1
|
Liang S, Zhao D, Liu X, Liu B, Li Y. The stomach, small intestine, and colon-specific gastrointestinal tract delivery systems for bioactive nutrients. Adv Colloid Interface Sci 2025; 341:103503. [PMID: 40209595 DOI: 10.1016/j.cis.2025.103503] [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: 07/29/2024] [Revised: 02/13/2025] [Accepted: 03/29/2025] [Indexed: 04/12/2025]
Abstract
Oral administration is a convenient way to deliver bioactive nutrients. However, the complex and dynamic environment of the gastrointestinal (GI) tract poses distinct challenges. These include the acidic environment of the stomach, limited transport across the GI mucosa, and the risk of enzymatic degradation, all of which can compromise the nutritional effectiveness of orally delivered nutrients. In response to these challenges, various GI tract delivery systems have been developed to target specific regions, such as the stomach, small intestine, or colon, to precisely control the release of bioactive nutrients and enhance their health-promoting benefits. This review critically examines the principles underlying stomach-, small intestine-, and colon-targeted delivery systems, highlighting the selection of appropriate wall materials and the interactions between delivery systems and the mucosal epithelial barrier. Moreover, we describe relevant biological models and quantitative analyses to measure these interactions. In particular, we emphasize the significant advantages offered by colon-targeted delivery systems in maintaining a healthy colonic microenvironment. This review aims to inspire novel concepts and stimulate further research into GI tract delivery systems, offering promising avenues for maximizing the therapeutic effects of bioactive nutrients in practical applications.
Collapse
Affiliation(s)
- Shuang Liang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; Central Laboratory, NMPA Key Laboratory for Dental Materials, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Dongyu Zhao
- Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xiangyu Liu
- Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Bin Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yuan Li
- Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
2
|
Yin L, Xu P, Huang Y, Gu X, Sun L, Zhou H, Zhou W, Xie C, Fan Q. Glutathione-Responsive Near-Infrared-II Fluorescence Probe for Early and Accurate Detection of In Situ and Metastatic Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503257. [PMID: 40434227 DOI: 10.1002/smll.202503257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 05/14/2025] [Indexed: 05/29/2025]
Abstract
In situ and metastatic malignant tumors are primary diseases that threaten human life. Among all the metastases, liver metastasis is the most difficult to detect. As most imaging probes have high liver accumulation, it is difficult to distinguish tiny metastases from normal liver tissue with strong background signal. In this study, the design of a novel second near-infrared window (NIR-II) fluorescence probe for precise detection of carcinoma in situ and liver metastases is presented. The probe called Tg-RGD utilizes a commercially available cyanine dye IR-806 as the signaling moiety, a disulfide bond linker as the responsive moiety, an RGD-capped poly(ethylene glycol) (PEG) as the water soluble enhancer, and the tumor targeting moiety. Tg-RGD shows good glutathione (GSH) responsiveness and selectivity, where its NIR-II fluorescence intensity can enhance 50-fold after activation. In vivo study indicates that Tg-RGD shows much better imaging and targeting effects than Tg-PEG with a similar structure but without RGD moiety for both orthotopic breast cancer and osteosarcoma. Most importantly, Tg-RGD can detect tiny liver metastases with high signal-to-background ratio (3.2). Thus, this study reports a high-performance tumor-specific NIR-II fluorescence probe for in situ and tiny metastatic tumor detection, and may further broaden the applications into related tumor lesions.
Collapse
Affiliation(s)
- Likun Yin
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Pu Xu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Yuxin Huang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Xuxuan Gu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Liwen Sun
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Hui Zhou
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Wen Zhou
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Chen Xie
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Quli Fan
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| |
Collapse
|
3
|
Lee D, Jeon HJ, Jang D, Lee D, Kim S, Han M, Jung SJ, Lee J, Choi J, Kim DH, Ahn DJ, Kim K, Kim S, Lee HJ, Suh S. Engineering Bacterial Secretion Systems for Enhanced Tumor Imaging and Surgical Guidance. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2504389. [PMID: 40395153 DOI: 10.1002/adma.202504389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/10/2025] [Indexed: 05/22/2025]
Abstract
Current imaging techniques suffer from a lack of specificity and resolution, leading to inaccurate tumor imaging and limited applicability of targeted contrast agents, as they require cancer-specific development. The need for enhanced contrast through improved tumor-to-background ratio (TBR) and the toxicity from repeated injections due to fading fluorescent signals further complicate the issue. Additionally, challenges in visualizing the entire 3D tumor with surface-stained contrast agents highlight the demand for advanced imaging solutions for more precise surgical guidance. A novel approach is proposed utilizing Streptavidin Associated Salmonella (SAS) as a contrast agent for image-guided surgeries. SAS selectively proliferates in cancerous tissues and secretes streptavidin upon induction, enabling the binding of subsequently injected biotin-conjugated fluorescent dyes. This approach enhances tumor visualization with a TBR of up to 15.3, far surpassing conventional agents (TBR ∼ 2), while enabling prolonged 3-day imaging, deep tumor penetration, and precise invasive margin delineation with a single contrast agent injection. Furthermore, biosafety evaluations confirmed efficient bacterial clearance, absence of systemic toxicity, and stable physiological responses, supporting its potential for safe clinical translation. This innovative method offers substantial improvements over existing fluorescent contrast agents and holds promise for both diagnostic and therapeutic applications in cancer surgery.
