1
|
Kang Y, Cao X, Fan Y, Li Y, Xu T, Zhou Q, He B. Exosome biomarkers in breast cancer: Systematic review and meta-analysis. Clin Chim Acta 2025; 574:120342. [PMID: 40311726 DOI: 10.1016/j.cca.2025.120342] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Breast cancer (BC) has become the primary cancer that threatens women's health and life expectancy. Early diagnosis is crucial for effective treatment and favourable prognosis. As a non-invasive and valuable liquid biopsy method, exosomes are promising for the diagnosis and prognosis of BC. The aim of this meta-analysis is to evaluate the diagnostic and prognostic value of exosome biomarkers in BC. METHODS A systematic search of relevant English literature was conducted in PubMed, Web of Science, and Cochrane library until August 2024 (diagnosis) and October 2024 (prognosis). QUADAS-2 and QUAPAS were used to assess the quality of the literature. Summary statistics and analyses of relevant effect sizes were conducted using STATA software. Subgroup analysis and sensitivity analysis were performed to identify potential sources of heterogeneity. RESULTS For diagnosis, a total of 31 articles with 3,778 patients and 2,722 controls were included, the pooled sensitivity (SEN), specificity (SPE), and area under the receiver operating characteristic curve (AUC) of overall exosome biomarkers were 0.89 (95 %CI: 0.86-0.91), 0.87 (95 %CI: 0.85-0.90), and 0.94 (95 %CI: 0.92-0.96), respectively, indicating a high diagnostic value of exosomes in BC patients. Subgroup analysis suggested that miRNAs in exosomes exhibited better diagnostic value compared to proteins and non-miRNAs, the SEN, SPE, and AUC were 0.89 (95 %CI: 0.82-0.93), 0.86 (95 %CI: 0.80-0.90), and 0.92 (95 %CI: 0.90-0.94), respectively. Among all miRNAs, the pooled SEN, SPE, and AUC of miR-21 were 0.86 (95 %CI: 0.67-0.95), 0.90 (95 %CI: 0.78-0.96), and 0.95 (95 %CI: 0.92-0.96), respectively. The diagnostic efficiency was improved when biomarkers were combined as a panel (SEN 0.91 versus 0.87, SPE 0.89 versus 0.86, AUC 0.96 versus 0.91). In terms of prognosis, we retrieved 14 articles with 2,781 patients. The pooled HR of overall survival (OS) and progression-free survival (PFS) were 1.41 (95 %CI: 0.92-1.90) and 4.39 (95 %CI: 1.87-6.91), respectively, indicating exosome biomarkers like soluble HLA-G, miR-1246, miR-155, and PSMA were a predictor of poor PFS in BC patients. Subgroup analysis in OS group revealed a significant association between the overexpression of exosome proteins (soluble HLA-G, AnxA2, NGF, CXCL13) and worse OS in BC patients (HR = 2.91, 95 %CI: 1.36-4.47). Similarly, the overexpression of miR-1246 and miR-155 was associated with worse PFS in BC patients (HR = 4.13, 95 %CI: 1.24-7.03). Moreover, when biomarkers were combined as a panel, the prognostic efficiency significantly improved in OS (HR = 4.05, 95 %CI: 2.26-5.84) outcome. CONCLUSION The meta-analysis revealed that exosome miR-21 might serve as a promising diagnostic biomarker in BC. Dysregulated exosome proteins and miRNAs could predict poor OS and PFS outcomes, respectively.
Collapse
Affiliation(s)
- Yurou Kang
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoqing Cao
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yujing Fan
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yimin Li
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Qing Zhou
- NHC Key Laboratory of Contraceptives Vigilance and Fertility Surveillance, Jiangsu Health Development Research Center, Jiangsu Provincial Medical Key Laboratory of Fertility Protection and Health Technology Assessment, NO.277 Fenghuang West Street, Nanjing, China.
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Li F, Jiang Y, Wang X, Gao Y, Lo CS, Su S, Wu Z, Jiang B, Zhao Z, Lin S, Xie Y, Chen J, Guo Q, Dong Z, Zhu J. Titanium nitride meta-biosensors targeting extracellular vesicles for high-sensitivity prostate cancer detection. Biosens Bioelectron 2025; 277:117288. [PMID: 39985905 DOI: 10.1016/j.bios.2025.117288] [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: 01/07/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Disposable plasmonic metasurfaces with high biosensing performance are urgently sought for clinical label-free detection. Low-cost aluminum (Al) and titanium nitride (TiN) offer promising alternatives to noble metals for constructing these metasurfaces. However, Al suffers from limited chemical stability, and TiN exhibits weak plasmonic effects, both of which hinder their application in meta-biosensing. Here we integrate their complementary advantages and propose the TiN/Al meta-biosensors. They not only empower the unique near-field enhancement for sensing by TiN/Al hybrid plasmonic modes, but also construct a robust TiN armor against external wear, heat, moisture and corrosion during the bio-detection process. Compared to traditional gold-based counterparts, our meta-biosensors offer superior optical sensitivity at a much lower cost and with fewer pretreatment steps. The excellent biosensing performance facilitates the development of a high-throughput detection system for serum small extracellular vesicles (sEVs), aiding in the diagnosis and follow-up of prostate cancer. The sEVs meta-biosensing demonstrates a diagnostic sensitivity of 100% for significantly distinguishing early cancer, breaking through the conventional testing limitation. Moreover, it doubles the prediction accuracy of cancer recurrence risk following surgery. Our research highlights the potential for large-scale development of powerful meta-biosensors based on non-noble materials, opening up significant opportunities in cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Fajun Li
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China; Xiamen Dili-Chip Technology Co. Ltd., Xiamen, 361000, China; School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, 361100, China
| | - Yuanyuan Jiang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Xuegang Wang
- Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Yuan Gao
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Ching Shu Lo
- School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, 361100, China
| | - Shengdong Su
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Zhilin Wu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Bingliang Jiang
- Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Zhongjie Zhao
- Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Shaowei Lin
- Department of Nuclear Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Yinong Xie
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Junjie Chen
- Analysis and Measurement Center, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Qiwei Guo
- Department of Central Laboratory, Department of Obstetrics and Gynecology, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Zhaogang Dong
- Quantum Innovation Centre, Agency for Science Technology and Research, 2 Fusionopolis Way, Innovis #08-03, Singapore, 138634, Republic of Singapore
| | - Jinfeng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China; Xiamen Dili-Chip Technology Co. Ltd., Xiamen, 361000, China; School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, 361100, China.
| |
Collapse
|
3
|
Zhang Y, Wang C, Shao H. Nanoplasmonic Sensing of Heterogeneous Extracellular Vesicles: From Bulk to Single Vesicles. SMALL METHODS 2025:e2500097. [PMID: 40391615 DOI: 10.1002/smtd.202500097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/16/2025] [Indexed: 05/22/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous nanoscale membrane vesicles released by almost all cell types into the circulation. Depending on their biogenesis and cells of origin, EVs show considerable heterogeneity in their biophysical and biomolecular composition and can serve as reflective and dynamic blood biomarkers for personalized medicine. Conventional analytical technologies, however, often lack the compatibility to reveal nanoscale EV features and resolve vesicle heterogeneity. The past decade has since witnessed the development of various nanoplasmonic technologies to empower EV analysis, through bulk and single-vesicle characterization, at an unprecedented scale and resolution. These platforms achieve versatile measurements that are not only size-matched to EV dimensions but can also probe multiplexed biomolecular contents, thereby providing new insights into EV heterogeneity and enabling transformative clinical opportunities. In this review, key characteristics of EVs and their remarkable heterogeneity are introduced. The sensing principles of plasmonic platforms are also discussed, with recent technology developments highlighted to resolve EV heterogeneity, through bulk analyses of EV subpopulations as well as high-resolution single-EV measurements. An outlook is further provided on emerging opportunities, at the interface of biomarker discovery and technology innovation, to develop empowering nanoplasmonic EV platforms for personalized medicine. biosensing; bulk analysis; extracellular vesicles; nanoplasmonics; single-vesicle analysis.
Collapse
Affiliation(s)
- Yan Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Chao Wang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Materials Science and Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117575, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 138673, Singapore
| |
Collapse
|
4
|
Ren TJ, Zhang YZ, Zhang Q, Tan M, Gu J, Tong Y, Wang Y, Yang C, Xu ZR. Accurate Cancer Diagnosis and Treatment Monitoring through Multiplexed Profiling of Protein Markers on Small Extracellular Vesicles. ACS NANO 2025; 19:18630-18643. [PMID: 40340378 DOI: 10.1021/acsnano.5c02864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
The detection of small extracellular vesicles (sEVs) is currently a pivotal liquid biopsy approach for noninvasive cancer diagnosis. However, the lack of adequate specificity and sensitivity, as well as labor-intensive purification and analysis procedures, present challenges in isolating and profiling sEVs. Here, we present a protein-specific enzymatic optical reporter deposition-based liquid biopsy assay for the rapid and efficient capture and ultrasensitive detection of sEVs using a minimal volume of initial biofluids (10 μL). Biotin aptamers were employed to label sEV proteins for peroxidase conjugation, catalyzing the conversion of fluorescein tyramine into highly reactive free radicals. Efficient signal conversion was achieved by depositing nanoheterolayers composed of covalent tyraminated complexes onto sEV surfaces. The present method offers a detection limit of 6.4 × 103 particles mL-1 with a linear range of 104-1010 particles mL-1 for sEVs. Two machine learning algorithms, principal coordinates analysis and principal component analysis, were subsequently applied for dimensionality reduction. In a clinical cohort of 84 patients, including 6 cancer types and noncancer cases, the assay achieved an overall accuracy of 100% (95% confidence interval) in distinguishing between cancer and noncancer controls and 96% in classifying cancer types. As drugs are frequently administered to patients to modulate the activity of tumor cells, we investigated the efficacy of this strategy in treatment monitoring, achieving an overall accuracy of 100%. This strategy demonstrates a cost-effective, rapid, and low sample volume consumption approach that holds significant potential for precise cancer diagnosis and auxiliary assessment of drug response in clinical settings.
Collapse
Affiliation(s)
- Ting-Ju Ren
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Ying-Zhi Zhang
- National Clinical Research Center for Laboratory Medicine, Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110819, China
| | - Qi Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Meilun Tan
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Jiahui Gu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Yuxiao Tong
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Yue Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Chunguang Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Zhang-Run Xu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| |
Collapse
|
5
|
Zhang L, Wong CY, Shao H. Integrated technologies for molecular profiling of genetic and modified biomarkers in extracellular vesicles. LAB ON A CHIP 2025. [PMID: 40135945 DOI: 10.1039/d5lc00053j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane vesicles actively released by cells into a variety of biofluids. EVs carry myriad molecular cargoes; these include classical genetic biomarkers inherited from the parent cells as well as EV modifications by other entities (e.g., small molecule drugs). Aided by these diverse cargoes, EVs enable long-distance intercellular communication and have been directly implicated in various disease pathologies. As such, EVs are being increasingly recognized as a source of valuable biomarkers for minimally-invasive disease diagnostics and prognostics. Despite the clinical potential, EV molecular profiling remains challenging, especially in clinical settings. Due to the nanoscale dimension of EVs as well as the abundance of contaminants in biofluids, conventional EV detection methods have limited resolution, require extensive sample processing and can lose rare biomarkers. To address these challenges, new micro- and nanotechnologies have been developed to discover EV biomarkers and empower clinical applications. In this review, we introduce EV biogenesis for different cargo incorporation, and discuss the use of various EV biomarkers for clinical applications. We also assess different chip-based integrated technologies developed to measure genetic and modified biomarkers in EVs. Finally, we highlight future opportunities in technology development to facilitate the clinical translation of various EV biomarkers.
Collapse
Affiliation(s)
- Li Zhang
- Institute for Health Innovation & Technology, National University of Singapore, MD6, 14 Medical Drive #14-01, Singapore 117599, Singapore.
| | - Chi Yan Wong
- Institute for Health Innovation & Technology, National University of Singapore, MD6, 14 Medical Drive #14-01, Singapore 117599, Singapore.
