1
|
Wang J, Xu J, Liu X, Li X, Xu Z. A microfluidic chip incorporating magnetic sorting and invasive separation for isolation, culture and telomerase analysis of circulating tumor cells. Talanta 2025; 285:127316. [PMID: 39644673 DOI: 10.1016/j.talanta.2024.127316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/28/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
Circulating tumor cells (CTCs) are a crucial indicator of cancer metastasis, and are vital for early diagnosis, disease monitoring, and treatment response evaluation. However, their extremely low concentration and the complexities of isolation techniques pose a significant challenge in capturing and analyzing CTCs. In this study, we developed a novel microfluidic system that integrates magnetic capture and invasive screening onto a single microfluidic chip. By attaching positively charged magnetic nanoparticles to negatively charged CTCs, the magnetic separation of CTCs within the chip effectively eliminates interference from blood cells. A total of 2 mL blood sample can be processed within 3 min, achieving an impressive tumor capture efficiency of 84 %. Using the chip, we also successfully achieved long-term culture of CTCs, and identified CTCs with high activity and invasive potential in blood samples from 11 patients with colorectal cancer. Finally, we analyzed telomerase activity in cultured CTCs on the microfluidic chip. Significantly higher invasive potential and telomerase activity were observed in CTCs from the malignant tumor group compared to the benign group (P < 0.01), highlighting their increased aggressiveness. This study offers a novel approach for efficient CTCs isolation, culture, and telomerase analysis, clarifying the crucial role of telomerase in tumor metastasis and providing profound insights for future research on telomerase-targeted tumor metastasis.
Collapse
Affiliation(s)
- Jie Wang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Jiali Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Xiaopeng Liu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Xin Li
- Department of Anesthesiology, Liaoning Cancer Hospital and Institute, Shenyang, 110042, PR China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China.
| |
Collapse
|
2
|
Zuo J, Fang Y, Wang R, Liang S. High-throughput solutions in tumor organoids: from culture to drug screening. Stem Cells 2025; 43:sxae070. [PMID: 39460616 PMCID: PMC11811636 DOI: 10.1093/stmcls/sxae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Tumor organoids have emerged as an ideal in vitro model for patient-derived tissues, as they recapitulate the characteristics of the source tumor tissue to a certain extent, offering the potential for personalized tumor therapy and demonstrating significant promise in pharmaceutical research and development. However, establishing and applying this model involves multiple labor-intensive and time-consuming experimental steps and lacks standardized protocols and uniform identification criteria. Thus, high-throughput solutions are essential for the widespread adoption of tumor organoid models. This review provides a comprehensive overview of current high-throughput solutions across the entire workflow of tumor organoids, from sampling and culture to drug screening. Furthermore, we explore various technologies that can control and optimize single-cell preparation, organoid culture, and drug screening with the ultimate goal of ensuring the automation and high efficiency of the culture system and identifying more effective tumor therapeutics.
Collapse
Affiliation(s)
- Jianing Zuo
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan, Dalian 116001, Liaoning, China
| | - Yanhua Fang
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan, Dalian 116001, Liaoning, China
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Ruoyu Wang
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan, Dalian 116001, Liaoning, China
| | - Shanshan Liang
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan, Dalian 116001, Liaoning, China
| |
Collapse
|
3
|
Huang D, Zhao Y, Cao C, Zhao J. Efficient Particle Manipulation Using Contraction-Expansion Microchannels Embedded with Hook-Shaped Arrays. MICROMACHINES 2025; 16:83. [PMID: 39858738 PMCID: PMC11767759 DOI: 10.3390/mi16010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025]
Abstract
Inertial microfluidics, as an efficient method for the manipulation of micro-/nanoparticles, has garnered significant attention due to its advantages of high throughput, structural simplicity, no need for external fields, and sheathless operation. Common structures include straight channels, contraction-expansion array (CEA) channels, spiral channels, and serpentine channels. In this study, we developed a CEA channel embedded with hook-shaped microstructures to modify the characteristics of vortices. Through experimental studies, we investigated the particles' migration mechanisms within the proposed structure. The findings indicated that, in comparison to conventional rectangular microstructures, the particles within the hook-shaped microstructured CEA channels experienced a more pronounced influence from inertial lift forces. Moreover, the magnitude of the second flow within the novel configuration was directly proportional to the channel width, the length of the expansion segment, and the embedding depth of the microstructure. The innovative structure was subsequently employed for particle trapping, focusing, and separation. The experimental outcomes revealed focusing efficiency of up to 99.1% and sorting efficiency of up to 97%. This research holds the potential to enhance the foundational theory of Dean flows and broaden the application spectrum of inertial contraction-expansion microfluidic chips.
Collapse
Affiliation(s)
- Di Huang
- School of Mechanical and Electrical Engineering, China University of Mining and Technology, Xuzhou 221116, China; (Y.Z.); (C.C.)
| | | | | | - Jiyun Zhao
- School of Mechanical and Electrical Engineering, China University of Mining and Technology, Xuzhou 221116, China; (Y.Z.); (C.C.)
| |
Collapse
|
4
|
Cai J, Chen B, He M, Yuan G, Hu B. An Integrated Inertial-Magnetophoresis Microfluidic Chip Online-Coupled with ICP-MS for Rapid Separation and Precise Detection of Circulating Tumor Cells. Anal Chem 2024; 96:14222-14229. [PMID: 39159467 DOI: 10.1021/acs.analchem.4c02876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Circulating tumor cells (CTCs) are recognized as promising targets for liquid biopsy, which play an important role in early diagnosis and efficacy monitoring of cancer. However, due to the extreme scarcity of CTCs and partial size overlap between CTCs and white blood cells (WBCs), the separation and detection of CTCs from blood remain a big challenge. To address this issue, we fabricated a microfluidic chip by integrating a passive contraction-expansion array (CEA) inertial sorting zone and an active magnetophoresis zone with the trapezoidal groove and online coupled it with inductively coupled plasma mass spectrometry (ICP-MS) for rapid separation and precise detection of MCF-7 cells (as a model CTC) in blood samples. In the integrated microfluidic chip, most of the small-sized WBCs can be rapidly removed in the circular CEA inertial sorter, while the rest of the magnetically labeled WBCs can be further captured in the trapezoidal groove under the magnetic field. As a result, the rapid separation of MCF-7 cells from blood samples was achieved with an average recovery of 91.6% at a sample flow rate of 200 μL min-1. The developed online integrated inertial-magnetophoresis microfluidic chip-ICP-MS system has been applied for the detection of CTCs in real clinical blood samples with a fast analysis speed (5 min per 1 mL blood). CTCs were detected in all 24 blood samples from patients with different types of cancer, exhibiting excellent application potential in clinical diagnosis.
Collapse
Affiliation(s)
- Jing Cai
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Beibei Chen
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Man He
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Guolin Yuan
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Bin Hu
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
5
|
Qiao Z, Teng X, Liu A, Yang W. Novel Isolating Approaches to Circulating Tumor Cell Enrichment Based on Microfluidics: A Review. MICROMACHINES 2024; 15:706. [PMID: 38930676 PMCID: PMC11206030 DOI: 10.3390/mi15060706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Circulating tumor cells (CTCs), derived from the primary tumor and carrying genetic information, contribute significantly to the process of tumor metastasis. The analysis and detection of CTCs can be used to assess the prognosis and treatment response in patients with tumors, as well as to help study the metastatic mechanisms of tumors and the development of new drugs. Since CTCs are very rare in the blood, it is a challenging problem to enrich CTCs efficiently. In this paper, we provide a comprehensive overview of microfluidics-based enrichment devices for CTCs in recent years. We explore in detail the methods of enrichment based on the physical or biological properties of CTCs; among them, physical properties cover factors such as size, density, and dielectric properties, while biological properties are mainly related to tumor-specific markers on the surface of CTCs. In addition, we provide an in-depth description of the methods for enrichment of single CTCs and illustrate the importance of single CTCs for performing tumor analyses. Future research will focus on aspects such as improving the separation efficiency, reducing costs, and increasing the detection sensitivity and accuracy.
Collapse
Affiliation(s)
- Zezheng Qiao
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China; (Z.Q.); (X.T.)
| | - Xiangyu Teng
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China; (Z.Q.); (X.T.)
| | - Anqin Liu
- School of Mechanical and Electrical Engineering, Yantai Institute of Technology, Yantai 264005, China
| | - Wenguang Yang
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China; (Z.Q.); (X.T.)
| |
Collapse
|
6
|
Nian M, Chen B, He M, Hu B. A Cascaded Phase-Transfer Microfluidic Chip with Magnetic Probe for High-Activity Sorting, Purification, Release, and Detection of Circulating Tumor Cells. Anal Chem 2024; 96:766-774. [PMID: 38158582 DOI: 10.1021/acs.analchem.3c03971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Microfluidic chips have emerged as a promising tool for sorting and enriching circulating tumor cells (CTCs) in blood, while the efficacy and purity of CTC sorting greatly depend on chip design. Herein, a novel cascaded phase-transfer microfluidic chip was developed for high-efficiency sorting, purification, release, and detection of MCF-7 cells (as a model CTC) in blood samples. MCF-7 cells were specifically captured by EpCAM aptamer-modified magnetic beads and then introduced into the designed cascaded phase-transfer microfluidic chip that consisted of three functional regions (sorting, purification, and release zone). In the sorting zone, the MCF-7 cells moved toward the inner wall of the channel and entered the purification zone for primary separation from white blood cells; in the purification zone, the MCF-7 cells were transferred to the phosphate-buffered saline flow under the interaction of Dean forces and central magnetic force, achieving high purification of MCF-7 cells from blood samples; in the release zone, MCF-7 cells were further transferred into the nuclease solution and fixed in groove by the strong magnetic force and hydrodynamic force, and the continuously flowing nuclease solution cleaved the aptamer on the trapped MCF-7 cells, causing gentle release of MCF-7 cells for subsequent inductively coupled plasma mass spectrometry (ICP-MS) detection or further cultivation. By measurement of the endogenous element Zn in the cells using ICP-MS for cell counting, an average cell recovery of 84% for MCF-7 cells was obtained in spiked blood samples. The developed method was applied in the analysis of real blood samples from healthy people and breast cancer patients, and CTCs were successfully detected in all tested patient samples (16/16). Additionally, the removal of the magnetic probes on the cell surface significantly improved cell viability up to 99.3%. Therefore, the developed cascaded phase-transfer microfluidic chip ICP-MS system possessed high integration for CTCs analysis with high cell viability, cell recovery, and purity, showing great advantages in early clinical cancer diagnosis.
