1
|
Wang X, Bai L, Kong L, Guo Z. Advances in circulating tumor cells for early detection, prognosis and metastasis reduction in lung cancer. Front Oncol 2024; 14:1411731. [PMID: 38974237 PMCID: PMC11224453 DOI: 10.3389/fonc.2024.1411731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
Globally, lung cancer stands as the leading type of cancer in terms of incidence and is the major source of mortality attributed to cancer. We have outlined the molecular biomarkers for lung cancer that are available clinically. Circulating tumor cells (CTCs) spread from the original location, circulate in the bloodstream, extravasate, and metastasize, forming secondary tumors by invading and establishing a favorable environment. CTC analysis is considered a common liquid biopsy method for lung cancer. We have enumerated both in vivo and ex vivo techniques for CTC separation and enrichment, examined the advantages and limitations of these methods, and also discussed the detection of CTCs in other bodily fluids. We have evaluated the value of CTCs, as well as CTCs in conjunction with other biomarkers, for their utility in the early detection and prognostic assessment of patients with lung cancer. CTCs engage with diverse cells of the metastatic process, interfering with the interaction between CTCs and various cells in metastasis, potentially halting metastasis and enhancing patient prognosis.
Collapse
Affiliation(s)
- Xiaochen Wang
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Lu Bai
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Linghui Kong
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Zhijuan Guo
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
2
|
Wei YJ, Wei X, Zhang X, Wu CX, Cai JY, Chen ML, Wang JH. A hydrodynamic-based dual-function microfluidic chip for high throughput discriminating tumor cells. Talanta 2024; 273:125884. [PMID: 38508128 DOI: 10.1016/j.talanta.2024.125884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
A hydrodynamic-based microfluidic chip consisted of two function units that could not only separate tumor cells (TCs) from whole blood but also remove residual blood cells was designed. The separation of TCs was achieved by a straight contraction-expansion array (CEA) microchannel on the front end of the chip. The addition of contractive structure brought a micro-vortex like Dean vortex that promoted cell focusing in the channel, while when cells entered the dilated region, the wall-induced lift force generated by the channel wall gave cells a push away from the wall. As the wall-induced lift force is proportional to the third power of the cell diameter, TCs with larger diameter will have a larger lateral migration under the wall-induced lift force, realizing the separation of TCs from blood sample. Fluorescent particles with diameters of 19.3 μm and 4.5 μm were used to simulate TCs and red blood cells, respectively, to verify the separation capacity of the proposed CEA microchannel for particles with different diameter. And a separation efficiency 98.7% for 19.3 μm particles and a removal rate 96.2% for 4.5 μm particles was observed at sample flow rate of 10 μL min-1 and sheath flow rate of 190 μL min-1. In addition, a separation efficiency about 96.1% for MCF-7 cells (stained with DiI) and removal rates of 96.2% for red blood cells (RBCs) and 98.7% for white blood cells (WBCs) were also obtained under the same condition. However, on account of the large number of blood cells in the blood, there will be a large number of blood cells remained in the isolated TCs, so a purification unit based on hydrodynamic filtration (HDF) was added after the separation microchannel. The purification channel is a size-dictated cell filter that can remove residual blood cells but retain TCs, thus achieving the purification of TCs. Combined the CEA microchannel and the purifier, the microchip facilitates sorting of MCF-7 cells from whole blood with a separation rate about 95.3% and a removal rate over 99.99% for blood cells at a sample flow rate of 10 μL min-1, sheath flow rate of 190 μL min-1 and washing flow rate of 63 μL min-1.
Collapse
Affiliation(s)
- Yu-Jia Wei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Xing Wei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Xuan Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Cheng-Xing Wu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Ji-Ying Cai
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Ming-Li Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China.
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China.
| |
Collapse
|
3
|
Mukherjee S, Mukherjee A, Bytesnikova Z, Ashrafi AM, Richtera L, Adam V. 2D graphene-based advanced nanoarchitectonics for electrochemical biosensors: Applications in cancer biomarker detection. Biosens Bioelectron 2024; 250:116050. [PMID: 38301543 DOI: 10.1016/j.bios.2024.116050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/01/2024] [Accepted: 01/17/2024] [Indexed: 02/03/2024]
Abstract
Low-cost, rapid, and easy-to-use biosensors for various cancer biomarkers are of utmost importance in detecting cancer biomarkers for early-stage metastasis control and efficient diagnosis. The molecular complexity of cancer biomarkers is overwhelming, thus, the repeatability and reproducibility of measurements by biosensors are critical factors. Electrochemical biosensors are attractive alternatives in cancer diagnosis due to their low cost, simple operation, and promising analytical figures of merit. Recently graphene-derived nanostructures have been used extensively for the fabrication of electrochemical biosensors because of their unique physicochemical properties, including the high electrical conductivity, adsorption capacity, low cost and ease of mass production, presence of oxygen-containing functional groups that facilitate the bioreceptor immobilization, increased flexibility and mechanical strength, low cellular toxicity. Indeed, these properties make them advantageous compared to other alternatives. However, some drawbacks must be overcome to extend their use, such as poor and uncontrollable deposition on the substrate due to the low dispersity of some graphene materials and irreproducibility of the results because of the differences in various batches of the produced graphene materials. This review has documented the most recently developed strategies for electrochemical sensor fabrication. It differs in the categorization method compared to published works to draw greater attention to the wide opportunities of graphene nanomaterials for biological applications. Limitations and future scopes are discussed to advance the integration of novel technologies such as artificial intelligence, the internet of medical things, and triboelectric nanogenerators to eventually increase efficacy and efficiency.
Collapse
Affiliation(s)
- Soumajit Mukherjee
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
| | - Atripan Mukherjee
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic; ELI Beamlines Facility, The Extreme Light Infrastructure ERIC, Za Radnici 835, 252 41, Dolni Breznany, Czech Republic
| | - Zuzana Bytesnikova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
| | - Amir M Ashrafi
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
| | - Lukas Richtera
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00, Brno, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic.
| |
Collapse
|
4
|
Liu LE, Xue L, Li Y, Ji J, Yuan X, Han H, Ding L, Wu Y, Yang R. MOFs-derived Co 3O 4@MnO 2@Carbon dots with enhanced nanozymes activity for photoelectrochemical detection of cancer cells in whole blood. Talanta 2024; 266:125095. [PMID: 37625292 DOI: 10.1016/j.talanta.2023.125095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Nanozymes have attracted widespread attention, and rationally designing high-activity nanozymes to improve their application performance are a long-term objective. Herein, taking metal-organic frameworks-derived Co3O4 polyhedron with large surface area and high porosity as nanoconfinement carriers, Co3O4@MnO2@CDs polyhedron was successfully synthesized by the room-temperature reduction of MnO4- ions and physical load of carbon dots (CDs). Through cancer cells-triggered double antibody sandwich strategy, the Co3O4@MnO2@CDs polyhedron were introduced to the TiO2 nanoparticle (NPs) modified electrode, leading to the decreased photocurrent. The Co3O4@MnO2@CDs polyhedron can not only quench the photocurrent of TiO2 NPs, also act as nanozymes to catalyze precipitates. Moreover, the precipitates can not only reduce the photoelectrochemical (PEC) response, also increase the quenching capacity of the Co3O4@MnO2@CDs polyhedron. Additionally, the steric hindrance effect of the Co3O4@MnO2@CDs-Ab conjugates further weaken the photocurrent. Based on the multifunctional Co3O4@MnO2@CDs polyhedron, the proposed PEC biosensor for the detection of A549 cancer cells exhibits a wide linear range from 102 to 106 cells/mL and a low detection limit of 11 cells/mL. Furthermore, this strategy can differentiate between lung cancer patients and healthy individuals. The designed multifunctional Co3O4@MnO2@CDs nanozymes provide a new horizon for PEC detection of cancer cells, and may have great potential in early clinical diagnosis and biomedical research.
Collapse
Affiliation(s)
- Li-E Liu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Linsheng Xue
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuling Li
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiangying Ji
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Xinxin Yuan
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Hangchen Han
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Ruiying Yang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
5
|
Zhan Q, Liu B, Situ X, Luo Y, Fu T, Wang Y, Xie Z, Ren L, Zhu Y, He W, Ke Z. New insights into the correlations between circulating tumor cells and target organ metastasis. Signal Transduct Target Ther 2023; 8:465. [PMID: 38129401 PMCID: PMC10739776 DOI: 10.1038/s41392-023-01725-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Organ-specific metastasis is the primary cause of cancer patient death. The distant metastasis of tumor cells to specific organs depends on both the intrinsic characteristics of the tumor cells and extrinsic factors in their microenvironment. During an intermediate stage of metastasis, circulating tumor cells (CTCs) are released into the bloodstream from primary and metastatic tumors. CTCs harboring aggressive or metastatic features can extravasate to remote sites for continuous colonizing growth, leading to further lesions. In the past decade, numerous studies demonstrated that CTCs exhibited huge clinical value including predicting distant metastasis, assessing prognosis and monitoring treatment response et al. Furthermore, increasingly numerous experiments are dedicated to identifying the key molecules on or inside CTCs and exploring how they mediate CTC-related organ-specific metastasis. Based on the above molecules, more and more inhibitors are being developed to target CTCs and being utilized to completely clean CTCs, which should provide promising prospects to administer advanced tumor. Recently, the application of various nanomaterials and microfluidic technologies in CTCs enrichment technology has assisted to improve our deep insights into the phenotypic characteristics and biological functions of CTCs as a potential therapy target, which may pave the way for us to make practical clinical strategies. In the present review, we mainly focus on the role of CTCs being involved in targeted organ metastasis, especially the latest molecular mechanism research and clinical intervention strategies related to CTCs.
