1
|
Notarbartolo M, Alfieri ML, Avolio R, Ball V, Errico ME, Massaro M, Puglisi R, Sànchez-Espejo R, Viseras C, Riela S. Design of innovative and low-cost dopamine-biotin conjugate sensor for the efficient detection of protein and cancer cells. J Colloid Interface Sci 2025; 678:766-775. [PMID: 39307064 DOI: 10.1016/j.jcis.2024.09.145] [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: 06/13/2024] [Revised: 08/30/2024] [Accepted: 09/15/2024] [Indexed: 10/27/2024]
Abstract
The rapid, precise identification and quantification of specific biomarkers, toxins, or pathogens is currently a key strategy for achieving more efficient diagnoses. Herein a dopamine-biotin monomer was synthetized and oxidized in the presence of hexamethylenediamine, to obtain adhesive coatings based on polydopamine-biotin (PDA-BT) on different materials to be used in targeted molecular therapy. Insight into the structure of the PDA-BT coating was obtained by solid-state 13C NMR spectroscopy acquired, for the first time, directly onto the coating, deposited on alumina spheres. The receptor binding capacity of the PDA-BT coating toward 4-hydroxyazobenzene-2-carboxylic acid/Avidin complex was verified by means of UV-vis spectroscopy. Different deposition cycles of avidin onto the PDA-BT coating by layer-by-layer assembly showed that the film retains its receptor binding capacity for at least eight consecutive cycles. Finally, the feasibility of PDA-BT coating to recognize cell lines with different grade of overexpression of biotin receptors (BR) was investigated by tumor cell capture experiments by using MCF-7 (BR+) and HL-60 (BR-) cell lines. The results show that the developed system can selectively capture MCF-7 cells indicating that it could represent a first approach for the development of future more sophisticated biosensors easily accessible, low cost and recyclable with the dual and rapid detection of both proteins and cells.
Collapse
Affiliation(s)
- Monica Notarbartolo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, Parco d'Orleans II, Ed. 16-17, 90128 Palermo, Italy
| | - Maria Laura Alfieri
- Department of Chemical Sciences, University of Naples "Federico II", I-80126 Naples, Italy.
| | - Roberto Avolio
- Institute of Chemistry and Technology of Polymers, National Council of Research (CNR), via Campi Flegrei 34, Pozzuoli I-80078, Italy
| | - Vincent Ball
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elizabeth, 67000 Strasbourg. France; Institut National de la Santé et de la Recherche Médicale, Unité mixte de rechere 1121. 1 rue Eugène Boeckel, 67084 Strasbourg Cedex, France
| | - Maria Emanuela Errico
- Institute of Chemistry and Technology of Polymers, National Council of Research (CNR), via Campi Flegrei 34, Pozzuoli I-80078, Italy
| | - Marina Massaro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, Parco d'Orleans II, Ed. 16-17, 90128 Palermo, Italy.
| | - Roberta Puglisi
- Dipartimento di Scienze Chimiche (DSC), Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Rita Sànchez-Espejo
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Campus Universitario de Cartuja, 18071 Granada, Spain
| | - César Viseras
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Campus Universitario de Cartuja, 18071 Granada, Spain; Andalusian Institute of Earth Sciences, CSIC-UGR, 18100 Armilla, Granada, Spain
| | - Serena Riela
- Dipartimento di Scienze Chimiche (DSC), Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| |
Collapse
|
2
|
Zhang G, Huang X, Gong Y, Ding Y, Wang H, Zhang H, Wu L, Su R, Yang C, Zhu Z. Fingerprint Profiling of Glycans on Extracellular Vesicles via Lectin-Induced Aggregation Strategy for Precise Cancer Diagnostics. J Am Chem Soc 2024; 146:29053-29063. [PMID: 39235449 DOI: 10.1021/jacs.4c10390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Extracellular vesicles (EVs) harbor abundant glycans that mediate various functions, such as intercellular communication and disease advancement, which play significant roles in disease progression. However, the presence of EV heterogeneity in body fluids and the complex nature of the glycan structures have posed challenges for the detection of EV glycans. In this study, we provide a streamlined method integrated, membrane-specific separation with lectin-induced aggregation strategy (MESSAGE), for multiplexed profiling of EV glycans. By leveraging a rationally designed lectin-induced aggregation strategy, the expression of EV glycans is converted to size-based signals. With the assistance learning machine algorithms, the MESSAGE strategy with high sensitivity, specificity, and simplicity can be used for early cancer diagnosis and classification, as well as monitoring cancer metastasis via 20 μL plasma sample within 2 h. Furthermore, our platform holds promise for advancing the field of EV-based liquid biopsy for clinical applications, opening new possibilities for the profiling of EV glycan signatures in various disease states.
Collapse
Affiliation(s)
- Guihua Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiaodan Huang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yanli Gong
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yue Ding
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hua Wang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Huimin Zhang
- Innovation Laboratory for Sciences, Technologies of Energy Materials of Fujian Province, Xiamen 361000, China
| | - Lingling Wu
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Rui Su
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Chaoyong Yang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhi Zhu
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
3
|
Ajiboye IO, Banerjee RK. Surface Reaction of Electroosmotic Flow-Driven Free Antigens With Immobilized Magnetic-Microbeads-Tagged-Antibodies in Microchannels. J Biomech Eng 2024; 146:091003. [PMID: 38511298 DOI: 10.1115/1.4065138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
Immunoassays based on reactions between target pathogen (antigen; Ag) and antibody (Ab) are frequently used for Ag detection. An external magnetic field was used to immobilize magnetic microbeads-tagged-antibodies (mMB-Ab) on the surface of a microchannel in the capture zone. The mMB-Ab was subsequently used for Ag detection. The objective of this numerical study, with experimental validation, is to assess the surface reaction between mMB-Ab and Ag in the presence of electro-osmotic flow (EOF). First, immobilization of mMB-Ab complex in the wall of the capture zone was achieved. Subsequently, the Ag was transported by EOF toward the capture zone to bind with the immobilized mMB-Ab. Lastly, mMB-Ab:Ag complex was formed and immobilized in the capture zone. A finite volume solver was used to implement the above steps. The surface reaction between the mMB-Ab and Ag was investigated in the presence of electric fields (E): 150 V/cm-450 V/cm and Ag concentrations: 0.001 M-1000 M. The depletion of mMB-Ab increases with time as the E decreases. Furthermore, as the concentration of Ag decreases, the depletion of mMB-Ab increases with time. These results quantify the detection of Ag using the EOF device; thus, signifying its potential for rapid throughput screening of Ag. This platform technology can lead to the development of portable devices for the detection of target cells, pathogens, and biomolecules for testing water systems, biological fluids, and biochemicals.
Collapse
Affiliation(s)
- Israel O Ajiboye
- Department of Mechanical and Materials Engineering, University of Cincinnati, Rhodes Hall 601, 2851 Woodside Drive, Cincinnati, OH 45219
| | - Rupak K Banerjee
- Department of Biomedical Engineering, University of Cincinnati, Veterans Affairs Medical Center, Rhodes Hall 593, 2851 Woodside Drive, Cincinnati, OH 45219
| |
Collapse
|
4
|
Fathi-Karkan S, Sargazi S, Shojaei S, Farasati Far B, Mirinejad S, Cordani M, Khosravi A, Zarrabi A, Ghavami S. Biotin-functionalized nanoparticles: an overview of recent trends in cancer detection. NANOSCALE 2024; 16:12750-12792. [PMID: 38899396 DOI: 10.1039/d4nr00634h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Electrochemical bio-sensing is a potent and efficient method for converting various biological recognition events into voltage, current, and impedance electrical signals. Biochemical sensors are now a common part of medical applications, such as detecting blood glucose levels, detecting food pathogens, and detecting specific cancers. As an exciting feature, bio-affinity couples, such as proteins with aptamers, ligands, paired nucleotides, and antibodies with antigens, are commonly used as bio-sensitive elements in electrochemical biosensors. Biotin-avidin interactions have been utilized for various purposes in recent years, such as targeting drugs, diagnosing clinically, labeling immunologically, biotechnology, biomedical engineering, and separating or purifying biomolecular compounds. The interaction between biotin and avidin is widely regarded as one of the most robust and reliable noncovalent interactions due to its high bi-affinity and ability to remain selective and accurate under various reaction conditions and bio-molecular attachments. More recently, there have been numerous attempts to develop electrochemical sensors to sense circulating cancer cells and the measurement of intracellular levels of protein thiols, formaldehyde, vitamin-targeted polymers, huwentoxin-I, anti-human antibodies, and a variety of tumor markers (including alpha-fetoprotein, epidermal growth factor receptor, prostate-specific Ag, carcinoembryonic Ag, cancer antigen 125, cancer antigen 15-3, etc.). Still, the non-specific binding of biotin to endogenous biotin-binding proteins present in biological samples can result in false-positive signals and hinder the accurate detection of cancer biomarkers. This review summarizes various categories of biotin-functional nanoparticles designed to detect such biomarkers and highlights some challenges in using them as diagnostic tools.
Collapse
Affiliation(s)
- Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166 Iran.
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd 9414974877, Iran.
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shirin Shojaei
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Bahareh Farasati Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran.
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Turkiye.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye.
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600 077, India
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
5
|
Cai Q, He Y, Zhou Y, Zheng J, Deng J. Nanomaterial-Based Strategies for Preventing Tumor Metastasis by Interrupting the Metastatic Biological Processes. Adv Healthc Mater 2024; 13:e2303543. [PMID: 38411537 DOI: 10.1002/adhm.202303543] [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: 10/17/2023] [Revised: 02/01/2024] [Indexed: 02/28/2024]
Abstract
Tumor metastasis is the primary cause of cancer-related deaths. The prevention of tumor metastasis has garnered notable interest and interrupting metastatic biological processes is considered a potential strategy for preventing tumor metastasis. The tumor microenvironment (TME), circulating tumor cells (CTCs), and premetastatic niche (PMN) play crucial roles in metastatic biological processes. These processes can be interrupted using nanomaterials due to their excellent physicochemical properties. However, most studies have focused on only one aspect of tumor metastasis. Here, the hypothesis that nanomaterials can be used to target metastatic biological processes and explore strategies to prevent tumor metastasis is highlighted. First, the metastatic biological processes and strategies involving nanomaterials acting on the TME, CTCs, and PMN to prevent tumor metastasis are briefly summarized. Further, the current challenges and prospects of nanomaterials in preventing tumor metastasis by interrupting metastatic biological processes are discussed. Nanomaterial-and multifunctional nanomaterial-based strategies for preventing tumor metastasis are advantageous for the long-term fight against tumor metastasis and their continued exploration will facilitate rapid progress in the prevention, diagnosis, and treatment of tumor metastasis. Novel perspectives are outlined for developing more effective strategies to prevent tumor metastasis, thereby improving the outcomes of patients with cancer.
