1
|
Moorthy DN, Dhinasekaran D, Rebecca PNB, Rajendran AR. Optical Biosensors for Detection of Cancer Biomarkers: Current and Future Perspectives. JOURNAL OF BIOPHOTONICS 2024:e202400243. [PMID: 39442779 DOI: 10.1002/jbio.202400243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/22/2024] [Accepted: 08/19/2024] [Indexed: 10/25/2024]
Abstract
Optical biosensors are emerging as a promising technique for the sensitive and accurate detection of cancer biomarkers, enabling significant advancements in the field of early diagnosis. This study elaborates on the latest developments in optical biosensors designed for detecting cancer biomarkers, highlighting their vital significance in early cancer diagnosis. When combined with targeted nanoparticles, the bio-fluids can help in the molecular stage diagnosis of cancer. This enhances the discrimination of disease from the normal subjects drastically. The optical sensor methods that are involved in the disease diagnosis and imaging of cancer taken for the present review are surface plasmon resonance, localized surface plasmon resonance, fluorescence resonance energy transfer, surface-enhanced Raman spectroscopy and colorimetric sensing. The article meticulously describes the specific biomarkers and analytes that optical biosensors target. Beyond elucidating the underlying principles and applications, this article furnishes an overview of recent breakthroughs and emerging trends in the field. This encompasses the evolution of innovative nanomaterials and nanostructures designed to augment sensitivity and the incorporation of microfluidics for facilitating point-of-care testing, thereby charting a course towards prospective advancements.
Collapse
Affiliation(s)
| | | | - P N Blessy Rebecca
- Functional Nano-Materials (FuN) Laboratory, Department of Physics and Nanotechnology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ajay Rakkesh Rajendran
- Functional Nano-Materials (FuN) Laboratory, Department of Physics and Nanotechnology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
2
|
Liu X, Zhang W, Gu J, Wang J, Wang Y, Xu Z. Single-cell SERS imaging of dual cell membrane receptors expression influenced by extracellular matrix stiffness. J Colloid Interface Sci 2024; 668:335-342. [PMID: 38678888 DOI: 10.1016/j.jcis.2024.04.170] [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: 02/09/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Membrane receptors perform a diverse range of cellular functions, accounting for more than half of all drug targets. The mechanical microenvironment regulates cell behaviors and phenotype. However, conventional analysis methods of membrane receptors often ignore the effects of the extracellular matrix stiffness, failing to reveal the heterogeneity of cell membrane receptors expression. Herein, we developed an in-situ surface-enhanced Raman scattering (SERS) imaging method to visualize single-cell membrane receptors on substrates with different stiffness. Two SERS substrates, Au@4-mercaptobenzonitrile@Ag@Sgc8c and Au@4-pethynylaniline@Ag@SYL3c, were employed to specifically target protein tyrosine kinase-7 (PTK7) and epithelial cell adhesion molecule (EpCAM), respectively. The polyacrylamide (PA) gels with tunable stiffness (2.5-25 kPa) were constructed to mimic extracellular matrix. The simultaneous SERS imaging of dual membrane receptors on single cancer cells on substrates with different stiffness was achieved. Our findings reveal decreased expression of PTK7 and EpCAM on cells cultured on stiffer substrates and higher migration ability of the cells. The results elucidate the heterogeneity of membrane receptors expression of cells cultured on the substrates with different stiffness. This single-cell analysis method offers an in-situ platform for investigating the impacts of extracellular matrix stiffness on the expression of membrane receptors, providing insights into the role of cell membrane receptors in cancer metastasis.
Collapse
Affiliation(s)
- Xiaopeng Liu
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Wenshu Zhang
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Jiahui Gu
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Jie Wang
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Yue Wang
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China.
| |
Collapse
|
3
|
Verdin A, Malherbe C, Eppe G. Designing SERS nanotags for profiling overexpressed surface markers on single cancer cells: A review. Talanta 2024; 276:126225. [PMID: 38749157 DOI: 10.1016/j.talanta.2024.126225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 06/14/2024]
Abstract
This review focuses on the chemical design and the use of Surface-Enhanced Raman Scattering (SERS)-active nanotags for measuring surface markers that can be overexpressed at the surface of single cancer cells. Indeed, providing analytical tools with true single-cell measurements capabilities is capital, especially since cancer research is increasingly leaning toward single-cell analysis, either to guide treatment decisions or to understand complex tumor behaviour including the single-cell heterogeneity and the appearance of treatment resistance. Over the past two decades, SERS nanotags have triggered significant interest in the scientific community owing their advantages over fluorescent tags, mainly because SERS nanotags resist photobleaching and exhibit sharper signal bands, which reduces possible spectral overlap and enables the discrimination between the SERS signals and the autofluorescence background from the sample itself. The extensive efforts invested in harnessing SERS nanotags for biomedical purposes, particularly in cancer research, highlight their potential as the next generation of optical labels for single-cell studies. The review unfolds in two main parts. The first part focuses on the structure of SERS nanotags, detailing their chemical composition and the role of each building block of the tags. The second part explores applications in measuring overexpressed surface markers on single-cells. The latter encompasses studies using single nanotags, multiplexed measurements, quantitative information extraction, monitoring treatment responses, and integrating phenotype measurements with SERS nanotags on single cells isolated from complex biological matrices. This comprehensive review anticipates SERS nanotags to persist as a pivotal technology in advancing single-cell analytical methods, particularly in the context of cancer research and personalized medicine.
Collapse
Affiliation(s)
- Alexandre Verdin
- Mass Spectrometry Laboratory, MolSys Research Unit, University of Liège, Belgium.
| | - Cedric Malherbe
- Mass Spectrometry Laboratory, MolSys Research Unit, University of Liège, Belgium
| | - Gauthier Eppe
- Mass Spectrometry Laboratory, MolSys Research Unit, University of Liège, Belgium
| |
Collapse
|
4
|
Lyu N, Hassanzadeh-Barforoushi A, Rey Gomez LM, Zhang W, Wang Y. SERS biosensors for liquid biopsy towards cancer diagnosis by detection of various circulating biomarkers: current progress and perspectives. NANO CONVERGENCE 2024; 11:22. [PMID: 38811455 PMCID: PMC11136937 DOI: 10.1186/s40580-024-00428-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/09/2024] [Indexed: 05/31/2024]
Abstract
Liquid biopsy has emerged as a promising non-invasive strategy for cancer diagnosis, enabling the detection of various circulating biomarkers, including circulating tumor cells (CTCs), circulating tumor nucleic acids (ctNAs), circulating tumor-derived small extracellular vesicles (sEVs), and circulating proteins. Surface-enhanced Raman scattering (SERS) biosensors have revolutionized liquid biopsy by offering sensitive and specific detection methodologies for these biomarkers. This review comprehensively examines the application of SERS-based biosensors for identification and analysis of various circulating biomarkers including CTCs, ctNAs, sEVs and proteins in liquid biopsy for cancer diagnosis. The discussion encompasses a diverse range of SERS biosensor platforms, including label-free SERS assay, magnetic bead-based SERS assay, microfluidic device-based SERS system, and paper-based SERS assay, each demonstrating unique capabilities in enhancing the sensitivity and specificity for detection of liquid biopsy cancer biomarkers. This review critically assesses the strengths, limitations, and future directions of SERS biosensors in liquid biopsy for cancer diagnosis.
Collapse
Affiliation(s)
- Nana Lyu
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | | | - Laura M Rey Gomez
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Wei Zhang
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Yuling Wang
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
5
|
Wang C, Weng G, Li J, Zhu J, Zhao J. A review of SERS coupled microfluidic platforms: From configurations to applications. Anal Chim Acta 2024; 1296:342291. [PMID: 38401925 DOI: 10.1016/j.aca.2024.342291] [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: 09/03/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/26/2024]
Abstract
Microfluidic systems have attracted considerable attention due to their low reagent consumption, short analysis time, and ease of integration in comparison to conventional methods, but still suffer from shortcomings in sensitivity and selectivity. Surface enhanced Raman scattering (SERS) offers several advantages in the detection of compounds, including label-free detection at the single-molecule level, and the narrow Raman peak width for multiplexing. Combining microfluidics with SERS is a viable way to improve their detection sensitivity. Researchers have recently developed several SERS coupled microfluidic platforms with substantial potential for biomolecular detection, cellular and bacterial analysis, and hazardous substance detection. We review the current development of SERS coupled microfluidic platforms, illustrate their detection principles and construction, and summarize the latest applications in biology, environmental protection and food safety. In addition, we innovatively summarize the current status of SERS coupled multi-mode microfluidic platforms with other detection technologies. Finally, we discuss the challenges and countermeasures during the development of SERS coupled microfluidic platforms, as well as predict the future development trend of SERS coupled microfluidic platforms.
Collapse
Affiliation(s)
- Chenyang Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, No. 28 Xianning West Road, Xi'an 710049, China
| | - Guojun Weng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, No. 28 Xianning West Road, Xi'an 710049, China.
| | - Jianjun Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, No. 28 Xianning West Road, Xi'an 710049, China
| | - Jian Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, No. 28 Xianning West Road, Xi'an 710049, China
| | - Junwu Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, No. 28 Xianning West Road, Xi'an 710049, China.
| |
Collapse
|
6
|
Li J, Qi Y, Yang R, Chen X, Chen Z, Zhu JJ. Intellectualized Visualization of Single-Particle Raman Spectra for Sensitive Detection and Simultaneous Multianalysis of Heavy Metal Ions. Anal Chem 2023; 95:14736-14745. [PMID: 37737121 DOI: 10.1021/acs.analchem.3c02851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Easy-to-use, reliable, and real-time methods for detecting heavy metal ion contamination are urgently required, which is a primary concern for water pollution control and human health. However, present methods for this aim are still unable to achieve simultaneous multianalysis for complex real sample detection. Herein, an intellectualized vision-based single-nanoparticle Raman imaging strategy combined with ion-responsive functional nucleic acids (FNAs) was proposed to address these issues. We reported a correspondence between the concentration of the analytes and the density of particles (DOP) of specifically captured nanoparticles to achieve sensitive detection and simultaneous multianalysis of heavy metal ions. The specific detection of Pb2+ (Hg2+) was obtained with a detection linear range from 100 pM to 100 nM (from 500 fM to 100 nM) and limit of detections low to 1 pM (100 fM), with the advantages of good specificity, excellent homogeneity, and reproducibility. Furthermore, the differentiation of different heavy metal ions (Pb2+/Hg2+) was achieved, i.e., the simultaneous multianalysis, based on Raman imaging of the single particle and intelligent machine vision method. Finally, the Raman imaging assay was utilized for real sample analysis, and it provided a powerful and reliable tool for detecting trace Pb2+/Hg2+ in real water samples and facilitated the portable on-site monitoring of heavy metal ions.