Collapse
Affiliation(s)
- Qinru Zhan
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Bixia Liu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Xiaohua Situ
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Yuting Luo
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Tongze Fu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Yanxia Wang
- Zhongshan School of Medicine, Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Zhongpeng Xie
- Zhongshan School of Medicine, Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Lijuan Ren
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Ying Zhu
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China.
| | - Weiling He
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, 10065, USA.
- School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, 361000, Xiamen, Fujian, P.R. China.
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China.
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|
6
|
Ouyang D, Ye N, Yang K, Wang Y, Hu L, Chao S, Toner M, Li Y. Precision Isolation of Circulating Leukemia Cells in Chronic Myelogenous Leukemia Patients Using a Novel Microfluidic Device and Its Clinical Applications. Cancers (Basel) 2023; 15:5696. [PMID: 38067399 PMCID: PMC10705219 DOI: 10.3390/cancers15235696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 02/12/2024] Open
Abstract
Chronic Myelogenous Leukemia (CML) is a prevalent hematologic malignancy characterized by the malignant transformation of myeloid cells and their proliferation in the peripheral blood. The management of CML poses significant challenges, particularly in detecting and eradicating minimal residual disease, which is crucial for preventing relapse and improving survival outcomes. Traditional minimal residual disease detection methods, such as bone marrow aspiration, are invasive and have limitations which include the potential for sampling errors and false negatives. This study introduces a novel label-free microfluidic chip designed for the segregation and recovery of circulating leukemia cells, offering a non-invasive liquid biopsy approach with potential applications in precision medicine. Over July 2021 to October 2023, we recruited 56 CML patients across various disease stages and collected blood samples for analysis using our microfluidic device. The device demonstrated high efficacy in isolating circulating leukemia cells, with an optimal capture efficiency of 78% at a sample flow rate of 3 mL/h. Our results indicate that the microfluidic device can efficiently segregate and quantify circulating leukemia cells, providing a detailed understanding of CML progression and treatment response. The significant reduction in circulating leukemia cell counts in patients in complete remission highlights the device's potential in monitoring treatment efficacy. Furthermore, the device's sensitivity in detecting minimal residual disease could offer a more reliable prognostic tool for therapeutic decision-making in CML management.
Collapse
Affiliation(s)
- Dongfang Ouyang
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston, MA 02129, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Ningxin Ye
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Kun Yang
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3E8, Canada
| | - Yiyang Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Lina Hu
- Department of Hematology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Shuen Chao
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston, MA 02129, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston, MA 02129, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Yonghua Li
- Department of Hematology, PLA General Hospital of Southern Theater Command, Guangzhou 510010, China
| |
Collapse
|
7
|
Wang W, Zheng Z, Lei J. CTC, ctDNA, and Exosome in Thyroid Cancers: A Review. Int J Mol Sci 2023; 24:13767. [PMID: 37762070 PMCID: PMC10530859 DOI: 10.3390/ijms241813767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Thyroid cancer has become more common in recent years all around the world. Many issues still need to be urgently addressed in the diagnosis, treatment, and prognosis of thyroid cancer. Liquid biopsy (mainly circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and circulating exosomes) may provide a novel and ideal approach to solve these issues, allows us to assess the features of diseases more comprehensively, and has a function in a variety of malignancies. Recently, liquid biopsy has been shown to be critical in thyroid cancer diagnosis, treatment, and prognosis in numerous previous studies. In this review, by testing CTCs, ctDNA, and exosomes, we focus on the possible clinical role of liquid biopsy in thyroid cancer, including diagnostic and prognostic biomarkers and response to therapy. We briefly review how liquid biopsy components have progressed in thyroid cancer by consulting the existing public information. We also discuss the clinical potential of liquid biopsy in thyroid cancer and provide a reference for liquid biopsy research. Liquid biopsy has the potential to be a useful tool in the early detection, monitoring, or prediction of response to therapies and prognosis in thyroid cancer, with promising clinical applications.
Collapse
Affiliation(s)
- Wenwen Wang
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyao Zheng
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianyong Lei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Yu N, Ma G, Chen Y, Huang S, Gong Y, Li S, Gu H, You H, Miao P. MnO 2 nanosheets and gold nanoparticles supported electrochemical detection of circulating tumor cells. Colloids Surf B Biointerfaces 2023; 229:113482. [PMID: 37523806 DOI: 10.1016/j.colsurfb.2023.113482] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/15/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
The concentration of circulating tumor cells (CTCs) in peripheral blood is strongly correlated with the progress of certain metastatic cancers. In this study, we have developed a novel and facile electrochemical biosensor for the detection of CTCs based on the use of manganese dioxide nanosheets (MnO2 NSs) and gold nanoparticles (AuNPs). Aptamer sequence of target cell is modified on the surface of AuNPs for specifical recognition. With low-speed centrifugation, numerous AuNPs@DNA can be removed from the supernatant. On the other hand, MnO2 NSs are modified on the electrode surface to capture unreacted AuNPs@DNA. The declined electrochemical signal intensity can be used to reflect the level of CTCs. This biosensor achieves a wide linear range from 10 to 104 cells mL-1 and a limit of detection as low as 3 cells mL-1. Due to the specific aptamer as the recognition element, interfering cells can be successfully distinguished and this method performs satisfactorily in clinical samples. Therefore, it has great potential to be used as a powerful tool benefiting rare cells analysis and the investigation of dynamics of cellular interactions.
Collapse
Affiliation(s)
- Nong Yu
- Department of Diagnostics, People's Hospital of Suzhou New District, Suzhou 215010, PR China
| | - Guifeng Ma
- Department of Diagnostics, People's Hospital of Suzhou New District, Suzhou 215010, PR China
| | - Yuyao Chen
- Department of Diagnostics, People's Hospital of Suzhou New District, Suzhou 215010, PR China
| | - Shan Huang
- Department of Diagnostics, People's Hospital of Suzhou New District, Suzhou 215010, PR China
| | - Yalei Gong
- Department of Diagnostics, People's Hospital of Suzhou New District, Suzhou 215010, PR China
| | - Shuangshuang Li
- Department of Diagnostics, People's Hospital of Suzhou New District, Suzhou 215010, PR China
| | - Haiqin Gu
- Department of Diagnostics, People's Hospital of Suzhou New District, Suzhou 215010, PR China
| | - Honglan You
- Department of Diagnostics, People's Hospital of Suzhou New District, Suzhou 215010, PR China.
| | - Peng Miao
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, PR China.
| |
Collapse
|
9
|
Halawa T, Baeesa S, Fadul MM, Badahdah AA, Enani M, Fathaddin AA, Kawass D, Alkhotani A, Bahakeem B, Kurdi M. The Role of Liquid Biopsy in the Diagnosis and Prognosis of WHO Grade 4 Astrocytoma. Cureus 2023; 15:e41221. [PMID: 37525780 PMCID: PMC10387356 DOI: 10.7759/cureus.41221] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/02/2023] Open
Abstract
Liquid biopsy, as a non-invasive diagnostic tool, has recently gained significant attention in the field of oncology. It involves the analysis of various biomarkers present in bodily fluids, such as blood or cerebrospinal fluid, to provide information about the underlying cancer. In the case of WHO grade 4 astrocytomas, liquid biopsy has the potential to significantly impact the diagnosis and prognosis of this aggressive malignant brain tumor. By detecting specific genetic mutations, such as IDH1 or EGFR, and monitoring levels of circulating tumor DNA, liquid biopsy can aid in the early detection and monitoring of disease progression. This innovative approach is gradually being acknowledged as a less invasive and cost-effective procedure for cancer diagnosis and management to improve patient outcomes and quality of life. Various kinds of biomarkers circulating in cerebrospinal fluid (CSF), such as circulating tumor cells (CTC) and different types of nucleic acids like cell-free DNA (cfDNA), cell-free RNA (ctRNA), and microRNAs (miRNA), have been identified. These biomarkers, which require dependable detection methods, are comparatively simple to obtain and allow for repeated measurements, making them significantly superior for disease monitoring. This review aims to compare the latest liquid biopsy analysis tools for both CSF and plasma in the central nervous system.
