51
|
Litti L, Colusso A, Pinto M, Ruli E, Scarsi A, Ventura L, Toffoli G, Colombatti M, Fracasso G, Meneghetti M. SERRS multiplexing with multivalent nanostructures for the identification and enumeration of epithelial and mesenchymal cells. Sci Rep 2020; 10:15805. [PMID: 32978492 PMCID: PMC7519640 DOI: 10.1038/s41598-020-72911-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/08/2020] [Indexed: 12/21/2022] Open
Abstract
Liquid biopsy represents a new frontier of cancer diagnosis and prognosis, which allows the isolation of tumor cells released in the blood stream. The extremely low abundance of these cells needs appropriate methodologies for their identification and enumeration. Herein we present a new protocol based on surface enhanced resonance Raman scattering (SERRS) gold multivalent nanostructures to identify and enumerate tumor cells with epithelial and mesenchimal markers. The validation of the protocol is obtained with spiked samples of peripheral blood mononuclear cells (PBMC). Gold nanostructures are functionalized with SERRS labels and with antibodies to link the tumor cells. Three types of such nanosystems were simultaneously used and the protocol allows obtaining the identification of all individual tumor cells with the help of a Random Forest ensemble learning method.
Collapse
Affiliation(s)
- Lucio Litti
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy
| | - Andrea Colusso
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy
| | - Marcella Pinto
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy
| | - Erlis Ruli
- Department of Statistical Sciences, University of Padova, via Battisti 241, 35121, Padua, Italy
| | - Alessia Scarsi
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy
| | - Laura Ventura
- Department of Statistical Sciences, University of Padova, via Battisti 241, 35121, Padua, Italy
| | - Giuseppe Toffoli
- SOC Farmacologia Sperimentale e Clinica, Centro di Riferimento Oncologico, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Marco Colombatti
- Department of Medicine, University of Verona, P.le L.A. Scuro, 37134, Verona, Italy
| | - Giulio Fracasso
- Department of Medicine, University of Verona, P.le L.A. Scuro, 37134, Verona, Italy.
| | - Moreno Meneghetti
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy.
| |
Collapse
|
52
|
Jian X, Xu J, Yang L, Zhao C, Xu J, Gao Z, Song YY. Intracellular Metal-Organic Frameworks: Integrating an All-In-One Semiconductor Electrode Chip for Therapy, Capture, and Quantification of Circulating Tumor Cells. Anal Chem 2020; 92:13319-13326. [PMID: 32897047 DOI: 10.1021/acs.analchem.0c02618] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Capture, analysis, and inactivation of circulating tumor cells (CTCs) have emerged as important issues for the early diagnosis and therapy of cancer. In this study, an all-in-one sensing device was developed by integrating magnetic metal-organic framework (magMOF) nanoparticles (NPs) and TiO2 nanotube arrays (TiNTs). The magMOF NPs are composed of a magnetic Fe3O4 core and a MIL-100(Fe) shell, which is loaded with glucose oxidase (GOD) and provides an intensive starvation therapy by catalyzing the consumption of cellular nutrients, thus accelerating the generation of intracellular iron ions by MIL-100(Fe) dissolution. Importantly, these iron ions not only lead to an intensive Fenton-like reaction but also establish an excellent correlation of electrochemical intensities with cancer cell numbers. Owing to the intracellular magMOF NPs, the CTCs were magnetically collected onto TiNTs. The exogenous ·OH radicals generated by TiNT photocatalysis trigger iron ions to be rapidly released out and subsequently detected via differential pulse voltammetry using TiNTs as the electrode. An excellent correlation of differential pulse voltammetry intensities with CTC numbers is obtained from 2 to 5000 cell mL-1. This nanoplatform not only paves a way to combine starvation therapy agents with Fenton-like reaction for chemodynamic therapy but also opens up new insights into the construction of all-in-one chips for CTC capture and diagnosis.
Collapse
Affiliation(s)
- Xiaoxia Jian
- College of Science, Northeastern University, Shenyang 110004, China
| | - Jing Xu
- College of Science, Northeastern University, Shenyang 110004, China
| | - Lingling Yang
- College of Science, Northeastern University, Shenyang 110004, China
| | - Chenxi Zhao
- College of Science, Northeastern University, Shenyang 110004, China
| | - Jingwen Xu
- College of Science, Northeastern University, Shenyang 110004, China
| | - Zhida Gao
- College of Science, Northeastern University, Shenyang 110004, China
| | - Yan-Yan Song
- College of Science, Northeastern University, Shenyang 110004, China
| |
Collapse
|
53
|
Park JE, Oh N, Nam H, Park JH, Kim S, Jeon JS, Yang M. Efficient Capture and Raman Analysis of Circulating Tumor Cells by Nano-Undulated AgNPs-rGO Composite SERS Substrates. SENSORS (BASEL, SWITZERLAND) 2020; 20:E5089. [PMID: 32906807 PMCID: PMC7570931 DOI: 10.3390/s20185089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/02/2020] [Accepted: 09/05/2020] [Indexed: 12/12/2022]
Abstract
The analysis of circulating tumor cells (CTCs) in the peripheral blood of cancer patients is critical in clinical research for further investigation of tumor progression and metastasis. In this study, we present a novel surface-enhanced Raman scattering (SERS) substrate for the efficient capture and characterization of cancer cells using silver nanoparticles-reduced graphene oxide (AgNPs-rGO) composites. A pulsed laser reduction of silver nanowire-graphene oxide (AgNW-GO) mixture films induces hot-spot formations among AgNPs and artificial biointerfaces consisting of rGOs. We also use in situ electric field-assisted fabrication methods to enhance the roughness of the SERS substrate. The AgNW-GO mixture films, well suited for the proposed process due to its inherent electrophoretic motion, is adjusted between indium tin oxide (ITO) transparent electrodes and the nano-undulated surface is generated by applying direct-current (DC) electric fields during the laser process. As a result, MCF7 breast cancer cells are efficiently captured on the AgNPs-rGO substrates, about four times higher than the AgNWs-GO films, and the captured living cells are successfully analyzed by SERS spectroscopy. Our newly designed bifunctional substrate can be applied as an effective system for the capture and characterization of CTCs.
Collapse
Affiliation(s)
- Jong-Eun Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
| | - Nuri Oh
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (N.O.); (J.-H.P.)
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hyeono Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (N.O.); (J.-H.P.)
| | - Sanha Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
| | - Jessie S. Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
| | - Minyang Yang
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; (J.-E.P.); (H.N.); (S.K.)
- Department of Mechanical Engineering, State University of New York Korea, Incheon 21985, Korea
| |
Collapse
|
54
|
Cheng J, Liu Y, Zhao Y, Zhang L, Zhang L, Mao H, Huang C. Nanotechnology-Assisted Isolation and Analysis of Circulating Tumor Cells on Microfluidic Devices. MICROMACHINES 2020; 11:E774. [PMID: 32823926 PMCID: PMC7465711 DOI: 10.3390/mi11080774] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022]
Abstract
Circulating tumor cells (CTCs), a type of cancer cell that spreads from primary tumors into human peripheral blood and are considered as a new biomarker of cancer liquid biopsy. It provides the direction for understanding the biology of cancer metastasis and progression. Isolation and analysis of CTCs offer the possibility for early cancer detection and dynamic prognosis monitoring. The extremely low quantity and high heterogeneity of CTCs are the major challenges for the application of CTCs in liquid biopsy. There have been significant research endeavors to develop efficient and reliable approaches to CTC isolation and analysis in the past few decades. With the advancement of microfabrication and nanomaterials, a variety of approaches have now emerged for CTC isolation and analysis on microfluidic platforms combined with nanotechnology. These new approaches show advantages in terms of cell capture efficiency, purity, detection sensitivity and specificity. This review focuses on recent progress in the field of nanotechnology-assisted microfluidics for CTC isolation and detection. Firstly, CTC isolation approaches using nanomaterial-based microfluidic devices are summarized and discussed. The different strategies for CTC release from the devices are specifically outlined. In addition, existing nanotechnology-assisted methods for CTC downstream analysis are summarized. Some perspectives are discussed on the challenges of current methods for CTC studies and promising research directions.
Collapse
Affiliation(s)
- Jie Cheng
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Liu
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Lina Zhang
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China;
| | - Lingqian Zhang
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Haiyang Mao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Chengjun Huang
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
55
|
Recognition and sensitive detection of CTCs using a controllable label-free electrochemical cytosensor. Mikrochim Acta 2020; 187:487. [PMID: 32761498 DOI: 10.1007/s00604-020-04452-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 07/12/2020] [Indexed: 02/06/2023]
Abstract
An excellent atomic layer deposition (ALD) method was adopted for the controllable systhesis of a xFe2O3-nPt (or nPt-xFe2O3)-coated graphene nanostructure (xFe2O3-nPt@graphene). The produced nanomaterials have been characterized by transmission electron microscopy (TEM), cyclic voltammetry (CV), and X-ray photoelectron spectroscopy (XPS). It is shown that xFe2O3 and nPt were effectively tailored and deposited on the graphene. A simple, rapid, and sensitive electrochemical cytosensor based on the controllable nanomaterials was successfully developed for MCF-7 cells detection by combining the high affinity and specificity of an aptamer. The prepared cytosensor displays a linear response to MCF-7 in the concentration range 18 to 1.5 × 106 cell mL-1 with the detection limit of 6 cell mL-1 (at an S/N of 3). This cytosensor was applied to detect circulating tumor cells (CTCs) in patient blood and the results were satisfied. The experimental results indicate that the proposed controllable electrochemical cytosensor is highly-sensitive, and convenient for clinical detection of breast CTCs. Graphical abstract.
