1
|
Neagu AN, Whitham D, Bruno P, Versaci N, Biggers P, Darie CC. Tumor-on-chip platforms for breast cancer continuum concept modeling. Front Bioeng Biotechnol 2024; 12:1436393. [PMID: 39416279 PMCID: PMC11480020 DOI: 10.3389/fbioe.2024.1436393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Our previous article entitled "Proteomics and its applications in breast cancer", proposed a Breast Cancer Continuum Concept (BCCC), including a Breast Cancer Cell Continuum Concept as well as a Breast Cancer Proteomic Continuum Concept. Breast cancer-on-chip (BCoC), breast cancer liquid biopsy-on-chip (BCLBoC), and breast cancer metastasis-on-chip (BCMoC) models successfully recapitulate and reproduce in vitro the principal mechanisms and events involved in BCCC. Thus, BCoC, BCLBoC, and BCMoC platforms allow for multiple cell lines co-cultivation to reproduce BC hallmark features, recapitulating cell proliferation, cell-to-cell communication, BC cell-stromal crosstalk and stromal activation, effects of local microenvironmental conditions on BC progression, invasion/epithelial-mesenchymal transition (EMT)/migration, intravasation, dissemination through blood and lymphatic circulation, extravasation, distant tissues colonization, and immune escape of cancer cells. Moreover, tumor-on-chip platforms are used for studying the efficacy and toxicity of chemotherapeutic drugs/nano-drugs or nutraceuticals. Therefore, the aim of this review is to summarize and analyse the main bio-medical roles of on-chip platforms that can be used as powerful tools to study the metastatic cascade in BC. As future direction, integration of tumor-on-chip platforms and proteomics-based specific approaches can offer important cues about molecular profile of the metastatic cascade, alowing for novel biomarker discovery. Novel microfluidics-based platforms integrating specific proteomic landscape of human milk, urine, and saliva could be useful for early and non-invasive BC detection. Also, risk-on-chip models may improve BC risk assessment and prevention based on the identification of biomarkers of risk. Moreover, multi-organ-on-chip systems integrating patient-derived BC cells and patient-derived scaffolds have a great potential to study BC at integrative level, due to the systemic nature of BC, for personalized and precision medicine. We also emphasized the strengths and weaknesses of BCoC and BCMoC platforms.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| | - Pathea Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| | - Nicholas Versaci
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| | - Peter Biggers
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| |
Collapse
|
2
|
Duan X, Qin W, Hao J, Yu X. Recent advances in the applications of DNA frameworks in liquid biopsy: A review. Anal Chim Acta 2024; 1308:342578. [PMID: 38740462 DOI: 10.1016/j.aca.2024.342578] [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: 12/20/2023] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 05/16/2024]
Abstract
Cancer is one of the serious threats to public life and health. Early diagnosis, real-time monitoring, and individualized treatment are the keys to improve the survival rate and prolong the survival time of cancer patients. Liquid biopsy is a potential technique for cancer early diagnosis due to its non-invasive and continuous monitoring properties. However, most current liquid biopsy techniques lack the ability to detect cancers at the early stage. Therefore, effective detection of a variety of cancers is expected through the combination of various techniques. Recently, DNA frameworks with tailorable functionality and precise addressability have attracted wide spread attention in biomedical applications, especially in detecting cancer biomarkers such as circulating tumor cells (CTCs), exosomes and circulating tumor nucleic acid (ctNA). Encouragingly, DNA frameworks perform outstanding in detecting these cancer markers, but also face some challenges and opportunities. In this review, we first briefly introduced the development of DNA frameworks and its typical structural characteristics and advantages. Then, we mainly focus on the recent progress of DNA frameworks in detecting commonly used cancer markers in liquid-biopsy. We summarize the advantages and applications of DNA frameworks for detecting CTCs, exosomes and ctNA. Furthermore, we provide an outlook on the possible opportunities and challenges for exploiting the structural advantages of DNA frameworks in the field of cancer diagnosis. Finally, we envision the marriage of DNA frameworks with other emerging materials and technologies to develop the next generation of disease diagnostic biosensors.
Collapse
Affiliation(s)
- Xueyuan Duan
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Science, China Jiliang University, Hangzhou, 310018, China
| | - Weiwei Qin
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Science, China Jiliang University, Hangzhou, 310018, China.
| | - Jicong Hao
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Science, China Jiliang University, Hangzhou, 310018, China
| | - Xiaoping Yu
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Science, China Jiliang University, Hangzhou, 310018, China.