Collapse
Affiliation(s)
- Dohee Lee
- Bionics Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Heung Jin Jeon
- Cancer Research Institute, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - Dohyub Jang
- Chemical and Biological integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Deukhee Lee
- Bionics Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Solbi Kim
- Cancer Research Institute, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - Minju Han
- Department of Medical Science and Cancer Research Institute, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - Sharon Jiyoon Jung
- Technological Convergence Support Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Junghyun Lee
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jia Choi
- KU-KIST Graduate School of Conversing Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Dong Ha Kim
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Dong June Ahn
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Keri Kim
- Bionics Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sehoon Kim
- KU-KIST Graduate School of Conversing Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyo-Jin Lee
- Cancer Research Institute, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - SeungBeum Suh
- Division of Bio-Medical Science & Technology, University of Science & Technology, Seoul, 02792, Republic of Korea
| |
Collapse
|
4
|
Li Z, Peng T, Yang M, Qiu Y, Ye P, Wang X, Jin H. Dual Functionality of [ 64Cu]Cu-NOTA-San A-Cy7 for Diagnostic Imaging and Surgical Guidance in Hsp90α-Positive Tumors. Mol Pharm 2025. [PMID: 40343454 DOI: 10.1021/acs.molpharmaceut.5c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Intraoperative fluorescence navigation in esophageal cancer enables the clinical translation of fluorescence imaging. Heat shock protein 90 alpha (Hsp90α) plays a vital role in the progression of malignant disease, and elevated Hsp90α expression has been reported in esophageal cancer. The aim of this study was to develop a dual-modality probe, [64Cu]Cu-NOTA-San A-Cy7, for imaging Hsp90α expression in vivo via both positron emission tomography (PET) and fluorescence imaging in esophageal cancer. In this study, the Hsp90α-targeting cyclopeptide Sansalvamide A (San A) was chemically modified with a Cy7 dye and NOTA chelator simultaneously. Experimental assays confirmed that NOTA-San A-Cy7 has a favorable affinity for Hsp90α-positive EC109 cells, with a dissociation constant (Kd) of 1.08 ± 0.19 μM. The probe [64Cu]Cu-NOTA-San A-Cy7 was successfully synthesized with 64CuCl2, achieving a high radiochemical purity of over 95%. Furthermore, the probe demonstrated excellent stability in both saline and serum solutions. The probe was subsequently evaluated in a Hsp90α-positive EC109 tumor-bearing model via PET imaging, which confirmed that Hsp90α-specific uptake was significantly reduced by the co-administration of an excess blocking agent. Biodistribution studies revealed that at 24 hours post-injection, the tumor uptake of the probe was 1.35 ± 0.29%ID/g in the nonblocking group and significantly decreased to 0.73 ± 0.15%ID/g in the blocking group (p < 0.05). Concurrent with the PET experiment, fluorescence imaging was conducted, revealing substantial tumor uptake in the EC109 model. As a proof of concept, imaging-guided surgery utilizing the fluorescent component of this probe was performed. This approach demonstrated the potential for providing surgical guidance in mice positive for Hsp90α, highlighting the dual functionality of the probe for both diagnostic imaging and intraoperative navigation. In summary, our findings unequivocally demonstrate that the dual-modality probe [64Cu]Cu-NOTA-San A-Cy7 holds significant promise as an agent for imaging Hsp90α-positive tumors in vivo, offering a valuable tool for the detection and potential management of such tumors.
Collapse
Affiliation(s)
- Zhijun Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
- Department of Urology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan Province 421001, China
| | - Tukang Peng
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Min Yang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yifan Qiu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Peizhen Ye
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Xiaohui Wang
- Department of Nuclear Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Hongjun Jin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| |
Collapse
|
5
|
Guo C, Gu T, Wen SH, Dang Y, Zhou Y, Ma J, Yu S. Intramolecular enhanced entropy-driven DNA-Au nanodevice for mRNA imaging in living cells. Mikrochim Acta 2025; 192:235. [PMID: 40095194 DOI: 10.1007/s00604-025-06997-2] [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: 11/26/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025]
Abstract
An intramolecular enhanced entropy-driven DNA amplifier-tethered gold nanoparticle (DNA-Au) nanodevice has been designed for highly sensitive in situ imaging of messenger ribonucleic acid (mRNA) in living cells. The DNA amplifier is immobilized on a same AuNP and the initial fluorescence of DNA-Au nanodevice is quenched. Upon internalized into the target cancer cells, the nanodevice can be activated by endogenous TK1 mRNA, and promptly release the fluorophore via the intramolecular enhanced DNA strand displacement reaction. The decreasing distance and increasing local concentration of the probes via intramolecular reaction can significantly improve the reaction kinetics of DNA-Au nanodevice, thus achieving the highly sensitive imaging of TK1 mRNA. The excellent sensitivity and selectivity allow the DNA-Au nanodevice to accurately discriminate different cell lines and monitor the variations in intracellular TK1 mRNA expression levels via fluorescence imaging. Therefore, the proposed intramolecular enhanced entropy-driven DNA-Au nanodevice will afford a reliable approach for accurate determination of mRNA in molecular diagnostic systems.
Collapse
Affiliation(s)
- Chao Guo
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China
| | - Tongnian Gu
- Sunresin New Materials Co. Ltd., Xi'an Hi-tech Industrial Development Zone, Xi'an, 710076, P.R. China
| | - Shao-Hua Wen
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China
| | - Yuan Dang
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China
| | - Yuanzhen Zhou
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China
- Engineering Research Center of Low-Carbon Energy Efficient Utilization, Universities of Shaanxi Province, Xi'an University of Architecture and Technology, Xi'an, 710055, China
| | - Junping Ma
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China.
| | - Sha Yu
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China.
| |
Collapse
|
6
|
Lv R, Hang S, Zhao Y, Gao W, Zhang P, Zheng K, Zhang Q, Ding C. Reactive Oxygen Species (ROS)-Tyrosinase Cascade-Activated Near-Infrared Fluorescent Probe for the Precise Imaging of Melanoma. Anal Chem 2025; 97:4241-4250. [PMID: 39946555 DOI: 10.1021/acs.analchem.5c00018] [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: 02/26/2025]
Abstract
As a highly aggressive malignancy, the issue of curing melanoma at an advanced stage could suffer from severe metastasis and a lower 5-year survival rate. Therefore, the early diagnosis of melanoma with high accuracy is vital and contributes to a significantly improved 5-year survival rate. This work reports a dual-locked receptor, m-BA-Hcy, which releases the near-infrared (NIR) fluorophore Hcy-OH upon the dual activation of reactive oxygen species (ROS) and tyrosinase (TYR). The substitution of boric acid on the phenyl ring was studied, which influences the feasibility of the performance of the envisaged cascade reaction. The sensing behavior was discussed in terms of optical spectroscopy and reaction mechanism, and imaging was fully performed at the cellular and organism levels. Receptor m-BA-Hcy was hence clarified to possess supreme sensitivity and accuracy for melanoma detection.
Collapse
Affiliation(s)
- Ruidian Lv
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Sitong Hang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Yuran Zhao
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Weijie Gao
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Peng Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Ke Zheng
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Qian Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Caifeng Ding
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| |
Collapse
|
7
|
Hou DY, Zhang NY, Zhang P, Li XP, Wu JC, Lv MY, Wang ZJ, Hu XJ, Liang JX, Wang HL, Wang YZ, You HH, An HW, Wang H, Xu W. In vivo self-assembled bispecific fluorescence probe for early detection of bladder cancer and metastasis. Sci Bull (Beijing) 2025; 70:407-418. [PMID: 39537458 DOI: 10.1016/j.scib.2024.10.032] [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: 08/20/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Tumor metastasis accounts for over 90% of tumor-related deaths, prompting the development of fluorescently labeled tumor-specific molecular imaging agents for differentiating tumors from normal tissues. However, early detection of metastasis lesions by tracking tumor markers alone has proven to be challenging. Herein, we reported a glycopeptide-based bispecific fluorescence probe (bsProbe) for earlier detection of bladder cancer and metastasis. By simultaneously recognition (tumor & tumor microenvironment) and in vivo self-assembly, the tumor accumulation of bsProbe (12.3% ID/g) was obviously increased by ∼6 fold compared with that in CXCR4 specific fluorescence probe (sProbe), indicating the obvious advantages of bsProbe over existing tumor metastasis detection probes. Additionally, bsProbe substantially broadens the tumor diagnosis window and enhances the detection signal to noise ratio (SNR: approximately 9.5), permitting early diagnosis of lung micro-metastasis (∼1 mm), precise identifying of tumor boundaries and micro-tumors in orthotopic tumor models. More importantly, bsProbe was demonstrated to distinguish malignant from benign specimen with a specificity of 90.48% and sensitivity of 92.22% in 195 clinical specimens of bladder cancer patients. Taken together, this novel synergetic targeting (CD206 × CXCR4) strategy provides an attractive method for earlier detection of bladder cancer and metastasis, which might be further extended to the imaging-guided surgery of clinical invisible tumors.