Collapse
Affiliation(s)
- Taher Halawa
- Department of Pediatrics, Faculty of Medicine King Abdulaziz University, Rabigh, SAU
| | - Saleh Baeesa
- Department of Neuroscience, King Faisal Specialist Hospital and Research Centre, Jeddah, SAU
| | - Motaz M Fadul
- Department of Pathology, Faculty of Medicine King Abdulaziz University, Rabigh, SAU
| | - Adnan A Badahdah
- Department of Internal Medicine, University of Jeddah, Jeddah, SAU
| | - Maryam Enani
- Department of Surgery, King Abdulaziz University Hospital, Jeddah, SAU
| | - Amany A Fathaddin
- Department of Pathology, College of Medicine, King Saud University, Riyadh, SAU
- Department of Pathology, King Saud University Medical City, Riyadh, SAU
| | - Dania Kawass
- Department of Family Medicine, Faculty of Medicine King Abdulaziz University, Jeddah, SAU
| | - Alaa Alkhotani
- Department of Pathology, Umm Al-Qura University, Makkah, SAU
| | - Basem Bahakeem
- Department of Internal Medicine, Umm Al-Qura University, Makkah, SAU
| | - Maher Kurdi
- Department of Pathology, Faculty of Medicine King Abdulaziz University, Rabigh, SAU
| |
Collapse
|
10
|
Wang X, Diwu W, Guo J, Yan M, Ma W, Yang M, Bi L, Han Y. Enhancement of antibacterial properties and biocompatibility of Ti 6Al 4V by graphene oxide/strontium nanocomposite electrodepositing. Biochem Biophys Res Commun 2023; 665:35-44. [PMID: 37156051 DOI: 10.1016/j.bbrc.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/19/2023] [Accepted: 05/01/2023] [Indexed: 05/10/2023]
Abstract
Ti6Al4V is a widely used orthopedic implant material in clinics. Due to its poor antibacterial properties, surface modification is required to prevent peri-implantation infection. However, chemical linkers used for surface modification have generally been reported to have detrimental effects on cell growth. In this work, by optimizing parameters related to electrodeposition, a composite structural coating with graphene oxide (GO) compact films in the inner layer and 35 nm diameter strontium (Sr) nanoparticles in the outer layer was constructed on the surface of Ti6Al4V without using substance harmful to bone marrow mesenchymal stem cells (BMSCs) growth. The antibacterial properties of Ti6Al4V are enhanced by the controlled release of Sr ions and incomplete masking of the GO surface, showing excellent antibacterial activity against Staphylococcus aureus in bacterial culture assays. The biomimetic GO/Sr coating has a reduced roughness of the implant surface and a water contact angle of 44.1°, improving the adhesion, proliferation and differentiation of BMSCs. Observations of synovial tissue and fluid in the joint in an implantation model of rabbit knee also point to the superior anti-infective properties of the novel GO/Sr coating. In summary, the novel GO/Sr nanocomposite coating on the surface of Ti6Al4V effectively prevents surface colonization of Staphylococcus aureus and eliminates local infections in vitro and in vivo.
Collapse
Affiliation(s)
- Xing Wang
- Department of Orthopedics, The First Affiliated Hospital of Air Force Military Medical University, People's Republic of China; Department of Medical Identification, The Air Force Medical Center, Beijing, People's Republic of China
| | - Weilong Diwu
- Department of Orthopedics, The First Affiliated Hospital of Air Force Military Medical University, People's Republic of China
| | - Jianbin Guo
- Department of Joint Surgery, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, People's Republic of China
| | - Ming Yan
- Department of Orthopedics, The First Affiliated Hospital of Air Force Military Medical University, People's Republic of China
| | - Wenrui Ma
- Department of Orthopedics, The First Affiliated Hospital of Air Force Military Medical University, People's Republic of China
| | - Min Yang
- Department of Orthopedics, The First Affiliated Hospital of Air Force Military Medical University, People's Republic of China
| | - Long Bi
- Department of Orthopedics, The First Affiliated Hospital of Air Force Military Medical University, People's Republic of China.
| | - Yisheng Han
- Department of Orthopedics, The First Affiliated Hospital of Air Force Military Medical University, People's Republic of China.
| |
Collapse
|
11
|
Li J, Dong C, Gan H, Gu X, Zhang J, Zhu Y, Xiong J, Song C, Wang L. Nondestructive separation/enrichment and rolling circle amplification-powered sensitive SERS enumeration of circulating tumor cells via aptamer recognition. Biosens Bioelectron 2023; 231:115273. [PMID: 37054599 DOI: 10.1016/j.bios.2023.115273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Nondestructive separation/enrichment and reliable detection of extremely rare circulating tumor cells (CTCs) in peripheral blood are of considerable importance in tumor precision diagnosis and treatment, yet this remains a big challenge. Herein, a novel strategy for nondestructive separation/enrichment and ultra-sensitive surface-enhanced Raman scattering (SERS)-based enumeration of CTCs is proposed via aptamer recognition and rolling circle amplification (RCA). In this work the magnetic beads modified with "Aptamer (Apt)-Primer" (AP) probes were utilized to specifically capture CTCs, and then after magnetic separation/enrichment, the RCA-powered SERS counting and benzonase nuclease cleavage-assisted nondestructive release of CTCs were realized, respectively. The AP was assembled by hybridizing the EpCAM-specific aptamer with a primer, and the optimal AP contains 4 mismatched bases. The RCA enhanced SERS signal nearly 4.5-fold, and the SERS strategy has good specificity, uniformity and reproducibility. The proposed SERS detection possesses a good linear relationship with the concentration of MCF-7 cells spiked in PBS with the limit of detection (LOD) of 2 cells/mL, which shows good potential practicality for detecting CTCs in blood with recoveries ranging from 100.56% to 116.78%. Besides, the released CTCs remained good cellular activity with the normal proliferation after re-culture for 48 h and normal growth for at least three generations. The proposed strategy of nondestructive separation/enrichment and SERS-based sensitive enumeration is promising for reliable analysis of EpCAM-positive CTCs in blood, which is expected to provide a powerful tool for analysis of extremely rare circulating tumor cells in complex peripheral blood for liquid biopsy.
Collapse
|
12
|
Lv S, Zheng D, Chen Z, Jia B, Zhang P, Yan J, Jiang W, Zhao X, Xu JJ. Near-Infrared Light-Responsive Size-Selective Lateral Flow Chip for Single-Cell Manipulation of Circulating Tumor Cells. Anal Chem 2023; 95:1201-1209. [PMID: 36541430 DOI: 10.1021/acs.analchem.2c03947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Accurately obtaining information on the heterogeneity of CTCs at the single-cell level is a very challenging task that may facilitate cancer pathogenesis research and personalized therapy. However, commonly used multicellular population capture and release assays tend to lose effective information on heterogeneity and cannot accurately assess molecular-level studies and drug resistance assessment of CTCs in different stages of tumor metastasis. Herein, we designed a near-infrared (NIR) light-responsive microfluidic chip for biocompatible single-cell manipulation and study the heterogeneity of CTCs by a combination of the lateral flow microarray (LFM) chip and photothermal response system. First, immunomagnetic labeling and a gradient magnetic field were combined to distribute CTCs in different regions of the chip according to the content of surface markers. Subsequently, the LFM chip achieves high single-cell capture efficiency and purity (even as low as 5 CTCs per milliliter of blood) under the influence of lateral fluid and magnetic fields. Due to the rapid dissolution of the gelatin capture structure at 37 °C and the photothermal properties of gold nanorods, the captured single CTC cell can be recovered in large quantities at physiological temperature or released individually at a specific point by NIR. The multifunctional NIR-responsive LFM chip demonstrates excellent performance in capture and site release of CTCs with high viability, which provides a robust and versatile means for CTCs heterogeneity study at the single-cell level.
Collapse
Affiliation(s)
- Songwei Lv
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.,School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Dong Zheng
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Zhaoxian Chen
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Bin Jia
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Peng Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jiaxuan Yan
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Wanlan Jiang
- Department of Rheumatology and Immunology, The First People's Hospital of Changzhou (The Third Affiliated Hospital of Soochow University), Changzhou 213003, China
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jing-Juan Xu
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
13
|
Chen Z, Xiong Y, Ma R, Chen P, Duan L, Yang S, Gisèle IU, You L, Xiao D. A novel magnetic fluid for ultra-fast and highly efficient extraction of N1-methylnicotinamide in urine samples. NEW J CHEM 2023. [DOI: 10.1039/d3nj00488k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Among the three pre-treatment materials, Fe3O4@HPMC@DMSA NPs were selected to be the best material and were used to perform MSPE-HPLC-UV.
Collapse
Affiliation(s)
- Zhuhui Chen
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Yue Xiong
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Ranran Ma
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Pei Chen
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Le Duan
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Shuying Yang
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Ineza Urujeni Gisèle
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Linjun You
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, 24 Tongjia Lane, Nanjing 210009, China
| | - Deli Xiao
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, 24 Tongjia Lane, Nanjing 210009, China
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
14
|
Reversible capture and release of circulating tumor cells on a three‐dimensional conductive interface to improve cell purity for gene mutation analysis. VIEW 2022. [DOI: 10.1002/viw.20220054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
15
|
Kang H, Xiong Y, Ma L, Yang T, Xu X. Recent advances in micro-/nanostructure array integrated microfluidic devices for efficient separation of circulating tumor cells. RSC Adv 2022; 12:34892-34903. [PMID: 36540264 PMCID: PMC9724214 DOI: 10.1039/d2ra06339e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/18/2022] [Indexed: 09/06/2023] Open
Abstract
Circulating tumor cells (CTCs) released from the primary tumor to peripheral blood are promising targets for liquid biopsies. Their biological information is vital for early cancer detection, efficacy assessment, and prognostic monitoring. Despite the tremendous clinical applications of CTCs, development of effective separation techniques are still demanding. Traditional separation methods usually use batch processing for enrichment, which inevitably destroy cell integrity and affect the complete information acquisition. Considering the rarity and heterogeneity of CTCs, it is urgent to develop effective separation methods. Microfluidic chips with precise fluid control at the micron level are promising devices for CTC separation. Their further combination with micro-/nanostructure arrays adds more biomolecule binding sites and exhibit unique fluid barrier effect, which significantly improve the CTC capture efficiency, purity, and sensitivity. This review summarized the recent advances in micro-/nanostructure array integrated microfluidic devices for CTC separation, including microrods, nanowires, and 3D micro-/nanostructures. The mechanisms by which these structures contribute to improved capture efficiency are discussed. Two major categories of separation methods, based on the physical and biological properties of CTCs, are discussed separately. Physical separation includes the design and preparation of micro-/nanostructure arrays, while chemical separation additionally involves the selection and modification of specific capture probes. These emerging technologies are expected to become powerful tools for disease diagnosis in the future.