| |
Collapse
|
3
|
Allen TA. The Role of Circulating Tumor Cells as a Liquid Biopsy for Cancer: Advances, Biology, Technical Challenges, and Clinical Relevance. Cancers (Basel) 2024; 16:1377. [PMID: 38611055 PMCID: PMC11010957 DOI: 10.3390/cancers16071377] [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: 02/27/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer remains a leading cause of mortality worldwide, with metastasis significantly contributing to its lethality. The metastatic spread of tumor cells, primarily through the bloodstream, underscores the importance of circulating tumor cells (CTCs) in oncological research. As a critical component of liquid biopsies, CTCs offer a non-invasive and dynamic window into tumor biology, providing invaluable insights into cancer dissemination, disease progression, and response to treatment. This review article delves into the recent advancements in CTC research, highlighting their emerging role as a biomarker in various cancer types. We explore the latest technologies and methods for CTC isolation and detection, alongside novel approaches to characterizing their biology through genomics, transcriptomics, proteomics, and epigenetic profiling. Additionally, we examine the clinical implementation of these findings, assessing how CTCs are transforming the landscape of cancer diagnosis, prognosis, and management. By offering a comprehensive overview of current developments and potential future directions, this review underscores the significance of CTCs in enhancing our understanding of cancer and in shaping personalized therapeutic strategies, particularly for patients with metastatic disease.
Collapse
|
4
|
Comparative application of microfluidic systems in circulating tumor cells and extracellular vesicles isolation; a review. Biomed Microdevices 2022; 25:4. [PMID: 36574057 DOI: 10.1007/s10544-022-00644-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 12/28/2022]
Abstract
Cancer is a prevalent cause of mortality globally, where early diagnosis leads to a reduced death rate. Many researchers' common strategies are based on personalized diagnostic methods with rapid response and high accuracy. This technology was developed by applying liquid biopsy instead of tissue biopsies in the case of tumor cell analysis that facilitates point-of-care testing for cancer diagnosis and treatment. In recent years, significant progress in microfluidic technology led to the successful isolation, analysis, and monitoring of cancer biomarkers in body liquid biopsy with merits like high sensitivity and flexibility, low sample usage, cost effective, and the ability of automation. The most critical and informative markers in body liquid refer to circulating tumor cells (CTCs) and extracellular vesicles derived from tumors (EVs) that carry various biomarkers in their structure (DNAs, proteins, and RNAs) as compared to ctDNA. The released ctDNA has a low half-life and decreased sensitivity due to large amounts of nucleic acid in serum. This review intends to highlight different cancer screening tests with a particular focus on the details regarding the only FDA-approved and awaiting technologies for FDA clearance to isolate CTCs and EVs based on microfluidics systems.
Collapse
|
5
|
Imparato G, Urciuolo F, Mazio C, Netti PA. Capturing the spatial and temporal dynamics of tumor stroma for on-chip optimization of microenvironmental targeting nanomedicine. LAB ON A CHIP 2022; 23:25-43. [PMID: 36305728 DOI: 10.1039/d2lc00611a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Malignant cells grow in a complex microenvironment that plays a key role in cancer progression. The "dynamic reciprocity" existing between cancer cells and their microenvironment is involved in cancer differentiation, proliferation, invasion, metastasis, and drug response. Therefore, understanding the molecular mechanisms underlying the crosstalk between cancer cells and their surrounding tissue (i.e., tumor stroma) and how this interplay affects the disease progression is fundamental to design and validate novel nanotherapeutic approaches. As an important regulator of tumor progression, metastasis and therapy resistance, the extracellular matrix of tumors, the acellular component of the tumor microenvironment, has been identified as very promising target of anticancer treatment, revolutionizing the traditional therapeutic paradigm that sees the neoplastic cells as the preferential objective to fight cancer. To design and to validate such a target therapy, advanced 3D preclinical models are necessary to correctly mimic the complex, dynamic and heterogeneous tumor microenvironment. In addition, the recent advancement in microfluidic technology allows fine-tuning and controlling microenvironmental parameters in tissue-on-chip devices in order to emulate the in vivo conditions. In this review, after a brief description of the origin of tumor microenvironment heterogeneity, some examples of nanomedicine approaches targeting the tumor microenvironment have been reported. Further, how advanced 3D bioengineered tumor models coupled with a microfluidic device can improve the design and testing of anti-cancer nanomedicine targeting the tumor microenvironment has been discussed. We highlight that the presence of a dynamic extracellular matrix, able to capture the spatiotemporal heterogeneity of tumor stroma, is an indispensable requisite for tumor-on-chip model and nanomedicine testing.
