of 94% while preserving cell functional integrity within 8 minutes. The microfluidic device is self-pumping and was optimized to increase flow rate and achieve near perfect capturing of rare cells enabled by having a trapping capacity above the acoustic vortex saturation concentration threshold. Our approach enables rapid isolation of CTCs, cCAFs and their associated clusters from blood samples of cancer patients at different stages. By examining the combined role of cCAFs and CTCs in early cancer onset and metastasis progression, the device accurately diagnoses both cancer and the metastatic propensity of breast cancer patients. This was confirmed by flow cytometry where we observed that metastatic breast cancer blood samples had significantly higher percentage of exhausted CD8+ T cells expressing programmed cell death protein 1 (PD1), higher number of CD4+ T regulatory cells and T helper cells. We show for the first time that our lateral cavity acoustic transducers (LCATs)-based approach can thus be developed into a metastatic propensity assay for clinical usage by elucidating cancer immunological responses and the complex relationships between CTCs and its companion tumor microenvironment.
Collapse
Affiliation(s)
- Ruoyu Jiang
- Biomedical Engineering, University of California, Irvine, CA 92697, USA.
| | - Sudhanshu Agrawal
- Department of Medicine, Division of Basic and Clinical Immunology, University of California, Irvine, CA 92697, USA
| | - Mohammad Aghaamoo
- Biomedical Engineering, University of California, Irvine, CA 92697, USA.
| | - Ritesh Parajuli
- Department of Medicine, Division of Hematology Oncology, University of California, Irvine, CA 92697, USA
| | - Anshu Agrawal
- Department of Medicine, Division of Basic and Clinical Immunology, University of California, Irvine, CA 92697, USA
| | - Abraham P Lee
- Biomedical Engineering, University of California, Irvine, CA 92697, USA. and Mechanical and Aerospace Engineering, University of California, Irvine, CA 92697, USA
| |
Collapse
|
38
|
Chen H, Li Y, Zhang Z, Wang S. Immunomagnetic separation of circulating tumor cells with microfluidic chips and their clinical applications. BIOMICROFLUIDICS 2020; 14:041502. [PMID: 32849973 PMCID: PMC7440929 DOI: 10.1063/5.0005373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/04/2020] [Indexed: 06/11/2023]
Abstract
Circulating tumor cells (CTCs) are tumor cells detached from the original lesion and getting into the blood and lymphatic circulation systems. They potentially establish new tumors in remote areas, namely, metastasis. Isolation of CTCs and following biological molecular analysis facilitate investigating cancer and coming out treatment. Since CTCs carry important information on the primary tumor, they are vital in exploring the mechanism of cancer, metastasis, and diagnosis. However, CTCs are very difficult to separate due to their extreme heterogeneity and rarity in blood. Recently, advanced technologies, such as nanosurfaces, quantum dots, and Raman spectroscopy, have been integrated with microfluidic chips. These achievements enable the next generation isolation technologies and subsequent biological analysis of CTCs. In this review, we summarize CTCs' separation with microfluidic chips based on the principle of immunomagnetic isolation of CTCs. Fundamental insights, clinical applications, and potential future directions are discussed.
Collapse
Affiliation(s)
- Hongmei Chen
- School of Mathematics and Physics of Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Yong Li
- School of Mathematics and Physics of Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Zhifeng Zhang
- Department of Engineering Science and Mechanics, The Pennsylvania State University, State College, Pennsylvania 16802, USA
| | - Shuangshou Wang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Maanshan 243002, China
| |
Collapse
|