Collapse
Affiliation(s)
- Jinxiang Li
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yongbing Qi
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, People's Republic of China
| | - Ruixin Yang
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, People's Republic of China
| | - Xueqin Chen
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, People's Republic of China
| | - Zixuan Chen
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, People's Republic of China
| | - Jun-Jie Zhu
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, People's Republic of China
- Shenzhen Research Institute of Nanjing University, Shenzhen 518000, People's Republic of China
| |
Collapse
|
7
|
Cutshaw G, Uthaman S, Hassan N, Kothadiya S, Wen X, Bardhan R. The Emerging Role of Raman Spectroscopy as an Omics Approach for Metabolic Profiling and Biomarker Detection toward Precision Medicine. Chem Rev 2023; 123:8297-8346. [PMID: 37318957 PMCID: PMC10626597 DOI: 10.1021/acs.chemrev.2c00897] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Omics technologies have rapidly evolved with the unprecedented potential to shape precision medicine. Novel omics approaches are imperative toallow rapid and accurate data collection and integration with clinical information and enable a new era of healthcare. In this comprehensive review, we highlight the utility of Raman spectroscopy (RS) as an emerging omics technology for clinically relevant applications using clinically significant samples and models. We discuss the use of RS both as a label-free approach for probing the intrinsic metabolites of biological materials, and as a labeled approach where signal from Raman reporters conjugated to nanoparticles (NPs) serve as an indirect measure for tracking protein biomarkers in vivo and for high throughout proteomics. We summarize the use of machine learning algorithms for processing RS data to allow accurate detection and evaluation of treatment response specifically focusing on cancer, cardiac, gastrointestinal, and neurodegenerative diseases. We also highlight the integration of RS with established omics approaches for holistic diagnostic information. Further, we elaborate on metal-free NPs that leverage the biological Raman-silent region overcoming the challenges of traditional metal NPs. We conclude the review with an outlook on future directions that will ultimately allow the adaptation of RS as a clinical approach and revolutionize precision medicine.
Collapse
Affiliation(s)
- Gabriel Cutshaw
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50012, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| | - Saji Uthaman
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50012, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| | - Nora Hassan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50012, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| | - Siddhant Kothadiya
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50012, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| | - Xiaona Wen
- Biologics Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50012, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| |
Collapse
|
8
|
Surappa S, Multani P, Parlatan U, Sinawang PD, Kaifi J, Akin D, Demirci U. Integrated "lab-on-a-chip" microfluidic systems for isolation, enrichment, and analysis of cancer biomarkers. LAB ON A CHIP 2023; 23:2942-2958. [PMID: 37314731 PMCID: PMC10834032 DOI: 10.1039/d2lc01076c] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The liquid biopsy has garnered considerable attention as a complementary clinical tool for the early detection, molecular characterization and monitoring of cancer over the past decade. In contrast to traditional solid biopsy techniques, liquid biopsy offers a less invasive and safer alternative for routine cancer screening. Recent advances in microfluidic technologies have enabled handling of liquid biopsy-derived biomarkers with high sensitivity, throughput, and convenience. The integration of these multi-functional microfluidic technologies into a 'lab-on-a-chip' offers a powerful solution for processing and analyzing samples on a single platform, thereby reducing the complexity, bio-analyte loss and cross-contamination associated with multiple handling and transfer steps in more conventional benchtop workflows. This review critically addresses recent developments in integrated microfluidic technologies for cancer detection, highlighting isolation, enrichment, and analysis strategies for three important sub-types of cancer biomarkers: circulating tumor cells, circulating tumor DNA and exosomes. We first discuss the unique characteristics and advantages of the various lab-on-a-chip technologies developed to operate on each biomarker subtype. This is then followed by a discussion on the challenges and opportunities in the field of integrated systems for cancer detection. Ultimately, integrated microfluidic platforms form the core of a new class of point-of-care diagnostic tools by virtue of their ease-of-operation, portability and high sensitivity. Widespread availability of such tools could potentially result in more frequent and convenient screening for early signs of cancer at clinical labs or primary care offices.
Collapse
Affiliation(s)
- Sushruta Surappa
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| | - Priyanka Multani
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| | - Ugur Parlatan
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| | - Prima Dewi Sinawang
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Jussuf Kaifi
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| | - Demir Akin
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
- Center for Cancer Nanotechnology Excellence for Translational Diagnostics (CCNE-TD), School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| |
Collapse
|
9
|
Ilyas A, Dyussupova A, Sultangaziyev A, Shevchenko Y, Filchakova O, Bukasov R. SERS immuno- and apta-assays in biosensing/bio-detection: Performance comparison, clinical applications, challenges. Talanta 2023; 265:124818. [PMID: 37453393 DOI: 10.1016/j.talanta.2023.124818] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
Surface Enhanced Raman Spectroscopy is increasingly used as a sensitive bioanalytical tool for detection of variety of analytes ranging from viruses and bacteria to cancer biomarkers and toxins, etc. This comprehensive review describes principles of operation and compares the performance of immunoassays and aptamer assays with Surface Enhanced Raman scattering (SERS) detection to each other and to some other bioassay methods, including ELISA and fluorescence assays. Both immuno- and aptamer-based assays are categorized into assay on solid substrates, assays with magnetic nanoparticles and assays in laminar flow or/and strip assays. The best performing and recent examples of assays in each category are described in the text and illustrated in the figures. The average performance, particularly, limit of detection (LOD) for each of those methods reflected in 9 tables of the manuscript and average LODs are calculated and compared. We found out that, on average, there is some advantage in terms of LOD for SERS immunoassays (0.5 pM median LOD of 88 papers) vs SERS aptamer-based assays (1.7 pM median LOD of 51 papers). We also tabulated and analyzed the clinical performance of SERS immune and aptamer assays, where selectivity, specificity, and accuracy are reported, we summarized the best examples. We also reviewed challenges to SERS bioassay performance and real-life application, including non-specific protein binding, nanoparticle aggregation, limited nanotag stability, sometimes, relatively long time to results, etc. The proposed solutions to those challenges are also discussed in the review. Overall, this review may be interesting not only to bioanalytical chemist, but to medical and life science researchers who are interested in improvement of bioanalyte detection and diagnostics.
Collapse
Affiliation(s)
- Aisha Ilyas
- Department of Chemistry, SSH, Nazarbayev University, Astana, Kazakhstan
| | | | | | - Yegor Shevchenko
- Department of Chemistry, SSH, Nazarbayev University, Astana, Kazakhstan
| | - Olena Filchakova
- Department of Biology, SSH, Nazarbayev University, Astana, Kazakhstan
| | - Rostislav Bukasov
- Department of Chemistry, SSH, Nazarbayev University, Astana, Kazakhstan.
| |
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
|
Wei J, Zhang S, Yuan J, Wang Z, Zong S, Cui Y. Nanoscale imaging of tumor cell exosomes by expansion single molecule localization microscopy (ExSMLM). Talanta 2023; 261:124641. [PMID: 37187025 DOI: 10.1016/j.talanta.2023.124641] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/17/2023]
Abstract
Tumor cell exosomes play a very important role in the process of tumor cell proliferation and metastasis. However, due to the nanoscale size and high heterogeneity of exosomes, in-depth understanding of their appearance and biological characteristics is still lacking. Expansion microscopy (ExM) is a method that embeds biological samples in a swellable gel to physically magnify the samples to improve the imaging resolution. Before the emergence of ExM, scientists had invented several super-resolution imaging techniques that could break the diffraction limit. Among them, single molecule localization microscopy (SMLM) usually has the best spatial resolution (20-50 nm). However, considering the small size of exosomes (30-150 nm), the resolution of SMLM is still not high enough for detailed imaging of exosomes. Hence, we propose a tumor cell exosomes imaging method that combines ExM and SMLM (i.e. Expansion SMLM, denoted as ExSMLM), which can realize the expansion and super-resolution imaging of tumor cell exosomes. In this technique, immunofluorescence was first performed to fluorescently label the protein markers on the exosomes, then the exosomes were polymerized into a swellable polyelectrolyte gel. The electrolytic nature of the gel made the fluorescently labeled exosomes undergo isotropic linear physical expansion. The expansion factor obtained in the experiment was about 4.6. Finally, SMLM imaging of the expanded exosomes was performed. Owing to the improved resolution of ExSMLM, nanoscale substructures of closely packed proteins were observed on single exosomes, which has never been achieved before. With such a high resolution, ExSMLM would have a great potential in detailed investigation of exosomes and exosome-related biological processes.
