1
|
Xu Q, Wang Y, Dou S, Xu Y, Xu Z, Xu H, Zhang Y, Xia Y, Xue Y, Li H, Ma X, Zhang K, Wang H, Ma F, Wang Q, Li B, Wang W. High-Viability Circulating Tumor Cells Sorting From Whole Blood at Single Cell Level Using Laser-Induced Forward Transfer-Assisted Microfiltration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414195. [PMID: 39868845 PMCID: PMC12079465 DOI: 10.1002/advs.202414195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/20/2025] [Indexed: 01/28/2025]
Abstract
The efficient isolation and molecular analysis of circulating tumor cells (CTCs) from whole blood at single-cell level are crucial for understanding tumor metastasis and developing personalized treatments. The viability of isolated cells is the key prerequisite for the downstream molecular analysis, especially for RNA sequencing. This study develops a laser-induced forward transfer -assisted microfiltration system (LIFT-AMFS) for high-viability CTC enrichment and retrieval from whole blood. The LIFT-compatible double-stepped microfilter (DSMF), central to this system, comprises two micropore layers: the lower layer's smaller micropores facilitate size-based cell separation, and the upper layer's larger micropores enable liquid encapsulating captured cells. By optimizing the design of the DSMFs, the system has a capture efficiency of 88% at the processing throughput of up to 15.0 mL min-1 during the microfilter-based size screening stage, with a single-cell yield of over 95% during the retrieval stage. The retrieved single cells, with high viability, are qualified for ex vivo culture and direct RNA sequencing. The cDNA yield from isolated CTCs surpassed 4.5 ng, sufficient for library construction. All single-cell sequencing data exhibited Q30 scores above 95.92%. The LIFT-AMFS shows promise in cellular and biomedical research.
Collapse
Affiliation(s)
- Qingmei Xu
- School of Integrated CircuitsPeking UniversityBeijing100871China
- Department of Electrical EngineeringTaiyuan Institute of TechnologyTaiyuan030008China
| | - Yuntong Wang
- Changchun Institute of OpticsFine Mechanics and PhysicsChinese Academy of SciencesChangchun130033China
- University of Chinese Academy of SciencesBeijing100049China
| | - Songtao Dou
- School of Integrated CircuitsPeking UniversityBeijing100871China
| | - Yang Xu
- Cancer Translational Medicine Research CenterThe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
- Department of Respiratory MedicineThe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
| | - Zhenhe Xu
- State Key Laboratory of Bioreactor EngineeringShanghai Frontiers Science Center of Optogenetic Techniques for Cell MetabolismEast China University of Science and TechnologyShanghai200237China
| | - Han Xu
- School of Integrated CircuitsPeking UniversityBeijing100871China
| | - Yi Zhang
- School of Integrated CircuitsPeking UniversityBeijing100871China
| | - Yanming Xia
- Guangzhou National LaboratoryGuangzhou510320China
| | - Ying Xue
- Hooke LaboratoryChangchun130033China
| | - Hang Li
- Hooke LaboratoryChangchun130033China
| | - Xiao Ma
- Hangzhou Branemagic Medical Technology Co. Ltd.Hangzhou310021China
| | | | - Huan Wang
- Changchun Institute of OpticsFine Mechanics and PhysicsChinese Academy of SciencesChangchun130033China
- University of Chinese Academy of SciencesBeijing100049China
| | - Fengzhou Ma
- School of Integrated CircuitsPeking UniversityBeijing100871China
- Hangzhou Branemagic Medical Technology Co. Ltd.Hangzhou310021China
| | - Qi Wang
- Cancer Translational Medicine Research CenterThe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
- Department of Respiratory MedicineThe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
| | - Bei Li
- Changchun Institute of OpticsFine Mechanics and PhysicsChinese Academy of SciencesChangchun130033China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wei Wang
- School of Integrated CircuitsPeking UniversityBeijing100871China
- National Key Laboratory of Advanced Micro and Nano Manufacture TechnologyBeijing100871China
- Beijing Advanced Innovation Center for Integrated CircuitsBeijing100871China
| |
Collapse
|
2
|
Sun J, Han S, Yang R, Guo L, Li J, Li C, Xu L, Liu H, Dong B. Combining hybrid cell membrane modified magnetic nanoparticles and inverted microfluidic chip for in situ CTCs capture and inactivation. Biosens Bioelectron 2024; 263:116575. [PMID: 39067413 DOI: 10.1016/j.bios.2024.116575] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/25/2024] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
Circulating tumor cells (CTCs) serve as crucial indicators for tumor occurrence, progression, and prognosis monitoring. However, achieving high sensitivity and high purity capture of CTCs remains challenging. Additionally, in situ capture and synchronous clearance hold promise as methods to impede tumor metastasis, but further exploration is needed. In this study, biomimetic cell membrane-coated magnetic nanoparticles (NPs) were designed to address the issue of nonspecific adsorption of capture probes by the immune system during blood circulation. Membranes from human breast cancer cells (tumor cell membranes, TMs) and leukocytes (white blood cell membranes, WMs) were extracted and fused to form a hybrid membrane (HM), which was further modified onto the surface of porous magnetic NPs loaded with indocyanine green (ICG). The incorporation of TM enhanced the material's target specificity, thus increasing capture efficiency, while WM coating reduced interference from homologous white blood cells (WBCs), further enhancing capture purity. Additionally, in conjunction with our novel inverted microfluidic chip, this work introduces the first use of polymer photonic crystals as the capture interface for CTCs. Besides providing an advantageous surface structure for CTC attachment, the 808 nm photonic bandgap effectively amplifies the 808 nm excitation light at the capture surface position. Therefore, upon capturing CTCs, the ICG molecules in the probes facilitate enhanced photothermal (PTT) and photodynamic (PDT) synergistic effects, directly inactivating the captured CTCs. This method achieves capture efficiency and purity exceeding 95% and permits in situ inactivation post-capture, providing an important approach for future research on impeding tumor metastasis in vivo.
Collapse
Affiliation(s)
- Jiao Sun
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, 2699 Qianjin Street, Changchun, 130012, China; Department of Cell Biology, College of Basic Medical Science, Jilin University, Changchun, 130021, China
| | - Songrui Han
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Rui Yang
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Lihua Guo
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Jiawei Li
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Chang Chun, 130021, China
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, China
| | - Lin Xu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Haipeng Liu
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Chang Chun, 130021, China.
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, 2699 Qianjin Street, Changchun, 130012, China.
| |
Collapse
|
3
|
Xu Y, Chen B, He M, Cui Z, Hu B. All-in-One Microfluidic Chip for Online Labeling, Separating, and Focusing Rare Circulating Tumor Cells from Blood Samples Followed by Inductively Coupled Plasma Mass Spectrometry Detection. Anal Chem 2023; 95:14061-14067. [PMID: 37677145 DOI: 10.1021/acs.analchem.3c02680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Circulating tumor cell (CTC) detection is essential for early cancer diagnosis and evaluating treatment efficacy. Despite the growing interest in isolating CTCs and further quantifying surface biomarkers at the single-cell level, highly efficient separation of rare CTCs from massive blood cells is still a big challenge. Here, we developed an all-in-one microfluidic chip system for the immunolabeling, magnetic separation, and focusing of HepG2 cells (as a CTC model) and online combined it with single cell-inductively coupled plasma mass spectrometry (SC-ICP-MS) for quantitative analysis of the asialoglycoprotein receptor (ASGPR) on single HepG2 cells. Lanthanide-labeled anti-ASGPR monoclonal antibody and antiepithelial cell adhesion molecule-modified magnetic beads were prepared as signal and magnetic probes, respectively. Target cells were highly efficiently labeled with signal and magnetic probes in the mixing zone of the microfluidic chip and then focused and sorted in the separation zone by specific magnetic separation techniques to avoid matrix contamination. The average cell recovery of HepG2 cells was derived to be 94.1 ± 5.7% with high separation efficiency and purity. The sorted cells with signal probes were detected for enumeration and quantification of ASGPR on their surface by SC-ICP-MS. The developed method showed good specificity and high sensitivity, detecting an average of (1.0 ± 0.2) × 105 ASGPR molecules per cell surface. This method can be used for absolute quantitative analysis of ASGPR on the surface of single hepatocellular carcinoma cells in real-world samples, providing a highly efficient analytical platform for studying targeted drug delivery in cancer therapy.
Collapse
Affiliation(s)
- Yan Xu
- 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
| | - Zewei Cui
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Bin Hu
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
4
|
Orrapin S, Thongkumkoon P, Udomruk S, Moonmuang S, Sutthitthasakul S, Yongpitakwattana P, Pruksakorn D, Chaiyawat P. Deciphering the Biology of Circulating Tumor Cells through Single-Cell RNA Sequencing: Implications for Precision Medicine in Cancer. Int J Mol Sci 2023; 24:12337. [PMID: 37569711 PMCID: PMC10418766 DOI: 10.3390/ijms241512337] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Circulating tumor cells (CTCs) hold unique biological characteristics that directly involve them in hematogenous dissemination. Studying CTCs systematically is technically challenging due to their extreme rarity and heterogeneity and the lack of specific markers to specify metastasis-initiating CTCs. With cutting-edge technology, single-cell RNA sequencing (scRNA-seq) provides insights into the biology of metastatic processes driven by CTCs. Transcriptomics analysis of single CTCs can decipher tumor heterogeneity and phenotypic plasticity for exploring promising novel therapeutic targets. The integrated approach provides a perspective on the mechanisms underlying tumor development and interrogates CTCs interactions with other blood cell types, particularly those of the immune system. This review aims to comprehensively describe the current study on CTC transcriptomic analysis through scRNA-seq technology. We emphasize the workflow for scRNA-seq analysis of CTCs, including enrichment, single cell isolation, and bioinformatic tools applied for this purpose. Furthermore, we elucidated the translational knowledge from the transcriptomic profile of individual CTCs and the biology of cancer metastasis for developing effective therapeutics through targeting key pathways in CTCs.
Collapse
Affiliation(s)
- Santhasiri Orrapin
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Patcharawadee Thongkumkoon
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Sasimol Udomruk
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
| | - Sutpirat Moonmuang
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Songphon Sutthitthasakul
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Petlada Yongpitakwattana
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Dumnoensun Pruksakorn
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
- Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
| | - Parunya Chaiyawat
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
| |
Collapse
|
5
|
Jou HJ, Lo PH, Ling PY. Recent Advances of Microfluidic Platform for Cell Based Non-Invasive Prenatal Diagnosis. Int J Mol Sci 2023; 24:ijms24020991. [PMID: 36674508 PMCID: PMC9865170 DOI: 10.3390/ijms24020991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
The purpose of the present review is to try to highlight recent advances in the application of microfluidic technology on non-invasive prenatal diagnosis (NIPD). The immunoaffinity based microfluidic technology is the most common approach for NIPD, followed by size-based microfluidic methods. Immunoaffinity microfluidic methods can enrich and isolate circulating fetal extravillous trophoblasts (fEVTs) or fetal nucleated red blood cells (fnRBCs) for NIPD by using specific antibodies, but size-based microfluidic systems are only applied to isolate fEVTs. Most studies based on the immunoaffinity microfluidic system gave good results. Enough fetal cells were obtained for chromosomal and/or genetic analysis in all blood samples. However, the results from studies using size-based microfluidic systems for NIPD are less than ideal. In conclusion, recent advances in microfluidic devices make the immunoaffinity based microfluidic system potentially a powerful tool for cell-based NIPD. However, more clinical validation is needed.