Collapse
Affiliation(s)
- Hanyue Kang
- School of Materials Science and Engineering, Tongji University Shanghai 201804 China
| | - Yuting Xiong
- School of Materials Science and Engineering, Tongji University Shanghai 201804 China
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University Hangzhou 310058 China
| | - Tongqing Yang
- School of Materials Science and Engineering, Tongji University Shanghai 201804 China
| | - Xiaobin Xu
- School of Materials Science and Engineering, Tongji University Shanghai 201804 China
| |
Collapse
|
16
|
Smolkova B, Kataki A, Earl J, Ruz-Caracuel I, Cihova M, Urbanova M, Buocikova V, Tamargo S, Rovite V, Niedra H, Schrader J, Kohl Y. Liquid biopsy and preclinical tools for advancing diagnosis and treatment of patients with pancreatic neuroendocrine neoplasms. Crit Rev Oncol Hematol 2022; 180:103865. [DOI: 10.1016/j.critrevonc.2022.103865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
|
17
|
Li N, Jin K, Chen T, Li X. A static force model to analyze the nuclear deformation on cell adhesion to vertical nanostructures. SOFT MATTER 2022; 18:6638-6644. [PMID: 36004571 DOI: 10.1039/d2sm00971d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Vertical nanostructures have been found to induce the deformation of the nuclear envelope during cell adhesion. However, there has been a lack of quantitative analysis of the influence of nanostructures morphology on the degree of nuclear deformation. Here, a theoretical model was proposed to investigate the mechanism of nuclear deformation by analyzing the mechanical force balance. Based on the established model, we analyzed the effects of the morphology of the nanopillar array on nuclear deformation and gave the quantitative relationship of the deformation depth of the nucleus with the pitch and radius of nanopillars. Our theoretical results seem to show broad agreements with experimental observations, which implies that the work can provide useful guidance to the design of nanostructures for biomedical applications.
Collapse
Affiliation(s)
- Nanxin Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Kun Jin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Tongsheng Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- SCNU Qingyuan Institute of Science and Technology Innovation Co., Ltd., South China Normal University, Qingyuan 511500, China
| | - Xinlei Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
18
|
Deng Z, Wu S, Wang Y, Shi D. Circulating tumor cell isolation for cancer diagnosis and prognosis. EBioMedicine 2022; 83:104237. [PMID: 36041264 PMCID: PMC9440384 DOI: 10.1016/j.ebiom.2022.104237] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/03/2022] Open
Abstract
Circulating tumor cells (CTCs) are tumor cells that shed from the primary tumor and intravasate into the peripheral blood circulation system responsible for metastasis. Sensitive detection of CTCs from clinical samples can serve as an effective tool in cancer diagnosis and prognosis through liquid biopsy. Current CTC detection technologies mainly reply on the biomarker-mediated platforms including magnetic beads, microfluidic chips or size-sensitive microfiltration which can compromise detection sensitivity due to tumor heterogeneity. A more sensitive, biomarker independent CTCs isolation technique has been recently developed with the surface-charged superparamagnetic nanoprobe capable of different EMT subpopulation CTC capture from 1 mL clinical blood. In this review, this new strategy is compared with the conventional techniques on biomarker specificity, impact of protein corona, effect of glycolysis on cell surface charge, and accurate CTC identification. Correlations between CTC enumeration and molecular profiling in clinical blood and cancer prognosis are provided for clinical cancer management.
Collapse
Affiliation(s)
- Zicheng Deng
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA; Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Shengming Wu
- The Institute for Translational Nanomedicine Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, School of Medicine Tongji University, Shanghai 200092, PR China
| | - Yilong Wang
- The Institute for Translational Nanomedicine Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, School of Medicine Tongji University, Shanghai 200092, PR China.
| | - Donglu Shi
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA.
| |
Collapse
|
19
|
Li J, Yuan Y, Gan H, Dong C, Cao B, Ni JL, Li X, Gu W, Song C, Wang L. Double-Tetrahedral DNA Probe Functionalized Ag Nanorod Biointerface for Effective Capture, Highly Sensitive Detection, and Nondestructive Release of Circulating Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:32869-32879. [PMID: 35839122 DOI: 10.1021/acsami.2c06005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Circulating tumor cells (CTCs) are indicative of tumorigenesis, metastasis, and recurrence; however, it is still a great challenge to efficiently analyze the extremely rare CTCs in peripheral blood. Herein, a novel nanobiointerface integrating high affinities of arrayed silver nanorods (Ag NRs) and double-tetrahedral DNA (DTDN) probes by a clever strategy is proposed for the efficient capture, highly sensitive detection, and nondestructive release of CTCs. Under the optimal conditions, the DTDN-probe-functionalized Ag NRs nanobiointerface can capture 90.2% of SGC-7901 cells in PBS, and the capture efficiency is 2.8 times and 50 times those of a DTDN-probe-functionalized Ag film and unfunctionalized Ag NRs, respectively, benefiting from the nanorough interface of the Ag NRs array and multivalent recognition of the DTDN probe. In addition, 93.4% of cells was released via Zn2+-assisted DNAzyme cleavage, and the viability of the postreleased CTCs is about 98.0%. The potential practicality of the nanobiointerface for testing CTCs in blood was further characterized by spiking SGC-7901 cells in leukocytes collected from human blood, and the results show that 83.8% capture efficiency, 91.2% release efficiency, and single-cell detection limit were achieved, which indicates that the nanobiointerface has great potential in clinical applications for reliable CTC analyses.
Collapse
Affiliation(s)
- Jinxiang Li
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, People's Republic of China
| | - Yugang Yuan
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Hongyu Gan
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, People's Republic of China
| | - Chen Dong
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, People's Republic of China
| | - Bin Cao
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Jin-Liang Ni
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Xueliang Li
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Wenjie Gu
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, People's Republic of China
| | - Chunyuan Song
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, People's Republic of China
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, People's Republic of China
| |
Collapse
|
20
|
Chen K, Zhang Z, Zhu X, Lin Z, Xie J, Dong Q, Fu Q, Zhang Y. In situ signal amplification improves the capture efficiency of circulating tumor cells with low expression of EpCAM. Anal Chim Acta 2022; 1221:340133. [DOI: 10.1016/j.aca.2022.340133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 11/01/2022]
|
21
|
Kong X, Sun Y, Zhang Q, Li S, Jia Y, Li R, Liu Y, Xie Z. Specific Tumor Cell Detection by a Metabolically Targeted Aggregation-Induced Emission-Based Gold Nanoprobe. ACS OMEGA 2022; 7:18073-18084. [PMID: 35664593 PMCID: PMC9161387 DOI: 10.1021/acsomega.2c01494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
Detection of circulating tumor cells (CTCs) could be widely used for early diagnosis and real-time monitoring of tumor progression in liquid biopsy samples. Compared with normal cells, tumor cells exhibit relatively strong negative surface charges due to the high rate of glycolysis. In this study, a cationic fluorescence "turn-on" aggregation-induced emission (AIE) nanoprobe based on gold nanorods (GNRs) was designed and tested to detect tumor cells specifically. In brief, tetraphenylethene (TPE), an AIE dye, was conjugated to the cationic polymer polyethylenimine (PEI) yielding TPEI. TPEI-PEG-SH was obtained by further functionalizing TPEI with a thiol group. TPEI-PEG-SH was grafted to the surface of GNRs, yielding the cationic AIE nanoprobe, named as GNRs-PEG-TPEI. The nanoprobe was characterized to have a uniform particle size of 172 nm, a strong positive surface charge (+54.87 mV), and a surface modification load of ∼40%. The in vitro stability of GNRs-PEG-TPEI was verified. The cellular imaging results demonstrated that the nanoprobe could efficiently recognize several types of tumor cells including MCF-7, HepG2, and Caco-2 while exhibiting specific fluorescence signals only after interacting with tumor cells and minimal background interference. In addition, the study investigated the toxicity of the nanoprobe to the captured cells and proved the safety of the nanoprobe. In conclusion, a specific and efficient nanoprobe was developed for capture and detection of different types of tumor cells based on their unique metabolic characteristics. It holds great promise for achieving early diagnosis and monitoring the tumor progression by detecting the CTCs in clinical liquid biopsy samples.
Collapse
Affiliation(s)
| | | | - Qian Zhang
- School of Pharmaceutical
Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Siju Li
- School of Pharmaceutical
Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yizhen Jia
- School of Pharmaceutical
Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Rui Li
- School of Pharmaceutical
Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yang Liu
- School of Pharmaceutical
Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Zhiyong Xie
- School of Pharmaceutical
Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
22
|
Recent progress on microfluidic devices with incorporated 1D nanostructures for enhanced extracellular vesicle (EV) separation. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00195-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
23
|
Ming R, Jiang Y, Fan J, An C, Li J, Chen T, Li X. High-Efficiency Capture of Cells by Softening Cell Membrane. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106547. [PMID: 35112794 DOI: 10.1002/smll.202106547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/30/2021] [Indexed: 06/14/2023]
Abstract
The capture of circulating tumor cells (CTCs) by nanostructured substrate surface is a useful method for early diagnosis of cancer. At present, most methods used to improve the cell capture efficiency are based on changing substrate surface properties. However, there are still some gaps between these methods and practical applications. Here, a method is presented for improving cell capture efficiency from a different perspective, that is, changing the properties of the cells. Concretely, the mechanical properties of the cell membrane are changed by adding Cytochalasin D to soften the cell membrane. Furthermore, a corresponding theoretical model is proposed to explain the experimental results. It is found that cell softening can reduce the resistance of cell adhesion, which makes the adhesion ability stronger. The high-efficiency capture of cells by softening the cell membrane provides a potential method to improve the detection performance of CTCs.