Collapse
Affiliation(s)
- Giorgia Imparato
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
| | - Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy
| | - Claudia Mazio
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
| | - Paolo A Netti
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy
| |
Collapse
|
6
|
Kang H, Xiong Y, Ma L, Yang T, Xu X. Recent advances in micro-/nanostructure array integrated microfluidic devices for efficient separation of circulating tumor cells. RSC Adv 2022; 12:34892-34903. [PMID: 36540264 PMCID: PMC9724214 DOI: 10.1039/d2ra06339e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/18/2022] [Indexed: 09/06/2023] Open
Abstract
Circulating tumor cells (CTCs) released from the primary tumor to peripheral blood are promising targets for liquid biopsies. Their biological information is vital for early cancer detection, efficacy assessment, and prognostic monitoring. Despite the tremendous clinical applications of CTCs, development of effective separation techniques are still demanding. Traditional separation methods usually use batch processing for enrichment, which inevitably destroy cell integrity and affect the complete information acquisition. Considering the rarity and heterogeneity of CTCs, it is urgent to develop effective separation methods. Microfluidic chips with precise fluid control at the micron level are promising devices for CTC separation. Their further combination with micro-/nanostructure arrays adds more biomolecule binding sites and exhibit unique fluid barrier effect, which significantly improve the CTC capture efficiency, purity, and sensitivity. This review summarized the recent advances in micro-/nanostructure array integrated microfluidic devices for CTC separation, including microrods, nanowires, and 3D micro-/nanostructures. The mechanisms by which these structures contribute to improved capture efficiency are discussed. Two major categories of separation methods, based on the physical and biological properties of CTCs, are discussed separately. Physical separation includes the design and preparation of micro-/nanostructure arrays, while chemical separation additionally involves the selection and modification of specific capture probes. These emerging technologies are expected to become powerful tools for disease diagnosis in the future.
Collapse
Affiliation(s)
- Hanyue Kang
- School of Materials Science and Engineering, Tongji University Shanghai 201804 China
| | - Yuting Xiong
- School of Materials Science and Engineering, Tongji University Shanghai 201804 China
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University Hangzhou 310058 China
| | - Tongqing Yang
- School of Materials Science and Engineering, Tongji University Shanghai 201804 China
| | - Xiaobin Xu
- School of Materials Science and Engineering, Tongji University Shanghai 201804 China
| |
Collapse
|
7
|
Xiang Y, Hu C, Wu G, Xu S, Li Y. Nanomaterial-based microfluidic systems for cancer biomarker detection: Recent applications and future perspectives. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
8
|
Chen Y, Yang Y, Feng J, Carrier AJ, Tyagi D, Yu X, Wang C, Oakes KD, Zhang X. A universal monoclonal antibody-aptamer conjugation strategy for selective non-invasive bioparticle isolation from blood on a regenerative microfluidic platform. Acta Biomater 2022; 152:210-220. [PMID: 36087870 DOI: 10.1016/j.actbio.2022.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022]
Abstract
Simultaneous isolation of various circulating tumor cell (CTC) subtypes from whole blood is useful in cancer diagnosis and prognosis. Microfluidic affinity separation devices are promising for CTC separation because of their high throughput capacity and automatability. However, current affinity agents, such as antibodies (mAbs) and aptamers (Apts) alone, are still suboptimal for efficient, consistent, and versatile cell analysis. By introducing a hybrid affinity agent, i.e., an aptamer-antibody (Apt-mAb) conjugate, we developed a universal and regenerative microchip with high efficiency and non-invasiveness in the separation and profiling of various CTCs from blood. The Apt-mAb conjugate consists of a monoclonal antibody that specifically binds the target cell receptor and a surface-bound aptamer that recognizes the conserved Fc region of the mAb. The aptamer then indirectly links the surface functionalization of the microfluidic channels to the mAbs. This hybrid affinity agent and the microchip platform may be widely useful for various bio-particle separations in different biological matrices. Further, the regeneration capability of the microchip improves data consistency between multiple uses and minimizes plastic waste while promoting environmental sustainability. STATEMENT OF SIGNIFICANCE: A hybrid affinity agent, Apt-mAb, consisting of a universal aptamer (Apt) that binds the conserved Fc region of monoclonal antibodies (mAbs) was developed. The invented nano-biomaterial combines the strengths and overcomes the weakness of both Apts and mAbs, thus changing the paradigm of affinity separation of cell subtypes. When Apt-mAb was used to fabricate microfluidic chips using a "universal screwdriver" approach, the microchip could be easily tuned to bind any cell type, exhibiting great universality. Besides high sensitivity and selectivity, the superior regenerative capacity of the microchips makes them reusable, which provides improved consistency and repeatability in cell profiling and opens a new approach towards in vitro diagnostic point-of-care testing devices with environmental sustainability and cost-effectiveness.
Collapse
Affiliation(s)
- Yongli Chen
- Department of Biological Applied Engineering, Shenzhen Key Laboratory of Fermentation Purification and Analysis, Shenzhen Polytechnic, Shenzhen, 518055, China
| | - Yikun Yang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, China.
| | - Jinglong Feng
- Department of Biological Applied Engineering, Shenzhen Key Laboratory of Fermentation Purification and Analysis, Shenzhen Polytechnic, Shenzhen, 518055, China
| | - Andrew J Carrier
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia, B1P 6L2, Canada
| | - Deependra Tyagi
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia, B1P 6L2, Canada
| | - Xin Yu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, China
| | - Chunguang Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, China
| | - Ken D Oakes
- Department of Biology, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia, B1P 6L2, Canada
| | - Xu Zhang
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia, B1P 6L2, Canada.
