1
|
He S, Yu J, Cheng P, Liu J, Zhang C, Xu C, Pu K, Zhang Y. Differential Optical Imaging of Antigen Presentation Machinery Using Molecular Optical Reporters. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2420393. [PMID: 40370186 DOI: 10.1002/adma.202420393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/15/2025] [Indexed: 05/16/2025]
Abstract
Detection of antigen presentation is central to understanding immunological processes and developing therapeutics for cancer, infectious diseases, and allergies. However, methods with the ability to dynamically and noninvasively distinguish between major histocompatibility complex class I (MHC-I) and MHC-II antigen presentations remain lacking. Herein, we develop activatable molecular optical reporters (MORs) for real-time differential imaging of antigen presentations in lymph nodes (LNs). These MORs are engineered to passively target LNs and activated through proteolytic cleavage by key enzymes in the MHC-I and MHC-II pathways, the immunoproteasome (iP) and cathepsin S (CTSS), respectively, triggering their chemiluminescent or fluorescent signals. Coupled with minimized signal crosstalk and high sensitivity, MORs delineate the subtle differences in the antigen presentation machinery across various disease models, including cancer and bacterial or viral infection, a feat unattainable for existing imaging methods. After systemic administration, MORs also allow real-time visualization of antigen presentation in the tumor microenvironment. Besides, MORs are validated to have potential for preclinical application in immunotherapeutics screening and clinical application in tissue biopsy. Thus, our study not only presents the first example of real-time, in vivo differential imaging of antigen presentation pathways but also opens new avenues for optical probes in immune contexture analysis.
Collapse
Affiliation(s)
- Shasha He
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
- College of Chemistry and Chemical Engineering, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, P. R. China
| | - Jie Yu
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Chi Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Yan Zhang
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
2
|
Dai J, Wei W, Yan C, Ji DK, Liu C, Huang J, Liang C, Liu J, Guo Z, Zhu WH. Multiplex imaging of amyloid-β plaques dynamics in living brains with quinoline-malononitrile-based probes. Nat Biomed Eng 2025:10.1038/s41551-025-01392-x. [PMID: 40360763 DOI: 10.1038/s41551-025-01392-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 04/03/2025] [Indexed: 05/15/2025]
Abstract
The dynamic behaviour of amyloid-β (Aβ) plaques in Alzheimer's disease remains poorly understood, and accumulation and distribution of Aβ plaques must be inferred from in vitro pathological changes in brain tissue. In situ detection of Aβ plaques in live imaging is challenging because of the lack of adequate probes. Here we report the design of unimolecular quinoline-malononitrile-based Aβ probes, termed QMFluor integrative framework, that binds in vivo to Aβ plaques, making them detectable via near-infrared fluorescence imaging, magnetic resonance imaging, positron emission tomography and computed tomography. QMFluor probes are permeable to the blood-brain barrier, and, upon systematic injection, enable real-time magnetic resonance imaging and positron emission tomography-computed tomography imaging of the Aβ biodistribution in the hippocampus and cerebral cortex, and accurately differentiate the brains of living Alzheimer's disease mouse models from wild-type controls. We further demonstrate the ability of QMFluor probes to reach the brain after intravenous injection in a large animal model. This strategy expands the toolbox of probes for in vivo visualization of amyloids in Alzheimer's disease pathological analysis, drug screening and clinical applications.
Collapse
Affiliation(s)
- Jianfeng Dai
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenxu Yan
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China.
| | - Ding-Kun Ji
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Caiqi Liu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Jialiang Huang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Chenyi Liang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiqian Guo
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China.
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.
| | - Wei-Hong Zhu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China.
- Center of Photosensitive Chemicals Engineering, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
3
|
Yi L, Liu Y, Wan C, Li S, Zhou M, Qi F, Xie H, Wang X, Su Y, Du W, Feng X, Li Y, Liu BF, Chen P. Programmable Manually Powered Microfluidics for Rapid Point-of-Care Diagnosis of Urinary Tract Infection. Anal Chem 2025; 97:9480-9491. [PMID: 40268684 DOI: 10.1021/acs.analchem.5c00847] [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: 04/25/2025]
Abstract
Point-of-care testing (POCT) for urinary tract infection (UTI) holds significant importance in the field of disease prevention and control, as well as the advancement of personalized precision medicine. However, conventional methods for detecting UTIs continue to face challenges such as time-consuming and labor-intensive detection processes, and reliance on specialized equipment and personnel rendering them unsuitable for point-of-care applications, especially in resource-limited areas. Here, we propose a novel flexible programmable manually powered microfluidic (FPM) for rapid point-of-care diagnosis of UTIs. For the first time, the proposed FPMs was achieved through a combined strategy of laser printing, cutting, and laminating, with the entire process completed in under 15 min at a cost of less than $0.5, which effectively circumvent the traditionally time-consuming and labor-intensive soft lithography techniques. By incorporating a modular structure-based design concept, we successfully developed various types of portable FPMs with functionalities including parallel pumping, simultaneous releasing, quantitative dispensing, sequential releasing, cyclic motion of multiple liquids and concentration gradient generating. As a proof-of-concept demonstration, we initially employed a high-throughput parallel dispensing design to analyze six urinary biochemical markers within 1 min, presenting potential applicability for future at-home testing. We then integrated a manually powered concentration gradient generator with spatial confinement signal enhancement to enable rapid phenotypic antimicrobial susceptibility testing (AST) within three to 5 h, while achieving clinical diagnostic accuracy rates of up to 95.56%. Therefore, our proposed FPMs eliminate the need for external pumps or actuators and could serve as an affordable hand-held POCT tool for UTI diagnosis. Moreover, in resource-poor areas, they have potential utility as robust POCT devices addressing diverse rapid detection needs.
Collapse
Affiliation(s)
- Longyu Yi
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuqi Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chao Wan
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shunji Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mengfan Zhou
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fukang Qi
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Han Xie
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xin Wang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yixiao Su
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wei Du
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaojun Feng
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
4
|
Zhang J, Luo H, Ma W, Lv J, Wang B, Sun F, Chi W, Fang Z, Yang Z. Semiconducting Open-Shell Radicals for Precise Tumor Activatable Phototheranostics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500293. [PMID: 40051340 PMCID: PMC12061280 DOI: 10.1002/advs.202500293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/12/2025] [Indexed: 05/10/2025]
Abstract
Semiconducting open-shell radicals (SORs) have promising potential for the development of phototheranostic agents, enabling tumor bioimaging and boosting tumorous reactive oxygen species (ROS). Herein, a new class of semiconducting perylene diimide (PDI), designated as PDI(Br)n with various numbers of bromine (Br) atoms modified on PDI's bay/ortho positions is reported. PDI(Br)n is demonstrated to transform into a radical anion, [PDI(Br)n]•-, in a reducing solution, with a typical g-value of 2.0022. Specifically, [PDI(Br)4/6]•- is generated in the weakly reductive tumor-mimicking solution and exhibits high stability in air. Quantum chemical kinetic simulation and ultrafast femtosecond transient absorption spectroscopy indicate that [PDI(Br)6]•- has a low π-π stacking energy (0.35 eV), a fast electron transfer rate (192.4 ps) and energy gap of PDI(Br)6 (ΔES1, T1 = 1.307 eV, ΔES1, T2 = 0.324 eV) respectively, which together result in excited-state charge transfer characters. The PDI(Br)6 nanoparticle radicals, [PDI(Br)6] NPs•-, specifically enable chemodynamic and type-I photodynamic ROS generation in tumors, including superoxide and hydroxyl radicals, which elicit immunogenic cell death effect. Also, [PDI(Br)6] NPs•- facilitate activatable bioimaging-guided therapy due to their photoacoustic signal at 808 nm and NIR-II emission at 1115 nm. The work paves the way for the design of SORs for precise cancer theranostics.
Collapse
Affiliation(s)
- Jie Zhang
- Strait Laboratory of Flexible Electronics (SLoFE)Fujian Key Laboratory of Flexible ElectronicsStrait Institute of Flexible Electronics (Future Technologies)Fujian Normal UniversityFuzhou350117China
| | - Haifen Luo
- Strait Laboratory of Flexible Electronics (SLoFE)Fujian Key Laboratory of Flexible ElectronicsStrait Institute of Flexible Electronics (Future Technologies)Fujian Normal UniversityFuzhou350117China
| | - Wen Ma
- Strait Laboratory of Flexible Electronics (SLoFE)Fujian Key Laboratory of Flexible ElectronicsStrait Institute of Flexible Electronics (Future Technologies)Fujian Normal UniversityFuzhou350117China
| | - Jingqi Lv
- Strait Laboratory of Flexible Electronics (SLoFE)Fujian Key Laboratory of Flexible ElectronicsStrait Institute of Flexible Electronics (Future Technologies)Fujian Normal UniversityFuzhou350117China
| | - Bo Wang
- Strait Laboratory of Flexible Electronics (SLoFE)Fujian Key Laboratory of Flexible ElectronicsStrait Institute of Flexible Electronics (Future Technologies)Fujian Normal UniversityFuzhou350117China
| | - Fengwei Sun
- Strait Laboratory of Flexible Electronics (SLoFE)Fujian Key Laboratory of Flexible ElectronicsStrait Institute of Flexible Electronics (Future Technologies)Fujian Normal UniversityFuzhou350117China
| | - Weijie Chi
- School of Chemistry and Chemical EngineeringHainan UniversityHaikou570228China
| | - Zhuting Fang
- Department of Oncology and Vascuar Interventonal TherapyClinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhou350014China
- Department of imterventional Radiologyshengli Clinical Medical College of Fujian Medical UniversityFuijian Provincial HospitalFuzhou University Affliated Provincial HospitalFuzhou350001China
| | - Zhen Yang
- Strait Laboratory of Flexible Electronics (SLoFE)Fujian Key Laboratory of Flexible ElectronicsStrait Institute of Flexible Electronics (Future Technologies)Fujian Normal UniversityFuzhou350117China
| |
Collapse
|
5
|
Lin Y, Huang J, Pu K. Near-Infrared Chemiluminescent Theranostics. Angew Chem Int Ed Engl 2025; 64:e202501681. [PMID: 40051035 DOI: 10.1002/anie.202501681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Molecular chemiluminescence probes with near-infrared (NIR) emission offer promising benefits in deciphering complex pathological processes in a living system, as NIR chemiluminescence minimizes autofluorescence, enhances deep-tissue penetration, and improves signal-to-noise ratio. Molecular engineering using single-luminophore design and dual-luminophore design with intramolecular energy transfer provides ways to develop conventional chemiluminophore scaffolds into NIR chemiluminescence probes with ideal chemiluminescence quantum yield and half-life. By virtue of the structural diversity, 1,2-dioxetane-based NIR chemiluminophores with biomarker activity have been developed. This review summarizes the molecular design strategies of NIR chemiluminescence theranostic probes (NCTPs), followed by introducing activatable NCTPs with their biomedical applications for disease theranostics. Lastly, future perspectives and potential challenges of NIR chemiluminescence imaging in preclinical research and clinical translational potential are discussed.
Collapse
Affiliation(s)
- Youshi Lin
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Jingsheng Huang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| |
Collapse
|
6
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
7
|
Ou X, Chen P, Liu BF. Liquid Biopsy on Microfluidics: From Existing Endogenous to Emerging Exogenous Biomarkers Analysis. Anal Chem 2025; 97:8625-8640. [PMID: 40247704 DOI: 10.1021/acs.analchem.4c05407] [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: 04/19/2025]
Abstract
Liquid biopsy is an appealing approach for early diagnosis and assessment of treatment efficacy in cancer. Typically, liquid biopsy involves the detection of endogenous biomarkers, including circulating tumor cells (CTCs), extracellular vesicles (EVs), circulating tumor DNA (ctDNA), circulating tumor RNA (ctRNA), and proteins. The levels of these endogenous biomarkers are higher in cancer patients compared to those in healthy individuals. However, the clinical application of liquid biopsy using endogenous biomarker analysis faces challenges due to its low abundance and poor stability in circulation. Recently, a promising strategy involving the engineering of exogenous probes has been developed to overcome these limitations. These exogenous probes are activated within the tumor microenvironment, generating distinct exogenous markers that can be easily distinguished from background biological signals. Alternatively, these exogenous probes can be labeled with intrinsic endogenous biomarkers in vivo and detected in vitro after metabolic processes. In this review, we primarily focus on microfluidic-based liquid biopsy techniques that allow for the transition from analyzing existing endogenous biomarkers to emerging exogenous ones. First, we introduce common endogenous biomarkers, as well as synthetic exogenous ones. Next, we discuss recent advancements in microfluidic-based liquid biopsy techniques for analyzing both existing endogenous and emerging exogenous biomarkers. Lastly, we provide insights into future directions for liquid biopsy on microfluidic systems.