Collapse
|
56
|
Surface Imprinted Layer of Cypermethrin upon Au Nanoparticle as a Specific and Selective Coating for the Detection of Template Pesticide Molecules. COATINGS 2020. [DOI: 10.3390/coatings10080751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The detection of specific pesticides on food products is essential as these substances pose health risks due to their toxicity. The use of surface-enhanced Raman spectroscopy (SERS) takes advantage of the straightforward technique to obtain fingerprint spectra of target analytes. In this study, SERS-active substrates are made using Au nanoparticles (NPs) coated with a layer of polymer and followed by imprinting with a pesticide–Cypermethrin, as a molecularly imprinted polymer (MIP). Cypermethrin was eventually removed and formed as template cavities, then denoted as Au NP/MIP, to capture the analogous molecules. The captured molecules situated in-between the areas of high electromagnetic field formed by plasmonic Au NPs result in an effect of SERS. The formation of Au NP/MIP was, respectively, studied through morphological analysis using transmission electron microscopy (TEM) and compositional analysis using X-ray photoelectron spectroscopy (XPS). Two relatively similar pesticides, Cypermethrin and Permethrin, were used as analytes. The results showed that Au NP/MIP was competent to detect both similar molecules despite the imprint being made only by Cypermethrin. Nevertheless, Au NP/MIP has a limited number of imprinted cavities that result in sensing only low concentrations of a pesticide solution. Au NP/MIP is thus a specific design for detecting analogous molecules similar to its template structure.
Collapse
|
57
|
Weth A, Krol I, Priesner K, Donato C, Pirker S, Wolf C, Aceto N, Baumgartner W. A novel device for elimination of cancer cells from blood specimens. Sci Rep 2020; 10:10181. [PMID: 32576883 PMCID: PMC7311454 DOI: 10.1038/s41598-020-67071-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/29/2020] [Indexed: 11/09/2022] Open
Abstract
Circulating tumor cells (CTCs) are derivatives of solid cancerous lesions that detach from the tumor mass and enter the blood circulation. CTCs are considered to be the precursors of metastasis in several cancer types. They are present in the blood of cancer patients as single cells or clusters, with the latter being associated with a higher metastatic potential. Methods to eliminate CTCs from the bloodstream are currently lacking. Here, we took advantage of the lower shear stress-resistance of cancer cells compared to blood cells, and developed a device that can eliminate cancer cells without blood damage. The device consists of an axial pump and a coupled rotating throttle, controllable to prevent local blood flow impairment, yet maintaining a constant shear performance. When processing cancer cells through our device, we observe cancer cell-cluster disruption and viability reduction of single cancer cells, without noticeable effects on human blood cells. When injecting cancer cell-containing samples into tumor-free recipient mice, processed samples fail to generate metastasis. Together, our data show that a selective disruption of cancer cells is possible while preserving blood cells, paving the way towards the development of novel, implantable tools for CTC disruption and metastasis prevention.
Collapse
Affiliation(s)
- Agnes Weth
- Institute of Biomedical Mechatronics, Johannes Kepler University of Linz, Altenbergerstr. 69, 4040, Linz, Austria
| | - Ilona Krol
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058, Basel, Switzerland
| | - Kurt Priesner
- Griesmühle Ltd, Griesmühlweg 14, 4111, Walding, Austria
| | - Cinzia Donato
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058, Basel, Switzerland
| | - Stefan Pirker
- Department of Particulate Flow Modelling, Johannes Kepler University of Linz, Altenbergerstr. 69, 4040, Linz, Austria
| | - Christoph Wolf
- Institute of Biomedical Mechatronics, Johannes Kepler University of Linz, Altenbergerstr. 69, 4040, Linz, Austria
| | - Nicola Aceto
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058, Basel, Switzerland.
| | - Werner Baumgartner
- Institute of Biomedical Mechatronics, Johannes Kepler University of Linz, Altenbergerstr. 69, 4040, Linz, Austria.
| |
Collapse
|
58
|
Lei KF. A Review on Microdevices for Isolating Circulating Tumor Cells. MICROMACHINES 2020; 11:E531. [PMID: 32456042 PMCID: PMC7281722 DOI: 10.3390/mi11050531] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/12/2020] [Accepted: 05/20/2020] [Indexed: 01/17/2023]
Abstract
Cancer metastasis is the primary cause of high mortality of cancer patients. Enumeration of circulating tumor cells (CTCs) in the bloodstream is a very important indicator to estimate the therapeutic outcome in various metastatic cancers. The aim of this article is to review recent developments on the CTC isolation technologies in microdevices. Based on the categories of biochemical and biophysical isolation approaches, a literature review and in-depth discussion will be included to provide an overview of this challenging topic. The current excellent developments suggest promising CTC isolation methods in order to establish a precise indicator of the therapeutic outcome of cancer patients.
Collapse
Affiliation(s)
- Kin Fong Lei
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan 333, Taiwan; ; Tel.: +886-3-2118800 (ext. 5345)
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou 333, Taiwan
| |
Collapse
|
59
|
Yang J, Li X, Jiang B, Yuan R, Xiang Y. In Situ-Generated Multivalent Aptamer Network for Efficient Capture and Sensitive Electrochemical Detection of Circulating Tumor Cells in Whole Blood. Anal Chem 2020; 92:7893-7899. [PMID: 32338500 DOI: 10.1021/acs.analchem.0c01195] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Monitoring circulating tumor cells (CTCs) in human blood can offer useful information for convenient metastasis diagnosis, prognosis, and treatment of cancers. However, it remains a substantial challenge to detect CTCs because of their particular scarcity in complex peripheral blood. Herein, we describe an in situ-generated multivalent aptamer network-modified electrode interface for efficiently capturing and sensitively detecting CTCs in whole blood by electrochemistry. Such an interface was fabricated via rolling circle amplification extension of the electrode-immobilized primer/circular DNA complexes for the yield of long ssDNA strands with many repeated aptamer segments, which could achieve efficient capture of rare CTCs in a multivalent cooperative manner. The antibody and horseradish peroxidase-functionalized gold nanoparticles further specifically associated with the surface-bound CTCs and generated electrocatalytically amplified current outputs for highly sensitive detection of CTCs with an attractive detection limit of five cells. Also, the multivalent aptamer network interface could successfully distinguish the target cells from other control cells and achieve CTC detection in whole blood, demonstrating its promising potential for monitoring different rare CTCs in human blood.
Collapse
Affiliation(s)
- Jianmei Yang
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China
| | - Xiaolong Li
- School of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Bingying Jiang
- School of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Ruo Yuan
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China
| | - Yun Xiang
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China
| |
Collapse
|
60
|
Ding P, Wang Z, Wu Z, Zhou Y, Sun N, Pei R. Natural Biointerface Based on Cancer Cell Membranes for Specific Capture and Release of Circulating Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2020; 12:20263-20270. [PMID: 32259427 DOI: 10.1021/acsami.0c03355] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Circulating tumor cells (CTCs) are an important part of liquid biopsy as they represent a potentially rich source of information for cancer diagnosis, monitoring, prognosis, and treatment guidance. It has been proved that the nanotopography interaction between cells and the surface of CTC detection platforms can significantly improve the capture efficiency of CTCs, whereas many mature nanostructure substrates have been developed based on chemistry materials. In this work, a natural biointerface with unique biological properties is fabricated for efficient isolation and nondestructive release of CTCs from blood samples using the cancer cell membranes. The cell membrane interfaces are proved to have a good antiadhesion property for nonspecific cells because of their own electronegativity. A natural surface nanostructure is provided by the cancer cell membrane to nicely match with the surface nanotopography of CTCs. Bovine serum albumin (BSA) as a linker and DNA aptamer against the epithelial cell adhesion molecule (EpCAM) as a specific affinity molecule are then introduced onto the cell membrane interfaces to achieve the highly efficient and specific capture of CTCs. Finally, the captured target cells can be intactly released from the substrate using the complementary DNA sequence with controlling the incubation time. This study provides a smart strategy in the development of a natural biological interface for the isolation and release of CTCs with high purity.