| |
Collapse
|
9
|
Wang Y, Li J, Pei Z, Pei Y. A glutathione activatable bioprobe for detection of hepatocellular carcinoma cells in peripheral blood via carbohydrate-protein interaction. Anal Chim Acta 2022; 1221:340106. [DOI: 10.1016/j.aca.2022.340106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/14/2022] [Accepted: 06/19/2022] [Indexed: 11/01/2022]
|
10
|
Wang Y, Gao Y, Yin Y, Pan Y, Wang Y, Song Y. Nanomaterial-assisted microfluidics for multiplex assays. Mikrochim Acta 2022; 189:139. [PMID: 35275267 DOI: 10.1007/s00604-022-05226-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/10/2022] [Indexed: 02/07/2023]
Abstract
Simultaneous detection of different biomarkers from a single specimen in a single test, allowing more rapid, efficient, and low-cost analysis, is of great significance for accurate diagnosis of disease and efficient monitoring of therapy. Recently, developments in microfabrication and nanotechnology have advanced the integration of nanomaterials in microfluidic devices toward multiplex assays of biomarkers, combining both the advantages of microfluidics and the unique properties of nanomaterials. In this review, we focus on the state of the art in multiplexed detection of biomarkers based on nanomaterial-assisted microfluidics. Following an overview of the typical microfluidic analytical techniques and the most commonly used nanomaterials for biochemistry analysis, we highlight in detail the nanomaterial-assisted microfluidic strategies for different biomarkers. These highly integrated platforms with minimum sample consumption, high sensitivity and specificity, low detection limit, enhanced signals, and reduced detection time have been extensively applied in various domains and show great potential in future point-of-care testing and clinical diagnostics.
Collapse
Affiliation(s)
- Yanping Wang
- Sino-French Engineer School, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Yanfeng Gao
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Yi Yin
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yongchun Pan
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Yuzhen Wang
- Key Laboratory of Flexible Electronics & Institute of Advanced Materials, Jiangsu National Synergistic Innovation Center for Advanced Materials, Nanjing Tech University, Nanjing, 211816, China
| | - Yujun Song
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
11
|
Qian H, Zhang Y, Xu J, He J, Gao W. Progress and application of circulating tumor cells in non-small cell lung cancer. Mol Ther Oncolytics 2021; 22:72-84. [PMID: 34514090 PMCID: PMC8408556 DOI: 10.1016/j.omto.2021.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) has the highest morbidity and mortality worldwide among malignant tumors. NSCLC is a great threat to health and well-being. Biopsy is the gold standard to diagnose lung cancer, but traditional biopsy methods cannot fully reflect the true condition of tumors. There is growing evidence that a single-point biopsy fails to reveal the complete landscape of the tumor due to intratumor heterogeneity, but it is impractical to complete multiple biopsies that are separated both spatially and temporally. Liquid biopsy heralds that a new era is coming. Circulating tumor cells (CTCs) are tumor cells that circulate in the peripheral blood after being shed from primary or metastatic tumors. CTCs constitute a considerable portion of a liquid biopsy, which contributes to the diagnosis, assessment of prognosis, and therapy of NSCLC. Herein, this review discusses the technologies for detection and enrichment of CTCs as well as clinical applications involving CTCs.
Collapse
Affiliation(s)
- Huizhu Qian
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Yue Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Jing Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Jing He
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Wen Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| |
Collapse
|
12
|
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: 0.8] [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
|
13
|
Wei X, Chen K, Guo S, Liu W, Zhao XZ. Emerging Microfluidic Technologies for the Detection of Circulating Tumor Cells and Fetal Nucleated Red Blood Cells. ACS APPLIED BIO MATERIALS 2021; 4:1140-1155. [DOI: 10.1021/acsabm.0c01325] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Xiaoyun Wei
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
- Key Laboratory of Medical Information and 3D Bioprinting of Zhejiang Province, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Keke Chen
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| | - Shishang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| |
Collapse
|
14
|
Chircov C, Bîrcă AC, Grumezescu AM, Andronescu E. Biosensors-on-Chip: An Up-to-Date Review. Molecules 2020; 25:E6013. [PMID: 33353220 PMCID: PMC7765790 DOI: 10.3390/molecules25246013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Generally, biosensors are designed to translate physical, chemical, or biological events into measurable signals, thus offering qualitative and/or quantitative information regarding the target analytes. While the biosensor field has received considerable scientific interest, integrating this technology with microfluidics could further bring significant improvements in terms of sensitivity and specificity, resolution, automation, throughput, reproducibility, reliability, and accuracy. In this manner, biosensors-on-chip (BoC) could represent the bridging gap between diagnostics in central laboratories and diagnostics at the patient bedside, bringing substantial advancements in point-of-care (PoC) diagnostic applications. In this context, the aim of this manuscript is to provide an up-to-date overview of BoC system development and their most recent application towards the diagnosis of cancer, infectious diseases, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Cristina Chircov
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (C.C.); (A.C.B.); (E.A.)
| | - Alexandra Cătălina Bîrcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (C.C.); (A.C.B.); (E.A.)