Collapse
Affiliation(s)
- Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yijia He
- School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yang Zhou
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
6
|
Li Q, Wang Y, Gao W, Qian G, Chen X, Liu Y, Yu S. A microfluidic device for enhanced capture and high activity release of heterogeneous CTCs from whole blood. Talanta 2024; 266:125007. [PMID: 37556952 DOI: 10.1016/j.talanta.2023.125007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
Circulating tumor cells (CTCs) are tumor cells that spontaneously detach from the primary focus, and early detection and characterization of CTCs is vital for cancer diagnosis and appropriate treatment. Current methods commonly use EpCAM to capture CTCs, but this results in a loss of information on other CTC subsets (EpCAM-negative cells) due to the heterogeneity of CTCs. Here, we report a novel microfluidic device that integrates the capture and release of heterogeneous CTCs directly from whole blood. A spiral chip was designed for the separation of differently sized cells, and larger CTCs were effectively separated from smaller blood cells with a 98% recovery rate. CD146-containing magnetic beads were used to complement the EpCAM-based CTC capture methods, and the capture efficiency of Fe3O4@Gelatin@CD146/EpCAM increased by 20% over Fe3O4@Gelatin@EpCAM. Finally, MMP-9 was employed to release CTCs with high efficiency and less damage by degrading gelatins on the surface of Fe3O4. The established method was successfully applied to CTC capture and release in a simulated patient's whole blood. The developed method achieved enhanced capture and high activity release of heterogeneous CTCs with less interference by blood cells, which contributes to the early detection and clinical downstream analysis of CTCs.
Collapse
Affiliation(s)
- Qiaoyu Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China; Institute of Mass Spectrometry, School of Material Science and Chemical Engineering; Ningbo University, Ningbo, Zhejiang, 315211, China; Department of Chemistry, Fudan University, Shanghai 200438, China
| | - Yanlin Wang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China; Institute of Mass Spectrometry, School of Material Science and Chemical Engineering; Ningbo University, Ningbo, Zhejiang, 315211, China; Department of Chemistry, Fudan University, Shanghai 200438, China
| | - Wenjing Gao
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering; Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Guoqing Qian
- Department of Infectious Diseases, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Xueqin Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yushan Liu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Shaoning Yu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China; Institute of Mass Spectrometry, School of Material Science and Chemical Engineering; Ningbo University, Ningbo, Zhejiang, 315211, China; Department of Chemistry, Fudan University, Shanghai 200438, China.
| |
Collapse
|
7
|
Nguyen TNA, Huang PS, Chu PY, Hsieh CH, Wu MH. Recent Progress in Enhanced Cancer Diagnosis, Prognosis, and Monitoring Using a Combined Analysis of the Number of Circulating Tumor Cells (CTCs) and Other Clinical Parameters. Cancers (Basel) 2023; 15:5372. [PMID: 38001632 PMCID: PMC10670359 DOI: 10.3390/cancers15225372] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Analysis of circulating tumor cells (CTCs) holds promise to diagnose cancer or monitor its development. Among the methods, counting CTC numbers in blood samples could be the simplest way to implement it. Nevertheless, its clinical utility has not yet been fully accepted. The reasons could be due to the rarity and heterogeneity of CTCs in blood samples that could lead to misleading results from assays only based on single CTC counts. To address this issue, a feasible direction is to combine the CTC counts with other clinical data for analysis. Recent studies have demonstrated the use of this new strategy for early detection and prognosis evaluation of cancers, or even for the distinguishment of cancers with different stages. Overall, this approach could pave a new path to improve the technical problems in the clinical applications of CTC counting techniques. In this review, the information relevant to CTCs, including their characteristics, clinical use of CTC counting, and technologies for CTC enrichment, were first introduced. This was followed by discussing the challenges and new perspectives of CTC counting techniques for clinical applications. Finally, the advantages and the recent progress in combining CTC counts with other clinical parameters for clinical applications have been discussed.
Collapse
Affiliation(s)
- Thi Ngoc Anh Nguyen
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (T.N.A.N.); (P.-S.H.); (P.-Y.C.)
| | - Po-Shuan Huang
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (T.N.A.N.); (P.-S.H.); (P.-Y.C.)
| | - Po-Yu Chu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (T.N.A.N.); (P.-S.H.); (P.-Y.C.)
| | - Chia-Hsun Hsieh
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei City Municipal TuCheng Hospital, New Taipei City 23652, Taiwan;
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33302, Taiwan
| | - Min-Hsien Wu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (T.N.A.N.); (P.-S.H.); (P.-Y.C.)
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei City Municipal TuCheng Hospital, New Taipei City 23652, Taiwan;
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33302, Taiwan
| |
Collapse
|
8
|
Chen Z, Yue Z, Yang K, Shen C, Cheng Z, Zhou X, Li S. Four Ounces Can Move a Thousand Pounds: The Enormous Value of Nanomaterials in Tumor Immunotherapy. Adv Healthc Mater 2023; 12:e2300882. [PMID: 37539730 DOI: 10.1002/adhm.202300882] [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: 03/19/2023] [Revised: 07/17/2023] [Indexed: 08/05/2023]
Abstract
The application of nanomaterials in healthcare has emerged as a promising strategy due to their unique structural diversity, surface properties, and compositional diversity. In particular, nanomaterials have found a significant role in improving drug delivery and inhibiting the growth and metastasis of tumor cells. Moreover, recent studies have highlighted their potential in modulating the tumor microenvironment (TME) and enhancing the activity of immune cells to improve tumor therapy efficacy. Various types of nanomaterials are currently utilized as drug carriers, immunosuppressants, immune activators, immunoassay reagents, and more for tumor immunotherapy. Necessarily, nanomaterials used for tumor immunotherapy can be grouped into two categories: organic and inorganic nanomaterials. Though both have shown the ability to achieve the purpose of tumor immunotherapy, their composition and structural properties result in differences in their mechanisms and modes of action. Organic nanomaterials can be further divided into organic polymers, cell membranes, nanoemulsion-modified, and hydrogel forms. At the same time, inorganic nanomaterials can be broadly classified as nonmetallic and metallic nanomaterials. The current work aims to explore the mechanisms of action of these different types of nanomaterials and their prospects for promoting tumor immunotherapy.
Collapse
Affiliation(s)
- Ziyin Chen
- Department of Urology, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Ziqi Yue
- Department of Forensic Medicine, Harbin Medical University, 150001, Harbin, P. R. China
| | - Kaiqi Yang
- Clinical Medicine, Harbin Medical University, 150001, Harbin, P. R. China
| | - Congrong Shen
- Department of Urology, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Zhe Cheng
- Department of Forensic Medicine, Harbin Medical University, 150001, Harbin, P. R. China
| | - Xiaofeng Zhou
- Department of Urology, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, 110042, Shenyang, P. R. China
- The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, Shenyang, 110042, China
| |
Collapse
|
9
|
Yang L, Zhang Y, Dong X, Wang H, Liu Z. Organic solvent-free solid-phase extraction of acetamiprid in food samples using Strep-Tag system integrated apta-magnetic sorbents. Food Chem 2023; 423:136398. [PMID: 37247526 DOI: 10.1016/j.foodchem.2023.136398] [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: 12/14/2022] [Revised: 05/10/2023] [Accepted: 05/14/2023] [Indexed: 05/31/2023]
Abstract
A novel magnetic sorbent, named ABA-(Strep-tag II)-STMB, was prepared by modifying the acetamiprid-binding aptamer (ABA) onto Strep-Tactin-coated magnetic beads (STMBs) via Strep-tag II. The integration of the Strep-tag system allows the elution of the target by the addition of D-biotin, which can compete with Strep-tag II for the Strep-Tactin sites on the magnetic beads, instead of organic solvents. The sorbent showed good selectivity and reusability, and the extraction efficiency could still reach 90.5 % after 8 reuses. Under the optimized conditions, the developed magnetic solid-phase extraction (MSPE) method exhibited good linearity in the range of 0.1-100 μM, with the limits of detection (LOD) of 0.017-0.019 μM, and the limits of quantification (LOQ) of 0.057-0.066 μM. The relative standard deviations (RSDs) were below 5.51 %. The spiked recoveries were 84.4 %-96.0 %. The analysis results were in good agreement with those of the QuEChERS method.
Collapse
Affiliation(s)
- Limin Yang
- State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao 266580, PR China.
| | - Yiping Zhang
- State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao 266580, PR China
| | - Xingdong Dong
- State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao 266580, PR China
| | - Hao Wang
- State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao 266580, PR China
| | - Zhen Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518055, PR China.
| |
Collapse
|
10
|
Xiang Y, Zhang H, Lu H, Wei B, Su C, Qin X, Fang M, Li X, Yang F. Bioorthogonal Microbubbles with Antifouling Nanofilm for Instant and Suspended Enrichment of Circulating Tumor Cells. ACS NANO 2023; 17:9633-9646. [PMID: 37144647 DOI: 10.1021/acsnano.3c03194] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Integrating clinical rare cell enrichment, culture, and single-cell phenotypic profiling is currently hampered by the lack of competent technologies, which typically suffer from weak cell-interface collision affinity, strong nonspecific adsorption, and the potential uptake. Here, we report cells-on-a-bubble, a bioinspired, self-powered bioorthogonal microbubble (click bubble) that leverages a clickable antifouling nanointerface and a DNA-assembled sucker-like polyvalent cell surface, to enable instant and suspended isolation of circulating tumor cells (CTCs) within minutes. Using this biomimetic engineering strategy, click bubbles achieve a capture efficiency of up to 98%, improved by 20% at 15 times faster over their monovalent counterparts. Further, the buoyancy-activated bubble facilitates self-separation, 3D suspension culture, and in situ phenotyping of the captured single cancer cells. By using a multiantibody design, this fast, affordable micromotor-like click bubble enables suspended enrichment of CTCs in a cohort (n = 42) across three cancer types and treatment response evaluation, signifying its great potential to enable single-cell analysis and 3D organoid culture.
Collapse
Affiliation(s)
- Yuanhang Xiang
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Hui Zhang
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Hao Lu
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Binqi Wei
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Cuiyun Su
- Department of Respiratory Oncology, Department of Clinical Laboratory, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xiaojie Qin
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Min Fang
- Department of Respiratory Oncology, Department of Clinical Laboratory, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xinchun Li
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Fan Yang
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
11
|
Guo L, Liu C, Qi M, Cheng L, Wang L, Li C, Dong B. Recent progress of nanostructure-based enrichment of circulating tumor cells and downstream analysis. LAB ON A CHIP 2023; 23:1493-1523. [PMID: 36776104 DOI: 10.1039/d2lc00890d] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The isolation and detection of circulating tumor cells (CTCs) play an important role in early cancer diagnosis and prognosis, providing easy access to identify metastatic cells before clinically detectable metastases. In the past 20 years, according to the heterogeneous expression of CTCs on the surface and their special physical properties (size, morphology, electricity, etc.), a series of in vitro enrichment methods of CTCs have been developed based on microfluidic chip technology, nanomaterials and various nanostructures. In recent years, the in vivo detection of CTCs has attracted considerable attention. Photoacoustic flow cytometry and fluorescence flow cytometry were used to detect CTCs in a noninvasive manner. In addition, flexible magnetic wire and indwelling intravascular non-circulating CTCs isolation system were developed for in vivo CTCs study. In the aspect of downstream analysis, gene analysis and drug sensitivity tests of enriched CTCs were developed based on various existing molecular analysis techniques. All of these studies constitute a complete study of CTCs. Although the existing reviews mainly focus on one aspect of capturing CTCs study, a review that includes the in vivo and in vitro capture and downstream analysis study of CTCs is highly needed. This review focuses on not only the classic work and latest research progress in in vitro capture but also includes the in vivo capture and downstream analysis, discussing the advantages and significance of the different research methods and providing new ideas for solving the heterogeneity and rarity of CTCs.