Collapse
Affiliation(s)
- Jinxiu Wei
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Siyao Zhang
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Jiangnan Yuan
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Zhuyuan Wang
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Shenfei Zong
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China.
| | - Yiping Cui
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China.
| |
Collapse
|
12
|
Clack K, Soda N, Kasetsirikul S, Mahmudunnabi RG, Nguyen NT, Shiddiky MJA. Toward Personalized Nanomedicine: The Critical Evaluation of Micro and Nanodevices for Cancer Biomarker Analysis in Liquid Biopsy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205856. [PMID: 36631277 DOI: 10.1002/smll.202205856] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/20/2022] [Indexed: 06/17/2023]
Abstract
Liquid biopsy for the analysis of circulating cancer biomarkers (CBs) is a major advancement toward the early detection of cancer. In comparison to tissue biopsy techniques, liquid biopsy is relatively painless, offering multiple sampling opportunities across easily accessible bodily fluids such as blood, urine, and saliva. Liquid biopsy is also relatively inexpensive and simple, avoiding the requirement for specialized laboratory equipment or trained medical staff. Major advances in the field of liquid biopsy are attributed largely to developments in nanotechnology and microfabrication that enables the creation of highly precise chip-based platforms. These devices can overcome detection limitations of an individual biomarker by detecting multiple markers simultaneously on the same chip, or by featuring integrated and combined target separation techniques. In this review, the major advances in the field of portable and semi-portable micro, nano, and multiplexed platforms for CB detection for the early diagnosis of cancer are highlighted. A comparative discussion is also provided, noting merits and drawbacks of the platforms, especially in terms of portability. Finally, key challenges toward device portability and possible solutions, as well as discussing the future direction of the field are highlighted.
Collapse
Affiliation(s)
- Kimberley Clack
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Narshone Soda
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Surasak Kasetsirikul
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Rabbee G Mahmudunnabi
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Nam-Trung Nguyen
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Muhammad J A Shiddiky
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| |
Collapse
|
13
|
Saikia A, Newar R, Das S, Singh A, Deuri DJ, Baruah A. Scopes and Challenges of Microfluidic Technology for Nanoparticle Synthesis, Photocatalysis and Sensor Applications: A Comprehensive Review. Chem Eng Res Des 2023. [DOI: 10.1016/j.cherd.2023.03.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
14
|
Li J, Dong C, Gan H, Gu X, Zhang J, Zhu Y, Xiong J, Song C, Wang L. Nondestructive separation/enrichment and rolling circle amplification-powered sensitive SERS enumeration of circulating tumor cells via aptamer recognition. Biosens Bioelectron 2023; 231:115273. [PMID: 37054599 DOI: 10.1016/j.bios.2023.115273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Nondestructive separation/enrichment and reliable detection of extremely rare circulating tumor cells (CTCs) in peripheral blood are of considerable importance in tumor precision diagnosis and treatment, yet this remains a big challenge. Herein, a novel strategy for nondestructive separation/enrichment and ultra-sensitive surface-enhanced Raman scattering (SERS)-based enumeration of CTCs is proposed via aptamer recognition and rolling circle amplification (RCA). In this work the magnetic beads modified with "Aptamer (Apt)-Primer" (AP) probes were utilized to specifically capture CTCs, and then after magnetic separation/enrichment, the RCA-powered SERS counting and benzonase nuclease cleavage-assisted nondestructive release of CTCs were realized, respectively. The AP was assembled by hybridizing the EpCAM-specific aptamer with a primer, and the optimal AP contains 4 mismatched bases. The RCA enhanced SERS signal nearly 4.5-fold, and the SERS strategy has good specificity, uniformity and reproducibility. The proposed SERS detection possesses a good linear relationship with the concentration of MCF-7 cells spiked in PBS with the limit of detection (LOD) of 2 cells/mL, which shows good potential practicality for detecting CTCs in blood with recoveries ranging from 100.56% to 116.78%. Besides, the released CTCs remained good cellular activity with the normal proliferation after re-culture for 48 h and normal growth for at least three generations. The proposed strategy of nondestructive separation/enrichment and SERS-based sensitive enumeration is promising for reliable analysis of EpCAM-positive CTCs in blood, which is expected to provide a powerful tool for analysis of extremely rare circulating tumor cells in complex peripheral blood for liquid biopsy.
Collapse
|
15
|
Su X, Liu X, Xie Y, Chen M, Zheng C, Zhong H, Li M. Integrated SERS-Vertical Flow Biosensor Enabling Multiplexed Quantitative Profiling of Serological Exosomal Proteins in Patients for Accurate Breast Cancer Subtyping. ACS NANO 2023; 17:4077-4088. [PMID: 36758150 DOI: 10.1021/acsnano.3c00449] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Protein profiles of exosomes (EXOs) in clinical samples of cancer patients have become a promising diagnostic and therapeutic biomarker. However, simultaneous quantitative analysis of multiple exosomal proteins of interest remains challenging. To address the unmet need, we develop a paper-based surface-enhanced Raman spectroscopy (SERS)-vertical flow biosensor, named iREX (integrated Raman spectroscopic EXO) biosensor, for multiplexed quantitative profiling of exosomal proteins in clinical serum samples of patients. Utilizing this iREX biosensor, we are able to quantitatively profile MUC1, HER2 and CEA in EXO samples derived from various breast cancer cell subtypes. The results show discriminative expression profiles of the three exosomal proteins in these cell subtypes, which allows for accurate diagnosis and molecular subtyping of breast cancer. We further validate the clinical utility of the iREX biosensor for simultaneous quantitative analysis of MUC1, HER2 and CEA in patient's blood serums, thereby aiding in noninvasive breast cancer subtyping and longitudinal treatment monitoring. Our iREX biosensor integrating the SERS detection in a vertical flow diagnostic device offers great advantages of high sensitivity, molecular specificity, powerful multiplexing capability, and high diagnostic accuracy. We believe that the iREX biosensor could be a promising clinical tool for comprehensive analysis of exosomal proteins in clinical samples for personalized diagnosis and precise management of breast cancer.
Collapse
Affiliation(s)
- Xiaoming Su
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China
| | - Xinyu Liu
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Yangcenzi Xie
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Mingyang Chen
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Chao Zheng
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Hong Zhong
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
16
|
Awiaz G, Lin J, Wu A. Recent advances of Au@Ag core-shell SERS-based biosensors. EXPLORATION (BEIJING, CHINA) 2023; 3:20220072. [PMID: 37323623 PMCID: PMC10190953 DOI: 10.1002/exp.20220072] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/18/2022] [Indexed: 06/17/2023]
Abstract
The methodological advancements in surface-enhanced Raman scattering (SERS) technique with nanoscale materials based on noble metals, Au, Ag, and their bimetallic alloy Au-Ag, has enabled the highly efficient sensing of chemical and biological molecules at very low concentration values. By employing the innovative various type of Au, Ag nanoparticles and especially, high efficiency Au@Ag alloy nanomaterials as substrate in SERS based biosensors have revolutionized the detection of biological components including; proteins, antigens antibodies complex, circulating tumor cells, DNA, and RNA (miRNA), etc. This review is about SERS-based Au/Ag bimetallic biosensors and their Raman enhanced activity by focusing on different factors related to them. The emphasis of this research is to describe the recent developments in this field and conceptual advancements behind them. Furthermore, in this article we apex the understanding of impact by variation in basic features like effects of size, shape varying lengths, thickness of core-shell and their influence of large-scale magnitude and morphology. Moreover, the detailed information about recent biological applications based on these core-shell noble metals, importantly detection of receptor binding domain (RBD) protein of COVID-19 is provided.
Collapse
Affiliation(s)
- Gul Awiaz
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical MaterialsNingbo Institute of Materials Technology and Engineering, CASNingboChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jie Lin
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical MaterialsNingbo Institute of Materials Technology and Engineering, CASNingboChina
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhouChina
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical MaterialsNingbo Institute of Materials Technology and Engineering, CASNingboChina
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhouChina
| |
Collapse
|
17
|
Isolation, Detection and Analysis of Circulating Tumour Cells: A Nanotechnological Bioscope. Pharmaceutics 2023; 15:pharmaceutics15010280. [PMID: 36678908 PMCID: PMC9864919 DOI: 10.3390/pharmaceutics15010280] [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: 12/17/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Cancer is one of the dreaded diseases to which a sizeable proportion of the population succumbs every year. Despite the tremendous growth of the health sector, spanning diagnostics to treatment, early diagnosis is still in its infancy. In this regard, circulating tumour cells (CTCs) have of late grabbed the attention of researchers in the detection of metastasis and there has been a huge surge in the surrounding research activities. Acting as a biomarker, CTCs prove beneficial in a variety of aspects. Nanomaterial-based strategies have been devised to have a tremendous impact on the early and rapid examination of tumor cells. This review provides a panoramic overview of the different nanotechnological methodologies employed along with the pharmaceutical purview of cancer. Initiating from fundamentals, the recent nanotechnological developments toward the detection, isolation, and analysis of CTCs are comprehensively delineated. The review also includes state-of-the-art implementations of nanotechnological advances in the enumeration of CTCs, along with future challenges and recommendations thereof.
Collapse
|
18
|
Gharib G, Bütün İ, Muganlı Z, Kozalak G, Namlı İ, Sarraf SS, Ahmadi VE, Toyran E, van Wijnen AJ, Koşar A. Biomedical Applications of Microfluidic Devices: A Review. BIOSENSORS 2022; 12:1023. [PMID: 36421141 PMCID: PMC9688231 DOI: 10.3390/bios12111023] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/30/2022] [Accepted: 11/08/2022] [Indexed: 05/26/2023]
Abstract
Both passive and active microfluidic chips are used in many biomedical and chemical applications to support fluid mixing, particle manipulations, and signal detection. Passive microfluidic devices are geometry-dependent, and their uses are rather limited. Active microfluidic devices include sensors or detectors that transduce chemical, biological, and physical changes into electrical or optical signals. Also, they are transduction devices that detect biological and chemical changes in biomedical applications, and they are highly versatile microfluidic tools for disease diagnosis and organ modeling. This review provides a comprehensive overview of the significant advances that have been made in the development of microfluidics devices. We will discuss the function of microfluidic devices as micromixers or as sorters of cells and substances (e.g., microfiltration, flow or displacement, and trapping). Microfluidic devices are fabricated using a range of techniques, including molding, etching, three-dimensional printing, and nanofabrication. Their broad utility lies in the detection of diagnostic biomarkers and organ-on-chip approaches that permit disease modeling in cancer, as well as uses in neurological, cardiovascular, hepatic, and pulmonary diseases. Biosensor applications allow for point-of-care testing, using assays based on enzymes, nanozymes, antibodies, or nucleic acids (DNA or RNA). An anticipated development in the field includes the optimization of techniques for the fabrication of microfluidic devices using biocompatible materials. These developments will increase biomedical versatility, reduce diagnostic costs, and accelerate diagnosis time of microfluidics technology.