Collapse
Affiliation(s)
- Pi Ding
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhili Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zeen Wu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Youxin Zhou
- The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Na Sun
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
61
|
Facile synthesis of 3D hierarchical micro-/nanostructures in capillaries for efficient capture of circulating tumor cells. J Colloid Interface Sci 2020; 575:108-118. [PMID: 32361043 DOI: 10.1016/j.jcis.2020.04.087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 01/28/2023]
Abstract
The efficient capture of rare circulating tumor cells (CTCs) with high viability is of great importance in cancer diagnosis. The integration of three-dimensional (3D) nanobiointerfaces with capillary flow channel platforms can efficiently improve CTC capture performance by providing abundant binding sites and increasing the likelihood of contact as samples flow through the microchannels. However, due to the complex preparation processes, facile synthesis of nanostructures for use as substrates in flow channels for biomedical applications is still challenging. To reduce the encapsulation steps in the fabricating of nanostructured flow channel devices, we chose the enclosed glass capillaries as flow channels and accomplished all the experiments in the microchannels, including 3D nanostructure synthesis, surface modification and capture/release of CTCs. In this work, we constructed a novel 3D Zn(OH)F/ZnO nanoforest array in capillaries for CTC isolation via a facile microfluidic wet chemistry method. Because of the abundant binding sites of the 3D Zn(OH)F/ZnO nanoforest array, the capture efficiency was remarkably enhanced compared with that of vertical nanowires (90.3% vs 69.1%). In addition, a high release efficiency and cell viability of released cells were achieved by grafting poly(N-isopropylacrylamide) (PNIPPAm). These results may provide evidence for a novel method to fabricate hierarchical 3D substrates with a combination of biomolecule recognition and topographical interaction for biomedical applications.
Collapse
|
62
|
Electrochemical biosensor for the epithelial cancer biomarker EpCAM based on reduced graphene oxide modified with nanostructured titanium dioxide. Mikrochim Acta 2020; 187:275. [DOI: 10.1007/s00604-020-04233-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 03/23/2020] [Indexed: 01/23/2023]
|
63
|
Safety assessment control on mouse fibroblast cells compared with various chemically synthesized graphene oxide nanocomposites. CHEMICAL PAPERS 2020. [DOI: 10.1007/s11696-020-01133-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
64
|
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: 12] [Impact Index Per Article: 2.4] [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
|
65
|
Lin Z, Luo G, Du W, Kong T, Liu C, Liu Z. Recent Advances in Microfluidic Platforms Applied in Cancer Metastasis: Circulating Tumor Cells' (CTCs) Isolation and Tumor-On-A-Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1903899. [PMID: 31747120 DOI: 10.1002/smll.201903899] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/13/2019] [Indexed: 05/03/2023]
Abstract
Cancer remains the leading cause of death worldwide despite the enormous efforts that are made in the development of cancer biology and anticancer therapeutic treatment. Furthermore, recent studies in oncology have focused on the complex cancer metastatic process as metastatic disease contributes to more than 90% of tumor-related death. In the metastatic process, isolation and analysis of circulating tumor cells (CTCs) play a vital role in diagnosis and prognosis of cancer patients at an early stage. To obtain relevant information on cancer metastasis and progression from CTCs, reliable approaches are required for CTC detection and isolation. Additionally, experimental platforms mimicking the tumor microenvironment in vitro give a better understanding of the metastatic microenvironment and antimetastatic drugs' screening. With the advancement of microfabrication and rapid prototyping, microfluidic techniques are now increasingly being exploited to study cancer metastasis as they allow precise control of fluids in small volume and rapid sample processing at relatively low cost and with high sensitivity. Recent advancements in microfluidic platforms utilized in various methods for CTCs' isolation and tumor models recapitulating the metastatic microenvironment (tumor-on-a-chip) are comprehensively reviewed. Future perspectives on microfluidics for cancer metastasis are proposed.
Collapse
Affiliation(s)
- Zhengjie Lin
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Guanyi Luo
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Weixiang Du
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Tiantian Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Changkun Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Zhou Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
66
|
Siemer S, Wünsch D, Khamis A, Lu Q, Scherberich A, Filippi M, Krafft MP, Hagemann J, Weiss C, Ding GB, Stauber RH, Gribko A. Nano Meets Micro-Translational Nanotechnology in Medicine: Nano-Based Applications for Early Tumor Detection and Therapy. NANOMATERIALS 2020; 10:nano10020383. [PMID: 32098406 PMCID: PMC7075286 DOI: 10.3390/nano10020383] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/03/2020] [Accepted: 02/15/2020] [Indexed: 02/07/2023]
Abstract
Nanomaterials have great potential for the prevention and treatment of cancer. Circulating tumor cells (CTCs) are cancer cells of solid tumor origin entering the peripheral blood after detachment from a primary tumor. The occurrence and circulation of CTCs are accepted as a prerequisite for the formation of metastases, which is the major cause of cancer-associated deaths. Due to their clinical significance CTCs are intensively discussed to be used as liquid biopsy for early diagnosis and prognosis of cancer. However, there are substantial challenges for the clinical use of CTCs based on their extreme rarity and heterogeneous biology. Therefore, methods for effective isolation and detection of CTCs are urgently needed. With the rapid development of nanotechnology and its wide applications in the biomedical field, researchers have designed various nano-sized systems with the capability of CTCs detection, isolation, and CTCs-targeted cancer therapy. In the present review, we summarize the underlying mechanisms of CTC-associated tumor metastasis, and give detailed information about the unique properties of CTCs that can be harnessed for their effective analytical detection and enrichment. Furthermore, we want to give an overview of representative nano-systems for CTC isolation, and highlight recent achievements in microfluidics and lab-on-a-chip technologies. We also emphasize the recent advances in nano-based CTCs-targeted cancer therapy. We conclude by critically discussing recent CTC-based nano-systems with high therapeutic and diagnostic potential as well as their biocompatibility as a practical example of applied nanotechnology.
Collapse
Affiliation(s)
- Svenja Siemer
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Désirée Wünsch
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Aya Khamis
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Qiang Lu
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Arnaud Scherberich
- Laboratory of Tissue Engineering, Universitätspital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland (M.F.)
| | - Miriam Filippi
- Laboratory of Tissue Engineering, Universitätspital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland (M.F.)
| | - Marie Pierre Krafft
- Institut Charles Sadron (CNRS), University of Strasbourg, 23 rue du Loess, 67034 Strasbourg Cedex, France
| | - Jan Hagemann
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Carsten Weiss
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Postfach 3640, 76021 Karlsruhe, Germany
| | - Guo-Bin Ding
- Institute for Biotechnology, Shanxi University, No. 92 Wucheng Road, 030006 Taiyuan, China
| | - Roland H. Stauber
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
- Institute for Biotechnology, Shanxi University, No. 92 Wucheng Road, 030006 Taiyuan, China
- Correspondence: (R.H.S.); (A.G.); Tel.: +49-6131-176030 (A.G.)
| | - Alena Gribko
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
- Correspondence: (R.H.S.); (A.G.); Tel.: +49-6131-176030 (A.G.)
| |
Collapse
|
67
|
Wang X, Wang X, Cheng S, Ye M, Zhang C, Xian Y. Near-Infrared Light-Switched MoS 2 Nanoflakes@Gelatin Bioplatform for Capture, Detection, and Nondestructive Release of Circulating Tumor Cells. Anal Chem 2020; 92:3111-3117. [PMID: 31968939 DOI: 10.1021/acs.analchem.9b04724] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The integrative bioplatform for capture, detection and release of circulating tumor cells (CTCs) is of great significance in clinical diagnosis and biomedical research. To fulfill this demand, we introduced a near-infrared (NIR) light-switched bioplatform for efficient isolation and downstream analysis of CTCs. The platform was created by first modifying the PEG-MoS2 nanoflakes (NFs)@gelatin nanocomposite on the ITO surface, and then introducing the MUC1 aptamer as a specific recognition element via coupling reaction between aptamer and gelatin to achieve the specific capture for CTCs. Subsequently, the captured cells are released under a NIR light irradiation (808 nm) by using MoS2 NFs as the NIR-regulated control element. Significantly, this platform could capture and release of CTCs with an excellent capture/release efficiency of 89.5% and 92.5%, respectively. Furthermore, the electrochemical bioplatform exhibited a wide linear range for the detection of CTCs from 50 to 1 × 106 cells mL-1 with a detection limit of 15 cells mL-1. After 5 days of reculture, the released cells still maintain good cell shape and proliferation capacity. Moreover, the bioplatfrom is a simple, versatile, and universal system for the recognition, capture, release, and detection of different types of CTCs. Therefore, this bioplatform shows potential applications on the early diagnosis of cancers.
Collapse
Affiliation(s)
- Xiuli Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| | - Xinjun Wang
- Shanghai Zhangjiang Institute of Medical Innovation , Shanghai 201204 , China
| | - Shasha Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| | - Mingqiang Ye
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| | - Cuiling Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| | - Yuezhong Xian
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| |
Collapse
|
68
|
Cui H, Liu Q, Li R, Wei X, Sun Y, Wang Z, Zhang L, Zhao XZ, Hua B, Guo SS. ZnO nanowire-integrated bio-microchips for specific capture and non-destructive release of circulating tumor cells. NANOSCALE 2020; 12:1455-1463. [PMID: 31808771 DOI: 10.1039/c9nr07349c] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Circulating tumor cells (CTCs) are one type of significant biomarker in cancer patients' blood that have been attracting attention from researchers for decades, and their efficient and viable isolation is of vital importance in cancer prevention and treatment. However, the development of efficient and low-cost bio-microchips still faces significant challenges. In this paper, we construct a novel three-dimensional micro-nano bio-microchip that has dual functions of specifically capturing and non-destructively releasing cancer cells. ZnO nanowire arrays were vertically grown on the surface of a polydimethylsiloxane (PDMS) pillar substrate with a gear structure (ZnO-coated G-PDMS pillar microchips). The gear structure provides more binding sites for antibodies and target cancer cells, while ZnO nanowires provide a rough surface for CTC attachment and size-specific effects for retaining CTCs. For subsequent culture and bioanalysis, the captured CTCs can be non-destructively released with high efficiency and good viability using a mild acidic solution treatment. Furthermore, the manufacturing process of the G-PDMS pillar microchips is convenient and low-cost, and the preparation approach of the ZnO nanowire is mature and simple to operate. In particular, the bio-microchips showed high capture efficiency (91.11% ± 5.53%) and excellent cell viability (96%) using a spiked cell sample. Moreover, we successfully achieved the specific fluorescent labeling of CTCs in 9 clinical breast cancer patients' samples. The ZnO-coated G-PDMS pillar microchips not only have great potential for new target drug development for cancer stem cells but also open up new opportunities for individualized treatment.