Collapse
Affiliation(s)
- Lihua Guo
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, P. R. China.
| | - Chang Liu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, P. R. China.
| | - Manlin Qi
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School of Stomatology, Jilin University, Changchun, 130021, P. R. China.
| | - Liang Cheng
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School of Stomatology, Jilin University, Changchun, 130021, P. R. China.
| | - Lin Wang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School of Stomatology, Jilin University, Changchun, 130021, P. R. China.
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China.
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, P. R. China.
| |
Collapse
|
12
|
Guo X, Hu F, Zhao S, Yong Z, Zhang Z, Peng N. Immunomagnetic Separation Method Integrated with the Strep-Tag II System for Rapid Enrichment and Mild Release of Exosomes. Anal Chem 2023; 95:3569-3576. [PMID: 36661256 DOI: 10.1021/acs.analchem.2c03470] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Exosomes are important participants in numerous pathophysiological processes and hold promising application value in cancer diagnosis, monitoring, and prognosis. However, the small size (40-160 nm) and high heterogeneity of exosomes make it still challenging to enrich exosomes efficiently from the complex biological fluid microenvironment, which has largely restricted their downstream analysis and clinical application. In this work, we introduced a novel method for rapid isolation and mild release of exosomes from the cell culture supernatant. A Strep-tag II-based immunomagnetic isolation (SIMI) system was constructed by modifying the capture antibodies onto magnetic nanoparticles through specific and reversible recognition between Strep-Tactin and Strep-tag II. Due to their high affinity and binding selectivity, exosomes could be isolated within 38 min with an isolation efficiency of 82.5% and a release efficiency of 62%. Compared with the gold-standard ultracentrifugation, the SIMI system could harvest nearly 59% more exosomes from the 293 T cell culture medium with shorter isolation time and higher purity. In addition, cellular uptake assay indicated that exosomes released from magnetic nanoparticles could maintain their high biological activity. These superior characteristics show that this novel method is a fast, efficient, and nondestructive exosome isolation tool and thus could potentially be further utilized in various exosome-related applications, e.g., disease diagnosis and drug delivery.
Collapse
Affiliation(s)
- Xiaoniu Guo
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Fei Hu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China.,Xi'an Key Laboratory of Biomedical Testing and High-End Equipment, Xi'an 710049, Shaanxi, China
| | - Shuhao Zhao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Zhang Yong
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Zengming Zhang
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Niancai Peng
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China.,Xi'an Key Laboratory of Biomedical Testing and High-End Equipment, Xi'an 710049, Shaanxi, China
| |
Collapse
|
13
|
Vidlarova M, Rehulkova A, Stejskal P, Prokopova A, Slavik H, Hajduch M, Srovnal J. Recent Advances in Methods for Circulating Tumor Cell Detection. Int J Mol Sci 2023; 24:3902. [PMID: 36835311 PMCID: PMC9959336 DOI: 10.3390/ijms24043902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/06/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
Circulating tumor cells (CTCs) are released from primary tumors and transported through the body via blood or lymphatic vessels before settling to form micrometastases under suitable conditions. Accordingly, several studies have identified CTCs as a negative prognostic factor for survival in many types of cancer. CTCs also reflect the current heterogeneity and genetic and biological state of tumors; so, their study can provide valuable insights into tumor progression, cell senescence, and cancer dormancy. Diverse methods with differing specificity, utility, costs, and sensitivity have been developed for isolating and characterizing CTCs. Additionally, novel techniques with the potential to overcome the limitations of existing ones are being developed. This primary literature review describes the current and emerging methods for enriching, detecting, isolating, and characterizing CTCs.
Collapse
Affiliation(s)
- Monika Vidlarova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University in Olomouc, 779 00 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital in Olomouc, 779 00 Olomouc, Czech Republic
| | - Alona Rehulkova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University in Olomouc, 779 00 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital in Olomouc, 779 00 Olomouc, Czech Republic
| | - Pavel Stejskal
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University in Olomouc, 779 00 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital in Olomouc, 779 00 Olomouc, Czech Republic
| | - Andrea Prokopova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University in Olomouc, 779 00 Olomouc, Czech Republic
| | - Hanus Slavik
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, 67000 Strasbourg, France
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University in Olomouc, 779 00 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital in Olomouc, 779 00 Olomouc, Czech Republic
| | - Josef Srovnal
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University in Olomouc, 779 00 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital in Olomouc, 779 00 Olomouc, Czech Republic
| |
Collapse
|
14
|
Yang L, Zhang Y, Dong X, Wang X, Li M, Jiang L, Liu Z. An impedance aptasensing method based on the Strep-Tag system integrated apta-magnetic probe for acetamiprid determination. Microchem J 2023. [DOI: 10.1016/j.microc.2023.108513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
15
|
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
|
16
|
Schwab FD, Scheidmann MC, Ozimski LL, Kling A, Armbrecht L, Ryser T, Krol I, Strittmatter K, Nguyen-Sträuli BD, Jacob F, Fedier A, Heinzelmann-Schwarz V, Wicki A, Dittrich PS, Aceto N. MyCTC chip: microfluidic-based drug screen with patient-derived tumour cells from liquid biopsies. MICROSYSTEMS & NANOENGINEERING 2022; 8:130. [PMID: 36561926 PMCID: PMC9763115 DOI: 10.1038/s41378-022-00467-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/22/2022] [Accepted: 10/24/2022] [Indexed: 06/17/2023]
Abstract
Cancer patients with advanced disease are characterized by intrinsic challenges in predicting drug response patterns, often leading to ineffective treatment. Current clinical practice for treatment decision-making is commonly based on primary or secondary tumour biopsies, yet when disease progression accelerates, tissue biopsies are not performed on a regular basis. It is in this context that liquid biopsies may offer a unique window to uncover key vulnerabilities, providing valuable information about previously underappreciated treatment opportunities. Here, we present MyCTC chip, a novel microfluidic device enabling the isolation, culture and drug susceptibility testing of cancer cells derived from liquid biopsies. Cancer cell capture is achieved through a label-free, antigen-agnostic enrichment method, and it is followed by cultivation in dedicated conditions, allowing on-chip expansion of captured cells. Upon growth, cancer cells are then transferred to drug screen chambers located within the same device, where multiple compounds can be tested simultaneously. We demonstrate MyCTC chip performance by means of spike-in experiments with patient-derived breast circulating tumour cells, enabling >95% capture rates, as well as prospective processing of blood from breast cancer patients and ascites fluid from patients with ovarian, tubal and endometrial cancer, where sensitivity to specific chemotherapeutic agents was identified. Together, we provide evidence that MyCTC chip may be used to identify personalized drug response patterns in patients with advanced metastatic disease and with limited treatment opportunities.
Collapse
Affiliation(s)
- Fabienne D. Schwab
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel, Basel, Switzerland
- Department of Gynaecologic Oncology, University Hospital Basel, Basel, Switzerland
| | - Manuel C. Scheidmann
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel, Basel, Switzerland
| | - Lauren L. Ozimski
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel, Basel, Switzerland
- Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - André Kling
- Department of Biosystems Science and Engineering, Swiss Federal Institute of Technology Zurich (ETH Zurich), Basel, Switzerland
| | - Lucas Armbrecht
- Department of Biosystems Science and Engineering, Swiss Federal Institute of Technology Zurich (ETH Zurich), Basel, Switzerland
| | - Till Ryser
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel, Basel, Switzerland
| | - Ilona Krol
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel, Basel, Switzerland
- Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Karin Strittmatter
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel, Basel, Switzerland
- Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Bich Doan Nguyen-Sträuli
- Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
- Department of Gynaecology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Francis Jacob
- Department of Biomedicine, Ovarian Cancer Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - André Fedier
- Department of Biomedicine, Ovarian Cancer Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Viola Heinzelmann-Schwarz
- Department of Gynaecologic Oncology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, Ovarian Cancer Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Andreas Wicki
- University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Petra S. Dittrich
- Department of Biosystems Science and Engineering, Swiss Federal Institute of Technology Zurich (ETH Zurich), Basel, Switzerland
| | - Nicola Aceto
- Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|
17
|
Detection of Circulating Tumor Cells Using the Attune NxT. Int J Mol Sci 2022; 24:ijms24010021. [PMID: 36613466 PMCID: PMC9820284 DOI: 10.3390/ijms24010021] [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: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Circulating tumor cells (CTCs) have been detected in many patients with different solid malignancies. It has been reported that presence of CTCs correlates with worse survival in patients with multiple types of cancer. Several techniques have been developed to detect CTCs in liquid biopsies. Currently, the only method for CTC detection that is approved by the Food and Drug Administration is CellSearch. Due to low abundance of CTCs in certain cancer types and in early stages of disease, its clinical application is currently limited to metastatic colorectal cancer, breast cancer and prostate cancer. Therefore, we aimed to develop a new method for the detection of CTCs using the Attune NxT-a flow cytometry-based application that was specifically developed to detect rare events in biological samples without the need for enrichment. When healthy donor blood samples were spiked with variable amounts of different EpCAM+EGFR+ tumor cell lines, recovery yield was on average 75%. The detection range was between 1000 and 10 cells per sample. Cell morphology was confirmed with the Attune CytPix. Analysis of blood samples from metastatic colorectal cancer patients, as well as lung cancer patients, demonstrated that increased EpCAM+EGFR+ events were detected in more than half of the patient samples. However, most of these cells showed no (tumor) cell-like morphology. Notably, CellSearch analysis of blood samples from a subset of colorectal cancer patients did not detect CTCs either, suggesting that these blood samples were negative for CTCs. Therefore, we anticipate that the Attune NxT is not superior to CellSearch in detection of low amounts of CTCs, although handling and analysis of samples is easier. Moreover, morphological confirmation is essential to distinguish between CTCs and false positive events.