Collapse
Affiliation(s)
- Hei-Jen Jou
- Departments of Obstetrics and Gynecology, Taiwan Adventist Hospital, Taipei 105404, Taiwan
- School of Nursing, National Taipei University of Nursing and Health Science, Taipei 112303, Taiwan
- Departments of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100225, Taiwan
- International College of Semiconductor Technology, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
- Correspondence: ; Tel.: +886-2-7718151 (ext. 2833)
| | - Pei-Hsuan Lo
- Departments of Obstetrics and Gynecology, Taiwan Adventist Hospital, Taipei 105404, Taiwan
| | - Pei-Ying Ling
- Departments of Obstetrics and Gynecology, Taiwan Adventist Hospital, Taipei 105404, Taiwan
| |
Collapse
|
6
|
Erickson A, Chiarelli PA, Huang J, Levengood SL, Zhang M. Electrospun nanofibers for 3-D cancer models, diagnostics, and therapy. NANOSCALE HORIZONS 2022; 7:1279-1298. [PMID: 36106417 DOI: 10.1039/d2nh00328g] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
As one of the leading causes of global mortality, cancer has prompted extensive research and development to advance efficacious drug discovery, sustained drug delivery and improved sensitivity in diagnosis. Towards these applications, nanofibers synthesized by electrospinning have exhibited great clinical potential as a biomimetic tumor microenvironment model for drug screening, a controllable platform for localized, prolonged drug release for cancer therapy, and a highly sensitive cancer diagnostic tool for capture and isolation of circulating tumor cells in the bloodstream and for detection of cancer-associated biomarkers. This review provides an overview of applied nanofiber design with focus on versatile electrospinning fabrication techniques. The influence of topographical, physical, and biochemical properties on the function of nanofiber assemblies is discussed, as well as current and foreseeable barriers to the clinical translation of applied nanofibers in the field of oncology.
Collapse
Affiliation(s)
- Ariane Erickson
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| | - Peter A Chiarelli
- The Saban Research Institute, University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Jianxi Huang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| | - Sheeny Lan Levengood
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
7
|
Wang D, Dong R, Wang X, Jiang X. Flexible Electronic Catheter Based on Nanofibers for the In Vivo Elimination of Circulating Tumor Cells. ACS NANO 2022; 16:5274-5283. [PMID: 35302351 DOI: 10.1021/acsnano.1c09807] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Clearing circulating tumor cells (CTCs) that are closely related to cancer metastasis and recurrence in peripheral blood helps to reduce the probability of cancer recurrence and metastasis. However, conventional therapies aiming at killing CTCs always cause damage to normal blood cells, tissues, and organs. Here, we report a flexible electronic catheter that can capture and kill CTCs via irreversible electroporation (IRE) with high efficiency. The flexible electronic catheter is assembled from nanofibers (NFs) with liquid metal-polymer conductor (MPC) electrodes. The NFs were modified with an epithelial cellular adhesion molecule (EpCAM) antibody on the surface to improve specific biorecognition and cell adhesion. Whole-body blood can be screened by the catheter repeatedly, during which the EpCAM antibody on a nanofiber can enrich CTCs to the surface of the catheter. Taking advantage of the high specific surface area, the capture efficiency of NF-based catheters for CTCs is 25 times higher than previously reported cases. Furthermore, the number of nonspecifically captured WBCs is less than 10 per mm2 areas of the catheter, compared to their original large number of 4-11 × 106 mL-1 of whole blood, showing good specificity of the flexible electronic catheter. The flexible and biocompatible MPC electrodes have a high killing efficiency of 100% for the captured CTCs in a rabbit model. No noticeable hematologic index and morphological changes of the vessels and major organs were observed, indicating that this electronic catheter had good biocompatibility. The present functional electronic catheter offers an alternative strategy for improving the efficiency of clinical cancer therapy.
Collapse
Affiliation(s)
- Dou Wang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088, Xueyuan Rd., Xili, Nanshan District, Shenzhen, Guangdong 518055, P. R. China
| | - Ruihua Dong
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088, Xueyuan Rd., Xili, Nanshan District, Shenzhen, Guangdong 518055, P. R. China
| | - Xuedong Wang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088, Xueyuan Rd., Xili, Nanshan District, Shenzhen, Guangdong 518055, P. R. China
| | - Xingyu Jiang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088, Xueyuan Rd., Xili, Nanshan District, Shenzhen, Guangdong 518055, P. R. China
| |
Collapse
|
8
|
Liu Y, Li R, Zhang L, Guo S. Nanomaterial-Based Immunocapture Platforms for the Recognition, Isolation, and Detection of Circulating Tumor Cells. Front Bioeng Biotechnol 2022; 10:850241. [PMID: 35360401 PMCID: PMC8964261 DOI: 10.3389/fbioe.2022.850241] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/18/2022] [Indexed: 01/10/2023] Open
Abstract
Circulating tumor cells (CTCs) are a type of cancer cells that circulate in the peripheral blood after breaking away from solid tumors and are essential for the establishment of distant metastasis. Up to 90% of cancer-related deaths are caused by metastatic cancer. As a new type of liquid biopsy, detecting and analyzing CTCs will provide insightful information for cancer diagnosis, especially the in-time disease status, which would avoid some flaws and limitations of invasive tissue biopsy. However, due to the extremely low levels of CTCs among a large number of hematologic cells, choosing immunocapture platforms for CTC detection and isolation will achieve good performance with high purity, selectivity, and viability. These properties are directly associated with precise downstream analysis of CTC profiling. Recently, inspired by the nanoscale interactions of cells in the tissue microenvironment, platforms based on nanomaterials have been widely explored to efficiently enrich and sensitively detect CTCs. In this review, various immunocapture platforms based on different nanomaterials for efficient isolation and sensitive detection of CTCs are outlined and discussed. First, the design principles of immunoaffinity nanomaterials are introduced in detail. Second, the immunocapture and release of platforms based on nanomaterials ranging from nanoparticles, nanostructured substrates, and immunoaffinity microfluidic chips are summarized. Third, recent advances in single-cell release and analysis of CTCs are introduced. Finally, some perspectives and challenges are provided in future trends of CTC studies.
Collapse
Affiliation(s)
- Yichao Liu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Rui Li
- Xinjiang Key Laboratory of Solid State Physics and Devices, Xinjiang University, Urumqi, China
| | - Lingling Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Lingling Zhang, ; Shishang Guo,
| | - Shishang Guo
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, China
- *Correspondence: Lingling Zhang, ; Shishang Guo,
| |
Collapse
|
9
|
Circulating tumour cells in the -omics era: how far are we from achieving the 'singularity'? Br J Cancer 2022; 127:173-184. [PMID: 35273384 PMCID: PMC9296521 DOI: 10.1038/s41416-022-01768-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/27/2022] [Accepted: 02/17/2022] [Indexed: 12/22/2022] Open
Abstract
Over the past decade, cancer diagnosis has expanded to include liquid biopsies in addition to tissue biopsies. Liquid biopsies can result in earlier and more accurate diagnosis and more effective monitoring of disease progression than tissue biopsies as samples can be collected frequently. Because of these advantages, liquid biopsies are now used extensively in clinical care. Liquid biopsy samples are analysed for circulating tumour cells (CTCs), cell-free DNA, RNA, proteins and exosomes. CTCs originate from the tumour, play crucial roles in metastasis and carry information on tumour heterogeneity. Multiple single-cell omics approaches allow the characterisation of the molecular makeup of CTCs. It has become evident that CTCs are robust biomarkers for predicting therapy response, clinical development of metastasis and disease progression. This review describes CTC biology, molecular heterogeneity within CTCs and the involvement of EMT in CTC dynamics. In addition, we describe the single-cell multi-omics technologies that have provided insights into the molecular features within therapy-resistant and metastasis-prone CTC populations. Functional studies coupled with integrated multi-omics analyses have the potential to identify therapies that can intervene the functions of CTCs.
Collapse
|
10
|
Ojha AK, Rajasekaran R, Pandey AK, Dutta A, Seesala VS, Das SK, Chaudhury K, Dhara S. Nanotheranostics: Nanoparticles Applications, Perspectives, and Challenges. BIOSENSING, THERANOSTICS, AND MEDICAL DEVICES 2022:345-376. [DOI: 10.1007/978-981-16-2782-8_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
|
11
|
He S, Wei J, Ding L, Yang X, Wu Y. State-of-the-arts techniques and current evolving approaches in the separation and detection of circulating tumor cell. Talanta 2021; 239:123024. [PMID: 34952370 DOI: 10.1016/j.talanta.2021.123024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/01/2023]
Abstract
Circulating tumor cells (CTCs) are cancer cells that shed from the primary tumor and then enter the circulatory system, a small part of which may evolve into metastatic cancer under appropriate microenvironment conditions. The detection of CTCs is a truly noninvasive, dynamic monitor for disease changes, which has considerable clinical implications in the selection of targeted drugs. However, their inherent rarity and heterogeneity pose significant challenges to their isolation and detection. Even the "gold standard", CellSearch™, suffers from high expenses, low capture efficiency, and the consumption of time. With the advancement of CTCs analysis technologies in recent years, the yield and efficiency of CTCs enrichment have gradually been improved, as well as detection sensitivity. In this review, the isolation and detection strategies of CTCs have been completely described and the potential directions for future research and development have also been highlighted through analyzing the challenges faced by current strategies.
Collapse
Affiliation(s)
- Sitian He
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| | - Jinlan Wei
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaonan Yang
- School of Information Engineering, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
12
|
Liu Y, Wang X, Zhou Y, Yang G, Hou J, Zhou S. Engineered multifunctional metal-phenolic nanocoatings for label-free capture and "self-release" of heterogeneous circulating tumor cells. NANOSCALE 2021; 13:16923-16931. [PMID: 34522934 DOI: 10.1039/d1nr04112f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Immunomagnetic beads have been widely explored as an important analytical tool for the rapid and sensitive detection of circulating tumor cells (CTCs). However, their clinical application is seriously hindered by the tedious preparation procedures and heterogeneous nature of CTCs. To this end, a designed multifunctional platform named Fe3O4@TA/CuII superparamagnetic nanoparticles (SPMNPs) is expected to have the following features: (i) the formation of a tannic acid-copper (II) ion (TA/CuII) coating which could be accomplished by a one-step method is very simple; (ii) the TA/CuII coating shows high affinity for heterogeneous CTCs and good resistance to nonspecific adhesion of blood cells; (iii) "self-release" of the captured cells could be achieved as the TA/CuII coating gradually degrades in the cell culture environment without any additional interventions. Therefore, the resulting Fe3O4@TA/CuII SPMNPs could capture various CTCs (MCF-7, HepG2 and HeLa cells) with different expression levels of the epithelial cell adhesion molecule (EpCAM). And the capture efficiency and cell purity can reach 88% and 87%, respectively. In addition, 68% of the captured cells are self-released after 6 h of incubation and most of the released cells show high cell proliferation activity. In particular, Fe3O4@TA/CuII SPMNPs can successfully detect 1-13 CTCs from 1 mL of blood of 14 patients with 6 types of cancers. Hence, we expect that the as-prepared Fe3O4@TA/CuII SPMNPs with simple, efficient, and universal yet cost-efficient characteristics could act as a promising analytical tool for clinical CTC detection.