Collapse
Affiliation(s)
- Ghazaleh Gharib
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
- Sabanci University Nanotechnology Research and Application Centre (SUNUM), Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - İsmail Bütün
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
| | - Zülâl Muganlı
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
| | - Gül Kozalak
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - İlayda Namlı
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
| | | | | | - Erçil Toyran
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
| | - Andre J. van Wijnen
- Department of Biochemistry, University of Vermont, 89 Beaumont Avenue, Burlington, VT 05405, USA
| | - Ali Koşar
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
- Sabanci University Nanotechnology Research and Application Centre (SUNUM), Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
19
|
Li J, Wuethrich A, Zhang Z, Wang J, Lin LL, Behren A, Wang Y, Trau M. SERS Multiplex Profiling of Melanoma Circulating Tumor Cells for Predicting the Response to Immune Checkpoint Blockade Therapy. Anal Chem 2022; 94:14573-14582. [PMID: 36222247 DOI: 10.1021/acs.analchem.2c02398] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Immune checkpoint blockade (ICB) therapy has achieved remarkable success in many cancers including melanoma. However, ICB therapy benefits only a small proportion of patients and produces severe side effects for some patients. Thus, there is an urgent need to identify patients who are more likely to respond to ICB therapy to improve outcomes and minimize side effects. To predict ICB therapy responses, we design a surface-enhanced Raman scattering (SERS) assay for multiplex profiling of circulating tumor cells (CTCs) under basal and interferon-γ (IFN-γ) stimulation. Through simultaneous ensemble and single-cell measurements of CTCs, the SERS assay can reveal tumor heterogeneity and offer a comprehensive CTC phenotype for decision-making. Anisotropic gold-silver alloy nanoboxes are utilized as SERS plasmonic substrates for improved signal readouts of CTC surface biomarkers. By generating a unique CTC signature with four surface biomarkers, the developed assay enables the differentiation of CTCs from three different patient-derived melanoma cell lines. Significantly, in a cohort of 14 melanoma patients who received programmed cell death-1 blockade therapy, the changes of CTC signature induced by IFN-γ stimulation to CTCs show the potential to predict responders. We expect that the SERS assay can help select patients for receiving ICB therapy in other cancers.
Collapse
Affiliation(s)
- Junrong Li
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan430079, P. R. China.,Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD4072, Australia
| | - Alain Wuethrich
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD4072, Australia
| | - Zhen Zhang
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD4072, Australia
| | - Jing Wang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou350007, P. R. China
| | - Lynlee L Lin
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD4102, Australia
| | - Andreas Behren
- Oliva Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, VIC3086, Australia.,Department of Medicine, University of Melbourne, Heidelberg, VIC3010, Australia
| | - Yuling Wang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW2109, Australia
| | - Matt Trau
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD4072, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD4072, Australia
| |
Collapse
|
20
|
Chen Z, Yin G, Wei J, Qi T, Qian Z, Wang Z, Zong S, Cui Y. Quantitative analysis of multiple breast cancer biomarkers using DNA-PAINT. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:3671-3679. [PMID: 36063064 DOI: 10.1039/d2ay00670g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Immunotherapy has become an efficient treatment method of breast cancer. Detection of proteins such as PD-L1 and CTLA-4, which are important immune checkpoint molecules, is attracting more and more attention as they play key roles in immunotherapy. Here, by combining the high resolution of DNA-PAINT (DNA points accumulation for imaging in nanoscale topography) with the qPAINT quantitative analysis method, accurate spatial localization and absolute quantification of PD-L1 and CTLA-4 on the membrane of breast cancer cells could be achieved. Meanwhile, exchange-PAINT was also conducted to count three other biomarkers (EpCAM, EGFR, and HER2). Simultaneous analysis of these biomarkers can greatly facilitate the differentiation of different kinds of breast cancer. Such a simple quantitative analysis method holds great potential in diagnosis and immunotherapy of cancers.
Collapse
Affiliation(s)
- Zengwei Chen
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Gaoqiang Yin
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Jinxiu Wei
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Tongsheng Qi
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Ziting Qian
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Zhuyuan Wang
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Shenfei Zong
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Yiping Cui
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| |
Collapse
|
21
|
Lenzi E, Henriksen-Lacey M, Molina B, Langer J, de Albuquerque CDL, Jimenez de Aberasturi D, Liz-Marzán LM. Combination of Live Cell Surface-Enhanced Raman Scattering Imaging with Chemometrics to Study Intracellular Nanoparticle Dynamics. ACS Sens 2022; 7:1747-1756. [PMID: 35671439 PMCID: PMC9237835 DOI: 10.1021/acssensors.2c00610] [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] [Indexed: 01/18/2023]
Abstract
Surface-enhanced Raman scattering (SERS)-encoded nanoparticles are used for bioimaging, on account of their well-defined Raman spectra and biocompatibility, which allow long incubation times with high signal stability and no cytotoxicity. However, reliable analysis of SERS bioimaging requires quantification of the amount of encoded nanoparticles that have been taken up by cells and the effect of subsequent dilution due to cellular division (mitosis). Although methods such as elemental analysis and flow cytometry can be used to quantify nanoparticle uptake, these are both end-point measurements in which a cell population is screened rather than looking at individual cells. In contrast, SERS imaging can be applied at multiple timepoints to the same individual cells without damaging the biological sample. We present the application of both supervised and unsupervised multivariate analyses, to quantify the intracellular amount of SERS tags in individual MCF7 living cells, toward the characterization of cellular uptake in vitro. The obtained results from both methodologies were validated by standard elemental analysis techniques.
Collapse
Affiliation(s)
- Elisa Lenzi
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain.,Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
| | - Malou Henriksen-Lacey
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain.,Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
| | - Beatriz Molina
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain
| | - Judith Langer
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain.,Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
| | - Carlos D L de Albuquerque
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain.,Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
| | - Dorleta Jimenez de Aberasturi
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain.,Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain.,Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Luis M Liz-Marzán
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain.,Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain.,Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
22
|
Xu X, Zhang M, Zhang X, Liu Y, Cai L, Zhang Q, Chen Q, Lin L, Lin S, Song Y, Zhu Z, Yang C. Decoding Expression Dynamics of Protein and Transcriptome at the Single-Cell Level in Paired Picoliter Chambers. Anal Chem 2022; 94:8164-8173. [PMID: 35650660 DOI: 10.1021/acs.analchem.1c05312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Simultaneous analysis of mRNAs and proteins at the single-cell level provides information about the dynamics and correlations of gene and protein expressions in individual cells, enabling a comprehensive study of cellular heterogeneity and expression patterns. Here, we present a platform for about 1000 cellular indexing of mRNAs and membrane proteins, named multi-Paired-seq, with high cell utilization, accurate molecular measurement, and low cost. Based on hydrodynamic differential flow resistance, multi-Paired-seq largely improves cell utilization in the percentage of cells measured in population (>95%). Combined with the pump/valve structure, cell-free antibodies and mRNAs can be removed completely for highly accurate detection (R = 0.96) of protein copies. The picoliter reaction chambers allow high detection sensitivity for both mRNA transcripts and protein copies and low sequencing cost. Using multi-Paired-seq, three clusters of known breast cancer cell types are identified according to multimodal measurements, and the expression correlations between mRNAs and proteins under altered conditions are quantified. Multi-Paired-seq provides multimodal measurements at the single-cell level, which offers a new tool for cell biology, developmental biology, drug discovery, and precision medicine.
Collapse
Affiliation(s)
- Xing Xu
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Mingxia Zhang
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China.,Suzhou Dynamic Biosystems Co., Ltd., Suzhou, Jiangsu 215000, China
| | - Xuebing Zhang
- Suzhou Dynamic Biosystems Co., Ltd., Suzhou, Jiangsu 215000, China
| | - Yilong Liu
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Linfeng Cai
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Qianqian Zhang
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Qin Chen
- Suzhou Dynamic Biosystems Co., Ltd., Suzhou, Jiangsu 215000, China
| | - Li Lin
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Shichao Lin
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Yanling Song
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Zhi Zhu
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Chaoyong Yang
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China.,Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| |
Collapse
|
23
|
Recent advances for cancer detection and treatment by microfluidic technology, review and update. Biol Proced Online 2022; 24:5. [PMID: 35484481 PMCID: PMC9052508 DOI: 10.1186/s12575-022-00166-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/18/2022] [Indexed: 12/16/2022] Open
Abstract
Numerous cancer-associated deaths are owing to a lack of effective diagnostic and therapeutic approaches. Microfluidic systems for analyzing a low volume of samples offer a precise, quick, and user-friendly technique for cancer diagnosis and treatment. Microfluidic devices can detect many cancer-diagnostic factors from biological fluids and also generate appropriate nanoparticles for drug delivery. Thus, microfluidics may be valuable in the cancer field due to its high sensitivity, high throughput, and low cost. In the present article, we aim to review recent achievements in the application of microfluidic systems for the diagnosis and treatment of various cancers. Although microfluidic platforms are not yet used in the clinic, they are expected to become the main technology for cancer diagnosis and treatment. Microfluidic systems are proving to be more sensitive and accurate for the detection of cancer biomarkers and therapeutic strategies than common assays. Microfluidic lab-on-a-chip platforms have shown remarkable potential in the designing of novel procedures for cancer detection, therapy, and disease follow-up as well as the development of new drug delivery systems for cancer treatment.