Collapse
Affiliation(s)
- Heng Cui
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, Hubei, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Xiao Y, Lin L, Shen M, Shi X. Design of DNA Aptamer-Functionalized Magnetic Short Nanofibers for Efficient Capture and Release of Circulating Tumor Cells. Bioconjug Chem 2020; 31:130-138. [PMID: 31855600 DOI: 10.1021/acs.bioconjchem.9b00816] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The isolation of viable circulating tumor cells (CTCs) from blood is of paramount significance for early stage detection and individualized therapy of cancer. Currently, CTCs isolated by conventional magnetic separation methods are tightly coated with magnetic materials even after attempted coating removal treatments, which is not conducive for subsequent analysis of CTCs. Herein, we developed DNA aptamer-functionalized magnetic short nanofibers (aptamer-MSNFs) for efficient capture and release of CTCs. In our work, polyethylenimine (PEI)-stabilized Fe3O4 nanoparticles with a mean diameter of 22.6 nm were first synthesized and encapsulated within PEI/poly(vinyl alcohol) nanofibers via a blended electrospinning process. After a homogenization treatment to acquire the MSNFs, surface conjugation of the DNA aptamer was performed through thiol-maleimide coupling. The formed aptamer-MSNFs, with a mean diameter of 350 nm and an average length of 9.6 μm, display a saturated magnetization of 12.3 emu g-1, are capable of specifically capturing cancer cells with an efficiency of 87%, and enable the nondestructive release of cancer cells with a release efficiency of 91% after nuclease treatment. In particular, the prepared aptamer-MSNFs displayed a significantly higher release efficiency than commercial magnetic beads. The designed aptamer-MSNFs may hold great promise for CTC capture and release as well as for other cell sorting applications.
Collapse
Affiliation(s)
- Yunchao Xiao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology , Donghua University , Shanghai 201620 , P. R. China
| | - Lizhou Lin
- Department of Ultrasound, Shanghai General Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200080 , P. R. China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology , Donghua University , Shanghai 201620 , P. R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology , Donghua University , Shanghai 201620 , P. R. China
| |
Collapse
|
70
|
|
71
|
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: 59] [Impact Index Per Article: 11.8] [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
|
72
|
Zhou Y, Dong Z, Andarge H, Li W, Pappas D. Nanoparticle modification of microfluidic cell separation for cancer cell detection and isolation. Analyst 2020; 145:257-267. [DOI: 10.1039/c9an01719d] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We present a nanoparticle surface modification approach to improve the microfluidic performance in detecting cancer cells. Multiple cancer cell lines were included in this work, and the capture ability of the chip with surface modification reached a significant increase.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Chemistry and Biochemistry
- Texas Tech University
- Lubbock
- USA
| | - Ziye Dong
- Department of Chemical Engineering
- Texas Tech University
- Lubbock
- USA
| | - Hermella Andarge
- Department of Chemistry and Biochemistry
- Texas Tech University
- Lubbock
- USA
| | - Wei Li
- Department of Chemical Engineering
- Texas Tech University
- Lubbock
- USA
| | - Dimitri Pappas
- Department of Chemistry and Biochemistry
- Texas Tech University
- Lubbock
- USA
| |
Collapse
|
73
|
Pallares RM, Thanh NTK, Su X. Sensing of circulating cancer biomarkers with metal nanoparticles. NANOSCALE 2019; 11:22152-22171. [PMID: 31555790 DOI: 10.1039/c9nr03040a] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The analysis of circulating cancer biomarkers, including cell-free and circulating tumor DNA, circulating tumor cells, microRNA and exosomes, holds promise in revolutionizing cancer diagnosis and prognosis using body fluid analysis, also known as liquid biopsy. To enable clinical application of these biomarkers, new analytical tools capable of detecting them in very low concentrations in complex sample matrixes are needed. Metal nanoparticles have emerged as extraordinary analytical scaffolds because of their unique optoelectronic properties and ease of functionalization. Hence, multiple analytical techniques have been developed based on these nanoparticles and their plasmonic properties. The aim of this review is to summarize and discuss the present development on the use of metal nanoparticles for the analysis of circulating cancer biomarkers. We examine how metal nanoparticles can be used as (1) analytical transducers in various sensing principles, such as aggregation induced colorimetric assays, plasmon resonance energy transfer, surface enhanced Raman spectroscopy, and refractive index sensing, and (2) signal amplification elements in surface plasmon resonance spectroscopy and electrochemical detection. We critically discuss the clinical relevance of each category of circulating biomarkers, followed by a thorough analysis of how these nanoparticle-based designs have overcome some of the main challenges that gold standard analytical techniques currently face, and what new directions the field may take in the future.
Collapse
Affiliation(s)
- Roger M Pallares
- Biophysics Group, Department of Physics and Astronomy, University College London, London, WC1E 6BT, UK.
| | | | | |
Collapse
|
74
|
Kanioura A, Petrou P, Kletsas D, Tserepi A, Chatzichristidi M, Gogolides E, Kakabakos S. Three-dimensional (3D) hierarchical oxygen plasma micro/nanostructured polymeric substrates for selective enrichment of cancer cells from mixtures with normal ones. Colloids Surf B Biointerfaces 2019; 187:110675. [PMID: 31810566 DOI: 10.1016/j.colsurfb.2019.110675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 10/16/2019] [Accepted: 11/25/2019] [Indexed: 12/30/2022]
Abstract
The enrichment of cancer cell population when in mixtures with normal ones is of great importance for cancer diagnosis. In this work, poly(methyl methacrylate) films have been processed applying different oxygen plasma conditions to fabricate surfaces with structure height ranging from 22 to more than 2000 nm. The surfaces were then evaluated with respect to adhesion and proliferation of both normal and cancer human cells. In particular, normal skin and lung fibroblasts, and four different cancer cell lines, A431 (skin cancer), HT1080 (fibrosarcoma), A549 (lung cancer), and PC3 (prostate cancer), have been employed. It was found that adhesion and proliferation of cancer cells was favored when cultured onto the hierarchical micro/nanostructured surfaces as compared to untreated ones with the maximum values obtained for substrates treated at -100 V for 3 min. On the other hand, although the adhesion of normal fibroblasts was not influenced by the micro/nanostructured surfaces, their morphology and proliferation was significantly impaired, especially after 3-day culture on these surfaces. The reduced proliferation rate of adherent fibroblasts was linked to reduced focal points formation, as it was verified through vinculin staining, and not to apoptosis. The micro/nanostructured surfaces prepared with plasma treatment at -100 V for 3 min (hierarchical topography with mean height of ∼800 nm) were selected as substrates for normal and cancer cell co-culture experiments. It was found that 25-80 times enrichment of cancer over the normal cells was achieved on the nanostructured surfaces after 3-day culture, while it was 5-8 times lower on the untreated ones. It should be noticed that this is the first time such high enrichment ratios are achieved without implementing surfaces modified with binding molecules specific for cancer cells. Thus, the nanostructured surfaces hold a strong promise as culture substrates for separation and enrichment of cancer cells from mixtures with normal ones that should find application in cancer diagnostics.
Collapse
Affiliation(s)
- Anastasia Kanioura
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | - Panagiota Petrou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | - Dimitris Kletsas
- Institute of Biosciences and Applications, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | - Angeliki Tserepi
- Institute of Nanoscience & Nanotechnology, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | | | - Evangelos Gogolides
- Institute of Nanoscience & Nanotechnology, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece
| | - Sotirios Kakabakos
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, NCSR "Demokritos", Aghia Paraskevi, 15341, Greece.
| |
Collapse
|
75
|
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
|
76
|
Beckwith KS, Ullmann S, Vinje J, Sikorski P. Influence of Nanopillar Arrays on Fibroblast Motility, Adhesion, and Migration Mechanisms. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1902514. [PMID: 31464377 DOI: 10.1002/smll.201902514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/25/2019] [Indexed: 06/10/2023]
Abstract
Surfaces decorated with high aspect ratio nanostructures are a promising tool to study cellular processes and design novel devices to control cellular behavior. However, little is known about the dynamics of cellular phenomenon such as adhesion, spreading, and migration on such surfaces. In particular, how these are influenced by the surface properties. In this work, fibroblast behavior is investigated on regular arrays of 1 µm high polymer nanopillars with varying pillar to pillar distance. Embryonic mouse fibroblasts (NIH-3T3) spread on all arrays, and on contact with the substrate engulf nanopillars independently of the array pitch. As the cells start to spread, different behavior is observed. On dense arrays which have a pitch equal or below 1 µm, cells are suspended on top of the nanopillars, making only sporadic contact with the glass support. Cells stay attached to the glass support and fully engulf nanopillars during spreading and migration on the sparse arrays which have a pitch of 2 µm and above. These alternate states have a profound effect on cell migration rates. Dynamic F-actin puncta colocalize with nanopillars during cell spreading and migration. Strong membrane association with engulfed nanopillars might explain the reduced migration rates on sparse arrays.