Collapse
Affiliation(s)
- Ruiqi Ming
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Ye Jiang
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Jiaqi Fan
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Chunchun An
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Jinqi Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Tongsheng Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- SCNU Qingyuan Institute of Science and Technology Innovation Co. Ltd., Qingyuan, 511517, China
| | - Xinlei Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| |
Collapse
|
24
|
Shi J, Zhao C, Shen M, Chen Z, Liu J, Zhang S, Zhang Z. Combination of microfluidic chips and biosensing for the enrichment of circulating tumor cells. Biosens Bioelectron 2022; 202:114025. [DOI: 10.1016/j.bios.2022.114025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 12/26/2022]
|
25
|
Liu Y, Lin Z, Zheng Z, Zhang Y, Shui L. Accurate Isolation of Circulating Tumor Cells via a Heterovalent DNA Framework Recognition Element-Functionalized Microfluidic Chip. ACS Sens 2022; 7:666-673. [PMID: 35113538 DOI: 10.1021/acssensors.1c02692] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Detection of circulating tumor cells (CTCs) has provided a noninvasive and efficient approach for early diagnosis, treatment, and prognosis of cancer. However, efficient capture of CTCs in the clinical environment is very challenging because of the extremely rare and heterogeneous expression of CTCs. Herein, we fabricated a multimarker microfluidic chip for the enrichment of heterogeneous CTCs from peripheral blood samples of breast cancer patients. The multimarker aptamer cocktail DNA nanostructures (TP-multimarker) were modified on a deterministic lateral displacement (DLD)-patterned microfluidic chip to enhance the capture efficiency through the size selection effect of DLD arrays and the synergistic effect of multivalent aptamers. As compared to a monovalent aptamer-modified chip, the multimarker chip exhibits enhanced capture efficiency toward both high and low epithelial cell adhesion molecule expression cell lines, and the DNA nanostructure-functionalized chip enables the accurate capture of different phenotypes of CTCs. In addition, the DNA nanoscaffold makes nucleases more accessible to the aptamers to release cells with molecular integrity and outstanding cell viability.
Collapse
Affiliation(s)
- Yan Liu
- Joint Laboratory of Optofluidic Technology and Systems, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Zhun Lin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ziwei Zheng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Lingling Shui
- Joint Laboratory of Optofluidic Technology and Systems, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Nanophotonic Functional Materials and Devices, School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China
| |
Collapse
|
26
|
Chen Z, Gu Y, Wang G, Liu Q, Li Y, Weng Y, Lu N, Yang G, Liu Y. Normal Force-Induced Highly Efficient Mechanical Sterilization of GaN Nanopillars. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:856-862. [PMID: 34990133 DOI: 10.1021/acs.langmuir.1c03066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Bacterial residue is one of the main causes of diseases and economic losses. In recent years, microfabrication technology has inspired the introduction of microstructures on the surfaces of relevant materials to provide antibacterial effects. This antibacterial method has become a popular research topic due to its safety, effectiveness, and stability. However, its exact mechanism is still under debate. In this study, normal force was introduced to bacteria on GaN nanopillars to investigate the mechanical sterilization effects and a computer simulation was conducted. The results show that the normal force induces highly efficient mechanical sterilization of the nanopillars, and their surfaces impede the attachment of bacteria. This study provides insights into the antibacterial effect of nanopillars and offers a potential antibacterial tool with high efficiency.
Collapse
Affiliation(s)
- Zhe Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Yan Gu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Gongbo Wang
- College of Mechanical Engineering, Tianjin University, Tianjin 300072, China
| | - Qingrun Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Yujie Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Yuyan Weng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Naiyan Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Science, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Guofeng Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Science, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Yu Liu
- Microsoft Quantum Materials Lab Copenhagen, 2800 Lyngby, Denmark
- Center for Quantum Devices, Niels Bohr Institute, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
27
|
Xu Y, Lin J, Wu X, Xu X, Zhang D, Xie Y, Pan T, He Y, Wu A, Shao G. TiO2-Based Bioprobe Enabling Excellent SERS Activity in Detection of Diverse Circulating Tumor Cells. J Mater Chem B 2022; 10:3808-3816. [DOI: 10.1039/d2tb00464j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Circulating tumor cells (CTCs), can be the seeds of tumor metastasis, and are closely linked to cancer-related death. Fast and effective detection of CTCs is important for early diagnosis of...
Collapse
|
28
|
Takahashi H, Baba Y, Yasui T. Oxide nanowire microfluidics addressing previously-unattainable analytical methods for biomolecules towards liquid biopsy. Chem Commun (Camb) 2021; 57:13234-13245. [PMID: 34825908 DOI: 10.1039/d1cc05096f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nanowire microfluidics using a combination of self-assembly and nanofabrication technologies is expected to be applied to various fields due to its unique properties. We have been working on the fabrication of nanowire microfluidic devices and the development of analytical methods for biomolecules using the unique phenomena generated by the devices. The results of our research are not just limited to the development of nanospace control with "targeted dimensions" in "targeted arrangements" with "targeted materials/surfaces" in "targeted spatial locations/structures" in microfluidic channels, but also cover a wide range of analytical methods for biomolecules (extraction, separation/isolation, and detection) that are impossible to achieve with conventional technologies. Specifically, we are working on the extraction technology "the cancer-related microRNA extraction method in urine," the separation technology "the ultrafast and non-equilibrium separation method for biomolecules," and the detection technology "the highly sensitive electrical measurement method." These research studies are not just limited to the development of biomolecule analysis technology using nanotechnology, but are also opening up a new academic field in analytical chemistry that may lead to the discovery of new pretreatment, separation, and detection principles.
Collapse
Affiliation(s)
- Hiromi Takahashi
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Yoshinobu Baba
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Institute of Quantum Life Science, National Institutes for Quantum Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Takao Yasui
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Japan Science and Technology Agency (JST), PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
29
|
Han X, Zhang Y, Tian J, Wu T, Li Z, Xing F, Fu S. Polymer‐based microfluidic devices: A comprehensive review on preparation and applications. POLYM ENG SCI 2021. [DOI: 10.1002/pen.25831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Xue Han
- School of Physics and Optoelectronic Engineering Shandong University of Technology Zibo China
| | - Yonghui Zhang
- School of Physics and Optoelectronic Engineering Shandong University of Technology Zibo China
| | - Jingkun Tian
- School of Physics and Optoelectronic Engineering Shandong University of Technology Zibo China
| | - Tiange Wu
- School of Physics and Optoelectronic Engineering Shandong University of Technology Zibo China
| | - Zongwen Li
- School of Physics and Optoelectronic Engineering Shandong University of Technology Zibo China
| | - Fei Xing
- School of Physics and Optoelectronic Engineering Shandong University of Technology Zibo China
| | - Shenggui Fu
- School of Physics and Optoelectronic Engineering Shandong University of Technology Zibo China
| |
Collapse
|
30
|
Wang J, Sui L, Huang J, Miao L, Nie Y, Wang K, Yang Z, Huang Q, Gong X, Nan Y, Ai K. MoS 2-based nanocomposites for cancer diagnosis and therapy. Bioact Mater 2021; 6:4209-4242. [PMID: 33997503 PMCID: PMC8102209 DOI: 10.1016/j.bioactmat.2021.04.021] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 04/05/2021] [Accepted: 04/11/2021] [Indexed: 12/24/2022] Open
Abstract
Molybdenum is a trace dietary element necessary for the survival of humans. Some molybdenum-bearing enzymes are involved in key metabolic activities in the human body (such as xanthine oxidase, aldehyde oxidase and sulfite oxidase). Many molybdenum-based compounds have been widely used in biomedical research. Especially, MoS2-nanomaterials have attracted more attention in cancer diagnosis and treatment recently because of their unique physical and chemical properties. MoS2 can adsorb various biomolecules and drug molecules via covalent or non-covalent interactions because it is easy to modify and possess a high specific surface area, improving its tumor targeting and colloidal stability, as well as accuracy and sensitivity for detecting specific biomarkers. At the same time, in the near-infrared (NIR) window, MoS2 has excellent optical absorption and prominent photothermal conversion efficiency, which can achieve NIR-based phototherapy and NIR-responsive controlled drug-release. Significantly, the modified MoS2-nanocomposite can specifically respond to the tumor microenvironment, leading to drug accumulation in the tumor site increased, reducing its side effects on non-cancerous tissues, and improved therapeutic effect. In this review, we introduced the latest developments of MoS2-nanocomposites in cancer diagnosis and therapy, mainly focusing on biosensors, bioimaging, chemotherapy, phototherapy, microwave hyperthermia, and combination therapy. Furthermore, we also discuss the current challenges and prospects of MoS2-nanocomposites in cancer treatment.
Collapse
Affiliation(s)
- Jianling Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Lihua Sui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jia Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Lu Miao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yubing Nie
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Zhichun Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Qiong Huang
- Department of Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xue Gong
- Department of Radiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Yayun Nan
- Geriatric Medical Center, Ningxia People's Hospital, Yinchuan, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|
31
|
Wang C, Xu Y, Zhao X, Li S, Qian Q, Wang W, Mi X. A double-tetrahedral DNA framework based electrochemical biosensor for ultrasensitive detection and release of circulating tumor cells. Analyst 2021; 146:6474-6481. [PMID: 34585683 DOI: 10.1039/d1an01470f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Detecting circulating tumor cells (CTCs) in patients' blood is essential for early diagnosis, precise treatment and prognosis of cancer. Yet due to CTCs being extremely rare in the peripheral blood of patients, it is still a challenge to detect CTCs with high sensitivity and high selectivity. Here, we developed a double-tetrahedral DNA framework (DTDF) based electrochemical biosensor system (E-CTC sensor system) for ultrasensitive detection and release of CTCs. In this work, an upright tetrahedral DNA framework (UTDF) was used as a rigid scaffold to modify a screen-printed gold electrode (SPGE), and an inverted tetrahedral DNA framework (ITDF) provided three vertex chains to multivalently bind with aptamers. Meanwhile, a streptavidin tagged horseradish peroxidase homopolymer (SA-polyHRP) was linked to biotin-modified aptamers to significantly amplify the signal. Moreover, the captured CTCs could be effectively released via benzonase nuclease with little cell damage. Our E-CTC sensor system achieved a linear range from 1 to 105 MCF-7 cells with an ultralow detection limit of 1 cell. The release efficiency reached 88.1%-97.6% and the viability of the released cells reached up to 98%. We also detected the MCF-7 cells in mimic whole blood samples, suggesting that the E-CTC sensor system shows promise for use in clinical research.