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, MD6, 14 Medical Drive #14-01, Singapore 117599, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117583, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Department of Materials Science and Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore
| |
Collapse
|
6
|
Kang JY, Mun D, Park M, Yoo G, Kim H, Yun N, Joung B. Injured Cardiac Tissue-Targeted Delivery of TGFβ1 siRNA by FAP Aptamer-Functionalized Extracellular Vesicles Promotes Cardiac Repair. Int J Nanomedicine 2025; 20:2575-2592. [PMID: 40046817 PMCID: PMC11881639 DOI: 10.2147/ijn.s497428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/23/2025] [Indexed: 05/13/2025] Open
Abstract
Purpose Small-interfering RNA (siRNA) therapy holds significant potential for treating cardiac injury; however, its clinical application is constrained by poor blood stability and insufficient cellular uptake. Extracellular vesicles (EVs) have emerged as an effective delivery system for siRNA in vivo; but their lack of specific cell or tissue-targeting ability remains a major challenge. Thus, we aimed to develop an EV-based delivery system capable of targeted delivery of therapeutic siRNA to injured cardiac tissue for cardiac repair. Methods To identify fibroblast activation protein (FAP) as a potential target for delivery to injured cardiac tissue, we analyzed cardiac tissues from patients with heart failure and angiotensin II (Ang II)-treated mice. Injured cardiac tissue-targeting EVs were developed by embedding a cholesterol-conjugated FAP aptamer, which specifically targets FAP, onto human serum-derived EVs (hEV). Results Our findings revealed that FAP is upregulated after cardiac injury, highlighting its potential as a target for siRNA delivery to injured cardiac tissues. We successfully developed FAP aptamer-functionalized hEV (hEV@FAP) and confirmed their typical EV characteristics, including morphology, size distribution, zeta potential, and marker protein expression. In addition, hEV@FAP demonstrated high targeting selectivity to FAP-positive regions both in vitro and in vivo. To treat cardiac injury, hEV@FAP were loaded with TGFβ1 siRNA (siTGFβ1), identified as a molecular target for cardiac repair. In Ang II-treated mice, intravenous administration of hEV@FAP-siTGFβ1 effectively reduced Ang II-induced TGFβ1 expression in cardiac tissues, attributed to the protective and targeting capabilities of hEV@FAP. Consequently, hEV@FAP-siTGFβ1 significantly improved cardiac function, reduced myocardial fibrosis, and decreased cardiomyocyte cross-sectional area (P < 0.05) without inducing systemic toxicity. Conclusion hEV@FAP represents a novel approach for targeted delivery of therapeutic siRNA to injured cardiac tissues, providing a promising nanomedicine for cardiac repair.
Collapse
Affiliation(s)
- Ji-Young Kang
- Division of Cardiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Dasom Mun
- Division of Cardiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Malgeum Park
- Division of Cardiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Gyeongseo Yoo
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyoeun Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Nuri Yun
- GNTPharma Science and Technology Center for Health, Incheon, 21983, Republic of Korea
| | - Boyoung Joung
- Division of Cardiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
7
|
Zhang G, Huang X, Liu S, Xu Y, Wang N, Yang C, Zhu Z. Demystifying EV heterogeneity: emerging microfluidic technologies for isolation and multiplexed profiling of extracellular vesicles. LAB ON A CHIP 2025; 25:1228-1255. [PMID: 39775292 DOI: 10.1039/d4lc00777h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous lipid containers carrying complex molecular cargoes, including proteins, nucleic acids, glycans, etc. These vesicles are closely associated with specific physiological characteristics, which makes them invaluable in the detection and monitoring of various diseases. However, traditional isolation methods are often labour-intensive, inefficient, and time-consuming. In addition, single biomarker analyses are no longer accurate enough to meet diagnostic needs. Routine isolation and molecular analysis of high-purity EVs in clinical applications is even more challenging. In this review, we discuss a promising solution, microfluidic-based techniques, that combine efficient isolation and multiplex detection of EVs, to further demystify EV heterogeneity. These microfluidic-based EV multiplexing platforms will hopefully facilitate development of liquid biopsies and offer promising opportunities for personalised therapy.
Collapse
Affiliation(s)
- Guihua Zhang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Xiaodan Huang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Sinong Liu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Yiling Xu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Nan Wang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, 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 & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, 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, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai 200127, China
| | - Zhi Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, 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
|
8
|
An L, Liu Y, Liu Y. Organ-on-a-Chip Applications in Microfluidic Platforms. MICROMACHINES 2025; 16:201. [PMID: 40047688 PMCID: PMC11857120 DOI: 10.3390/mi16020201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 03/09/2025]
Abstract
Microfluidic technology plays a crucial role in organ-on-a-chip (OoC) systems by replicating human physiological processes and disease states, significantly advancing biomedical research and drug discovery. This article reviews the design and fabrication processes of microfluidic devices. It also explores how these technologies are integrated into OoC platforms to simulate human physiological environments, highlighting key principles, technological advances, and diverse applications. Through case studies involving the simulation of multiple organs such as the heart, liver, and lungs, the article evaluates the impact of OoC systems' integrated microfluidic technology on drug screening, toxicity assessment, and personalized medicine. In addition, this article considers technical challenges, ethical issues, and future directions, and looks ahead to further optimizing the functionality and biomimetic precision of OoCs through innovation, emphasizing its critical role in promoting personalized medicine and precision treatment strategies.
Collapse
Affiliation(s)
- Ling An
- School of Engineering, Dali University, Dali 671003, China;
| | - Yi Liu
- School of Engineering, Dali University, Dali 671003, China;
| | - Yaling Liu
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015, USA
| |
Collapse
|
9
|
Zhang H, Wu B, Wang Y, Du H, Fang L. Extracellular Vesicles as Mediators and Potential Targets in Combating Cancer Drug Resistance. Molecules 2025; 30:498. [PMID: 39942602 PMCID: PMC11819960 DOI: 10.3390/molecules30030498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Extracellular vesicles (EVs) are key mediators in the communication between cancer cells and their microenvironment, significantly influencing drug resistance. This review provides a comprehensive analysis of the roles of EVs in promoting drug resistance through mechanisms such as drug efflux, apoptosis resistance, autophagy imbalance, and tumor microenvironment modulation. Despite extensive research, details of EVs biogenesis, cargo selection, and specific pathways in EVs-mediated drug resistance are not fully understood. This review critically examines recent advancements, highlighting key studies that elucidate the molecular mechanisms of EVs functions. Additionally, innovative therapeutic strategies targeting EVs are explored, including inhibiting EVs biogenesis, engineering EVs for drug delivery, and identifying resistance-inhibiting molecules within EVs. By integrating insights from primary research and proposing new directions for future studies, this review aims to advance the understanding of EVs in cancer biology and foster effective interventions to mitigate drug resistance in cancer therapy.
Collapse
Affiliation(s)
- Haodong Zhang
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; (H.Z.); (H.D.)
| | - Bohan Wu
- Westa College, Southwest University, Chongqing 400715, China; (B.W.); (Y.W.)
| | - Yanheng Wang
- Westa College, Southwest University, Chongqing 400715, China; (B.W.); (Y.W.)
| | - Huamao Du
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; (H.Z.); (H.D.)
| | - Liaoqiong Fang
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; (H.Z.); (H.D.)
- National Engineering Research Center of Ultrasound Medicine, Chongqing 401121, China
| |
Collapse
|
10
|
Lin X, Zhu J, Shen J, Zhang Y, Zhu J. Advances in exosome plasmonic sensing: Device integration strategies and AI-aided diagnosis. Biosens Bioelectron 2024; 266:116718. [PMID: 39216205 DOI: 10.1016/j.bios.2024.116718] [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: 06/14/2024] [Revised: 08/11/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Exosomes, as next-generation biomarkers, has great potential in tracking cancer progression. They face many detection limitations in cancer diagnosis. Plasmonic biosensors have attracted considerable attention at the forefront of exosome detection, due to their label-free, real-time, and high-sensitivity features. Their advantages in multiplex immunoassays of minimal liquid samples establish the leading position in various diagnostic studies. This review delineates the application principles of plasmonic sensing technologies, highlighting the importance of exosomes-based spectrum and image signals in disease diagnostics. It also introduces advancements in miniaturizing plasmonic biosensing platforms of exosomes, which can facilitate point-of-care testing for future healthcare. Nowadays, inspired by the surge of artificial intelligence (AI) for science and technology, more and more AI algorithms are being adopted to process the exosome spectrum and image data from plasmonic detection. Using representative algorithms of machine learning has become a mainstream trend in plasmonic biosensing research for exosome liquid biopsy. Typically, these algorithms process complex exosome datasets efficiently and establish powerful predictive models for precise diagnosis. This review further discusses critical strategies of AI algorithm selection in exosome-based diagnosis. Particularly, we categorize the AI algorithms into the interpretable and uninterpretable groups for exosome plasmonic detection applications. The interpretable AI enhances the transparency and reliability of diagnosis by elucidating the decision-making process, while the uninterpretable AI provides high diagnostic accuracy with robust data processing by a "black-box" working mode. We believe that AI will continue to promote significant progress of exosome plasmonic detection and mobile healthcare in the near future.
Collapse
Affiliation(s)
- Xiangyujie Lin
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Jiaheng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Jiaqing Shen
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Youyu Zhang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| | - Jinfeng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| |
Collapse
|
11
|
Liu W, Chung K, Yu S, Lee LP. Nanoplasmonic biosensors for environmental sustainability and human health. Chem Soc Rev 2024; 53:10491-10522. [PMID: 39192761 DOI: 10.1039/d3cs00941f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Monitoring the health conditions of the environment and humans is essential for ensuring human well-being, promoting global health, and achieving sustainability. Innovative biosensors are crucial in accurately monitoring health conditions, uncovering the hidden connections between the environment and human well-being, and understanding how environmental factors trigger autoimmune diseases, neurodegenerative diseases, and infectious diseases. This review evaluates the use of nanoplasmonic biosensors that can monitor environmental health and human diseases according to target analytes of different sizes and scales, providing valuable insights for preventive medicine. We begin by explaining the fundamental principles and mechanisms of nanoplasmonic biosensors. We investigate the potential of nanoplasmonic techniques for detecting various biological molecules, extracellular vesicles (EVs), pathogens, and cells. We also explore the possibility of wearable nanoplasmonic biosensors to monitor the physiological network and healthy connectivity of humans, animals, plants, and organisms. This review will guide the design of next-generation nanoplasmonic biosensors to advance sustainable global healthcare for humans, the environment, and the planet.
Collapse
Affiliation(s)
- Wenpeng Liu
- Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA.
| | - Kyungwha Chung
- Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA.
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Subin Yu
- Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA.
| | - Luke P Lee
- Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA 94720, USA
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Korea
| |
Collapse
|
12
|
Tang H, Yu D, Zhang J, Wang M, Fu M, Qian Y, Zhang X, Ji R, Gu J, Zhang X. The new advance of exosome-based liquid biopsy for cancer diagnosis. J Nanobiotechnology 2024; 22:610. [PMID: 39380060 PMCID: PMC11463159 DOI: 10.1186/s12951-024-02863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Liquid biopsy is a minimally invasive method that uses biofluid samples instead of tissue samples for cancer diagnosis. Exosomes are small extracellular vesicles secreted by donor cells and act as mediators of intercellular communication in human health and disease. Due to their important roles, exosomes have been considered as promising biomarkers for liquid biopsy. However, traditional methods for exosome isolation and cargo detection methods are time-consuming and inefficient, limiting their practical application. In the past decades, many new strategies, such as microfluidic chips, nanowire arrays and electrochemical biosensors, have been proposed to achieve rapid, accurate and high-throughput detection and analysis of exosomes. In this review, we discussed about the new advance in exosome-based liquid biopsy technology, including isolation, enrichment, cargo detection and analysis approaches. The comparison of currently available methods is also included. Finally, we summarized the advantages and limitations of the present strategies and further gave a perspective to their future translational use.
Collapse
Affiliation(s)
- Haozhou Tang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
- Department of Orthopaedics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
| | - Dan Yu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jiahui Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Maoye Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Min Fu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yu Qian
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaoxin Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Runbi Ji
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jianmei Gu
- Departmemt of Clinical Laboratory Medicine, Nantong Tumor Hospital/Affiliated Tumor Hospital of Nantong University, Nantong, 226300, China.