Collapse
Affiliation(s)
- Xiaowen Ou
- Hubei Key Laboratory of Purification and Application of Plant Anti-Cancer Active Ingredients, Department of Chemistry and Life Science, Hubei University of Education, Wuhan, 430205, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
8
|
Fan Y, Pei Y, Hu D, Wu Y, Sun K, Chen L, Yin J, Yan W, Shi M, Feng W, Liu X, Li F. A Lifetime Nanosensor for In Vivo pH Quantitative Imaging and Monitoring. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2502806. [PMID: 40269572 DOI: 10.1002/smll.202502806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Non-invasive, in vivo quantitative imaging for long-term biomarker monitoring is crucial for elucidating disease mechanisms, advancing precision medicine, and transforming diagnostics and therapeutic strategies. However, developing chemical sensors for sustained in vivo quantitative monitoring despite sensor concentration fluctuations, excitation variability, and tissue interference remains a major challenge. Here, a long-lifetime nanosensor based on a lanthanide-dye nanocomposite is presented that overcomes these limitations, enabling precise quantitative in vivo pH monitoring. Benefiting from a 64-fold reversible change in the dye's molar extinction coefficient, this nanosensor enables the dynamic tuning of reversible non-radiative energy transfer (RNET) efficiency (6.42%-35.23%) and luminescence lifetime (265-383 µs). This nanosensor enables 4 h of monitoring of gastrointestinal pH dynamics in mice following proton pump inhibitor (PPI) administration, offering new insights into pharmacodynamic effects across different administration routes and dosages and inter-individual variability in drug efficacy. Moreover, coordination with lanthanide nanocrystals induces a significant shift in the dye's pKa, highlighting the importance of nanomaterial interface engineering. This work establishes a versatile platform for in vivo diagnostics and therapeutic monitoring, marking a significant step forward in precision medicine.
Collapse
Affiliation(s)
- Yiwei Fan
- Academy for Engineering and Technology, Fudan University, Shanghai, 200433, China
| | - Yuetian Pei
- Academy for Engineering and Technology, Fudan University, Shanghai, 200433, China
| | - Donghao Hu
- School of Chemistry and Chemical Engineering & Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yukai Wu
- School of Chemistry and Chemical Engineering & Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Kuangshi Sun
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Lei Chen
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Jiamiao Yin
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Weigang Yan
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Mei Shi
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Wei Feng
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Xin Liu
- Academy for Engineering and Technology, Fudan University, Shanghai, 200433, China
| | - Fuyou Li
- School of Chemistry and Chemical Engineering & Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
9
|
Zhou Y, Chen J, Zhou Y, Liu Y, Yang X, Wang X, Chen W, Xu W, Cai H, Huang J. Renal-Clearable Molecular Reporters for Near-Infrared Fluorescence Imaging and Urinalysis of Pulmonary Metastatic Tumor. Anal Chem 2025; 97:8459-8467. [PMID: 40216593 DOI: 10.1021/acs.analchem.5c00392] [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: 04/23/2025]
Abstract
Despite approximately 40% of all patients with cancer developing pulmonary metastases in the course of their disease, it remains a diagnostic challenge in clinical practice. Herein, we propose a fluorogenic probe (CPRG) with gamma-glutamyl transferase (GGT)-triggered signal turn-on for near-infrared fluorescence imaging (NIRF) and urinalysis of orthotopic pulmonary metastatic tumors in living mice. CPRG comprised four key moieties: a GGT-reactive moiety, a hemicyanine-based signal unit, a polyethylene glycol linker, and an active tumor targeting moiety. Such a tailored probe is intrinsically nonfluorescent and only activates its NIRF signals in the presence of GGT. After intratracheal administration into the lungs of living tumor-bearing mice, CPRG can efficiently accumulate in the pulmonary tumors and sensitively turn-on the NIRF signal for real-time imaging. Relying on the high renal clearance efficiency (∼70% ID), it can be rapidly excreted through kidneys for urinalysis and assessed by the chemotherapeutic efficacy of cisplatin. This study not only reports fluorogenic tracers for imaging of pulmonary metastatic tumors but also provides guidelines for the development of molecular probes for companion diagnosis of metastatic cancer.
Collapse
Affiliation(s)
- Ya Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiangxian Chen
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yang Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Yi Liu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xingyue Yang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiaoxiao Wang
- Department of Pharmacy, Chongqing University Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing 400030, China
| | - Wanyi Chen
- Department of Pharmacy, Chongqing University Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing 400030, China
| | - Weiping Xu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Jiaguo Huang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
10
|
Li Y, Jiang G, Zhou J, Shi L, Yuan L. Recent Advances in Enzyme-Activated Dual-Locked Probes for Biological Applications. Chem Asian J 2025; 20:e202401647. [PMID: 39742394 DOI: 10.1002/asia.202401647] [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: 11/19/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/03/2025]
Abstract
Enzymes catalyze reactions involved in diverse physiological, pathological, and pharmacological processes. By employing the optical probe, fluorescence imaging enables non-invasive, real-time detection and assessment of disease states based on enzymatic activity. However, most enzyme-activated probes are single-locked probes that respond to a single biomarker. In comparison to single-locked probes, enzyme-activated dual-locked probes can effectively minimize the occurrence of false-positive signals, circumvent the problem of low specificity associated with biologically active substances, and facilitate precise imaging. This review systematically summarizes the design and application of dual-locked probes in disease diagnosis, with the aim of providing inspiration for researchers in the field.
Collapse
Affiliation(s)
- Yanhua Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Gangwei Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Junliang Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Ling Shi
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
11
|
Zhang H, Xu X, Cao Y, Chen Z, Liu W, Lu X, Li C. Unlocking the Power of Photothermal Agents: A Universal Platform for Smart Immune NIR-Agonists for Precise Cancer Therapy. Angew Chem Int Ed Engl 2025; 64:e202424830. [PMID: 39907354 DOI: 10.1002/anie.202424830] [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: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/06/2025]
Abstract
Selective ablation of tumor cells allows safe eradication, thereby minimizing off-target damage, while specifically inducing immunogenic cell death (ICD) rather than commonly non-immunogenic apoptosis of tumor cells enables activation of anti-tumor immune response against residual cancer cells, including metastatic lesions. Herein, we present a general strategy leveraging a novel photothermal agent (PTA) that concomitantly enables precise tumor killing and activation of anti-tumor immunity. The unique PTA scaffold exhibits unexpected inherent endoplasmic reticulum (ER)-targeting capability and potent near-infrared (NIR) photothermal activity, inducing NIR-controlled immunogenic pyroptosis in various tumor cell lines via targeting ER stress in an oxygen-independent manner. Moreover, both ER-targeting and NIR-activity of our scaffold can be modulated on demand by chemical caging/uncaging, allowing quick activation with diverse biological and bioorthogonal molecular triggers. The potency of this universal platform is demonstrated via its application to develop a membrane protein-activatable NIR-agonist that selectively activates ICD in tumor sites while priming anti-tumor immunity, minimizing off-target effects and enhancing efficacy against mouse breast tumors. This versatile approach could lead to customization of various personalized and effective immune NIR-agonists for specific photoimmunotherapy applicable to diverse solid tumors.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Xiaona Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Yahui Cao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Zihui Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Weiqing Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Xinyi Lu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Changhua Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
12
|
Li J, Guo L, Feng Y, Li G, Sun H, Huang W, Tian J, Du Y, An Y. Optical-magnetic Imaging for Optimizing Lymphodepletion-TIL Combination Therapy in Breast Cancer. Mol Imaging Biol 2025; 27:260-273. [PMID: 39909989 DOI: 10.1007/s11307-025-01985-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/20/2024] [Accepted: 01/19/2025] [Indexed: 02/07/2025]
Abstract
PURPOSE Lymphodepletion before tumor-infiltrating lymphocytes (TIL) infusion can activate the immune system, enhance the release of homeostatic cytokines, and decrease the number of immunosuppressive cells. This process is crucial for improving the therapeutic efficacy of TIL therapy. However, the challenge of in vivo assessing TILs targeting tumors limits the optimization of lymphodepleting conditioning regimen (LDC). PROCEDURES This study aims to employ magnetic particle imaging (MPI) and fluorescence molecular imaging (FMI) to monitor TIL biodistribution in vivo and optimize LDC in triple-negative breast cancer TIL therapy. MPI provides quantitative imaging capabilities without depth limitations, effectively complementing the high sensitivity of FMI. The efficacy of different LDCs in enhancing TIL therapy was assessed using FMI, and MPI quantified the number of TILs accumulated in the 4T1 tumor. RESULTS TILs preserved viability, phenotypes, and anti-tumor efficacy after being labeled with superparamagnetic iron oxide and fluorescence dye DiR. The dual-modality imaging system effectively discerned variations in LDC treatments that enhanced TIL therapy. Compared to TIL monotherapy, lymphodepletion with TIL therapy improves tumor dual-modality imaging signal intensity, increases the expression of monocyte chemotactic protein-1 in serum and tumor tissue, and enhances the therapeutic effect of TILs. CONCLUSION Our results confirm the utility of optical-magnetic dual-modality imaging for tracking the biodistribution of TILs in vivo. With the help of optical-magnetic dual-modality imaging, we successfully optimize TIL combination therapy. Optical-magnetic dual-modality imaging provides a new approach to develop personalized immunotherapy strategies and mine potential therapeutic mechanisms for TIL.
Collapse
Affiliation(s)
- Jiaqian Li
- School of Engineering Medicine & School of Biological Science and Medicine Engineering, Beihang University, Beijing, 100191, China
- The Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, 100191, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Lishuang Guo
- School of Engineering Medicine & School of Biological Science and Medicine Engineering, Beihang University, Beijing, 100191, China
- The Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, 100191, China
| | - Yuan Feng
- School of Engineering Medicine & School of Biological Science and Medicine Engineering, Beihang University, Beijing, 100191, China
- The Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, 100191, China
| | - Guanghui Li
- School of Engineering Medicine & School of Biological Science and Medicine Engineering, Beihang University, Beijing, 100191, China
- The Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, 100191, China
| | - He Sun
- School of Engineering Medicine & School of Biological Science and Medicine Engineering, Beihang University, Beijing, 100191, China
- The Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, 100191, China
| | - Wei Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Jie Tian
- School of Engineering Medicine & School of Biological Science and Medicine Engineering, Beihang University, Beijing, 100191, China.
- The Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, 100191, China.
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- The University of Chinese Academy of Sciences, Beijing, 100080, China.
| | - Yu An
- School of Engineering Medicine & School of Biological Science and Medicine Engineering, Beihang University, Beijing, 100191, China.
- The Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, 100191, China.