Collapse
|
24
|
Bhat MP, Thendral V, Uthappa UT, Lee KH, Kigga M, Altalhi T, Kurkuri MD, Kant K. Recent Advances in Microfluidic Platform for Physical and Immunological Detection and Capture of Circulating Tumor Cells. BIOSENSORS 2022; 12:220. [PMID: 35448280 PMCID: PMC9025399 DOI: 10.3390/bios12040220] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 05/05/2023]
Abstract
CTCs (circulating tumor cells) are well-known for their use in clinical trials for tumor diagnosis. Capturing and isolating these CTCs from whole blood samples has enormous benefits in cancer diagnosis and treatment. In general, various approaches are being used to separate malignant cells, including immunomagnets, macroscale filters, centrifuges, dielectrophoresis, and immunological approaches. These procedures, on the other hand, are time-consuming and necessitate multiple high-level operational protocols. In addition, considering their low efficiency and throughput, the processes of capturing and isolating CTCs face tremendous challenges. Meanwhile, recent advances in microfluidic devices promise unprecedented advantages for capturing and isolating CTCs with greater efficiency, sensitivity, selectivity and accuracy. In this regard, this review article focuses primarily on the various fabrication methodologies involved in microfluidic devices and techniques specifically used to capture and isolate CTCs using various physical and biological methods as well as their conceptual ideas, advantages and disadvantages.
Collapse
Affiliation(s)
- Mahesh Padmalaya Bhat
- Centre for Research in Functional Materials (CRFM), Jain Global Campus, Jain University, Bengaluru 562112, Karnataka, India; (M.P.B.); (V.T.); (M.K.)
- Agricultural Automation Research Center, Chonnam National University, Gwangju 61186, Korea;
| | - Venkatachalam Thendral
- Centre for Research in Functional Materials (CRFM), Jain Global Campus, Jain University, Bengaluru 562112, Karnataka, India; (M.P.B.); (V.T.); (M.K.)
| | | | - Kyeong-Hwan Lee
- Agricultural Automation Research Center, Chonnam National University, Gwangju 61186, Korea;
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Korea
| | - Madhuprasad Kigga
- Centre for Research in Functional Materials (CRFM), Jain Global Campus, Jain University, Bengaluru 562112, Karnataka, India; (M.P.B.); (V.T.); (M.K.)
| | - Tariq Altalhi
- Department of Chemistry, Faculty of Science, Taif University, Taif 21944, Saudi Arabia;
| | - Mahaveer D. Kurkuri
- Centre for Research in Functional Materials (CRFM), Jain Global Campus, Jain University, Bengaluru 562112, Karnataka, India; (M.P.B.); (V.T.); (M.K.)
| | - Krishna Kant
- Departamento de Química Física, Campus Universitario, CINBIO Universidade de Vigo, 36310 Vigo, Spain
| |
Collapse
|
25
|
Liu B, Zheng S, Li H, Xu J, Tang H, Wang Y, Wang Y, Sun F, Zhao X. Ultrasensitive and facile detection of multiple trace antibiotics with magnetic nanoparticles and core-shell nanostar SERS nanotags. Talanta 2022; 237:122955. [PMID: 34736680 DOI: 10.1016/j.talanta.2021.122955] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 01/13/2023]
Abstract
Ultrasensitive, multiplex, rapid, and accurate quantitative determination of trace antibiotics remains a challenging issue, which is of importance to public health and safety. Herein, we presented a multiplex strategy based on magnetic nanoparticles and surface-enhanced Raman scattering (SERS) nanotags for simultaneous detection of chloramphenicol (CAP) and tetracycline (TTC). In practice, SERS nanotags based on Raman reporter probes (RRPs) encoded gold-silver core-shell nanostars were used as detection labels for identifying different types of antibiotics, and the magnetic nanoparticles could be separated simply by magnetic force, which significantly improves the detection efficiency, reduces the analysis cost, and simplifies the operation. Our results demonstrate that the as-proposed assay possesses the capacities of high sensitivity and multiplexing with the limits of detection (LODs) for CAP and TTC of 159.49 and 294.12 fg mL-1, respectively, as well as good stability and reproducibility, and high selectivity and reliability. We believe that this strategy holds a great promising perspective for the detection of trace amounts of antibiotics in microsystems, which is crucial to our life. Additionally, the assay can also be used to detect other illegal additives by altering the appropriate antibodies or aptamers.
Collapse
Affiliation(s)
- Bing Liu
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, 226001, China.
| | - Shiya Zheng
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Haitao Li
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, 226001, China
| | - Junjie Xu
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, 226001, China
| | - Hanyu Tang
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, 226001, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou, 310058, China
| | - Yingchao Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, China
| | - Fei Sun
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, 226001, China.
| | - Xiangwei Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Southeast University Shenzhen Research Institute, Shenzhen, 518000, China.
| |
Collapse
|
26
|
Zhang J, Huang Y, Sun M, Wan S, Yang C, Song Y. Recent Advances in Aptamer-Based Liquid Biopsy. ACS APPLIED BIO MATERIALS 2022; 5:1954-1979. [PMID: 35014838 DOI: 10.1021/acsabm.1c01202] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Liquid biopsy capable of noninvasive and real-time molecular profiling is considered as a breakthrough technology, endowing an opportunity for precise diagnosis of individual patients. Extracellular vesicles (EVs) and circulating tumor cells (CTCs) consisting of substantial disease-related molecular information play an important role in liquid biopsy. Therefore, it is critically significant to exploit high-performance recognition ligands for efficient isolation and analysis of EVs and CTCs from complex body fluids. Aptamers exhibit extraordinary merits of high specificity and affinity, which are considered as superior recognition ligands for liquid biopsy. In this review, we first summarize recent advanced strategies for the evolution of high-performance aptamers and the construction of various aptamer-based recognition elements. Subsequently, we mainly discuss the isolation and analysis of EVs and CTCs based on the aptamer functioned biomaterials/biointerface. Ultimately, we envision major challenges and future direction of aptamer-based liquid biopsy for clinical utilities.
Collapse
Affiliation(s)
- Jialu Zhang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yihao Huang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Miao Sun
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shuang Wan
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, 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
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
27
|
Caballero D, Reis RL, Kundu SC. Current Trends in Microfluidics and Biosensors for Cancer Research Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:81-112. [DOI: 10.1007/978-3-031-04039-9_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
28
|
Liu X, Ma L, Yan W, Aazmi A, Fang M, Xu X, Kang H, Xu X. A review of recent progress toward the efficient separation of circulating tumor cells via micro‐/nanostructured microfluidic chips. VIEW 2022. [DOI: 10.1002/viw.20210013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Xiaoshi Liu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Liang Ma
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou P. R. China
| | - Wenyuan Yan
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou P. R. China
| | - Minghe Fang
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Xiuzhen Xu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Hanyue Kang
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Xiaobin Xu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| |
Collapse
|
29
|
Mitchell KR, Esene JE, Woolley AT. Advances in multiplex electrical and optical detection of biomarkers using microfluidic devices. Anal Bioanal Chem 2022; 414:167-180. [PMID: 34345949 PMCID: PMC8331214 DOI: 10.1007/s00216-021-03553-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023]
Abstract
Microfluidic devices can provide a versatile, cost-effective platform for disease diagnostics and risk assessment by quantifying biomarkers. In particular, simultaneous testing of several biomarkers can be powerful. Here, we critically review work from the previous 4 years up to February 2021 on developing microfluidic devices for multiplexed detection of biomarkers from samples. We focus on two principal approaches: electrical and optical detection methods that can distinguish and quantify biomarkers. Both electrical and spectroscopic multiplexed detection strategies are being employed to reach limits of detection below clinical sample levels. Some of the most promising strategies for point-of-care assays involve inexpensive materials such as paper-based microfluidic devices, or portable and accessible detectors such as smartphones. This review does not comprehensively cover all multiplexed microfluidic biomarker studies, but rather provides a critical evaluation of key work and suggests promising prospects for future advancement in this field. Electrical and optical multiplexing are powerful approaches for microfluidic biomarker analysis.
Collapse
Affiliation(s)
- Kaitlynn R Mitchell
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Joule E Esene
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Adam T Woolley
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA.
| |
Collapse
|
30
|
Lin T, Song YL, Kuang P, Chen S, Mao Z, Zeng TT. Nanostructure-based surface-enhanced Raman scattering for diagnosis of cancer. Nanomedicine (Lond) 2021; 16:2389-2406. [PMID: 34530631 DOI: 10.2217/nnm-2021-0298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cancer is a malignant disease that seriously affects human health and life. Early diagnosis and timely treatment can significantly improve the survival rate of cancer patients. Surface-enhanced Raman scattering (SERS) is an optical technology that can detect and image samples at the single-molecule level. It has the advantages of rapidity, high specificity, high sensitivity and no damage to the sample. The performance of SERS is highly dependent on the properties, size and morphology of the SERS substrate. Preparation of SERS substrates with good reproducibility and chemical stability is a key factor in realizing the wide application of SERS technology in cancer diagnosis. In this review we provide a detailed presentation of the latest research on SERS in cancer diagnosis and the detection of cancer biomarkers, mainly focusing on nanotechnological approaches in cancer diagnosis by using SERS. We also consider the future development of nanostructure-based SERS in cancer diagnosis.