- Affiliated Cancer Hospital of Nantong University, Nantong, 226300, China.
| | - Xu Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
13
|
Luo X, Yan S, Chen G, Wang Y, Zhang X, Lan J, Chen J, Yao X. A cavity induced mode hybridization plasmonic sensor for portable detection of exosomes. Biosens Bioelectron 2024; 261:116492. [PMID: 38870828 DOI: 10.1016/j.bios.2024.116492] [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: 01/14/2024] [Revised: 03/20/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024]
Abstract
Exosomes have been considered as promising biomarkers for cancer diagnosis due to their abundant information from originating cells. However, sensitive and reliable detection of exosomes is still facing technically challenges due to the lack of a sensing platform with high sensitivity and reproducibility. To address the challenges, here we propose a portable surface plasmon resonance (SPR) sensing of exosomes with a three-layer Au mirror/SiO2 spacer/Au nanohole sensor, fabricated by an economical polystyrene nanosphere self-assembly method. The SiO2 spacer can act as an optical cavity and induce mode hybridization, leading to excellent optimization of both sensitivity and full width at half maximum compared with normal single layer Au nanohole sensors. When modified with CD63 or EpCAM aptamers, a detection of limit (LOD) of as low as 600 particles/μL was achieved. The sensors showed good capability to distinguish between non-tumor derived L02 exosomes and tumor derived HepG2 exosomes. Additionally, high reproducibility was also achieved in detection of artificial serum samples with RSD as low as 2%, making it feasible for clinical applications. This mode hybridization plasmonic sensor provides an effective approach to optimize the detection sensitivity of exosomes, pushing SPR sensing one step further towards cancer diagnosis.
Collapse
Affiliation(s)
- Xinming Luo
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350108, China
| | - Sen Yan
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Guanyu Chen
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350108, China
| | - Yuxin Wang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350108, China
| | - Xi Zhang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350108, China; Innovative Drug Research Institute, Fujian Medical University, Fuzhou, 350108, China
| | - Jianming Lan
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350108, China; Innovative Drug Research Institute, Fujian Medical University, Fuzhou, 350108, China
| | - Jinghua Chen
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350108, China; Innovative Drug Research Institute, Fujian Medical University, Fuzhou, 350108, China.
| | - Xu Yao
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350108, China; Innovative Drug Research Institute, Fujian Medical University, Fuzhou, 350108, China.
| |
Collapse
|
14
|
Chen Y, Zhang L, Wu X, Sun X, Sundah NR, Wong CY, Natalia A, Tam JKC, Lim DWT, Chowbay B, Ang BT, Tang C, Loh TP, Shao H. Magnetic augmentation through multi-gradient coupling enables direct and programmable profiling of circulating biomarkers. Nat Commun 2024; 15:8410. [PMID: 39333499 PMCID: PMC11437193 DOI: 10.1038/s41467-024-52754-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
Conventional magnetic biosensing technologies have reduced analytical capacity for magnetic field dimensionality and require extensive sample processing. To address these challenges, we spatially engineer 3D magnetic response gradients for direct and programmable molecular detection in native biofluids. Named magnetic augmentation through triple-gradient coupling for high-performance detection (MATCH), the technology comprises gradient-distributed magnetic nanoparticles encapsulated within responsive hydrogel pillars and suspended above a magnetic sensor array. This configuration enables multi-gradient matching to achieve optimal magnetic activation, response and transduction, respectively. Through focused activation by target biomarkers, the platform preferentially releases sensor-proximal nanoparticles, generating response gradients that complement the sensor's intrinsic detection capability. By implementing an upstream module that recognizes different biomarkers and releases universal activation molecules, the technology achieves programmable detection of various circulating biomarkers in native plasma. It bypasses conventional magnetic labeling, completes in <60 minutes and achieves sensitive detection (down to 10 RNA and 1000 protein copies). We apply the MATCH to measure RNAs and proteins directly in patient plasma, achieving accurate cancer classification.
Collapse
Affiliation(s)
- Yuan Chen
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Li Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Xingjie Wu
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Xuecheng Sun
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Noah R Sundah
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Chi Yan Wong
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Auginia Natalia
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - John K C Tam
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Darren Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Centre for Clinician-Scientist Development, Duke-NUS Medical School, Singapore, Singapore
| | - Balram Chowbay
- Centre for Clinician-Scientist Development, Duke-NUS Medical School, Singapore, Singapore
- Clinical Pharmacology Laboratory, National Cancer Centre Singapore, Singapore, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Beng Ti Ang
- National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Carol Tang
- National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- SG Enable, Innovation, Singapore, Singapore
| | - Tze Ping Loh
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
15
|
Lang T, Geaghan S, Loh TP, Mak C, Papassotiriou I, Kyriakopoulou LG. Considerations for applying emerging technologies in paediatric laboratory medicine. Clin Chem Lab Med 2024; 62:1938-1949. [PMID: 39044644 DOI: 10.1515/cclm-2023-1408] [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: 03/21/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024]
Abstract
Emerging technology in laboratory medicine can be defined as an analytical method (including biomarkers) or device (software, applications, and algorithms) that by its stage of development, translation into broad routine clinical practice, or geographical adoption and implementation has the potential to add value to clinical diagnostics. Paediatric laboratory medicine itself may be considered an emerging area of specialisation that is established relatively recently following increased appreciation and understanding of the unique physiology and healthcare needs of the children. Through four clinical (neonatal hypoglycaemia, neonatal hyperbilirubinaemia, sickle cell disorder, congenital adrenal hyperplasia) and six technological (microassays, noninvasive testing, alternative matrices, next generation sequencing, exosome analysis, machine learning) illustrations, key takeaways of application of emerging technology for each area are summarised. Additionally, nine key considerations when applying emerging technology in paediatric laboratory medicine setting are discussed.
Collapse
Affiliation(s)
- Tim Lang
- Department of Blood Sciences, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Sharon Geaghan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tze Ping Loh
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Chloe Mak
- Division of Chemical Pathology, Hong Kong Children's Hospital, Kowloon, Hong Kong SAR, China
| | - Ioannis Papassotiriou
- First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | | |
Collapse
|
16
|
Xia X, Yang X, Gao W, Huang W, Xia X, Yan D. A novel HER2 targeting nanoagent self-assembled from affibody-epothilone B conjugate for cancer therapy. J Nanobiotechnology 2024; 22:502. [PMID: 39169343 PMCID: PMC11337599 DOI: 10.1186/s12951-024-02754-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Epothilone B (Epo B), a promising antitumor compound effective against various types of cancer cells in vitro. However, its poor selectivity for tumor cells and inadequate therapeutic windows significantly limit its potential clinical application. Affibody is a class of non-immunoglobulin affinity proteins with precise targeting capability to overexpressed molecular receptors on cancer cells, has been intensively investigated due to its exceptional affinity properties. In this study, we present a targeted nanoagent self-assembled from the precursor of an affibody conjugated with Epo B via a linker containing the thioketal (tk) group that is sensitive to reactive oxygen species (ROS). The core-shell structure of the ZHER2:342-Epo B Affibody-Drug Conjugate Nanoagent (Z-E ADCN), with the cytotoxin Epo B encapsulated within the ZHER2:342 affibody corona, leads to significantly reduced side effects on normal organs. Moreover, the abundant presence of ZHER2:342 on the surface effectively enhances the targeting capacity and tumor accumulation of the drug. Z-E ADCN can be internalized by cancer cells via HER2 receptor-mediated endocytosis followed by Epo B release in response to high levels of ROS, resulting in excellent anticancer efficacy in HER2-positive tumor models.
Collapse
Affiliation(s)
- Xuelin Xia
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Xiaoyuan Yang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Wenhui Gao
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Xiaoxia Xia
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
17
|
Zeng Y, Gao Y, He L, Ge W, Wang X, Ma T, Xie X. Smart delivery vehicles for cancer: categories, unique roles and therapeutic strategies. NANOSCALE ADVANCES 2024; 6:4275-4308. [PMID: 39170969 PMCID: PMC11334973 DOI: 10.1039/d4na00285g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/19/2024] [Indexed: 08/23/2024]
Abstract
Chemotherapy and surgery remain the primary treatment modalities for cancers; however, these techniques have drawbacks, such as cancer recurrence and toxic side effects, necessitating more efficient cancer treatment strategies. Recent advancements in research and medical technology have provided novel insights and expanded our understanding of cancer development; consequently, scholars have investigated several delivery vehicles for cancer therapy to improve the efficiency of cancer treatment and patient outcomes. Herein, we summarize several types of smart therapeutic carriers and elaborate on the mechanism underlying drug delivery. We reveal the advantages of smart therapeutic carriers for cancer treatment, focus on their effectiveness in cancer immunotherapy, and discuss the application of smart cancer therapy vehicles in combination with other emerging therapeutic strategies for cancer treatment. Finally, we summarize the bottlenecks encountered in the development of smart cancer therapeutic vehicles and suggest directions for future research. This review will promote progress in smart cancer therapy and facilitate related research.
Collapse
Affiliation(s)
- Yiyu Zeng
- Department of Stomatology, The Second Xiangya Hospital, Central South University Changsha 410011 P. R. China
| | - Yijun Gao
- Department of Stomatology, The Second Xiangya Hospital, Central South University Changsha 410011 P. R. China
| | - Liming He
- Department of Stomatology, Changsha Stomatological Hospital Changsha 410004 P. R. China
| | - Wenhui Ge
- Department of Stomatology, The Second Xiangya Hospital, Central South University Changsha 410011 P. R. China
| | - Xinying Wang
- Department of Stomatology, The Second Xiangya Hospital, Central South University Changsha 410011 P. R. China
| | - Tao Ma
- Department of Stomatology, The Second Xiangya Hospital, Central South University Changsha 410011 P. R. China
| | - Xiaoyan Xie
- Department of Stomatology, The Second Xiangya Hospital, Central South University Changsha 410011 P. R. China
| |
Collapse
|
18
|
Dong L, Liu M, Fang M, Lu Q, Li X, Ma Y, Zhao T. Nucleation-Inhibited Emulsion Interfacial Assembled Polydopamine Microvesicles as Artificial Antigen-Presenting Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400714. [PMID: 38593314 DOI: 10.1002/smll.202400714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/31/2024] [Indexed: 04/11/2024]
Abstract
Albeit microemulsion systems have emerged as efficient platforms for fabricating tunable nano/microstructures, lack of understanding on the emulsion-interfacial assembly hindered the control of fabrication. Herein, a nucleation-inhibited microemulsion interfacial assembly method is proposed, which deviates from conventional interfacial nucleation approaches, for the synthesis of polydopamine microvesicles (PDA MVs). These PDA MVs exhibit an approximate diameter of 1 µm, showcasing a pliable structure reminiscent of cellular morphology. Through modifications of antibodies on the surface of PDA MVs, their capacity as artificial antigen presentation cells is evaluated. In comparison to solid nanoparticles, PDA MVs with cell-like structures show enhanced T-cell activation, resulting in a 1.5-fold increase in CD25 expression after 1 day and a threefold surge in PD-1 positivity after 7 days. In summary, the research elucidates the influence of nucleation and interfacial assembly in microemulsion polymerization systems, providing a direct synthesis method for MVs and substantiating their effectiveness as artificial antigen-presenting cells.