Collapse
Affiliation(s)
- Yiling Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Xiaoshan Wang
- Cancer Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuwei Zhou
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Guang Yang
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jianwen Hou
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
13
|
Li R, Gong Z, Liu Y, Zhao X, Guo S. Detection of circulating tumor cells and single cell extraction technology: principle, effect and application prospect. NANO FUTURES 2021; 5:032002. [DOI: 10.1088/2399-1984/ac1325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Wang J, Li Y, Wang R, Han C, Xu S, You T, Li Y, Xia J, Xu X, Wang D, Tang H, Yang C, Chen X, Peng Z. A Fully Automated and Integrated Microfluidic System for Efficient CTC Detection and Its Application in Hepatocellular Carcinoma Screening and Prognosis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:30174-30186. [PMID: 34142547 DOI: 10.1021/acsami.1c06337] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Analysis of circulating tumor cells (CTCs) is regarded as a useful diagnostic index to monitor tumor development and guide precision medicine. Although the immunoassay is a common strategy for CTC identification and heterogeneity characterization, it is challenged by poor reaction efficiency and laborious manipulations in microdevices, which hinder the sensitivity, throughput, simplification, and applicability. To meet the need for rapid, sensitive, and simple CTC analysis, we developed an efficient CTC detection system by integrating a 3D printed off-chip multisource reagent platform, a bubble retainer, and a single CTC capture microchip, which can achieve CTC capture and identification within 90 min. Compared with traditional CTC identification methods, this system decreases immunostaining time and antibody consumption by 90% and performs the on-chip immunoassay in a fully automated manner. Using this system, CTCs from the peripheral blood of 19 patients with various cancers were captured, detected, and compared with clinical data. The system shows great potential for early screening, real-time monitoring, and precision medicine for hepatocellular carcinoma (HCC). With the advantages of automation, stability, economy, and user-friendly operation, the proposed system is promising for clinical scenarios.
Collapse
Affiliation(s)
- Jie Wang
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Yang Li
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Rui Wang
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Chao Han
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiquan Xu
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Tingting You
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Yuhuan Li
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Junjie Xia
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Xing Xu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Dongmei Wang
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Huamei Tang
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - 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, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China
| |
Collapse
|
15
|
Circulating Tumor Cells from Enumeration to Analysis: Current Challenges and Future Opportunities. Cancers (Basel) 2021; 13:cancers13112723. [PMID: 34072844 PMCID: PMC8198976 DOI: 10.3390/cancers13112723] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 05/25/2021] [Indexed: 01/19/2023] Open
Abstract
Simple Summary With estimated numbers of 1–10 per mL of blood, circulating tumor cells (CTCs) are extremely rare compared to white (a few million) or red (billions) blood cells. Given their critical role in metastasis, CTCs have enormous potential as a biomarker for cancer diagnosis, prognosis, and monitoring of treatment response. There are now efforts to characterize CTCs more precisely through molecular and functional analysis, expanding the CTC effort from one of diagnosis and prognosis to now include the use of CTCs to specifically target cancers and discover therapeutic solutions, establishing CTCs as critical in precision medicine. This article summarizes current knowledge about CTC isolation technologies and discusses the translational benefits of different types of downstream analysis approaches, including single-CTC analysis, ex vivo expansion of CTCs, and characterization of CTC-associated cells. Abstract Circulating tumor cells (CTCs) have been recognized as a major contributor to distant metastasis. Their unique role as metastatic seeds renders them a potential marker in the circulation for early cancer diagnosis and prognosis as well as monitoring of therapeutic response. In the past decade, researchers mainly focused on the development of isolation techniques for improving the recovery rate and purity of CTCs. These developed techniques have significantly increased the detection sensitivity and enumeration accuracy of CTCs. Currently, significant efforts have been made toward comprehensive molecular characterization, ex vivo expansion of CTCs, and understanding the interactions between CTCs and their associated cells (e.g., immune cells and stromal cells) in the circulation. In this review, we briefly summarize existing CTC isolation technologies and specifically focus on advances in downstream analysis of CTCs and their potential applications in precision medicine. We also discuss the current challenges and future opportunities in their clinical utilization.
Collapse
|
16
|
Sun N, Lee YT, Kim M, Wang JJ, Zhang C, Teng PC, Qi D, Zhang RY, Tran BV, Lee YT, Ye J, Palomique J, Nissen NN, Han SHB, Sadeghi S, Finn RS, Saab S, Busuttil RW, Posadas EM, Liang L, Pei R, Yang JD, You S, Agopian VG, Tseng HR, Zhu Y. Covalent Chemistry-Mediated Multimarker Purification of Circulating Tumor Cells Enables Noninvasive Detection of Molecular Signatures of Hepatocellular Carcinoma. ADVANCED MATERIALS TECHNOLOGIES 2021; 6:2001056. [PMID: 34212072 PMCID: PMC8240468 DOI: 10.1002/admt.202001056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Indexed: 05/02/2023]
Abstract
Transcriptomic profiling of tumor tissues introduces a large database, which has led to improvements in the ability of cancer diagnosis, treatment, and prevention. However, performing tumor transcriptomic profiling in the clinical setting is very challenging since the procurement of tumor tissues is inherently limited by invasive sampling procedures. Here, we demonstrated the feasibility of purifying hepatocellular carcinoma (HCC) circulating tumor cells (CTCs) from clinical patient samples with improved molecular integrity using Click Chips in conjunction with a multimarker antibody cocktail. The purified CTCs were then subjected to mRNA profiling by NanoString nCounter platform, targeting 64 HCC-specific genes, which were generated from an integrated data analysis framework with 8 tissue-based prognostic gene signatures from 7 publicly available HCC transcriptomic studies. After bioinformatics analysis and comparison, the HCC CTC-derived gene signatures showed high concordance with HCC tissue-derived gene signatures from TCGA database, suggesting that HCC CTCs purified by Click Chips could enable the translation of HCC tissue molecular profiling into a noninvasive setting.
Collapse
Affiliation(s)
- Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Minhyung Kim
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jasmine J Wang
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ceng Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Pai-Chi Teng
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dongping Qi
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Ryan Y Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Benjamin V Tran
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Yue Tung Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Jinglei Ye
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Juvelyn Palomique
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nicholas N Nissen
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Steven-Huy B Han
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Saeed Sadeghi
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Richard S Finn
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Sammy Saab
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Ronald W Busuttil
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Edwin M Posadas
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Li Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, P.R. China
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou 215123, P.R. China
| | - Ju Dong Yang
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sungyong You
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Vatche G Agopian
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| |
Collapse
|
17
|
Cheng SB, Chen MM, Wang YK, Sun ZH, Qin Y, Tian S, Dong WG, Xie M, Huang WH. A Three-Dimensional Conductive Scaffold Microchip for Effective Capture and Recovery of Circulating Tumor Cells with High Purity. Anal Chem 2021; 93:7102-7109. [PMID: 33908770 DOI: 10.1021/acs.analchem.1c00785] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effective acquirement of highly pure circulating tumor cells (CTCs) is very important for CTC-related research. However, it is a great challenge since abundant white blood cells (WBCs) are always co-collected with CTCs because of nonspecific bonding or low depletion rate of WBCs in various CTC isolation platforms. Herein, we designed a three-dimensional (3D) conductive scaffold microchip for highly effective capture and electrochemical release of CTCs with high purity. The conductive 3D scaffold was prepared by dense immobilization of gold nanotubes (Au NTs) on porous polydimethylsiloxane and was functionalized with a CTC-specific biomolecule facilitated by a Au-S bond before embedding into a microfluidic device. The spatially distributed 3D macroporous structure compelled cells to change migration from linear to chaotic and the densely covered Au NTs enhanced the topographic interaction between cells and the substrate, thus synergistically improving the CTC capture efficiency. The Au NT-coated 3D scaffold had good electrical conductivity and the Au-S bond was breakable by voltage exposure so that captured CTCs could be specifically released by electrochemical stimulation while nonspecifically bonded WBCs were not responsive to this process, facilitating recovery of CTCs with high purity. The 3D conductive scaffold microchip was successfully applied to obtain highly pure CTCs from cancer patients' blood, benefiting the downstream analysis of CTCs.
Collapse
Affiliation(s)
- Shi-Bo Cheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Miao-Miao Chen
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Yi-Ke Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Zi-Han Sun
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yu Qin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Shan Tian
- Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei-Guo Dong
- Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Min Xie
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
18
|
Electrochemical sensing technology for liquid biopsy of circulating tumor cells-a review. Bioelectrochemistry 2021; 140:107823. [PMID: 33915341 DOI: 10.1016/j.bioelechem.2021.107823] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/01/2021] [Accepted: 04/11/2021] [Indexed: 02/06/2023]
Abstract
In recent years, a lot of new detection techniques for circulating tumor cells (CTCs) have been developed. Among them, electrochemical sensing technology has gradually developed because of its advantages of good selectivity, high sensitivity, low cost and rapid detection. Especially in the latest decade, the field of electrochemical biosensing has witnessed great progress, thanks to the merging of biosensing research area with nanotechnology, immunotechnology, nucleic acid technology, and microfluidic technology. In this review, the recent progress for the detection of CTCs according to the principle of detection was summarized and how they can contribute to the enhanced performance of such biosensors was explained. The latest electrode construction strategies such as rolling circle amplification reaction, DNA walker and microfluidic technology and their advantages were also introduced emphatically. Moreover, the main reasonswhy the existing biosensors have not been widely used clinically and the next research points were clearly put forward.