Collapse
Affiliation(s)
- Miaoxiang Nian
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Beibei Chen
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Man He
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Bin Hu
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
7
|
Seyfoori A, Seyyed Ebrahimi SA, Samandari M, Samiei E, Stefanek E, Garnis C, Akbari M. Microfluidic-Assisted CTC Isolation and In Situ Monitoring Using Smart Magnetic Microgels. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205320. [PMID: 36720798 DOI: 10.1002/smll.202205320] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/16/2022] [Indexed: 06/18/2023]
Abstract
Capturing rare disease-associated biomarkers from body fluids can offer an early-stage diagnosis of different cancers. Circulating tumor cells (CTCs) are one of the major cancer biomarkers that provide insightful information about the cancer metastasis prognosis and disease progression. The most common clinical solutions for quantifying CTCs rely on the immunomagnetic separation of cells in whole blood. Microfluidic systems that perform magnetic particle separation have reported promising outcomes in this context, however, most of them suffer from limited efficiency due to the low magnetic force generated which is insufficient to trap cells in a defined position within microchannels. In this work, a novel method for making soft micromagnet patterns with optimized geometry and magnetic material is introduced. This technology is integrated into a bilayer microfluidic chip to localize an external magnetic field, consequently enhancing the capture efficiency (CE) of cancer cells labeled with the magnetic nano/hybrid microgels that are developed in the previous work. A combined numerical-experimental strategy is implemented to design the microfluidic device and optimize the capturing efficiency and to maximize the throughput. The proposed design enables high CE and purity of target cells and real-time time on-chip monitoring of their behavior. The strategy introduced in this paper offers a simple and low-cost yet robust opportunity for early-stage diagnosis and monitoring of cancer-associated biomarkers.
Collapse
Affiliation(s)
- Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
- Advanced Magnetic Materials Research Center, College of Engineering, University of Tehran, Tehran, Iran
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC V8P 5C2, Canada
| | | | - Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ehsan Samiei
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Evan Stefanek
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Cathie Garnis
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC V8P 5C2, Canada
- Bitechnology Center, Silesian University of Technology, Akademicka 2A, 44-100, Gliwice, Poland
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90024, USA
| |
Collapse
|
8
|
Tran Thi YV, Hoang BA, Thanh HT, Nguyen TH, Ngoc TP, Thu HB, Hoang NN, Bui TT, Duc TC, Do Quang L. Design and Numerical Study on a Microfluidic System for Circulating Tumor Cells Separation From Whole Blood Using Magnetophoresis and Dielectrophoresis Techniques. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
9
|
Xu K, Jiao X, Wang P, Chen C, Chen C. Isolation of circulating tumor cells based on magnetophoresis. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2022. [DOI: 10.1016/j.cjac.2022.100058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
10
|
Abstract
Magnetic cell separation has become a key methodology for the isolation of target cell populations from biological suspensions, covering a wide spectrum of applications from diagnosis and therapy in biomedicine to environmental applications or fundamental research in biology. There now exists a great variety of commercially available separation instruments and reagents, which has permitted rapid dissemination of the technology. However, there is still an increasing demand for new tools and protocols which provide improved selectivity, yield and sensitivity of the separation process while reducing cost and providing a faster response. This review aims to introduce basic principles of magnetic cell separation for the neophyte, while giving an overview of recent research in the field, from the development of new cell labeling strategies to the design of integrated microfluidic cell sorters and of point-of-care platforms combining cell selection, capture, and downstream detection. Finally, we focus on clinical, industrial and environmental applications where magnetic cell separation strategies are amongst the most promising techniques to address the challenges of isolating rare cells.
Collapse
|
11
|
Chelakkot C, Yang H, Shin YK. Relevance of Circulating Tumor Cells as Predictive Markers for Cancer Incidence and Relapse. Pharmaceuticals (Basel) 2022; 15:75. [PMID: 35056131 PMCID: PMC8781286 DOI: 10.3390/ph15010075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023] Open
Abstract
Shedding of cancer cells from the primary site or undetectable bone marrow region into the circulatory system, resulting in clinically overt metastasis or dissemination, is the hallmark of unfavorable invasive cancers. The shed cells remain in circulation until they extravasate to form a secondary metastatic lesion or undergo anoikis. The circulating tumor cells (CTCs) found as single cells or clusters carry a plethora of information, are acknowledged as potential biomarkers for predicting cancer prognosis and cancer progression, and are supposed to play key roles in determining tailored therapies for advanced diseases. With the advent of novel technologies that allow the precise isolation of CTCs, more and more clinical trials are focusing on the prognostic and predictive potential of CTCs. In this review, we summarize the role of CTCs as a predictive marker for cancer incidence, relapse, and response to therapy.
Collapse
Affiliation(s)
- Chaithanya Chelakkot
- Bio-MAX/N-Bio, Bio-MAX Institute, Seoul National University, Seoul 08226, Korea
- Genobio Corp., Seoul 08394, Korea
| | - Hobin Yang
- Research Institute of Pharmaceutical Science, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08226, Korea
| | - Young Kee Shin
- Bio-MAX/N-Bio, Bio-MAX Institute, Seoul National University, Seoul 08226, Korea
- Research Institute of Pharmaceutical Science, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08226, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08226, Korea
| |
Collapse
|
12
|
Burr R, Edd JF, Chirn B, Mishra A, Haber DA, Toner M, Maheswaran S. Negative-Selection Enrichment of Circulating Tumor Cells from Peripheral Blood Using the Microfluidic CTC-iChip. Methods Mol Biol 2022; 2471:309-321. [PMID: 35175606 DOI: 10.1007/978-1-0716-2193-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The ability to isolate and analyze rare circulating tumor cells (CTCs) holds the potential to increase our understanding of cancer evolution and allows monitoring of disease and therapeutic responses through a relatively non-invasive blood-based biopsy. While many methods have been described to isolate CTCs from the blood, the vast majority rely on size-based sorting or positive selection of CTCs based on surface markers, which introduces bias into the downstream product by making assumptions about these heterogenous cells. Here we describe a negative-selection protocol for enrichment of CTCs through removal of blood components including red blood cells, platelets, and white blood cells. This procedure results in a product that is amenable to downstream single-cell analytics including RNA-Seq, ATAC-Seq and DNA methylation, droplet digital PCR (ddPCR) for tumor specific transcripts, staining and extensive image analysis, and ex vivo culture of patient-derived CTCs.
Collapse
Affiliation(s)
- Risa Burr
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, MA, USA
| | - Jon F Edd
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian Chirn
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, MA, USA
| | - Avanish Mishra
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel A Haber
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, MA, USA
- Howard Hughes Medical Institute, Bethesda, MD, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mehmet Toner
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Shriners Hospital for Children, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shyamala Maheswaran
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, MA, USA.
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Li F, Xu H, Zhao Y. Magnetic particles as promising circulating tumor cell catchers assisting liquid biopsy in cancer diagnosis: A review. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
14
|
Wang C, Ma Y, Pei Z, Song F, Zhong J, Wang Y, Yan X, Dai P, Jiang Y, Qiu J, Shi M, Wu X. Sheathless acoustic based flow cell sorter for enrichment of rare cells. Cytometry A 2021; 101:311-324. [PMID: 34806837 DOI: 10.1002/cyto.a.24521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/12/2022]
Abstract
Cell enrichment is a powerful tool in many kinds of cell research, especially in applications with low abundance cell types. In this work, we developed a microfluidic fluorescence activated cell sorting device that was able to perform on-demand, low loss cell detection, and sorting. The chip utilizes three-dimensional acoustic standing waves to position all cells in the same fluid velocity regime without sheath. When the cells pass through a laser interrogation region, the scattering and fluorescent signals are detected, translated and transported to software. The target cells are then identified by gating on the plots. Short bursts of standing acoustic waves are triggered by order from PC to sort target cells within predefined gating region. For very low abundance and rare labeled lymphocytes mixed with high concentration unlabeled white blood cells (WBCs), (1-100 labeled lymphocytes are diluted in 106 WBCs in 1 ml volume fluid), the device is able to remove more than 98% WBCs and recover labeled lymphocytes with efficiency of 80%. We further demonstrated that this device worked with real clinical samples by successfully isolating fetal nucleated red blood cells (FNRBCs) in the blood samples from pregnant women with male fetus. The obtained cells were sequenced and the expressions of (sex determining region Y) SRY genes were tested to determine fetal cell proportion. In genetic analysis, the proportion of fetal cells in the final picked sample is up to 40.64%. With this ability, the device proposed could be valuable for biomedical applications involving fetal cells, circulating tumor cells, and stem cells.