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (C.C.); (A.C.B.); (E.A.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (C.C.); (A.C.B.); (E.A.)
| |
Collapse
|
15
|
A dual recognition strategy for accurate detection of CTCs based on novel branched PtAuRh trimetallic nanospheres. Biosens Bioelectron 2020; 176:112893. [PMID: 33342693 DOI: 10.1016/j.bios.2020.112893] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Accurate detection of circulating tumor cells (CTCs) has a pivotal role in the metastasis monitoring and prognosis of tumor. In this work, an ultrasensitive electrochemical cytosensor was developed based on excellent electrocatalytic materials and a dual recognition strategy. Herein, novel branched PtAuRh trimetallic nanospheres (b-PtAuRh TNS) were synthesized for the first time by a facile one-pot method, which had a huge specific surface area and outstanding catalytic activity. B-PtAuRh TNS linked with aptamers targeting mucin1 (MUC1) were served as signal tags to amplify the signal. As electrode modified material, the nanocomposites of Cabot carbon black (BP2000) and AuNPs were used to improve the electron transfer efficiency of electrode. In addition to using b-PtAuRh TNS labeled anti-MUC1 aptamers as signal probes, anti-EpCAM antibodies were worked as capture probes to achieve dual recognition of target cells. In other words, only cells expressing both MUC1 and EpCAM could produce electrochemical signal. The constructed cytosensor presented a wide linear range (5 - 1 × 106 cells mL-1) and a low detection limit (1 cell mL-1). It was worth noting that the proposed cytosensor could detect CTCs in clinical blood samples. To sum up, the developed cytosensor might become a promising detection platform for cancer diagnosis and tumor metastasis.
Collapse
|
16
|
Cheng J, Liu Y, Zhao Y, Zhang L, Zhang L, Mao H, Huang C. Nanotechnology-Assisted Isolation and Analysis of Circulating Tumor Cells on Microfluidic Devices. MICROMACHINES 2020; 11:E774. [PMID: 32823926 PMCID: PMC7465711 DOI: 10.3390/mi11080774] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022]
Abstract
Circulating tumor cells (CTCs), a type of cancer cell that spreads from primary tumors into human peripheral blood and are considered as a new biomarker of cancer liquid biopsy. It provides the direction for understanding the biology of cancer metastasis and progression. Isolation and analysis of CTCs offer the possibility for early cancer detection and dynamic prognosis monitoring. The extremely low quantity and high heterogeneity of CTCs are the major challenges for the application of CTCs in liquid biopsy. There have been significant research endeavors to develop efficient and reliable approaches to CTC isolation and analysis in the past few decades. With the advancement of microfabrication and nanomaterials, a variety of approaches have now emerged for CTC isolation and analysis on microfluidic platforms combined with nanotechnology. These new approaches show advantages in terms of cell capture efficiency, purity, detection sensitivity and specificity. This review focuses on recent progress in the field of nanotechnology-assisted microfluidics for CTC isolation and detection. Firstly, CTC isolation approaches using nanomaterial-based microfluidic devices are summarized and discussed. The different strategies for CTC release from the devices are specifically outlined. In addition, existing nanotechnology-assisted methods for CTC downstream analysis are summarized. Some perspectives are discussed on the challenges of current methods for CTC studies and promising research directions.
Collapse
Affiliation(s)
- Jie Cheng
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Liu
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Lina Zhang
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China;
| | - Lingqian Zhang
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Haiyang Mao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Chengjun Huang
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
17
|
Khizar S, Ben Halima H, Ahmad NM, Zine N, Errachid A, Elaissari A. Magnetic nanoparticles in microfluidic and sensing: From transport to detection. Electrophoresis 2020; 41:1206-1224. [DOI: 10.1002/elps.201900377] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Sumera Khizar
- Université de Lyon LAGEP, UMR‐5007, CNRS, Université Lyon 1, 5007 43 Bd 11 Novembre 1918 Villeurbanne F‐69622 France
- Polymer Research Lab School of Chemical and Materials Engineering (SCME) National University of Sciences and Technology (NUST) H‐12 Sector Islamabad 44000 Pakistan
| | - Hamdi Ben Halima
- Université de Lyon Institut des Science Analytiques UMR 5280, CNRS Université Lyon 1 ENS Lyon-5, rue de la Doua Villeurbanne F‐69100 France
| | - Nasir M. Ahmad
- Polymer Research Lab School of Chemical and Materials Engineering (SCME) National University of Sciences and Technology (NUST) H‐12 Sector Islamabad 44000 Pakistan
| | - Nadia Zine
- Université de Lyon Institut des Science Analytiques UMR 5280, CNRS Université Lyon 1 ENS Lyon-5, rue de la Doua Villeurbanne F‐69100 France