Collapse
|
19
|
Casanova-Salas I, Athie A, Boutros PC, Del Re M, Miyamoto DT, Pienta KJ, Posadas EM, Sowalsky AG, Stenzl A, Wyatt AW, Mateo J. Quantitative and Qualitative Analysis of Blood-based Liquid Biopsies to Inform Clinical Decision-making in Prostate Cancer. Eur Urol 2021; 79:762-771. [PMID: 33422353 DOI: 10.1016/j.eururo.2020.12.037] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022]
Abstract
CONTEXT Genomic stratification can impact prostate cancer (PC) care through diagnostic, prognostic, and predictive biomarkers that aid in clinical decision-making. The temporal and spatial genomic heterogeneity of PC together with the challenges of acquiring metastatic tissue biopsies hinder implementation of tissue-based molecular profiling in routine clinical practice. Blood-based liquid biopsies are an attractive, minimally invasive alternative. OBJECTIVE To review the clinical value of blood-based liquid biopsy assays in PC and identify potential applications to accelerate the development of precision medicine. EVIDENCE ACQUISITION A systematic review of PubMed/MEDLINE was performed to identify relevant literature on blood-based circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and extracellular vesicles (EVs) in PC. EVIDENCE SYNTHESIS Liquid biopsy has emerged as a practical tool to profile tumor dynamics over time, elucidating features that evolve (genome, epigenome, transcriptome, and proteome) with tumor progression. Liquid biopsy tests encompass analysis of DNA, RNA, and proteins that can be detected in CTCs, ctDNA, or EVs. Blood-based liquid biopsies have demonstrated promise in the context of localized tumors (diagnostic signatures, risk stratification, and disease monitoring) and advanced disease (response/resistance biomarkers and prognostic markers). CONCLUSIONS Liquid biopsies have value as a source of prognostic, predictive, and response biomarkers in PC. Most clinical applications have been developed in the advanced metastatic setting, where CTC and ctDNA yields are significantly higher. However, standardization of assays and analytical/clinical validation is necessary prior to clinical implementation. PATIENT SUMMARY Traces of tumors can be isolated from blood samples from patients with prostate cancer either as whole cells or as DNA fragments. These traces provide information on tumor features. These minimally invasive tests can guide diagnosis and treatment selection.
Collapse
Affiliation(s)
- Irene Casanova-Salas
- Vall d'Hebron Institute of Oncology (VHIO) and Vall d'Hebron University Hospital, Barcelona, Spain
| | - Alejandro Athie
- Vall d'Hebron Institute of Oncology (VHIO) and Vall d'Hebron University Hospital, Barcelona, Spain
| | - Paul C Boutros
- Departments of Human Genetics and Urology, Institute for Precision Health and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - David T Miyamoto
- Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edwin M Posadas
- Translational Oncology Program & Urologic Oncology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, Tübingen, Germany
| | - Alexander W Wyatt
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Joaquin Mateo
- Vall d'Hebron Institute of Oncology (VHIO) and Vall d'Hebron University Hospital, Barcelona, Spain.
| |
Collapse
|
20
|
Wang H, Wu J, Zhang Q, Hao J, Wang Y, Li Z, Niu H, Zhang H, Zhang S. A Modified Method to Isolate Circulating Tumor Cells and Identify by a Panel of Gene Mutations in Lung Cancer. Technol Cancer Res Treat 2021; 20:1533033821995275. [PMID: 34032165 PMCID: PMC8155778 DOI: 10.1177/1533033821995275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/29/2020] [Accepted: 01/27/2021] [Indexed: 01/01/2023] Open
Abstract
The CellSearch system is the only FDA approved and successful used detection technology for circulating tumor cells(CTCs). However, the process for identification of CTCs by CellSearch appear to damage the cells, which may adversely affects subsequent molecular biology assays. We aimed to explore and establish a membrane-preserving method for immunofluorescence identification of CTCs that keeping the isolated cells intact. 98 patients with lung cancer were enrolled, and the efficacy of clinical detection of CTCs was examined. Based on the CellSearch principle, we optimized an anti-EpCAM antibody and improved cell membrane rupture. A 5 ml peripheral blood sample was used to enrich CTCs with EpCAM immunomagnetic beads. Fluorescence signals were amplified with secondary antibodies against anti-EpCAM antibody attached on immunomagnetic beads. After identifying CTCs, single CTCs were isolated by micromanipulation. To confirm CTCs, genomic DNA was extracted and amplified at the single cell level to sequence 72 target genes of lung cancer and analyze the mutation copy number variations (CNVs) and gene mutations. A goat anti-mouse polyclonal antibody conjugated with Dylight 488 was selected to stain tumor cells. We identified CTCs based on EpCAM+ and CD45+ cells to exclude white blood cells. In the 98 lung cancer patients, the detection rate of CTCs (≥1 CTC) per 5 ml blood was 87.76%, the number of detections was 1-36, and the median was 2. By sequencing 72 lung cancer-associated genes, we found a high level of CNVs and gene mutations characteristic of tumor cells. We established a new CTCs staining scheme that significantly improves the detection rate and allows further analysis of CTCs characteristics at the genetic level.
Collapse
Affiliation(s)
- Helin Wang
- Department of Oncology, The First Affiliated Hospital of Xinxiang
Medical University, Henan, China
| | - Jieqing Wu
- Department of Oncology, The First Affiliated Hospital of Xinxiang
Medical University, Henan, China
| | - Qi Zhang
- Department of Oncology, Beijing Chaoyang Huanxing Cancer Hospital,
Beijing, China
| | - Jianqing Hao
- Department of Respiratory Medicine, Qingyang People’s Hospital,
Gansu, China
| | - Ying Wang
- Department of Oncology, The First Affiliated Hospital of Xinxiang
Medical University, Henan, China
| | - Zhuoran Li
- Department of Oncology, The First Affiliated Hospital of Xinxiang
Medical University, Henan, China
| | - Hongrui Niu
- Department of Oncology, The First Affiliated Hospital of Xinxiang
Medical University, Henan, China
| | - Hongtao Zhang
- Department of Central Laboratory, Beijing Chest Hospital, Capital
Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute,
Beijing, China
| | - Shucai Zhang
- Department of Oncology, Beijing Chest Hospital, Capital Medical
University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing,
China
| |
Collapse
|
21
|
Xu L, Li R, Wang Z, Cui H, Li W, Yu M, Guo SS, Zhao XZ. Electrospun degradable Zn-Mn oxide hierarchical nanofibers for specific capture and efficient release of circulating tumor cells. NANOTECHNOLOGY 2020; 31:495102. [PMID: 32990263 DOI: 10.1088/1361-6528/abb48b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Constructing biological affinity devices is considered as an effective strategy for isolating circulating tumor cells (CTCs), and electrospun nanofibers (ESNFs) have recently received attention. However, the current research focuses on polymer fibers, and fabricating stimuli-responsive inorganic nanofibers for cancer diagnosis and analysis is still challenging. In this work, Zn-Mn oxide nanofibers (ZnMnNFs) are used to capture and purify cancer cells after modification with specific antibodies. Then, the hierarchical nanofibers are degraded by reductive weak acid to release the captured cells efficiently without residues. Fusion of Zn and Mn, two transition metals, enhances the surface activity of oxides so that ZnMnNFs are easier to be degraded and modified. By using MCF-7 cancer cells, the cell capture efficiency of ZnMnNFs is up to 88.2%. Furthermore, by using citric acid, it is discovered that, by comparison with Mn oxide nanofibers, the cell release efficiency of ZnMnNFs is improved to 95.1% from 15.4%. In addition, the viability of released cells exceeds 90%. Lastly, the robustness of ZnMnNFs substrates is tested in peripheral blood from breast cancer patients (BCP) and colorectal cancer patients (CCP). Combined with fluorescence labeling, CTCs are confirmed to be isolated from all the clinical samples. This is the first trial of using ternary inorganic ESNFs for cancer cell capture. It is anticipated that the degradable ESNFs will provide biocompatible theranostic platforms and overcome the current limitations of cell release for high-precision gene analysis.
Collapse
Affiliation(s)
- Longguang Xu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Rui Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Zixiang Wang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Heng Cui
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Wei Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Mingxia Yu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, People's Republic of China
| | - Shi-Shang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| |
Collapse
|
22
|
Rezaei Z, Mahmoudifard M. Pivotal role of electrospun nanofibers in microfluidic diagnostic systems - a review. J Mater Chem B 2020; 7:4602-4619. [PMID: 31364667 DOI: 10.1039/c9tb00682f] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recently, the usage of electrospinning technology for the fabrication of fine fibers with a good deal of variation in morphology and structure has drawn the attention of many researchers around the world. These fibers have found their way in the many fields of science including medical diagnosis, tissue engineering, drug delivery, replica molding, solar cells, catalysts, energy conversion and storage, physical and chemical sensors and other applications. Among all applications, biosensing with the aim of rapid and sensitive biomarker detection is an area that warrants attention. Electrospun nanofibrous membranes enjoy numerous factors which benefit them to be used as potential candidates in biosensing platforms. Some of these factors include a high surface to volume ratio, analogous scale compared to bioactive molecules and relatively defect-free properties of nanofibers (NFs). In this review, we focused on the recent advances in electrospun nanofibrous membrane-based micro-analytical devices with an application as diagnostic systems. Hence, a study on the electrospun nanofiber usage in lab-on-a-chip and paper-based point-of-care devices, with an opening introduction to biosensors, nanofibers, the electrospinning method, and microfluidics as the principles of the intended subject, is provided. It is anticipated that the given examples in this paper will provide sufficient evidence for the potential of electrospun NFs for being used as a substrate in the commercial fabrication of highly sensitive and selective biosensors.
Collapse
Affiliation(s)
- Zahra Rezaei
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran and Chemical & Petroleum Engineering Department, Sharif University of Technology, Tehran, Iran.
| | - Matin Mahmoudifard
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
23
|
Qian W, Miao Z, Zhang XJ, Yang XT, Tang YY, Tang YY, Hu LY, Li S, Zhu D, Cheng H. Functionalized reduced graphene oxide with aptamer macroarray for cancer cell capture and fluorescence detection. Mikrochim Acta 2020; 187:407. [PMID: 32594259 DOI: 10.1007/s00604-020-04402-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/17/2020] [Indexed: 12/29/2022]
Abstract
An integrated aptamer macroarray functionalized with reduced graphene oxide (rGO) to specifically capture and sensitively detect cancer cells is reported. The capture for cancer cells is based on effective recognition of the modified rGO surface through the aptamer against epithelial cell adhesion molecule (EpCAM). The rough structure of rGO enhances morphologic interactions between rGO film interface and the cancer cells, while super-hydrophilicity of modified rGO hinders nonspecific cell capture. The synergistic interactions offer the aptamer macroarray high efficiency of cancer cell capture. By means of a turn-on fluorescence strategy based on the conformation change of the aptamer induced by the target recognition, the enriched cancer cells can be directly read out at excitation/emission wavelengths of 550/680 nm without washing, separation, and dying steps. The working range is 1 × 102 to 2 × 104 cells per mL with a detection limit of 22 cells per mL. The results indicate that the aptamer macroarray has a considerable foreground for early diagnosis, therapy, and monitoring of cancer. Graphical abstract.
Collapse
Affiliation(s)
- Wenhui Qian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Zhaoyi Miao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Xiao-Jing Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Xiao-Tong Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Ying-Ying Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Yu Ying Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Lin Yu Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Su Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Dong Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China. .,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing, China.