Collapse
Affiliation(s)
- Da-Yong Hou
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China; Department of PET-CT/MRI, Harbin Medical University Cancer Hospital, Harbin 150001, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Ni-Yuan Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Peng Zhang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Xiang-Peng Li
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Jiong-Cheng Wu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Mei-Yu Lv
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Zhi-Jia Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Xing-Jie Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Jian-Xiao Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Hong-Lei Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Yue-Ze Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Hui-Hui You
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Hong-Wei An
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Wanhai Xu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
8
|
Lu Y, Shan B, Li L, Jiang R, Li M. Tumor-Homing Biomimetic Near-Infrared II SERS Probes for Targeted Intraoperative Resection Guidance of Orthotopic Glioblastoma. NANO LETTERS 2025; 25:2325-2333. [PMID: 39884958 DOI: 10.1021/acs.nanolett.4c05622] [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: 02/01/2025]
Abstract
In vivo optical imaging holds great potential for surgical guidance with the ability to intraoperatively identify tumor lesions in a surgical bed and navigate their surgical excision in real time. Nevertheless, its full potential remains underexploited, mainly due to the dearth of high-performance optical probes. Herein, hybrid cell membrane-biomimetic near-infrared II surface-enhanced Raman spectroscopy (NIR-II SERS) probes are reported for intraoperative resection guidance of orthotopic glioblastoma. A novel class of plasmonic Au nanorod (AuNR)@Au-Ag frames is developed with remarkable plasmonic properties tunable beyond 1700 nm. We demonstrate the exceptional NIR-II SERS performance both in vitro and in vivo of the biomimetic NIR-II SERS probes created with AuNR@Au-Ag frames and hybrid cell membranes. The biomimetic NIR-II SERS probes are successfully applied in an orthotopic glioblastoma mouse model for intraoperative resection guidance with complete tumor removal and improved surgical outcomes. This study presents a promising strategy for precise NIR-II SERS surgical navigation.
Collapse
Affiliation(s)
- Yaxuan Lu
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Beibei Shan
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Linhu Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Renting Jiang
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
9
|
Bi X, Feng J, Feng X, Li D, Wang Y, Zhao S, Zhang L. APE1-Activated and NIR-II Photothermal-Enhanced Chemodynamic Therapy Guided by Amplified Fluorescence Imaging. Anal Chem 2025; 97:1748-1757. [PMID: 39808438 DOI: 10.1021/acs.analchem.4c05274] [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: 01/16/2025]
Abstract
The development of intelligent nanotheranostic technology that integrates diagnostic and therapeutic functions holds great promise for personalized nanomedicine. However, most of the nanotheranostic agents exhibit "always-on" properties and do not involve an amplification step, which may largely limit imaging contrast and restrict therapeutic efficacy. Herein, we construct a novel nanotheranostic platform (Hemin/DHPs/PDA@CuS nanocomposite) by assembling DNA hairpin probes (DHPs) and hemin on the surface of PDA@CuS nanosheets that enables amplified fluorescence imaging and activatable chemodynamic therapy (CDT) of tumors. The cancer-relevant APE1 triggers nucleic acid amplification with DHPs to generate activatable and amplified fluorescence signals for discriminating cancer cells from normal cells. Meanwhile, excessive G-quadruplex/hemin-based DNAzyme are also activated, and they function as Fenton-like catalysts to catalyze the production of highly toxic hydroxyl radicals (•OH) for CDT. Moreover, owing to the excellent photothermal conversion efficiency in the near-infrared-II (NIR-II) window, the PDA@CuS not only improves the catalytic performance of CDT but also furnishes PTT. A remarkable antitumor therapeutic effect is demonstrated both in vitro and in vivo. Therefore, the Hemin/DHPs/PDA@CuS nanocomposite is expected to provide a promising avenue for precise imaging-guided antitumor therapy.
Collapse
Affiliation(s)
- Xiaofeng Bi
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Jinyue Feng
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Xiyuan Feng
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Dianpeng Li
- Guangxi Key Laboratory of Plant Functional Phytochemicals and Sustainable Utilization, Guangxi Institute of Botany, Guangxi Zhuang Autonomous Region and Chinese Academy of Sciences, Guilin 541006, PR China
| | - Yumin Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Shulin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| | - Liangliang Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, PR China
| |
Collapse
|
10
|
Keizers B, Nijboer TS, van der Fels CAM, van den Heuvel MC, van Dam GM, Kruijff S, Jan de Jong I, Witjes MJH, Voskuil FJ, Gorpas D, Browne WR, van der Zaag PJ. Systematic comparison of fluorescence imaging in the near-infrared and shortwave-infrared spectral range using clinical tumor samples containing cetuximab-IRDye800CW. JOURNAL OF BIOMEDICAL OPTICS 2025; 30:S13708. [PMID: 39553388 PMCID: PMC11566260 DOI: 10.1117/1.jbo.30.s1.s13708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/25/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Significance Shortwave-infrared (SWIR) imaging is reported to yield better contrast in fluorescence-guided surgery than near-infrared (NIR) imaging, due to a reduction in scattering. This benefit of SWIR was shown in animal studies, however not yet in clinical studies with patient samples. Aim We investigate the potential benefit of SWIR to NIR imaging in clinical samples containing cetuximab-IRDye800CW in fluorescence-guided surgery. Approach The potential of the epidermal growth factor-targeted NIR dye cetuximab-IRDye800CW in the shortwave range was examined by recording the absorption and emission spectrum. An ex vivo comparison of NIR and SWIR images using clinical tumor samples of patients with penile squamous cell carcinoma (PSCC) and head and neck squamous cell carcinoma (HNSCC) containing cetuximab-IRDye800CW was performed. The comparison was based on the tumor-to-background ratio and an adapted contrast-to-noise ratio (aCNR) using the standard of care pathology tissue assessment as the golden standard. Results Based on the emission spectrum, cetuximab-IRDye800CW can be detected in the SWIR range. In clinical PSCC samples, overall SWIR imaging was found to perform similarly to NIR imaging (NIR imaging is better than SWIR in the 2/7 criteria examined, and SWIR is better than NIR in the 3/7 criteria). However, when inspecting HNSCC data, NIR is better than SWIR in nearly all (5/7) examined criteria. This difference seems to originate from background autofluorescence overwhelming the off-peak SWIR fluorescence signal in HNSCC tissue. Conclusion SWIR imaging using the targeted tracer cetuximab-IRDye800CW currently does not provide additional benefit over NIR imaging in ex vivo clinical samples. Background fluorescence in the SWIR region, resulting in a higher background signal, limits SWIR imaging in HNSCC samples. However, SWIR shows potential in increasing the contrast of tumor borders in PSCC samples, as shown by a higher aCNR over a line.