Collapse
Affiliation(s)
- Ting Lin
- Department of Hematology, Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ya-Li Song
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pu Kuang
- Department of Hematology, Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Si Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhigang Mao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting-Ting Zeng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
31
|
Shi H, Wang M, Gong Y, Huang Y, Ning L, Xiang Y, Yin Y, Li G. Rapid Naked-Eye Tracking of On-Cell Phenotype Based on Dual-Aptamer-Weaved Cascade Assembly of Nanostructures. Anal Chem 2021; 93:11159-11166. [PMID: 34347435 DOI: 10.1021/acs.analchem.1c01668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Phenotypic plasticity is an emerging paradigm for providing biological and clinical insights into cancer initiation, progression, and resistance to therapy. However, it is a great challenge to track phenotypic information on live cells with high levels of sensitivity, specificity, and simplicity, when a specific cancer-cell subset is being targeted. In this work, we have successfully achieved cascade assembly of nanoparticles on the surface of specific cancer cells by designing a dual-aptamer-weaved molecular AND logic system. Taking advantage of spatial addressability, precise controllability, and targeting recognition of the nanostructure assemblies, we can precisely label the target-cell subset in a large population of similar cells and rapidly obtain phenotypic information in response to the surface changes of captured cancer cells. Without sophisticated instruments, we can know the phenotypic information on HepG2 cells in whole blood with a high level of sensitivity and rapid naked-eye tracking of on-cell phenotype changes of HepG2 cells undergoing epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Hai Shi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Mengjiao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Youjing Gong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yue Huang
- Department of Food Science and Engineering, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, P. R. China
| | - Limin Ning
- College of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Yang Xiang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Genxi Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.,Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
32
|
Haldavnekar R, Venkatakrishnan K, Tan DB. Boosting the sub-cellular biomolecular cancer signals by self-functionalized tag-free nano sensor. Biosens Bioelectron 2021; 190:113407. [PMID: 34134072 DOI: 10.1016/j.bios.2021.113407] [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: 10/13/2020] [Revised: 05/19/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023]
Abstract
Surface Enhanced Raman Scattering (SERS)-based sub-cellular cancer diagnosis can simultaneously obtain multiple biomolecular signals crucial in diagnostic platform for a heterogeneous disease like cancer. But, SERS-probes being typically tagged with chemical functionalization demonstrate limitations due to adverse biocompatibility, ineffective cellular internalization, SERS-signal quenching and spectral contamination. Although, tag-free SERS-probes overcome these limitations; complexity in spectral interpretation and detection insensitivity make it disadvantageous. In this study, we have exploited the inherent charges of cellular biomolecules and introduced self-functionalized complementary charged, tag-free SERS nano probes for biomolecule-specific investigation. Extremely small nano probes (sub 10 nm), synthesized with multiphoton ionization were functionalized with charge by physical synthesis without any ligands or chemical processes. The probes demonstrated significant SERS (EF~106) with analyte molecules (4ATP & 4MBA). Multifold signal boost was achieved for the signals of cellular components - amplification of ~7 fold for DNA, ~16 fold for proteins and ~24 fold for lipids with the commentary charged nano probes as compared to the neutral nano probes. The signal boost was attributed to the efficient delivery of extremely small, complementary charged probes to the cellular biomolecules of interest enabling simultaneous detection of sub-cellular biomolecules such as DNA, proteins and lipids and with high reproducibility. Cancer classification and investigation of drug resistance in cancer with single cell sensitivity was demonstrated. Such biomolecule-specific investigation of cancer from intact cells will open pathways for comprehensive cancer diagnosis.
Collapse
Affiliation(s)
- Rupa Haldavnekar
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Canada; Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Nanocharacterization Laboratory, Department of Aerospace Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Department of Biomedical Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada
| | - Krishnan Venkatakrishnan
- Keenan Research Center for Biomedical Science, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada; Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Nanocharacterization Laboratory, Department of Aerospace Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Department of Biomedical Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada.
| | - Dr Bo Tan
- Keenan Research Center for Biomedical Science, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada; Department of Biomedical Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada
| |
Collapse
|
33
|
Wang X, Cheng S, Wang X, Wei L, Kong Q, Ye M, Luo X, Xu J, Zhang C, Xian Y. pH-Sensitive Dye-Based Nanobioplatform for Colorimetric Detection of Heterogeneous Circulating Tumor Cells. ACS Sens 2021; 6:1925-1932. [PMID: 33881313 DOI: 10.1021/acssensors.1c00314] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The efficient capture and sensitive detection of circulating tumor cells (CTCs) play a vital role in cancer diagnosis and prognosis. However, CTCs in the peripheral blood are very rare and heterogeneous, which make them difficult to isolate and detect. Herein, a novel colorimetric nanobioplatform was successfully developed for the highly efficient capture and highly sensitive detection of heterogeneous CTCs, which consisted of two parts: the multivalent aptamer-modified gold nanoparticles as the capture unit and two kinds of aptamer-functionalized pH-sensitive allochroic dyes (thymolphthalein and curcumin) @ molybdenum disulfide nanoflakes (MoS2 NFs) acting as the visual simultaneous detection of heterogeneous CTCs. Using MCF-7 and HeLa cells as the CTC models, the capture unit can effectively isolate the CTCs due to the multivalent probe with improved affinity. The two allochroic dyes can display obvious color changes under alkaline conditions (pH 12.5) in the presence of MCF-7 and HeLa cells, which provided a rapid and sensitive strategy for visualizing simultaneous detection of heterogeneous CTCs as low as 5 cells mL-1. This nanoplatform possessed a high sensitivity toward CTC detection owing to high dye loading capacity of MoS2 NFs and allochroic dyes with excellent pH sensitivity. It can successfully distinguish and quantitatively detect the targeted heterogeneous CTCs from numerous interfering cells in diluted whole blood. It can also be used to detect CTCs from lysed blood samples from cancer patients, indicating promising application for cancer diagnosis.
Collapse
Affiliation(s)
- Xiuli Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Shasha Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Xinjun Wang
- Shanghai Zhangjiang Institute of Medical Innovation, Shanghai 201204, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Liran Wei
- Shanghai Zhangjiang Institute of Medical Innovation, Shanghai 201204, China
| | - Qianqian Kong
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Mingqiang Ye
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Xianzhu Luo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Jiao Xu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Cuiling Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Yuezhong Xian
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| |
Collapse
|
34
|
Kang K, Zhou X, Zhang Y, Zhu N, Li G, Yi Q, Wu Y. Cell-Released Magnetic Vesicles Capturing Metabolic Labeled Rare Circulating Tumor Cells Based on Bioorthogonal Chemistry. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007796. [PMID: 33749110 DOI: 10.1002/smll.202007796] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/24/2021] [Indexed: 06/12/2023]
Abstract
Capture of circulating tumor cells (CTCs) with high efficiency and high purity holds great value for potential clinical applications. Besides the existing problems of contamination from blood cells and plasma proteins, unknown/down-regulated expression of targeting markers (e.g., antigen, receptor, etc.) of CTCs have questioned the reliability and general applicability of current CTCs capture methodologies based on immune/aptamer-affinity. Herein, a cell-engineered strategy is designed to break down such barriers by employing the cell metabolism as the leading force to solve key problems. Generally, through an extracellular vesicle generation way, the cell-released magnetic vesicles inherited parent cellular membrane characteristics are produced, and then functionalized with dibenzoazacyclooctyne to target and isolate the metabolic labeled rare CTCs. This strategy offers good reliability and broader possibilities to capture different types of tumor cells, as proven by the capture efficiency above 84% and 82% for A549 and HepG2 cell lines as well as an extremely low detection limitation of 5 cells. Moreover, it enabled high purity enrichment of CTCs from 1 mL blood samples of tumor-bearing mice, only ≈5-757 white blood cells are non-specific caught, ignoring the potential phenotypic fluctuation associated with the cancer progression.
Collapse
Affiliation(s)
- Ke Kang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xiaoxi Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yujia Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Nanhang Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Guohao Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qiangying Yi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
35
|
Stanciu LA, Wei Q, Barui AK, Mohammad N. Recent Advances in Aptamer-Based Biosensors for Global Health Applications. Annu Rev Biomed Eng 2021; 23:433-459. [PMID: 33872519 DOI: 10.1146/annurev-bioeng-082020-035644] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Since aptamers were first reported in the early 2000s, research on their use for the detection of health-relevant analytical targets has exploded. This review article provides a brief overview of the most recent developments in the field of aptamer-based biosensors for global health applications. The review provides a description of general aptasensing principles and follows up with examples of recent reports of diagnostics-related applications. These applications include detection of proteins and small molecules, circulating cancer cells, whole-cell pathogens, extracellular vesicles, and tissue diagnostics. The review also discusses the main challenges that this growing technology faces in the quest of bringing these new devices from the laboratory to the market.
Collapse
Affiliation(s)
- Lia A Stanciu
- School of Materials Engineering, Purdue University, West Lafayette, Indiana 47907-2045, USA; .,Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, USA
| | - Qingshan Wei
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, USA
| | - Amit K Barui
- School of Materials Engineering, Purdue University, West Lafayette, Indiana 47907-2045, USA; .,Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, USA
| | - Noor Mohammad
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, USA
| |
Collapse
|
36
|
Xia L, Li G. Recent progress of microfluidics in surface-enhanced Raman spectroscopic analysis. J Sep Sci 2021; 44:1752-1768. [PMID: 33630352 DOI: 10.1002/jssc.202001196] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/20/2021] [Accepted: 02/20/2021] [Indexed: 12/21/2022]
Abstract
Surface-enhanced Raman spectroscopy is a significant analytical tool capable of fingerprint identification of molecule in a rapid and ultrasensitive manner. However, it is still hard to meet the requirements of practical sample analysis. The introduction of microfluidics can effectively enhance the performance of surface-enhanced Raman spectroscopy in complex sample analysis including reproducibility, selectivity, sensitivity, and speed. This review summarizes the recent progress of microfluidics in surface-enhanced Raman spectroscopic analysis through four combination approaches. First, microfluidic synthetic techniques offer uniform nano-/microparticle fabrication approaches for reproductive surface-enhanced Raman spectroscopic analysis. Second, the integration of microchip and surface-enhanced Raman spectroscopic substrate provides advanced devices for sensitive and efficient detection. Third, microfluidic sample preparations enable rapid separation and preconcentration of analyte prior to surface-enhanced Raman spectroscopic detection. Fourth, highly integrated microfluidic devices can be employed to realize multistep surface-enhanced Raman spectroscopic analysis containing material fabrication, sample preparation, and detection processes. Furthermore, the challenges and outlooks of the application of microfluidics in surface-enhanced Raman spectroscopic analysis are discussed.