| | - Abdelhamid Errachid
- Université de Lyon Institut des Science Analytiques UMR 5280, CNRS Université Lyon 1 ENS Lyon-5, rue de la Doua Villeurbanne F‐69100 France
| | - Abdelhamid Elaissari
- Université de Lyon LAGEP, UMR‐5007, CNRS, Université Lyon 1, 5007 43 Bd 11 Novembre 1918 Villeurbanne F‐69622 France
| |
Collapse
|
18
|
Yang J, Li X, Jiang B, Yuan R, Xiang Y. In Situ-Generated Multivalent Aptamer Network for Efficient Capture and Sensitive Electrochemical Detection of Circulating Tumor Cells in Whole Blood. Anal Chem 2020; 92:7893-7899. [PMID: 32338500 DOI: 10.1021/acs.analchem.0c01195] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Monitoring circulating tumor cells (CTCs) in human blood can offer useful information for convenient metastasis diagnosis, prognosis, and treatment of cancers. However, it remains a substantial challenge to detect CTCs because of their particular scarcity in complex peripheral blood. Herein, we describe an in situ-generated multivalent aptamer network-modified electrode interface for efficiently capturing and sensitively detecting CTCs in whole blood by electrochemistry. Such an interface was fabricated via rolling circle amplification extension of the electrode-immobilized primer/circular DNA complexes for the yield of long ssDNA strands with many repeated aptamer segments, which could achieve efficient capture of rare CTCs in a multivalent cooperative manner. The antibody and horseradish peroxidase-functionalized gold nanoparticles further specifically associated with the surface-bound CTCs and generated electrocatalytically amplified current outputs for highly sensitive detection of CTCs with an attractive detection limit of five cells. Also, the multivalent aptamer network interface could successfully distinguish the target cells from other control cells and achieve CTC detection in whole blood, demonstrating its promising potential for monitoring different rare CTCs in human blood.
Collapse
Affiliation(s)
- Jianmei Yang
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China
| | - Xiaolong Li
- School of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Bingying Jiang
- School of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Ruo Yuan
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China
| | - Yun Xiang
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China
| |
Collapse
|
19
|
Unni M, Zhang J, George TJ, Segal MS, Fan ZH, Rinaldi C. Engineering magnetic nanoparticles and their integration with microfluidics for cell isolation. J Colloid Interface Sci 2020; 564:204-215. [PMID: 31911225 PMCID: PMC7023483 DOI: 10.1016/j.jcis.2019.12.092] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 01/09/2023]
Abstract
Isolation of cancer cells, bacteria, and viruses from peripheral blood has important applications in cancer diagnosis, therapy monitoring, and drug development. Magnetic particles functionalized with antibodies that target receptors of cancer cells have been shown to isolate such entities using magnetic field gradients. Here, we report enhancement in capture efficiency and specificity by engineering magnetic nanoparticles and integrating them with microfluidics for the enumeration of tumor cells. Nanoparticles were made from iron oxide, coated with poly(ethylene glycol), and conjugated through avidin-biotin chemistry with antibody specifically against epithelial cell adhesion molecule (EpCAM). On exposure of targeted nanoparticles to tumor cells, specific uptake by EpCAM-expressing tumor cells (e.g., BxPC3, a pancreatic cancer cell) was observed, whereas there was negligible uptake by cells with low EpCAM expression (e.g., CCRF-CEM, a leukemia cell). Using an arrangement of magnets called a Halbach array, capture efficiency and specificity towards BxPC3 cells tagged with magnetic nanoparticles were enhanced, compared to conditions without the magnetic field gradient and/or without magnetic nanoparticles, either in buffer or in whole blood. These results illustrate that engineered magnetic nanoparticles and their integration with microfluidics have great potential for tumor cell enumeration and cancer prognosis.
Collapse
Affiliation(s)
- Mythreyi Unni
- Department of Chemical Engineering, Gainesville, FL 32611, USA
| | - Jinling Zhang
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL 32611, USA
| | - Thomas J George
- Department of Medicine, Gainesville, FL 32611, USA; University of Florida Health Cancer Center, Gainesville, FL 32611, USA
| | - Mark S Segal
- Department of Medicine, Gainesville, FL 32611, USA
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL 32611, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, Gainesville, FL 32611, USA; Department of Chemistry, University of Florida, Gainesville, FL 32611, USA; University of Florida Health Cancer Center, Gainesville, FL 32611, USA.
| | - Carlos Rinaldi
- Department of Chemical Engineering, Gainesville, FL 32611, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, Gainesville, FL 32611, USA; University of Florida Health Cancer Center, Gainesville, FL 32611, USA.