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
13
|
Banstola A, Lin ZT, Li Y, Wu MX. PhotoChem Interplays: Lighting the Way for Drug Delivery and Diagnosis. Adv Drug Deliv Rev 2025; 219:115549. [PMID: 39986440 PMCID: PMC11903148 DOI: 10.1016/j.addr.2025.115549] [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: 07/29/2024] [Revised: 12/23/2024] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Light, a non-invasive tool integrated with cutting-edge nanotechnologies, has driven transformative advancements in imaging-based diagnosis and drug delivery for cancer and bacterial treatments. This review discusses recent progress in these areas, beginning with emerging imaging technologies. Unlike traditional photosensors activated by visible light, alternative energy sources such as near-infrared (NIR) light, X-rays, and ultrasound have been extensively investigated to activate various photosensors, achieving high sensitivity, wavelength versatility, and spatial resolution for deep-tissue imaging. Moreover, to address challenges like tissue autofluorescence in real-time fluorescence imaging, afterglow luminescent nanoparticles are being developed by integrating these alternative energy sources for real-time imaging and sensing in deep tissue for precise cancer diagnosis and treatment beyond superficial tissues. In addition to deep tissue imaging, light-responsive nanomedicines are revolutionizing anticancer and antimicrobial phototherapy by enabling spatially and temporally controlled drug release. These smart nanoparticles are engineered to release therapeutic cargo at target sites in response to microenvironmental cues specific to tumors or infections. In anticancer phototherapy, these nanoparticles facilitate controlled drug release via photoisomerization, photothermal, and photodynamic processes. To enhance circulation time and specific targeting, biomimetic nanoparticles, which mimic natural anti-tumor responses by our body, have attracted increasing attention. In antimicrobial phototherapy, research has been focused on the chemical modification of the photosensitizer to enable targeted drug delivery. An intriguing strategy has recently emerged involving the development of "pro-photosensitizers" that are specifically activated within bacterial cells upon light irradiation, offering a high margin of safety. These advancements leverage photochemical reactions and nanotechnology to enhance precision therapy and diagnosis in addressing critical health challenges.
Collapse
Affiliation(s)
- Asmita Banstola
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH), Department of Dermatology, Harvard Medical School (HMS), Boston, MA 02114, USA
| | - Zuan-Tao Lin
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH), Department of Dermatology, Harvard Medical School (HMS), Boston, MA 02114, USA
| | - Yongli Li
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH), Department of Dermatology, Harvard Medical School (HMS), Boston, MA 02114, USA
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH), Department of Dermatology, Harvard Medical School (HMS), Boston, MA 02114, USA.
| |
Collapse
|
14
|
Wu Z, Liu R, Chen J, Cai X, Yi J, Wang J, Wang D, Hu M. An enzymatic cleavage-triggered minimally invasive nanosensor for urine-based detection of early atherosclerosis. SCIENCE ADVANCES 2025; 11:eadu7614. [PMID: 40085714 PMCID: PMC11908493 DOI: 10.1126/sciadv.adu7614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
Timely detection of early atherosclerosis (AS) is crucial for improving cardiovascular outcomes, creating a growing demand for diagnostic tools that are simple, sensitive, and cost-effective. Here, we introduce a synthetic nanosensor for early AS detection that leverages the fluorescence and renal clearance properties of carbon quantum dots (CQDs). This nanosensor, designed to respond to the proteolytic activity of AS-associated dysregulated enzymes, entails CQDs as signal reporters to convert AS-associated proteolytic activity to fluorometric readings enabling a sensitive and cost-effective urine-based assay for early AS detection. Our findings demonstrated that the nanosensor provided distinct signals in atherosclerotic versus healthy mice at early AS stages, indicating its diagnostic potential. Moreover, toxicity tests showed no notable adverse effects, supporting its safety for diagnostic applications. This minimally invasive diagnostic approach could facilitate personalized therapy design and continuous efficacy assessment. It is expected that such a modular nanosensor platform can be integrated with simple urine tests to offer cost-effective detection of various diseases.
Collapse
Affiliation(s)
- Zhina Wu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| | - Rui Liu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Jianai Chen
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xueying Cai
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | | | - Jiasi Wang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| |
Collapse
|
15
|
Liu Y, Zhang Z, Hou X, Ding Q, Zeng S, Shen H, Gong W, Ding T, Mahmood Z, Zeng X, Ren B, Hu W, Hong X, Ding D, Xiao Y. Aggregation-Mediated Photoacoustic/NIR-II and Photodynamic Properties of pH-Reversible Thiopyrylium Agents: A Computational and Experimental Approach. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2420006. [PMID: 39895403 DOI: 10.1002/adma.202420006] [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: 12/19/2024] [Indexed: 02/04/2025]
Abstract
Aggregation profoundly influences the photophysical properties of molecules. Here, a new series of thiopyrylium-based hemicyanine near-infrared II (NIR-II) fluorophores is developed by meticulously adjusting their aggregation states. Notably, the star molecule HTPA exhibits a remarkable pH-responsive behavior and a significant increase in photoacoustic (PA) intensity when aggregated. Additionally, their behavior and pH reversibility during aggregation formation are systematically investigated, including computational optimization, femtosecond transient absorption spectroscopy, NMR analysis, and single crystal analysis. Finally, an innovative "off " nanoparticle specifically is designed for effective tumor-targeted PA/NIR-II dual-modal imaging and photodynamic therapy by utilizing a pH-responsive polymer. The signal-to-background ratio (SBR) of PA signals significantly increased to 169 in the region of interest (ROI) in the mouse model when irradiated at 1064 nm. These findings not only provide a promising avenue for future studies of NIR-II small molecules but also pave the way for significant advances in the field of integrated cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Yishen Liu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- State Key Laboratory of Drug Research, 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, 264117, China
| | - Zhiyun Zhang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- State Key Laboratory of Drug Research, 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, 264117, China
| | - Xiaowen Hou
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- State Key Laboratory of Drug Research, 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, 264117, China
| | - Qihang Ding
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Silue Zeng
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Department of Hepatobiliary Surgery I, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Hanchen Shen
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, 999077, China
| | - Wanxia Gong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Taotao Ding
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- State Key Laboratory of Drug Research, 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, 264117, China
| | - Zafar Mahmood
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, 510006, China
| | - Xiaodong Zeng
- State Key Laboratory of Drug Research, 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, 264117, China
| | - Bingtao Ren
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Wenbo Hu
- Frontiers Science Center for Flexible Electronics, and Xi'an Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xuechuan Hong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- State Key Laboratory of Drug Research, 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, 264117, China
- Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Dan Ding
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yuling Xiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- State Key Laboratory of Drug Research, 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, 264117, China
- Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| |
Collapse
|
16
|
Sivakumar A, Phuengkham H, Rajesh H, Mac QD, Rogers LC, Silva Trenkle AD, Bawage SS, Hincapie R, Li Z, Vainikos S, Lee I, Xue M, Qiu P, Finn MG, Kwong GA. AND-gated protease-activated nanosensors for programmable detection of anti-tumour immunity. NATURE NANOTECHNOLOGY 2025; 20:441-450. [PMID: 39753733 PMCID: PMC11922657 DOI: 10.1038/s41565-024-01834-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/30/2024] [Indexed: 03/20/2025]
Abstract
The forward design of biosensors that implement Boolean logic to improve detection precision primarily relies on programming genetic components to control transcriptional responses. However, cell- and gene-free nanomaterials programmed with logical functions may present lower barriers for clinical translation. Here we report the design of activity-based nanosensors that implement AND-gate logic without genetic parts via bi-labile cyclic peptides. These actuate by releasing a reporter if and only if cleaved by a specific pair of proteases. AND-gated nanosensors that detect the concomitant activity of the granzyme B protease secreted by CD8 T cells and matrix metalloproteinases overexpressed by cancer cells identify the unique condition of cytotoxic T cell killing of tumour cells. In preclinical mouse models, AND-gated nanosensors discriminate tumours that are responsive to immune checkpoint blockade therapy from B2m-/- tumours that are resistant to it, minimize signals from tissues without co-localized protease expression including the lungs during acute influenza infection, and release a reporter locally in tissue or distally in the urine for facile detection.
Collapse
Affiliation(s)
- Anirudh Sivakumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - Hathaichanok Phuengkham
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - Hitha Rajesh
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - Quoc D Mac
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - Leonard C Rogers
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - Aaron D Silva Trenkle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - Swapnil Subhash Bawage
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - Robert Hincapie
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhonghan Li
- Department of Chemistry, University of California Riverside, Riverside, CA, USA
| | - Sofia Vainikos
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - Inho Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - Min Xue
- Department of Chemistry, University of California Riverside, Riverside, CA, USA
| | - Peng Qiu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA
| | - M G Finn
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Gabriel A Kwong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, GA, USA.
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA.
- Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA, USA.
- The Georgia Immunoengineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
17
|
Hu Y, Liu J, Xu M, Pu K. Dual-Locked Fluorescence Probe for Monitoring the Dynamic Transition of Pulmonary Macrophages. J Am Chem Soc 2025; 147:7148-7157. [PMID: 39946549 DOI: 10.1021/jacs.5c00506] [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: 02/27/2025]
Abstract
Pulmonary macrophages undergo dynamic changes in population, proportion, and polarization during respiratory diseases. Monitoring these changes is critical for understanding their roles in pathology, improving the diagnosis, and guiding drug development. However, current analytic methods based on tissue biopsy are invasive and static, limiting their ability to provide such dynamic information. Herein, we report a dual-locked macrophage-specific renal-clearable probe (DMRPNOCas) for the dynamic monitoring of pulmonary macrophages during influenza A virus (IAV) infection. DMRPNOCas activates fluorescence in the presence of two biomarkers (caspase-1 and NO) only coexpressed by M1 macrophages. To optimize the NO reactivity, the scaffold of DMRPNOCas is screened from the hemicyanine derivatives with an o-phenylenediamine group positioned differently on the indole ring. Notably, the para-substituted o-phenylenediamine demonstrates a higher NO-activated fluorescence compared to its meta-substituted counterpart. This enhancement, as revealed by quantum chemical calculations, is attributed to differential inhibition of twisted intramolecular charge transfer induced by the NO reaction. DMRPNOCas specifically distinguishes M1 macrophages from other leukocytes including T cells, neutrophils, and M2 macrophages, a capability unmatched by single-locked control probes and other reported probes. Consequently, DMRPNOCas enables in vivo dynamic monitoring of pulmonary macrophages, uncovering extensive recruitment and M1 polarization of monocyte-derived macrophages within 48 h of IAV infection. This process is accompanied by a significant reduction in alveolar macrophages. These findings provide new insights into macrophage-mediated pulmonary inflammation and underscore the potential of dual-locked probes for precise diagnosis and monitoring of pathological processes.
Collapse
Affiliation(s)
- Yuxuan Hu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| |
Collapse
|
18
|
Ran XY, Wei YF, Wu YL, Dai LR, Xia WL, Zhou PZ, Li K. Xanthene-based NIR organic phototheranostics agents: design strategies and biomedical applications. J Mater Chem B 2025; 13:2952-2977. [PMID: 39898613 DOI: 10.1039/d4tb02480j] [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: 02/04/2025]
Abstract
Fluorescence imaging and phototherapy in the near-infrared window (NIR, 650-1700 nm) have attracted great attention for biomedical applications due to their minimal invasiveness, ultra-low photon scattering and high spatial-temporal precision. Among NIR emitting/absorbing organic dyes, xanthene derivatives with controllable molecular structures and optical properties, excellent fluorescence quantum yields, high molar absorption coefficients and remarkable chemical stability have been extensively studied and explored in the field of biological theranostics. The present study was aimed at providing a comprehensive summary of the progress in the development and design strategies of xanthene derivative fluorophores for advanced biological phototheranostics. This study elucidated several representative controllable strategies, including electronic programming strategies, extension of conjugated backbones, and strategic establishment of activatable fluorophores, which enhance the NIR fluorescence of xanthene backbones. Subsequently, the development of xanthene nanoplatforms based on NIR fluorescence for biological applications was detailed. Overall, this work outlines future efforts and directions for improving NIR xanthene derivatives to meet evolving clinical needs. It is anticipated that this contribution could provide a viable reference for the strategic design of organic NIR fluorophores, thereby enhancing their potential clinical practice in future.