Collapse
|
18
|
Jia L, Zhen X, Chen L, Feng Q, Yuan W, Bu Y, Wang S, Xie X. Bioinspired nano-plate-coral platform enabled efficient detection of circulating tumor cells via the synergistic capture of multivalent aptamer and tumor cell membrane. J Colloid Interface Sci 2022; 631:55-65. [DOI: 10.1016/j.jcis.2022.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/31/2022] [Accepted: 11/06/2022] [Indexed: 11/10/2022]
|
19
|
Tian R, Li X, Zhang H, Ma L, Zhang H, Wang Z. Ulex Europaeus Agglutinin-I-Based Magnetic Isolation for the Efficient and Specific Capture of SW480 Circulating Colorectal Tumor Cells. ACS OMEGA 2022; 7:30405-30411. [PMID: 36061664 PMCID: PMC9435041 DOI: 10.1021/acsomega.2c03702] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
The efficient and specific capture of circulating tumor cells (CTCs) from patients' peripheral blood is of significant value in precise cancer diagnosis and cancer therapy. As fine targeting molecules, lectins can recognize cancer cells specifically due to the abnormal glycosylation of molecules on the cancer cell membrane and the specific binding of lectin with glycoconjugates. Herein, a Ulex europaeus agglutinin-I (UEA-I)-based magnetic isolation strategy was developed to efficiently and specifically capture α-1,2-fucose overexpression CTCs from colorectal cancer (CRC) patients' peripheral blood. Using UEA-I-modified Fe3O4 magnetic beads (termed MB-UEA-I), up to 94 and 89% of target cells (i.e., SW480 CRC cells) were captured from the cell spiking complete cell culture medium and whole blood, respectively. More than 90% of captured cells show good viability and proliferation ability without detaching from MB-UEA-I. In combination with three-color immunocytochemistry (ICC) identification, MB-UEA-I has been successfully used to capture CTCs from CRC patients' peripheral blood. The experimental results indicate a correlation between CTC characterization and tumor metastasis. Specifically, MB-UEA-I can be applied to screen early CRC by capturing CTCs when served as a liquid biopsy. The presented work offers a new insight into developing cost-effective lectin-functionalized methods for biomedical applications.
Collapse
Affiliation(s)
- Rongrong Tian
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- University
of Science and Technology of China, Jinzhai Road, Baohe District, Hefei, Anhui 230026, P. R. China
| | - Xiaodong Li
- Department
of Radiology, The First Hospital of Jilin
University, Changchun, Jilin 130021, P. R. China
| | - Hua Zhang
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Lina Ma
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Huimao Zhang
- Department
of Radiology, The First Hospital of Jilin
University, Changchun, Jilin 130021, P. R. China
| | - Zhenxin Wang
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- University
of Science and Technology of China, Jinzhai Road, Baohe District, Hefei, Anhui 230026, P. R. China
| |
Collapse
|
20
|
Liu Y, Li R, Zhang L, Guo S. Nanomaterial-Based Immunocapture Platforms for the Recognition, Isolation, and Detection of Circulating Tumor Cells. Front Bioeng Biotechnol 2022; 10:850241. [PMID: 35360401 PMCID: PMC8964261 DOI: 10.3389/fbioe.2022.850241] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/18/2022] [Indexed: 01/10/2023] Open
Abstract
Circulating tumor cells (CTCs) are a type of cancer cells that circulate in the peripheral blood after breaking away from solid tumors and are essential for the establishment of distant metastasis. Up to 90% of cancer-related deaths are caused by metastatic cancer. As a new type of liquid biopsy, detecting and analyzing CTCs will provide insightful information for cancer diagnosis, especially the in-time disease status, which would avoid some flaws and limitations of invasive tissue biopsy. However, due to the extremely low levels of CTCs among a large number of hematologic cells, choosing immunocapture platforms for CTC detection and isolation will achieve good performance with high purity, selectivity, and viability. These properties are directly associated with precise downstream analysis of CTC profiling. Recently, inspired by the nanoscale interactions of cells in the tissue microenvironment, platforms based on nanomaterials have been widely explored to efficiently enrich and sensitively detect CTCs. In this review, various immunocapture platforms based on different nanomaterials for efficient isolation and sensitive detection of CTCs are outlined and discussed. First, the design principles of immunoaffinity nanomaterials are introduced in detail. Second, the immunocapture and release of platforms based on nanomaterials ranging from nanoparticles, nanostructured substrates, and immunoaffinity microfluidic chips are summarized. Third, recent advances in single-cell release and analysis of CTCs are introduced. Finally, some perspectives and challenges are provided in future trends of CTC studies.
Collapse
Affiliation(s)
- Yichao Liu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Rui Li
- Xinjiang Key Laboratory of Solid State Physics and Devices, Xinjiang University, Urumqi, China
| | - Lingling Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Lingling Zhang, ; Shishang Guo,
| | - Shishang Guo
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, China
- *Correspondence: Lingling Zhang, ; Shishang Guo,
| |
Collapse
|
21
|
Ginghina O, Hudita A, Zamfir M, Spanu A, Mardare M, Bondoc I, Buburuzan L, Georgescu SE, Costache M, Negrei C, Nitipir C, Galateanu B. Liquid Biopsy and Artificial Intelligence as Tools to Detect Signatures of Colorectal Malignancies: A Modern Approach in Patient's Stratification. Front Oncol 2022; 12:856575. [PMID: 35356214 PMCID: PMC8959149 DOI: 10.3389/fonc.2022.856575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/16/2022] [Indexed: 01/19/2023] Open
Abstract
Colorectal cancer (CRC) is the second most frequently diagnosed type of cancer and a major worldwide public health concern. Despite the global efforts in the development of modern therapeutic strategies, CRC prognosis is strongly correlated with the stage of the disease at diagnosis. Early detection of CRC has a huge impact in decreasing mortality while pre-lesion detection significantly reduces the incidence of the pathology. Even though the management of CRC patients is based on robust diagnostic methods such as serum tumor markers analysis, colonoscopy, histopathological analysis of tumor tissue, and imaging methods (computer tomography or magnetic resonance), these strategies still have many limitations and do not fully satisfy clinical needs due to their lack of sensitivity and/or specificity. Therefore, improvements of the current practice would substantially impact the management of CRC patients. In this view, liquid biopsy is a promising approach that could help clinicians screen for disease, stratify patients to the best treatment, and monitor treatment response and resistance mechanisms in the tumor in a regular and minimally invasive manner. Liquid biopsies allow the detection and analysis of different tumor-derived circulating markers such as cell-free nucleic acids (cfNA), circulating tumor cells (CTCs), and extracellular vesicles (EVs) in the bloodstream. The major advantage of this approach is its ability to trace and monitor the molecular profile of the patient's tumor and to predict personalized treatment in real-time. On the other hand, the prospective use of artificial intelligence (AI) in medicine holds great promise in oncology, for the diagnosis, treatment, and prognosis prediction of disease. AI has two main branches in the medical field: (i) a virtual branch that includes medical imaging, clinical assisted diagnosis, and treatment, as well as drug research, and (ii) a physical branch that includes surgical robots. This review summarizes findings relevant to liquid biopsy and AI in CRC for better management and stratification of CRC patients.
Collapse
Affiliation(s)
- Octav Ginghina
- Department II, University of Medicine and Pharmacy “Carol Davila” Bucharest, Bucharest, Romania
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | - Ariana Hudita
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Marius Zamfir
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | - Andrada Spanu
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | - Mara Mardare
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | - Irina Bondoc
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | | | - Sergiu Emil Georgescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Carolina Negrei
- Department of Toxicology, University of Medicine and Pharmacy “Carol Davila” Bucharest, Bucharest, Romania
| | - Cornelia Nitipir
- Department II, University of Medicine and Pharmacy “Carol Davila” Bucharest, Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, Bucharest, Romania
| | - Bianca Galateanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| |
Collapse
|
22
|
Abstract
Magnetic cell separation has become a key methodology for the isolation of target cell populations from biological suspensions, covering a wide spectrum of applications from diagnosis and therapy in biomedicine to environmental applications or fundamental research in biology. There now exists a great variety of commercially available separation instruments and reagents, which has permitted rapid dissemination of the technology. However, there is still an increasing demand for new tools and protocols which provide improved selectivity, yield and sensitivity of the separation process while reducing cost and providing a faster response. This review aims to introduce basic principles of magnetic cell separation for the neophyte, while giving an overview of recent research in the field, from the development of new cell labeling strategies to the design of integrated microfluidic cell sorters and of point-of-care platforms combining cell selection, capture, and downstream detection. Finally, we focus on clinical, industrial and environmental applications where magnetic cell separation strategies are amongst the most promising techniques to address the challenges of isolating rare cells.
Collapse
|
23
|
Chelakkot C, Yang H, Shin YK. Relevance of Circulating Tumor Cells as Predictive Markers for Cancer Incidence and Relapse. Pharmaceuticals (Basel) 2022; 15:75. [PMID: 35056131 PMCID: PMC8781286 DOI: 10.3390/ph15010075] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023] Open
Abstract
Shedding of cancer cells from the primary site or undetectable bone marrow region into the circulatory system, resulting in clinically overt metastasis or dissemination, is the hallmark of unfavorable invasive cancers. The shed cells remain in circulation until they extravasate to form a secondary metastatic lesion or undergo anoikis. The circulating tumor cells (CTCs) found as single cells or clusters carry a plethora of information, are acknowledged as potential biomarkers for predicting cancer prognosis and cancer progression, and are supposed to play key roles in determining tailored therapies for advanced diseases. With the advent of novel technologies that allow the precise isolation of CTCs, more and more clinical trials are focusing on the prognostic and predictive potential of CTCs. In this review, we summarize the role of CTCs as a predictive marker for cancer incidence, relapse, and response to therapy.
Collapse
Affiliation(s)
- Chaithanya Chelakkot
- Bio-MAX/N-Bio, Bio-MAX Institute, Seoul National University, Seoul 08226, Korea
- Genobio Corp., Seoul 08394, Korea
| | - Hobin Yang
- Research Institute of Pharmaceutical Science, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08226, Korea
| | - Young Kee Shin
- Bio-MAX/N-Bio, Bio-MAX Institute, Seoul National University, Seoul 08226, Korea
- Research Institute of Pharmaceutical Science, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08226, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08226, Korea
| |
Collapse
|
24
|
Li F, Xu H, Zhao Y. Magnetic particles as promising circulating tumor cell catchers assisting liquid biopsy in cancer diagnosis: A review. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
25
|
Libring S, Enríquez Á, Lee H, Solorio L. In Vitro Magnetic Techniques for Investigating Cancer Progression. Cancers (Basel) 2021; 13:4440. [PMID: 34503250 PMCID: PMC8430481 DOI: 10.3390/cancers13174440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/28/2021] [Accepted: 08/29/2021] [Indexed: 12/24/2022] Open
Abstract
Worldwide, there are currently around 18.1 million new cancer cases and 9.6 million cancer deaths yearly. Although cancer diagnosis and treatment has improved greatly in the past several decades, a complete understanding of the complex interactions between cancer cells and the tumor microenvironment during primary tumor growth and metastatic expansion is still lacking. Several aspects of the metastatic cascade require in vitro investigation. This is because in vitro work allows for a reduced number of variables and an ability to gather real-time data of cell responses to precise stimuli, decoupling the complex environment surrounding in vivo experimentation. Breakthroughs in our understanding of cancer biology and mechanics through in vitro assays can lead to better-designed ex vivo precision medicine platforms and clinical therapeutics. Multiple techniques have been developed to imitate cancer cells in their primary or metastatic environments, such as spheroids in suspension, microfluidic systems, 3D bioprinting, and hydrogel embedding. Recently, magnetic-based in vitro platforms have been developed to improve the reproducibility of the cell geometries created, precisely move magnetized cell aggregates or fabricated scaffolding, and incorporate static or dynamic loading into the cell or its culture environment. Here, we will review the latest magnetic techniques utilized in these in vitro environments to improve our understanding of cancer cell interactions throughout the various stages of the metastatic cascade.