Collapse
Affiliation(s)
- Lingkai Dong
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Minchao Liu
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Meng Fang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China
| | - Qianqian Lu
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Xingjin Li
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Yanming Ma
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Tiancong Zhao
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
19
|
Ji L, Ruan H, Fu Y, Xiong S. A study of antigen selection by extracellular vesicles as vaccine candidates against Mycobacterium tuberculosis infection. J Med Microbiol 2024; 73. [PMID: 39133547 DOI: 10.1099/jmm.0.001865] [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] [Indexed: 08/13/2024] Open
Abstract
Introduction. Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (M. tb), remains a significant global public health concern. It is crucial to develop more effective vaccines for TB in order to achieve global control of the disease. Extracellular vesicles (EVs) are spherical membrane-bound structures released by pathogens and host cells. During the course of an infection, both pathogen- and host-derived EVs are produced and play important roles in determining the course of the infection. EVs offer intriguing tools as potential vaccines due to their ability to deliver multiple pathogen or host antigens.Hypothesis /Gap Statement. We hypothesized that EVs derived from M. tb and EVs from M. tb-infected macrophages may serve as potential vaccine candidates against M. tb infection.Aim. This study aims to compare the immunogenicity and immune protection between M. tb EVs and M. tb-infected macrophage-derived EVs.Methodology. In this study, EVs were extracted from culture supernatants of M. tb and M. tb-infected macrophages, respectively. Mass spectrometry was employed to explore the antigen composition of H37Rv-Mφ-EVs and H37Rv-EVs. Cytokine profiling and antibody response assays were used to analyse the immunogenicity offered by EVs. Additionally, we used histological examination to evaluate and protective efficacy of the EVs.Results. Our results demonstrated that mice immunized by EVs released from M. tb-infected macrophages induced stronger inflammatory cytokine response than M. tb. Moreover, EVs from M. tb-infected macrophages reinforced T-cell activation and antibody response compared to M. tb EVs. Proteomic analysis revealed that EVs from M. tb-infected macrophages containing immunodominant cargos have stronger binding ability with major histocompatibility complex molecules, which may contribute to the protection from M. tb infection. Indeed, immunization of EVs released from M. tb-infected macrophages significantly reduced the bacterial load and better protection against M. tb infection than EVs from M. tb. Importantly, the selected antigens (Ag85B, ESAT-6 and the Rv0580c) from EVs of M. tb-infected macrophages exhibited effective immunogenicity.Conclusion. Our results suggested that EVs derived from M. tb-infected macrophages might serve as a proper antigenic library for vaccine candidates against M. tb challenge.
Collapse
Affiliation(s)
- Lin Ji
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
| | - Hang Ruan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
| | - Yuxuan Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
- The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu 215123, PR China
| |
Collapse
|
20
|
Xie M, Wu Y, Zhang Y, Lu R, Zhai Z, Huang Y, Wang F, Xin C, Rong G, Zhao C, Jiang K, Zhou X, Zhou X, Zhu X, Hong J, Zhang C. Membrane Fusion-Mediated Loading of Therapeutic siRNA into Exosome for Tissue-Specific Application. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403935. [PMID: 38889294 DOI: 10.1002/adma.202403935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/30/2024] [Indexed: 06/20/2024]
Abstract
Tissue-specific delivery of oligonucleotide therapeutics beyond the liver remains a key challenge in nucleic acid drug development. To address this issue, exploiting exosomes as a novel carrier has emerged as a promising approach for efficient nucleic acid drug delivery. However, current exosome-based delivery systems still face multiple hurdles in their clinical applications. Herein, this work presents a strategy for constructing a hybrid exosome vehicle (HEV) through a DNA zipper-mediated membrane fusion approach for tissue-specific siRNA delivery. As a proof-of-concept, this work successfully fuses a liposome encapsulating anti-NFKBIZ siRNAs with corneal epithelium cell (CEC)-derived exosomes to form a HEV construct for the treatment of dry eye disease (DED). With homing characteristics inherited from exosomes, the siRNA-bearing HEV can target its parent cells and efficiently deliver the siRNA payloads to the cornea. Subsequently, the NFKBIZ gene silencing significantly reduces pro-inflammatory cytokine secretions from the ocular surface, reshapes its inflammatory microenvironment, and ultimately achieves an excellent therapeutic outcome in a DED mouse model. As a versatile platform, this hybrid exosome with targeting capability and designed therapeutic siRNAs may hold great potential in various disease treatments.
Collapse
Affiliation(s)
- Miao Xie
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yuqing Wu
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Yilun Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Ruiyang Lu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Zimeng Zhai
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Yangyang Huang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Fujun Wang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Changchang Xin
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Guangyu Rong
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Chen Zhao
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Kai Jiang
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Xujiao Zhou
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Xingtao Zhou
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Jiaxu Hong
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
- Department of Ophthalmology, Children's Hospital of Fudan University, Shanghai, 201102, P. R. China
- Key Laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, 200031, P. R. China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, P. R. China
| | - Chuan Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
21
|
Zhao Z, Liu M, Duan L, Lin R, Wang L, Zhang P, Li J, Ma B, Yang Y, Bu F, Wang R, Zhou W, Chao D, Zhao Y, Yin S, Tang L, Zhang W, Li X, Zhao D. Ultrafine Asymmetric Soft/Stiff Nanohybrids with Tunable Patchiness via a Dynamic Surface-Mediated Assembly. J Am Chem Soc 2024. [PMID: 39025826 DOI: 10.1021/jacs.4c05072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Asymmetric soft-stiff patch nanohybrids with small size, spatially separated organics and inorganics, controllable configuration, and appealing functionality are important in applications, while the synthesis remains a great challenge. Herein, based on polymeric single micelles (the smallest assembly subunit of mesoporous materials), we report a dynamic surface-mediated anisotropic assembly approach to fabricate a new type of small asymmetric organic/inorganic patch nanohybrid for the first time. The size of this asymmetric organic/inorganic nanohybrid is ∼20 nm, which contains dual distinct subunits of a soft organic PS-PVP-PEO single micelle nanosphere (12 nm in size and 632 MPa in Young' modulus) and stiff inorganic SiO2 nanobulge (∼8 nm, 2275 MPa). Moreover, the number of SiO2 nanobulges anchored on each micelle can be quantitatively controlled (from 1 to 6) by dynamically tuning the density (fluffy or dense state) of the surface cap organic groups. This small asymmetric patch nanohybrid also exhibits a dramatically enhanced uptake level of which the total amount of intracellular endocytosis is about three times higher than that of the conventional nanohybrids.
Collapse
Affiliation(s)
- Zaiwang Zhao
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Mengli Liu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Linlin Duan
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Runfeng Lin
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Lipeng Wang
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Pengfei Zhang
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Jun Li
- Henan Institute of Advanced Technology, College of Chemistry, Zhengzhou University, Zhengzhou 450052, China
| | - Bing Ma
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Yang Yang
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Fanxing Bu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Ruicong Wang
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Wanhai Zhou
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Dongliang Chao
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Yujuan Zhao
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Sixing Yin
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Lei Tang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| | - Weian Zhang
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
| | - Xiaomin Li
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| | - Dongyuan Zhao
- College of Energy Materials and Chemistry, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010070, P. R. China
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM and State Key Laboratory of Molecular Engineering of Polymers, College of Chemistry and Materials, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
22
|
Liu Y, Sundah NR, Ho NRY, Shen WX, Xu Y, Natalia A, Yu Z, Seet JE, Chan CW, Loh TP, Lim BY, Shao H. Bidirectional linkage of DNA barcodes for the multiplexed mapping of higher-order protein interactions in cells. Nat Biomed Eng 2024; 8:909-923. [PMID: 38898172 DOI: 10.1038/s41551-024-01225-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/05/2024] [Indexed: 06/21/2024]
Abstract
Capturing the full complexity of the diverse hierarchical interactions in the protein interactome is challenging. Here we report a DNA-barcoding method for the multiplexed mapping of pairwise and higher-order protein interactions and their dynamics within cells. The method leverages antibodies conjugated with barcoded DNA strands that can bidirectionally hybridize and covalently link to linearize closely spaced interactions within individual 3D protein complexes, encoding and decoding the protein constituents and the interactions among them. By mapping protein interactions in cancer cells and normal cells, we found that tumour cells exhibit a larger diversity and abundance of protein complexes with higher-order interactions. In biopsies of human breast-cancer tissue, the method accurately identified the cancer subtype and revealed that higher-order protein interactions are associated with cancer aggressiveness.
Collapse
Affiliation(s)
- Yu Liu
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Noah R Sundah
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Nicholas R Y Ho
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
| | - Wan Xiang Shen
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yun Xu
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Auginia Natalia
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Zhonglang Yu
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Ju Ee Seet
- Department of Pathology, National University Hospital, Singapore, Singapore
| | - Ching Wan Chan
- Department of Surgery, National University Hospital, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tze Ping Loh
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Brian Y Lim
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore.
- Department of Computer Science, School of Computing, National University of Singapore, Singapore, Singapore.
| | - Huilin Shao
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
23
|
Lim W, Lee S, Koh M, Jo A, Park J. Recent advances in chemical biology tools for protein and RNA profiling of extracellular vesicles. RSC Chem Biol 2024; 5:483-499. [PMID: 38846074 PMCID: PMC11151817 DOI: 10.1039/d3cb00200d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized vesicles secreted by cells that contain various cellular components such as proteins, nucleic acids, and lipids from the parent cell. EVs are abundant in body fluids and can serve as circulating biomarkers for a variety of diseases or as a regulator of various biological processes. Considering these characteristics of EVs, analysis of the EV cargo has been spotlighted for disease diagnosis or to understand biological processes in biomedical research. Over the past decade, technologies for rapid and sensitive analysis of EVs in biofluids have evolved, but detection and isolation of targeted EVs in complex body fluids is still challenging due to the unique physical and biological properties of EVs. Recent advances in chemical biology provide new opportunities for efficient profiling of the molecular contents of EVs. A myriad of chemical biology tools have been harnessed to enhance the analytical performance of conventional assays for better understanding of EV biology. In this review, we will discuss the improvements that have been achieved using chemical biology tools.
Collapse
Affiliation(s)
- Woojeong Lim
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
| | - Soyeon Lee
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
| | - Minseob Koh
- Department of Chemistry, Pusan National University Busan 46241 Republic of Korea
| | - Ala Jo
- Center for Nanomedicine, Institute for Basic Science Seoul 03722 Republic of Korea
| | - Jongmin Park
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University Chuncheon 24341 Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University Chuncheon 24341 Republic of Korea
| |
Collapse
|
24
|
Lei Y, Fei X, Ding Y, Zhang J, Zhang G, Dong L, Song J, Zhuo Y, Xue W, Zhang P, Yang C. Simultaneous subset tracing and miRNA profiling of tumor-derived exosomes via dual-surface-protein orthogonal barcoding. SCIENCE ADVANCES 2023; 9:eadi1556. [PMID: 37792944 PMCID: PMC10550235 DOI: 10.1126/sciadv.adi1556] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/31/2023] [Indexed: 10/06/2023]
Abstract
The clinical potential of miRNA-based liquid biopsy has been largely limited by the heterogeneous sources in plasma and tedious assay processes. Here, we develop a precise and robust one-pot assay called dual-surface-protein-guided orthogonal recognition of tumor-derived exosomes and in situ profiling of microRNAs (SORTER) to detect tumor-derived exosomal miRNAs and enhance the diagnostic accuracy of prostate cancer (PCa). The SORTER uses two allosteric aptamers against exosomal marker CD63 and tumor marker EpCAM to create an orthogonal labeling barcode and achieve selective sorting of tumor-specific exosome subtypes. Furthermore, the labeled barcode on tumor-derived exosomes initiated targeted membrane fusion with liposome probes to import miRNA detection reagents, enabling in situ sensitive profiling of tumor-derived exosomal miRNAs. With a signature of six miRNAs, SORTER differentiated PCa and benign prostatic hyperplasia with an accuracy of 100%. Notably, the diagnostic accuracy reached 90.6% in the classification of metastatic and nonmetastatic PCa. We envision that the SORTER will promote the clinical adaptability of miRNA-based liquid biopsy.
Collapse
Affiliation(s)
- Yanmei Lei
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaochen Fei
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yue Ding
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jianhui Zhang
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Guihua Zhang
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Liang Dong
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jia Song
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ying Zhuo
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Wei Xue
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Peng Zhang
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chaoyong Yang
- Institute of Molecular Medicine, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, 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
|
25
|
Kollipara PS, Li X, Li J, Chen Z, Ding H, Kim Y, Huang S, Qin Z, Zheng Y. Hypothermal opto-thermophoretic tweezers. Nat Commun 2023; 14:5133. [PMID: 37612299 PMCID: PMC10447564 DOI: 10.1038/s41467-023-40865-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 08/10/2023] [Indexed: 08/25/2023] Open
Abstract
Optical tweezers have profound importance across fields ranging from manufacturing to biotechnology. However, the requirement of refractive index contrast and high laser power results in potential photon and thermal damage to the trapped objects, such as nanoparticles and biological cells. Optothermal tweezers have been developed to trap particles and biological cells via opto-thermophoresis with much lower laser powers. However, the intense laser heating and stringent requirement of the solution environment prevent their use for general biological applications. Here, we propose hypothermal opto-thermophoretic tweezers (HOTTs) to achieve low-power trapping of diverse colloids and biological cells in their native fluids. HOTTs exploit an environmental cooling strategy to simultaneously enhance the thermophoretic trapping force at sub-ambient temperatures and suppress the thermal damage to target objects. We further apply HOTTs to demonstrate the three-dimensional manipulation of functional plasmonic vesicles for controlled cargo delivery. With their noninvasiveness and versatile capabilities, HOTTs present a promising tool for fundamental studies and practical applications in materials science and biotechnology.