Collapse
Affiliation(s)
- Xiao-Yun Ran
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, P. R. China.
| | - Yuan-Feng Wei
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan-Ling Wu
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, P. R. China.
| | - Li-Rui Dai
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wen-Li Xia
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, P. R. China.
| | - Pei-Zhi Zhou
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, P. R. China.
| |
Collapse
|
19
|
Li T, Zong Q, Dong H, Ullah I, Pan Z, Yuan Y. Non-invasive in vivo monitoring of PROTAC-mediated protein degradation using an environment-sensitive reporter. Nat Commun 2025; 16:1892. [PMID: 39987172 PMCID: PMC11846984 DOI: 10.1038/s41467-025-57191-0] [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: 06/05/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
Proteolysis targeting chimeras (PROTACs) represent a groundbreaking therapeutic technology for selectively degrading proteins of interest (POIs). The structural variations in PROTACs unpredictably influence their protein degradation efficiency, which is predominantly assessed by quantifying POIs abundance through western blotting. This approach, however, falls short of enabling non-invasive monitoring of protein degradation within living cells let alone assessing directly the degradation effects in vivo. Herein, we develop an environment-sensitive reporter (ESR) for the quantification of protein degradation events triggered by PROTACs in vivo. By simultaneously integrating POIs targeting ligand and an environment-sensitive fluorophore, the ESR signals exhibit a strong fluorescence correlation with the levels of POIs. This non-invasive monitoring reporter offers a high-throughput and convenient way to screen POIs targeting degraders and predict PROTACs-mediated therapeutic outcomes in mouse models. These properties suggest the potential of ESR strategy as a general modular scheme for non-invasive quantification of protein degradation of cancer-related therapeutic targets.
Collapse
Affiliation(s)
- Tao Li
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, PR China
| | - Qingyu Zong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, PR China
| | - He Dong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, PR China
| | - Ihsan Ullah
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, PR China
| | - Zhenhai Pan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, PR China
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, PR China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, PR China.
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, PR China.
| |
Collapse
|
20
|
Liu J, Cheng P, Xu C, Pu K. Molecular probes for in vivo optical imaging of immune cells. Nat Biomed Eng 2025:10.1038/s41551-024-01275-7. [PMID: 39984703 DOI: 10.1038/s41551-024-01275-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/23/2024] [Indexed: 02/23/2025]
Abstract
Advancing the understanding of the various roles and components of the immune system requires sophisticated methods and technology for the detection of immune cells in their natural states. Recent advancements in the development of molecular probes for optical imaging have paved the way for non-invasive visualization and real-time monitoring of immune responses and functions. Here we discuss recent progress in the development of molecular probes for the selective imaging of specific immune cells. We emphasize the design principles of the probes and their comparative performance when using various optical modalities across disease contexts. We highlight molecular probes for imaging tumour-infiltrating immune cells, and their applications in drug screening and in the prediction of therapeutic outcomes of cancer immunotherapies. We also discuss the use of these probes in visualizing immune cells in atherosclerosis, lung inflammation, allograft rejection and other immune-related conditions, and the translational opportunities and challenges of using optical molecular probes for further understanding of the immune system and disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
21
|
Luan X, Gao Y, Pan Y, Huang Z, Zeng F, He G, He B, Ye D, Song Y. Bifunctional Nanoassembly Enables Metabolism-Driven Microfluidic Blood Screening Guided by MRI Localization for Cancer Monitoring. Anal Chem 2025; 97:3395-3403. [PMID: 39900559 DOI: 10.1021/acs.analchem.4c05427] [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: 02/05/2025]
Abstract
Early detection and precise tumor localization are critical for improving treatment outcomes and enabling more targeted and minimally invasive therapies as biotechnology evolves. However, endogenous biomarkers from early lesions face significant challenges, such as short circulation times and blood dilution, which hinder early diagnostic efforts. In this study, we present a multimodal nanosensor specifically engineered to target cancer by responding to CD44 and tumor-associated enzymes within the microenvironment. Following systemic administration, the nanosensor selectively accumulates at the disease site, delivering hexaminolevulinate (HAL) to produce protoporphyrin IX (PpIX) as a synthetic biomarker, thus amplifying disease signals for analysis via a microfluidics-based device. Concurrently, embedded Gd2O3 nanoclusters facilitate tumor visualization through magnetic resonance imaging (MRI). Beyond tumor diagnosis, this innovative methodology supports the multimodal monitoring of drug response through the assessment of blood reporter signals and MRI imaging. This multifunctional system addresses critical limitations in traditional cancer diagnostics, which typically rely on sequential blood biomarker tests, followed by imaging. Our approach enhances diagnostic efficiency, minimizes the need for invasive procedures, and promotes more accurate and personalized cancer care.
Collapse
Affiliation(s)
- Xiaowei Luan
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Yanfeng Gao
- School of Medical Imaging, Wannan Medical College, Wuhu 241002, China
| | - Yongchun Pan
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Zheng Huang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Fei Zeng
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Guanzhong He
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yujun Song
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| |
Collapse
|
22
|
Liu Y, Yao Y, Sha J, Liang G, Sun X. Dual-Locked Enzyme-Activatable Fluorescence Probes for Precise Bioimaging. ACS Biomater Sci Eng 2025; 11:730-741. [PMID: 39841057 DOI: 10.1021/acsbiomaterials.4c01858] [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: 01/23/2025]
Abstract
Real-time visualization of endogenous enzymes not only helps reveal the underlying biological principles but also provides pathological information for cancer/disease diagnosis and even treatment guidance. To this end, enzyme-activatable fluorescence probes are frequently fabricated that turn their fluorescence signals "on" exclusively at the enzyme-rich region, thus enabling noninvasive and real-time imaging of enzymes of interest at the molecular level with superior sensitivity and selectivity. However, in a complex biological context, commonly used single enzyme-activatable (i.e., single-locked) probes may suffer from "false positive" signals at healthy tissues and be insufficient to accurately indicate the occurrence of certain diseases. Therefore, dual-locked fluorescence probes have been promoted to address these issues. Using dual enzymes (or an enzyme with another stimulus) as "keys", they permit simultaneous detection of distinct biomarkers, offering significantly enhanced imaging precision toward certain biological events. Considering that recent reviews on these probes remain scarce, we thus provide this review. We summarize the recent progress, particularly highlighting the breakthroughs in the last three years, and discuss the challenges in this field.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Yuchen Yao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Junhui Sha
- School of Life Science and Technology, Southeast University, Nanjing 211189, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
- Handan Norman Technology Company, Limited, Guantao 057750, China
| | - Xianbao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
23
|
Xu L, Xiong W, Zhang S, Pan J, Liu Y, Liu Q, Wu H, Li L, Zhu JJ, Zheng F. Near-Infrared Photothermally Assisted Nanoprobes Boost Signal Amplification for Fluorescence Imaging and Urinalysis of Tumor. Anal Chem 2025; 97:2463-2471. [PMID: 39865715 DOI: 10.1021/acs.analchem.4c06166] [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: 01/28/2025]
Abstract
Early diagnosis of tumors allows effective treatment of primary cancers through localized therapeutic interventions. However, developing diagnostic tools for sensitive, simple, and early tumor (especially less than 2 mm in diameter) detection remains a challenge. Herein, we presented a biomarker-activatable nanoprobe that enabled a near-infrared (NIR) photothermally amplified signal for fluorescence imaging and urinalysis of tumor. This activatable nanoprobe was constructed by encapsulating renal-clearable NIR ZW800 dyes into a CuS@mSiO2 core-shell nanoparticle with hyaluronic acid. The nanoprobes could accumulate at the tumor site and respond to tumor-associated hyaluronidase, undergoing in situ enzyme-catalyzed decapsulation to release renal-clearable ZW800 dyes for fluorescence imaging and uranalysis of tumor in living mice. Notedly, with the aid of NIR laser irradiation, the photothermal effect of CuS core boosted signal amplification for tumor diagnosis via photothermally enhanced ZW800 release, providing high specificity and sensitivity. On account of the biomarker specificity and the spatiotemporally controlled NIR irradiation, the nanoprobes could selectively activate their NIR fluorescence (NIRF) signal to visualize tumors in living mice and allow for the easy translation of the nanoprobe as artificial urinary biomarker probes for in vitro diagnosis of tumor progression. In 4T1 tumor-bearing mice models, the activatable nanoprobes enabled ultrasensitive detection of tumors (1.9 mm in diameter). This study offers a noninvasive and photothermally signal-amplified approach for early tumor diagnosis via NIRF imaging or simple urine tests.
Collapse
Affiliation(s)
- Limei Xu
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Weiwei Xiong
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Shiling Zhang
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Jiajia Pan
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Yingqi Liu
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Qiulin Liu
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Haojie Wu
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| | - Lingling Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Xianlin Ave 163, Nanjing, Jiangsu 210023, China
| | - Fenfen Zheng
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu 212003, China
| |
Collapse
|
24
|
Zhu J, Zhao L, An W, Miao Q. Recent advances and design strategies for organic afterglow agents to enhance autofluorescence-free imaging performance. Chem Soc Rev 2025; 54:1429-1452. [PMID: 39714452 DOI: 10.1039/d4cs01060d] [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: 12/24/2024]
Abstract
Long-lasting afterglow luminescence imaging that detects photons slowly being released from chemical defects has emerged, eliminating the need for real-time photoexcitation and enabling autofluorescence-free in vivo imaging with high signal-to-background ratios (SBRs). Organic afterglow nano-systems are notable for their tunability and design versatility. However, challenges such as unsatisfactory afterglow intensity, short emission wavelengths, limited activatable strategies, and shallow tissue penetration depth hinder their widespread biomedical applications and clinical translation. Such contradiction between promising prospects and insufficient properties has spurred researchers' efforts to improve afterglow performance. In this review, we briefly outline the general composition and mechanisms of organic afterglow luminescence, with a focus on design strategies and an in-depth understanding of the structure-property relationship to advance afterglow luminescence imaging. Furthermore, pending issues and future perspectives are discussed.
Collapse
Affiliation(s)
- Jieli Zhu
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China.
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Liangyou Zhao
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China.
| | - Weihao An
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China.
| | - Qingqing Miao
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China.
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
25
|
Zhang H, Chen P, Shi W, Qu A, Sun M, Kuang H. Renal Clearable Chiral Manganese Oxide Supraparticles for In Vivo Detection of Metalloproteinase-9 in Early Cancer Diagnosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415656. [PMID: 39713947 DOI: 10.1002/adma.202415656] [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: 10/14/2024] [Revised: 11/29/2024] [Indexed: 12/24/2024]
Abstract
In this study, polypeptide TGGGPLGVARGKGGC-induced chiral manganese dioxide supraparticles (MnO2 SPs) are prepared for sensitive quantification of matrix metalloproteinase-9 (MMP-9) in vitro and in vivo. The results show that L-type manganese dioxide supraparticles (L-MnO2 SPs) exhibited twice the affinity for the cancer cell membrane receptor CD47 (cluster of differentiation, integrin-associated protein) than D-type manganese dioxide supraparticles (D-MnO2 SPs) to accumulate at the tumor site after surface modification of the internalizing arginine-glycine-aspartic acid (iRGD) ligand, specifically reacting with the MMP-9, disassembling into ultrasmall nanoparticles (NPs), and efficiently underwent renal clearance. Furthermore, L-MnO2 facilitates the quantification of MMP-9 in mouse tumor xenografts, as demonstrated by circular dichroism (CD) and magnetic resonance imaging (MRI) within 2 h. A strong linear relationship is observed between MMP-9 concentration and both CD and MRI intensity, ranging from 0.01 to 10 ng mL-1. The corresponding limits of detection (LOD) are 0.0054 ng mL-1 for CD and 0.0062 ng mL-1 for MRI, respectively. hese SPs provide a new approach for exploring chiral advanced biosensors for early diagnosis of cancer.
Collapse
Affiliation(s)
- Hongyu Zhang
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- Department of Instrumentation and Analytical Chemistry, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, 116023, China
| | - Panpan Chen
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Wenxiong Shi
- Institute for New Energy Materials and Low Carbon Technologies, School of Materials Science and Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Aihua Qu
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maozhong Sun
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hua Kuang
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
26
|
Huang Y, Zheng J, Yu M. Nanoparticle Transport in Proximal Tubules with Rhabdomyolysis-Induced Necrosis. Angew Chem Int Ed Engl 2025; 64:e202417024. [PMID: 39423345 DOI: 10.1002/anie.202417024] [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: 09/04/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
Renal-clearable engineered nanoparticles are being explored for their potential to deliver therapeutic agents for kidney disease treatment. A fundamental understanding of how these nanoparticles accumulate in diseased kidneys at the cellular level is essential to enhance their effectiveness and minimize side effects on adjacent healthy tissues. Herein, we report that the accumulation of glutathione-coated, near-infrared emitting gold nanoparticles (GS-AuNPs) correlates strongly with the necrotic stages of injured proximal tubular cells. Using a rhabdomyolysis-induced acute kidney injury (AKI) mouse model, we observed that GS-AuNPs were significantly accumulated in the extracellular lumen of proximal tubular epithelial cells (PTECs) at advanced necrotic stage, where cellular debris and released intracellular contents impeded their clearance. In contrast, during early necrosis, GS-AuNPs were still cleared through the unobstructed lumen. Additionally, intracellular uptake of GS-AuNPs was significantly reduced across all necrotic stages. These findings underscore the need for new strategies to design nanoparticles that can effectively target and be taken up by the diseased tubular cells before extensive necrosis occurs; so that nanoparticle-mediated drug delivery for kidney disease treatment can be achieved with desired efficacy and precision.