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, the First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
24
|
Cheng SB, Wang M, Zhang C, Chen MM, Wang YK, Tian S, Zhan N, Dong WG, Xie M, Huang WH. Flexible Three-Dimensional Net for Intravascular Fishing of Circulating Tumor Cells. Anal Chem 2020; 92:5447-5455. [PMID: 32162513 DOI: 10.1021/acs.analchem.0c00203] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Current strategies for in vitro isolation of circulating tumor cells (CTCs) fail to detect extremely rare CTCs heterogeneously distributed in blood. It is possible to devise methods for in vivo capture of CTCs based on processing almost all of the blood in the human body to improve detection sensitivity, but the complicated manipulation, biosafety concerns, and limited capture efficiency of conventional detection strategies prohibit their implementation in the clinic. Herein, we present a flexible three-dimensional (3-D) CTC-Net probe for intravascular collection of CTCs. The CTC-Net, consisting of a 3-D elastic scaffold with an interconnected, spatially distributed network accommodates a large quantity of immobilized antibodies and provides an enhanced substrate-cell contact frequency, which results in an enhanced capture efficiency and effective detection of heterogeneous CTCs. The as-prepared CTC-Net can be readily compressed and injected into blood vessels and fully unfolded to form a 3-D "fishing-net" structure for capture of the CTCs, and then retracted for imaging and downstream gene analysis of the captured CTCs. Significant advantages for the CTC-Net over currently available in vitro and in vivo procedures are demonstrated for detection of extremely rare CTCs from wild-type rats and successful capture of CTCs and CTC clusters before metastasis in the case of tumor-bearing rats. Our research demonstrates for the first time the use of a 3-D scaffold CTC-Net probe for in vivo capture of CTCs. The method shows exceptional performance for cell capture, which is readily implemented and holds great potential in the clinic for early diagnosis of cancer.
Collapse
Affiliation(s)
- Shi-Bo Cheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Ming Wang
- Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chi Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | - Miao-Miao Chen
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yi-Ke Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Shan Tian
- Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Na Zhan
- Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei-Guo Dong
- Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Min Xie
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
25
|
Zhu Y, Zou C, Zhang J, Jiang W, Guan F, Tang K, Li S, Li G, Wang J, Ke Z. Dynamically Monitoring the Clonal Evolution of Lung Cancer Based on the Molecular Characterization of Circulating Tumor Cells Using Aptamer Cocktail-Modified Nanosubstrates. ACS APPLIED MATERIALS & INTERFACES 2020; 12:5671-5679. [PMID: 31940177 DOI: 10.1021/acsami.9b22234] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Dynamically monitoring the clonal evolution of lung cancer and performing molecular analyses on tumor cells are challenging but necessary tasks to adjust therapeutic interventions and evaluate treatment efficacy. Circulating tumor cells (CTCs), as a "liquid biopsy", may offer an auxiliary tool to identify phenotypic transformation of solid tumors at primary or metastatic sites and uncover their corresponding molecular variation. Herein, we developed an aptamer-modified PEG-PLGA-nanofiber (PPN) microfluidic system optimized for recognizing rare CTC subtypes in lung cancer patients. This unique purification system can be adopted to monitor the clonal evolution of solid tumors by following the intrinsic immunophenotypes of CTCs, while significantly enhancing capture efficiency for polyclonal-derived tumor cells, further facilitating therapeutic evaluation via dynamic CTC enumeration. Combining with downstream single-cell sequencing, the aptamer-modified-PPN microfluidic system was able to provide early insight into tumor heterogeneity and predict histologic transformation in advance, broadening its clinical applications in lung cancer patients.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Pathology or Precision Medicine Institute , The First Affiliated Hospital of Sun Yat-Sen University , Guangzhou , Guangdong 510080 , P.R. China
| | - Chang Zou
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University , the First Affiliated Hospital of Southern University, Shenzhen People's Hospital , Shenzhen , Guangdong 518020 , P.R. China
| | - Jian Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital , Sun Yat-sen University , Guangzhou , Guangdong 510630 , P.R. China
| | - Wenting Jiang
- Department of Pathology or Precision Medicine Institute , The First Affiliated Hospital of Sun Yat-Sen University , Guangzhou , Guangdong 510080 , P.R. China
| | - Fanglin Guan
- Health Science Center , Xi'an Jiaotong University , Xi'an , Shanxi 710061 , P.R. China
| | - Kejing Tang
- Department of Pathology or Precision Medicine Institute , The First Affiliated Hospital of Sun Yat-Sen University , Guangzhou , Guangdong 510080 , P.R. China
| | - Shuhua Li
- Department of Pathology or Precision Medicine Institute , The First Affiliated Hospital of Sun Yat-Sen University , Guangzhou , Guangdong 510080 , P.R. China
| | - Guannan Li
- Beijing National Laboratory for Molecular Sciences (BNLMS) , Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Jianhong Wang
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University , the First Affiliated Hospital of Southern University, Shenzhen People's Hospital , Shenzhen , Guangdong 518020 , P.R. China
| | - Zunfu Ke
- Department of Pathology or Precision Medicine Institute , The First Affiliated Hospital of Sun Yat-Sen University , Guangzhou , Guangdong 510080 , P.R. China
| |
Collapse
|
26
|
Dong J, Chen JF, Smalley M, Zhao M, Ke Z, Zhu Y, Tseng HR. Nanostructured Substrates for Detection and Characterization of Circulating Rare Cells: From Materials Research to Clinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903663. [PMID: 31566837 PMCID: PMC6946854 DOI: 10.1002/adma.201903663] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/02/2019] [Indexed: 05/03/2023]
Abstract
Circulating rare cells in the blood are of great significance for both materials research and clinical applications. For example, circulating tumor cells (CTCs) have been demonstrated as useful biomarkers for "liquid biopsy" of the tumor. Circulating fetal nucleated cells (CFNCs) have shown potential in noninvasive prenatal diagnostics. However, it is technically challenging to detect and isolate circulating rare cells due to their extremely low abundance compared to hematologic cells. Nanostructured substrates offer a unique solution to address these challenges by providing local topographic interactions to strengthen cell adhesion and large surface areas for grafting capture agents, resulting in improved cell capture efficiency, purity, sensitivity, and reproducibility. In addition, rare-cell retrieval strategies, including stimulus-responsiveness and additive reagent-triggered release on different nanostructured substrates, allow for on-demand retrieval of the captured CTCs/CFNCs with high cell viability and molecular integrity. Several nanostructured substrate-enabled CTC/CFNC assays are observed maturing from enumeration and subclassification to molecular analyses. These can one day become powerful tools in disease diagnosis, prognostic prediction, and dynamic monitoring of therapeutic response-paving the way for personalized medical care.
Collapse
Affiliation(s)
- Jiantong Dong
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jie-Fu Chen
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
27
|
Soltani S, Khanian N, Choong TSY, Rashid U. Recent progress in the design and synthesis of nanofibers with diverse synthetic methodologies: characterization and potential applications. NEW J CHEM 2020. [DOI: 10.1039/d0nj01071e] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The advancements of nanotechnology, particularly nanomaterials science, have produced a broad range of nanomaterials including nanofibers, nanorods, nanowires and etc., which have been technically and practically examined over various applications.
Collapse
Affiliation(s)
- Soroush Soltani
- Department of Chemical and Environmental Engineering
- Universiti Putra Malaysia
- Malaysia
| | | | | | - Umer Rashid
- Institute of Advanced Technology
- Universiti Putra Malaysia
- Malaysia
| |
Collapse
|
28
|
Jiménez-Zenteno AK, Cerf A. Liquid Biopsy Based on Circulating Cancer-Associated Cells: Bridging the Gap from an Emerging Concept to a Mainstream Tool in Precision Medicine. ACTA ACUST UNITED AC 2019; 4:e1900164. [PMID: 32293131 DOI: 10.1002/adbi.201900164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/15/2019] [Indexed: 01/01/2023]
Abstract
The concept of liquid biopsy and the isolation and analysis of circulating biomarkers from blood samples is proposed as a surrogate to solid biopsies and can have the potential to revolutionize the management of patients with cancer. The relevance of circulating tumor cells (CTCs) and the importance of the information they carry is acknowledged by the medical community. But what are the barriers to clinical adoption? This review draws a panorama of the biological implications of CTCs, their physical and biochemical properties, and the current technological bottlenecks for their analysis in relation with the medical needs. Keys and considerations to bridge the technological and clinical gaps that still need to be overcome to be able to introduce CTCs in clinical routine are finally synthesized.
Collapse
Affiliation(s)
| | - Aline Cerf
- Université de Toulouse, CNRS, 7 Avenue du Colonel Roche, 31400, Toulouse, France
| |
Collapse
|
29
|
General Study and Gene Expression Profiling of Endotheliocytes Cultivated on Electrospun Materials. MATERIALS 2019; 12:ma12244082. [PMID: 31817735 PMCID: PMC6947544 DOI: 10.3390/ma12244082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/20/2019] [Accepted: 12/03/2019] [Indexed: 12/27/2022]
Abstract
Endothelization of the luminal surface of vascular grafts is required for their long-term functioning. Here, we have cultivated human endothelial cells (HUVEC) on different 3D matrices to assess cell proliferation, gene expression and select the best substrate for endothelization. 3D matrices were produced by electrospinning from solutions of poly(D,L-lactide-co-glycolide) (PLGA), polycaprolactone (PCL), and blends of PCL with gelatin (Gl) in hexafluoroisopropanol. Structure and surface properties of 3D matrices were characterized by SEM, AFM, and sessile drop analysis. Cell adhesion, viability, and proliferation were studied by SEM, Alamar Blue staining, and 5-ethynyl-2’-deoxyuridine (EdU) assay. Gene expression profiling was done on an Illumina HiSeq 2500 platform. Obtained data indicated that 3D matrices produced from PCL with Gl and treated with glutaraldehyde provide the most suitable support for HUVEC adhesion and proliferation. Transcriptome sequencing has demonstrated a minimal difference of gene expression profile in HUVEC cultivated on the surface of these matrices as compared to tissue culture plastic, thus confirming these matrices as the best support for endothelization.
Collapse
|
30
|
Wei X, Chen K, Cai B, Rao L, Wang Z, Sun Y, Yu M, Liu W, Guo S, Zhao XZ. An Acoustic Droplet-Induced Enzyme Responsive Platform for the Capture and On-Demand Release of Single Circulating Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:41118-41126. [PMID: 31612699 DOI: 10.1021/acsami.9b16566] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The recovery of rare single circulating tumor cells (CTCs) from patients has great potential to facilitate the study of cell heterogeneity and cancer metastasis, which may promote the development of individualized cancer immunotherapy. Herein, a versatile single-cell recovery approach that utilizes an acoustic droplet-induced enzyme responsive platform for the capture and on-demand release of single CTCs is proposed. The platform combines a multifunctional enzyme-responsive gelatin nanoparticle (GNP)-decorated substrate (GNP-chip) for specific capture with an acoustic droplet positioning technique to realize on-demand release of single CTCs. The acoustic droplet dispenser is employed to generate oxidized alginate microdroplets containing the MMP-9 enzyme (OA-MMP-9) with controllable size and precise positioning upon the cell-attached GNP-chip, allowing controlled cell-surface biodegradation under enzymatic reactions followed by calcium chloride (CaCl2) solution treatment to form single-cell encapsulated calcium alginate hydrogels. Benefitting from the existence of hydrogels, the released cells could be efficiently recovered by microcapillary. Results demonstrate that the encapsulated cells maintain good cell morphology in the hydrogels, which allow further single-cell nucleic acid analysis. As a proof-of-concept platform, this approach enables reliable and efficient retrieval of single CTCs and holds the potential for versatility in single-cell analysis systems.