Collapse
Affiliation(s)
- Kai S Beckwith
- Centre of Molecular Inflammation Research, Department of Molecular and Clinical Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Sindre Ullmann
- Centre of Molecular Inflammation Research, Department of Molecular and Clinical Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Jakob Vinje
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Pawel Sikorski
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| |
Collapse
|
77
|
Xiong Z, Shen M, Shi X. Zwitterionic Modification of Nanomaterials for Improved Diagnosis of Cancer Cells. Bioconjug Chem 2019; 30:2519-2527. [DOI: 10.1021/acs.bioconjchem.9b00543] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Zhijuan Xiong
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
| |
Collapse
|
78
|
Liu H, Wang Z, Chen C, Ding P, Sun N, Pei R. Dual-antibody Modified PLGA Nanofibers for Specific Capture of Epithelial and Mesenchymal CTCs. Colloids Surf B Biointerfaces 2019; 181:143-148. [DOI: 10.1016/j.colsurfb.2019.05.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/13/2019] [Accepted: 05/14/2019] [Indexed: 12/20/2022]
|
79
|
Wang Y, Han X, Cui Z, Shi D. Bioelectricity, Its Fundamentals, Characterization Methodology, and Applications in Nano-Bioprobing and Cancer Diagnosis. ACTA ACUST UNITED AC 2019; 3:e1900101. [PMID: 32648718 DOI: 10.1002/adbi.201900101] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/01/2019] [Indexed: 12/11/2022]
Abstract
Bioelectricity is an essential characteristic of a biological system that has played an important role in medical diagnosis particularly in cancer liquid biopsy. However, its biophysical origin and measurements have presented great challenges in experimental methodologies. For instance, in dynamic cell processes, bioelectricity cannot be accurately determined as a static electrical potential via electrophoresis. Cancer cells fundamentally differ from normal cells by having a much higher rate of glycolysis resulting in net negative charges on cell surfaces. The most recent investigations on cancer cell surface charge that is the direct bio-electrical manifestation of the "Warburg Effect," which can be directly monitored by specially designed nanoprobes, has been provided. The most up-to-date research results from charge-mediated cell targeting are reviewed. Correlations between the cell surface charge and cancer cell metabolism are established based on cell/probe electrostatic interactions. Bioelectricity is utilized not only as an analyte for investigation of the metabolic state of the cancer cells, but also applied in electrostatically and magnetically capturing of the circulating tumor cells from whole blood. Also reviewed is on the isolation of Candida albicans via bioelectricity-driven nanoparticle binding on fungus with surface charges.
Collapse
Affiliation(s)
- Yilong Wang
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200092, P. R. China
| | - Xiao Han
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200092, P. R. China
| | - Zheng Cui
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200092, P. R. China.,Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Donglu Shi
- Materials Science and Engineering Program, Department of Mechanical and Materials Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, 45221, USA
| |
Collapse
|
80
|
Stelcer E, Konkol M, Głȩboka A, Suchorska WM. Liquid Biopsy in Oligometastatic Prostate Cancer-A Biologist's Point of View. Front Oncol 2019; 9:775. [PMID: 31475117 PMCID: PMC6702517 DOI: 10.3389/fonc.2019.00775] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa) is the main cause of cancer-related mortality in males and the diagnosis, treatment, and care of these patients places a great burden on healthcare systems globally. Clinically, PCa is highly heterogeneous, ranging from indolent tumors to highly aggressive disease. In many cases treatment-generally either radiotherapy (RT) or surgery-can be curative. Several key genetic and demographic factors such as age, family history, genetic susceptibility, and race are associated with a high incidence of PCa. While our understanding of PCa, which is mainly based on the tools of molecular biology-has improved dramatically in recent years, efforts to better understand this complex disease have led to the identification of a new type of PCa-oligometastatic PCa. Oligometastatic disease should be considered an individual, heterogeneous entity with distinct metastatic phenotypes and, consequently, wide prognostic variability. In general, patients with oligometastatic disease typically present less biologically aggressive tumors whose metastatic potential is more limited and which are slow-growing. These patients are good candidates for more aggressive treatment approaches. The main aim of the presented review was to evaluate the utility of liquid biopsy for diagnostic purposes in PCa and for use in monitoring disease progression and treatment response, particularly in patients with oligometastatic PCa. Liquid biopsies offer a rapid, non-invasive approach whose use t is expected to play an important role in routine clinical practice to benefit patients. However, more research is needed to resolve the many existing discrepancies with regard to the definition and isolation method for specific biomarkers, as well as the need to determine the most appropriate markers. Consequently, the current priority in this field is to standardize liquid biopsy-based techniques. This review will help to improve understanding of the biology of PCa, particularly the recently defined condition known as "oligometastatic PCa". The presented review of the body of evidence suggests that additional research in molecular biology may help to establish novel treatments for oligometastatic PCa. In the near future, the treatment of PCa will require an interdisciplinary approach involving active cooperation among clinicians, physicians, and biologists.
Collapse
Affiliation(s)
- Ewelina Stelcer
- Radiobiology Laboratory, Greater Poland Cancer Centre, Poznan, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Konkol
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
- Radiation Oncology Department, Greater Poland Cancer Centre, Poznan, Poland
| | | | - Wiktoria Maria Suchorska
- Radiobiology Laboratory, Greater Poland Cancer Centre, Poznan, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
81
|
Specific cell capture and noninvasive release via moderate electrochemical oxidation of boronic ester linkage. Biosens Bioelectron 2019; 138:111316. [DOI: 10.1016/j.bios.2019.111316] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 12/11/2022]
|
82
|
Stine KJ. Nanoporous Gold and Other Related Materials. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1080. [PMID: 31357629 PMCID: PMC6723733 DOI: 10.3390/nano9081080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 12/22/2022]
Abstract
The field of nanomaterials continues to expand with the discovery of new nanostructures opening up new possibilities for both the study of unique physical properties and new applications [...].
Collapse
Affiliation(s)
- Keith J Stine
- Department of Chemistry and Biochemistry, University of Missouri, Saint Louis, MO 63121, USA.
| |
Collapse
|
83
|
Sun D, Lu J, Zhang L, Chen Z. Aptamer-based electrochemical cytosensors for tumor cell detection in cancer diagnosis: A review. Anal Chim Acta 2019; 1082:1-17. [PMID: 31472698 DOI: 10.1016/j.aca.2019.07.054] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 01/25/2023]
Abstract
Circulating tumor cells, a type of viable cancer cell circulating from primary or metastatic tumors in the blood stream, can lead to the parallel development of primary tumors and metastatic lesions. Highly selective and sensitive detection of tumor cells has become a hot research topic and can provide a basis for early diagnosis of cancers and anticancer drug evaluation to develop the best treatment plan. Aptamers are single-stranded oligonucleotides that can bind to target tumor cells in unique three-dimensional structures with high specificity and affinity. Aptamer-based methods or signal amplification methods using aptamers show great potential in improving the selectivity and sensitivity of electrochemical (EC) cytosensors for tumor cell detection. This review covers the remarkable developments in aptamer-based EC cytosensors for the identification of cell type, cell counting and detection of crucial proteins on the cell surface. Various EC techniques have been developed for cancer cell detection, including common voltammetry or impedance, electrochemiluminescence and photoelectrochemistry in a direct approach (aptamer-target cell), sandwich approach (capture probe-target cell-signaling probe) or other approach. The current challenges and promising opportunities in the establishment of EC aptamer cytosensors for tumor cell detection are also discussed.
Collapse
Affiliation(s)
- Duanping Sun
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China; School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Jing Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Luyong Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zuanguang Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
84
|
Liu C, Yang B, Chen X, Hu Z, Dai Z, Yang D, Zheng X, She X, Liu Q. Capture and separation of circulating tumor cells using functionalized magnetic nanocomposites with simultaneous in situ chemotherapy. NANOTECHNOLOGY 2019; 30:285706. [PMID: 30849773 DOI: 10.1088/1361-6528/ab0e25] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Circulating tumor cells (CTCs) are a type of rare cell that are firstly shed from solid tumors and then exist in the bloodstream. The effective capture and separation of CTCs has significant meaning in cancer diagnosis and prognosis. In this study, novel Fe3O4-FePt magnetic nanocomposites (Fe3O4-FePt MNCs) were constructed by integrating face centered cubic (fcc) FePt nanoparticles (NPs) onto the surface of the Fe3O4@SiO2 core. After further modification with NH2-PEG-COOH and the tumor-targeting molecule tLyP-1, the acquired Fe3O4-FePt MNCs possesses excellent biocompatibility and stability and could efficiently target and capture tLyP-1 receptor-positive CTCs. Based on the acidic microenvironment within cancer cells, the FePt layer could rapidly release active Fe2+ ions, which could catalyze H2O2 into reactive oxygen species (ROS) and further induce in situ apoptosis in cancer cells while having no distinct cytotoxicity to normal cells. Moreover, the Fe3O4@SiO2 core with its intrinsic magnetism has huge potential for the bioseparation of CTCs. The in vitro ROS fluorescence imaging experiments and cell capture and separation experiments indicated that the Fe3O4-FePt MNCs could specifically capture and separate cancer cells in the CTCs model and further induce in situ apoptosis. Therefore, the Fe3O4-FePt MNCs could serve as a promising multifunctional nanoseparator for efficiently capturing CTCs and simultaneously inducing in situ chemotherapy.