Collapse
Affiliation(s)
- Yingyu Huang
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 W. Campbell Rd., Richardson, TX 75080, USA
| | - Jie Zheng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 W. Campbell Rd., Richardson, TX 75080, USA
| | - Mengxiao Yu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 W. Campbell Rd., Richardson, TX 75080, USA
| |
Collapse
|
27
|
Zhou Y, Xu W, Ruan B, Zhu L, Jiang Y, Cai H, Huang J. Molecular Imaging of Renin Activity using Fluorogenic Nanoprobes for Precision Antihypertensive Therapy. Angew Chem Int Ed Engl 2025; 64:e202416002. [PMID: 39279688 DOI: 10.1002/anie.202416002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/18/2024]
Abstract
Life-threatening hypertension remains inadequately controlled in clinics due to its heterogeneous renin levels. Rapid stratification of hypertension through renin analysis is crucial for effective personalized treatment, yet an ultrasensitive detection approach is currently lacking. Here, we report activatable renin nanoprobes (ARNs) for non-invasive and ultrasensitive profiling of renin activity and guiding antihypertensive treatment decision through near-infrared fluorescence (NIRF) in vivo imaging and in vitro urinalysis. ARNs are intrinsically non-fluorescent due to NIRF reporter connected to a gold nanocluster through a renin-responsive peptide. In hyperreninemia mouse models, ARNs specifically react with renin to liberate the renal clearable NIRF reporter for accurate renin detection that outperforms the gold standard radioimmunoassay. Such specific and sensitive detection also enables imaging-based high-throughput screening of antihypertensive drugs. In hypertensive rat models, ARNs enable ultrasensitive detection of both plasma and urinary renin, facilitating renin-guided precision treatment and significantly improving hypertension control rate (90 % versus 58 %). Our nanoprobe platform holds great potential for assisting clinicians in rapidly and accurately classifying hypertensive patients and improving outcomes through tailored treatment selection.
Collapse
Affiliation(s)
- Ya Zhou
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Weiping Xu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Bankang Ruan
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lijuan Zhu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuyan Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA-94305, USA
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen, 518107, China
| | - Jiaguo Huang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key La-boratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| |
Collapse
|
28
|
Dong L, Wang C, Tong T, Ling B, Liu P, Yang Y, Liu Z, Wang C, Yuan Y. Self-Activated Cascade-Tailored Small Molecule for Cancer Therapy, Companion Diagnostics, and "Theranostic Correlation" Evaluation. Anal Chem 2024; 96:20147-20151. [PMID: 39652365 DOI: 10.1021/acs.analchem.4c05313] [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: 12/25/2024]
Abstract
Companion diagnostics (CDx) have emerged as valuable tools for monitoring biomarkers essential for drug activation and therapeutic response, enabling personalized treatment strategies. However, the current FDA-approved CDx is limited to in vitro testing, making it challenging to assess the real-time drug efficacy. Moreover, evaluation of treatment responses solely based on drug release or activation may disregard tumor heterogeneity. To address these challenges, we have developed a cascade-responsive small molecule Cbl-DEVD-Hcy for simultaneous cancer therapy and the timely evaluation of therapy effectiveness in vivo. Upon cleavage by tumor-cell-overexpressed carboxylesterase, chlorambucil (Cbl) can be released to induce tumor cell apoptosis and activate caspase-3. This activation triggers the production of the near-infrared dye Hcy-NH2, generating both near-infrared fluorescence and photoacoustic signals for monitoring the apoptosis process. The excellent "theranostic correlation" between the imaging signal and therapeutic response, as demonstrated in orthotopic breast tumors, highlights the potential of Cbl-DEVD-Hcy for effective tumor therapy and precise CDx in the body.
Collapse
Affiliation(s)
- Ling Dong
- Department of the Interventional Medical Center, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
- Department of Chemical and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui 230061, China
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Chenchen Wang
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Tong Tong
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Bo Ling
- Department of Chemical and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui 230061, China
| | - Pingping Liu
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yanyun Yang
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zhengjie Liu
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Congxiao Wang
- Department of the Interventional Medical Center, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Yue Yuan
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
29
|
Xu M, Hu Y, Wu J, Liu J, Pu K. Sonodynamic Nano-LYTACs Reverse Tumor Immunosuppressive Microenvironment for Cancer Immunotherapy. J Am Chem Soc 2024; 146:34669-34680. [PMID: 39644208 DOI: 10.1021/jacs.4c13022] [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: 12/09/2024]
Abstract
Extracellular and transmembrane proteins, which account for the products of approximately 40% of all protein-encoding genes in tumors, play a crucial role in shaping the tumor immunosuppressive microenvironment (TIME). While protein degradation therapy has been applied to membrane proteins of cancer cells, it has rarely been extended to immune cells. We herein report a polymeric nanolysosome targeting chimera (nano-LYTAC) that undergoes membrane protein degradation on M2 macrophages and generates a sonodynamic effect for combinational cancer immunotherapy. Nano-LYTAC is found to have higher degradation efficacy to the interleukin 4 receptor (IL-4R) compared to traditional inhibitors. More importantly, it is revealed that the effect of nano-LYTAC on the function of the M2 macrophage is concentration-dependent: downregulating CD206 expression and interleukin 10 (IL-10) secretion from M2 macrophages at low concentration, while triggering their apoptosis at high concentration. Moreover, nano-LYTAC is found to possess long tumor retention (>48 h), allowing for multiple sonodynamic treatments with a single dose. Such a synergistic sonodynamic immunotherapy mediated by nano-LYTAC effectively reprograms the TIME via inhibiting the functions of M2 macrophages and regulatory T cells (Tregs), as well as promoting the maturation of dendritic cells (DCs) and tumor infiltration of T effector cells (Teffs), completely suppressing tumor growth, inhibiting pulmonary metastasis, and preventing recurrence under preclinical animal models.
Collapse
Affiliation(s)
- Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Yuxuan Hu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Jiayan Wu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921
| |
Collapse
|
30
|
Shi M, Zhang Y, Chen JX, Wu Y, Wang Z, Shi PF, Jin X, Wang XQ. A Bicyclic Dioxetane Chemiluminescence Nanoprobe for Peroxynitrite Imaging in Vivo. Anal Chem 2024. [PMID: 39565840 DOI: 10.1021/acs.analchem.4c04510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Peroxynitrite (ONOO-) is a critical biomarker associated with a wide array of diseases including cancer, inflammatory conditions, and neurodegenerative disorders. This study introduces an innovative chemiluminescence nanoprobe (CLNP) based on a bicyclic dioxetane structure, designed for highly sensitive and specific in vivo imaging of ONOO-. Our CLNP demonstrates exceptional capabilities in generating high-contrast imaging of disease lesions, with applications verified across tumor models, acute inflammation, and acute liver injury scenarios. Key findings highlight the probe's rapid response to oxidative species, superior tissue penetration, and high signal-to-noise ratio, underscoring its potential for real-time diagnostic applications. This work represents an important advance in the field of diagnostic imaging using CL probes, offering promising avenues for the early detection and treatment of ONOO--related pathologies.
Collapse
Affiliation(s)
- Min Shi
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, P. R. China
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, Shandong 276000, P. R. China
| | - Yun Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Jia-Xing Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Yanyuan Wu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zongping Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Peng-Fei Shi
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, Shandong 276000, P. R. China
| | - Xudong Jin
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, P. R. China
| | - Xue-Qiang Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
31
|
Wei K, Xu Y, Nie C, Wei Q, Xie P, Chen T, Jiang J, Chu X. A Multifunctional Peptide Nucleic Acid/Peptide Copolymer-Based Dual-Mode Biosensor with Macrophage-Hitchhiking for Enhanced Tumor Imaging and Urinalysis. J Am Chem Soc 2024. [PMID: 39563630 DOI: 10.1021/jacs.4c10562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Biosensors are capable of diagnosing tumors through imaging in vivoor liquid biopsy, but they face the challenges of inefficient delivery into tumor sites and the lack of reliable tumor-associated biomarkers. Herein, we constructed a dual-mode biosensor based on a multifunctional peptide nucleic acid (PNA)/peptide copolymer and DNA tetrahedron for tumor imaging and urinalysis. The biosensor could enter the cancer cells to initiate a microRNA-21-specific catalytic hairpin assembly reaction after cleavage by matrix-metalloprotease (MMP) in the tumor microenvironment, and the MMP cleavage product was released into the bloodstream and then was filtered out by the kidney. As PNA was a synthetic DNA analogue that could not be degraded by nucleases and proteases, it could serve as a reliable synthetic biomarker and be easily detected by high-performance liquid chromatography in urine. Importantly, the biosensor was hitchhiked on the macrophage membrane to realize efficient delivery in the depth of tumor utilizing the macrophage ability of actively homing to the tumor site and infiltrating into the tumor. The results indicated that the signal output of the biosensor was improved remarkably and mice with a tumor volume as little as 30-40 mm3 could be reliably discriminated through urine assay. This innovative macrophage-hitchhiking dual-mode biosensor holds a great potential as a non-invasive and convenient tool for tumor diagnosis and tumor progression evaluation.
Collapse
Affiliation(s)
- Kaiji Wei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yu Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Cunpeng Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Qiaomei Wei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Ping Xie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Tingting Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jianhui Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xia Chu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
32
|
Ma X, Mao M, Liu Z, Liang C, He J, Qu Y, Xu L, Cheng R, Zhuang W, Lei Y, Nie W, Yuan L, Pang DW, Xie HY. AND-Gate Logic Förster Resonance Energy Transfer/Magnetic Resonance Tuning Nanoprobe for Programmable Antitumor Immunity Imaging. J Am Chem Soc 2024; 146:31873-31884. [PMID: 39504515 DOI: 10.1021/jacs.4c11072] [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: 11/08/2024]
Abstract
Simultaneous detection of different biomarkers related to the spatiotemporally dynamic immune events is of particular importance for the accurate evaluation of antitumor immune effects. Here, we have developed an AND-gate logic dual resonance energy transfer nanoprobe (named DRET) for dynamic monitoring of programmed CD8+ T cell activation and tumor cell apoptosis. Immunotherapy-induced granzyme B secretion from CD8+ T cells and the subsequent caspase-3 release from apoptotic tumor cells individually activate one of the tiers of the "AND-gate" logic DRET. The resulting fluorescence recovery and magnetic resonance T1 enhancement can be used for precise immunomodulatory drug screening, early efficacy prediction, and immune stratification. Particularly, not only "Responders" can be distinguished from "Non-responders", but also "Acquired resistance" can be identified from "Maintain responders", providing a novel approach to put forward the accurate evaluation of antitumor immunity.