Collapse
Affiliation(s)
- Chenguang Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Xu
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xiaoshuang Zhao
- Key Laboratory of Functional Materials for Informatics, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Shuainai Li
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiuling Qian
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Wang
- Shanghai Pudong New District Zhoupu Hospital (Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital), Shanghai 201318, China.
| | - Xianqiang Mi
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Key Laboratory of Functional Materials for Informatics, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China. .,CAS Center for Excellence in Superconducting Electronics (CENSE), Shanghai 200050, China.,Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 310024 Hangzhou, China
| |
Collapse
|
32
|
Yamamoto H, Elbadawy M, Fujisaka K, Sato Y, Ohmori T, Shinohara Y, Hatano Y, Kobayashi D, Gomyo A, Sudo Y, Azakami D, Uchide T, Fukushima R, Morita S, Abugomaa A, Yamawaki H, Kaneda M, Usui T, Sasaki K. Evaluation of the Safety and Feasibility of Apheresis in Dogs: For Application in Metastatic Cancer Research. Animals (Basel) 2021; 11:2770. [PMID: 34679792 PMCID: PMC8532909 DOI: 10.3390/ani11102770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/18/2021] [Accepted: 09/18/2021] [Indexed: 11/17/2022] Open
Abstract
In patients with solid tumors, circulating tumor cells (CTCs) spread in their blood and function as a seed for metastases. However, the study of CTCs has been limited by their rarity, low frequency, and heterogeneity. The efficient collection of CTCs will contribute to further research of metastatic cancers. Apheresis is a process in which the whole blood of an individual is passed through a machine that isolates a particular constituent and returns the remainder to the circulation. In the present study, we investigated the safety and feasibility of apheresis to separate peripheral blood monocytes (PBMCs), whose density is closely similar to that of CTCs, and to capture intravenously administered human breast cancer cells, MCF7s, from the dogs. No life-threatening events were observed in dogs during the apheresis process. The changes in the hemogram were transient and recovered gradually within a few days after apheresis. During apheresis, 50 mL of PBMCs could be collected from each dog. Notably, a thrombus was formed along the circuit wall during apheresis, which decreased the blood collection pressure. MCF7 cells were successfully captured by the apheresis machine. The captured cells were regrown in vitro and characterized compared with the original cells. In conclusion, apheresis could be safely performed in dogs to isolate CTCs with precautions to maintain hemodynamic stability.
Collapse
Affiliation(s)
- Haru Yamamoto
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (H.Y.); (M.E.); (K.F.); (Y.S.); (Y.S.); (A.A.); (K.S.)
| | - Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (H.Y.); (M.E.); (K.F.); (Y.S.); (Y.S.); (A.A.); (K.S.)
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Koudai Fujisaka
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (H.Y.); (M.E.); (K.F.); (Y.S.); (Y.S.); (A.A.); (K.S.)
| | - Yomogi Sato
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (H.Y.); (M.E.); (K.F.); (Y.S.); (Y.S.); (A.A.); (K.S.)
| | - Takahiro Ohmori
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (T.O.); (R.F.); (S.M.)
| | - Yuta Shinohara
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (H.Y.); (M.E.); (K.F.); (Y.S.); (Y.S.); (A.A.); (K.S.)
- Pet Health & Food Division, Iskara Industry Co., Ltd., 1-14-2, Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| | - Yui Hatano
- Laboratory of Clinical Oncology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (Y.H.); (D.K.); (A.G.); (Y.S.)
| | - Daichi Kobayashi
- Laboratory of Clinical Oncology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (Y.H.); (D.K.); (A.G.); (Y.S.)
| | - Ayana Gomyo
- Laboratory of Clinical Oncology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (Y.H.); (D.K.); (A.G.); (Y.S.)
| | - Yuji Sudo
- Laboratory of Clinical Oncology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (Y.H.); (D.K.); (A.G.); (Y.S.)
| | - Daigo Azakami
- Laboratory of Clinical Oncology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (Y.H.); (D.K.); (A.G.); (Y.S.)
| | - Tsuyoshi Uchide
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8538, Japan;
| | - Ryuji Fukushima
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (T.O.); (R.F.); (S.M.)
| | - Shohei Morita
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (T.O.); (R.F.); (S.M.)
| | - Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (H.Y.); (M.E.); (K.F.); (Y.S.); (Y.S.); (A.A.); (K.S.)
- Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Dakahliya, Egypt
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, 35-1, Higashi 23 Ban-cho, Towada, Aomori 034-8628, Japan;
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan;
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (H.Y.); (M.E.); (K.F.); (Y.S.); (Y.S.); (A.A.); (K.S.)
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (H.Y.); (M.E.); (K.F.); (Y.S.); (Y.S.); (A.A.); (K.S.)
| |
Collapse
|
33
|
Zhang X, Wei X, Men X, Wu CX, Bai JJ, Li WT, Yang T, Chen ML, Wang JH. Dual-Multivalent-Aptamer-Conjugated Nanoprobes for Superefficient Discerning of Single Circulating Tumor Cells in a Microfluidic Chip with Inductively Coupled Plasma Mass Spectrometry Detection. ACS APPLIED MATERIALS & INTERFACES 2021; 13:43668-43675. [PMID: 34473482 DOI: 10.1021/acsami.1c11953] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The efficient recognition of circulating tumor cells (CTCs) with an aptamer probe confers numerous benefits; however, the stability and binding affinity of aptamers are significantly hampered in real biological sample matrices. Inspired by the efficient preying mechanism by multiplex tubing feet and endoskeletons of sea urchins, we engineered a superefficient biomimetic single-CTC recognition platform by conjugating dual-multivalent-aptamers (DMAs) Sgc8 and SYL3C onto AuNPs to form a sea urchin-like nanoprobe (sea urchin-DMA-AuNPs). Aptamers Sgc8 and SYL3C selectively bind with the biomarker proteins PTK7 and EpCAM expressed on the surface of CTCs. CTCs were captured with 100% efficiency, followed by sorting on a specially designed multifunctional microfluidic configuration, integrating a single-CTC separation unit and a hydrodynamic filtrating purification unit. After sorting, background-free analysis of biomarker proteins in single CTCs was undertaken with inductively coupled plasma mass spectrometry by measuring the amount of 197Au isotope in sea urchin-DMA-AuNPs. With respect to a single-aptamer nanoprobe/-interface, the dual-aptamer nanoprobe improves the binding efficiency by more than 200% (Kd < 0.35 nM). The microchip facilitates the recognition of single CTCs with a sorting separation rate of 93.6% at a flow rate of 60 μL min-1, and it exhibits 73.8 ± 5.0% measurement efficiency for single CTCs. The present strategy ensures the manipulation and detection of a single CTC in 100 μL of whole blood within 1 h.
Collapse
Affiliation(s)
- Xuan Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Xing Wei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Xue Men
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Cheng-Xin Wu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jun-Jie Bai
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Wei-Tao Li
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Ting Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Ming-Li Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
34
|
Recent Development of Nanomaterials-Based Cytosensors for the Detection of Circulating Tumor Cells. BIOSENSORS-BASEL 2021; 11:bios11080281. [PMID: 34436082 PMCID: PMC8391755 DOI: 10.3390/bios11080281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
The accurate analysis of circulating tumor cells (CTCs) holds great promise in early diagnosis and prognosis of cancers. However, the extremely low abundance of CTCs in peripheral blood samples limits the practical utility of the traditional methods for CTCs detection. Thus, novel and powerful strategies have been proposed for sensitive detection of CTCs. In particular, nanomaterials with exceptional physical and chemical properties have been used to fabricate cytosensors for amplifying the signal and enhancing the sensitivity. In this review, we summarize the recent development of nanomaterials-based optical and electrochemical analytical techniques for CTCs detection, including fluorescence, colorimetry, surface-enhanced Raman scattering, chemiluminescence, electrochemistry, electrochemiluminescence, photoelectrochemistry and so on.
Collapse
|
35
|
Sun N, Lee YT, Kim M, Wang JJ, Zhang C, Teng PC, Qi D, Zhang RY, Tran BV, Lee YT, Ye J, Palomique J, Nissen NN, Han SHB, Sadeghi S, Finn RS, Saab S, Busuttil RW, Posadas EM, Liang L, Pei R, Yang JD, You S, Agopian VG, Tseng HR, Zhu Y. Covalent Chemistry-Mediated Multimarker Purification of Circulating Tumor Cells Enables Noninvasive Detection of Molecular Signatures of Hepatocellular Carcinoma. ADVANCED MATERIALS TECHNOLOGIES 2021; 6:2001056. [PMID: 34212072 PMCID: PMC8240468 DOI: 10.1002/admt.202001056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Indexed: 05/02/2023]
Abstract
Transcriptomic profiling of tumor tissues introduces a large database, which has led to improvements in the ability of cancer diagnosis, treatment, and prevention. However, performing tumor transcriptomic profiling in the clinical setting is very challenging since the procurement of tumor tissues is inherently limited by invasive sampling procedures. Here, we demonstrated the feasibility of purifying hepatocellular carcinoma (HCC) circulating tumor cells (CTCs) from clinical patient samples with improved molecular integrity using Click Chips in conjunction with a multimarker antibody cocktail. The purified CTCs were then subjected to mRNA profiling by NanoString nCounter platform, targeting 64 HCC-specific genes, which were generated from an integrated data analysis framework with 8 tissue-based prognostic gene signatures from 7 publicly available HCC transcriptomic studies. After bioinformatics analysis and comparison, the HCC CTC-derived gene signatures showed high concordance with HCC tissue-derived gene signatures from TCGA database, suggesting that HCC CTCs purified by Click Chips could enable the translation of HCC tissue molecular profiling into a noninvasive setting.