Collapse
Affiliation(s)
- Chunmiao Liu
- School of Materials Science and Engineering, Qingdao University, Qingdao 266071, People's Republic of China. Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Chen M, Liu A, Chen B, Zhu DM, Xie W, Deng FF, Ji LW, Chen LB, Huang HM, Fu YR, Liu W, Wang FB. Erythrocyte-derived vesicles for circulating tumor cell capture and specific tumor imaging. NANOSCALE 2019; 11:12388-12396. [PMID: 31215952 DOI: 10.1039/c9nr01805k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The precise diagnosis of cancer remains a great challenge; therefore, it is our research interest to develop safe, tumor-specific reagents. In this study, we designed nanovesicles derived from erythrocyte membranes; the nanovesicles are capable of recognizing tumor cells for both circulating tumor cell (CTC) capture and tumor imaging. The tumor-targeting molecules folic acid (FA) and fluorescein Cy5 were modified on the nanovesicle surface. The developed nanovesicles exhibit excellent tumor targeting ability both in vitro and in vivo for CTC capture and in tumor imaging. Compared with traditional immunomagnetic beads, the proposed nanovesicles are capable of avoiding non-specific adsorption as a derivative of red blood cells. Combined with a non-invasive means of micromanipulation, the nanometer-sized vesicles show a high purity of CTC capture (over 90%). In vivo, the nanovesicles can also be employed for efficient tumor imaging without obvious toxicity and side effects. In brief, the nanovesicles prepared herein show potential clinical application for integrated diagnosis in vitro and in vivo.
Collapse
Affiliation(s)
- Ming Chen
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Ao Liu
- Huazhong Agricultural University, College of Plant Science and Technology, Wuhan, China
| | - Bei Chen
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Dao-Ming Zhu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Wei Xie
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Fang-Fang Deng
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Li-Wei Ji
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Li-Ben Chen
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Hui-Ming Huang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - You-Rong Fu
- Department of Blood Transfusion, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Fu-Bing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
86
|
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
|
87
|
Niciński K, Krajczewski J, Kudelski A, Witkowska E, Trzcińska-Danielewicz J, Girstun A, Kamińska A. Detection of circulating tumor cells in blood by shell-isolated nanoparticle - enhanced Raman spectroscopy (SHINERS) in microfluidic device. Sci Rep 2019; 9:9267. [PMID: 31239487 PMCID: PMC6592934 DOI: 10.1038/s41598-019-45629-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 06/11/2019] [Indexed: 01/22/2023] Open
Abstract
Isolation and detection of circulating tumor cells (CTCs) from human blood plays an important role in non- invasive screening of cancer evolution and in predictive therapeutic treatment. Here, we present the novel tool utilizing: (i) the microfluidic device with (ii) incorporated photovoltaic (PV) based SERS-active platform, and (iii) shell-isolated nanoparticles (SHINs) for simultaneous separation and label-free analysis of circulating tumour cells CTCs in the blood specimens with high specificity and sensitivity. The proposed microfluidic chip enables the efficient size - based inertial separation of circulating cancer cells from the whole blood samples. The SERS-active platform incorporated into the microfluidic device permits the label-free detection and identification of isolated cells through the insight into their molecular and biochemical structure. Additionally, the silver nanoparticles coated with an ultrathin shell of silica (Ag@SiO2) was used to improve the detection accuracy and sensitivity of analysed tumor cells via taking advantages of shell-isolated nanoparticle-enhanced Raman spectroscopy (SHINERS). The empirical analysis of SHINERS spectra revealed that there are some differences among studied (HeLa), renal cell carcinoma (Caki-1), and blood cells. Unique SHINERS features and differences in bands intensities between healthy and cancer cells might be associated with the variations in the quantity and quality of molecules such as lipid, protein, and DNA or their structure during the metastasis cancer formation. To demonstrate the statistical efficiency of the developed method and improve the differentiation for circulating tumors cells detection the principal component analysis (PCA) has been performed for all SHINERS data. PCA method has been applied to recognize the most significant differences in SHINERS data among the three analyzed cells: Caki-1, HeLa, and blood cells. The proposed approach challenges the current multi-steps CTCs detection methods in the terms of simplicity, sensitivity, invasiveness, destructivity, time and cost of analysis, and also prevents the defragmentation/damage of tumor cells and thus leads to improving the accuracy of analysis. The results of this research work show the potential of developed SERS based tool for the separation of tumor cells from whole blood samples in a simple and minimally invasive manner, their detection and molecular characterization using one single technology.
Collapse
Affiliation(s)
- K Niciński
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224, Warsaw, Poland
| | - J Krajczewski
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland
| | - A Kudelski
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland
| | - E Witkowska
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224, Warsaw, Poland
| | - J Trzcińska-Danielewicz
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - A Girstun
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - A Kamińska
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224, Warsaw, Poland.
| |
Collapse
|
88
|
Gribko A, Künzel J, Wünsch D, Lu Q, Nagel SM, Knauer SK, Stauber RH, Ding GB. Is small smarter? Nanomaterial-based detection and elimination of circulating tumor cells: current knowledge and perspectives. Int J Nanomedicine 2019; 14:4187-4209. [PMID: 31289440 PMCID: PMC6560927 DOI: 10.2147/ijn.s198319] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cells (CTCs) are disseminated cancer cells. The occurrence and circulation of CTCs seem key for metastasis, still the major cause of cancer-associated deaths. As such, CTCs are investigated as predictive biomarkers. However, due to their rarity and heterogeneous biology, CTCs’ practical use has not made it into the clinical routine. Clearly, methods for the effective isolation and reliable detection of CTCs are urgently needed. With the development of nanotechnology, various nanosystems for CTC isolation and enrichment and CTC-targeted cancer therapy have been designed. Here, we summarize the relationship between CTCs and tumor metastasis, and describe CTCs’ unique properties hampering their effective enrichment. We comment on nanotechnology-based systems for CTC isolation and recent achievements in microfluidics and lab-on-a-chip technologies. We discuss recent advances in CTC-targeted cancer therapy exploiting the unique properties of nanomaterials. We conclude by introducing developments in CTC-directed nanosystems and other advanced technologies currently in (pre)clinical research.
Collapse
Affiliation(s)
- Alena Gribko
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Julian Künzel
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Désirée Wünsch
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Qiang Lu
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Sophie Madeleine Nagel
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Shirley K Knauer
- Department of Molecular Biology II, Center for Medical Biotechnology (ZMB)/Center for Nanointegration (CENIDE), University Duisburg-Essen, Essen 45117, Germany
| | - Roland H Stauber
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Guo-Bin Ding
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ; .,Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, People's Republic of China,
| |
Collapse
|
89
|
Kim TH, Wang Y, Oliver CR, Thamm DH, Cooling L, Paoletti C, Smith KJ, Nagrath S, Hayes DF. A temporary indwelling intravascular aphaeretic system for in vivo enrichment of circulating tumor cells. Nat Commun 2019; 10:1478. [PMID: 30932020 PMCID: PMC6443676 DOI: 10.1038/s41467-019-09439-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 03/11/2019] [Indexed: 12/18/2022] Open
Abstract
Circulating tumor cells (CTCs) have become an established biomarker for prognosis in patients with various carcinomas. However, current ex vivo CTC isolation technologies rely on small blood volumes from a single venipuncture limiting the number of captured CTCs. This produces statistical variability and inaccurate reflection of tumor cell heterogeneity. Here, we describe an in vivo indwelling intravascular aphaeretic CTC isolation system to continuously collect CTCs directly from a peripheral vein. The system returns the remaining blood products after CTC enrichment, permitting interrogation of larger blood volumes than classic phlebotomy specimens over a prolonged period of time. The system is validated in canine models showing capability to screen 1-2% of the entire blood over 2 h. Our result shows substantial increase in CTC capture, compared with serial blood draws. This technology could potentially be used to analyze large number of CTCs to facilitate translation of analytical information into future clinical decisions.
Collapse
Affiliation(s)
- Tae Hyun Kim
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yang Wang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - C Ryan Oliver
- Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Douglas H Thamm
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Laura Cooling
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Costanza Paoletti
- Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI, 48109, USA
| | - Kaylee J Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Daniel F Hayes
- Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
90
|
Li F, Hu S, Zhang R, Gu Y, Li Y, Jia Y. Porous Graphene Oxide Enhanced Aptamer Specific Circulating-Tumor-Cell Sensing Interface on Light Addressable Potentiometric Sensor: Clinical Application and Simulation. ACS APPLIED MATERIALS & INTERFACES 2019; 11:8704-8709. [PMID: 30762335 DOI: 10.1021/acsami.8b21101] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The circulating-tumor-cell (CTC) specific aptamer is believed to be a power recognition factor to realize clinical CTC assay. However, the limited sensing range is still one of the challenges in its real application. The porous-graphene-oxide (PGO) enhanced aptamer specific CTC sensing interface is studied on the platform of light-addressable-potentiometric-sensor (LAPS) to provide a clinical available method for CTC detection. The underlying mechanism of this sensing interface on LAPS is modeled and simulated. It is confirmed to be a promising candidate for CTC assay by the linear responding for 5-5000 spiked cells, as well as the satisfactory sensitivity for clinical samples.