Collapse
Affiliation(s)
- Bas Keizers
- University Medical Centre Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Surgery, Groningen, The Netherlands
| | - Thomas S. Nijboer
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | | | - Marius C. van den Heuvel
- University Medical Centre Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | | | - Schelto Kruijff
- University Medical Centre Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Surgery, Groningen, The Netherlands
| | - Igle Jan de Jong
- University Medical Centre Groningen, Department of Urology, Groningen, The Netherlands
| | - Max J. H. Witjes
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Floris J. Voskuil
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Dimitris Gorpas
- Helmholtz Zentrum München, Institute of Biological and Medical Imaging, Neuherberg, Germany
- Technische Universität München, School of Medicine and Health, Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), München, Germany
| | - Wesley R. Browne
- University of Groningen, Stratingh Institute for Chemistry, Faculty of Science and Engineering, Groningen, The Netherlands
| | - Pieter J. van der Zaag
- University Medical Centre Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University of Groningen, Zernike Institute, Molecular Biophysics, Groningen, The Netherlands
| |
Collapse
|
11
|
Zhang C, Cha R, Long K, Liu Y, Dong Y, Zhang Y, Wang X, Jiang X. Functionalized Iron Oxide Nanoparticles for Both Dual-Modal Imaging and Erythropoiesis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:68905-68917. [PMID: 39656520 DOI: 10.1021/acsami.4c15206] [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: 12/20/2024]
Abstract
Cancer-related anemia (CRA), a complication of cancer, is considered the primary cause of high mortality for cancer patients. Safe and effective theranostics are desirable for realizing the high diagnostic accuracy of tumors and ameliorating CRA in the clinic. However, the available theranostics do not support dual-modal imaging and the amelioration of CRA at the same time. In this study, we synthesized functionalized iron oxide nanoparticles (Fe3O4 NPs) modified with protoporphyrin IX (PPIX) and folic acid (FA) by a one-step modification strategy (Fe3O4@NH-PPIX&FA NPs) or a step-by-step strategy (Fe3O4@NH-PPIX-FA NPs), aiming at both magnetic resonance imaging/fluorescence imaging (MRI/FI) and erythropoiesis. Fe3O4@NH-PPIX-FA NPs displayed better ability of MRI/FI than Fe3O4@NH-PPIX&FA NPs and had an efficient tumor targeting of 45 min after tail vein injection owing to the reduction of the steric effect and extension of FA groups. Fe3O4@NH-PPIX-FA NPs exhibited satisfactory erythropoiesis with up to 20% elevation of red blood cell (RBC) counts and hemoglobin concentrations in mice with CRA, which provided a safe alternative to RBC transfusions, especially for patients needing recurrent RBC transfusions. With excellent performance in both dual-modal imaging and erythropoiesis, Fe3O4@NH-PPIX-FA NPs could be a powerful tool for the theranostics of cancer patients with anemia.
Collapse
Affiliation(s)
- Chunliang Zhang
- Laboratory of Theoretical and Computational Nanoscience, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Haidian District, Beijing 100190, P. R. China
- The Ninth Medical Center of PLA General Hospital, No. 9 Anxiang Beili, Chaoyang District, Beijing 100101, P. R. China
| | - Ruitao Cha
- Laboratory of Theoretical and Computational Nanoscience, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Haidian District, Beijing 100190, P. R. China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, No. 2 Tiantan Xi Li, Dongcheng District, Beijing 100050, P. R. China
| | - Keying Long
- Laboratory of Theoretical and Computational Nanoscience, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Haidian District, Beijing 100190, P. R. China
| | - Yang Liu
- Laboratory of Theoretical and Computational Nanoscience, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Haidian District, Beijing 100190, P. R. China
| | - Yanrong Dong
- Institute of Health Service and Transfusion Medicine, Beijing 100850, P. R. China
| | - Yulong Zhang
- Institute of Health Service and Transfusion Medicine, Beijing 100850, P. R. China
| | - Xiaohui Wang
- Institute of Health Service and Transfusion Medicine, Beijing 100850, P. R. China
| | - Xingyu Jiang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, P. R. China
| |
Collapse
|
12
|
Surendran A, Tintu R, Das KS, Nair VJA, Varghese P. Biomedical and Anticancer Potential of Green Synthesized Chalcogenide Zinc Sulfide Nanoparticles Using Different Plant Extracts as the Capping Agent. BRAZILIAN JOURNAL OF PHYSICS 2024; 54:224. [DOI: 10.1007/s13538-024-01591-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/23/2024] [Indexed: 01/05/2025]
|
13
|
Xu M, Li P, Wei J, Yan P, Zhang Y, Guo X, Liu C, Yang X. Progress of fluorescence imaging in lymph node dissection surgery for prostate and bladder cancer. Front Oncol 2024; 14:1395284. [PMID: 39429471 PMCID: PMC11486700 DOI: 10.3389/fonc.2024.1395284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Fluorescence imaging is a relatively new imaging method used to visualize different tissue structures to help guide intraoperative operations, which has potential advantages with high sensitivity and contrast compared to conventional imaging. In this work, we review fluorescent contrast agents and devices used for lymphatic system imaging. Indocyanine green is the most widely utilized due to its high sensitivity, specificity, low background fluorescence, and safety profile. In prostate and bladder cancer lymph node dissection, the complex lymphatic drainage can result in missed metastatic nodes and extensive dissection increases the risk of complications like lymphocele, presenting a significant challenge for urologists. Fluorescence-guided sentinel lymph node dissection facilitates precise tumor staging. The combination of fluorescence and radiographic imaging improves the accuracy of lymph node staging. Multimodal imaging presents new potential for precisely identifying metastatic pelvic lymph nodes.