Collapse
Affiliation(s)
- Ce Wang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Yuting Ma
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Zhiguo Pei
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Feifei Song
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Jinfeng Zhong
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Yao Wang
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Xintao Yan
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Pu Dai
- Department of Otolaryngology, Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, China
| | - Yi Jiang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jianping Qiu
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Mengdie Shi
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Xiaodong Wu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| |
Collapse
|
15
|
Liu Y, Zhao W, Cheng R, Harris BN, Murrow JR, Hodgson J, Egan M, Bankey A, Nikolinakos PG, Laver T, Meichner K, Mao L. Fundamentals of integrated ferrohydrodynamic cell separation in circulating tumor cell isolation. LAB ON A CHIP 2021; 21:1706-1723. [PMID: 33720269 PMCID: PMC8102387 DOI: 10.1039/d1lc00119a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Methods to separate circulating tumor cells (CTCs) from blood samples were intensively researched in order to understand the metastatic process and develop corresponding clinical assays. However current methods faced challenges that stemmed from CTCs' heterogeneity in their biological markers and physical morphologies. To this end, we developed integrated ferrohydrodynamic cell separation (iFCS), a scheme that separated CTCs independent of their surface antigen expression and physical characteristics. iFCS integrated both diamagnetophoresis of CTCs and magnetophoresis of blood cells together via a magnetic liquid medium, ferrofluid, whose magnetization could be tuned by adjusting its magnetic volume concentration. In this paper, we presented the fundamental theory of iFCS and its specific application in CTC separation. Governing equations of iFCS were developed to guide its optimization process. Three critical parameters that affected iFCS's cell separation performance were determined and validated theoretically and experimentally. These parameters included the sample flow rate, the volumetric concentration of magnetic materials in the ferrofluid, and the gradient of the magnetic flux density. We determined these optimized parameters in an iFCS device that led to a high recovery CTC separation in both spiked and clinical samples.
Collapse
Affiliation(s)
- Yang Liu
- Department of Chemistry, The University of Georgia, Athens, GA 30602, USA
| | - Wujun Zhao
- Department of Chemistry, The University of Georgia, Athens, GA 30602, USA
| | - Rui Cheng
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, GA 30602, USA.
| | - Bryana N Harris
- Department of Chemical Engineering, Auburn University, Auburn, AL 36830, USA
| | - Jonathan R Murrow
- Department of Medicine, Augusta University - The University of Georgia Medical Partnership, Athens, GA 30602, USA
| | - Jamie Hodgson
- University Cancer & Blood Center, LLC, Athens, GA 30607, USA
| | - Mary Egan
- University Cancer & Blood Center, LLC, Athens, GA 30607, USA
| | | | | | - Travis Laver
- Small Animal Medicine and Surgery, Veterinary Teaching Hospital, The University of Georgia, Athens, GA 30602, USA
| | - Kristina Meichner
- Department of Pathology, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA
| | - Leidong Mao
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
16
|
Huang D, Xiang N. Rapid and precise tumor cell separation using the combination of size-dependent inertial and size-independent magnetic methods. LAB ON A CHIP 2021; 21:1409-1417. [PMID: 33605279 DOI: 10.1039/d0lc01223h] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Circulating tumor cells (CTCs) play a significant role in cancer diagnosis and treatment monitoring. One of the major challenges in isolating and detecting rare CTCs from blood is that white blood cells (WBCs) have a size overlap with the target CTCs. To address this issue, we constructed a three-stage i-Mag device integrated with passive inertial microfluidics and active magnetophoresis, enabling rapid and precise separation of tumor cells from blood. The first-stage spiral inertial sorter was applied to rapidly remove small-sized red blood cells (RBCs), and then the second-stage serpentine inertial focuser and the third-stage magnetic sorter were used for removing the magnetically labeled WBCs size-independently, to significantly purify the captured tumor cells. Then, the separation performance of our i-Mag device was explored. The results indicated rapid and precise separation of breast cancer cells from diluted whole blood at a high separation efficiency of 93.84% and at a high purity of 51.47%. The purity of the collected tumor cells could be further improved to 93.60% when the blood dilution ratio was increased. We also successfully applied our i-Mag device for the isolation and detection of trace tumor cells. Our i-Mag device has numerous advantages, such as enabling high-throughput processing and high-precision separation, requiring easy manufacturing at a low cost, and providing tumor antigen-independent operation. We believe that the i-Mag device has great potential to act as a precise tool for separating various bioparticles.
Collapse
Affiliation(s)
- Di Huang
- School of Mechatronic Engineering, China University of Mining and Technology, Xuzhou 221116, P.R. China
| | | |
Collapse
|
17
|
Pang W, Ding S, Lin L, Wang C, Lei M, Xu J, Wang X, Qu J, Wei X, Gu B. Noninvasive and real-time monitoring of Au nanoparticle promoted cancer metastasis using in vivo flow cytometry. BIOMEDICAL OPTICS EXPRESS 2021; 12:1846-1857. [PMID: 33996202 PMCID: PMC8086442 DOI: 10.1364/boe.420123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 06/12/2023]
Abstract
Cancer is the second leading cause of mortality globally, while cancer metastasis, which accounts for about 90% of cancer-related mortality, presents an extremely poor prognosis. Thus, various nanomedicines were designed and synthesized for cancer treatment, but nanomaterials could lead to endothelial leakiness and consequently facilitate intravasation and extravasation of cancer cells to form circulating tumor cells (CTCs), which were regarded as the potential metastatic seeds, possibly accelerating cancer metastasis. Neither possible metastatic sites were observed nor rare CTCs could be measured using common methods at the early stage of cancer metastasis, it is urgent to explore new technology to dynamically monitor nanomedicine promoted cancer metastasis with high sensitivity, which would be beneficial for cancer treatment as well as design and synthesis of nanomedicine. Herein, a novel optical biopsy tool i.e. in vivo flow cytometry (IVFC) was constructed to noninvasively and real-time monitor CTCs of tumor-bearing mice treated with various concentrations of Au nanoparticles. The in vivo experimental results demonstrated the promoted CTCs were Au nanoparticles dose-dependent consistent with the in vitro results, which showed Au nanoparticles induced dose-dependent gaps in the blood vessel endothelial walls to accelerate CTCs formation, making IVFC a promising biopsy tool in fundamental, pre-clinical and clinical investigation of nanomedicine and cancer metastasis.
Collapse
Affiliation(s)
- Wen Pang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Contributed equally to this work
| | - Shihui Ding
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Contributed equally to this work
| | - Liyun Lin
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chen Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Man Lei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jiale Xu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xintong Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xunbin Wei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Biomedical Engineering Department, Peking University, Beijing 100081, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Bobo Gu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
18
|
Li F, Wang M, Cai H, He Y, Xu H, Liu Y, Zhao Y. Nondestructive capture, release, and detection of circulating tumor cells with cystamine-mediated folic acid decorated magnetic nanospheres. J Mater Chem B 2021; 8:9971-9979. [PMID: 33174893 DOI: 10.1039/d0tb01091j] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Circulating tumor cell (CTC) detection and enumeration have been considered as a noninvasive biopsy method for the diagnosis, characterization, and monitoring of various types of cancers. However, CTCs are exceptionally rare, which makes CTC detection technologically challenging. In the past few decades, much effort has been focused on highly efficient CTC capture, while the activity of CTCs has often been ignored. Here, we develop an effective method for nondestructive CTC capture, release, and detection. Folic acid (FA), as a targeting molecule, is conjugated on magnetic nanospheres through a cleavable disulfide bond-containing linker (cystamine) and a polyethylene glycol (PEG2k) linker, forming MN@Cys@PEG2k-FA nanoprobes, which can bind with folate receptor (FR) positive CTCs specifically and efficiently, leading to the capture of CTCs with an external magnetic field. When approximately 150 and 10 model CTCs were spiked in 1 mL of lysis blood, 93.1 ± 2.9% and 80.0 ± 9.7% CTCs were recovered, respectively. In total, 81.3 ± 2.6% captured CTCs can be released from MN@Cys@PEG2k-FA magnetic nanospheres by treatment with dithiothreitol. The released CTCs are easily identified from blood cells for specific detection and enumeration combined with immunofluorescence staining with a limit of detection of 10 CTC mL-1 lysed blood. Moreover, the released cells remain healthy with high viability (98.6 ± 0.78%) and can be cultured in vitro without detectable changes in morphology or behavior compared with healthy untreated cells. The high viability of the released CTCs may provide the possibility for downstream proteomics research of CTCs; therefore, cultured CTCs were collected for proteomics. As a result, 3504 proteins were identified. In conclusion, the MN@Cys@PEG2k-FA magnetic nanospheres prepared in this study may be a promising tool for early-stage cancer diagnosis and provide the possibility for downstream analysis of CTCs.
Collapse
Affiliation(s)
- Fulai Li
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China. and Department of Chemical Biology, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Minning Wang
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China. and Department of Chemical Biology, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Huahuan Cai
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China. and Department of Chemical Biology, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Yaohui He
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, P. R. China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, P. R. China
| | - Yan Liu
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China. and Department of Chemical Biology, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Yufen Zhao
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China. and Department of Chemical Biology, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China and Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang 315221, P. R. China
| |
Collapse
|
19
|
Huang D, Man J, Jiang D, Zhao J, Xiang N. Inertial microfluidics: Recent advances. Electrophoresis 2020; 41:2166-2187. [PMID: 33027533 DOI: 10.1002/elps.202000134] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/19/2020] [Accepted: 10/02/2020] [Indexed: 02/24/2024]
Abstract
Inertial microfluidics has attracted significant attentions in last decade due to its superior advantages of high throughput, label- and external field-free operation, simplicity, and low cost. A wide variety of channel geometry designs were demonstrated for focusing, concentrating, isolating, or separating of various bioparticles such as blood components, circulating tumor cells, bacteria, and microalgae. In this review, we first briefly introduce the physics of inertial migration and Dean flow for allowing the readers with diverse backgrounds to have a better understanding of the fundamental mechanisms of inertial microfluidics. Then, we present a comprehensive review of the recent advances and applications of inertial microfluidic devices according to different channel geometries ranging from straight channels, curved channels to contraction-expansion-array channels. Finally, the challenges and future perspective of inertial microfluidics are discussed. Owing to its superior benefit for particle manipulation, the inertial microfluidics will play a more important role in biology and medicine applications.