Collapse
Affiliation(s)
- Sarah Libring
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (Á.E.)
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
| | - Ángel Enríquez
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (Á.E.)
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
- Center for Implantable Devices, Purdue University, West Lafayette, IN 47907, USA
| | - Hyowon Lee
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (Á.E.)
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
- Center for Implantable Devices, Purdue University, West Lafayette, IN 47907, USA
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (Á.E.)
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
26
|
Zhang Y, Wang W, Guo H, Liu M, Zhu H, Sun H. Hyaluronic acid-functionalized redox responsive immunomagnetic nanocarrier for circulating tumor cell capture and release. NANOTECHNOLOGY 2021; 32:475102. [PMID: 33494073 DOI: 10.1088/1361-6528/abdf8c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/25/2021] [Indexed: 06/12/2023]
Abstract
Detection of circulating tumor cells (CTCs) in peripheral blood holds significant insights for cancer diagnosis, prognosis evaluation, and precision medicine. To efficiently capture and release CTCs with high viability, we reported the development of hyaluronic acid (HA)-functionalized redox responsive immunomagnetic nanocarrier (Fe3O4@SiO2-SS-HA). First, Fe3O4nanoparticles were prepared and modified with tetraethyl orthosilicate (TEOS), 3-mercaptopropyltrimethoxysilane (MPTMS) and 2,2'-dithiodipyridine (DDPy) to form the magnetic substrate (Fe3O4@SiO2-SSPy). Modified with targeted segment HA-functionalized L-cysteine ethyl ester hydrochloride (HA-Cys) via disulfide exchange reaction, the Fe3O4@SiO2-SS-HA was formed. The nanocarrier with prominent magnetic property, targeting ligand, and redox-sensitive disulfide linkages was able to specially capture MCF-7 cells with an efficiency of 92% and effectively release captured cells with an efficiency of 81.4%. Furthermore, the Fe3O4@SiO2-SS-HA could successfully be used for the capture of MCF-7 cells, and the captured cells could be diferntiated from the blood cells. Almost all of released tumor cells kept good viability and a robust proliferative capacity after being re-cultured. It is likely that the as-prepared nanocarrier will serve as a new weapon against CD44 receptor-overexpressed cancer cells.
Collapse
Affiliation(s)
- Yi Zhang
- School of Bioengineering and Food, Key Laboratory of Fermentation Engineering (Ministry of Education), Key Laboratory of Industrial Microbiology in Hubei, National '111' Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Wenjing Wang
- School of Bioengineering and Food, Key Laboratory of Fermentation Engineering (Ministry of Education), Key Laboratory of Industrial Microbiology in Hubei, National '111' Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Huiling Guo
- School of Bioengineering and Food, Key Laboratory of Fermentation Engineering (Ministry of Education), Key Laboratory of Industrial Microbiology in Hubei, National '111' Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Mingxing Liu
- School of Bioengineering and Food, Key Laboratory of Fermentation Engineering (Ministry of Education), Key Laboratory of Industrial Microbiology in Hubei, National '111' Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Hongda Zhu
- School of Bioengineering and Food, Key Laboratory of Fermentation Engineering (Ministry of Education), Key Laboratory of Industrial Microbiology in Hubei, National '111' Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Hongmei Sun
- School of Bioengineering and Food, Key Laboratory of Fermentation Engineering (Ministry of Education), Key Laboratory of Industrial Microbiology in Hubei, National '111' Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan 430068, People's Republic of China
| |
Collapse
|
27
|
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
|
28
|
Heredia-Soto V, Rodríguez-Salas N, Feliu J. Liquid Biopsy in Pancreatic Cancer: Are We Ready to Apply It in the Clinical Practice? Cancers (Basel) 2021; 13:1986. [PMID: 33924143 PMCID: PMC8074327 DOI: 10.3390/cancers13081986] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits the poorest prognosis of all solid tumors, with a 5-year survival of less than 10%. To improve the prognosis, it is necessary to advance in the development of tools that help us in the early diagnosis, treatment selection, disease monitoring, evaluation of the response and prognosis. Liquid biopsy (LB), in its different modalities, represents a particularly interesting tool for these purposes, since it is a minimally invasive and risk-free procedure that can detect both the presence of genetic material from the tumor and circulating tumor cells (CTCs) in the blood and therefore distantly reflect the global status of the disease. In this work we review the current status of the main LB modalities (ctDNA, exosomes, CTCs and cfRNAs) for detecting and monitoring PDAC.
Collapse
Affiliation(s)
- Victoria Heredia-Soto
- Translational Oncology Research Laboratory, Biomedical Research Institute, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain; (V.H.-S.); (N.R.-S.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Monforte de Lemos 5, 28029 Madrid, Spain
| | - Nuria Rodríguez-Salas
- Translational Oncology Research Laboratory, Biomedical Research Institute, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain; (V.H.-S.); (N.R.-S.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Monforte de Lemos 5, 28029 Madrid, Spain
- Cátedra UAM-AMGEN, Medical Oncology Department, La Paz University Hospital, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Jaime Feliu
- Translational Oncology Research Laboratory, Biomedical Research Institute, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain; (V.H.-S.); (N.R.-S.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Monforte de Lemos 5, 28029 Madrid, Spain
- Cátedra UAM-AMGEN, Medical Oncology Department, La Paz University Hospital, Paseo de la Castellana 261, 28046 Madrid, Spain
| |
Collapse
|
29
|
SHEN CC, WU CK, CHEN YH, WANG JX, YANG MH, ZHANG H. Advance in Novel Methods for Enrichment and Precise Analysis of Circulating Tumor Cells. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2021. [DOI: 10.1016/s1872-2040(21)60089-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
30
|
Liu Y, Xu H, Li T, Wang W. Microtechnology-enabled filtration-based liquid biopsy: challenges and practical considerations. LAB ON A CHIP 2021; 21:994-1015. [PMID: 33710188 DOI: 10.1039/d0lc01101k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Liquid biopsy, an important enabling technology for early diagnosis and dynamic monitoring of cancer, has drawn extensive attention in the past decade. With the rapid developments of microtechnology, it has been possible to manipulate cells at the single-cell level, which dramatically improves the liquid biopsy capability. As the microtechnology-enabled liquid biopsy matures from proof-of-concept demonstrations towards practical applications, a main challenge it is facing now is to process clinical samples which are usually of a large volume while containing very rare targeted cells in complex backgrounds. Therefore, a high-throughput liquid biopsy which is capable of processing liquid samples with a large volume in a reasonable time along with a high recovery rate of rare targeted cells from complex clinical liquids is in high demand. Moreover, the purity, viability and release feasibility of recovered targeted cells are the other three key impact factors requiring careful considerations. To date, among the developed techniques, micropore-type filtration has been acknowledged as the most promising solution to address the aforementioned challenges in practical applications. However, the presently reported studies about micropore-type filtration are mostly based on trial and error for device designs aiming at different cancer types, which requires lots of efforts. Therefore, there is an urgent need to investigate and elaborate the fundamental theories of micropore-type filtration and key features that influence the working performances in the liquid biopsy of real clinical samples to promote the application efficacy in practical applications. In this review, the state of the art of microtechnology-enabled filtration is systematically and comprehensively summarized. Four key features of the filtration, including throughput, purity, viability and release feasibility of the captured targeted cells, are elaborated to provide the guidelines for filter designs. The recent progress in the filtration mode modulation and sample standardization to improve the filtration performance of real clinical samples is also discussed. Finally, this review concludes with prospective views for future developments of filtration-based liquid biopsy to promote its application efficacy in clinical practice.
Collapse
Affiliation(s)
- Yaoping Liu
- Institute of Microelectronics, Peking University, Beijing, 100871, China.
| | | | | | | |
Collapse
|
31
|
Ding P, Wang Z, Wu Z, Zhu W, Liu L, Sun N, Pei R. Aptamer-based nanostructured interfaces for the detection and release of circulating tumor cells. J Mater Chem B 2021; 8:3408-3422. [PMID: 32022083 DOI: 10.1039/c9tb02457c] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Analysis of circulating tumor cells (CTCs) can provide significant clinical information for tumors, which has proven to be helpful for cancer diagnosis, prognosis monitoring, treatment efficacy, and personalized therapy. However, CTCs are an extremely rare cell population, which challenges the isolation of CTCs from patient blood. Over the last few decades, many strategies for CTC detection have been developed based on the physical and biological properties of CTCs. Among them, nanostructured interfaces have been widely applied as CTC detection platforms to overcome the current limitations associated with CTC capture. Furthermore, aptamers have attracted significant attention in the detection of CTCs due to their advantages, including good affinity, low cost, easy modification, excellent stability, and low immunogenicity. In addition, effective and nondestructive release of CTCs can be achieved by aptamer-mediated methods that are used under mild conditions. Herein, we review some progress in the detection and release of CTCs through aptamer-functionalized nanostructured interfaces.
Collapse
Affiliation(s)
- Pi Ding
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | | | | | | | | | | | | |
Collapse
|
32
|
Rushton AJ, Nteliopoulos G, Shaw JA, Coombes RC. A Review of Circulating Tumour Cell Enrichment Technologies. Cancers (Basel) 2021; 13:cancers13050970. [PMID: 33652649 PMCID: PMC7956528 DOI: 10.3390/cancers13050970] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Circulating tumour cells (CTCs) are cancer cells shed into the bloodstream from tumours and their analysis can provide important insights into cancer detection and monitoring, with the potential to direct personalised therapies for the patient. These CTCs are rare in the blood, which makes their detection and enrichment challenging and to date, only one technology (the CellSearch) has gained FDA approval for determining the prognosis of patients with advanced breast, prostate and colorectal cancers. Here, we review the wide range of enrichment technologies available to isolate CTCs from other blood components and highlight the important characteristics that new technologies should possess for routine clinical use. Abstract Circulating tumour cells (CTCs) are the precursor cells for the formation of metastatic disease. With a simple blood draw, liquid biopsies enable the non-invasive sampling of CTCs from the blood, which have the potential to provide important insights into cancer detection and monitoring. Since gaining FDA approval in 2004, the CellSearch system has been used to determine the prognosis of patients with metastatic breast, prostate and colorectal cancers. This utilises the cell surface marker Epithelial Cell Adhesion Molecule (EpCAM), to enrich CTCs, and many other technologies have adopted this approach. More recently, the role of mesenchymal-like CTCs in metastasis formation has come to light. It has been suggested that these cells are more aggressive metastatic precursors than their epithelial counterparts; however, mesenchymal CTCs remain undetected by EpCAM-based enrichment methods. This has prompted the development of a variety of ‘label free’ enrichment technologies, which exploit the unique physical properties of CTCs (such as size and deformability) compared to other blood components. Here, we review a wide range of both immunocapture and label free CTC enrichment technologies, summarising the most significant advantages and disadvantages of each. We also highlight the important characteristics that technologies should possess for routine clinical use, since future developments could have important clinical implications, with the potential to direct personalised therapies for patients with cancer.