Collapse
Affiliation(s)
- Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Zhenya Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yun Qu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Luzheng Xu
- Medical and Health Analysis Center, Peking University, Beijing 100191, P. R. China
| | - Ran Cheng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Wanru Zhuang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Yao Lei
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Lan Yuan
- Medical and Health Analysis Center, Peking University, Beijing 100191, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
33
|
Yang FK, Cao J, Zhang T, Jiang HX, Cui HB, Wang K. Dual-Activated Photoacoustic Probe for Reliably Detecting Hydroxyl Radical in Ischemic Cardiovascular Disease in Mouse and Human Samples. ACS Sens 2024; 9:5445-5453. [PMID: 39364916 DOI: 10.1021/acssensors.4c01665] [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: 10/05/2024]
Abstract
Cardiovascular disease (CVD) is a chronic disease characterized by the accumulation of lipids and fibrous tissue within the arterial walls, potentially leading to vascular obstruction and an increased risk of heart disease and stroke. Hydroxyl radicals play a significant role in the formation and progression of CVD as they can instigate lipid peroxidation, resulting in cellular damage and inflammatory responses. However, precisely detecting hydroxyl radicals in CVD lesions presents significant challenges due to their high reactivity and short lifespan. Herein, we present the development and application of a novel activatable optical probe, Cy-OH-LP, designed to detect hydroxyl radicals in lipid-rich environments specifically. Built on the Cy7 molecular skeleton, Cy-OH-LP exhibits near-infrared absorption and fluorescence characteristics, and its specific response to hydroxyl radicals enables a turn-on signal in both photoacoustic and fluorescence spectra. The probe demonstrated excellent selectivity and stability in various tests. Furthermore, Cy-OH-LP was successfully applied in an in vivo model to detect hydroxyl radicals in mouse models, providing a potential tool for diagnosing and monitoring AS. The biosafety of Cy-OH-LP was also verified, showing low cytotoxicity and no significant organ damage in mice. The findings suggest that Cy-OH-LP is a promising tool for the specific detection of hydroxyl radicals in lipid-rich environments, providing new possibilities for research and clinical applications in the field of oxidative stress-related diseases.
Collapse
Affiliation(s)
- Fang-Kun Yang
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Jie Cao
- The Fifth Dental Center, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Ting Zhang
- Wuxi Maternity and Chield Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi 214002, China
| | - Hao-Xiang Jiang
- Affiliated Children's Hospital of Jiangnan University, Wuxi 214023, China
| | - Han-Bin Cui
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Kai Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
- Affiliated Children's Hospital of Jiangnan University, Wuxi 214023, China
| |
Collapse
|
34
|
Wang J, Cao M, Han L, Shangguan P, Liu Y, Zhong Y, Chen C, Wang G, Chen X, Lin M, Lu M, Luo Z, He M, Sung HHY, Niu G, Lam JWY, Shi B, Tang BZ. Blood-Brain Barrier-Penetrative Fluorescent Anticancer Agents Triggering Paraptosis and Ferroptosis for Glioblastoma Therapy. J Am Chem Soc 2024; 146:28783-28794. [PMID: 39394087 DOI: 10.1021/jacs.4c07785] [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: 10/13/2024]
Abstract
Currently used drugs for glioblastoma (GBM) treatments are ineffective, primarily due to the significant challenges posed by strong drug resistance, poor blood-brain barrier (BBB) permeability, and the lack of tumor specificity. Here, we report two cationic fluorescent anticancer agents (TriPEX-ClO4 and TriPEX-PF6) capable of BBB penetration for efficient GBM therapy via paraptosis and ferroptosis induction. These aggregation-induced emission (AIE)-active agents specifically target mitochondria, effectively triggering ATF4/JNK/Alix-regulated paraptosis and GPX4-mediated ferroptosis. Specifically, they rapidly induce substantial mitochondria-derived vacuolation, accompanied by reactive oxygen species generation, decreased mitochondrial membrane potential, and intracellular Ca2+ overload, thereby disrupting metabolisms and inducing nonapoptotic cell death. In vivo imaging revealed that TriPEX-ClO4 and TriPEX-PF6 successfully traversed the BBB to target orthotopic glioma and initiated effective synergistic therapy postintravenous injection. Our AIE drugs emerged as the pioneering paraptosis inducers against drug-resistant GBM, significantly extending survival up to 40 days compared to Temozolomide (20 days) in drug-resistant GBM-bearing mice. These compelling results open up new venues for the development of fluorescent anticancer drugs and innovative treatments for brain diseases.
Collapse
Affiliation(s)
- Jiefei Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Mingyue Cao
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, P. R. China
| | - Lulu Han
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Ping Shangguan
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Yisheng Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Yong Zhong
- Key Laboratory for Special Functional Materials of Ministry of Education, Henan University, Kaifeng 475004, P. R. China
| | - Chaoyue Chen
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, Division of Life Science, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, P. R. China
| | - Gaoyang Wang
- Key Laboratory for Special Functional Materials of Ministry of Education, Henan University, Kaifeng 475004, P. R. China
| | - Xiaoyu Chen
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Ming Lin
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Mengya Lu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Zhengqun Luo
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Mu He
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Herman H Y Sung
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, Division of Life Science, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, P. R. China
| | - Guangle Niu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jacky W Y Lam
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, Division of Life Science, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, P. R. China
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen 518172, P. R. China
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, Division of Life Science, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, P. R. China
| |
Collapse
|
35
|
Cheng X, He C, Huang J, Li J, Hu Z, Wang L, Wei T, Cui L, Lu M, Mi P, Xu J. A Tumor-Homing Nanoframework for Synergistic Microwave Tumor Ablation and Provoking Strong Anticancer Immunity Against Metastasis. ACS NANO 2024; 18:29121-29139. [PMID: 39387481 DOI: 10.1021/acsnano.4c10603] [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: 10/15/2024]
Abstract
Microwave thermotherapy (MT) is a clinical local tumor ablation modality, but its applications are limited by its therapeutic efficacy and safety. Therefore, developing sensitizers to optimize the outcomes of MT is in demand in clinical practice. Herein, we engineered a special nanoframework (i.e., FdMI) based on a fucoidan-decorated zirconium metal-organic framework incorporating manganese ions and liquid physisorption for microwave tumor ablation. The monodisperse nanoframework exhibited both microwave thermal effects and microwave dynamic effects, which could effectively kill cancer cells by efficient intracellular drug delivery. Through fucoidan-mediated targeting of P-selectin in the tumor microenvironment (TME), the FdMI effectively accumulated in tumor regions, leading to significant eradication of orthotropic triple-negative breast cancer (TNBC) and aggressive Hepa1-6 liver tumors by the synergistic effects of microwave thermotherapy/dynamic therapy (MT/MDT). The eradication of primary tumors could activate systemic immune responses, which effectively inhibited distant TNBC tumors and lung metastasis of Hepa1-6 liver tumors, respectively. This work not only engineered nanoparticle sensitizers for tumor-targeted synergistic MT/MDT but also demonstrated that nanocarrier-based microwave tumor ablation could stimulate antitumor immunity to effectively inhibit distant and metastatic tumors, demonstrating the high potential for effectively managing advanced malignant tumors.
Collapse
Affiliation(s)
- Xueqing Cheng
- Department of Ultrasound & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Chuanshi He
- Department of Ultrasound & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Jiangbo Huang
- Department of Ultrasound, Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Juan Li
- Department of Ultrasound & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Ziyue Hu
- Department of Ultrasound & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Lu Wang
- Department of Ultrasound & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Ting Wei
- Department of Ultrasound & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Likun Cui
- Department of Ultrasound & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Man Lu
- Department of Ultrasound & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Jinshun Xu
- Department of Ultrasound & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| |
Collapse
|
36
|
Wan Y, Gao Y, Wei WC, Lee KW, Tan JH, Chen CY, Chen H, Li S, Wong KT, Lee CS. Facilely Achieving Near-Infrared-II J-Aggregates through Molecular Bending on a Donor-Acceptor Fluorophore for High-Performance Tumor Phototheranostics. ACS NANO 2024; 18:27949-27961. [PMID: 39364674 DOI: 10.1021/acsnano.4c05546] [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: 10/05/2024]
Abstract
Constructing J-aggregated organic dyes represents a promising strategy for obtaining biomedical second near-infrared (NIR-II) emissive materials, as they exhibit red-shifted spectroscopic properties upon assembly into nanoparticles (NPs) in aqueous environments. However, currently available NIR-II J-aggregates primarily rely on specific molecular backbones with intricate design strategies and are susceptible to fluorescence quenching during assembly. A facile approach for constructing bright NIR-II J-aggregates using prevalent donor-acceptor (D-A) molecules is still lacking. In this study, we present a facile method that transforms D-A molecules into J-aggregates by simply bending the molecule through introducing a methyl group, enabling high-performance NIR-II phototheranostics. The TAA-BT-CN molecule exhibits hypsochromic-shift absorption upon forming H-aggregated NPs, while the designed mTAA-BT-CN with a bent structure demonstrates a bathochromic shift of over 100 nm in absorption upon forming J-aggregated NPs, leading to much enhanced NIR-II emission beyond 1100 nm. With respect to its H-aggregated counterpart with the aggregation-caused quenching (ACQ) phenomenon, the J-aggregated mTAA-BT-CN NPs exhibit a 7-fold increase in NIR-II fluorescence owing to their aggregation-induced emission (AIE) property as well as efficient generation of heat and reactive oxygen species under 808 nm light excitation. Finally, the mTAA-BT-CN NPs are employed for whole-body blood vessel imaging using NIR-II technology as well as imaging-guided tumor phototherapies. This study will facilitate the flourishing advancement of J-aggregates based on prevalent D-A-type molecules.
Collapse
Affiliation(s)
- Yingpeng Wan
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China
- Center of Super-Diamond and Advanced Films (COSDAF), Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon 999077, Hong Kong SAR, P. R. China
| | - Yijian Gao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Wei-Chih Wei
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Ka-Wai Lee
- Center of Super-Diamond and Advanced Films (COSDAF), Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon 999077, Hong Kong SAR, P. R. China
| | - Ji-Hua Tan
- Center of Super-Diamond and Advanced Films (COSDAF), Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon 999077, Hong Kong SAR, P. R. China
| | - Chung-Yu Chen
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Huan Chen
- Center of Super-Diamond and Advanced Films (COSDAF), Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon 999077, Hong Kong SAR, P. R. China
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Ken-Tsung Wong
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Institute of Atomic and Molecular Science, Academia Sinica, Taipei 10617, Taiwan
| | - Chun-Sing Lee
- Center of Super-Diamond and Advanced Films (COSDAF), Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon 999077, Hong Kong SAR, P. R. China
| |
Collapse
|
37
|
Chen X, Li Y, Su J, Zhang L, Liu H. Progression in Near-Infrared Fluorescence Imaging Technology for Lung Cancer Management. BIOSENSORS 2024; 14:501. [PMID: 39451714 PMCID: PMC11506746 DOI: 10.3390/bios14100501] [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: 08/28/2024] [Revised: 10/01/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024]
Abstract
Lung cancer is a major threat to human health and a leading cause of death. Accurate localization of tumors in vivo is crucial for subsequent treatment. In recent years, fluorescent imaging technology has become a focal point in tumor diagnosis and treatment due to its high sensitivity, strong selectivity, non-invasiveness, and multifunctionality. Molecular probes-based fluorescent imaging not only enables real-time in vivo imaging through fluorescence signals but also integrates therapeutic functions, drug screening, and efficacy monitoring to facilitate comprehensive diagnosis and treatment. Among them, near-infrared (NIR) fluorescence imaging is particularly prominent due to its improved in vivo imaging effect. This trend toward multifunctionality is a significant aspect of the future advancement of fluorescent imaging technology. In the past years, great progress has been made in the field of NIR fluorescence imaging for lung cancer management, as well as the emergence of new problems and challenges. This paper generally summarizes the application of NIR fluorescence imaging technology in these areas in the past five years, including the design, detection principles, and clinical applications, with the aim of advancing more efficient NIR fluorescence imaging technologies to enhance the accuracy of tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Xinglong Chen
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China; (X.C.); (Y.L.); (J.S.)
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Yuning Li
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China; (X.C.); (Y.L.); (J.S.)