Collapse
Affiliation(s)
- Mingquan Xu
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, ;China
| | - Panpan Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Jinzheng Wei
- Department of Orthopedics, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Pengyu Yan
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Yunmeng Zhang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Xinyu Guo
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Chao Liu
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, ;China
| | - Xiaofeng Yang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, ;China
| |
Collapse
|
14
|
Vaselli M, Gabriels RY, Schmidt I, Sterkenburg AJ, Kats-Ugurlu G, Nagengast WB, de Boer JF. Ex vivo optical coherence tomography combined with near infrared targeted fluorescence: towards in-vivo esophageal cancer detection. BIOMEDICAL OPTICS EXPRESS 2024; 15:5706-5722. [PMID: 39421768 PMCID: PMC11482167 DOI: 10.1364/boe.537828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 10/19/2024]
Abstract
Early detection of (pre)malignant esophageal lesions is critical to improve esophageal cancer morbidity and mortality rates. In patients with advanced esophageal adenocarcinoma (EAC) who undergo neoadjuvant chemoradiation therapy, the efficacy of therapy could be optimized and unnecessary surgery prevented by the reliable assessment of residual tumors after therapy. Optical coherence tomography (OCT) provides structural images at a (sub)-cellular level and has the potential to visualize morphological changes in tissue. However, OCT lacks molecular imaging contrast, a feature that enables the study of biological processes at a cellular level and can enhance esophageal cancer diagnostic accuracy. We combined OCT with near-infrared fluorescence molecular imaging using fluorescently labelled antibodies (immuno-OCT). The main goal of this proof of principle study is to investigate the feasibility of immuno-OCT for esophageal cancer imaging. We aim to assess whether the sensitivity of our immuno-OCT device is sufficient to detect the tracer uptake using an imaging dose (∼100 times smaller than a dose with therapeutic effects) of a targeted fluorescent agent. The feasibility of immuno-OCT was demonstrated ex-vivo on dysplastic lesions resected from Barrett's patients and on esophageal specimens resected from patients with advanced EAC, who were respectively topically and intravenously administrated with the tracer bevacizumab-800CW. The detection sensitivity of our system (0.3 nM) is sufficient to detect increased tracer uptake with micrometer resolution using an imaging dose of labelled antibodies. Moreover, the absence of layered structures that are typical of normal esophageal tissue observed in OCT images of dysplastic/malignant esophageal lesions may further aid their detection. Based on our preliminary results, immuno-OCT could improve the detection of dysplastic esophageal lesions.
Collapse
Affiliation(s)
- Margherita Vaselli
- Department of Physics and Astronomy, LaserLab Amsterdam, Vrije Universiteit de Boelelaan 1081,, Amsterdam, The Netherlands
| | - Ruben Y. Gabriels
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iris Schmidt
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrea J. Sterkenburg
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gursah Kats-Ugurlu
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wouter B. Nagengast
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johannes F. de Boer
- Department of Physics and Astronomy, LaserLab Amsterdam, Vrije Universiteit de Boelelaan 1081,, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Bhattacharya T, Preetam S, Mukherjee S, Kar S, Roy DS, Singh H, Ghose A, Das T, Mohapatra G. Anticancer activity of quantum size carbon dots: opportunities and challenges. DISCOVER NANO 2024; 19:122. [PMID: 39103694 DOI: 10.1186/s11671-024-04069-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024]
Abstract
Research into the anticancer activity of quantum-sized carbon dots (CDs) has emerged as a promising avenue in cancer research. This CDs delves into the opportunities and challenges associated with harnessing the potential of these nanostructures for combating cancer. Quantum-sized carbon dots, owing to their unique physicochemical properties, exhibit distinct advantages as potential therapeutic agents. Opportunities lie in their tunable size, surface functionalization capabilities, and biocompatibility, enabling targeted drug delivery and imaging in cancer cells. However, we include challenges, a comprehensive understanding of the underlying mechanisms, potential toxicity concerns, and the optimization of synthesis methods for enhanced therapeutic efficacy. A succinct summary of the state of the research in this area is given in this review, emphasizing the exciting possibilities and ongoing challenges in utilizing quantum-sized carbon dots as a novel strategy for cancer treatment.
Collapse
Affiliation(s)
- Tanima Bhattacharya
- Faculty of Applied Science, Lincoln University College, 47301, Petaling Jaya, Selangor Darul Ehsan, Malaysia.
| | - Subham Preetam
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Sohini Mukherjee
- Department of Environmental Science, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019, India
| | - Sanjukta Kar
- Dietetics and Applied Nutrition, Amity University Kolkata, Kadampukur, India
| | | | - Harshita Singh
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Arak Ghose
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Tanmoy Das
- Faculty of Engineering, Lincoln University College, 47301, Petaling Jaya, Selangor Darul Ehsan, Malaysia.
| | - Gautam Mohapatra
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| |
Collapse
|
16
|
Qian K, Gao S, Jiang Z, Ding Q, Cheng Z. Recent advances in mitochondria-targeting theranostic agents. EXPLORATION (BEIJING, CHINA) 2024; 4:20230063. [PMID: 39175881 PMCID: PMC11335472 DOI: 10.1002/exp.20230063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/07/2024] [Indexed: 08/24/2024]
Abstract
For its vital role in maintaining cellular activity and survival, mitochondrion is highly involved in various diseases, and several strategies to target mitochondria have been developed for specific imaging and treatment. Among these approaches, theranostic may realize both diagnosis and therapy with one integrated material, benefiting the simplification of treatment process and candidate drug evaluation. A variety of mitochondria-targeting theranostic agents have been designed based on the differential structure and composition of mitochondria, which enable more precise localization within cellular mitochondria at disease sites, facilitating the unveiling of pathological information while concurrently performing therapeutic interventions. Here, progress of mitochondria-targeting theranostic materials reported in recent years along with background information on mitochondria-targeting and therapy have been briefly summarized, determining to deliver updated status and design ideas in this field to readers.
Collapse
Affiliation(s)
- Kun Qian
- State Key Laboratory of Drug ResearchMolecular Imaging CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Shu Gao
- State Key Laboratory of Drug ResearchMolecular Imaging CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingChina
| | - Zhaoning Jiang
- State Key Laboratory of Drug ResearchMolecular Imaging CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingChina
- Shandong Laboratory of Yantai Drug DiscoveryBohai Rim Advanced Research Institute for Drug DiscoveryYantaiShandongChina
| | - Qihang Ding
- Department of ChemistryKorea UniversitySeoulRepublic of Korea
| | - Zhen Cheng
- State Key Laboratory of Drug ResearchMolecular Imaging CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingChina
- Shandong Laboratory of Yantai Drug DiscoveryBohai Rim Advanced Research Institute for Drug DiscoveryYantaiShandongChina
| |
Collapse
|
17
|
Wang R, Hua S, Xing Y, Wang R, Wang H, Jiang T, Yu F. Organic dye-based photosensitizers for fluorescence imaging-guided cancer phototheranostics. Coord Chem Rev 2024; 513:215866. [DOI: 10.1016/j.ccr.2024.215866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
|
18
|
Ullah Z, Roy S, Muhammad S, Yu C, Huang H, Chen D, Long H, Yang X, Du X, Guo B. Fluorescence imaging-guided surgery: current status and future directions. Biomater Sci 2024; 12:3765-3804. [PMID: 38961718 DOI: 10.1039/d4bm00410h] [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: 07/05/2024]
Abstract
Surgery is one of the most important paradigms for tumor therapy, while fluorescence imaging (FI) offers real-time intraoperative guidance, greatly boosting treatment prognosis. The imaging fidelity heavily relies on not only imaging facilities but also probes for imaging-guided surgery (IGS). So far, a great number of IGS probes with emission in visible (400-700 nm) and near-infrared (NIR 700-1700 nm) windows have been developed for pinpointing disease margins intraoperatively. Herein, the state-of-the-art fluorescent probes for IGS are timely updated, with a special focus on the fluorescent probes under clinical examination. For a better demonstration of the superiority of NIR FI over visible FI, both imaging modalities are critically compared regarding signal-to-background ratio, penetration depth, resolution, tissue autofluorescence, photostability, and biocompatibility. Various types of fluorescence IGS have been summarized to demonstrate its importance in the medical field. Furthermore, the most recent progress of fluorescent probes in NIR-I and NIR-II windows is summarized. Finally, an outlook on multimodal imaging, FI beyond NIR-II, efficient tumor targeting, automated IGS, the use of AI and machine learning for designing fluorescent probes, and the fluorescence-guided da Vinci surgical system is given. We hope this review will stimulate interest among researchers in different areas and expedite the translation of fluorescent probes from bench to bedside.