Collapse
Affiliation(s)
- Xiaoyun Wei
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan 430072 , China
| | - Keke Chen
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan 430072 , China
| | - Bo Cai
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Lang Rao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan 430072 , China
| | - Zixiang Wang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan 430072 , China
| | - Yue Sun
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan 430072 , China
| | - Mingxia Yu
- Department of Clinical Laboratory , Zhongnan Hospital of Wuhan University , Wuhan 430071 , China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan 430072 , China
| | - Shishang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan 430072 , China
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan 430072 , China
| |
Collapse
|
31
|
Li W, Wang H, Zhao Z, Gao H, Liu C, Zhu L, Wang C, Yang Y. Emerging Nanotechnologies for Liquid Biopsy: The Detection of Circulating Tumor Cells and Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805344. [PMID: 30589111 DOI: 10.1002/adma.201805344] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/29/2018] [Indexed: 05/18/2023]
Abstract
Liquid biopsy enables noninvasive and dynamic analysis of molecular or cellular biomarkers, and therefore holds great potential for the diagnosis, prognosis, monitoring of disease progress and treatment efficacy, understanding of disease mechanisms, and identification of therapeutic targets for drug development. In this review, the recent progress in nanomaterials, nanostructures, nanodevices, and nanosensors for liquid biopsy is summarized, with a focus on the detection and molecular characterization of circulating tumor cells (CTCs) and extracellular vesicles (EVs). The developments and advances of nanomaterials and nanostructures in enhancing the sensitivity, specificity, and purity for the detection of CTCs and EVs are discussed. Sensing techniques for signal transduction and amplification as well as visualization strategies are also discussed. New technologies for the reversible release of the isolated CTCs and EVs and for single-CTC/EV analysis are summarized. Emerging microfluidic platforms for the integral on-chip isolation, detection, and molecular analysis are also included. The opportunities, challenges, and prospects of these innovative materials and technologies, especially with regard to their feasibility in clinical applications, are discussed. The applications of nanotechnology-based liquid biopsy will bring new insight into the clinical practice in monitoring and treatment of tumor and other significant diseases.
Collapse
Affiliation(s)
- Wenzhe Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Huayi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zijian Zhao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Houqian Gao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Changliang Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
32
|
Rossi E, Zamarchi R. Single-Cell Analysis of Circulating Tumor Cells: How Far Have We Come in the -Omics Era? Front Genet 2019; 10:958. [PMID: 31681412 PMCID: PMC6811661 DOI: 10.3389/fgene.2019.00958] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor cells detach from the primary tumor or metastatic sites and enter the peripheral blood, often causing metastasis. These cells, named Circulating Tumor Cells (CTCs), display the same spatial and temporal heterogeneity as the primary tumor. Since CTCs are involved in tumor progression, they represent a privileged window to disclose mechanisms of metastases, while -omic analyses at the single-cell level allow dissection of the complex relationships between the tumor subpopulations and the surrounding normal tissue. However, in addition to reporting the proof of concept that we can query CTCs to reveal tumor evolution throughout the continuum of treatment for early detection of resistance to therapy, the scientific literature has also been highlighting the disadvantages of CTCs, which hampers a routine use of this approach in clinical practice. To date, an increasing number of CTC technologies, as well as -omics methods, have been employed, mostly lacking strong comparative analyses. The rarity of CTCs also represents a major challenge, because there is no consensus regarding the minimal criteria necessary and sufficient to define an event as CTC; moreover, we cannot often compare data from of one study with that of another. Finally, the availability of an individual tumor profile undermines the traditional histology-based treatment. Applying molecular data for patient benefit implies a collective effort by biologists, bioengineers, and clinicians, to create tools to interpret molecular data and manage precision medicine in every single patient. Herein, we focus on the most recent findings in CTC −omics to learn how far we have come.
Collapse
Affiliation(s)
- Elisabetta Rossi
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy.,Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Rita Zamarchi
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
33
|
Kim O, Lee D, Chungwon Lee A, Lee Y, Bae HJ, Lee HB, Kim RN, Han W, Kwon S. Whole Genome Sequencing of Single Circulating Tumor Cells Isolated by Applying a Pulsed Laser to Cell-Capturing Microstructures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1902607. [PMID: 31240868 DOI: 10.1002/smll.201902607] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Indexed: 06/09/2023]
Abstract
Single cell analysis of heterogeneous circulating tumor cells (CTCs), by which the genomic profiles of rare single CTCs are connected to the clinical status of cancer patients, is crucial for understanding cancer metastasis and the clinical impact on patients. However, the heterogeneity in genotypes and phenotypes and rarity of CTCs have limited extensive single CTC genome research, further hindering clinical investigation. Despite recent efforts to build platforms that separate CTCs, the investigation on CTCs is difficult due to the lack of a retrieval process at the single cell level. In this study, laser-induced isolation of microstructures on an optomechanically-transferrable-chip and sequencing (LIMO-seq) is applied for whole genome sequencing of single CTCs. Also, the whole genome sequences and the molecular profiles of the isolated single cells from the whole blood of a breast cancer patient are analyzed.
Collapse
Affiliation(s)
- Okju Kim
- Department of Electrical and Computer Engineering Institute of Engineering Research, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Daewon Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
- BK21+ Creative Research Engineer Development for IT, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Amos Chungwon Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yongju Lee
- Department of Electrical and Computer Engineering Institute of Engineering Research, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Hyung Jong Bae
- Nano Systems Institute, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Han-Byoel Lee
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Ryong Nam Kim
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Sunghoon Kwon
- Department of Electrical and Computer Engineering Institute of Engineering Research, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Institutes of Entrepreneurial BioConvergence, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
- Inter-University Semiconductor Research Center, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| |
Collapse
|
34
|
Cheng SB, Chen MM, Wang YK, Sun ZH, Xie M, Huang WH. Current techniques and future advance of microfluidic devices for circulating tumor cells. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Iliescu FS, Poenar DP, Yu F, Ni M, Chan KH, Cima I, Taylor HK, Cima I, Iliescu C. Recent advances in microfluidic methods in cancer liquid biopsy. BIOMICROFLUIDICS 2019; 13:041503. [PMID: 31431816 PMCID: PMC6697033 DOI: 10.1063/1.5087690] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/24/2019] [Indexed: 05/04/2023]
Abstract
Early cancer detection, its monitoring, and therapeutical prediction are highly valuable, though extremely challenging targets in oncology. Significant progress has been made recently, resulting in a group of devices and techniques that are now capable of successfully detecting, interpreting, and monitoring cancer biomarkers in body fluids. Precise information about malignancies can be obtained from liquid biopsies by isolating and analyzing circulating tumor cells (CTCs) or nucleic acids, tumor-derived vesicles or proteins, and metabolites. The current work provides a general overview of the latest on-chip technological developments for cancer liquid biopsy. Current challenges for their translation and their application in various clinical settings are discussed. Microfluidic solutions for each set of biomarkers are compared, and a global overview of the major trends and ongoing research challenges is given. A detailed analysis of the microfluidic isolation of CTCs with recent efforts that aimed at increasing purity and capture efficiency is provided as well. Although CTCs have been the focus of a vast microfluidic research effort as the key element for obtaining relevant information, important clinical insights can also be achieved from alternative biomarkers, such as classical protein biomarkers, exosomes, or circulating-free nucleic acids. Finally, while most work has been devoted to the analysis of blood-based biomarkers, we highlight the less explored potential of urine as an ideal source of molecular cancer biomarkers for point-of-care lab-on-chip devices.
Collapse
Affiliation(s)
- Florina S. Iliescu
- School of Applied Science, Republic Polytechnic, Singapore 738964, Singapore
| | - Daniel P. Poenar
- VALENS-Centre for Bio Devices and Signal Analysis, School of EEE, Nanyang Technological University, Singapore 639798, Singapore
| | - Fang Yu
- Singapore Institute of Manufacturing Technology, A*STAR, Singapore 138634, Singapore
| | - Ming Ni
- School of Biological Sciences and Engineering, Yachay Technological University, San Miguel de Urcuquí 100105, Ecuador
| | - Kiat Hwa Chan
- Division of Science, Yale-NUS College, Singapore 138527, Singapore
| | | | - Hayden K. Taylor
- Department of Mechanical Engineering, University of California, Berkeley, California 94720, USA
| | - Igor Cima
- DKFZ-Division of Translational Oncology/Neurooncology, German Cancer Consortium (DKTK), Heidelberg and University Hospital Essen, Essen 45147, Germany
| | | |
Collapse
|
36
|
Dong J, Jan YJ, Cheng J, Zhang RY, Meng M, Smalley M, Chen PJ, Tang X, Tseng P, Bao L, Huang TY, Zhou D, Liu Y, Chai X, Zhang H, Zhou A, Agopian VG, Posadas EM, Shyue JJ, Jonas SJ, Weiss PS, Li M, Zheng G, Yu HH, Zhao M, Tseng HR, Zhu Y. Covalent chemistry on nanostructured substrates enables noninvasive quantification of gene rearrangements in circulating tumor cells. SCIENCE ADVANCES 2019; 5:eaav9186. [PMID: 31392269 PMCID: PMC6669017 DOI: 10.1126/sciadv.aav9186] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 06/26/2019] [Indexed: 05/07/2023]
Abstract
Well-preserved mRNA in circulating tumor cells (CTCs) offers an ideal material for conducting molecular profiling of tumors, thereby providing a noninvasive diagnostic solution for guiding treatment intervention and monitoring disease progression. However, it is technically challenging to purify CTCs while retaining high-quality mRNA.Here, we demonstrate a covalent chemistry-based nanostructured silicon substrate ("Click Chip") for CTC purification that leverages bioorthogonal ligation-mediated CTC capture and disulfide cleavage-driven CTC release. This platform is ideal for CTC mRNA assays because of its efficient, specific, and rapid purification of pooled CTCs, enabling downstream molecular quantification using reverse transcription Droplet Digital polymerase chain reaction. Rearrangements of ALK/ROS1 were quantified using CTC mRNA and matched with those identified in biopsy specimens from 12 patients with late-stage non-small cell lung cancer. Moreover, CTC counts and copy numbers of ALK/ROS1 rearrangements could be used together for evaluating treatment responses and disease progression.