| |
Collapse
|
20
|
Engineering microfluidic chip for circulating tumor cells: From enrichment, release to single cell analysis. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.03.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Sun D, Lu J, Zhang L, Chen Z. Aptamer-based electrochemical cytosensors for tumor cell detection in cancer diagnosis: A review. Anal Chim Acta 2019; 1082:1-17. [PMID: 31472698 DOI: 10.1016/j.aca.2019.07.054] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 01/25/2023]
Abstract
Circulating tumor cells, a type of viable cancer cell circulating from primary or metastatic tumors in the blood stream, can lead to the parallel development of primary tumors and metastatic lesions. Highly selective and sensitive detection of tumor cells has become a hot research topic and can provide a basis for early diagnosis of cancers and anticancer drug evaluation to develop the best treatment plan. Aptamers are single-stranded oligonucleotides that can bind to target tumor cells in unique three-dimensional structures with high specificity and affinity. Aptamer-based methods or signal amplification methods using aptamers show great potential in improving the selectivity and sensitivity of electrochemical (EC) cytosensors for tumor cell detection. This review covers the remarkable developments in aptamer-based EC cytosensors for the identification of cell type, cell counting and detection of crucial proteins on the cell surface. Various EC techniques have been developed for cancer cell detection, including common voltammetry or impedance, electrochemiluminescence and photoelectrochemistry in a direct approach (aptamer-target cell), sandwich approach (capture probe-target cell-signaling probe) or other approach. The current challenges and promising opportunities in the establishment of EC aptamer cytosensors for tumor cell detection are also discussed.
Collapse
Affiliation(s)
- Duanping Sun
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China; School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Jing Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Luyong Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zuanguang Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
22
|
LeValley PJ, Tibbitt MW, Noren B, Kharkar P, Kloxin AM, Anseth KS, Toner M, Oakey J. Immunofunctional photodegradable poly(ethylene glycol) hydrogel surfaces for the capture and release of rare cells. Colloids Surf B Biointerfaces 2019; 174:483-492. [PMID: 30497010 PMCID: PMC6545105 DOI: 10.1016/j.colsurfb.2018.11.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022]
Abstract
Circulating tumor cells (CTCs) play a central role in cancer metastasis and represent a rich source of data for cancer prognostics and therapeutic guidance. Reliable CTC recovery from whole blood therefore promises a less invasive and more sensitive approach to cancer diagnosis and progression tracking. CTCs, however, are exceedingly rare in whole blood, making their quantitative recovery challenging. Several techniques capable of isolating these rare cells have been introduced and validated, yet most suffer from low CTC purity or viability, both of which are essential to develop a clinically viable CTC isolation platform. To address these limitations, we introduce a patterned, immunofunctional, photodegradable poly(ethylene glycol) (PEG) hydrogel capture surface for the isolation and selective release of rare cell populations. Flat and herringbone capture surfaces were successfully patterned via PDMS micromolding and photopolymerization of photolabile PEG hydrogels. Patterned herringbone surfaces, designed to convectively transport cells to the capture surface, exhibited improved capture density relative to flat surfaces for target cell capture from buffer, buffy coat, and whole blood. Uniquely, captured cells were released for collection by degrading the hydrogel capture surface with either bulk or targeted irradiation with cytocompatible doses of long wavelength UV light. Recovered cells remained viable following capture and release and exhibited similar growth rates as untreated control cells. The implementation of molded photodegradable PEG hydrogels as a CTC capture surface provides a micropatternable, cytocompatible platform that imparts the unique ability to recover pure, viable CTC samples by selectively releasing target cells.
Collapse
Affiliation(s)
- Paige J LeValley
- Department of Chemical Engineering, University of Wyoming, Laramie, WY 82071, United States; Department of Biomolecular and Chemical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Mark W Tibbitt
- Department of Chemical and Biological Engineering, University of Colorado Boulder,Boulder, CO 80309, United States; Department of Mechanical and Process Engineering, ETH Zürich, Zürich, 8092, Switzerland
| | - Ben Noren
- Department of Chemical Engineering, University of Wyoming, Laramie, WY 82071, United States
| | - Prathamesh Kharkar
- Department of Biomolecular and Chemical Engineering, University of Delaware, Newark, DE 19716, United States
| | - April M Kloxin
- Department of Biomolecular and Chemical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder,Boulder, CO 80309, United States; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, United States; Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Mehmet Toner
- BioMEMS Resource Center, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Shriners Hospital for Children and Harvard Medical School, Boston, MA 02114, United States
| | - John Oakey
- Department of Chemical Engineering, University of Wyoming, Laramie, WY 82071, United States.
| |
Collapse
|
23
|
He Y, Qin J, Wu S, Yang H, Wen H, Wang Y. Cancer cell–nanomaterial interface: role of geometry and surface charge of nanocomposites in the capture efficiency and cell viability. Biomater Sci 2019; 7:2759-2768. [DOI: 10.1039/c9bm00037b] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The nanomaterial–cell interface plays an important role in biodetection and therapy. The experimental results in this study indicated that the magnetic nanocomposites with strong positive surface charge but different geometry interacted with cancer cells in different ways, leading to various cell capture efficiency and cytotoxicity.