Collapse
Affiliation(s)
- Zia Ullah
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Shubham Roy
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Saz Muhammad
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
- School of System Design and Intelligent Manufacturing, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chen Yu
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Haiyan Huang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Dongxiang Chen
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Haodong Long
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Xiulan Yang
- School of Computer Science and Engineering, Yulin Normal University, Yulin, 537000, China.
| | - Xuelian Du
- Department of Gynecology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1, Fuhua Road, Futian District, Shenzhen, 518033, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| |
Collapse
|
19
|
Chen S, Zhuang D, Jia Q, Guo B, Hu G. Advances in Noninvasive Molecular Imaging Probes for Liver Fibrosis Diagnosis. Biomater Res 2024; 28:0042. [PMID: 38952717 PMCID: PMC11214848 DOI: 10.34133/bmr.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/08/2024] [Indexed: 07/03/2024] Open
Abstract
Liver fibrosis is a wound-healing response to chronic liver injury, which may lead to cirrhosis and cancer. Early-stage fibrosis is reversible, and it is difficult to precisely diagnose with conventional imaging modalities such as magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography, and ultrasound imaging. In contrast, probe-assisted molecular imaging offers a promising noninvasive approach to visualize early fibrosis changes in vivo, thus facilitating early diagnosis and staging liver fibrosis, and even monitoring of the treatment response. Here, the most recent progress in molecular imaging technologies for liver fibrosis is updated. We start by illustrating pathogenesis for liver fibrosis, which includes capillarization of liver sinusoidal endothelial cells, cellular and molecular processes involved in inflammation and fibrogenesis, as well as processes of collagen synthesis, oxidation, and cross-linking. Furthermore, the biological targets used in molecular imaging of liver fibrosis are summarized, which are composed of receptors on hepatic stellate cells, macrophages, and even liver collagen. Notably, the focus is on insights into the advances in imaging modalities developed for liver fibrosis diagnosis and the update in the corresponding contrast agents. In addition, challenges and opportunities for future research and clinical translation of the molecular imaging modalities and the contrast agents are pointed out. We hope that this review would serve as a guide for scientists and students who are interested in liver fibrosis imaging and treatment, and as well expedite the translation of molecular imaging technologies from bench to bedside.
Collapse
Affiliation(s)
- Shaofang Chen
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Danping Zhuang
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Qingyun Jia
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application,
Harbin Institute of Technology, Shenzhen 518055, China
| | - Genwen Hu
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| |
Collapse
|
20
|
White HW, Naveed AB, Campbell BR, Lee YJ, Baik FM, Topf M, Rosenthal EL, Hom ME. Infrared Fluorescence-guided Surgery for Tumor and Metastatic Lymph Node Detection in Head and Neck Cancer. Radiol Imaging Cancer 2024; 6:e230178. [PMID: 38940689 PMCID: PMC11287229 DOI: 10.1148/rycan.230178] [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: 10/09/2023] [Revised: 05/13/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024]
Abstract
In patients with head and neck cancer (HNC), surgical removal of cancerous tissue presents the best overall survival rate. However, failure to obtain negative margins during resection has remained a steady concern over the past 3 decades. The need for improved tumor removal and margin assessment presents an ongoing concern for the field. While near-infrared agents have long been used in imaging, investigation of these agents for use in HNC imaging has dramatically expanded in the past decade. Targeted tracers for use in primary and metastatic lymph node detection are of particular interest, with panitumumab-IRDye800 as a major candidate in current studies. This review aims to provide an overview of intraoperative near-infrared fluorescence-guided surgery techniques used in the clinical detection of malignant tissue and sentinel lymph nodes in HNC, highlighting current applications, limitations, and future directions for use of this technology within the field. Keywords: Molecular Imaging-Cancer, Fluorescence © RSNA, 2024.
Collapse
Affiliation(s)
- Haley W. White
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Abdullah Bin Naveed
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Benjamin R. Campbell
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Yu-Jin Lee
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Fred M. Baik
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Michael Topf
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Eben L. Rosenthal
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Marisa E. Hom
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| |
Collapse
|
21
|
Qin T, Huang M, Wei W, Zhou W, Tang Q, Huang Q, Tang N, Gai S. PLAUR facilitates the progression of clear cell renal cell carcinoma by activating the PI3K/AKT/mTOR signaling pathway. PeerJ 2024; 12:e17555. [PMID: 38948215 PMCID: PMC11214736 DOI: 10.7717/peerj.17555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/21/2024] [Indexed: 07/02/2024] Open
Abstract
Background PLAUR has been found upregulated in various tumors and closely correlated with the malignant phenotype of tumor cells. The aim of this study was to investigate the relationship between PLAUR and clear cell renal cell carcinoma (ccRCC) and its potential mechanism of promoting tumor progression. Methods The expression levels and clinical significance of PLAUR, along with the associated signaling pathways, were extensively investigated in ccRCC samples obtained from The Cancer Genome Atlas (TCGA). PLAUR expression in 20 pairs of ccRCC tumor tissues and the adjacent tissues was assessed using qRT-PCR and IHC staining. Additionally, a series of in vitro experiments were conducted to investigate the impact of PLAUR suppression on cellular proliferation, migration, invasion, cell cycle progression, and apoptosis in ccRCC. The Western blot analysis was employed to investigate the expression levels of pivotal genes associated with the PI3K/AKT/mTOR signaling pathway. Results The expression of PLAUR was significantly upregulated in ccRCC compared to normal renal tissues, and higher PLAUR expression in ccRCC was associated with a poorer prognosis than low expression. The in-vitro functional investigations demonstrated that knockdown of PLAUR significantly attenuated the proliferation, migration, and invasion capabilities of ccRCC cells. Concurrently, PLAUR knockdown effectively induced cellular apoptosis, modulated the cell cycle, inhibited the EMT process, and attenuated the activation of the PI3K/AKT/mTOR signaling pathway. PLAUR may represent a key mechanism underlying ccRCC progression. Conclusions The involvement of PLAUR in ccRCC progression may be achieved through the activation of the PI3K/AKT/mTOR signaling pathway, making it a reliable biomarker for the identification and prediction of ccRCC.