Collapse
Affiliation(s)
- Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Minhyung Kim
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jasmine J Wang
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ceng Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Pai-Chi Teng
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dongping Qi
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Ryan Y Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Benjamin V Tran
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Yue Tung Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Jinglei Ye
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Juvelyn Palomique
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nicholas N Nissen
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Steven-Huy B Han
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Saeed Sadeghi
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Richard S Finn
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Sammy Saab
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Ronald W Busuttil
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Edwin M Posadas
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Li Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, P.R. China
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou 215123, P.R. China
| | - Ju Dong Yang
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sungyong You
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Vatche G Agopian
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| |
Collapse
|
36
|
Cheng SB, Chen MM, Wang YK, Sun ZH, Qin Y, Tian S, Dong WG, Xie M, Huang WH. A Three-Dimensional Conductive Scaffold Microchip for Effective Capture and Recovery of Circulating Tumor Cells with High Purity. Anal Chem 2021; 93:7102-7109. [PMID: 33908770 DOI: 10.1021/acs.analchem.1c00785] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effective acquirement of highly pure circulating tumor cells (CTCs) is very important for CTC-related research. However, it is a great challenge since abundant white blood cells (WBCs) are always co-collected with CTCs because of nonspecific bonding or low depletion rate of WBCs in various CTC isolation platforms. Herein, we designed a three-dimensional (3D) conductive scaffold microchip for highly effective capture and electrochemical release of CTCs with high purity. The conductive 3D scaffold was prepared by dense immobilization of gold nanotubes (Au NTs) on porous polydimethylsiloxane and was functionalized with a CTC-specific biomolecule facilitated by a Au-S bond before embedding into a microfluidic device. The spatially distributed 3D macroporous structure compelled cells to change migration from linear to chaotic and the densely covered Au NTs enhanced the topographic interaction between cells and the substrate, thus synergistically improving the CTC capture efficiency. The Au NT-coated 3D scaffold had good electrical conductivity and the Au-S bond was breakable by voltage exposure so that captured CTCs could be specifically released by electrochemical stimulation while nonspecifically bonded WBCs were not responsive to this process, facilitating recovery of CTCs with high purity. The 3D conductive scaffold microchip was successfully applied to obtain highly pure CTCs from cancer patients' blood, benefiting the downstream analysis of CTCs.
Collapse
Affiliation(s)
- Shi-Bo Cheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Miao-Miao Chen
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Yi-Ke Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Zi-Han Sun
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yu Qin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Shan Tian
- Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei-Guo Dong
- Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Min Xie
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
37
|
Zhang H, Yuan F, Qi Y, Liu B, Chen Q. Circulating Tumor Cells for Glioma. Front Oncol 2021; 11:607150. [PMID: 33777749 PMCID: PMC7987781 DOI: 10.3389/fonc.2021.607150] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/11/2021] [Indexed: 12/13/2022] Open
Abstract
Liquid biopsy has entered clinical applications for several cancers, including metastatic breast, prostate, and colorectal cancer for CTC enumeration and NSCLC for EGFR mutations in ctDNA, and has improved the individualized treatment of many cancers, but relatively little progress has been made in validating circulating biomarkers for brain malignancies. So far, data on circulating tumor cells about glioma are limited, the application of circulating tumor cells as biomarker for glioma patients has only just begun. This article reviews the research status and application prospects of circulating tumor cells in gliomas. Several detection methods and research results of circulating tumor cells about clinical research in gliomas are briefly discussed. The wide application prospect of circulating tumor cells in glioma deserves further exploration, and the research on more sensitive and convenient detection methods is necessary.
Collapse
Affiliation(s)
- Huikai Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fanen Yuan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yangzhi Qi
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
38
|
|
39
|
Wei X, Chen K, Guo S, Liu W, Zhao XZ. Emerging Microfluidic Technologies for the Detection of Circulating Tumor Cells and Fetal Nucleated Red Blood Cells. ACS APPLIED BIO MATERIALS 2021; 4:1140-1155. [DOI: 10.1021/acsabm.0c01325] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
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
- Key Laboratory of Medical Information and 3D Bioprinting of Zhejiang Province, Hangzhou Dianzi University, Hangzhou 310018, 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
| | - Shishang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, 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
| | - 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
|
40
|
Wang J, Zhang R, Ji X, Wang P, Ding C. SERS and fluorescence detection of circulating tumor cells (CTCs) with specific capture-release mode based on multifunctional gold nanomaterials and dual-selective recognition. Anal Chim Acta 2021; 1141:206-213. [PMID: 33248653 DOI: 10.1016/j.aca.2020.10.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 10/16/2020] [Indexed: 01/05/2023]
Abstract
Herein, a dual-selective recognition and multi-enhanced surface-enhanced Raman scattering (SERS)-fluorescence dual mode detection platform is designed for the detection of circulating tumor cells (CTCs). The gold nanoflowers (AuNFs) substrate was synthesized and the CTCs were captured on the surface area of AuNFs/ITO substrate by aptamers modified. At the same time, the novel nanoprobe was designed, anti-EpCAM (AE) and trigger DNA were modified onto the surface of gold nanostars (AuNSs) through a PEG linker. The novel nanoprobe identified CTCs through the specific recognition reaction between AE and the cell epithelial adhesion molecule of the CTCs. The dual-recognition cellular mechanism of the aptamers and AE improves selectivity. Then, the complementary sequence (CS) hybridize with aptamers to release the captured CTCs into the culture medium. The number of CTCs released was detected by SERS and fluorescence. The limit of SERS detection was 5 cells/mL with a linear relationship from 5 to 200 cells/mL. The limit of fluorescence detection was 10 cells/mL with a linear relationship from 10 to 200 cells/mL. Thus, the developed CTCs detection platform demonstrates promising applications for clinical diagnosis.
Collapse
Affiliation(s)
- Jing Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Ruiyuan Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Xiaoting Ji
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Peipei Wang
- Qingdao Central Hospital, Qingdao, 266042, China
| | - Caifeng Ding
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China.
| |
Collapse
|
41
|
Xu L, Li R, Wang Z, Cui H, Li W, Yu M, Guo SS, Zhao XZ. Electrospun degradable Zn-Mn oxide hierarchical nanofibers for specific capture and efficient release of circulating tumor cells. NANOTECHNOLOGY 2020; 31:495102. [PMID: 32990263 DOI: 10.1088/1361-6528/abb48b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Constructing biological affinity devices is considered as an effective strategy for isolating circulating tumor cells (CTCs), and electrospun nanofibers (ESNFs) have recently received attention. However, the current research focuses on polymer fibers, and fabricating stimuli-responsive inorganic nanofibers for cancer diagnosis and analysis is still challenging. In this work, Zn-Mn oxide nanofibers (ZnMnNFs) are used to capture and purify cancer cells after modification with specific antibodies. Then, the hierarchical nanofibers are degraded by reductive weak acid to release the captured cells efficiently without residues. Fusion of Zn and Mn, two transition metals, enhances the surface activity of oxides so that ZnMnNFs are easier to be degraded and modified. By using MCF-7 cancer cells, the cell capture efficiency of ZnMnNFs is up to 88.2%. Furthermore, by using citric acid, it is discovered that, by comparison with Mn oxide nanofibers, the cell release efficiency of ZnMnNFs is improved to 95.1% from 15.4%. In addition, the viability of released cells exceeds 90%. Lastly, the robustness of ZnMnNFs substrates is tested in peripheral blood from breast cancer patients (BCP) and colorectal cancer patients (CCP). Combined with fluorescence labeling, CTCs are confirmed to be isolated from all the clinical samples. This is the first trial of using ternary inorganic ESNFs for cancer cell capture. It is anticipated that the degradable ESNFs will provide biocompatible theranostic platforms and overcome the current limitations of cell release for high-precision gene analysis.
Collapse
Affiliation(s)
- Longguang Xu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Rui Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Zixiang Wang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Heng Cui
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Wei Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Mingxia Yu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, People's Republic of China
| | - Shi-Shang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| |
Collapse
|
42
|
Kong J, Liu Y, Du X, Wang K, Chen W, Huang D, Wei Y, Mao H. Effect of cell-nanostructured substrate interactions on the capture efficiency of HeLa cells. Biomed Mater 2020; 16. [PMID: 33260171 DOI: 10.1088/1748-605x/abcf5c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/01/2020] [Indexed: 11/11/2022]
Abstract
Circulating tumour cells (CTCs) are regarded as an effective biomarker for cancer detection, diagnosis and prognosis monitoring. CTCs capture based on nanostructured substrates is a powerful technique. Some specific adhesion molecule antibody-coated on the surface of nanostructured substrates, such as EpCAM, is commonly used to enhance CTCs capture efficiency. Substrate nanotopographies regulate the interaction between the substrates and captured cells, further influencing cell capture efficiency. However, the relationship between cell capture efficiency and cell-substrate interaction remains poorly understood. Here, we explored the relationship between cell capture efficiency and cell-substrate interaction based on two sets of nanostructures with different nanotopographies without antibody conjugation. Given the urgent demand of improving capture efficiency of EpCAM-negative cells, we used HeLa (EpCAM-negative) cells as the main targets. We demonstrated that HeLa cells could be more effectively captured by two nanostructural substrates, especially by DCNFs. Therefore, the morphological and migrating interaction between HeLa cells and distinct substrates were associated with cell capture efficiency. Our findings demonstrated the potential mechanism for optimizing the nanotopography for higher capture efficiency, and provide a potential foundation for cancer detection, diagnosis and treatment.