Collapse
Affiliation(s)
- Amelia J. Rushton
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
- Correspondence:
| | - Georgios Nteliopoulos
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
| | - Jacqueline A. Shaw
- Leicester Cancer Research Centre, University of Leicester, Leicester LE2 7LX, UK;
| | - R. Charles Coombes
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
| |
Collapse
|
33
|
Singh B, Arora S, D'Souza A, Kale N, Aland G, Bharde A, Quadir M, Calderón M, Chaturvedi P, Khandare J. Chemo-specific designs for the enumeration of circulating tumor cells: advances in liquid biopsy. J Mater Chem B 2021; 9:2946-2978. [PMID: 33480960 DOI: 10.1039/d0tb02574g] [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
Advanced materials and chemo-specific designs at the nano/micrometer-scale have ensured revolutionary progress in next-generation clinically relevant technologies. For example, isolating a rare population of cells, like circulating tumor cells (CTCs) from the blood amongst billions of other blood cells, is one of the most complex scientific challenges in cancer diagnostics. The chemical tunability for achieving this degree of exceptional specificity for extra-cellular biomarker interactions demands the utility of advanced entities and multistep reactions both in solution and in the insoluble state. Thus, this review delineates the chemo-specific substrates, chemical methods, and structure-activity relationships (SARs) of chemical platforms used for isolation and enumeration of CTCs in advancing the relevance of liquid biopsy in cancer diagnostics and disease management. We highlight the synthesis of cell-specific, tumor biomarker-based, chemo-specific substrates utilizing functionalized linkers through chemistry-based conjugation strategies. The capacity of these nano/micro substrates to enhance the cell interaction specificity and efficiency with the targeted tumor cells is detailed. Furthermore, this review accounts for the importance of CTC capture and other downstream processes involving genotypic and phenotypic CTC analysis in real-time for the detection of the early onset of metastases progression and chemotherapy treatment response, and for monitoring progression free-survival (PFS), disease-free survival (DFS), and eventually overall survival (OS) in cancer patients.
Collapse
Affiliation(s)
- Balram Singh
- Actorius Innovations and Research Pvt. Ltd, Pune, 411057, India.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kumar T, Soares RRG, Ali Dholey L, Ramachandraiah H, Aval NA, Aljadi Z, Pettersson T, Russom A. Multi-layer assembly of cellulose nanofibrils in a microfluidic device for the selective capture and release of viable tumor cells from whole blood. NANOSCALE 2020; 12:21788-21797. [PMID: 33103175 DOI: 10.1039/d0nr05375a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
According to reports by the World Health Organization (WHO), cancer-related deaths reached almost 10 million in 2018. Nearly 65% of these deaths occurred in low- to middle-income countries, a trend that is bound to increase since cancer diagnostics are not currently considered a priority in resource-limited settings (RLS). Thus, cost-effective and specific cancer screening and diagnostics tools are in high demand, particularly in RLS. The selective isolation and up-concentration of rare cells while maintaining cell viability and preventing phenotypic changes is a powerful tool to allow accurate and sensitive downstream analysis. Here, multi-layer cellulose nanofibril-based coatings functionalized with anti-EpCAM antibodies on the surface of disposable microfluidic devices were optimized for specific capture of target cells, followed by efficient release without significant adverse effects. HCT 116 colon cancer cells were captured in a single step with >97% efficiency at 41.25 μL min-1 and, when spiked in whole blood, an average enrichment factor of ∼200-fold relative to white blood cells was achieved. The release of cells was performed by enzymatic digestion of the cellulose nanofibrils which had a negligible impact on cell viability. In particular, >80% of the cells were recovered with at least 97% viability in less than 30 min. Such performance paves the way to expand and improve clinical diagnostic applications by simplifying the isolation of circulating tumor cells (CTCs) and other rare cells directly from whole blood.
Collapse
Affiliation(s)
- Tharagan Kumar
- KTH Royal Institute of Technology, Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, Solna, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Chen Y, Chen X, Li M, Fan P, Wang B, Zhao S, Yu W, Zhang S, Tang Y, Gao T. A new analytical platform for potential point-of-care testing of circulating tumor cells. Biosens Bioelectron 2020; 171:112718. [PMID: 33059165 DOI: 10.1016/j.bios.2020.112718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 01/21/2023]
Abstract
It is of significance to detect circulating tumor cells (CTCs) in whole blood using transportable instruments at the point of care to assist evaluating chemotherapeutic efficacy and recurrence risk of cancer patients. However, the current widely used detection methods either require expensive and complex equipments, need complicated enrichment steps, or produce high rates of false positive and/or negative results. Aiming for solving the two critical challenges involved in instrumentation miniaturization and simplification of sample preparation for POCT of CTCs without sacrificing the detection sensitivity and accuracy, this work reports a custom-built, automatic, large field-of-view microscopic CTC cytometer and a novel enrichment strategy based on a synthesized peptide ligand discovered from One-Bead One-Compound library screening. The custom-built microscope has compact size, low weight and efficient cost while still maintaining a detection limit of as low as 5 target objects. The simplified sample preparation utilized a novel peptide LXW7 functionalized to magnetic beads and allows for rapid, highly selective and sensitive detection of CTCs. This analytical platform may fulfill the unmet need for possible point-of-care CTC counting, and provide a new option for early diagnosis of cancers and convenient evaluation of chemotherapeutic efficacy and cancer recurrence.
Collapse
Affiliation(s)
- Yangfei Chen
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Xuqi Chen
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Mengna Li
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Pengwei Fan
- Clinical Laboratory Center, Wuhan No. 7 Hospital, Wuhan, 430071, Hubei, China
| | - Bin Wang
- Clinical Laboratory Center, Wuhan No. 7 Hospital, Wuhan, 430071, Hubei, China
| | - Sheng Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Shaohua Zhang
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Yuchen Tang
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Tingjuan Gao
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, And Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan, 430079, Hubei, China.
| |
Collapse
|
36
|
Tian C, Xu X, Wang Y, Li D, Lu H, Yang Z. Development and Clinical Prospects of Techniques to Separate Circulating Tumor Cells from Peripheral Blood. Cancer Manag Res 2020; 12:7263-7275. [PMID: 32884342 PMCID: PMC7434565 DOI: 10.2147/cmar.s248380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Detection of circulating tumor cells (CTC) is an important liquid biopsy technique that has advanced considerably in recent years. To further advance the development of technology for curing cancer, several CTC technologies have been proposed by various research groups. Despite their potential role in early cancer diagnosis and prognosis, CTC methods are currently used for research purposes only, and very few methods have been accepted for clinical applications because of difficulties, including CTC heterogeneity, CTC separation from the blood, and a lack of thorough clinical validation. Although current CTC technologies have not been truly implemented, they possess high potential as future clinical diagnostic techniques for individualized cancer. Here, we review current developments in CTC separation technology. We also explore new CTC detection methods based on telomerase and nanomaterials, such as in vivo flow cytometry. In addition, we discuss the difficulties that must be overcome before CTC can be applied in clinical settings.
Collapse
Affiliation(s)
- Cheng Tian
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Xinhua Xu
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Yuke Wang
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Dailong Li
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Haiyan Lu
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| | - Ziwei Yang
- Yichang Central People's Hospital, First Clinical Medical College of Three Gorges University, Yichang 443000, People's Republic of China
| |
Collapse
|
37
|
Hazra RS, Kale N, Aland G, Qayyumi B, Mitra D, Jiang L, Bajwa D, Khandare J, Chaturvedi P, Quadir M. Cellulose Mediated Transferrin Nanocages for Enumeration of Circulating Tumor Cells for Head and Neck Cancer. Sci Rep 2020; 10:10010. [PMID: 32561829 PMCID: PMC7305211 DOI: 10.1038/s41598-020-66625-2] [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: 06/03/2019] [Accepted: 05/20/2020] [Indexed: 01/05/2023] Open
Abstract
Herein we report a hierarchically organized, water-dispersible 'nanocage' composed of cellulose nanocrystals (CNCs), which are magnetically powered by iron oxide (Fe3O4) nanoparticles (NPs) to capture circulating tumor cells (CTCs) in blood for head and neck cancer (HNC) patients. Capturing CTCs from peripheral blood is extremely challenging due to their low abundance and its account is clinically validated in progression-free survival of patients with HNC. Engaging multiple hydroxyl groups along the molecular backbone of CNC, we co-ordinated Fe3O4 NPs onto CNC scaffold, which was further modified by conjugation with a protein - transferrin (Tf) for targeted capture of CTCs. Owing to the presence of Fe3O4 nanoparticles, these nanocages were magnetic in nature, and CTCs could be captured under the influence of a magnetic field. Tf-CNC-based nanocages were evaluated using HNC patients' blood sample and compared for the CTC capturing efficiency with clinically relevant Oncoviu platform. Conclusively, we observed that CNC-derived nanocages efficiently isolated CTCs from patient's blood at 85% of cell capture efficiency to that of the standard platform. Capture efficiency was found to vary with the concentration of Tf and Fe3O4 nanoparticles immobilized onto the CNC scaffold. We envision that, Tf-CNC platform has immense connotation in 'liquid biopsy' for isolation and enumeration of CTCs for early detection of metastasis in cancer.
Collapse
Affiliation(s)
- Raj Shankar Hazra
- Department of Mechanical Engineering, Materials and Nanotechnology Program, North Dakota State University, Fargo, 58108, ND, USA
| | - Narendra Kale
- Maharashtra Institute of Technology-WPU, School of Pharmacy, Pune, India
| | | | - Burhanuddin Qayyumi
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, 400012, Maharashtra, India
| | - Dipankar Mitra
- Department of Electrical and Computer Engineering, North Dakota State University, Fargo, 58108, ND, USA
| | - Long Jiang
- Department of Mechanical Engineering, Materials and Nanotechnology Program, North Dakota State University, Fargo, 58108, ND, USA
| | - Dilpreet Bajwa
- Department of Mechanical and Industrial Engineering, Montana State University, Bozeman, MT, 59717-3800, USA
| | - Jayant Khandare
- Maharashtra Institute of Technology-WPU, School of Pharmacy, Pune, India. .,Actorius Innovations and Research (AIR) Pvt. Ltd., Pune, India.
| | - Pankaj Chaturvedi
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, 400012, Maharashtra, India
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, 58108, ND, USA.
| |
Collapse
|
38
|
Luo L, He Y. Magnetically driven microfluidics for isolation of circulating tumor cells. Cancer Med 2020; 9:4207-4231. [PMID: 32325536 PMCID: PMC7300401 DOI: 10.1002/cam4.3077] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Circulating tumor cells (CTCs) largely contribute to cancer metastasis and show potential prognostic significance in cancer isolation and detection. Miniaturization has progressed significantly in the last decade which in turn enabled the development of several microfluidic systems. The microfluidic systems offer a controlled microenvironment for studies of fundamental cell biology, resulting in the rapid development of microfluidic isolation of CTCs. Due to the inherent ability of magnets to provide forces at a distance, the technology of CTCs isolation based on the magnetophoresis mechanism has become a routine methodology. This historical review aims to introduce two principles of magnetic isolation and recent techniques, facilitating research in this field and providing alternatives for researchers in their study of magnetic isolation. Researchers intend to promote effective CTC isolation and analysis as well as active development of next-generation cancer treatment. The first part of this review summarizes the primary principles based on positive and negative magnetophoretic isolation and describes the metrics for isolation performance. The second part presents a detailed overview of the factors that affect the performance of CTC magnetic isolation, including the magnetic field sources, functionalized magnetic nanoparticles, magnetic fluids, and magnetically driven microfluidic systems.