Collapse
Affiliation(s)
- Ling Xia
- School of Chemistry, Sun Yat-sen University, Guangzhou, P. R. China
| | - Gongke Li
- School of Chemistry, Sun Yat-sen University, Guangzhou, P. R. China
| |
Collapse
|
37
|
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
|
38
|
Wu L, Wang Y, Xu X, Liu Y, Lin B, Zhang M, Zhang J, Wan S, Yang C, Tan W. Aptamer-Based Detection of Circulating Targets for Precision Medicine. Chem Rev 2021; 121:12035-12105. [PMID: 33667075 DOI: 10.1021/acs.chemrev.0c01140] [Citation(s) in RCA: 254] [Impact Index Per Article: 84.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The past decade has witnessed ongoing progress in precision medicine to improve human health. As an emerging diagnostic technique, liquid biopsy can provide real-time, comprehensive, dynamic physiological and pathological information in a noninvasive manner, opening a new window for precision medicine. Liquid biopsy depends on the sensitive and reliable detection of circulating targets (e.g., cells, extracellular vesicles, proteins, microRNAs) from body fluids, the performance of which is largely governed by recognition ligands. Aptamers are single-stranded functional oligonucleotides, capable of folding into unique tertiary structures to bind to their targets with superior specificity and affinity. Their mature evolution procedure, facile modification, and affinity regulation, as well as versatile structural design and engineering, make aptamers ideal recognition ligands for liquid biopsy. In this review, we present a broad overview of aptamer-based liquid biopsy techniques for precision medicine. We begin with recent advances in aptamer selection, followed by a summary of state-of-the-art strategies for multivalent aptamer assembly and aptamer interface modification. We will further describe aptamer-based micro-/nanoisolation platforms, aptamer-enabled release methods, and aptamer-assisted signal amplification and detection strategies. Finally, we present our perspectives regarding the opportunities and challenges of aptamer-based liquid biopsy for precision medicine.
Collapse
Affiliation(s)
- Lingling Wu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yidi Wang
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xing Xu
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yilong Liu
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Bingqian Lin
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Mingxia Zhang
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jialu Zhang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuang Wan
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Chaoyong Yang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Weihong Tan
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China.,The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
39
|
Liu LS, Wang F, Ge Y, Lo PK. Recent Developments in Aptasensors for Diagnostic Applications. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9329-9358. [PMID: 33155468 DOI: 10.1021/acsami.0c14788] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Aptamers are exciting smart molecular probes for specific recognition of disease biomarkers. A number of strategies have been developed to convert target-aptamer binding into physically detectable signals. Since the aptamer sequence was first discovered, a large variety of aptamer-based biosensors have been developed, with considerable attention paid to their potential applications in clinical diagnostics. So far, a variety of techniques in combination with a wide range of functional nanomaterials have been used for the design of aptasensors to further improve the sensitivity and detection limit of target determination. In this paper, the advantages of aptamers over traditional antibodies as the molecular recognition components in biosensors for high-throughput screening target molecules are highlighted. Aptamer-target pairing configurations are predominantly single- or dual-site binding; the design of recognition modes of each aptamer-target pairing configuration is described. Furthermore, signal transduction strategies including optical, electrical, mechanical, and mass-sensitive modes are clearly explained together with examples. Finally, we summarize the recent progress in the development of aptamer-based biosensors for clinical diagnosis, including detection of cancer and disease biomarkers and in vivo molecular imaging. We then conclude with a discussion on the advanced development and challenges of aptasensors.
Collapse
Affiliation(s)
- Ling Sum Liu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Fei Wang
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Yonghe Ge
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Pik Kwan Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
40
|
Affiliation(s)
- Keke Hu
- Department of Chemistry and Molecular Biology, University of Gothenburg, Kemivägen 10, 41296 Gothenburg, Sweden
| | - Tho D. K. Nguyen
- Department of Chemistry and Molecular Biology, University of Gothenburg, Kemivägen 10, 41296 Gothenburg, Sweden
| | - Stefania Rabasco
- Department of Chemistry and Molecular Biology, University of Gothenburg, Kemivägen 10, 41296 Gothenburg, Sweden
| | - Pieter E. Oomen
- Department of Chemistry and Molecular Biology, University of Gothenburg, Kemivägen 10, 41296 Gothenburg, Sweden
- ParaMedir B.V., 1e Energieweg 13, 9301 LK Roden, The Netherlands
| | - Andrew G. Ewing
- Department of Chemistry and Molecular Biology, University of Gothenburg, Kemivägen 10, 41296 Gothenburg, Sweden
| |
Collapse
|
41
|
Kim HS, Lee T, Yun J, Lee G, Hong Y. Cancer protein biomarker identification and quantification using nanoforest substrate and hand-held Raman spectrometer. Microchem J 2021. [DOI: 10.1016/j.microc.2020.105632] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
42
|
Guo QR, Zhang LL, Liu JF, Li Z, Li JJ, Zhou WM, Wang H, Li JQ, Liu DY, Yu XY, Zhang JY. Multifunctional microfluidic chip for cancer diagnosis and treatment. Nanotheranostics 2021; 5:73-89. [PMID: 33391976 PMCID: PMC7738943 DOI: 10.7150/ntno.49614] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Microfluidic chip is not a chip in the traditional sense. It is technologies that control fluids at the micro level. As a burgeoning biochip, microfluidic chips integrate multiple disciplines, including physiology, pathology, cell biology, biophysics, engineering mechanics, mechanical design, materials science, and so on. The application of microfluidic chip has shown tremendous promise in the field of cancer therapy in the past three decades. Various types of cell and tissue cultures, including 2D cell culture, 3D cell culture and tissue organoid culture could be performed on microfluidic chips. Patient-derived cancer cells and tissues can be cultured on microfluidic chips in a visible, controllable, and high-throughput manner, which greatly advances the process of personalized medicine. Moreover, the functionality of microfluidic chip is greatly expanding due to the customizable nature. In this review, we introduce its application in developing cancer preclinical models, detecting cancer biomarkers, screening anti-cancer drugs, exploring tumor heterogeneity and producing nano-drugs. We highlight the functions and recent development of microfluidic chip to provide references for advancing cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Qiao-Ru Guo
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Ling-Ling Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Ji-Fang Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Zhen Li
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R.China
| | - Jia-Jun Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Wen-Min Zhou
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Hui Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, P.R.China
| | - Jing-Quan Li
- The First Affiliated Hospital, Hainan Medical University, Haikou, P.R.China
| | - Da-Yu Liu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R.China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Jian-Ye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China.,The First Affiliated Hospital, Hainan Medical University, Haikou, P.R.China
| |
Collapse
|
43
|
Li R, Gui B, Mao H, Yang Y, Chen D, Xiong J. Self-Concentrated Surface-Enhanced Raman Scattering-Active Droplet Sensor with Three-Dimensional Hot Spots for Highly Sensitive Molecular Detection in Complex Liquid Environments. ACS Sens 2020; 5:3420-3431. [PMID: 32929960 DOI: 10.1021/acssensors.0c01276] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this work, a surface-enhanced Raman scattering (SERS)-active droplet with three-dimensional (3D) hot spots prepared from a superhydrophobic SERS substrate, which is inspired by the nut wizard strategy, was developed for ultrasensitive detection in complex liquid environments. The SERS substrate was composed of silver-capped parylene C-coated carbon nanoparticles (Ag-PC@CNPs). Such a SERS substrate was prepared by candle-soot deposition to provide a porous carbon nanoparticle layer followed by deposition of a parylene C film to protect the CNPs and then sputtering of silver nanoparticles. Similar to a nut wizard, a droplet rolling on the Ag-PC@CNP-coated substrate picked up the Ag-PC@CNPs. In this way, a self-concentrated and extremely sensitive SERS-active droplet sensor with 3D hot spots was formed. The sensor did not require precise laser focusing and showed relatively high repeatability and much higher sensitivity than those of a corresponding SERS substrate with two-dimensional hot spots. The sensor also achieved high sensitivity and specificity in complex liquid environments; in addition, bovine serum albumin with a concentration as low as 1 pM can be achieved. Consequently, an extremely simple, flexible, and highly sensitive SERS detection technique applicable to liquid biopsy analysis is anticipated.
Collapse
Affiliation(s)
- Ruirui Li
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, P.R. China
- National Key Laboratory for Electronic Measurement Technology, North University of China, Taiyuan 030051, P.R. China
| | - Bo Gui
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, P.R. China
| | - Haiyang Mao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, P.R. China
- Advanced Sensing Department, Wuxi Internet of Things Innovation Center Co. Ltd., Wuxi 214001, P.R. China
| | - Yudong Yang
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, P.R. China
| | - Dapeng Chen
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, P.R. China
- Advanced Sensing Department, Wuxi Internet of Things Innovation Center Co. Ltd., Wuxi 214001, P.R. China
| | - Jijun Xiong
- National Key Laboratory for Electronic Measurement Technology, North University of China, Taiyuan 030051, P.R. China
| |
Collapse
|
44
|
Pei H, Li L, Han Z, Wang Y, Tang B. Recent advances in microfluidic technologies for circulating tumor cells: enrichment, single-cell analysis, and liquid biopsy for clinical applications. LAB ON A CHIP 2020; 20:3854-3875. [PMID: 33107879 DOI: 10.1039/d0lc00577k] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Circulating tumor cells (CTCs) detach from primary or metastatic lesions and circulate in the peripheral blood, which is considered to be the cause of distant metastases. CTC analysis in the form of liquid biopsy, enumeration and molecular analysis provide significant clinical information for cancer diagnosis, prognosis and therapeutic strategies. Despite the great clinical value, CTC analysis has not yet entered routine clinical practice due to lack of efficient technologies to perform CTC isolation and single-cell analysis. Taking the rarity and inherent heterogeneity of CTCs into account, reliable methods for CTC isolation and detection are in urgent demand for obtaining valuable information on cancer metastasis and progression from CTCs. Microfluidic technology, featuring microfabricated structures, can precisely control fluids and cells at the micrometer scale, thus making itself a particularly suitable method for rare CTC manipulation. Besides the enrichment function, microfluidic chips can also realize the analysis function by integrating multiple detection technologies. In this review, we have summarized the recent progress in CTC isolation and detection using microfluidic technologies, with special attention to emerging direct enrichment and enumeration in vivo. Further, few insights into single CTC molecular analysis are also demonstrated. We have provided a review of potential clinical applications of CTCs, ranging from early screening and diagnosis, tumor progression and prognosis, treatment and resistance monitoring, to therapeutic evaluation. Through this review, we conclude that the clinical utility of CTCs will be expanded as the isolation and analysis techniques are constantly improving.