Collapse
Affiliation(s)
- Jinlong Kong
- Taiyuan University of Technology, Taiyuan University of Technology, Taiyuan, Shanxi , 030024, CHINA
| | - Yang Liu
- Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, Beijing, 100864, CHINA
| | - Xiangbin Du
- Taiyuan University of Technology, Taiyuan University of Technology, Taiyuan, Shanxi , 030024, CHINA
| | - Kaiqun Wang
- Taiyuan University of Technology, Taiyuan University of Technology, Taiyuan, Shanxi , 030024, CHINA
| | - Weiyi Chen
- Taiyuan University of Technology, Taiyuan University of Technology, Taiyuan, Shanxi , 030024, CHINA
| | - Di Huang
- Taiyuan University of Technology, Taiyuan University of Technology, Taiyuan, Shanxi , 030024, CHINA
| | - Yan Wei
- Taiyuan University of Technology, Taiyuan University of Technology, Taiyuan, 030024, CHINA
| | - Haiyang Mao
- Key Laboratory of Microelectronics Devices and Integrated Technology, Institute of Microelectronics pf Chinese Academy of Sciences, Beijing, Beijing, 100029, CHINA
| |
Collapse
|
43
|
Pei H, Li L, Han Z, Wang Y, Tang B. Recent advances in microfluidic technologies for circulating tumor cells: enrichment, single-cell analysis, and liquid biopsy for clinical applications. LAB ON A CHIP 2020; 20:3854-3875. [PMID: 33107879 DOI: 10.1039/d0lc00577k] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Circulating tumor cells (CTCs) detach from primary or metastatic lesions and circulate in the peripheral blood, which is considered to be the cause of distant metastases. CTC analysis in the form of liquid biopsy, enumeration and molecular analysis provide significant clinical information for cancer diagnosis, prognosis and therapeutic strategies. Despite the great clinical value, CTC analysis has not yet entered routine clinical practice due to lack of efficient technologies to perform CTC isolation and single-cell analysis. Taking the rarity and inherent heterogeneity of CTCs into account, reliable methods for CTC isolation and detection are in urgent demand for obtaining valuable information on cancer metastasis and progression from CTCs. Microfluidic technology, featuring microfabricated structures, can precisely control fluids and cells at the micrometer scale, thus making itself a particularly suitable method for rare CTC manipulation. Besides the enrichment function, microfluidic chips can also realize the analysis function by integrating multiple detection technologies. In this review, we have summarized the recent progress in CTC isolation and detection using microfluidic technologies, with special attention to emerging direct enrichment and enumeration in vivo. Further, few insights into single CTC molecular analysis are also demonstrated. We have provided a review of potential clinical applications of CTCs, ranging from early screening and diagnosis, tumor progression and prognosis, treatment and resistance monitoring, to therapeutic evaluation. Through this review, we conclude that the clinical utility of CTCs will be expanded as the isolation and analysis techniques are constantly improving.
Collapse
Affiliation(s)
- Haimeng Pei
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | | | | | | | | |
Collapse
|
44
|
Jiang W, Han L, Yang L, Xu T, He J, Peng R, Liu Z, Zhang C, Yu X, Jia L. Natural Fish Trap-Like Nanocage for Label-Free Capture of Circulating Tumor Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002259. [PMID: 33240774 PMCID: PMC7675191 DOI: 10.1002/advs.202002259] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/04/2020] [Indexed: 06/11/2023]
Abstract
Nanomaterials have achieved several breakthroughs in the capture of circulating tumor cells (CTCs) over the past decades. However, artificial fabrication of label-free nanomaterials used for high-efficiency CTC capture is still a challenge. Through billions of years of evolution and natural selection, various complicated and precise hierarchical structures are developed. Here, a novel fish trap-like "nanocage" structure derived from the natural Chrysanthemum pollen is reported and a nanocage-featured film for the label-free capture of CTCs and CTC clusters is constructed. The nanocage-featured film effectively captures 92% rare cancer cells with a broad spectrum of cancer types, due to the synergistic effect of nanocage-CTC filopodia matching, high contact area, and strong adhesion force between the cancer cells and the nanocage. Furthermore, the nanocage-featured film successfully detects CTCs and CTC clusters in 2 or 4 mL blood taken from 21 cancer patients (stages I-IV) suffering from various types of cancers. This novel, abundant, and economical fish trap-like "nanocage" may provide new perspectives for the application of natural nanomaterials in clinical CTC capture and analysis.
Collapse
Affiliation(s)
- Wenning Jiang
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Lulu Han
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Liwei Yang
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Ting Xu
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Jiabei He
- Department of OncologyThe Dalian Municipal Central Hospital Affiliated of Dalian Medical UniversityDalian116033P. R. China
| | - Ruilian Peng
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Ziyu Liu
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Chong Zhang
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| | - Xiaomin Yu
- Department of OncologyThe Dalian Municipal Central Hospital Affiliated of Dalian Medical UniversityDalian116033P. R. China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and ImagingSchool of BioengineeringDalian University of TechnologyDalian116023P. R. China
| |
Collapse
|
45
|
Shenoy BM, Hegde G, Roy Mahapatra D. Field enhancement in microfluidic semiconductor nanowire array. BIOMICROFLUIDICS 2020; 14:064102. [PMID: 33163137 PMCID: PMC7609134 DOI: 10.1063/5.0028899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/15/2020] [Indexed: 06/11/2023]
Abstract
Nano-material integrated microfluidic platforms are increasingly being considered to accelerate biological sample preparation and molecular diagnostics. A major challenge in this context is the generation of high electric fields for electroporation of cell membranes. In this paper, we have studied a novel mechanism of generating a high electric field in the microfluidic channels by using an array of semiconductor nanowires. When an electrostatic field is applied across a semiconductor nanowire array, the electric field is localized near the nanowires and the field strength is higher than what was reported previously with various other micro-geometries. Nanowires made of ZnO, Si, and Si-SiO2 and their orientation and array spacing are considered design parameters. It is observed that for a given ratio of the spacing between nanowires to the diameter, the electric field enhancement near the edges of ZnO nanowires is nearly 30 times higher compared to Si or Si-SiO2 nanowire arrays. This enhancement is a combined effect of the unique geometry with a pointed tip with a hexagonal cross section, the piezoelectric and the spontaneous polarization in the ZnO nanowires, and the electro-kinetics of the interface fluid. Considering the field localization phenomena, the trajectories of E. coli cells in the channel are analyzed. For a given inter-nanowire spacing and an applied electric field, the channels with ZnO nanowire arrays have a greater probability of cell lysis in comparison to Si-based nanowire arrays. Detailed correlations between the cell lysis probability with the inter-nanowire spacing and the applied electric field are reported.
Collapse
Affiliation(s)
- Bhamy Maithry Shenoy
- Department of Aerospace Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Gopalkrishna Hegde
- BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - D. Roy Mahapatra
- Department of Aerospace Engineering, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
46
|
Qin W, Chen L, Wang Z, Li Q, Fan C, Wu M, Zhang Y. Bioinspired DNA Nanointerface with Anisotropic Aptamers for Accurate Capture of Circulating Tumor Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000647. [PMID: 33042737 PMCID: PMC7539197 DOI: 10.1002/advs.202000647] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/29/2020] [Indexed: 05/08/2023]
Abstract
The capture and analysis of circulating tumor cells (CTCs) have provided a non-invasive entry for cancer diagnosis and disease monitoring. Despite recent development in affinity-based CTCs isolation, it remains challenging to achieve efficient capture toward CTCs with dynamic surface expression. Enlightened by the synergistic effect insideimmune synapses, the development of a nanointerface engineered with topology-defined anisotropic aptamers programmed by DNA scaffold (DNA nanosynapse), for accurate CTCs isolation, is herein reported. As compared to isotropic aptamers, the DNA nanosynapse exhibits enhanced anchoring on the cell membrane with both high and low epithelial cell adhesion molecule (EpCAM) expression. This nanointerface enables accurate capture toward CTCs of heterogeneous EpCAM, without dramatically proportional change inside the mixture of diverse phenotypes. By applying this nanoplatform, CTCs detection as well as downstream analysis for measuring disease status can be achieved in clinical samples from breast cancer patients.
Collapse
Affiliation(s)
- Weiwei Qin
- Guangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhouGuangdong510006China
- College of Materials and EnergySouth China Agricultural UniversityGuangzhouGuangdong510642China
- State Key Laboratory of Chemo/Biosensing and ChemometricsHunan UniversityChangsha410082China
| | - Liang Chen
- Guangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhouGuangdong510006China
| | - Zhiru Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhouGuangdong510006China
| | - Qian Li
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Chunhai Fan
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Minhao Wu
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Yuanqing Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhouGuangdong510006China
| |
Collapse
|
47
|
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: 2.5] [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
|
48
|
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: 6.5] [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
|
49
|
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: 1.3] [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
|
50
|
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: 26] [Impact Index Per Article: 6.5] [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
|