Collapse
Affiliation(s)
- Fang Li
- College of Electronic Information and Optical Engineering , Nankai University , Tianjin 300071 , China
| | - Shihui Hu
- College of Electronic Information and Optical Engineering , Nankai University , Tianjin 300071 , China
| | - Rong Zhang
- College of Electronic Information and Optical Engineering , Nankai University , Tianjin 300071 , China
| | - Yajun Gu
- School of Medical Laboratory , Tianjin Medical University , Tianjin 300070 , China
| | - Yueguo Li
- Department of Clinical Laboratory , Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer , Tianjin 300060 , China
| | - Yunfang Jia
- College of Electronic Information and Optical Engineering , Nankai University , Tianjin 300071 , China
| |
Collapse
|
91
|
Detection of Circulating Tumor Cells Using Membrane-Based SERS Platform: A New Diagnostic Approach for 'Liquid Biopsy'. NANOMATERIALS 2019; 9:nano9030366. [PMID: 30841516 PMCID: PMC6473992 DOI: 10.3390/nano9030366] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/18/2019] [Accepted: 02/26/2019] [Indexed: 01/05/2023]
Abstract
The detection and monitoring of circulating tumor cells (CTCs) in blood is an important strategy for early cancer evidence, analysis, monitoring of therapeutic response, and optimization of cancer therapy treatments. In this work, tailor-made membranes (MBSP) for surface-enhanced Raman spectroscopy (SERS)-based analysis, which permitted the separation and enrichment of CTCs from blood samples, were developed. A thin layer of SERS-active metals deposited on polymer mat enhanced the Raman signals of CTCs and provided further insight into CTCs molecular and biochemical composition. The SERS spectra of all studied cells—prostate cancer (PC3), cervical carcinoma (HeLa), and leucocytes as an example of healthy (normal) cell—revealed significant differences in both the band positions and/or their relative intensities. The multivariate statistical technique based on principal component analysis (PCA) was applied to identify the most significant differences (marker bands) in SERS data among the analyzed cells and to perform quantitative analysis of SERS data. Based on a developed PCA algorithm, the studied cell types were classified with an accuracy of 95% in 2D PCA to 98% in 3D PCA. These results clearly indicate the diagnostic efficiency for the discrimination between cancer and normal cells. In our approach, we exploited the one-step technology that exceeds most of the multi-stage CTCs analysis methods used and enables simultaneous filtration, enrichment, and identification of the tumor cells from blood specimens.
Collapse
|
92
|
Cho H, Kim J, Song H, Sohn KY, Jeon M, Han KH. Microfluidic technologies for circulating tumor cell isolation. Analyst 2019; 143:2936-2970. [PMID: 29796523 DOI: 10.1039/c7an01979c] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metastasis is the main cause of tumor-related death, and the dispersal of tumor cells through the circulatory system is a critical step in the metastatic process. Early detection and analysis of circulating tumor cells (CTCs) is therefore important for early diagnosis, prognosis, and effective treatment of cancer, enabling favorable clinical outcomes in cancer patients. Accurate and reliable methods for isolating and detecting CTCs are necessary to obtain this clinical information. Over the past two decades, microfluidic technologies have demonstrated great potential for isolating and detecting CTCs from blood. The present paper reviews current advanced microfluidic technologies for isolating CTCs based on various biological and physical principles, and discusses their fundamental advantages and drawbacks for subsequent cellular and molecular assays. Owing to significant genetic heterogeneity among CTCs, microfluidic technologies for isolating individual CTCs have recently been developed. We discuss these single-cell isolation methods, as well as approaches to overcoming the limitations of current microfluidic CTC isolation technologies. Finally, we provide an overview of future innovative microfluidic platforms.
Collapse
Affiliation(s)
- Hyungseok Cho
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae 621-749, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
93
|
Kozminsky M, Fouladdel S, Chung J, Wang Y, Smith DC, Alva A, Azizi E, Morgan T, Nagrath S. Detection of CTC Clusters and a Dedifferentiated RNA-Expression Survival Signature in Prostate Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801254. [PMID: 30693182 PMCID: PMC6343066 DOI: 10.1002/advs.201801254] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/19/2018] [Indexed: 05/20/2023]
Abstract
Rates of progression and treatment response in advanced prostate cancer are highly variable, necessitating non-invasive methods to assess the molecular characteristics of these tumors in real time. The unique potential of circulating tumor cells (CTCs) to serve as a clinically useful liquid biomarker is due to their ability to inform via both enumeration and RNA expression. A microfluidic graphene oxide-based device (GO Chip) is used to isolate CTCs and CTC clusters from the whole blood of 41 men with metastatic castration-resistant prostate cancer. Additionally, the expression of 96 genes of interest is determined by RT-qPCR. Multivariate analyses are conducted to determine the genes most closely associated with overall survival, PSA progression, and radioclinical progression. A preliminary signature, comprising high expression of stemness genes and low expression of epithelial and mesenchymal genes, potentially implicates an undifferentiated CTC phenotype as a marker of poor prognosis in this setting.
Collapse
Affiliation(s)
- Molly Kozminsky
- Department of Chemical EngineeringUniversity of Michigan2300 Hayward StreetAnn ArborMI48109USA
- Translational Oncology ProgramUniversity of Michigan1600 Huron PkwyAnn ArborMI48109USA
- Biointerfaces InstituteUniversity of Michigan2800 Plymouth RoadAnn ArborMI48109USA
| | - Shamileh Fouladdel
- Translational Oncology ProgramUniversity of Michigan1600 Huron PkwyAnn ArborMI48109USA
- Biointerfaces InstituteUniversity of Michigan2800 Plymouth RoadAnn ArborMI48109USA
- Department of Internal MedicineUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Jae‐Seung Chung
- Department of UrologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Yugang Wang
- Department of UrologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - David C. Smith
- Department of Internal MedicineDivision of Hematology/OncologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Ajjai Alva
- Department of Internal MedicineDivision of Hematology/OncologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Ebrahim Azizi
- Translational Oncology ProgramUniversity of Michigan1600 Huron PkwyAnn ArborMI48109USA
- Biointerfaces InstituteUniversity of Michigan2800 Plymouth RoadAnn ArborMI48109USA
- Department of Internal MedicineUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Todd Morgan
- Department of UrologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Sunitha Nagrath
- Department of Chemical EngineeringUniversity of Michigan2300 Hayward StreetAnn ArborMI48109USA
- Translational Oncology ProgramUniversity of Michigan1600 Huron PkwyAnn ArborMI48109USA
- Biointerfaces InstituteUniversity of Michigan2800 Plymouth RoadAnn ArborMI48109USA
| |
Collapse
|
94
|
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
|
95
|
Gao T, Li L, Chen T, Shi L, Yang Y, Li G. DNA-Oriented Shaping of Cell Features for the Detection of Rare Disseminated Tumor Cells. Anal Chem 2018; 91:1126-1132. [DOI: 10.1021/acs.analchem.8b04783] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Tao Gao
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, P. R. China
| | - Lingling Li
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Tianshu Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Liu Shi
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biochemistry, Nanjing University, Nanjing 210093, P. R. China
| | - Yang Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, P. R. China
| | - Genxi Li
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biochemistry, Nanjing University, Nanjing 210093, P. R. China
| |
Collapse
|
96
|
He W, Hou M, Zhang H, Zeng C, He S, Chen X, Xu M, Sun C, Jiang W, Wang H, Shen H, Zhang Y, Liu J, Sun S, Jiang N, Cui Y, Sun Y, Chen Y, Cao J, Wang C, Li M, Zhang Y, Wang L, Wang J, Lin M, Ke Z. Clinical significance of circulating tumor cells in predicting disease progression and chemotherapy resistance in patients with gestational choriocarcinoma. Int J Cancer 2018; 144:1421-1431. [PMID: 30070688 PMCID: PMC6587450 DOI: 10.1002/ijc.31742] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 05/28/2018] [Accepted: 06/20/2018] [Indexed: 12/22/2022]
Abstract
Gestational choriocarcinoma (GC) is a highly aggressive tumor. In our study, we systematically investigated EpCAM/CD147 expression characteristics in patients with GC and assessed the role of circulating tumor cells (CTCs) in predicting chemotherapy response and disease progression. GC tissues were positive for either epithelial cellular adhesion molecule (EpCAM) or CD147, and all samples exhibited strong human chorionic gonadotropin (HCG) expression. Among all the recruited patients (n = 115), 103 had at least 1 CTC in a 7.5‐mL peripheral blood sample, and the percentage of patients with ≥4 CTCs in a particular FIGO stage group increased with a higher FIGO stage (p < 0.001). Furthermore, the pretreatment CTC count was related to tumor size (r = 0.225, p = 0.015) and the number of metastases (r = 0.603, p < 0.001). A progression analysis showed that among the 115 included patients who qualified for further examination, 52 of the 64 patients defined as progressive had ≥4 pretreatment CTCs, while only 7 of the 51 non‐progressive patients had ≥4 pretreatment CTCs (p < 0.001). In multivariate analysis, CTCs (≥4) remained the strongest predictor of PFS when other prognostic markers, FIGO score and FIGO stage were included. Moreover, based on the chemotherapy response, patients with ≥4 CTCs were more likely to be resistant to chemotherapy than those with <4 CTCs (P < 0.001). These findings demonstrates the feasibility of CTC detection in cases of GC by adopting EpCAM/CD147 antibodies together as capturing antibodies. The CTC count is a promising indicator in the evaluation of biological activities and the chemotherapy response in GC patients. What's new? Gestational choriocarcinoma tumor cells tend to spread to distant organs by hematogenous dissemination. This study shows that circulating tumor cells (CTCs) in patients with gestational choriocarcinomas can be readily captured by targeting the highly expressed membrane antigens EpCAM and CD147. Elevated CTC levels, defined as 4 or more CTCs per 7.5 ml of peripheral blood, were found to predict chemotherapy resistance and to more effectively predict disease progression where compared with traditional β‐human chorionic gonadotropin. The findings suggest that CTC enumeration could be used to stratify gestational choriocarcinoma patients for personalized clinical intervention.