Collapse
Affiliation(s)
- Haimeng Pei
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | | | | | | | | |
Collapse
|
45
|
Zhang Q, Rong Y, Yi K, Huang L, Chen M, Wang F. Circulating tumor cells in hepatocellular carcinoma: single-cell based analysis, preclinical models, and clinical applications. Theranostics 2020; 10:12060-12071. [PMID: 33204329 PMCID: PMC7667686 DOI: 10.7150/thno.48918] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Circulating tumor cells (CTCs) are shed into the bloodstream from primary tumors and metastatic lesions and provide significant information about tumor progression and metastasis. CTCs contribute to tumor metastasis through the epithelial-to-mesenchymal transition (EMT). CTC clusters and stem-like phenotypes lead to a more aggressive and metastatic potential. CTCs retain the heterogeneity and imitate the nature of corresponding primary tumors. Therefore, it is important to use single-cell based analysis to obtain information on tumor heterogeneity and biology. CTCs are also good candidates for building preclinical models (especially 3D organoid cultures) for drug screening, disease modeling, genome editing, tumor immunity research, and organ-like biobank establishment. In this article, we summarize the current CTC capture technology, dissect the phenotypes associated with CTC metastasis, and review the progress in single-cell based analysis and preclinical modeling of the pattern and kinetics of CTCs. In particular, we discuss the use of CTCs to assess the progression of hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
| | | | | | | | | | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| |
Collapse
|
46
|
Cheng J, Liu Y, Zhao Y, Zhang L, Zhang L, Mao H, Huang C. Nanotechnology-Assisted Isolation and Analysis of Circulating Tumor Cells on Microfluidic Devices. MICROMACHINES 2020; 11:E774. [PMID: 32823926 PMCID: PMC7465711 DOI: 10.3390/mi11080774] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022]
Abstract
Circulating tumor cells (CTCs), a type of cancer cell that spreads from primary tumors into human peripheral blood and are considered as a new biomarker of cancer liquid biopsy. It provides the direction for understanding the biology of cancer metastasis and progression. Isolation and analysis of CTCs offer the possibility for early cancer detection and dynamic prognosis monitoring. The extremely low quantity and high heterogeneity of CTCs are the major challenges for the application of CTCs in liquid biopsy. There have been significant research endeavors to develop efficient and reliable approaches to CTC isolation and analysis in the past few decades. With the advancement of microfabrication and nanomaterials, a variety of approaches have now emerged for CTC isolation and analysis on microfluidic platforms combined with nanotechnology. These new approaches show advantages in terms of cell capture efficiency, purity, detection sensitivity and specificity. This review focuses on recent progress in the field of nanotechnology-assisted microfluidics for CTC isolation and detection. Firstly, CTC isolation approaches using nanomaterial-based microfluidic devices are summarized and discussed. The different strategies for CTC release from the devices are specifically outlined. In addition, existing nanotechnology-assisted methods for CTC downstream analysis are summarized. Some perspectives are discussed on the challenges of current methods for CTC studies and promising research directions.
Collapse
Affiliation(s)
- Jie Cheng
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Liu
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Lina Zhang
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China;
| | - Lingqian Zhang
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Haiyang Mao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Chengjun Huang
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
47
|
Li N, Zong S, Zhang Y, Wang Z, Wang Y, Zhu K, Yang K, Wang Z, Chen B, Cui Y. A SERS-colorimetric dual-mode aptasensor for the detection of cancer biomarker MUC1. Anal Bioanal Chem 2020; 412:5707-5718. [PMID: 32632516 DOI: 10.1007/s00216-020-02790-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022]
Abstract
Human mucin-1 (MUC1) has attracted considerable attention owing to its overexpression in diverse malignancies. Here, for the rapid and efficient detection of MUC1, we present a SERS-colorimetric dual-mode aptasensor, by integrating SERS probes with magnetic separation, which has several distinctive advantages. Using such a dual-mode aptasensor, the colorimetric functionality is distinguishable by the naked eye, providing a fast and straightforward screening ability for the detection of MUC1. Moreover, SERS-based detection greatly improves the detection sensitivity, reaching a limit of detection of 0.1 U/mL. In addition, the combination of SERS and colorimetric method holds the advantages of these two techniques and thereby increases the reliability and efficiency of MUC1 detection. On the one hand, the magnetic nanobeads functionalized with MUC1-specific aptamer were utilized as an efficient capturing substrate for separating MUC1 from biological complex medium. On the other hand, the gold-silver core-shell nanoparticles modified with Raman reporters and the complementary sequences of MUC1 were used as the signal indicator, which could simultaneously report the SERS signal and colorimetric change. This strategy can achieve a good detection range and realize MUC1 analysis in real patients' samples. Thus, we anticipate that this kind of aptasensor would provide promising potential applications in the diagnosis and prognosis of cancers. Graphical abstract.
Collapse
Affiliation(s)
- Na Li
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Shenfei Zong
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Yizhi Zhang
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Zhile Wang
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Yujie Wang
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Kai Zhu
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Kuo Yang
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Zhuyuan Wang
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing, 210096, Jiangsu, China.
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Yiping Cui
- Advanced Photonics Center, School of Electronic Science and Engineering, Southeast University, Nanjing, 210096, Jiangsu, China
| |
Collapse
|
48
|
Wu L, Teixeira A, Garrido-Maestu A, Muinelo-Romay L, Lima L, Santos LL, Prado M, Diéguez L. Profiling DNA mutation patterns by SERS fingerprinting for supervised cancer classification. Biosens Bioelectron 2020; 165:112392. [PMID: 32729513 DOI: 10.1016/j.bios.2020.112392] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/04/2020] [Accepted: 06/13/2020] [Indexed: 12/14/2022]
Abstract
Profiling DNA mutation patterns for cancer classification plays an essential role in precision and personalized medicine. Conventional PCR-based mutation assay is limited by the extensive labour on target amplification. We herein create an amplification-free surface enhanced Raman spectroscopy (SERS) biochip which enables direct and simultaneous identification of multiple point mutations in tumor cells. Without pre-amplifying the target sequences, the SERS assay reads out the presence of cellular mutations through the interpretation of Raman fingerprints. The SERS sensor is integrated into a microfluidic chip, achieving one-step multiplex analysis within 40 min. Importantly, by combining SERS spectra encoding technique with supervised learning algorithm, a panel of nucleotide mixtures can be well distinguished according to their mutation profiles. We initially demonstrate an excellent levels of classification in samples from colorectal cancer and melanoma cell lines. For final clinical validation, the system performance is verified by classifying cancer patient samples, which shows an accuracy above 90%. Due to the simplicity and rapidness, the SERS biosensor is expected to become a promising tool for clinical point-of-care diagnosis towards precision medicine.
Collapse
Affiliation(s)
- Lei Wu
- Medical Devices, International Iberian Nanotechnology Laboratory - INL, 4715-330 Braga, Portugal
| | - Alexandra Teixeira
- Medical Devices, International Iberian Nanotechnology Laboratory - INL, 4715-330 Braga, Portugal
| | - Alejandro Garrido-Maestu
- Food Quality and Safety, International Iberian Nanotechnology Laboratory - INL, 4715-330 Braga, Portugal
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706, Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Luis Lima
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, R. Dr. António Bernardino de Almeida 865, 4200-072 Porto, Portugal; Glycobiology in Cancer, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), R. Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, R. Dr. António Bernardino de Almeida 865, 4200-072 Porto, Portugal; Health School of University Fernando Pessoa, Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology, Porto, Portugal
| | - Marta Prado
- Food Quality and Safety, International Iberian Nanotechnology Laboratory - INL, 4715-330 Braga, Portugal
| | - Lorena Diéguez
- Medical Devices, International Iberian Nanotechnology Laboratory - INL, 4715-330 Braga, Portugal.
| |
Collapse
|
49
|
Wu L, Zhang Z, Tang M, Zhu D, Dong X, Hu J, Qi C, Tang H, Pang D. Spectrally Combined Encoding for Profiling Heterogeneous Circulating Tumor Cells Using a Multifunctional Nanosphere‐Mediated Microfluidic Platform. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Ling‐Ling Wu
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Zhi‐Ling Zhang
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Man Tang
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Dong‐Liang Zhu
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Xiao‐Juan Dong
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Jiao Hu
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Chu‐Bo Qi
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Hong‐Wu Tang
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Dai‐Wen Pang
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
- State Key Laboratory of Medicinal Chemical Biology Tianjin Key Laboratory of Biosensing and Molecular Recognition Research Center for Analytical Sciences College of Chemistry Nankai University Tianjin 300071 P. R. China
| |
Collapse
|
50
|
Wu L, Zhang Z, Tang M, Zhu D, Dong X, Hu J, Qi C, Tang H, Pang D. Spectrally Combined Encoding for Profiling Heterogeneous Circulating Tumor Cells Using a Multifunctional Nanosphere‐Mediated Microfluidic Platform. Angew Chem Int Ed Engl 2020; 59:11240-11244. [DOI: 10.1002/anie.201914468] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/15/2020] [Indexed: 01/08/2023]
Affiliation(s)
- Ling‐Ling Wu
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Zhi‐Ling Zhang
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Man Tang
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Dong‐Liang Zhu
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Xiao‐Juan Dong
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Jiao Hu
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Chu‐Bo Qi
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Hong‐Wu Tang
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
| | - Dai‐Wen Pang
- College of Chemistry and Molecular Sciences The Institute for Advanced Studies Wuhan University Wuhan 430072 P. R. China
- State Key Laboratory of Medicinal Chemical Biology Tianjin Key Laboratory of Biosensing and Molecular Recognition Research Center for Analytical Sciences College of Chemistry Nankai University Tianjin 300071 P. R. China
| |
Collapse
|