Collapse
Affiliation(s)
- Tianzi Qin
- The First Clinical Medical College of Jinan University, Guangzhou, China
- Department of Urology, the Affiliated Hospital of Youjinag Medical University for Nationalities, Baise, China
| | - Minyu Huang
- Department of Urology, the Affiliated Hospital of Youjinag Medical University for Nationalities, Baise, China
| | - Wenjuan Wei
- Department of Ultrasound department, the Affiliated Hospital of Youjinag Medical University for Nationalities, Baise, China
| | - Wei Zhou
- Department of Urology, the Affiliated Hospital of Youjinag Medical University for Nationalities, Baise, China
| | - Qianli Tang
- The First Clinical Medical College of Jinan University, Guangzhou, China
- The Affiliated Hospital of Youjinag Medical University for Nationalities, Baise, China
| | - Qun Huang
- Department of Urology, the Affiliated Hospital of Youjinag Medical University for Nationalities, Baise, China
| | - Ning Tang
- Youjinag Medical University for Nationalities, Baise, China
| | - Shasha Gai
- Youjinag Medical University for Nationalities, Baise, China
| |
Collapse
|
22
|
Xie X, Zhai J, Zhou X, Guo Z, Lo PC, Zhu G, Chan KWY, Yang M. Magnetic Particle Imaging: From Tracer Design to Biomedical Applications in Vasculature Abnormality. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306450. [PMID: 37812831 DOI: 10.1002/adma.202306450] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/14/2023] [Indexed: 10/11/2023]
Abstract
Magnetic particle imaging (MPI) is an emerging non-invasive tomographic technique based on the response of magnetic nanoparticles (MNPs) to oscillating drive fields at the center of a static magnetic gradient. In contrast to magnetic resonance imaging (MRI), which is driven by uniform magnetic fields and projects the anatomic information of the subjects, MPI directly tracks and quantifies MNPs in vivo without background signals. Moreover, it does not require radioactive tracers and has no limitations on imaging depth. This article first introduces the basic principles of MPI and important features of MNPs for imaging sensitivity, spatial resolution, and targeted biodistribution. The latest research aiming to optimize the performance of MPI tracers is reviewed based on their material composition, physical properties, and surface modifications. While the unique advantages of MPI have led to a series of promising biomedical applications, recent development of MPI in investigating vascular abnormalities in cardiovascular and cerebrovascular systems, and cancer are also discussed. Finally, recent progress and challenges in the clinical translation of MPI are discussed to provide possible directions for future research and development.
Collapse
Affiliation(s)
- Xulin Xie
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Jiao Zhai
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Zhengjun Guo
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
- Department of Oncology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Pui-Chi Lo
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Kannie W Y Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| |
Collapse
|
23
|
Situ J, Yang Y, Zhang L, Yan H, Cheng Y. Integration of O 2-economised tumour-targeted photosensitive magnetic nanomaterials in the diagnosis and therapy of gastric cancer. RSC Adv 2024; 14:9920-9932. [PMID: 38528931 PMCID: PMC10961965 DOI: 10.1039/d4ra00497c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/09/2024] [Indexed: 03/27/2024] Open
Abstract
Hypoxia in the tumour microenvironment is a major limiting factor in photodynamic therapy. The present study employed a novel O2-economised photosensitizer, ACSN, to effectively curtail oxygen consumption by impeding the aerobic respiration of tumour cells, thereby increasing the reactive oxygen species (ROS) production in photodynamic therapy. To enhance the efficacy of photodynamic therapy, the active targeting peptide iRGD was employed to facilitate drug accumulation in the tumour tissue. Therefore, we constructed a targeted drug platform, ACSN/Fe3O4@MSNs-iRGD, that integrates diagnosis and treatment. The drug exhibited excellent active targeting ability towards gastric cancer MGC-803 cells and can efficiently penetrate the mitochondria upon cellular internalisation. The photosensitizer ACSN, released from the drug, effectively suppressed mitochondrial aerobic respiration to conserve oxygen and exhibited robust ROS production upon laser excitation. The core-shell structure comprises Fe3O4, which offers excellent T2 dark contrast for real-time tumour monitoring through MRI imaging. By incorporating excellent photodynamic therapy and MRI imaging capabilities, this drug can serve as an effective platform for the integration of tumour diagnosis and treatment, thus addressing the limitations associated with conventional tumour therapies. It is anticipated that this approach will soon be clinically translated.
Collapse
Affiliation(s)
- JinRong Situ
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
| | - Yingying Yang
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
| | - Lingle Zhang
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
| | - Hongzhang Yan
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
| | - Yingsheng Cheng
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
- Tongji Hospital Affiliated to Tongji University Shanghai China
| |
Collapse
|
24
|
Sharma A, Verwilst P, Li M, Ma D, Singh N, Yoo J, Kim Y, Yang Y, Zhu JH, Huang H, Hu XL, He XP, Zeng L, James TD, Peng X, Sessler JL, Kim JS. Theranostic Fluorescent Probes. Chem Rev 2024; 124:2699-2804. [PMID: 38422393 PMCID: PMC11132561 DOI: 10.1021/acs.chemrev.3c00778] [Citation(s) in RCA: 86] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.
Collapse
Affiliation(s)
- Amit Sharma
- Amity
School of Chemical Sciences, Amity University
Punjab, Sector 82A, Mohali 140 306, India
| | - Peter Verwilst
- Rega
Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49, Box 1041, 3000 Leuven, Belgium
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Dandan Ma
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Nem Singh
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Jiyoung Yoo
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Ying Yang
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Jing-Hui Zhu
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Haiqiao Huang
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xi-Le Hu
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiao-Peng He
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- National
Center for Liver Cancer, the International Cooperation Laboratory
on Signal Transduction, Eastern Hepatobiliary
Surgery Hospital, Shanghai 200438, China
| | - Lintao Zeng
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- School
of Chemistry and Chemical Engineering, Henan
Normal University, Xinxiang 453007, China
| | - Xiaojun Peng
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- State
Key Laboratory of Fine Chemicals, Dalian
University of Technology, Dalian 116024, China
| | - Jonathan L. Sessler
- Department
of Chemistry, The University of Texas at
Austin, Texas 78712-1224, United
States
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- TheranoChem Incorporation, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
25
|
Fu Q, Yang X, Wang M, Zhu K, Wang Y, Song J. Activatable Probes for Ratiometric Imaging of Endogenous Biomarkers In Vivo. ACS NANO 2024; 18:3916-3968. [PMID: 38258800 DOI: 10.1021/acsnano.3c10659] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Dynamic variations in the concentration and abnormal distribution of endogenous biomarkers are strongly associated with multiple physiological and pathological states. Therefore, it is crucial to design imaging systems capable of real-time detection of dynamic changes in biomarkers for the accurate diagnosis and effective treatment of diseases. Recently, ratiometric imaging has emerged as a widely used technique for sensing and imaging of biomarkers due to its advantage of circumventing the limitations inherent to conventional intensity-dependent signal readout methods while also providing built-in self-calibration for signal correction. Here, the recent progress of ratiometric probes and their applications in sensing and imaging of biomarkers are outlined. Ratiometric probes are classified according to their imaging mechanisms, and ratiometric photoacoustic imaging, ratiometric optical imaging including photoluminescence imaging and self-luminescence imaging, ratiometric magnetic resonance imaging, and dual-modal ratiometric imaging are discussed. The applications of ratiometric probes in the sensing and imaging of biomarkers such as pH, reactive oxygen species (ROS), reactive nitrogen species (RNS), glutathione (GSH), gas molecules, enzymes, metal ions, and hypoxia are discussed in detail. Additionally, this Review presents an overview of challenges faced in this field along with future research directions.