Collapse
Affiliation(s)
- Jiantong Dong
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yu Jen Jan
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ju Cheng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ryan Y. Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Meng Meng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pin-Jung Chen
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xinghong Tang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Patrick Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lirong Bao
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tzu-Yang Huang
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Dongjing Zhou
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Yupin Liu
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Xiaoshu Chai
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Haibo Zhang
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Anqi Zhou
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vatche G. Agopian
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Edwin M. Posadas
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jing-Jong Shyue
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Steven J. Jonas
- Department of Pediatrics, David Geffen School of Medicine, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, and Children’s Discovery and Innovation Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Paul S. Weiss
- California NanoSystems Institute, Departments of Chemistry and Biochemistry and Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mengyuan Li
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Guangjuan Zheng
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Hsiao-hua Yu
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
37
|
Lu YT, Delijani K, Mecum A, Goldkorn A. Current status of liquid biopsies for the detection and management of prostate cancer. Cancer Manag Res 2019; 11:5271-5291. [PMID: 31239778 PMCID: PMC6559244 DOI: 10.2147/cmar.s170380] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022] Open
Abstract
In recent years, new therapeutic options have become available for prostate cancer (PC) patients, generating an urgent need for better biomarkers to guide the choice of therapy and monitor treatment response. Liquid biopsies, including circulating tumor cells (CTCs), circulating nucleic acids, and exosomes, have been developed as minimally invasive assays allowing oncologists to monitor PC patients with real-time cellular or molecular information. While CTC counts remain the most extensively validated prognostic biomarker to monitor treatment response, recent advances demonstrate that CTC morphology and androgen receptor characterization can provide additional information to guide the choice of treatment. Characterization of cell-free DNA (cfDNA) is another rapidly emerging field with novel technologies capable of monitoring the evolution of treatment relevant alterations such as those in DNA damage repair genes for poly (ADP-ribose) polymerase (PARP) inhibition. In addition, several new liquid biopsy fields are emerging, including the characterization of heterogeneity, CTC RNA sequencing, the culture and xenografting of CTCs, and the characterization of extracellular vesicles (EVs) and circulating microRNAs. This review describes the clinical utilization of liquid biopsies in the management of PC patients and emerging liquid biopsy technologies with the potential to advance personalized cancer therapy.
Collapse
Affiliation(s)
- Yi-Tsung Lu
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Kevin Delijani
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Andrew Mecum
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Amir Goldkorn
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
38
|
Abstract
Electrospinning is a versatile and viable technique for generating ultrathin fibers. Remarkable progress has been made with regard to the development of electrospinning methods and engineering of electrospun nanofibers to suit or enable various applications. We aim to provide a comprehensive overview of electrospinning, including the principle, methods, materials, and applications. We begin with a brief introduction to the early history of electrospinning, followed by discussion of its principle and typical apparatus. We then discuss its renaissance over the past two decades as a powerful technology for the production of nanofibers with diversified compositions, structures, and properties. Afterward, we discuss the applications of electrospun nanofibers, including their use as "smart" mats, filtration membranes, catalytic supports, energy harvesting/conversion/storage components, and photonic and electronic devices, as well as biomedical scaffolds. We highlight the most relevant and recent advances related to the applications of electrospun nanofibers by focusing on the most representative examples. We also offer perspectives on the challenges, opportunities, and new directions for future development. At the end, we discuss approaches to the scale-up production of electrospun nanofibers and briefly discuss various types of commercial products based on electrospun nanofibers that have found widespread use in our everyday life.
Collapse
Affiliation(s)
- Jiajia Xue
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Tong Wu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Yunqian Dai
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, People’s Republic of China
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- School of Chemistry and Biochemistry, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
39
|
Wang X, Sun L, Zhang H, Wei L, Qu W, Zeng Z, Liu Y, Zhu Z. Microfluidic chip combined with magnetic-activated cell sorting technology for tumor antigen-independent sorting of circulating hepatocellular carcinoma cells. PeerJ 2019; 7:e6681. [PMID: 30972256 PMCID: PMC6448555 DOI: 10.7717/peerj.6681] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/25/2019] [Indexed: 12/29/2022] Open
Abstract
Purpose We aimed to generate a capture platform that integrates a deterministic lateral displacement (DLD) microfluidic structure with magnetic-activated cell sorting (MACS) technology for miniaturized, efficient, tumor antigen-independent circulating tumor cell (CTC) separation. Methods The microfluidic structure was based on the theory of DLD and was designed to remove most red blood cells and platelets. Whole Blood CD45 MicroBeads and a MACS separator were then used to remove bead-labeled white blood cells. We established HepG2 human liver cancer cells overexpressing green fluorescent protein by lentiviral transfection to simulate CTCs in blood, and these cells were then used to determine the CTC isolation efficiency of the device. The performance and clinical value of our platform were evaluated by comparison with the Abnova CytoQuest™ CR system in the separating of blood samples from 12 hepatocellular carcinoma patients undergoing liver transplantation in a clinical follow-up experiment. The isolated cells were stained and analyzed by confocal laser scanning microscopy. Results Using our integrated platform at the optimal flow rates for the specimen (60 µl/min) and buffer (100 µl/min per chip), we achieved an CTC yield of 85.1% ± 3.2%. In our follow-up of metastatic patients, CTCs that underwent epithelial–mesenchymal transition were found. These CTCs were missed by the CytoQuest™ CR bulk sorting approach, whereas our platform displayed increased sensitivity to EpCAMlow CTCs. Conclusions Our platform, which integrates microfluidic and MACS technology, is an attractive method for high-efficiency CTC isolation regardless of surface epitopes.
Collapse
Affiliation(s)
- Xuebin Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Liying Sun
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Haiming Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Lin Wei
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Wei Qu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Zhigui Zeng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Ying Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Zhijun Zhu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| |
Collapse
|
40
|
Yu CC, Chen YW, Yeh PY, Hsiao YS, Lin WT, Kuo CW, Chueh DY, You YW, Shyue JJ, Chang YC, Chen P. Random and aligned electrospun PLGA nanofibers embedded in microfluidic chips for cancer cell isolation and integration with air foam technology for cell release. J Nanobiotechnology 2019; 17:31. [PMID: 30782169 PMCID: PMC6379968 DOI: 10.1186/s12951-019-0466-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/11/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) comprise the high metastatic potential population of cancer cells in the blood circulation of humans; they have become the established biomarkers for cancer diagnosis, individualized cancer therapy, and cancer development. Technologies for the isolation and recovery of CTCs can be powerful cancer diagnostic tools for liquid biopsies, allowing the identification of malignancies and guiding cancer treatments for precision medicine. METHODS We have used an electrospinning process to prepare poly(lactic-co-glycolic acid) (PLGA) nanofibrous arrays in random or aligned orientations on glass slips. We then fabricated poly(methyl methacrylate) (PMMA)-based microfluidic chips embedding the PLGA nanofiber arrays and modified their surfaces through sequential coating with using biotin-(PEG)7-amine through EDC/NHS activation, streptavidin (SA), and biotinylated epithelial-cell adhesion-molecule antibody (biotin-anti-EpCAM) to achieve highly efficient CTC capture. When combined with an air foam technology that induced a high shear stress and, thereby, nondestructive release of the captured cells from the PLGA surfaces, the proposed device system operated with a high cell recovery rate. RESULTS The morphologies and average diameters of the electrospun PLGA nanofibers were characterized using scanning electron microscopy (SEM) and confocal Raman imaging. The surface chemistry of the PLGA nanofibers conjugated with the biotin-(PEG)7-amine was confirmed through time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging. The chip system was studied for the effects of the surface modification density of biotin-(PEG)7-amine, the flow rates, and the diameters of the PLGA nanofibers on the capture efficiency of EpCAM-positive HCT116 cells from the spiked liquid samples. To assess their CTC capture efficiencies in whole blood samples, the aligned and random PLGA nanofiber arrays were tested for their abilities to capture HCT116 cells, providing cancer cell capture efficiencies of 66 and 80%, respectively. With the continuous injection of air foam into the microfluidic devices, the cell release efficiency on the aligned PLGA fibers was 74% (recovery rate: 49%), while it was 90% (recovery rate: 73%) on the random PLGA fibers, from tests of 200 spiked cells in 2 mL of whole blood from healthy individuals. Our study suggests that integrated PMMA microfluidic chips embedding random PLGA nanofiber arrays may be suitable devices for the efficient capture and recovery of CTCs from whole blood samples.
Collapse
Affiliation(s)
- Chia-Cheng Yu
- Department of Materials Engineering, Ming Chi University of Technology, Taishan, New Taipei City, 24301, Taiwan
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-Wen Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Po-Ying Yeh
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yu-Sheng Hsiao
- Department of Materials Engineering, Ming Chi University of Technology, Taishan, New Taipei City, 24301, Taiwan.
| | - Wei-Ting Lin
- Department of Materials Engineering, Ming Chi University of Technology, Taishan, New Taipei City, 24301, Taiwan
| | - Chiung-Wen Kuo
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Di-Yen Chueh
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yun-Wen You
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Jing-Jong Shyue
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Ying-Chih Chang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan.
| | - Peilin Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
41
|
Wongkaew N. Nanofiber-integrated miniaturized systems: an intelligent platform for cancer diagnosis. Anal Bioanal Chem 2019; 411:4251-4264. [PMID: 30706075 DOI: 10.1007/s00216-019-01589-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/26/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022]
Abstract
Cancer diagnostic tools enabling screening, diagnosis, and effective disease management are essential elements to increase the survival rate of diagnosed patients. Low abundance of cancer markers present in large amounts of interferences remains the major issue. Moreover, current diagnostic technologies are restricted to high-resourced settings only. Integrating nanofibers into miniaturized analytical systems holds a significant promise to address these challenges as demonstrated by recent publications. A large surface area, three-dimensional porous network, and diverse range of functional chemistries make nanofibers an excellent candidate as immobilization support and/or transduction elements, enabling high capture yield and ultrasensitive detection in miniaturized devices. Functional nanofibers have thus been used to isolate and detect various cancer-related biomarkers with a high degree of success in both on-chip and off-chip platforms. In fact, the chemical and functional adaptability of nanofibers has been exploited to address the technical challenges unique to each of the cancer markers in body fluids, where circulating tumor cells are prominently investigated among others (proteins, nucleic acids, and exosomes). So far, none of the work has exploited the nanofibers for cancer-derived exosomes, opening an avenue for further research effort. The trend and future prospects signal possibilities to strengthen the implementation of nanofiber-miniaturized system hybrid for a next generation of cancer diagnostic platforms both in clinical and point-of-care testing.