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Jialin Su
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China; (X.C.); (Y.L.); (J.S.)
| | - Lemeng Zhang
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China; (X.C.); (Y.L.); (J.S.)
| | - Hongwen Liu
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China;
| |
Collapse
|
38
|
Cheng P, Pu K. Enzyme-responsive, multi-lock optical probes for molecular imaging and disease theranostics. Chem Soc Rev 2024; 53:10171-10188. [PMID: 39229642 DOI: 10.1039/d4cs00335g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Optical imaging is an indispensable tool for non-invasive visualization of biomolecules in living organisms, thereby offering a sensitive approach for disease diagnosis and image-guided disease treatment. Single-lock activatable optical probes (SOPs) that specifically switch on optical signals in the presence of biomarkers-of-interest have shown both higher detection sensitivity and imaging quality as compared to conventional "always-on" optical probes. However, such SOPs can still show "false-positive" results in disease diagnosis due to non-specific biomarker expression in healthy tissues. By contrast, multi-lock activatable optical probes (MOPs) that simultaneously detect multiple biomarkers-of-interest could improve detection specificity towards certain biomolecular events or pathological conditions. In this Review, we discuss the recent advancements of enzyme-responsive MOPs, with a focus on their biomedical applications. The higher detection specificity of MOPs could in turn enhance disease diagnosis accuracy and improve treatment efficacy in image-guided disease therapy with minimal toxicity in the surrounding healthy tissues. Finally, we discuss the current challenges and suggest future applications of MOPs.
Collapse
Affiliation(s)
- Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, 637457 Singapore, Singapore.
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, 637457 Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| |
Collapse
|
39
|
Qi Y, Xu M, Lu H, Wang X, Peng Y, Wang Z, Liang F, Jiang X, Du B. Hepatic Biotransformation of Renal Clearable Gold Nanoparticles for Noninvasive Detection of Liver Glutathione Level via Urinalysis. Angew Chem Int Ed Engl 2024; 63:e202409477. [PMID: 38877855 DOI: 10.1002/anie.202409477] [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: 05/20/2024] [Indexed: 07/31/2024]
Abstract
Renal clearable nanoparticles have been drawing much attention as they can avoid prolonged accumulation in the body by efficiently clearing through the kidneys. While much effort has been made to understand their interactions within the kidneys, it remains unclear whether their transport could be influenced by other organs, such as the liver, which plays a crucial role in metabolizing and eliminating both endogenous and exogenous substances through various biotransformation processes. Here, by utilizing renal clearable IRDye800CW conjugated gold nanocluster (800CW4-GS18-Au25) as a model, we found that although 800CW4-GS18-Au25 strongly resisted serum-protein binding and exhibited minimal accumulation in the liver, its surface was still gradually modified by hepatic glutathione-mediated biotransformation when passing through the liver, resulting in the dissociation of IRDye800CW from Au25 and biotransformation-generated fingerprint message of 800CW4-GS18-Au25 in urine, which allowed us to facilely quantify its urinary biotransformation index (UBI) via urine chromatography analysis. Moreover, we observed the linear correlation between UBI and hepatic glutathione concentration, offering us a noninvasive method for quantitative detection of liver glutathione level through a simple urine test. Our discoveries would broaden the fundamental understanding of in vivo transport of nanoparticles and advance the development of urinary probes for noninvasive biodetection.
Collapse
Affiliation(s)
- Yuming Qi
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, P.R. China
| | - Mingze Xu
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, P.R. China
| | - Huixu Lu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Xiaoxian Wang
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, P.R. China
| | - Yexi Peng
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, P.R. China
| | - Ziyuan Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Fengying Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Xingya Jiang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
| | - Bujie Du
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, P.R. China
| |
Collapse
|
40
|
He J, Liang C, Yu XH, Ma X, Qu Y, Zhuang WR, Li W, Nie W, Ren Y, Lei Y, Dong Y, Xie HY. Chemistry-Enabled Intercellular Enzymatic Labeling for Monitoring the Immune Effects of Cytotoxic T Lymphocytes In Vivo. Anal Chem 2024. [PMID: 39140208 DOI: 10.1021/acs.analchem.4c02862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Monitoring the effector function of cytotoxic T lymphocytes (CTLs) in vivo remains a great challenge. Here, we develop a chemistry-enabled enzymatic labeling approach to evaluate the tumor-specific immune response of CTLs by precisely monitoring the interaction between CTLs and tumor cells. Staphylococcus aureus sortase A (SrtA) is linked to the CTL surface through bioconjugate chemistry and then catalyzes the transfer of fluorescent-labeled substrate, 5-Tamra-LPETG, to CTLs. Meanwhile, the tumor cells are specifically decorated with N-terminal glycine residues (G5 peptide) through the inherent glycolmetabolism of cathepsin B-specific cleavable triacetylated N-azidoacetyl-d-mannosamine (CB-Ac3ManNAz) and click chemistry. After the infiltration of engineered CTLs into the tumor tissues, the immune-synapse-mediated specific interaction of CTLs and tumor cells leads to the accurate fluorescent labeling of tumor cells through the SrtA-catalyzed 5-Tamra-LPETG transfer. Therefore, the immune effect of CTLs as well as the performance of immune drugs can be determined, providing a novel strategy for pushing ahead immunotherapy.
Collapse
Affiliation(s)
- Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Xin-He Yu
- State Key Laboratory of Green Pesticide, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yun Qu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Wan-Ru Zhuang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing 100191, P. R. China
| | - Wenzhe Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing 100191, P. R. China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yue Ren
- Beijing Key Laboratory of Construction Tailorable Advanced Functional Materials and Green Applications, School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yao Lei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing 100191, P. R. China
| | - Yuping Dong
- Beijing Key Laboratory of Construction Tailorable Advanced Functional Materials and Green Applications, School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
41
|
Wang X, Liew SS, Huang J, Hu Y, Wei X, Pu K. Dual-Locked Enzyme-Activatable Bioorthogonal Fluorescence Turn-On Imaging of Senescent Cancer Cells. J Am Chem Soc 2024; 146:22689-22698. [PMID: 39101919 DOI: 10.1021/jacs.4c07286] [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: 08/06/2024]
Abstract
Bioorthogonal pretargeting optical imaging shows the potential for enhanced diagnosis and prognosis. However, the bioorthogonal handles, known for being "always reactive", may engage in reactions at unintended sites with their counterparts, resulting in nonspecific fluorescence activation and diminishing detection specificity. Meanwhile, despite the importance of detecting senescent cancer cells in cancer therapy, current methods mainly rely on common single senescence-associated biomarkers, which lack specificity for differentiating between various types of senescent cells. Herein, we report a dual-locked enzyme-activatable bioorthogonal fluorescence (DEBOF) turn-on imaging approach for the specific detection of senescent cancer cells. A dual-locked bioorthogonal targeting agent (DBTA) and a bioorthogonally activatable fluorescent imaging probe (BAP) are synthesized as the biorthogonal pair. DBTA is a tetrazine derivative dually caged by two enzyme-cleavable moieties, respectively, associated with senescence and cancer, which ensures that its bioorthogonal reactivity ("clickability") is only triggered in the presence of senescent cancer cells. BAP is a fluorophore caged by trans-cyclooctane (TCO), whose fluorescence is only activated upon bioorthogonal reaction between its TCO and the decaged tetrazine of DBTA. As such, the DEBOF imaging approach differentiates senescent cancer cells from nonsenescent cancer cells or other senescent cells, allowing noninvasive tracking of the population fluctuation of senescent cancer cells in the tumor of living mice to guide cancer therapies. This study thus provides a general molecular strategy for biomarker-activatable in vivo bioorthogonal pretargeting imaging with the potential to be applied to other imaging modalities beyond optics.
Collapse
Affiliation(s)
- Xinzhu Wang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Si Si Liew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Jingsheng Huang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Yuxuan Hu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Xin Wei
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| |
Collapse
|
42
|
Shen Y, Li W, Zhou Z, Xu J, Li Y, Li H, Zheng X, Liu S, Zhang XB, Yuan L. Dual-Locked Fluorescent Probes Activated by Aminopeptidase N and the Tumor Redox Environment for High-Precision Imaging of Tumor Boundaries. Angew Chem Int Ed Engl 2024; 63:e202406332. [PMID: 38781113 DOI: 10.1002/anie.202406332] [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: 04/03/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 05/25/2024]
Abstract
Clear delineation of tumor margins is essential for accurate resection and decreased recurrence rate in the clinic. Fluorescence imaging is emerging as a promising alternative to traditional visual inspection by surgeons for intraoperative imaging. However, traditional probes lack accuracy in tumor diagnosis, making it difficult to depict tumor boundaries accurately. Herein, we proposed an offensive and defensive integration (ODI) strategy based on the "attack systems (invasive peptidase) and defense systems (reductive microenvironment)" of multi-dimensional tumor characteristics to design activatable fluorescent probes for imaging tumor boundaries precisely. Screened out from a series of ODI strategy-based probes, ANQ performed better than traditional probes based on tumor unilateral correlation by distinguishing between tumor cells and normal cells and minimizing false-positive signals from living metabolic organs. To further improve the signal-to-background ratio in vivo, derivatized FANQ, was prepared and successfully applied to distinguish orthotopic hepatocellular carcinoma tissues from adjacent tissues in mice models and clinical samples. This work highlights an innovative strategy to develop activatable probes for rapid diagnosis of tumors and high-precision imaging of tumor boundaries, providing more efficient tools for future clinical applications in intraoperative assisted resection.
Collapse
Affiliation(s)
- Yang Shen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Wei Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
- School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, P. R. China
| | - Zhixuan Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Junchao Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Yuhang Li
- Department of Hepatobiliary Surgery/ Central Laboratory, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, P. R. China
| | - Haiyan Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Xudong Zheng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Sulai Liu
- Department of Hepatobiliary Surgery/ Central Laboratory, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, P. R. China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
43
|
Xiang K, Pan J, Yu J, Xiao L, Sun SK, Cheng R. A hemicyanine-based near-infrared fluorescent probe with large Stokes shift for non-invasive bioimaging of brown adipose tissue. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:5272-5279. [PMID: 39016035 DOI: 10.1039/d4ay00658e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Brown adipose tissue (BAT), characterized by the presence of numerous mitochondria, plays a key role in metabolism and energy expenditure. Accurately reporting the presence and activation of BAT is beneficial to study obesity, diabetes, and other metabolic disorders. Near-infrared (NIR) fluorescence imaging has the advantages of high sensitivity, non-radioactivity, and simple operation. However, most NIR probes for BAT imaging exhibit small Stokes shifts, which may lead to self-quenching, reducing the signal-to-noise ratio, and introducing cross-talk. Herein, we rationally designed and synthesized a series of hemicyanine-based NIR fluorescent probes HCYBAT-1-3. Among them, HCYBAT-1 demonstrated favorable properties such as near-infrared emission (776 nm), large Stokes shift (139 nm), good biocompatibility and specific mitochondrial targeting (Pearson's colocalization coefficient of 0.87). Meanwhile, HCYBAT-1 was successfully employed to differentiate BAT from white adipose tissue (WAT). Quantitative analysis of NIR fluorescent images showed that HCYBAT-1 could be used for real-time monitoring of BAT activation in mice stimulated by norepinephrine (NE) and cold exposure. Overall, probe HCYBAT-1 showcased its efficacy in non-invasive evaluation of BAT metabolism in vivo with high selectivity and sensitivity.
Collapse
Affiliation(s)
- Ke Xiang
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Jinbin Pan
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jiaojiao Yu
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Lehui Xiao
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Ran Cheng
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China.
| |
Collapse
|
44
|
Yue R, Li Z, Liu H, Wang Y, Li Y, Yin R, Yin B, Qian H, Kang H, Zhang X, Song G. Imaging-guided companion diagnostics in radiotherapy by monitoring APE1 activity with afterglow and MRI imaging. Nat Commun 2024; 15:6349. [PMID: 39068156 PMCID: PMC11283504 DOI: 10.1038/s41467-024-50688-0] [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: 11/15/2023] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
Companion diagnostics using biomarkers have gained prominence in guiding radiotherapy. However, biopsy-based techniques fail to account for real-time variations in target response and tumor heterogeneity. Herein, we design an activated afterglow/MRI probe as a companion diagnostics tool for dynamically assessing biomarker apurinic/apyrimidinic endonuclease 1(APE1) during radiotherapy in vivo. We employ ultrabright afterglow nanoparticles and ultrasmall FeMnOx nanoparticles as dual contrast agents, significantly broadening signal change range and enhancing the sensitivity of APE1 imaging (limit of detection: 0.0092 U/mL in afterglow imaging and 0.16 U/mL in MRI). We devise longitudinally and transversely subtraction-enhanced imaging (L&T-SEI) strategy to markedly enhance MRI contrast and signal-to-noise ratio between tumor and normal tissue of living female mice. The combined afterglow and MRI facilitate both anatomical and functional imaging of APE1 activity. This probe enables correlation of afterglow and MRI signals with APE1 expression, radiation dosage, intratumor ROS, and DNA damage, enabling early prediction of radiotherapy outcomes (as early as 3 h), significantly preceding tumor size reduction (6 days). By monitoring APE1 levels, this probe allows for early and sensitive detection of liver organ injury, outperforming histopathological analysis. Furthermore, MRI evaluates APE1 expression in radiation-induced abscopal effects provides insights into underlying mechanisms, and supports the development of treatment protocols.