Collapse
Affiliation(s)
| | - Xiuying Li
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Jingang Li
- Materials Science and Engineering Program and Texas Materials Institute, The University of Texas at Austin, Austin, TX, 78712, USA
- Laser Thermal Laboratory, Department of Mechanical Engineering, University of California, Berkeley, CA, 94720, USA
| | - Zhihan Chen
- Materials Science and Engineering Program and Texas Materials Institute, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Hongru Ding
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Youngsun Kim
- Materials Science and Engineering Program and Texas Materials Institute, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Suichu Huang
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing of Ministry of Education and School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 15001, China
| | - Zhenpeng Qin
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Yuebing Zheng
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA.
- Materials Science and Engineering Program and Texas Materials Institute, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
26
|
Pan S, Ding A, Li Y, Sun Y, Zhan Y, Ye Z, Song N, Peng B, Li L, Huang W, Shao H. Small-molecule probes from bench to bedside: advancing molecular analysis of drug-target interactions toward precision medicine. Chem Soc Rev 2023; 52:5706-5743. [PMID: 37525607 DOI: 10.1039/d3cs00056g] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Over the past decade, remarkable advances have been witnessed in the development of small-molecule probes. These molecular tools have been widely applied for interrogating proteins, pathways and drug-target interactions in preclinical research. While novel structures and designs are commonly explored in probe development, the clinical translation of small-molecule probes remains limited, primarily due to safety and regulatory considerations. Recent synergistic developments - interfacing novel chemical probes with complementary analytical technologies - have introduced and expedited diverse biomedical opportunities to molecularly characterize targeted drug interactions directly in the human body or through accessible clinical specimens (e.g., blood and ascites fluid). These integrated developments thus offer unprecedented opportunities for drug development, disease diagnostics and treatment monitoring. In this review, we discuss recent advances in the structure and design of small-molecule probes with novel functionalities and the integrated development with imaging, proteomics and other emerging technologies. We further highlight recent applications of integrated small-molecule technologies for the molecular analysis of drug-target interactions, including translational applications and emerging opportunities for whole-body imaging, tissue-based measurement and blood-based analysis.
Collapse
Affiliation(s)
- Sijun Pan
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yisi Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yaxin Sun
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yueqin Zhan
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Zhenkun Ye
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Ning Song
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Wei Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore 117599, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117583, Singapore
| |
Collapse
|
27
|
Yan M, Chen Q, Liu T, Li X, Pei P, Zhou L, Zhou S, Zhang R, Liang K, Dong J, Wei X, Wang J, Terasaki O, Chen P, Gu Z, Jiang L, Kong B. Site-selective superassembly of biomimetic nanorobots enabling deep penetration into tumor with stiff stroma. Nat Commun 2023; 14:4628. [PMID: 37532754 PMCID: PMC10397308 DOI: 10.1038/s41467-023-40300-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 06/24/2023] [Indexed: 08/04/2023] Open
Abstract
Chemotherapy remains as the first-choice treatment option for triple-negative breast cancer (TNBC). However, the limited tumor penetration and low cellular internalization efficiency of current nanocarrier-based systems impede the access of anticancer drugs to TNBC with dense stroma and thereby greatly restricts clinical therapeutic efficacy, especially for TNBC bone metastasis. In this work, biomimetic head/hollow tail nanorobots were designed through a site-selective superassembly strategy. We show that nanorobots enable efficient remodeling of the dense tumor stromal microenvironments (TSM) for deep tumor penetration. Furthermore, the self-movement ability and spiky head markedly promote interfacial cellular uptake efficacy, transvascular extravasation, and intratumoral penetration. These nanorobots, which integrate deep tumor penetration, active cellular internalization, near-infrared (NIR) light-responsive release, and photothermal therapy capacities into a single nanodevice efficiently suppress tumor growth in a bone metastasis female mouse model of TNBC and also demonstrate potent antitumor efficacy in three different subcutaneous tumor models.
Collapse
Affiliation(s)
- Miao Yan
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Qing Chen
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, 200032, Shanghai, P. R. China
| | - Tianyi Liu
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Xiaofeng Li
- Department of Chemistry, The University of Hong Kong, Hong Kong, 999077, P. R. China
| | - Peng Pei
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Lei Zhou
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, 200032, Shanghai, P. R. China
| | - Shan Zhou
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Runhao Zhang
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Kang Liang
- School of Chemical Engineering and Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, 200032, Shanghai, P. R. China
| | - Xunbin Wei
- Biomedical Engineering Department and Cancer Hospital and Institute, Key Laboratory of Carcinogenesis and Translational Research, Peking University, 100081, Beijing, P. R. China
| | - Jinqiang Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, 310063, Hangzhou, P. R. China
| | - Osamu Terasaki
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, P. R. China
| | - Pu Chen
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, 310063, Hangzhou, P. R. China
| | - Libo Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, 200032, Shanghai, P. R. China.
| | - Biao Kong
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China.
- Yiwu Research Institute of Fudan University, 322000, Yiwu, Zhejiang, P. R. China.
- Shandong Research Institute, Fudan University, 250103, Jinan, Shandong, P. R. China.
| |
Collapse
|
28
|
Zhang Y, Wong CY, Lim CZJ, Chen Q, Yu Z, Natalia A, Wang Z, Pang QY, Lim SW, Loh TP, Ang BT, Tang C, Shao H. Multiplexed RNA profiling by regenerative catalysis enables blood-based subtyping of brain tumors. Nat Commun 2023; 14:4278. [PMID: 37460561 DOI: 10.1038/s41467-023-39844-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/29/2023] [Indexed: 07/20/2023] Open
Abstract
Current technologies to subtype glioblastoma (GBM), the most lethal brain tumor, require highly invasive brain biopsies. Here, we develop a dedicated analytical platform to achieve direct and multiplexed profiling of circulating RNAs in extracellular vesicles for blood-based GBM characterization. The technology, termed 'enzyme ZIF-8 complexes for regenerative and catalytic digital detection of RNA' (EZ-READ), leverages an RNA-responsive transducer to regeneratively convert and catalytically enhance signals from rare RNA targets. Each transducer comprises hybrid complexes - protein enzymes encapsulated within metal organic frameworks - to configure strong catalytic activity and robust protection. Upon target RNA hybridization, the transducer activates directly to liberate catalytic complexes, in a target-recyclable manner; when partitioned within a microfluidic device, these complexes can individually catalyze strong chemifluorescence reactions for digital RNA quantification. The EZ-READ platform thus enables programmable and reliable RNA detection, across different-sized RNA subtypes (miRNA and mRNA), directly in sample lysates. When clinically evaluated, the EZ-READ platform established composite signatures for accurate blood-based GBM diagnosis and subtyping.
Collapse
Affiliation(s)
- Yan Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Chi Yan Wong
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Carine Z J Lim
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Qingchang Chen
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Zhonglang Yu
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Auginia Natalia
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Zhigang Wang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Qing You Pang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - See Wee Lim
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Tze Ping Loh
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Beng Ti Ang
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Carol Tang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- National Neuroscience Institute, Singapore, Singapore.
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
29
|
Jalali M, Del Real Mata C, Montermini L, Jeanne O, I Hosseini I, Gu Z, Spinelli C, Lu Y, Tawil N, Guiot MC, He Z, Wachsmann-Hogiu S, Zhou R, Petrecca K, Reisner WW, Rak J, Mahshid S. MoS 2-Plasmonic Nanocavities for Raman Spectra of Single Extracellular Vesicles Reveal Molecular Progression in Glioblastoma. ACS NANO 2023. [PMID: 37366177 DOI: 10.1021/acsnano.2c09222] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Extracellular vesicles (EVs) are continually released from cancer cells into biofluids, carrying actionable molecular fingerprints of the underlying disease with considerable diagnostic and therapeutic potential. The scarcity, heterogeneity and intrinsic complexity of tumor EVs present a major technological challenge in real-time monitoring of complex cancers such as glioblastoma (GBM). Surface-enhanced Raman spectroscopy (SERS) outputs a label-free spectroscopic fingerprint for EV molecular profiling. However, it has not been exploited to detect known biomarkers at the single EV level. We developed a multiplex fluidic device with embedded arrayed nanocavity microchips (MoSERS microchip) that achieves 97% confinement of single EVs in a minute amount of fluid (<10 μL) and enables molecular profiling of single EVs with SERS. The nanocavity arrays combine two featuring characteristics: (1) An embedded MoS2 monolayer that enables label-free isolation and nanoconfinement of single EVs due to physical interaction (Coulomb and van der Waals) between the MoS2 edge sites and the lipid bilayer; and (2) A layered plasmonic cavity that enables sufficient electromagnetic field enhancement inside the cavities to obtain a single EV level signal resolution for stratifying the molecular alterations. We used the GBM paradigm to demonstrate the diagnostic potential of the SERS single EV molecular profiling approach. The MoSERS multiplexing fluidic achieves parallel signal acquisition of glioma molecular variants (EGFRvIII oncogenic mutation and MGMT expression) in GBM cells. The detection limit of 1.23% was found for stratifying these key molecular variants in the wild-type population. When interfaced with a convolutional neural network (CNN), MoSERS improved diagnostic accuracy (87%) with which GBM mutations were detected in 12 patient blood samples, on par with clinical pathology tests. Thus, MoSERS demonstrates the potential for molecular stratification of cancer patients using circulating EVs.
Collapse
Affiliation(s)
- Mahsa Jalali
- Department of Bioengineering, McGill University, Montreal, Quebec H3A 0E9, Canada
| | | | - Laura Montermini
- Research Institute of the McGill University Health Centre (RIMUHC), Montreal, Quebec H4A 3J1, Canada
| | - Olivia Jeanne
- Department of Bioengineering, McGill University, Montreal, Quebec H3A 0E9, Canada
| | - Imman I Hosseini
- Department of Bioengineering, McGill University, Montreal, Quebec H3A 0E9, Canada
- Department of Physics, McGill University, Montreal, Quebec H3A 2T8, Canada
| | - Zonglin Gu
- College of Physical Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Cristiana Spinelli
- Research Institute of the McGill University Health Centre (RIMUHC), Montreal, Quebec H4A 3J1, Canada
| | - Yao Lu
- Department of Bioengineering, McGill University, Montreal, Quebec H3A 0E9, Canada
| | - Nadim Tawil
- Research Institute of the McGill University Health Centre (RIMUHC), Montreal, Quebec H4A 3J1, Canada
| | - Marie Christine Guiot
- Department of Neuropathology, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Zhi He
- Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, Hangzhou, 310058 China
| | | | - Ruhong Zhou
- Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, Hangzhou, 310058 China
| | - Kevin Petrecca
- Department of Neuropathology, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Walter W Reisner
- Department of Physics, McGill University, Montreal, Quebec H3A 2T8, Canada
| | - Janusz Rak
- Research Institute of the McGill University Health Centre (RIMUHC), Montreal, Quebec H4A 3J1, Canada
| | - Sara Mahshid
- Department of Bioengineering, McGill University, Montreal, Quebec H3A 0E9, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
30
|
Fu X, Song J, Yan W, Downs BM, Wang W, Li J. The biological function of tumor-derived extracellular vesicles on metabolism. Cell Commun Signal 2023; 21:150. [PMID: 37349803 PMCID: PMC10286389 DOI: 10.1186/s12964-023-01111-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/24/2023] [Indexed: 06/24/2023] Open
Abstract
Multiple studies have shown that extracellular vesicles (EVs) play a key role in the process of information transfer and material transport between cells. EVs are classified into different types according to their sizes, which includes the class of exosomes. In comparison to normal EVs, tumor-derived EVs (TDEs) have both altered components and quantities of contents. TDEs have been shown to help facilitate an environment conducive to the occurrence and development of tumor by regulation of glucose, lipids and amino acids. Furthermore, TDEs can also affect the host metabolism and immune system. EVs have been shown to have multiple clinically useful properties, including the use of TDEs as biomarkers for the early diagnosis of diseases and using the transport properties of exosomes for drug delivery. Targeting the key bioactive cargoes of exosomes could be applied to provide new strategies for the treatment of tumors. In this review, we summarize the finding of studies focused on measuring the effects of TDE on tumor-related microenvironment and systemic metabolism. Video Abstract.