Collapse
Affiliation(s)
- Di Huang
- College of Mechanical and Electrical Engineering, China University of Mining and Technology, Xuzhou, P. R. China
- Jiangsu Province and Education Ministry Co-sponsored Collaborative Innovation Center of Intelligent Mining Equipment, China University of Mining and Technology, Xuzhou, P. R. China
| | - Jiaxiang Man
- College of Mechanical and Electrical Engineering, China University of Mining and Technology, Xuzhou, P. R. China
- Jiangsu Province and Education Ministry Co-sponsored Collaborative Innovation Center of Intelligent Mining Equipment, China University of Mining and Technology, Xuzhou, P. R. China
| | - Di Jiang
- School of Mechanical and Electronic Engineering, Nanjing Forestry University, Nanjing, P. R. China
| | - Jiyun Zhao
- College of Mechanical and Electrical Engineering, China University of Mining and Technology, Xuzhou, P. R. China
- Jiangsu Province and Education Ministry Co-sponsored Collaborative Innovation Center of Intelligent Mining Equipment, China University of Mining and Technology, Xuzhou, P. R. China
| | - Nan Xiang
- School of Mechanical Engineering, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, P. R. China
| |
Collapse
|
20
|
Brunato S, Mastrotto F, Bellato F, Garofalo M, Göddenhenrich T, Mantovani G, Alexander C, Gross S, Salmaso S, Caliceti P. Thermosensitive "Smart" Surfaces for Biorecognition Based Cell Adhesion and Controlled Detachment. Macromol Biosci 2020; 21:e2000277. [PMID: 33146950 DOI: 10.1002/mabi.202000277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/20/2020] [Indexed: 11/07/2022]
Abstract
The biorecognition-based control of attachment/detachment of MCF-7 cancer cells from polymer-coated surfaces is demonstrated. A glass surface is coated with a thermoresponsive statistical copolymer of poly(N-isopropylacrylamide-co-acrylamide) [p(NIPAm-co-Am)], which is end-capped with the Gly-Arg-Gly-Asp-Ser (GRGDS) peptide, and the hydrophilic polymer poly(ethylene glycol) (PEG). Below the lower critical solution temperature (LCST) of p(NIPAm-co-Am) (38 °C), the copolymers are in the extended conformation, allowing for accessibility of the GRGDS peptides to membrane-associated integrins thus enabling cell attachment. Above the LCST, the p(NIPAm-co-Am) polymers collapse into globular conformations, resulting in the shielding of the GRGDS peptides into the PEG brush with consequent inaccessibility to cell-surface integrins, causing cell detachment. The surface coating is carried out by a multi-step procedure that included: glass surface amination with 3-aminopropyltriethoxysilane; reaction of mPEG5kDa -N-hydroxysuccinimide (NHS) and p(NIPam-co-Am)15.1kDa -bis-NHS with the surface aminopropyl groups and conjugation of GRGDS to the carboxylic acid termini of p(NIPam-co-Am)15.1kDa -COOH. A range of spectrophotometric, surface, and microscopy assays confirmed the identity of the polymer-coated substrates. Competition studies prove that MCF-7 cancer cells are attached via peptide recognition at the coated surfaces according to the mPEG5kDa /p(NIPam-co-Am)15.1kDa -GRGDS molar ratio. These data suggest the system can be exploited to modulate cell integrin/GRGDS binding for controlled cell capture and release.
Collapse
Affiliation(s)
- Silvia Brunato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, Padova, 35131, Italy
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, Padova, 35131, Italy
| | - Federica Bellato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, Padova, 35131, Italy
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, Padova, 35131, Italy
| | - Thomas Göddenhenrich
- Institute of Physics, Giessen University, Heinrich-Buff-Ring 16, Giessen, 35392, Germany
| | - Giuseppe Mantovani
- Molecular Therapeutics and Formulation Division, School of Pharmacy, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Cameron Alexander
- Molecular Therapeutics and Formulation Division, School of Pharmacy, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Silvia Gross
- Department of Chemical Sciences, University of Padova, via F. Marzolo 1, Padova, 35131, Italy
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, Padova, 35131, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, Padova, 35131, Italy
| |
Collapse
|
21
|
Luo L, He Y. Magnetically driven microfluidics for isolation of circulating tumor cells. Cancer Med 2020; 9:4207-4231. [PMID: 32325536 PMCID: PMC7300401 DOI: 10.1002/cam4.3077] [Citation(s) in RCA: 39] [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: 11/19/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Circulating tumor cells (CTCs) largely contribute to cancer metastasis and show potential prognostic significance in cancer isolation and detection. Miniaturization has progressed significantly in the last decade which in turn enabled the development of several microfluidic systems. The microfluidic systems offer a controlled microenvironment for studies of fundamental cell biology, resulting in the rapid development of microfluidic isolation of CTCs. Due to the inherent ability of magnets to provide forces at a distance, the technology of CTCs isolation based on the magnetophoresis mechanism has become a routine methodology. This historical review aims to introduce two principles of magnetic isolation and recent techniques, facilitating research in this field and providing alternatives for researchers in their study of magnetic isolation. Researchers intend to promote effective CTC isolation and analysis as well as active development of next-generation cancer treatment. The first part of this review summarizes the primary principles based on positive and negative magnetophoretic isolation and describes the metrics for isolation performance. The second part presents a detailed overview of the factors that affect the performance of CTC magnetic isolation, including the magnetic field sources, functionalized magnetic nanoparticles, magnetic fluids, and magnetically driven microfluidic systems.
Collapse
Affiliation(s)
- Laan Luo
- School of Chemical EngineeringKunming University of Science and TechnologyKunmingChina
| | - Yongqing He
- School of Chemical EngineeringKunming University of Science and TechnologyKunmingChina
- Chongqing Key Laboratory of Micro‐Nano System and Intelligent SensingChongqing Technology and Business UniversityChongqingChina
| |
Collapse
|
22
|
Alnaimat F, Karam S, Mathew B, Mathew B. Magnetophoresis and Microfluidics: A Great Union. IEEE NANOTECHNOLOGY MAGAZINE 2020; 14:24-41. [DOI: 10.1109/mnano.2020.2966029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
23
|
A homogeneous magnetic bead-based impedance immunosensor for highly sensitive detection of Escherichia coli O157:H7. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107513] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
24
|
Continuous Microfluidic Purification of DNA Using Magnetophoresis. MICROMACHINES 2020; 11:mi11020187. [PMID: 32054004 PMCID: PMC7074667 DOI: 10.3390/mi11020187] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/13/2022]
Abstract
Automatic microfluidic purification of nucleic acid is predictable to reduce the input of original samples and improve the throughput of library preparation for sequencing. Here, we propose a novel microfluidic system using an external NdFeB magnet to isolate DNA from the polymerase chain reaction (PCR) mixture. The DNA was purified and isolated when the DNA-carrying beads transported to the interface of multi-laminar flow under the influence of magnetic field. Prior to the DNA recovery experiments, COMSOL simulations were carried out to study the relationship between trajectory of beads and magnet positions as well as fluid velocities. Afterwards, the experiments to study the influence of varying velocities and input of samples on the DNA recovery were conducted. Compared to experimental results, the relative error of the final position of beads is less than 10%. The recovery efficiency decreases with increase of input or fluid velocity, and the maximum DNA recovery efficiency is 98.4% with input of l00 ng DNA at fluid velocity of 1.373 mm/s. The results show that simulations significantly reduce the time for parameter adjustment in experiments. In addition, this platform uses a basic two-layer chip to realize automatic DNA isolation without any other liquid switch value or magnet controller.
Collapse
|
25
|
Han SI, Kim HS, Han KH, Han A. Digital quantification and selection of high-lipid-producing microalgae through a lateral dielectrophoresis-based microfluidic platform. LAB ON A CHIP 2019; 19:4128-4138. [PMID: 31755503 DOI: 10.1039/c9lc00850k] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Microalgae are promising alternatives to petroleum as renewable biofuel sources, however not sufficiently economically competitive yet. Here, a label-free lateral dielectrophoresis-based microfluidic sorting platform that can digitally quantify and separate microalgae into six outlets based on the degree of their intracellular lipid content is presented. In this microfluidic system, the degree of cellular lateral displacement is inversely proportional to the intracellular lipid level, which was successfully demonstrated using Chlamydomonas reinhardtii cells. Using this functionality, a quick digital quantification of sub-populations that contain different intracellular lipid level in a given population was achieved. In addition, the degree of lateral displacement of microalgae could be readily controlled by simply changing the applied DEP voltage, where the level of gating in the intracellular lipid-based sorting decision could be easily adjusted. This allowed for selecting only a very small percentage of a given population that showed the highest degree of intracellular lipid content. In addition, this approach was utilized through an iterative selection process on natural and chemically mutated microalgal populations, successfully resulting in enrichment of high-lipid-accumulating microalgae. In summary, the developed platform can be exploited to quickly quantify microalgae lipid distribution in a given population in real-time and label-free, as well as to enrich a cell population with high-lipid-producing cells, or to select high-lipid-accumulating microalgal variants from a microalgal library.