Collapse
Affiliation(s)
- Laan Luo
- School of Chemical EngineeringKunming University of Science and TechnologyKunmingChina
| | - Yongqing He
- School of Chemical EngineeringKunming University of Science and TechnologyKunmingChina
- Chongqing Key Laboratory of Micro‐Nano System and Intelligent SensingChongqing Technology and Business UniversityChongqingChina
| |
Collapse
|
39
|
Cui H, Liu Q, Li R, Wei X, Sun Y, Wang Z, Zhang L, Zhao XZ, Hua B, Guo SS. ZnO nanowire-integrated bio-microchips for specific capture and non-destructive release of circulating tumor cells. NANOSCALE 2020; 12:1455-1463. [PMID: 31808771 DOI: 10.1039/c9nr07349c] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Circulating tumor cells (CTCs) are one type of significant biomarker in cancer patients' blood that have been attracting attention from researchers for decades, and their efficient and viable isolation is of vital importance in cancer prevention and treatment. However, the development of efficient and low-cost bio-microchips still faces significant challenges. In this paper, we construct a novel three-dimensional micro-nano bio-microchip that has dual functions of specifically capturing and non-destructively releasing cancer cells. ZnO nanowire arrays were vertically grown on the surface of a polydimethylsiloxane (PDMS) pillar substrate with a gear structure (ZnO-coated G-PDMS pillar microchips). The gear structure provides more binding sites for antibodies and target cancer cells, while ZnO nanowires provide a rough surface for CTC attachment and size-specific effects for retaining CTCs. For subsequent culture and bioanalysis, the captured CTCs can be non-destructively released with high efficiency and good viability using a mild acidic solution treatment. Furthermore, the manufacturing process of the G-PDMS pillar microchips is convenient and low-cost, and the preparation approach of the ZnO nanowire is mature and simple to operate. In particular, the bio-microchips showed high capture efficiency (91.11% ± 5.53%) and excellent cell viability (96%) using a spiked cell sample. Moreover, we successfully achieved the specific fluorescent labeling of CTCs in 9 clinical breast cancer patients' samples. The ZnO-coated G-PDMS pillar microchips not only have great potential for new target drug development for cancer stem cells but also open up new opportunities for individualized treatment.
Collapse
Affiliation(s)
- Heng Cui
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, Hubei, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhang Y, Ouyang M, Ray A, Liu T, Kong J, Bai B, Kim D, Guziak A, Luo Y, Feizi A, Tsai K, Duan Z, Liu X, Kim D, Cheung C, Yalcin S, Ceylan Koydemir H, Garner OB, Di Carlo D, Ozcan A. Computational cytometer based on magnetically modulated coherent imaging and deep learning. LIGHT, SCIENCE & APPLICATIONS 2019; 8:91. [PMID: 31645935 PMCID: PMC6804677 DOI: 10.1038/s41377-019-0203-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 09/05/2019] [Accepted: 09/12/2019] [Indexed: 05/08/2023]
Abstract
Detecting rare cells within blood has numerous applications in disease diagnostics. Existing rare cell detection techniques are typically hindered by their high cost and low throughput. Here, we present a computational cytometer based on magnetically modulated lensless speckle imaging, which introduces oscillatory motion to the magnetic-bead-conjugated rare cells of interest through a periodic magnetic force and uses lensless time-resolved holographic speckle imaging to rapidly detect the target cells in three dimensions (3D). In addition to using cell-specific antibodies to magnetically label target cells, detection specificity is further enhanced through a deep-learning-based classifier that is based on a densely connected pseudo-3D convolutional neural network (P3D CNN), which automatically detects rare cells of interest based on their spatio-temporal features under a controlled magnetic force. To demonstrate the performance of this technique, we built a high-throughput, compact and cost-effective prototype for detecting MCF7 cancer cells spiked in whole blood samples. Through serial dilution experiments, we quantified the limit of detection (LoD) as 10 cells per millilitre of whole blood, which could be further improved through multiplexing parallel imaging channels within the same instrument. This compact, cost-effective and high-throughput computational cytometer can potentially be used for rare cell detection and quantification in bodily fluids for a variety of biomedical applications.
Collapse
Affiliation(s)
- Yibo Zhang
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Mengxing Ouyang
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Aniruddha Ray
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Department of Physics and Astronomy, University of Toledo, Toledo, OH 43606 USA
| | - Tairan Liu
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Janay Kong
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Bijie Bai
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Donghyuk Kim
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Alexander Guziak
- Department of Physics and Astronomy, University of California, Los Angeles, CA 90095 USA
| | - Yi Luo
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Alborz Feizi
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Yale School of Medicine, New Haven, CT 06510 USA
| | - Katherine Tsai
- Department of Biochemistry, University of California, Los Angeles, CA 90095 USA
| | - Zhuoran Duan
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
| | - Xuewei Liu
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
| | - Danny Kim
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Chloe Cheung
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Sener Yalcin
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
| | - Hatice Ceylan Koydemir
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Omai B. Garner
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095 USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095 USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095 USA
| | - Aydogan Ozcan
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
41
|
Hu X, Zhu D, Chen M, Chen K, Liu H, Liu W, Yang Y. Precise and non-invasive circulating tumor cell isolation based on optical force using homologous erythrocyte binding. LAB ON A CHIP 2019; 19:2549-2556. [PMID: 31263813 DOI: 10.1039/c9lc00361d] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Precise isolation of circulating tumor cells (CTCs) is proved to be significant for early cancer diagnosis and downstream analysis. Most of the existing strategies yield low purity or cause unexpected damage to cells because of foreign material introduction. To avoid foreign material caused damage and achieve high efficiency simultaneously, this work presents an innovative strategy using tumor cell targeting molecules to bind homologous red blood cells (RBCs) with tumor cells, which results in obvious optical constant differences (both size and mean refractive index) between CC-RBCs (RBC conjugated CTCs) and other blood cells. Then the modified CTCs can be precisely separated under laser illumination in an optofluidic system. Experiments show that CTCs are efficiently modified with erythrocytes and finally isolated from blood at high purity (more than 92%) and a high recovery rate (over 90%). In the whole process, CTCs are proved to keep membrane and function integrity. The combination of homologous RBC binding and an optofluidic system will provide a convenient tool for cancer early diagnosis and treatment monitoring, which exhibits good performance in CTC non-invasive and precise isolation, thus showing great potential.
Collapse
Affiliation(s)
- Xuejia Hu
- School of Physics & Technology, Key Laboratory of Artificial Micro/Nano Structure of Ministry of Education, Wuhan University, Wuhan 430072, China. and Shenzhen Research Institute, Wuhan University, Shenzhen 518000, China
| | - Daoming Zhu
- School of Physics & Technology, Key Laboratory of Artificial Micro/Nano Structure of Ministry of Education, Wuhan University, Wuhan 430072, China.
| | - Ming Chen
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| | - Keke Chen
- School of Physics & Technology, Key Laboratory of Artificial Micro/Nano Structure of Ministry of Education, Wuhan University, Wuhan 430072, China.
| | - Hailiang Liu
- School of Physics & Technology, Key Laboratory of Artificial Micro/Nano Structure of Ministry of Education, Wuhan University, Wuhan 430072, China.
| | - Wei Liu
- School of Physics & Technology, Key Laboratory of Artificial Micro/Nano Structure of Ministry of Education, Wuhan University, Wuhan 430072, China.
| | - Yi Yang
- School of Physics & Technology, Key Laboratory of Artificial Micro/Nano Structure of Ministry of Education, Wuhan University, Wuhan 430072, China. and Shenzhen Research Institute, Wuhan University, Shenzhen 518000, China
| |
Collapse
|
42
|
Cheng SB, Chen MM, Wang YK, Sun ZH, Xie M, Huang WH. Current techniques and future advance of microfluidic devices for circulating tumor cells. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Bankó P, Lee SY, Nagygyörgy V, Zrínyi M, Chae CH, Cho DH, Telekes A. Technologies for circulating tumor cell separation from whole blood. J Hematol Oncol 2019; 12:48. [PMID: 31088479 PMCID: PMC6518774 DOI: 10.1186/s13045-019-0735-4] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022] Open
Abstract
The importance of early cancer diagnosis and improved cancer therapy has been clear for years and has initiated worldwide research towards new possibilities in the care strategy of patients with cancer using technological innovations. One of the key research fields involves the separation and detection of circulating tumor cells (CTC) because of their suggested important role in early cancer diagnosis and prognosis, namely, providing easy access by a liquid biopsy from blood to identify metastatic cells before clinically detectable metastasis occurs and to study the molecular and genetic profile of these metastatic cells. Provided the opportunity to further progress the development of technology for treating cancer, several CTC technologies have been proposed in recent years by various research groups and companies. Despite their potential role in cancer healthcare, CTC methods are currently mainly used for research purposes, and only a few methods have been accepted for clinical application because of the difficulties caused by CTC heterogeneity, CTC separation from the blood, and a lack of thorough clinical validation. Therefore, the standardization and clinical application of various developed CTC technologies remain important subsequent necessary steps. Because of their suggested future clinical benefits, we focus on describing technologies using whole blood samples without any pretreatment and discuss their advantages, use, and significance. Technologies using whole blood samples utilize size-based, immunoaffinity-based, and density-based methods or combinations of these methods as well as positive and negative enrichment during separation. Although current CTC technologies have not been truly implemented yet, they possess high potential as future clinical diagnostic techniques for the individualized therapy of patients with cancer. Thus, a detailed discussion of the clinical suitability of these new advanced technologies could help prepare clinicians for the future and can be a foundation for technologies that would be used to eliminate CTCs in vivo.