Collapse
Affiliation(s)
- Weiling He
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China.,Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Minzhi Hou
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China.,Department of Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Hui Zhang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Chao Zeng
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China.,Department of Pathology, Guangdong Medical College, Dongguan, Guangdong, People's Republic of China
| | - Shanyang He
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xinlin Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Manman Xu
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Cong Sun
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Wenting Jiang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Han Wang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Hongwei Shen
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yang Zhang
- Biomedical Engineering, The University of Texas at El Paso, El Paso, TX
| | - Jing Liu
- Department of Anesthesiology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Shijun Sun
- Molecular Diagnosis Center, The Affiliated Zhongshan Hospital, Sun Yat-Sen University, Zhongshan, Guangdong, People's Republic of China
| | - Neng Jiang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yongmei Cui
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yu Sun
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yangshan Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jessica Cao
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Mengzhen Li
- MyGene Diagnostics, Guangzhou International Biotech Island, Guangdong, People's Republic of China
| | - Yi Zhang
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Liantang Wang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jianhong Wang
- Pricision Medicine Center, Shenzhen People's Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Millicent Lin
- Department of Genetics, Harvard Medical School, Boston, MA
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
97
|
Kim TH, Yoon HJ, Fouladdel S, Wang Y, Kozminsky M, Burness ML, Paoletti C, Zhao L, Azizi E, Wicha MS, Nagrath S. Characterizing Circulating Tumor Cells Isolated from Metastatic Breast Cancer Patients Using Graphene Oxide Based Microfluidic Assay. ACTA ACUST UNITED AC 2018; 3:e1800278. [PMID: 32627379 DOI: 10.1002/adbi.201800278] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Indexed: 12/12/2022]
Abstract
The enumeration of circulating tumor cells (CTCs) has shown prognostic importance in patients with breast cancer. However, CTCs are highly heterogeneous with diverse functional properties, which may also be clinically relevant. To provide a comprehensive landscape of the patient's disease, further CTC analysis is required. Here, a highly sensitive and reproducible graphene oxide based CTC assay is utilized to isolate and characterize CTCs from 47 metastatic breast cancer patients. The CTCs are captured with high purity, requiring only a few milliliters of blood and enabling efficient enumeration and subsequent analysis at both the protein and the transcription level. The results show that patient clinical outcomes correlate with the associated CTC profile and clearly demonstrate the potential use of the assay in the clinical setting. Collectively, these findings suggest that beyond simple enumeration, CTC characterization may provide further information that improves the diagnosis of the patients' disease status for proper treatment decisions. Moreover, this thorough validation study will facilitate the translation of the CTC assay into future clinical applications to broaden the utility of liquid biopsy.
Collapse
Affiliation(s)
- Tae Hyun Kim
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI, 48109, USA.,Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109, USA.,Translational Oncology Program, University of Michigan, 1600 Huron Pkwy, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Hyeun Joong Yoon
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI, 48109, USA.,Translational Oncology Program, University of Michigan, 1600 Huron Pkwy, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.,Department of Electrical Engineering and Computer Science, South Dakota State University, 1175 Medary Ave, Brookings, SD, 57006, USA
| | - Shamileh Fouladdel
- Translational Oncology Program, University of Michigan, 1600 Huron Pkwy, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.,Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Yang Wang
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI, 48109, USA.,Translational Oncology Program, University of Michigan, 1600 Huron Pkwy, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Molly Kozminsky
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI, 48109, USA.,Translational Oncology Program, University of Michigan, 1600 Huron Pkwy, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Monika L Burness
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Costanza Paoletti
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Ebrahim Azizi
- Translational Oncology Program, University of Michigan, 1600 Huron Pkwy, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.,Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Max S Wicha
- Translational Oncology Program, University of Michigan, 1600 Huron Pkwy, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.,Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI, 48109, USA.,Translational Oncology Program, University of Michigan, 1600 Huron Pkwy, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| |
Collapse
|
98
|
Xing T, Wang B, Song Y, Zhang S, Ma L. Candle soot-templated silica nanobiointerface chip for detecting circulating tumour cells from patients with urologic malignancies. RSC Adv 2018; 8:34566-34572. [PMID: 35548613 PMCID: PMC9086980 DOI: 10.1039/c8ra05807e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/20/2018] [Indexed: 12/16/2022] Open
Abstract
Liquid biopsy, known as fluid biopsy or fluid phase biopsy, is of great clinical significance in cancer diagnosis and treatment monitoring. However, traditional techniques still meet restrictions when aiming for the detection of circulating tumour cells (CTCs) with high efficiency and low cost. Herein, we applied an easily prepared silica nanobiointerface chip for detecting CTCs in prostate cancer (PCa) and clear cell renal cell carcinoma (ccRCC) patients with high efficiency. The silica nanobiointerface chip was fabricated by depositing candle soot on a glass slide, followed by chemical vapour deposition, and then by modifying anti-epithelial cell adhesion molecule (EpCAM) antibody. The silica nanobiointerface chips exhibited excellent abilities to capture PC3 PCa cell lines, with average efficiency of 81.2 ± 1.4%. We demonstrate that the strong topographic interaction between targeted cells and nanostructured surface is critical to enhancing the capture efficiency of CTCs. We further tested peripheral blood samples from 10 preoperative PCa and 7 ccRCC patients. The results show that CTCs from 7 PCa cases and 4 ccRCC cases were successfully detected. We believe that the nanobiointerface chip will provide great potential for the clinical application of CTC.
Collapse
Affiliation(s)
- Tianying Xing
- Department of Urology, Peking University Third Hospital Beijing China
| | - Binshuai Wang
- Department of Urology, Peking University Third Hospital Beijing China
| | - Yimeng Song
- Department of Urology, Peking University Third Hospital Beijing China
| | - Shudong Zhang
- Department of Urology, Peking University Third Hospital Beijing China
| | - Lulin Ma
- Department of Urology, Peking University Third Hospital Beijing China
| |
Collapse
|
99
|
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: 303] [Impact Index Per Article: 43.3] [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
|
100
|
Dong Z, Yu D, Liu Q, Ding Z, Lyons VJ, Bright RK, Pappas D, Liu X, Li W. Enhanced capture and release of circulating tumor cells using hollow glass microspheres with a nanostructured surface. NANOSCALE 2018; 10:16795-16804. [PMID: 30160287 PMCID: PMC6693900 DOI: 10.1039/c8nr04434a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Self-floating hollow glass microspheres (HGMS) modified with tumor-specific antibodies have been developed for the capture of circulating tumor cells (CTCs), and have demonstrated effective cell isolation and good viability of isolated cancer cells. However, the capture efficiency decreases dramatically if the spiked cell concentration is low, possibly due to insufficient interactions between cancer cells and the HGMS surface. In order to apply HGMS-based CTC isolation to clinically relevant samples, it is desirable to create nanostructures on the surface of HGMS to enhance cell-surface interactions. Nevertheless, current microfabrication methods cannot generate nanostructured-surfaces on microspheres. The authors have developed a new HGMS with a controlled nanotopographical surface structure (NSHGMS), and demonstrated isolation and recovery of rare cancer cells. NSHGMS are achieved by applying layer-by-layer (LbL) assembly of negatively charged SiO2 nanoparticles and positively charged poly-l-arginine molecules, then sheathing the surface with an enzymatically degradable LbL film made from biotinylated alginate and poly-l-arginine, and capping with anti-EpCAM antibodies and anti-fouling PEG molecules. Compared to smooth-surfaced HGMS, NSHGMS showed shorter isolation time (20 min), enhanced capture efficiency (93.6 ± 4.9%) and lower detection limit (30 cells per mL) for commonly used cancer cell lines (MCF7, SK-BR-3, PC-3, A549 and CCRF-CEM). This NSHGMS-based CTC isolation method does not require specialized lab equipment or an external power source, and thus, can be used for the separation of targeted cells from blood or other body fluids in a resource-limited environment.
Collapse
Affiliation(s)
- Ziye Dong
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|