Collapse
Affiliation(s)
- Xiaoyu Fu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei China
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei China
| | - Junlong Song
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei China
| | - Wei Yan
- School of Life Science, Wuhan University, Wuhan, 430072 Hubei China
| | - Bradley M. Downs
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei China
| | - Juanjuan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei China
| |
Collapse
|
31
|
Natalia A, Zhang L, Sundah NR, Zhang Y, Shao H. Analytical device miniaturization for the detection of circulating biomarkers. NATURE REVIEWS BIOENGINEERING 2023; 1:1-18. [PMID: 37359772 PMCID: PMC10064972 DOI: 10.1038/s44222-023-00050-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 06/28/2023]
Abstract
Diverse (sub)cellular materials are secreted by cells into the systemic circulation at different stages of disease progression. These circulating biomarkers include whole cells, such as circulating tumour cells, subcellular extracellular vesicles and cell-free factors such as DNA, RNA and proteins. The biophysical and biomolecular state of circulating biomarkers carry a rich repertoire of molecular information that can be captured in the form of liquid biopsies for disease detection and monitoring. In this Review, we discuss miniaturized platforms that allow the minimally invasive and rapid detection and analysis of circulating biomarkers, accounting for their differences in size, concentration and molecular composition. We examine differently scaled materials and devices that can enrich, measure and analyse specific circulating biomarkers, outlining their distinct detection challenges. Finally, we highlight emerging opportunities in biomarker and device integration and provide key future milestones for their clinical translation.
Collapse
Affiliation(s)
- Auginia Natalia
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Li Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Noah R. Sundah
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Yan Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
32
|
Xu X, Wang Q, Qian X, Wu Y, Wang J, Li J, Li Y, Zhang Z. Spatial-Drug-Laden Protease-Activatable M1 Macrophage System Targets Lung Metastasis and Potentiates Antitumor Immunity. ACS NANO 2023; 17:5354-5372. [PMID: 36877635 DOI: 10.1021/acsnano.2c08834] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Lung metastasis is a critical cause of cancer mortality and its therapy is largely challenged by the limited drug delivery efficiency and robust immunosuppression in metastatic tumors. Herein, we designed a spatial-drug-laden M1 macrophage system with liposomal R848 inside and fibroblast activation protein protease (FAP)-sensitive phospholipid-DM4 conjugate on the membrane of M1 macrophage (RDM). RDM could preferentially accumulate at the metastatic lesions in lungs and responsively release the therapeutic agents as free drug molecules or drug-loaded nanovesicles. RDM treatment notably enhanced the infiltration of CD3+CD8+ T cells to lung metastasis and, respectively, caused an 8.54-, 12.87- and 2.85-fold improvement of the granzyme-B-, interferon-γ-, and Ki67-positive subtypes versus negative control. Moreover, RDM treatment produced a 90.99% inhibition of lung metastasis in 4T1 models and significant prolongation of survival in three murine lung metastatic models. Therefore, the drug-laden FAP-sensitive M1 macrophage system represents a feasible strategy to target lung metastasis and boost antitumor immunity for antimetastasis therapy.
Collapse
Affiliation(s)
- Xiaoxuan Xu
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiang Wang
- Department of Cardiothoracic Surgery, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 200125, China
| | - Xindi Qian
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Wu
- School of Pharmacy and Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai 201203, China
| | - Jiaoying Wang
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiwen Zhang
- School of Pharmacy and Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai 201203, China
| |
Collapse
|
33
|
Wang M, Zhang Y. Activatable molecular fluorescence probes for the imaging and detection of ischemic stroke. BRAIN SCIENCE ADVANCES 2023. [DOI: 10.26599/bsa.2023.9050003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
The real-time, noninvasive, nonionizing, high spatiotemporal resolution, and flexibility characteristics of molecular fluorescence imaging provide a uniquely powerful approach to imaging and monitoring the physiology and pathophysiology of ischemic stroke. Currently, various fluorescence probes have been synthesized with the aim of improving quantitative and quantitative studies of the pathologic processes of ischemic stroke in living animals. In this review, we present an overview of current activatable fluorescence probes for the imaging and diagnosis of ischemic stroke in animal models. We categorize the probes based on their activatable signals from the biomarkers associated with ischemic stroke, and we present representative examples of their functional mechanisms. Finally, we briefly discuss future perspectives in this field.
Collapse
Affiliation(s)
- Mengdie Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yan Zhang
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| |
Collapse
|
34
|
Zhao H, Pan S, Natalia A, Wu X, Ong CAJ, Teo MCC, So JBY, Shao H. A hydrogel-based mechanical metamaterial for the interferometric profiling of extracellular vesicles in patient samples. Nat Biomed Eng 2023; 7:135-148. [PMID: 36303008 DOI: 10.1038/s41551-022-00954-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 09/15/2022] [Indexed: 11/09/2022]
Abstract
The utility of mechanical metamaterials for biomedical applications has seldom been explored. Here we show that a metamaterial that is mechanically responsive to antibody-mediated biorecognition can serve as an optical interferometric mask to molecularly profile extracellular vesicles in ascites fluid from patients with cancer. The metamaterial consists of a hydrogel responsive to temperature and redox activity functionalized with antibodies to surface biomarkers on extracellular vesicles, and is patterned into micrometric squares on a gold-coated glass substrate. Through plasmonic heating, the metamaterial is maintained in a transition state between a relaxed form and a buckled state. Binding of extracellular vesicles from the patient samples to the antibodies on the hydrogel causes it to undergo crosslinking, induced by free radicals generated via the activity of horseradish peroxidase conjugated to the antibodies. Hydrogel crosslinking causes the metamaterial to undergo fast chiral re-organization, inducing amplified changes in its mechanical deformation and diffraction patterns, which are detectable by a smartphone camera. The mechanical metamaterial may find broad utility in the sensitive optical immunodetection of biomolecules.
Collapse
Affiliation(s)
- Haitao Zhao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Sijun Pan
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Auginia Natalia
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Xingjie Wu
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Chin-Ann J Ong
- Division of Surgical Oncology, National Cancer Centre, Singapore, Singapore
| | - Melissa C C Teo
- Division of Surgical Oncology, National Cancer Centre, Singapore, Singapore
| | - Jimmy B Y So
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Division of Surgical Oncology, National University Cancer Institute, Singapore, Singapore
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore. .,Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore. .,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
35
|
Li J, Wu Y, Wang J, Xu X, Zhang A, Li Y, Zhang Z. Macrophage Membrane-Coated Nano-Gemcitabine Promotes Lymphocyte Infiltration and Synergizes AntiPD-L1 to Restore the Tumoricidal Function. ACS NANO 2023; 17:322-336. [PMID: 36525314 DOI: 10.1021/acsnano.2c07861] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The limited lymphocyte infiltration and exhaustion of tumoricidal functions in solid tumors remain a formidable obstacle to cancer immunotherapy. Herein, we designed a macrophage membrane-coated nano-gemcitabine system (MNGs) to promote lymphocyte infiltration and then synergized anti-programmed death ligand 1 (antiPD-L1) to reinvigorate the exhausted lymphocytes. MNGs exhibited effective intratumor-permeating and responsive drug-releasing capacity, produced notable elimination of versatile immunosuppressive cells, and promoted lymphocyte infiltration into cancer cell regions in tumors, but over 50% of these infiltrated lymphocytes were in the exhausted state. Compared with MNG monotherapy, the MNGs+antiPD-L1 combination produced 31.77% and 30.63% reduction of exhausted CD3+CD8+ T cells and natural killer (NK) cells and 2.83- and 3.17-fold increases of interferon-γ (IFN-γ)-positive subtypes, respectively, thereby resulting in considerable therapeutic benefits in several tumor models. Thus, MNGs provide an encouraging strategy to promote lymphocyte infiltration and synergize antiPD-L1 to restore their tumoricidal function for cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China
| | - Yao Wu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy & Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai 201203, China
| | - Jiaoying Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoxuan Xu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ao Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Zhiwen Zhang
- School of Pharmacy & Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai 201203, China
| |
Collapse
|
36
|
Zhang R, Gao J, Zhao G, Zhou L, Kong F, Jiang T, Jiang H. Tetrazine bioorthogonal chemistry makes nanotechnology a powerful toolbox for biological applications. NANOSCALE 2023; 15:461-469. [PMID: 36533721 DOI: 10.1039/d2nr06056f] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Bioorthogonal chemistry enables researchers to manipulate bioactive molecules in living systems. These highly selective and biocompatible reactions can be carried out in various complex environments. Over the past two decades, a considerable number of strides have been made to expand the capacities of bioorthogonal chemistry coupled with the aim to fine-tune present reactions for specific applications. The good points of bioorthogonal chemistry have pushed material chemists to integrate bioorthogonal chemistry with nanotechnologies to broaden the biological applications of nanomaterials. Notably, bioorthogonal nanotechnologies fundamentally rely on, more than half, according to our investigation, tetrazine bioorthogonal chemistry (TBC) to function as bioorthogonal handles to react with target agents owing to the extremely rapid kinetics and high selectivities of TBC. Its utilization in combination with nanotechnologies has led to developments in various areas of biomedicine, such as in situ drug activation and targeted delivery, bioimaging and biosensing, and the understanding of cell-biomolecule interactions. Given the fantastic past achievements and the rapid developments in tetrazine bioorthogonal technologies, the future is certainly very bright.
Collapse
Affiliation(s)
- Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
- Cancer Institute, Affiliated Hospital of Qingdao University, 266071, China
| | - Jiake Gao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Gaoxiang Zhao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
- Cancer Institute, Affiliated Hospital of Qingdao University, 266071, China
| | - Liman Zhou
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530006, China.
| | - Fandong Kong
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530006, China.
| | - Tao Jiang
- Key Laboratory of Marine Drugs Chinese Ministry of Education, Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
- Cancer Institute, Affiliated Hospital of Qingdao University, 266071, China
| |
Collapse
|
37
|
Zhang K, Li R, Chen X, Yan H, Li H, Zhao X, Huang H, Chen S, Liu Y, Wang K, Han Z, Han Z, Kong D, Chen X, Li Z. Renal Endothelial Cell-Targeted Extracellular Vesicles Protect the Kidney from Ischemic Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204626. [PMID: 36416304 PMCID: PMC9875634 DOI: 10.1002/advs.202204626] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/24/2022] [Indexed: 05/02/2023]
Abstract
Endothelial cell injury plays a critical part in ischemic acute kidney injury (AKI) and participates in the progression of AKI. Targeting renal endothelial cell therapy may ameliorate vascular injury and further improve the prognosis of ischemic AKI. Here, P-selectin as a biomarker of ischemic AKI in endothelial cells is identified and P-selectin binding peptide (PBP)-engineered extracellular vesicles (PBP-EVs) with imaging and therapeutic functions are developed. The results show that PBP-EVs exhibit a selective targeting tendency to injured kidneys, while providing spatiotemporal information for the early diagnosis of AKI by quantifying the expression of P-selectin in the kidneys by molecular imaging. Meanwhile, PBP-EVs reveal superior nephroprotective functions in the promotion of renal repair and inhibition of fibrosis by alleviating inflammatory infiltration, improving reparative angiogenesis, and ameliorating maladaptive repair of the renal parenchyma. In conclusion, PBP-EVs, as an ischemic AKI theranostic system that is designed in this study, provide a spatiotemporal diagnosis in the early stages of AKI to help guide personalized therapy and exhibit superior nephroprotective effects, offering proof-of-concept data to design EV-based theranostic strategies to promote renal recovery and further improve long-term outcomes following AKI.