Collapse
Affiliation(s)
- Song-I Han
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA.
| | - Hyun Soo Kim
- Korea Institute of Machinery and Materials, Daegu Research Center for Medical Devices and Rehabilitation, Daegu, 42994, Republic of Korea
| | - Ki-Ho Han
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae, 50834, Republic of Korea
| | - Arum Han
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA. and Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
26
|
Xu M, Zhao H, Chen J, Liu W, Li E, Wang Q, Zhang L. An Integrated Microfluidic Chip and Its Clinical Application for Circulating Tumor Cell Isolation and Single-Cell Analysis. Cytometry A 2019; 97:46-53. [PMID: 31595638 DOI: 10.1002/cyto.a.23902] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 12/16/2022]
Abstract
Circulating tumor cells (CTCs) represent invasive tumor cell populations and provide a noninvasive solution to the clinical management and research of tumors. Characterization of CTCs at single-cell resolution enables the comprehensive understanding of tumor heterogeneity and may benefit the diagnosis and treatment of cancer patients. However, most efforts have been made on enumeration and detection of CTCs, while little focus has been directed to single-cell study. Herein, an integrated microfluidic platform for single-cell isolation and analysis was established. After validating this platform on lung cancer cell lines, we detected and isolated single CTCs from the peripheral blood samples of 20 cancer patients before and after one treatment cycle. Furthermore, we performed single-cell whole-exome DNA sequencing on a single CTC from the peripheral blood sample of a representative early stage lung cancer patient. Among the blood samples of 20 patients, 15 of them were positive for CTC detection (75.0% detectable rate). Single-cell analysis revealed detailed genetic variations of the CTC, while six new gene mutations were detected in both single CTC and surgical specimen. This study provides a useful tool for the isolation and analysis of single CTCs from peripheral blood samples, which not only facilitates the early diagnosis of cancers but also helps to unravel the genetic information of tumor at a single-cell level. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Mingxin Xu
- Department of Respiratory Medicine, The Second Hospital Affiliated to Dalian Medical University, Dalian, 116044, China
| | - Hui Zhao
- Department of Respiratory Medicine, The Second Hospital Affiliated to Dalian Medical University, Dalian, 116044, China
| | - Jun Chen
- Department of Oncology, The Second Hospital Affiliated to Dalian Medical University, Dalian, 116044, China
| | - Wenwen Liu
- Department of Respiratory Medicine, The Second Hospital Affiliated to Dalian Medical University, Dalian, 116044, China
| | - Encheng Li
- Department of Respiratory Medicine, The Second Hospital Affiliated to Dalian Medical University, Dalian, 116044, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital Affiliated to Dalian Medical University, Dalian, 116044, China
| | - Lichuan Zhang
- Department of Respiratory Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| |
Collapse
|
27
|
Li R, Jia F, Zhang W, Shi F, Fang Z, Zhao H, Hu Z, Wei Z. Device for whole genome sequencing single circulating tumor cells from whole blood. LAB ON A CHIP 2019; 19:3168-3178. [PMID: 31455953 DOI: 10.1039/c9lc00473d] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Whole-genome sequencing on circulating tumor cells (CTCs) at the single cell level has recently been found helpful for precision medicine, as the oncogenic profiles of single CTCs are useful for discovering oncogenic mutation heterogeneities and guiding/adjusting cancer treatment. To overcome the limits of existing methods of single CTC sequencing, in which CTC enrichment, identification and gene amplification are performed by discrete modules, this study presents a novel method in which all processing steps from blood sample collection to preparation of gene amplification products for sequencers are finished in a single microfluidic chip. This microfluidic chip comprehensively performs blood filtering, CTC enrichment, CTC identification/isolation, CTC lysis and whole genome amplification (WGA) at the single cell level. By sequencing single CTCs from clinical blood samples with pointing key driver and drug-resistance mutations, the novel microfluidic chip was validated to be capable of genetically profiling single CTCs with minimum cell loss/human labor, and more importantly, high accuracy and repeatability, which are crucial factors for promoting clinical application of single CTC sequencing.
Collapse
Affiliation(s)
- Ren Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Tavakoli H, Zhou W, Ma L, Perez S, Ibarra A, Xu F, Zhan S, Li X. Recent advances in microfluidic platforms for single-cell analysis in cancer biology, diagnosis and therapy. Trends Analyt Chem 2019; 117:13-26. [PMID: 32831435 PMCID: PMC7434086 DOI: 10.1016/j.trac.2019.05.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Understanding molecular, cellular, genetic and functional heterogeneity of tumors at the single-cell level has become a major challenge for cancer research. The microfluidic technique has emerged as an important tool that offers advantages in analyzing single-cells with the capability to integrate time-consuming and labour-intensive experimental procedures such as single-cell capture into a single microdevice at ease and in a high-throughput fashion. Single-cell manipulation and analysis can be implemented within a multi-functional microfluidic device for various applications in cancer research. Here, we present recent advances of microfluidic devices for single-cell analysis pertaining to cancer biology, diagnostics, and therapeutics. We first concisely introduce various microfluidic platforms used for single-cell analysis, followed with different microfluidic techniques for single-cell manipulation. Then, we highlight their various applications in cancer research, with an emphasis on cancer biology, diagnosis, and therapy. Current limitations and prospective trends of microfluidic single-cell analysis are discussed at the end.
Collapse
Affiliation(s)
- Hamed Tavakoli
- College of Environmental Science and Engineering, Nankai
University, Tianjin 300071, People’s Republic of China
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
| | - Wan Zhou
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
| | - Lei Ma
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
| | - Stefani Perez
- Biomedical Engineering, Border Biomedical Research Center,
Environmental Science & Engineering, University of Texas at El Paso, 500 West
University Ave, El Paso, TX 79968, USA
| | - Andrea Ibarra
- Biomedical Engineering, Border Biomedical Research Center,
Environmental Science & Engineering, University of Texas at El Paso, 500 West
University Ave, El Paso, TX 79968, USA
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center,
Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of
China
| | - Sihui Zhan
- College of Environmental Science and Engineering, Nankai
University, Tianjin 300071, People’s Republic of China
| | - XiuJun Li
- College of Environmental Science and Engineering, Nankai
University, Tianjin 300071, People’s Republic of China
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
- Biomedical Engineering, Border Biomedical Research Center,
Environmental Science & Engineering, University of Texas at El Paso, 500 West
University Ave, El Paso, TX 79968, USA
| |
Collapse
|
30
|
Raillon C, Che J, Thill S, Duchamp M, Desbiolles BXE, Millet A, Sollier E, Renaud P. Toward Microfluidic Label-Free Isolation and Enumeration of Circulating Tumor Cells from Blood Samples. Cytometry A 2019; 95:1085-1095. [PMID: 31364817 DOI: 10.1002/cyto.a.23868] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 12/18/2022]
Abstract
The isolation, analysis, and enumeration of circulating tumor cells (CTCs) from cancer patient blood samples are a paradigm shift for cancer patient diagnosis, prognosis, and treatment monitoring. Most methods used to isolate and enumerate these target cells rely on the expression of cell surface markers, which varies between patients, cancer types, tumors, and stages. Here, we propose a label-free high-throughput platform to isolate, enumerate, and size CTCs on two coupled microfluidic devices. Cancer cells were purified through a Vortex chip and subsequently flowed in-line to an impedance chip, where a pair of electrodes measured fluctuations of an applied electric field generated by cells passing through. A proof-of-concept of the coupling of those two devices was demonstrated with beads and cells. First, the impedance chip was tested as a stand-alone device: (1) with beads (mean counting error of 1.0%, sizing information clearly separated three clusters for 8, 15, and 20 um beads, respectively) as well as (2) with cancer cells (mean counting error of 3.5%). Second, the combined setup was tested with beads, then with cells in phosphate-buffered saline, and finally with cancer cells spiked in healthy blood. Experiments demonstrated that the Vortex HT chip enriched the cancer cells, which then could be counted and differentiated from smaller blood cells by the impedance chip based on size information. Further discrimination was shown with dual high-frequency measurements using electric opacity, highlighting the potential application of this combined setup for a fully integrated label-free isolation and enumeration of CTCs from cancer patient samples. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Camille Raillon
- STI-IMT-LMIS4, EPFL, 1015, Lausanne, Switzerland.,Vortex Biosciences, Inc., Pleasanton, California, 94588
| | - James Che
- Vortex Biosciences, Inc., Pleasanton, California, 94588
| | - Sandy Thill
- STI-IMT-LMIS4, EPFL, 1015, Lausanne, Switzerland
| | | | | | - Arnaud Millet
- Team Mechanobiology, Immunity and Cancer, Institute for Advanced Biosciences, INSERM U1209 CNRS UMR5309, Grenoble, France.,Grenoble Alpes University, Grenoble, France
| | | | | |
Collapse
|
31
|
Seyfoori A, Seyyed Ebrahimi SA, Samiei E, Akbari M. Multifunctional Hybrid Magnetic Microgel Synthesis for Immune-Based Isolation and Post-Isolation Culture of Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:24945-24958. [PMID: 31268286 DOI: 10.1021/acsami.9b02959] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Circulating tumor cells are of utmost importance among various biomarkers in liquid biopsies as a prognosis indicator of metastasis as well as in chemotherapeutic monitoring. This study introduces an efficient tool composed of soft nano/hybrid immune microgels for magnetic isolation of targeted tumor cells. The development process involves the in situ synthesis of magnetic nanoparticles within the three-dimensional matrix of thermoresponsive microgels. Surface modification and anti-EpCAM conjugation are adjusted by changing the temperature, and a conjugation efficiency of around 70% is achieved by using a protein G linker. Anti-EpCAM-conjugated nano/hybrid magnetic microgels are used to isolate EpCAM-expressing breast adenocarcinoma MCF-7 cells from culture media and whole blood with an efficiency of 75 and 70%, respectively. Furthermore, we demonstrate the ability of the hybrid microgels to isolate cancer cells with a purity of 65% and culture the cells post-isolation for further drug studies. The multifunctional hybrid microcarriers reported in this work can be potentially used for continuous monitoring of cancers and in personalized medicine.