Collapse
Affiliation(s)
- Yishu He
- School of Chemical Engineering
- Northwest university
- Xi'an
- 710069 P.R. China
- The Institute for Translational Nanomedicine
| | - Jingwen Qin
- The Institute for Translational Nanomedicine
- Shanghai East Hospital
- the Institute for Biomedical Engineering & Nano Science
- Tongji University School of Medicine
- Shanghai 200092
| | - Shengming Wu
- The Institute for Translational Nanomedicine
- Shanghai East Hospital
- the Institute for Biomedical Engineering & Nano Science
- Tongji University School of Medicine
- Shanghai 200092
| | - Haocheng Yang
- The Institute for Translational Nanomedicine
- Shanghai East Hospital
- the Institute for Biomedical Engineering & Nano Science
- Tongji University School of Medicine
- Shanghai 200092
| | - Huiyun Wen
- School of Chemical Engineering
- Northwest university
- Xi'an
- 710069 P.R. China
| | - Yilong Wang
- The Institute for Translational Nanomedicine
- Shanghai East Hospital
- the Institute for Biomedical Engineering & Nano Science
- Tongji University School of Medicine
- Shanghai 200092
| |
Collapse
|
24
|
Zhang Y, Zheng H, Zhan Y, Long M, Liu S, Lu J, Zang H, Fan S. Detection and application of circulating tumor cell and circulating tumor DNA in the non-small cell lung cancer. Am J Cancer Res 2018; 8:2377-2386. [PMID: 30662798 PMCID: PMC6325475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/02/2018] [Indexed: 06/09/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death in both men and women. The ability of cancer cells to break-off from the primary tumor and spread to distant organs is the main cause of death of cancer patients. The detection of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) is a considerable part of liquid biopsy, which contributes to the diagnosis, treatment and prognosis, and especially to identify the targetable mutations of NSCLC. This review is to discuss the detection and application of CTC and ctDNA in the diagnosis, prognostic evaluation and guiding targeted therapy of NSCLC.
Collapse
Affiliation(s)
- Yuting Zhang
- Department of Pathology, The Second Xiangya Hospital, Central South University Changsha, Hunan, China
| | - Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University Changsha, Hunan, China
| | - Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University Changsha, Hunan, China
| | - Mengping Long
- Department of Pathology, The Second Xiangya Hospital, Central South University Changsha, Hunan, China
| | - Sile Liu
- Department of Pathology, The Second Xiangya Hospital, Central South University Changsha, Hunan, China
| | - Junmi Lu
- Department of Pathology, The Second Xiangya Hospital, Central South University Changsha, Hunan, China
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital, Central South University Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University Changsha, Hunan, China
| |
Collapse
|
25
|
Competitive electrochemical platform for ultrasensitive cytosensing of liver cancer cells by using nanotetrahedra structure with rolling circle amplification. Biosens Bioelectron 2018; 120:8-14. [PMID: 30142479 DOI: 10.1016/j.bios.2018.08.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/29/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023]
Abstract
In this work, a competitive and label-free electrochemical platform was performed for the ultrasensitive cytosensing of liver cancer cells based on DNA nanotetrahedron (NTH) structure and rolling circle amplification (RCA) directed DNAzyme strategy. The multifunctional nanoprobes were fabricated through a DNA primer probe, carboxyfluorescein (FAM) functionalized TLS11a aptamer and horseradish peroxidase (HRP) immobilized on the surfaces of the platinum nanoparticles (PtNPs). Then the NTH-based complementary DNA (cDNA) probe, complementary to the TLS11a aptamer, was attached on a disposable screen-printed gold electrode (SPGE) for increasing the reactivity and accessibility with the prepared nanoprobes. Due to the primer probe and the circular probe with G-quadruplex sequences for RCA, it can lead to the formation of numerous G-quadruplex/hemin DNAzyme, thus generating a remarkable electrochemical response. When the target cells were present, the nanoprobes were released from the SPGE due to the specific recognition of TLS11a aptamers for HepG2 cells, resulting in the electrochemical signal changes. The cytosensor was ultrasensitive for HepG2 tumor cell detection with a detection limit of 3 cell per mL. Furthermore, this strategy was also demonstrated to be applicable for cancer cell imaging. In summary, this electrochemical cytosensor holds great potential for circulating tumor cell detection in the early cancer diagnose.
Collapse
|
26
|
Fan M, Wang F, Wang C. Reflux Precipitation Polymerization: A New Platform for the Preparation of Uniform Polymeric Nanogels for Biomedical Applications. Macromol Biosci 2018; 18:e1800077. [DOI: 10.1002/mabi.201800077] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/19/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Mingliang Fan
- State Key Laboratory of Molecular Engineering of Polymers; Department of Macromolecular Science; Fudan University; 220 Han Dan Road Shanghai 200433 China
| | - Fang Wang
- State Key Laboratory of Molecular Engineering of Polymers; Department of Macromolecular Science; Fudan University; 220 Han Dan Road Shanghai 200433 China
| | - Changchun Wang
- State Key Laboratory of Molecular Engineering of Polymers; Department of Macromolecular Science; Fudan University; 220 Han Dan Road Shanghai 200433 China
| |
Collapse
|