Collapse
Affiliation(s)
- Renye Yue
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, PR China
| | - Zhe Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Huiyi Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Youjuan Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Yuhang Li
- Department of Hepatobiliary Surgery/Central Laboratory, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, PR China
| | - Rui Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Baoli Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, PR China
| | - Heemin Kang
- Department of Materials Science and Engineering and College of Medicine, Korea University, Seoul, South Korea
| | - Xiaobing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China.
- Shenzhen Research Institute, Hunan University, Shenzhen, China.
| |
Collapse
|
45
|
Fu J, Xi H, Cai S, Peng Y, Liu Q, Qiu L, Lin J. Development of Granzyme B-targeted Smart Positron Emission Tomography Probes for Monitoring Tumor Early Response to Immunotherapy. ACS NANO 2024; 18:18910-18921. [PMID: 39001856 DOI: 10.1021/acsnano.4c01157] [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: 07/15/2024]
Abstract
Granzyme B is an immune-related biomarker that closely correlates with cytotoxic T lymphocytes (CTLs), and hence detecting the expression level of granzyme B can provide a dependable scheme for clinical immune response assessment. In this study, two positron emission tomography (PET) probes [18F]SF-M-14 and [18F]SF-H-14 targeting granzyme B are designed based on the intramolecular cyclization scaffold SF. [18F]SF-M-14 and [18F]SF-H-14 can respond to granzyme B and glutathione (GSH) to conduct intramolecular cyclization and self-assemble into nanoaggregates to enhance the retention of probe at the target site. Both probes are prepared with high radiochemical purity (>98%) and high stability in PBS and mouse serum. In 4T1 cells cocultured with T lymphocytes, [18F]SF-M-14 and [18F]SF-H-14 reach the maximum uptake of 6.71 ± 0.29 and 3.47 ± 0.09% ID/mg at 0.5 h, respectively, but they remain below 1.95 ± 0.22 and 1.47 ± 0.21% ID/mg in 4T1 cells without coculture of T lymphocytes. In vivo PET imaging shows that the tumor uptake in 4T1-tumor-bearing mice after immunotherapy is significantly higher (3.5 times) than that in the untreated group. The maximum tumor uptake of [18F]SF-M-14 and [18F]SF-H-14 in the mice treated with BEC was 4.08 ± 0.16 and 3.43 ± 0.12% ID/g, respectively, while that in the untreated mice was 1.04 ± 0.79 and 1.41 ± 0.11% ID/g, respectively. These results indicate that both probes have great potential in the early evaluation of clinical immunotherapy efficacy.
Collapse
Affiliation(s)
- Jiayu Fu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hongjie Xi
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Shuyue Cai
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ying Peng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Qingzhu Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Ling Qiu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jianguo Lin
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
46
|
Jiao M, Li X, Liu H, Cai P, Yang X, McHugh KJ, Zheng B, Sun J, Zhang P, Luo X, Jing L. Aqueous Grown Quantum Dots with Robust Near-Infrared Fluorescence for Integrated Traumatic Brain Injury Diagnosis and Surgical Monitoring. ACS NANO 2024; 18:19038-19053. [PMID: 38979966 DOI: 10.1021/acsnano.4c03123] [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: 07/10/2024]
Abstract
Surgical intervention is the most common first-line treatment for severe traumatic brain injuries (TBIs) associated with high intracranial pressure, while the complexity of these surgical procedures often results in complications. Surgeons often struggle to comprehensively evaluate the TBI status, making it difficult to select the optimal intervention strategy. Here, we introduce a fluorescence imaging-based technology that uses high-quality silver indium selenide-based quantum dots (QDs) for integrated TBI diagnosis and surgical guidance. These engineered, poly(ethylene glycol)-capped QDs emit in the near-infrared region, are resistant to phagocytosis, and importantly, are ultrastable after the epitaxial growth of an aluminum-doped zinc sulfide shell in the aqueous phase that renders the QDs resistant to long-term light irradiation and complex physiological environments. We found that intravenous injection of QDs enabled both the precise diagnosis of TBI in a mouse model and, more importantly, the comprehensive evaluation of the TBI status before, during, and after an operation to distinguish intracranial from superficial hemorrhages, provide real-time monitoring of the secondary hemorrhage, and guide the decision making on the evacuation of intracranial hematomas. This QD-based diagnostic and monitoring system could ultimately complement existing clinical tools for treating TBI, which may help surgeons improve patient outcomes and avoid unnecessary procedures.
Collapse
Affiliation(s)
- Mingxia Jiao
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Xiaoqi Li
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Hui Liu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Peng Cai
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Xiling Yang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Kevin J McHugh
- Departments of Bioengineering and Chemistry, Rice University, Houston, Texas 77005, United States
| | - Bowen Zheng
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Jiachen Sun
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Center for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Peisen Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Center for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Xiliang Luo
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Lihong Jing
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Center for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| |
Collapse
|
47
|
Wen X, Zhang C, Tian Y, Miao Y, Liu S, Xu JJ, Ye D, He J. Smart Molecular Imaging and Theranostic Probes by Enzymatic Molecular In Situ Self-Assembly. JACS AU 2024; 4:2426-2450. [PMID: 39055152 PMCID: PMC11267545 DOI: 10.1021/jacsau.4c00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Enzymatic molecular in situ self-assembly (E-MISA) that enables the synthesis of high-order nanostructures from synthetic small molecules inside a living subject has emerged as a promising strategy for molecular imaging and theranostics. This strategy leverages the catalytic activity of an enzyme to trigger probe substrate conversion and assembly in situ, permitting prolonging retention and congregating many molecules of probes in the targeted cells or tissues. Enhanced imaging signals or therapeutic functions can be achieved by responding to a specific enzyme. This E-MISA strategy has been successfully applied for the development of enzyme-activated smart molecular imaging or theranostic probes for in vivo applications. In this Perspective, we discuss the general principle of controlling in situ self-assembly of synthetic small molecules by an enzyme and then discuss the applications for the construction of "smart" imaging and theranostic probes against cancers and bacteria. Finally, we discuss the current challenges and perspectives in utilizing the E-MISA strategy for disease diagnoses and therapies, particularly for clinical translation.
Collapse
Affiliation(s)
- Xidan Wen
- Department
of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing 210008, China
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Chao Zhang
- Department
of Neurosurgery, Zhujiang Hospital, Southern
Medical University, Guangzhou 510282, China
| | - Yuyang Tian
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Yinxing Miao
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Shaohai Liu
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Jing-Juan Xu
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Deju Ye
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Jian He
- Department
of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing 210008, China
| |
Collapse
|
48
|
Zhou D, Zhang Z, Pan L, Wang Y, Yang J, Gao Y, Song Y. Sucrose-Powered Liposome Nanosensors for Urinary Glucometer-Based Monitoring of Cancer. Angew Chem Int Ed Engl 2024; 63:e202404493. [PMID: 38687277 DOI: 10.1002/anie.202404493] [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: 03/05/2024] [Revised: 04/12/2024] [Accepted: 04/30/2024] [Indexed: 05/02/2024]
Abstract
Timely detection of early-stage cancer holds immense potential in enhancing prognostic outcomes. There is an increasing desire for versatile tools to enable simple, sensitive, and cost-effective cancer detection. By exploiting the extraintestinal metabolic inertness and efficiency renal clearance of sucrose, we designed a liposome nanosensor using sucrose as a messenger to convert tumor-specific esterase activity into glucose meter readout, enabling economical and sensitive urinalysis for cancer detection in point-of-care testing (POCT). Our results demonstrate that the nanosensors exhibited significant signal differences between tumor-bearing and healthy mice in both orthotopic and metastatic tumor models. Additionally, efficient elimination of the nanosensors through the hepatobiliary pathway was observed with no significant toxicity. Such a non-invasive diagnostic modality significantly assists in personalized pharmacological treatment and follow-up efficacy assessment. We envision that this modular liposome nanosensor platform might be applied for economically detecting diverse diseases via a simple urinary test.
Collapse
Affiliation(s)
- Dongtao Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, China
| | - Zhibin Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, China
| | - Liqing Pan
- State Key Laboratory of Analytical Chemistry for Life Science, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, China
| | - Yanyi Wang
- State Key Laboratory of Analytical Chemistry for Life Science, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, China
| | - Jingjing Yang
- Department of Biochemistry and Molecular Biology Department, School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanfeng Gao
- School of Medical Imaging, Wannan Medical College, Wuhu, 241002, China
| | - Yujun Song
- State Key Laboratory of Analytical Chemistry for Life Science, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
49
|
Wei X, Xu C, Cheng P, Hu Y, Liu J, Xu M, Huang J, Zhang Y, Pu K. Leveraging Long-Distance Singlet-Oxygen Transfer for Bienzyme-Locked Afterglow Imaging of Intratumoral Granule Enzymes. J Am Chem Soc 2024; 146:17393-17403. [PMID: 38860693 DOI: 10.1021/jacs.4c05012] [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: 06/12/2024]
Abstract
Dual-locked activatable optical probes, leveraging the orthogonal effects of two biomarkers, hold great promise for the specific imaging of biological processes. However, their design approaches are limited to a short-distance energy or charge transfer mechanism, while the signal readout relies on fluorescence, which inevitably suffers from tissue autofluorescence. Herein, we report a long-distance singlet oxygen transfer approach to develop a bienzyme-locked activatable afterglow probe (BAAP) that emits long-lasting self-luminescence without real-time light excitation for the dynamic imaging of an intratumoral granule enzyme. Composed of an immuno-biomarker-activatable singlet oxygen (1O2) donor and a cancer-biomarker-activatable 1O2 acceptor, BAAP is initially nonafterglow. Only in the presence of both immune and cancer biomarkers can 1O2 be generated by the activated donor and subsequently diffuse toward the activated acceptor, resulting in bright near-infrared afterglow with a high signal-to-background ratio and specificity toward an intratumoral granule enzyme. Thus, BAAP allows for real-time tracking of tumor-infiltrating cytotoxic T lymphocytes, enabling the evaluation of cancer immunotherapy and the differentiation of tumor from local inflammation with superb sensitivity and specificity, which are unachievable by single-locked probes. Thus, this study not only presents the first dual-locked afterglow probe but also proposes a new design way toward dual-locked probes via reactive oxygen species transfer processes.
Collapse
Affiliation(s)
- Xin Wei
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Yuxuan Hu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Jingsheng Huang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | - Yan Zhang
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| |
Collapse
|
50
|
Xu L, Deng Y, Gao H, Yao Y, Liu X, Zhan W, Liang G, Sun X. Near-infrared AIEgens for sulfatase imaging in breast cancer in vivo. NANOSCALE 2024; 16:11538-11541. [PMID: 38841880 DOI: 10.1039/d4nr01314j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Aggregation-induced emission luminogens (AIEgens) enable highly sensitive and in situ visualization of sulfatase to benefit the early diagnosis of breast cancer (BC), but current sulfatase AIEgens always emit visible light (<650 nm). Herein, a near-infrared (NIR) AIEgen QMT-SFA is developed for sulfatase imaging in vivo. Hydrophilic QMT-SFA is cleaved by sulfatase to yield hydrophobic QMT-OH, which subsequently aggregates into nanoparticles to turn the AIE fluorescence "on", enabling sensitive sulfatase imaging in 4T1 cells and mouse models.
Collapse
Affiliation(s)
- Lingling Xu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Yu Deng
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Hang Gao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yuchen Yao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Wenjun Zhan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
- Handan Norman Technology Co., Ltd, Guantao 057750, China
| | - Xianbao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
| |
Collapse
|