Collapse
Affiliation(s)
- Amir Seyfoori
- Advanced Magnetic Materials Research Center, College of Engineering , University of Tehran , Tehran 14399-57131 , Iran
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute , ACECR , Tehran 1665659911 , Iran
| | - S A Seyyed Ebrahimi
- Advanced Magnetic Materials Research Center, College of Engineering , University of Tehran , Tehran 14399-57131 , Iran
| | | | | |
Collapse
|
32
|
Petchakup C, Tay HM, Li KHH, Hou HW. Integrated inertial-impedance cytometry for rapid label-free leukocyte isolation and profiling of neutrophil extracellular traps (NETs). LAB ON A CHIP 2019; 19:1736-1746. [PMID: 31020286 DOI: 10.1039/c9lc00250b] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Circulating leukocytes are indispensable components of the immune system, and rapid analysis of their native state or functionalities can help to unravel their pathophysiological roles and identify novel prognostic biomarkers in health and diseases. Herein we report a novel high throughput "sample-in-answer-out" integrated platform for continuous leukocyte sorting and single-cell electrical profiling in a label-free manner. The multi-staged platform enables isolation of neutrophils and monocytes from diluted or lysed blood samples directly within minutes based on Dean flow fractionation (DFF) (stage 1). Next DFF-purified leukocytes are inertially focused in serpentine channels into a single stream (stage 2) prior to impedance detection (stage 3). As a proof-of-concept for neutrophil functional characterization towards diabetes testing, we characterized the formation of neutrophil extracellular traps (NETosis) of healthy and glucose-treated neutrophils and observed significant changes in dielectric properties (size and opacity) between both groups. Interestingly, the NETosis profiles induced by calcium ionophore (CaI) and phorbol 12-myristate 13-acetate (PMA) were also electrically different, which could be attributed to the differential rates of cell enlargement and attenuated membrane permeability. Taken together, these results clearly demonstrated the potential of the developed platform for rapid (∼mins) and label-free leukocyte profiling and the use of impedance signatures as novel functional biomarkers for point-of-care testing in diabetes.
Collapse
Affiliation(s)
- Chayakorn Petchakup
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Block N3, 639798 Singapore.
| | | | | | | |
Collapse
|
33
|
Cho H, Kim J, Song H, Sohn KY, Jeon M, Han KH. Microfluidic technologies for circulating tumor cell isolation. Analyst 2019; 143:2936-2970. [PMID: 29796523 DOI: 10.1039/c7an01979c] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metastasis is the main cause of tumor-related death, and the dispersal of tumor cells through the circulatory system is a critical step in the metastatic process. Early detection and analysis of circulating tumor cells (CTCs) is therefore important for early diagnosis, prognosis, and effective treatment of cancer, enabling favorable clinical outcomes in cancer patients. Accurate and reliable methods for isolating and detecting CTCs are necessary to obtain this clinical information. Over the past two decades, microfluidic technologies have demonstrated great potential for isolating and detecting CTCs from blood. The present paper reviews current advanced microfluidic technologies for isolating CTCs based on various biological and physical principles, and discusses their fundamental advantages and drawbacks for subsequent cellular and molecular assays. Owing to significant genetic heterogeneity among CTCs, microfluidic technologies for isolating individual CTCs have recently been developed. We discuss these single-cell isolation methods, as well as approaches to overcoming the limitations of current microfluidic CTC isolation technologies. Finally, we provide an overview of future innovative microfluidic platforms.
Collapse
Affiliation(s)
- Hyungseok Cho
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae 621-749, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Advances in microfluidic techniques have prompted researchers to study the inherent heterogeneity of single cells in cell populations.
Collapse
Affiliation(s)
- Qiushi Huang
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Sifeng Mao
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Mashooq Khan
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Jin-Ming Lin
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| |
Collapse
|
35
|
Negishi R, Iwata R, Tanaka T, Kisailus D, Maeda Y, Matsunaga T, Yoshino T. Gel-based cell manipulation method for isolation and genotyping of single-adherent cells. Analyst 2019; 144:990-996. [DOI: 10.1039/c8an01456f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The simple and rapid method for isolation of single-adherent cells from a culture dish was developed and applied to genetic analysis of single-cells.
Collapse
Affiliation(s)
- Ryo Negishi
- Division of Biotechnology and Life Science
- Institute of Engineering
- Tokyo University of Agriculture and Technology
- Tokyo
- Japan
| | - Reito Iwata
- Division of Biotechnology and Life Science
- Institute of Engineering
- Tokyo University of Agriculture and Technology
- Tokyo
- Japan
| | - Tsuyoshi Tanaka
- Division of Biotechnology and Life Science
- Institute of Engineering
- Tokyo University of Agriculture and Technology
- Tokyo
- Japan
| | - David Kisailus
- Department of Chemical and Environmental Engineering
- University of California
- Riverside
- Riverside
- USA
| | - Yoshiaki Maeda
- Division of Biotechnology and Life Science
- Institute of Engineering
- Tokyo University of Agriculture and Technology
- Tokyo
- Japan
| | - Tadashi Matsunaga
- Division of Biotechnology and Life Science
- Institute of Engineering
- Tokyo University of Agriculture and Technology
- Tokyo
- Japan
| | - Tomoko Yoshino
- Division of Biotechnology and Life Science
- Institute of Engineering
- Tokyo University of Agriculture and Technology
- Tokyo
- Japan
| |
Collapse
|
36
|
Alam MK, Koomson E, Zou H, Yi C, Li CW, Xu T, Yang M. Recent advances in microfluidic technology for manipulation and analysis of biological cells (2007–2017). Anal Chim Acta 2018; 1044:29-65. [DOI: 10.1016/j.aca.2018.06.054] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/17/2022]
|
37
|
Shen Z, Wu A, Chen X. Current detection technologies for circulating tumor cells. Chem Soc Rev 2018; 46:2038-2056. [PMID: 28393954 DOI: 10.1039/c6cs00803h] [Citation(s) in RCA: 294] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Circulating tumor cells (CTCs) are cancer cells that circulate in the blood stream after being naturally shed from original or metastatic tumors, and can lead to a new fatal metastasis. CTCs have become a hotspot research field during the last decade. Detection of CTCs, as a liquid biopsy of tumors, can be used for early diagnosis of cancers, earlier evaluation of cancer recurrence and chemotherapeutic efficacy, and choice of individual sensitive anti-cancer drugs. Therefore, CTC detection is a crucial tool to fight against cancer. Herein, we classify the currently reported CTC detection technologies, introduce some representative samples for each technology, conclude the advantages and limitations, and give a future perspective including the challenges and opportunities of CTC detection.
Collapse
Affiliation(s)
- Zheyu Shen
- CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, Zhejiang 315201, China.
| | | | | |
Collapse
|
38
|
Negishi R, Takai K, Tanaka T, Matsunaga T, Yoshino T. High-Throughput Manipulation of Circulating Tumor Cells Using a Multiple Single-Cell Encapsulation System with a Digital Micromirror Device. Anal Chem 2018; 90:9734-9741. [DOI: 10.1021/acs.analchem.8b00896] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Ryo Negishi
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Kaori Takai
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Tsuyoshi Tanaka
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Tadashi Matsunaga
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Tomoko Yoshino
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| |
Collapse
|
39
|
Wang Y, Guan A, Wickramasekara S, Phillips KS. Analytical Chemistry in the Regulatory Science of Medical Devices. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2018; 11:307-327. [PMID: 29579404 DOI: 10.1146/annurev-anchem-061417-125556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In the United States, regulatory science is the science of developing new tools, standards, and approaches to assess the safety, efficacy, quality, and performance of all Food and Drug Administration-regulated products. Good regulatory science facilitates consumer access to innovative medical devices that are safe and effective throughout the Total Product Life Cycle (TPLC). Because the need to measure things is fundamental to the regulatory science of medical devices, analytical chemistry plays an important role, contributing to medical device technology in two ways: It can be an integral part of an innovative medical device (e.g., diagnostic devices), and it can be used to support medical device development throughout the TPLC. In this review, we focus on analytical chemistry as a tool for the regulatory science of medical devices. We highlight recent progress in companion diagnostics, medical devices on chips for preclinical testing, mass spectrometry for postmarket monitoring, and detection/characterization of bacterial biofilm to prevent infections.