Collapse
Affiliation(s)
- Petra Bankó
- Department of Biochemical Engineering, Budapest University of Technology and Economics, Budapest, Hungary
| | - Sun Young Lee
- Department of Radiation Oncology, Chonbuk National University Hospital, Jeonju, Republic of Korea
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical, Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | | | - Miklós Zrínyi
- Laboratory of Nanochemistry, Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Chang Hoon Chae
- Laboratory of Nanochemistry, Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Dong Hyu Cho
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical, Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
- Department of Obstetrics and Gynecology, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - András Telekes
- Department of Oncology, St. Lazarus Hospital, Salgótarján, Hungary
| |
Collapse
|
44
|
Sun H, Han L, Yang L, Yang Y, Jiang W, Xu T, Jia L. Modular Chamber Assembled with Cell-Replicated Surface for Capture of Cancer Cells. ACS Biomater Sci Eng 2019; 5:2647-2656. [PMID: 33405768 DOI: 10.1021/acsbiomaterials.8b01605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The capture of circulating tumor cells (CTCs) is mainly carried out with a small volume of blood using magnetic nanoparticles and complex microfluidics. In this study, we propose a CTC-capture apparatus based on a modular design and called this apparatus as the CTC chamber. Distinct from other CTC-capture apparatuses, the capacity of the CTC chamber could be altered by varying the number of CTC-capture modules to accommodate the different volumes of blood sample. The core component of the CTC-capture module was a polydimethylsiloxane (PDMS) film with cell-replicated topological structure and anti-EpCAM antibody coating. Both synergistic roles can enhance the capture yield of cancer cells. Furthermore, the CTC chamber was assembled with one or three CTC-capture modules for the capture of cancer cells from spiked blood samples representing late-stage (3 mL of blood, 10 cancer cells mL-1) or middle-early stage (9 mL of blood, 1 cancer cell mL-1) cancer. The results showed that high capture yield (EpCAM-positive, ∼80%; EpCAM-negative, ∼65%) and purity (EpCAM-positive, ∼90%; EpCAM-negative, ∼80%) could be obtained within 1 h. This economic and facile CTC chamber could therefore open up opportunities for designing the next-generation CTC detection devices suitable for the diagnosis of different stages of cancer.
Collapse
Affiliation(s)
- He Sun
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Life Science and Biotechnology, Dalian University of Technology, No. 2 Linggong Road, Dalian, Liaoning 116023, P. R. China
| | - Lulu Han
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Life Science and Biotechnology, Dalian University of Technology, No. 2 Linggong Road, Dalian, Liaoning 116023, P. R. China
| | - Liwei Yang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Life Science and Biotechnology, Dalian University of Technology, No. 2 Linggong Road, Dalian, Liaoning 116023, P. R. China
| | - Yan Yang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Life Science and Biotechnology, Dalian University of Technology, No. 2 Linggong Road, Dalian, Liaoning 116023, P. R. China
| | - Wenning Jiang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Life Science and Biotechnology, Dalian University of Technology, No. 2 Linggong Road, Dalian, Liaoning 116023, P. R. China
| | - Ting Xu
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Life Science and Biotechnology, Dalian University of Technology, No. 2 Linggong Road, Dalian, Liaoning 116023, P. R. China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Life Science and Biotechnology, Dalian University of Technology, No. 2 Linggong Road, Dalian, Liaoning 116023, P. R. China
| |
Collapse
|
45
|
Vinothini K, Rajendran NK, Munusamy MA, Alarfaj AA, Rajan M. Development of biotin molecule targeted cancer cell drug delivery of doxorubicin loaded κ-carrageenan grafted graphene oxide nanocarrier. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:676-687. [PMID: 30948104 DOI: 10.1016/j.msec.2019.03.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 02/22/2019] [Accepted: 03/04/2019] [Indexed: 01/17/2023]
Abstract
Cervical cancer is one of the most occurring cancers and the fourth leading occurrence of cancer in women, worldwide. In this study, we planned to synthesis κ-Carrageenan grafted graphene oxide nanocarrier conjugated with biotin (GO-κ-Car-biotin) for targeted cervical cancer. Doxorubicin (DOX) is a well-known anticancer drug for any type of cancer and it is used to entrap over on the graphene oxide surface via π-π stacking interaction. The chemical function and crystalline nature of the synthesized nanocarrier was characterized by Fourier Transformed Infrared Spectroscopy (FT-IR) and X-ray diffraction Analysis (XRD). The surface morphological study was carried out through Scanning Electron Microscopy (SEM), Transmission electron microscopy (TEM) and Atomic force microscopy (AFM). The in-vitro drug release profile of DOX was carried out by UV-Vis spectrometer at the λmax value of 480 nm. The entrapment of DOX on GO-κ-car-biotin has been observed at 94%. The hydrophilic DOX drug has excellent pH-sensitive drug released in an in-vitro study. The anticancer efficiency of the synthesized GO-based nanocarrier was examined using HeLa cell line in-vitro. Cell viability, proliferation, cytotoxicity, and nuclear chromatin condensation was studied by trypan blue assay, triphosphate assay (ATP), lactate dehydrogenase assay (LDH) and Hoechst staining respectively. Finally, biotin leading GO-κ-Car carrier demonstrated is a promising drug delivery system for cervical cancer treatment.
Collapse
Affiliation(s)
- Kandasamy Vinothini
- Biomaterials in Medicinal Chemistry Laboratory, Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai 625021, Tamil Nadu, India
| | - Naresh Kumar Rajendran
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Murugan A Munusamy
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia.
| | - Abdulla A Alarfaj
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mariappan Rajan
- Biomaterials in Medicinal Chemistry Laboratory, Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai 625021, Tamil Nadu, India
| |
Collapse
|
46
|
Zhou X, Luo B, Kang K, Ma S, Sun X, Lan F, Yi Q, Wu Y. Multifunctional luminescent immuno-magnetic nanoparticles: toward fast, efficient, cell-friendly capture and recovery of circulating tumor cells. J Mater Chem B 2018; 7:393-400. [PMID: 32254726 DOI: 10.1039/c8tb02701c] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Highly efficient isolation and recovery of viable circulating tumor cells (CTCs) from the blood of patients is an important precondition to address the current dilemma of insufficient CTC studies, and can promote the development of individualized antitumor therapies. Herein, a cell-friendly CTC isolation and recovery nanoplatform with luminescent labelling was established using a layer-by-layer (LbL) assembly technique and a stimulated cellular-release strategy. In particular, the anti-epithelial cell adhesion molecule (anti-EpCAM) antibody was introduced with a disulfide bond-containing linker for further bio-friendly recovery of the CTCs. Quantum dots (QDs) were deposited onto fast magnet-responsive Fe3O4 nanoparticles through a facile LbL assembly method to monitor the capture and recovery process in real time. The obtained PEGlyated immuno-magnetic nanospheres (PIMNs) can all be magnetically collected within 2 min. Capture efficiencies above 90% can be achieved from blood samples with 5-200 CTCs per mL after only 1-2 min incubation. Nearly all PIMNs on the surface of the CTCs were detached after 15 min of glutathione (GSH) treatment with the disappearance of QD signals. Recovered CTCs could be directly used for culture (cell viability, ∼98%), and their invasiveness and migration characteristics remained unchanged. Furthermore, the PIMNs were successfully applied to isolate CTCs in cancer patients' peripheral blood samples, and an average of 8.6 ± 5.8 CTCs per mL was detected. The results above suggested that PIMNs may serve as a powerful nanoplatform for CTC screening, isolation and recovery.
Collapse
Affiliation(s)
- Xiaoxi Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Rd., Chengdu 610064, Sichuan, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Chen Y, Wang L, Guo D, Sheng C, Dai H, Shi X, Zhang W, Huang Q, Peng C, Chen W. A rapid and efficient technique for liposomal and nonliposomal drug pharmacokinetics studies using magnetic nanoprobes and its application to leakage kinetics of liposomes. J Chromatogr A 2018; 1580:2-11. [PMID: 30391033 DOI: 10.1016/j.chroma.2018.10.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/17/2018] [Accepted: 10/17/2018] [Indexed: 10/28/2022]
Abstract
Currently, the pharmacokinetics of liposomes was researched in vivo by measuring the total amount of drug in plasma. This method of using the total drug amount instead of the free drug amount virtually increase the apparent exposure and apparent biological distribution. To solve this problem, we developed a rapid and efficient method by using well-established streptavidin-functional Fe3O4@PDA as the separation nanoprobes to efficiently isolate biotin-labeled DTX-liposomes over 75% from plasma in the presence of magnetic field. The isolation procedure takes only 20 min and the concentration of DTX in liposomes from plasma was determined by LC-MS/MS. The method for the determination of DTX in plasma was linear in the range of 5-5000 ng/mL, and the correlation coefficient was 0.9989. Results obtained in this study clearly demonstrated that the pharmacokinetic parameters of non-liposomal drug and total drug are different in vivo. Therefore, traditional method for studying the pharmacokinetics of liposomes in vivo is unreasonable, and the new method mentioned here provided a strategy for studying the pharmacokinetics of liposomes.
Collapse
Affiliation(s)
- Yunna Chen
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
| | - Lei Wang
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China.
| | - Dongdong Guo
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China
| | - Chenming Sheng
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
| | - Haozhi Dai
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
| | - Xiaoyan Shi
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
| | - Wenjing Zhang
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
| | - Qianqian Huang
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
| | - Can Peng
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
| | - Weidong Chen
- Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China.
| |
Collapse
|
48
|
Functional and biocompatible polymeric ionic liquid (PIL) - Decorated immunomagnetic nanospheres for the efficient capture of rare number CTCs. Anal Chim Acta 2018; 1044:162-173. [DOI: 10.1016/j.aca.2018.07.066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/22/2018] [Accepted: 07/25/2018] [Indexed: 11/22/2022]
|
49
|
Shen Z, Wu A, Chen X. Current detection technologies for circulating tumor cells. Chem Soc Rev 2018; 46:2038-2056. [PMID: 28393954 DOI: 10.1039/c6cs00803h] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Circulating tumor cells (CTCs) are cancer cells that circulate in the blood stream after being naturally shed from original or metastatic tumors, and can lead to a new fatal metastasis. CTCs have become a hotspot research field during the last decade. Detection of CTCs, as a liquid biopsy of tumors, can be used for early diagnosis of cancers, earlier evaluation of cancer recurrence and chemotherapeutic efficacy, and choice of individual sensitive anti-cancer drugs. Therefore, CTC detection is a crucial tool to fight against cancer. Herein, we classify the currently reported CTC detection technologies, introduce some representative samples for each technology, conclude the advantages and limitations, and give a future perspective including the challenges and opportunities of CTC detection.
Collapse
Affiliation(s)
- Zheyu Shen
- CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, Zhejiang 315201, China.
| | | | | |
Collapse
|
50
|
Yu Y, Yang Y, Ding J, Meng S, Li C, Yin X. Design of a Biocompatible and Ratiometric Fluorescent probe for the Capture, Detection, Release, and Reculture of Rare Number CTCs. Anal Chem 2018; 90:13290-13298. [DOI: 10.1021/acs.analchem.8b02625] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu P.R.China
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu P.R.China
| | - Yuan Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu P.R.China
| | - Jinhua Ding
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu P.R.China
| | - Si Meng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu P.R.China
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu P.R.China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu P.R.China
| |
Collapse
|