Collapse
Affiliation(s)
- Kaiyue Zhang
- School of MedicineNankai UniversityTianjin300071China
- The Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Rongrong Li
- School of MedicineNankai UniversityTianjin300071China
- The Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Xiaoniao Chen
- Beijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical UniversityBeijing100730China
- State Key Laboratory of Kidney DiseasesChinese PLA General HospitalBeijing100853China
| | - Hongyu Yan
- The Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Huifang Li
- School of MedicineNankai UniversityTianjin300071China
| | - Xiaotong Zhao
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityXinxiangHenan453003China
| | - Haoyan Huang
- School of MedicineNankai UniversityTianjin300071China
| | - Shang Chen
- School of MedicineNankai UniversityTianjin300071China
| | - Yue Liu
- School of MedicineNankai UniversityTianjin300071China
| | - Kai Wang
- The Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Zhibo Han
- Jiangxi Engineering Research Center for Stem CellShangraoJiangxi334000China
- Tianjin Key Laboratory of Engineering Technologies for Cell PharmaceuticalNational Engineering Research Center of Cell ProductsAmCellGene Co., LtdTianjin300457China
| | - Zhong‐Chao Han
- Jiangxi Engineering Research Center for Stem CellShangraoJiangxi334000China
- Tianjin Key Laboratory of Engineering Technologies for Cell PharmaceuticalNational Engineering Research Center of Cell ProductsAmCellGene Co., LtdTianjin300457China
- Beijing Engineering Laboratory of Perinatal Stem CellsBeijing Institute of Health and Stem CellsHealth & Biotech CoBeijing100176China
| | - Deling Kong
- The Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Xiang‐Mei Chen
- State Key Laboratory of Kidney DiseasesChinese PLA General HospitalBeijing100853China
| | - Zongjin Li
- School of MedicineNankai UniversityTianjin300071China
- The Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
- State Key Laboratory of Kidney DiseasesChinese PLA General HospitalBeijing100853China
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityXinxiangHenan453003China
- Tianjin Key Laboratory of Human Development and Reproductive RegulationTianjin Central Hospital of Gynecology ObstetricsNankai University Affiliated Hospital of Obstetrics and GynecologyTianjin300100China
| |
Collapse
|
38
|
Qian X, Wang Y, Xie H, Wang C, Li J, Lei Y, Liu H, Wu Y, Li Y, Zhang Z. Bioinspired nanovehicle of furoxans-oxaliplatin improves tumoral distribution for chemo-radiotherapy. J Control Release 2023; 353:447-461. [PMID: 36470332 DOI: 10.1016/j.jconrel.2022.11.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/13/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
The spatiotemporal distribution of therapeutic agents in tumors remains an essential challenge of radiation-mediated therapy. Herein, we rationally designed a macrophage microvesicle-inspired nanovehicle of nitric oxide donor-oxaliplatin (FO) conjugate (M-PFO), aiming to promote intratumor permeation and distribution profiles for chemo-radiotherapy. FO was responsively released from M-PFO in intracellular acidic environments, and then be activated by glutathione (GSH) into active oxaliplatin and NO molecules in a programmed manner. M-PFO exhibited notable accumulation, permeation and cancer cell accessibility in tumor tissues. Upon radiation, the reactive peroxynitrite species (ONOO-) were largely produced, which could diffuse into regions over 400 μm away from the tumor vessels and be detectable after 24 h of radiation, thereby exhibiting superior efficacy in improving the spatiotemporal distribution in tumors versus common reactive oxygen species (ROS). Moreover, M-PFO mediated chemo-radiotherapy caused notable inhibition of tumor growth, with an 89.45% inhibition in HT-29 tumor models and a 92.69% suppression in CT-26 tumor models. Therefore, this bioinspired design provides an encouraging platform to improve intratumor spatiotemporal distribution to synergize chemo-radiotherapy.
Collapse
Affiliation(s)
- Xindi Qian
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuqi Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Honglei Xie
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong, 264000, China
| | - Chen Wang
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong, 264000, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ying Lei
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201203, China
| | - Huanzhen Liu
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201203, China
| | - Yao Wu
- School of Pharmacy& Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Zhiwen Zhang
- School of Pharmacy& Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai 201203, China; Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong, 264000, China.
| |
Collapse
|
39
|
Wu M, Wang M, Jia H, Wu P. Extracellular vesicles: emerging anti-cancer drugs and advanced functionalization platforms for cancer therapy. Drug Deliv 2022; 29:2513-2538. [PMID: 35915054 PMCID: PMC9347476 DOI: 10.1080/10717544.2022.2104404] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Increasing evidences show that unmodified extracellular vesicles (EVs) derived from various cells can effectively inhibit the malignant progression of different types of tumors by delivering the bioactive molecules. Therefore, EVs are expected to be developed as emerging anticancer drugs. Meanwhile, unmodified EVs as an advanced and promising nanocarrier that is frequently used in targeted delivery therapeutic cargos and personalized reagents for the treatment and diagnosis of cancer. To improve the efficacy of EV-based treatments, researchers are trying to engineering EVs as an emerging nanomedicine translational therapy platform through biological, physical and chemical approaches, which can be broaden and altered to enhance their therapeutic capability. EVs loaded with therapeutic components such as tumor suppressor drugs, siRNAs, proteins, peptides, and conjugates exhibit significantly enhanced anti-tumor effects. Moreover, the design and preparation of tumor-targeted modified EVs greatly enhance the specificity and effectiveness of tumor therapy, and these strategies are expected to become novel ideas for tumor precision medicine. This review will focus on reviewing the latest research progress of functionalized EVs, clarifying the superior biological functions and powerful therapeutic potential of EVs, for researchers to explore new design concepts based on EVs and build next-generation nanomedicine therapeutic platforms.
Collapse
Affiliation(s)
- Manling Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of UST C, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
- Anhui Provincial Children’s Hospital, Hefei, Anhui, P.R. China
| | - Min Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Haoyuan Jia
- Department of Clinical Laboratory, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, Jiangsu, P.R. China
| | - Peipei Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of UST C, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
- Anhui Provincial Children’s Hospital, Hefei, Anhui, P.R. China
| |
Collapse
|
40
|
Wei X, Cai L, Chen H, Shang L, Zhao Y, Sun W. Noninvasive Multiplexed Analysis of Bladder Cancer-Derived Urine Exosomes via Janus Magnetic Microspheres. Anal Chem 2022; 94:18034-18041. [PMID: 36519619 DOI: 10.1021/acs.analchem.2c04408] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Bladder cancer greatly endangers human health, and its early diagnosis is of vital importance. Exosomes, which contain proteins and nucleic acids related to their source cells, are expected to be an emerging biomarker for bladder cancer detection. Here, we propose a novel system for multiplexed analysis of bladder cancer-derived urine exosomes based on Janus magnetic microspheres as barcoded microcarriers. The microcarriers are constructed by droplet-templated coassembly of colloidal silica nanoparticles and magnetic nanoparticles under a magnetic field. The microcarriers possess one hemisphere with structural color and the other hemisphere with magneto-responsiveness. Benefiting from the unique structure, these Janus microcarriers could serve as barcodes and could move controllably in a sample solution, thus realizing the multiplex detection of exosomes with high sensitivity. Notably, the present platform is noninvasive since a urine specimen, as an ideal source of bladder cancer-derived exosomes, is employed as the sample solution. This feature, together with the good sensitivity, specificity, low sample consumption, and easy operation, indicates the great potential of the platform for bladder cancer diagnosis in clinical applications.
Collapse
Affiliation(s)
- Xiaowei Wei
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou325027, China.,Laboratory Medicine Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing210011, China
| | - Lijun Cai
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing210096, China
| | - Hanxu Chen
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing210096, China
| | - Luoran Shang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-Laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
| | - Yuanjin Zhao
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing210096, China
| | - Weijian Sun
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou325027, China
| |
Collapse
|
41
|
Yong T, Wei Z, Gan L, Yang X. Extracellular-Vesicle-Based Drug Delivery Systems for Enhanced Antitumor Therapies through Modulating the Cancer-Immunity Cycle. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201054. [PMID: 35726204 DOI: 10.1002/adma.202201054] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/10/2022] [Indexed: 06/15/2023]
Abstract
Although immunotherapy harnessing activity of the immune system against tumors has made great progress, the treatment efficacy remains limited in most cancers. Current anticancer immunotherapy is primarily based on T-cell-mediated cellular immunity, which highly relies on efficiency of triggering the cancer-immunity cycle, namely, tumor antigen release, antigen presentation by antigen presenting cells, T cell activation, recruitment and infiltration of T cells into tumors, and recognition and killing of tumor cells by T cells. Unfortunately, these immunotherapies are restricted by inefficient drug delivery and acting on only a single step of the cancer-immunity cycle. Due to high biocompatibility, low immunogenicity, intrinsic cell targeting, and easy chemical and genetic manipulation, extracellular vesicle (EV)-based drug delivery systems are widely used to amplify anticancer immune responses by serving as an integrated platform for multiple drugs or therapeutic strategies to synergistically activate several steps of cancer-immunity cycle. This review summarizes various mechanisms related to affecting cancer-immunity cycle disorders. Meanwhile, preparation and application of EV-based drug delivery systems in modulating cancer-immunity cycle are introduced, especially in the improvement of T cell recruitment and infiltration into tumors. Finally, opportunities and challenges of EV-based drug delivery systems in translational clinical applications are briefly discussed.
Collapse
Affiliation(s)
- Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
42
|
Sun K, Zheng X, Jin H, Yu F, Zhao W. Exosomes as CNS Drug Delivery Tools and Their Applications. Pharmaceutics 2022; 14:pharmaceutics14102252. [PMID: 36297688 PMCID: PMC9609403 DOI: 10.3390/pharmaceutics14102252] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Central nervous system (CNS) diseases threaten the health of people all over the world. However, due to the structural and functional particularities of the brain and spinal cord, CNS-targeted drug development is rather challenging. Exosomes are small cellular vesicles with lipid bilayers that can be secreted by almost all cells and play important roles in intercellular communication. The advantages of low immunogenicity, the ability to cross the blood-brain barrier, and the flexibility of drug encapsulation make them stand out among CNS drug delivery tools. Herein, we reviewed the research on exosomes in CNS drug delivery over the past decade and outlined the impact of the drug loading mode, administration route, and engineered modification on CNS targeting. Finally, we highlighted the problems and prospects of exosomes as CNS drug delivery tools.
Collapse
Affiliation(s)
- Ke Sun
- College of Pharmacy, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Xue Zheng
- College of Pharmacy, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Hongzhen Jin
- College of Pharmacy, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
- Correspondence: (H.J.); (F.Y.)
| | - Fan Yu
- College of Life Sciences, Nankai University, Weijin Road, Nankai District, Tianjin 300350, China
- Correspondence: (H.J.); (F.Y.)
| | - Wei Zhao
- College of Pharmacy, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| |
Collapse
|
43
|
Extracellular vesicle isolation, purification and evaluation in cancer diagnosis. Expert Rev Mol Med 2022; 24:e41. [PMID: 36268744 DOI: 10.1017/erm.2022.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Strategies for non-invasive biomarker discovery in early detection of cancer are an urgent need. Extracellular vesicles (EVs) have generated increasing attention from the scientific community and are under intensive investigations due to their unique biological profiles and their non-invasive nature. EVs are membrane-enclosed vesicles with variable sizes and function. Such vesicles are actively secreted from multiple cell types and are considered as key vehicles for inter-cellular communications and signalling. The stability and potential to easily cross biological barriers enable EVs for exerting durable effects on target cells. These along with easy access to such vesicles, the consistent secretion from tumour during all stages of tumorigenesis and their content providing a reservoir of molecules as well as mirroring the identity of the cell of origin are virtues that have made EVs appealing to be assessed in liquid biopsy approaches and for using as a promising resource of biomarkers in cancer diagnosis and therapy and monitoring targeted cancer therapy. Early detection of EVs will guide time-scheduled personalised therapy. Surveying reliable and sensitive methods for rapid isolation of EVs from biofluids, the purity of isolated vesicles and their molecular profiling and marker specification for clinical translation in patients with cancer are issues in the area and the hot topics of many recent studies. Here, the focus is over methods for EV isolation and stratification for digging more information about liquid biopsy-based diagnosis. Extending knowledge regarding EV-based strategies is a key to validate independent patient follow-up for cancer diagnosis at early stages and inspecting the efficacy of therapeutics.