Collapse
Affiliation(s)
- Yi Wang
- Division of Biology, Chemistry, and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Office of Medical Products and Tobacco, US Food and Drug Administration, Silver Spring, Maryland 20993, USA;
| | - Allan Guan
- Division of Biology, Chemistry, and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Office of Medical Products and Tobacco, US Food and Drug Administration, Silver Spring, Maryland 20993, USA;
| | - Samanthi Wickramasekara
- Division of Biology, Chemistry, and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Office of Medical Products and Tobacco, US Food and Drug Administration, Silver Spring, Maryland 20993, USA;
| | - K Scott Phillips
- Division of Biology, Chemistry, and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Office of Medical Products and Tobacco, US Food and Drug Administration, Silver Spring, Maryland 20993, USA;
| |
Collapse
|
40
|
Cheng D, Yu Y, Han C, Cao M, Yang G, Liu J, Chen X, Peng Z. A simple microdevice for single cell capture, array, release, and fast staining using oscillatory method. BIOMICROFLUIDICS 2018; 12:034105. [PMID: 29861808 PMCID: PMC5955720 DOI: 10.1063/1.5025677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/01/2018] [Indexed: 06/08/2023]
Abstract
Microchips that perform single cell capture, array, and identification have become powerful tools for single cell studies, which can reveal precise underlying mechanisms among bulk cell populations. However, current single cell capture and on-chip immunostaining methods consume more time and reagent than desired. To optimize this technology, we designed a novel trap structure for single cell capture, array, and release, and meanwhile an oscillatory method was used to perform rapid on-chip cell immunostaining. The trap structure array used equal distribution of lateral flow to achieve single cell array in high velocity flows and decrease the risk of clogging. A length of glass capillary with a sealed bubble was inserted into the outlet so that it could act in a manner analogous to that of a capacitor in an RC circuit. By applying one periodic air pressure to the inlet, oscillation motion was generated, which significantly enhanced the on-chip reaction efficiency. In addition, the oscillation performance could be easily regulated by changing the length of the capillary. The trapped cells could maintain their positions during oscillation; hence, they were able to be tracked in real time. Through our trap microchip, 12 μm microbeads were successfully trapped to form a microarray with a capture efficiency of ∼92.7% and 2 μm microbeads were filtered. With an optimized oscillation condition (Ppush = 0.03 MPa, f = 1 Hz, L = 3 cm), fast on-chip immunostaining was achieved with the advantages of less time (5 min) and reagent (2 μl) consumption. The effectiveness of this method was demonstrated through quantitative microbead and qualitative Caco-2 cell experiments. The device is simple, flexible, and efficient, which we believe provides a promising approach to single cell heterogeneity studies, drug screening, and clinical diagnosis.
Collapse
Affiliation(s)
- Dantong Cheng
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yang Yu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Chao Han
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Mengjia Cao
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guang Yang
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jingquan Liu
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiang Chen
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhihai Peng
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
41
|
Lee Y, Kim DM, Li Z, Kim DE, Kim SJ. Pulsatile plasma filtration and cell-free DNA amplification using a water-head-driven point-of-care testing chip. LAB ON A CHIP 2018; 18:915-922. [PMID: 29445802 DOI: 10.1039/c7lc01328k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
We demonstrate a microfiltration chip that separates blood plasma by using water-head-driven pulsatile pressures rather than any external equipment and use it for on-chip amplification of nucleic acids. The chip generates pulsatile pressures to significantly reduce filter clogging without hemolysis, and consists of an oscillator, a plasma-extraction pump, and filter units. The oscillator autonomously converts constant water-head pressure to pulsatile pressure, and the pump uses the pulsatile pressure to extract plasma through the filter. Because the pulsatile pressure can periodically clear blood cells from the filter surface, filter clogging can be effectively reduced. In this way, we achieve plasma extraction with 100% purity and 90% plasma recovery at 15% hematocrit. During a 10 min period, the volume of plasma extracted was 43 μL out of a 243 μL extraction volume at 15% hematocrit. We also studied the influence of the pore size and diameter of the filter, blood loading volume, oscillation period, and hematocrit level on the filtration performance. To demonstrate the utility of our chip for point-of-care testing (POCT) applications, we successfully implemented on-chip amplification of a nucleic acid (miDNA21) in plasma filtered from blood. We expect our chip to be useful not only for POCT applications but also for other bench-top analysis tools using blood plasma.
Collapse
Affiliation(s)
- Yonghun Lee
- Department of Mechanical Engineering, Konkuk University, Seoul, 05029, Republic of Korea.
| | | | | | | | | |
Collapse
|
42
|
Wu CP, Wu P, Zhao HF, Liu WL, Li WP. Clinical Applications of and Challenges in Single-Cell Analysis of Circulating Tumor Cells. DNA Cell Biol 2018; 37:78-89. [PMID: 29265876 DOI: 10.1089/dna.2017.3981] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Chang-peng Wu
- Department of Neurosurgery, Shenzhen Second People's Hospital, Clinical Medicine College of Anhui Medical University, Shenzhen, China
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Peng Wu
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Department of Urology, Shenzhen, China
| | - Hua-fu Zhao
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wen-lan Liu
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Wei-ping Li
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
43
|
Zhu XD, Chu J, Wang YH. Advances in Microfluidics Applied to Single Cell Operation. Biotechnol J 2018; 13. [DOI: 10.1002/biot.201700416] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 11/11/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Xu-Dong Zhu
- National Engineering Centre for Biotechnology (Shanghai); College of Biotechnology; East China University of Science and Technology; 130 Meilong Road Shanghai 200237 China
| | - Ju Chu
- National Engineering Centre for Biotechnology (Shanghai); College of Biotechnology; East China University of Science and Technology; 130 Meilong Road Shanghai 200237 China
| | - Yong-Hong Wang
- National Engineering Centre for Biotechnology (Shanghai); College of Biotechnology; East China University of Science and Technology; 130 Meilong Road Shanghai 200237 China
| |
Collapse
|
44
|
Narayanan K, Mishra S, Singh S, Pei M, Gulyas B, Padmanabhan P. Engineering Concepts in Stem Cell Research. Biotechnol J 2017; 12. [PMID: 28901712 DOI: 10.1002/biot.201700066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/07/2017] [Indexed: 12/15/2022]
Abstract
The field of regenerative medicine integrates advancements made in stem cells, molecular biology, engineering, and clinical methodologies. Stem cells serve as a fundamental ingredient for therapeutic application in regenerative medicine. Apart from stem cells, engineering concepts have equally contributed to the success of stem cell based applications in improving human health. The purpose of various engineering methodologies is to develop regenerative and preventive medicine to combat various diseases and deformities. Explosion of stem cell discoveries and their implementation in clinical setting warrants new engineering concepts and new biomaterials. Biomaterials, microfluidics, and nanotechnology are the major engineering concepts used for the implementation of stem cells in regenerative medicine. Many of these engineering technologies target the specific niche of the cell for better functional capability. Controlling the niche is the key for various developmental activities leading to organogenesis and tissue homeostasis. Biomimetic understanding not only helped to improve the design of the matrices or scaffolds by incorporating suitable biological and physical components, but also ultimately aided adoption of designs that helped these materials/devices have better function. Adoption of engineering concepts in stem cell research improved overall achievement, however, several important issues such as long-term effects with respect to systems biology needs to be addressed. Here, in this review the authors will highlight some interesting breakthroughs in stem cell biology that use engineering methodologies.
Collapse
Affiliation(s)
- Karthikeyan Narayanan
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, PO Box 9196, One Medical Center Drive, 2 Morgantown, WV 26505-9196, USA
| | - Sachin Mishra
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Satnam Singh
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, PO Box 9196, One Medical Center Drive, 2 Morgantown, WV 26505-9196, USA
| | - Balazs Gulyas
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| |
Collapse
|
45
|
Salvianti F, Pinzani P. The diagnostic potential of mutation detection from single circulating tumor cells in cancer patients. Expert Rev Mol Diagn 2017; 17:975-981. [PMID: 28931314 DOI: 10.1080/14737159.2017.1381561] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Circulating tumor cells (CTCs) have gained importance in the oncology field as biomarkers of tumor development. The most relevant observation that emerged from the recent studies on CTCs is their heterogeneity, which can be investigated by new technologies for single cell analysis. Areas covered: This review considers the most recent advances (limited to the last two years) in the mutational analysis of single CTCs with a critical point of view on the technical challenges still to be faced and the steps needed to reach a standardization of the procedures able to translate these new approaches into clinical practice. Expert commentary: CTCs represent a surrogate tumor sample obtained by a minimally invasive procedure allowing the serial monitoring of the patient during the follow-up period or after treatment. Notwithstanding that, the analysis of CTCs is not so widespread; in fact, a limited number of centers can be equipped and possess the expertise for the development of workflows able to identify, enrich and isolate CTCs from blood. Moreover, the lack of standardized procedures and guidelines limits the study of CTCs to 'research use only' approaches.