Collapse
Affiliation(s)
- Nongnoot Wongkaew
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany.
| |
Collapse
|
42
|
Xiao Y, Wang M, Lin L, Du L, Shen M, Shi X. Specific capture and release of circulating tumor cells using a multifunctional nanofiber-integrated microfluidic chip. Nanomedicine (Lond) 2019; 14:183-199. [DOI: 10.2217/nnm-2018-0150] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To develop a multifunctional nanofibrous mat-embedded microfluidic chip system for specific capture and intact release of circulating tumor cells. Materials & methods: Electrospun polyethylenimine/polyvinyl alcohol nanofibers were functionalized with zwitterions to reduce the nonspecific adhesion of blood cells, followed by modification with arginine-glycine-aspartic acid peptide via an acid-sensitive benzoic imine bond. Results: The nanofiber-embedded microchip can be applied for capturing various types of cancer cells and circulating tumor cells with high efficiency and considerable purity. The captured cancer cells can be released from the nanofibrous substrates within 30 min. Conclusion: The developed multifunctional nanofiber-embedded microfluidic chip may have a great potential for clinical applications.
Collapse
Affiliation(s)
- Yunchao Xiao
- State Key Laboratory for Modification of Chemical Fibers & Polymer Materials, College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Mengyuan Wang
- State Key Laboratory for Modification of Chemical Fibers & Polymer Materials, College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Lizhou Lin
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Lianfang Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers & Polymer Materials, College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers & Polymer Materials, College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| |
Collapse
|
43
|
Wu J, Chen Q, Lin JM. Microfluidic technologies in cell isolation and analysis for biomedical applications. Analyst 2018; 142:421-441. [PMID: 27900377 DOI: 10.1039/c6an01939k] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Efficient platforms for cell isolation and analysis play an important role in applied and fundamental biomedical studies. As cells commonly have a size of around 10 microns, conventional handling approaches at a large scale are still challenged in precise control and efficient recognition of cells for further performance of isolation and analysis. Microfluidic technologies have become more prominent in highly efficient cell isolation for circulating tumor cells (CTCs) detection, single-cell analysis and stem cell separation, since microfabricated devices allow for the spatial and temporal control of complex biochemistries and geometries by matching cell morphology and hydrodynamic traps in a fluidic network, as well as enabling specific recognition with functional biomolecules in the microchannels. In addition, the fabrication of nano-interfaces in the microchannels has been increasingly emerging as a very powerful strategy for enhancing the capability of cell capture by improving cell-interface interactions. In this review, we focus on highlighting recent advances in microfluidic technologies for cell isolation and analysis. We also describe the general biomedical applications of microfluidic cell isolation and analysis, and finally make a prospective for future studies.
Collapse
Affiliation(s)
- Jing Wu
- School of Science, China University of Geosciences (Beijing), Beijing 100083, China.
| | - Qiushui Chen
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing 100084, China.
| | - Jin-Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
44
|
Wongkaew N, Simsek M, Griesche C, Baeumner AJ. Functional Nanomaterials and Nanostructures Enhancing Electrochemical Biosensors and Lab-on-a-Chip Performances: Recent Progress, Applications, and Future Perspective. Chem Rev 2018; 119:120-194. [DOI: 10.1021/acs.chemrev.8b00172] [Citation(s) in RCA: 305] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Nongnoot Wongkaew
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93053 Regensburg, Germany
| | - Marcel Simsek
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93053 Regensburg, Germany
| | - Christian Griesche
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93053 Regensburg, Germany
| | - Antje J. Baeumner
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
45
|
Kulasinghe A, Wu H, Punyadeera C, Warkiani ME. The Use of Microfluidic Technology for Cancer Applications and Liquid Biopsy. MICROMACHINES 2018; 9:E397. [PMID: 30424330 PMCID: PMC6187606 DOI: 10.3390/mi9080397] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/03/2018] [Accepted: 08/07/2018] [Indexed: 12/11/2022]
Abstract
There is growing awareness for the need of early diagnostic tools to aid in point-of-care testing in cancer. Tumor biopsy remains the conventional means in which to sample a tumor and often presents with challenges and associated risks. Therefore, alternative sources of tumor biomarkers is needed. Liquid biopsy has gained attention due to its non-invasive sampling of tumor tissue and ability to serially assess disease via a simple blood draw over the course of treatment. Among the leading technologies developing liquid biopsy solutions, microfluidics has recently come to the fore. Microfluidic platforms offer cellular separation and analysis platforms that allow for high throughout, high sensitivity and specificity, low sample volumes and reagent costs and precise liquid controlling capabilities. These characteristics make microfluidic technology a promising tool in separating and analyzing circulating tumor biomarkers for diagnosis, prognosis and monitoring. In this review, the characteristics of three kinds of circulating tumor markers will be described in the context of cancer, circulating tumor cells (CTCs), exosomes, and circulating tumor DNA (ctDNA). The review will focus on how the introduction of microfluidic technologies has improved the separation and analysis of these circulating tumor markers.
Collapse
Affiliation(s)
- Arutha Kulasinghe
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia.
| | - Hanjie Wu
- The School of Biomedical Engineering, Faculty of Engineering and Internet Technology, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Chamindie Punyadeera
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia.
| | - Majid Ebrahimi Warkiani
- The School of Biomedical Engineering, Faculty of Engineering and Internet Technology, University of Technology Sydney, Ultimo, NSW 2007, Australia.
- Institute of Molecular Medicine, Sechenov First Moscow State University, Moscow 119991, Russia.
| |
Collapse
|
46
|
Wu LL, Tang M, Zhang ZL, Qi CB, Hu J, Ma XY, Pang DW. Chip-Assisted Single-Cell Biomarker Profiling of Heterogeneous Circulating Tumor Cells Using Multifunctional Nanospheres. Anal Chem 2018; 90:10518-10526. [DOI: 10.1021/acs.analchem.8b02585] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Ling-Ling Wu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Man Tang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Zhi-Ling Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Chu-Bo Qi
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Jiao Hu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Xu-Yan Ma
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Dai-Wen Pang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| |
Collapse
|
47
|
Gao X, Li Q, Wang F, Liu X, Liu D. Dual-Responsive Self-Assembled Monolayer for Specific Capture and On-Demand Release of Live Cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:8145-8153. [PMID: 29933692 DOI: 10.1021/acs.langmuir.8b00676] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
We report a dual-responsive self-assembled monolayer (SAM) on a well-defined rough gold substrate for dynamic capture and release of live cells. By incorporating 5'-triphosphate (ATP) aptamer into a SAM, we can accurately isolate specific cell types and subsequently release captured cells at either population or desired-group (or even single-cell) levels. On one hand, the whole SAMs can be disassembled through addition of ATP solution, leading to the entire release of the captured cells from the supported substrate. On the other hand, desired cells can be selectively released using near-infrared light irradiation, with relatively high spatial and temporal precision. The proposed dual-responsive cell capture-and-release system is biologically friendly and is reusable with another round of modification, showing great usefulness in cancer diagnosis and molecular analysis.
Collapse
Affiliation(s)
- Xia Gao
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , Tianjin 300071 , China
| | - Qiang Li
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Fengchao Wang
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Xuehui Liu
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Dingbin Liu
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , Tianjin 300071 , China
| |
Collapse
|
48
|
Xu L, Zhao Y, Owusu KA, Zhuang Z, Liu Q, Wang Z, Li Z, Mai L. Recent Advances in Nanowire-Biosystem Interfaces: From Chemical Conversion, Energy Production to Electrophysiology. Chem 2018. [DOI: 10.1016/j.chempr.2018.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
49
|
Cui H, Wang B, Wang W, Hao Y, Liu C, Song K, Zhang S, Wang S. Frosted Slides Decorated with Silica Nanowires for Detecting Circulating Tumor Cells from Prostate Cancer Patients. ACS APPLIED MATERIALS & INTERFACES 2018; 10:19545-19553. [PMID: 29770688 DOI: 10.1021/acsami.8b06072] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Developing low-cost and highly efficient nanobiochips are important for liquid biopsies, real-time monitoring, and precision medicine. By in situ growth of silica nanowires on a commercial frosted slide, we develop a biochip for effective circulating tumor cells (CTCs) detection after modifying epithelial cell adhesion molecule antibody (anti-EpCAM). The biochip shows the specificity and high capture efficiency of 85.4 ± 8.3% for prostate cancer cell line (PC-3). The microsized frosted slides and silica nanowires allow enhanced efficiency in capture EpCAM positive cells by synergistic topographic interactions. And the capture efficiency of biochip increased with the increase of silica nanowires length on frosted slide. The biochip shows that micro/nanocomposite structures improve the capture efficiency of PC-3 more than 70% toward plain slide. Furthermore, the nanobiochip has been successfully applied to identify CTCs from whole blood specimens of prostate cancer patients. Thus, this frosted slide-based biochip may provide a cheap and effective way of clinical monitoring of CTCs.
Collapse
Affiliation(s)
- Haijun Cui
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Binshuai Wang
- Department of Urology , Peking University Third Hospital , Beijing 100191 , China
| | - Wenshuo Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yuwei Hao
- University of Chinese Academy of Sciences , Beijing 100049 , China
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Green Printing, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China
| | - Chuanyong Liu
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Kai Song
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
| | - Shudong Zhang
- Department of Urology , Peking University Third Hospital , Beijing 100191 , China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
50
|
Park ES, Yan JP, Ang RA, Lee JH, Deng X, Duffy SP, Beja K, Annala M, Black PC, Chi KN, Wyatt AW, Ma H. Isolation and genome sequencing of individual circulating tumor cells using hydrogel encapsulation and laser capture microdissection. LAB ON A CHIP 2018; 18:1736-1749. [PMID: 29762619 DOI: 10.1039/c8lc00184g] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Circulating tumor cells (CTCs) are malignant cells released into the bloodstream with the potential to form metastases in secondary sites. These cells, acquired non-invasively, represent a sample of highly relevant tumor tissue that is an alternative to difficult and low-yield tumor biopsies. In recent years, there has been growing interest in genomic profiling of CTCs to enable longitudinal monitoring of the tumor's adaptive response to therapy. However, due to their extreme rarity, genotyping CTCs has proved challenging. Relevant mutations can be masked by leukocyte contamination in isolates. Heterogeneity between subpopulations of tumor cells poses an additional obstacle. Recent advances in single-cell sequencing can overcome these limitations but isolation of single CTCs is prone to cell loss and is prohibitively difficult and time consuming. To address these limitations, we developed a single cell sample preparation and genome sequencing pipeline that combines biophysical enrichment and single cell isolation using laser capture microdissection (LCM). A key component of this process is the encapsulation of enriched CTC sample in a hydrogel matrix, which enhances the efficiency of single-cell isolation by LCM, and is compatible with downstream sequencing. We validated this process by sequencing of single CTCs and cell free DNA (cfDNA) from a single patient with castration resistant prostate cancer. Identical mutations were observed in prostate cancer driver genes (TP53, PTEN, FOXA1) in both single CTCs and cfDNA. However, two independently isolated CTCs also had identical missense mutations in the genes for ATR serine/threonine kinase, KMT2C histone methyltransferase, and FANCC DNA damage repair gene. These mutations may be missed by bulk sequencing libraries, whereas single cell sequencing could potentially enable the characterization of key CTC subpopulations that arise during metastasis.
Collapse
Affiliation(s)
- Emily S Park
- Department of Mechanical Engineering, University of British Columbia, Canada.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|