Collapse
|
44
|
Liu Z, Li T, Wang Z, Liu J, Huang S, Min BH, An JY, Kim KM, Kim S, Chen Y, Liu H, Kim Y, Wong DT, Huang TJ, Xie YH. Gold Nanopyramid Arrays for Non-Invasive Surface-Enhanced Raman Spectroscopy-Based Gastric Cancer Detection via sEVs. ACS APPLIED NANO MATERIALS 2022; 5:12506-12517. [PMID: 36185166 PMCID: PMC9513748 DOI: 10.1021/acsanm.2c01986] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/12/2022] [Indexed: 05/05/2023]
Abstract
Gastric cancer (GC) is one of the most common and lethal types of cancer affecting over one million people, leading to 768,793 deaths globally in 2020 alone. The key for improving the survival rate lies in reliable screening and early diagnosis. Existing techniques including barium-meal gastric photofluorography and upper endoscopy can be costly and time-consuming and are thus impractical for population screening. We look instead for small extracellular vesicles (sEVs, currently also referred as exosomes) sized ⌀ 30-150 nm as a candidate. sEVs have attracted a significantly higher level of attention during the past decade or two because of their potentials in disease diagnoses and therapeutics. Here, we report that the composition information of the collective Raman-active bonds inside sEVs of human donors obtained by surface-enhanced Raman spectroscopy (SERS) holds the potential for non-invasive GC detection. SERS was triggered by the substrate of gold nanopyramid arrays we developed previously. A machine learning-based spectral feature analysis algorithm was developed for objectively distinguishing the cancer-derived sEVs from those of the non-cancer sub-population. sEVs from the tissue, blood, and saliva of GC patients and non-GC participants were collected (n = 15 each) and analyzed. The algorithm prediction accuracies were reportedly 90, 85, and 72%. "Leave-a-pair-of-samples out" validation was further performed to test the clinical potential. The area under the curve of each receiver operating characteristic curve was 0.96, 0.91, and 0.65 in tissue, blood, and saliva, respectively. In addition, by comparing the SERS fingerprints of individual vesicles, we provided a possible way of tracing the biogenesis pathways of patient-specific sEVs from tissue to blood to saliva. The methodology involved in this study is expected to be amenable for non-invasive detection of diseases other than GC.
Collapse
Affiliation(s)
- Zirui Liu
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| | - Tieyi Li
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| | - Zeyu Wang
- Department
of Mechanical Engineering and Material Science, Duke University, Durham, North Carolina 27708, United States
| | - Jun Liu
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| | - Shan Huang
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| | - Byoung Hoon Min
- Department
of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | - Ji Young An
- Department
of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | - Kyoung Mee Kim
- Department
of Pathology and Translational Genomics, Sungkyunkwan University School
of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | - Sung Kim
- Department
of Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | - Yiqing Chen
- Department
of Bioengineering, University of California, Riverside, Riverside, California 92521, United States
| | - Huinan Liu
- Department
of Bioengineering, University of California, Riverside, Riverside, California 92521, United States
| | - Yong Kim
- UCLA
School of Dentistry, 10833 Le Conte Ave. Box 951668, Los Angeles, California 90095-1668, United States
| | - David T.W. Wong
- UCLA
School of Dentistry, 10833 Le Conte Ave. Box 951668, Los Angeles, California 90095-1668, United States
| | - Tony Jun Huang
- Department
of Mechanical Engineering and Material Science, Duke University, Durham, North Carolina 27708, United States
| | - Ya-Hong Xie
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
45
|
Słomka A, Wang B, Mocan T, Horhat A, Willms AG, Schmidt-Wolf IGH, Strassburg CP, Gonzalez-Carmona MA, Lukacs-Kornek V, Kornek MT. Extracellular Vesicles and Circulating Tumour Cells - complementary liquid biopsies or standalone concepts? Theranostics 2022; 12:5836-5855. [PMID: 35966579 PMCID: PMC9373826 DOI: 10.7150/thno.73400] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/06/2022] [Indexed: 12/11/2022] Open
Abstract
Liquid biopsies do promise a lot, but are they keeping it? In the past decade, additional novel biomarkers qualified to be called like that, of which, some took necessary hurdles resulting in FDA approval and clinical use. Some others are since a while around, well known and were once regarded to be a game changer in cancer diagnosis or cancer screening. But, during their clinical use limitations were observed from statistical significance and questions raised regarding their robustness, that eventually led to be dropped from associated clinical guidelines for certain applications including cancer diagnosis. The purpose of this review isn't to give a broad overview of all current liquid biopsy as biomarkers, weight them and promise a brighter future in cancer prevention, but rather to take a deeper look on two of those who do qualify to be called liquid biopsies now or then. These two are probably of greatest interest conceptually and methodically, and likely have the highest chances to be in clinical use soon, with a portfolio extension over their original conceptual usage. We aim to dig deeper beyond cancer diagnosis or cancer screening. Actually, we aim to review in depth extracellular vesicles (EVs) and compare with circulating tumour cells (CTCs). The latter methodology is partially FDA approved and in clinical use. We will lay out similarities as taking advantage of surface antigens on EVs and CTCs in case of characterization and quantification. But drawing readers' attention to downstream application based on capture/isolation methodology and simply on their overall nature, here apparently being living material eventually recoverable as CTCs are vs. dead material with transient effects on recipient cell as in case of EVs. All this we try to bring in perspective, compare and conclude towards which future direction we are aiming for, or should aim for. Do we announce a winner between CTCs vs EVs? No, but we provide good reasons to intensify research on them.
Collapse
Affiliation(s)
- Artur Słomka
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, 85-067 Bydgoszcz, Poland
| | - Bingduo Wang
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany.,Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Tudor Mocan
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Adelina Horhat
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Arnulf G Willms
- Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany.,Department of General, Visceral and Vascular Surgery, German Armed Forces Hospital Hamburg, 22049 Hamburg, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Christian P Strassburg
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Maria A Gonzalez-Carmona
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Veronika Lukacs-Kornek
- Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Miroslaw T Kornek
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| |
Collapse
|
46
|
Deng J, Zhao S, Li J, Cheng Y, Liu C, Liu Z, Li L, Tian F, Dai B, Sun J. One-Step Thermophoretic AND Gate Operation on Extracellular Vesicles Improves Diagnosis of Prostate Cancer. Angew Chem Int Ed Engl 2022; 61:e202207037. [PMID: 35749531 DOI: 10.1002/anie.202207037] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Indexed: 01/19/2023]
Abstract
Circulating extracellular vesicles (EVs) have emerged as a valuable source of cancer biomarkers. However, the high degree of EV heterogeneity and the complexity of clinical samples pose a challenge in the sensitive identification of tumor-derived EVs. Here we introduce a one-step thermophoretic AND gate operation (Tango) assay that integrates polyethylene glycol (PEG)-enhanced thermophoretic accumulation of EVs and simultaneous AND gate operation on EV membranes by dual-aptamers recognition. By using the Tango assay to detect tumor-derived EVs with co-presence of EpCAM and PSMA directly from serum in a homogeneous, separation-free format, we can discriminate prostate cancer (PCa) patients from benign prostatic hyperplasia (BPH) patients in the diagnostic gray zone with an accuracy of 91 % in 15 min. Our approach streamlines EV enrichment and AND gate operation on EVs in a single assay, providing a rapid, straightforward, and powerful method for precise and non-invasive diagnosis of cancer.
Collapse
Affiliation(s)
- Jinqi Deng
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuai Zhao
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junhong Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yangchang Cheng
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chao Liu
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zheng Liu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lele Li
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei Tian
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jiashu Sun
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
47
|
Ren Z, Zhang Z, Wei J, Dong B, Lee C. Wavelength-multiplexed hook nanoantennas for machine learning enabled mid-infrared spectroscopy. Nat Commun 2022; 13:3859. [PMID: 35790752 PMCID: PMC9256719 DOI: 10.1038/s41467-022-31520-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 06/03/2022] [Indexed: 12/19/2022] Open
Abstract
Infrared (IR) plasmonic nanoantennas (PNAs) are powerful tools to identify molecules by the IR fingerprint absorption from plasmon-molecules interaction. However, the sensitivity and bandwidth of PNAs are limited by the small overlap between molecules and sensing hotspots and the sharp plasmonic resonance peaks. In addition to intuitive methods like enhancement of electric field of PNAs and enrichment of molecules on PNAs surfaces, we propose a loss engineering method to optimize damping rate by reducing radiative loss using hook nanoantennas (HNAs). Furthermore, with the spectral multiplexing of the HNAs from gradient dimension, the wavelength-multiplexed HNAs (WMHNAs) serve as ultrasensitive vibrational probes in a continuous ultra-broadband region (wavelengths from 6 μm to 9 μm). Leveraging the multi-dimensional features captured by WMHNA, we develop a machine learning method to extract complementary physical and chemical information from molecules. The proof-of-concept demonstration of molecular recognition from mixed alcohols (methanol, ethanol, and isopropanol) shows 100% identification accuracy from the microfluidic integrated WMHNAs. Our work brings another degree of freedom to optimize PNAs towards small-volume, real-time, label-free molecular recognition from various species in low concentrations for chemical and biological diagnostics. Infrared spectroscopy with plasmonic nanoantennas is limited by small overlap between molecules and hot spots, and sharp resonance peaks. The authors demonstrate spectral multiplexing of hook nanoantennas with gradient dimensions as ultrasensitive vibrational probes in a continuous ultra-broadband region and utilize machine learning for enhanced sensing performance.
Collapse
|
48
|
Bioprobes-regulated precision biosensing of exosomes: From the nanovesicle surface to the inside. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
49
|
Fan Z, Jiang C, Wang Y, Wang K, Marsh J, Zhang D, Chen X, Nie L. Engineered extracellular vesicles as intelligent nanosystems for next-generation nanomedicine. NANOSCALE HORIZONS 2022; 7:682-714. [PMID: 35662310 DOI: 10.1039/d2nh00070a] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Extracellular vesicles (EVs), as natural carriers of bioactive cargo, have a unique micro/nanostructure, bioactive composition, and characteristic morphology, as well as fascinating physical, chemical and biochemical features, which have shown promising application in the treatment of a wide range of diseases. However, native EVs have limitations such as lack of or inefficient cell targeting, on-demand delivery, and therapeutic feedback. Recently, EVs have been engineered to contain an intelligent core, enabling them to (i) actively target sites of disease, (ii) respond to endogenous and/or exogenous signals, and (iii) provide treatment feedback for optimal function in the host. These advances pave the way for next-generation nanomedicine and offer promise for a revolution in drug delivery. Here, we summarise recent research on intelligent EVs and discuss the use of "intelligent core" based EV systems for the treatment of disease. We provide a critique about the construction and properties of intelligent EVs, and challenges in their commercialization. We compare the therapeutic potential of intelligent EVs to traditional nanomedicine and highlight key advantages for their clinical application. Collectively, this review aims to provide a new insight into the design of next-generation EV-based theranostic platforms for disease treatment.
Collapse
Affiliation(s)
- Zhijin Fan
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P. R. China
| | - Cheng Jiang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Yichao Wang
- Department of Clinical Laboratory Medicine, Tai Zhou Central Hospital (Taizhou University Hospital), Taizhou 318000, P. R. China
| | - Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Jade Marsh
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an 710049, P. R. China.
| | - Liming Nie
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P. R. China
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
| |
Collapse
|
50
|
Li H, Huang T, Lu L, Yuan H, Zhang L, Wang H, Yu B. Ultrasensitive Detection of Exosomes Using an Optical Microfiber Decorated with Plasmonic MoSe 2-Supported Gold Nanorod Nanointerfaces. ACS Sens 2022; 7:1926-1935. [DOI: 10.1021/acssensors.2c00598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Hongtao Li
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
- Guangxi Key Laboratory of Nuclear Physics and Nuclear Technology, College of Physics Science and Technology, Guangxi Normal University, Guilin 541004, People’s Republic of China
| | - Tianqi Huang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| | - Liang Lu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| | - Hao Yuan
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| | - Lei Zhang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| | - Hongzhi Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230031, People’s Republic of China
- Institute of Urology, Anhui Medical University Hefei, 230031, People’s Republic of China
| | - Benli Yu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| |
Collapse
|