Collapse
Affiliation(s)
- Francesca Salvianti
- a Department of Clinical and Experimental Biomedical Sciences 'Mario Serio' , University of Florence , Florence , Italy
| | - Pamela Pinzani
- a Department of Clinical and Experimental Biomedical Sciences 'Mario Serio' , University of Florence , Florence , Italy
| |
Collapse
|
46
|
Zhao W, Cheng R, Jenkins BD, Zhu T, Okonkwo NE, Jones CE, Davis MB, Kavuri SK, Hao Z, Schroeder C, Mao L. Label-free ferrohydrodynamic cell separation of circulating tumor cells. LAB ON A CHIP 2017; 17:3097-3111. [PMID: 28809987 PMCID: PMC5595667 DOI: 10.1039/c7lc00680b] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Circulating tumor cells (CTCs) have significant implications in both basic cancer research and clinical applications. To address the limited availability of viable CTCs for fundamental and clinical investigations, effective separation of extremely rare CTCs from blood is critical. Ferrohydrodynamic cell separation (FCS), a label-free method that conducted cell sorting based on cell size difference in biocompatible ferrofluids, has thus far not been able to enrich low-concentration CTCs from cancer patients' blood because of technical challenges associated with processing clinical samples. In this study, we demonstrated the development of a laminar-flow microfluidic FCS device that was capable of enriching rare CTCs from patients' blood in a biocompatible manner with a high throughput (6 mL h-1) and a high rate of recovery (92.9%). Systematic optimization of the FCS devices through a validated analytical model was performed to determine optimal magnetic field and its gradient, ferrofluid properties, and cell throughput that could process clinically relevant amount of blood. We first validated the capability of the FCS devices by successfully separating low-concentration (∼100 cells per mL) cancer cells using six cultured cell lines from undiluted white blood cells (WBCs), with an average 92.9% cancer cell recovery rate and an average 11.7% purity of separated cancer cells, at a throughput of 6 mL per hour. Specifically, at ∼100 cancer cells per mL spike ratio, the recovery rates of cancer cells were 92.3 ± 3.6% (H1299 lung cancer), 88.3 ± 5.5% (A549 lung cancer), 93.7 ± 5.5% (H3122 lung cancer), 95.3 ± 6.0% (PC-3 prostate cancer), 94.7 ± 4.0% (MCF-7 breast cancer), and 93.0 ± 5.3% (HCC1806 breast cancer), and the corresponding purities of separated cancer cells were 11.1 ± 1.2% (H1299 lung cancer), 10.1 ± 1.7% (A549 lung cancer), 12.1 ± 2.1% (H3122 lung cancer), 12.8 ± 1.6% (PC-3 prostate cancer), 11.9 ± 1.8% (MCF-7 breast cancer), and 12.2 ± 1.6% (HCC1806 breast cancer). Biocompatibility study on H1299 cell line and HCC1806 cell line showed that separated cancer cells had excellent short-term viability, normal proliferation and unaffected key biomarker expressions. We then demonstrated the enrichment of CTCs in blood samples obtained from two patients with newly diagnosed advanced non-small cell lung cancer (NSCLC). While still at its early stage of development, FCS could become a complementary tool for CTC separation for its high recovery rate and excellent biocompatibility, as well as its potential for further optimization and integration with other separation methods.
Collapse
Affiliation(s)
- Wujun Zhao
- Department of Chemistry, The University of Georgia, Athens, GA 30602, USA
| | - Rui Cheng
- College of Engineering, The University of Georgia, Athens, GA 30602, USA
| | | | - Taotao Zhu
- Department of Chemistry, The University of Georgia, Athens, GA 30602, USA
| | - Nneoma E. Okonkwo
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Courtney E. Jones
- College of Engineering and Computer Science, Syracuse University, Syracuse, NY 13210, USA
| | - Melissa B. Davis
- Department of Genetics, The University of Georgia, Athens, GA 30602, USA
| | - Sravan K. Kavuri
- Department of Pathology, Augusta University, Augusta, GA 30912, USA
| | - Zhonglin Hao
- Department of Medicine, Augusta University, Augusta, GA 30912, USA
| | | | - Leidong Mao
- College of Engineering, The University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
47
|
Paolillo C, Mu Z, Rossi G, Schiewer MJ, Nguyen T, Austin L, Capoluongo E, Knudsen K, Cristofanilli M, Fortina P. Detection of Activating Estrogen Receptor Gene ( ESR1) Mutations in Single Circulating Tumor Cells. Clin Cancer Res 2017; 23:6086-6093. [PMID: 28679775 DOI: 10.1158/1078-0432.ccr-17-1173] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/20/2017] [Accepted: 06/30/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Early detection is essential for treatment plans before onset of metastatic disease. Our purpose was to demonstrate feasibility to detect and monitor estrogen receptor 1 (ESR1) gene mutations at the single circulating tumor cell (CTC) level in metastatic breast cancer (MBC).Experimental Design: We used a CTC molecular characterization approach to investigate heterogeneity of 14 hotspot mutations in ESR1 and their correlation with endocrine resistance. Combining the CellSearch and DEPArray technologies allowed recovery of 71 single CTCs and 12 WBC from 3 ER-positive MBC patients. Forty CTCs and 12 WBC were subjected to whole genome amplification by MALBAC and Sanger sequencing.Results: Among 3 selected patients, 2 had an ESR1 mutation (Y537). One showed two different ESR1 variants in a single CTC and another showed loss of heterozygosity. All mutations were detected in matched cell-free DNA (cfDNA). Furthermore, one had 2 serial blood samples analyzed and showed changes in both cfDNA and CTCs with emergence of mutations in ESR1 (Y537S and T570I), which has not been reported previously.Conclusions: CTCs are easily accessible biomarkers to monitor and better personalize management of patients with previously demonstrated ER-MBC who are progressing on endocrine therapy. We showed that single CTC analysis can yield important information on clonal heterogeneity and can be a source of discovery of novel and potential driver mutations. Finally, we also validate a workflow for liquid biopsy that will facilitate early detection of ESR1 mutations, the emergence of endocrine resistance and the choice of further target therapy. Clin Cancer Res; 23(20); 6086-93. ©2017 AACR.
Collapse
Affiliation(s)
- Carmela Paolillo
- Department of Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia Pennsylvania.,Institute of Biochemistry and Clinical Biochemistry, Laboratory of Clinical Molecular and Personalized Diagnostics, Catholic University of the Sacred Heart, Rome, Italy
| | - Zhaomei Mu
- Department of Medicine-Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Giovanna Rossi
- Department of Medicine-Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Matthew J Schiewer
- Department of Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia Pennsylvania
| | - Thomas Nguyen
- Department of Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia Pennsylvania
| | - Laura Austin
- Department of Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia Pennsylvania
| | - Ettore Capoluongo
- Institute of Biochemistry and Clinical Biochemistry, Laboratory of Clinical Molecular and Personalized Diagnostics, Catholic University of the Sacred Heart, Rome, Italy
| | - Karen Knudsen
- Department of Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia Pennsylvania
| | - Massimo Cristofanilli
- Department of Medicine-Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - Paolo Fortina
- Department of Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia Pennsylvania. .,Department of Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
48
|
Li M, Anand RK. High-Throughput Selective Capture of Single Circulating Tumor Cells by Dielectrophoresis at a Wireless Electrode Array. J Am Chem Soc 2017; 139:8950-8959. [PMID: 28609630 DOI: 10.1021/jacs.7b03288] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We demonstrate continuous high-throughput selective capture of circulating tumor cells by dielectrophoresis at arrays of wireless electrodes (bipolar electrodes, BPEs). The use of BPEs removes the requirement of ohmic contact to individual array elements, thus enabling otherwise unattainable device formats. Capacitive charging of the electrical double layer at opposing ends of each BPE allows an AC electric field to be transmitted across the entire device. Here, two such designs are described and evaluated. In the first design, BPEs interconnect parallel microchannels. Pockets extruding from either side of the microchannels volumetrically control the number of cells captured at each BPE tip and enhance trapping. High-fidelity single-cell capture was achieved when the pocket dimensions were matched to those of the cells. A second, open design allows many non-targeted cells to pass through. These devices enable high-throughput capture of rare cells and single-cell analysis.
Collapse
Affiliation(s)
- Min Li
- Department of Chemistry, Iowa State University , Ames, Iowa 50010, United States
| | - Robbyn K Anand
- Department of Chemistry, Iowa State University , Ames, Iowa 50010, United States
| |
Collapse
|
49
|
Chen Q, Li D, Lin J, Wang M, Xuan X. Simultaneous Separation and Washing of Nonmagnetic Particles in an Inertial Ferrofluid/Water Coflow. Anal Chem 2017; 89:6915-6920. [PMID: 28548482 DOI: 10.1021/acs.analchem.7b01608] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Magnetic fluids (e.g., paramagnetic solutions and ferrofluids) have been increasingly used for label-free separation of nonmagnetic particles in microfluidic devices. Their biocompatibility, however, becomes a concern in high-throughput or large-volume applications. One way to potentially resolve this issue is resuspending the particles that are separated in a magnetic fluid immediately into a biocompatible buffer. We demonstrate herein the proof-of-principle of the first integration of negative magnetophoresis and inertial focusing for a simultaneous separation and washing of nonmagnetic particles in coflowing ferrofluid and water streams. The two operations take place in parallel in a simple T-shaped rectangular microchannel with a nearby permanent magnet. We find that the larger and smaller particles' exiting positions (and hence their separation distance) in the sheath water and ferrofluid suspension, respectively, vary with the total flow rate or the flow rate ratio between the two streams.
Collapse
Affiliation(s)
- Qi Chen
- Department of Mechanical Engineering, Clemson University , Clemson, South Carolina 29634-0921, United States.,MOA Key Laboratory of Agricultural Information Acquisition Technology (Beijing), China Agricultural University , Beijing 10083, China
| | - Di Li
- Department of Mechanical Engineering, Clemson University , Clemson, South Carolina 29634-0921, United States
| | - Jianhan Lin
- MOA Key Laboratory of Agricultural Information Acquisition Technology (Beijing), China Agricultural University , Beijing 10083, China
| | - Maohua Wang
- MOA Key Laboratory of Agricultural Information Acquisition Technology (Beijing), China Agricultural University , Beijing 10083, China
| | - Xiangchun Xuan
- Department of Mechanical Engineering, Clemson University , Clemson, South Carolina 29634-0921, United States
| |
Collapse
|
50
|
Antfolk M, Laurell T. Continuous flow microfluidic separation and processing of rare cells and bioparticles found in blood – A review. Anal Chim Acta 2017; 965:9-35. [DOI: 10.1016/j.aca.2017.02.017] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 01/31/2017] [Accepted: 02/03/2017] [Indexed: 12/12/2022]
|