Collapse
Affiliation(s)
- Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Mengzhen Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Kang Zhu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
26
|
Zhang DG, Pan YJ, Chen BQ, Lu XC, Xu QX, Wang P, Kankala RK, Jiang NN, Wang SB, Chen AZ. Protein-guided biomimetic nanomaterials: a versatile theranostic nanoplatform for biomedical applications. NANOSCALE 2024; 16:1633-1649. [PMID: 38168813 DOI: 10.1039/d3nr05495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Over the years, bioinspired mineralization-based approaches have been applied to synthesize multifunctional organic-inorganic nanocomposites. These nanocomposites can address the growing demands of modern biomedical applications. Proteins, serving as vital biological templates, play a pivotal role in the nucleation and growth processes of various organic-inorganic nanocomposites. Protein-mineralized nanomaterials (PMNMs) have attracted significant interest from researchers due to their facile and convenient preparation, strong physiological activity, stability, impressive biocompatibility, and biodegradability. Nevertheless, few comprehensive reviews have expounded on the progress of these nanomaterials in biomedicine. This article systematically reviews the principles and strategies for constructing nanomaterials using protein-directed biomineralization and biomimetic mineralization techniques. Subsequently, we focus on their recent applications in the biomedical field, encompassing areas such as bioimaging, as well as anti-tumor, anti-bacterial, and anti-inflammatory therapies. Furthermore, we discuss the challenges encountered in practical applications of these materials and explore their potential in future applications. This review aspired to catalyze the continued development of these bioinspired nanomaterials in drug development and clinical diagnosis, ultimately contributing to the fields of precision medicine and translational medicine.
Collapse
Affiliation(s)
- Da-Gui Zhang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Yu-Jing Pan
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Biao-Qi Chen
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Xiao-Chang Lu
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Qin-Xi Xu
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Pei Wang
- Jiangxi Provincial Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Ranjith Kumar Kankala
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Ni-Na Jiang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Shi-Bin Wang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Ai-Zheng Chen
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| |
Collapse
|
27
|
Fan Y, Liu S, Gao E, Guo R, Dong G, Li Y, Gao T, Tang X, Liao H. The LMIT: Light-mediated minimally-invasive theranostics in oncology. Theranostics 2024; 14:341-362. [PMID: 38164160 PMCID: PMC10750201 DOI: 10.7150/thno.87783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/18/2023] [Indexed: 01/03/2024] Open
Abstract
Minimally-invasive diagnosis and therapy have gradually become the trend and research hotspot of current medical applications. The integration of intraoperative diagnosis and treatment is a development important direction for real-time detection, minimally-invasive diagnosis and therapy to reduce mortality and improve the quality of life of patients, so called minimally-invasive theranostics (MIT). Light is an important theranostic tool for the treatment of cancerous tissues. Light-mediated minimally-invasive theranostics (LMIT) is a novel evolutionary technology that integrates diagnosis and therapeutics for the less invasive treatment of diseased tissues. Intelligent theranostics would promote precision surgery based on the optical characterization of cancerous tissues. Furthermore, MIT also requires the assistance of smart medical devices or robots. And, optical multimodality lay a solid foundation for intelligent MIT. In this review, we summarize the important state-of-the-arts of optical MIT or LMIT in oncology. Multimodal optical image-guided intelligent treatment is another focus. Intraoperative imaging and real-time analysis-guided optical treatment are also systemically discussed. Finally, the potential challenges and future perspectives of intelligent optical MIT are discussed.
Collapse
Affiliation(s)
- Yingwei Fan
- School of Medical Technology, Beijing Institute of Technology, Beijing, China, 100081
| | - Shuai Liu
- School of Medical Technology, Beijing Institute of Technology, Beijing, China, 100081
| | - Enze Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing, China, 100081
| | - Rui Guo
- School of Medical Technology, Beijing Institute of Technology, Beijing, China, 100081
| | - Guozhao Dong
- School of Medical Technology, Beijing Institute of Technology, Beijing, China, 100081
| | - Yangxi Li
- Dept. of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China, 100084
| | - Tianxin Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing, China, 100081
| | - Xiaoying Tang
- School of Medical Technology, Beijing Institute of Technology, Beijing, China, 100081
| | - Hongen Liao
- Dept. of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China, 100084
| |
Collapse
|
28
|
Zhang P, Li W, Liu C, Qin F, Lu Y, Qin M, Hou Y. Molecular imaging of tumour-associated pathological biomarkers with smart nanoprobe: From "Seeing" to "Measuring". EXPLORATION (BEIJING, CHINA) 2023; 3:20230070. [PMID: 38264683 PMCID: PMC10742208 DOI: 10.1002/exp.20230070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/18/2023] [Indexed: 01/25/2024]
Abstract
Although the extraordinary progress has been made in molecular biology, the prevention of cancer remains arduous. Most solid tumours exhibit both spatial and temporal heterogeneity, which is difficult to be mimicked in vitro. Additionally, the complex biochemical and immune features of tumour microenvironment significantly affect the tumour development. Molecular imaging aims at the exploitation of tumour-associated molecules as specific targets of customized molecular probe, thereby generating image contrast of tumour markers, and offering opportunities to non-invasively evaluate the pathological characteristics of tumours in vivo. Particularly, there are no "standard markers" as control in clinical imaging diagnosis of individuals, so the tumour pathological characteristics-responsive nanoprobe-based quantitative molecular imaging, which is able to visualize and determine the accurate content values of heterogeneous distribution of pathological molecules in solid tumours, can provide criteria for cancer diagnosis. In this context, a variety of "smart" quantitative molecular imaging nanoprobes have been designed, in order to provide feasible approaches to quantitatively visualize the tumour-associated pathological molecules in vivo. This review summarizes the recent achievements in the designs of these nanoprobes, and highlights the state-of-the-art technologies in quantitative imaging of tumour-associated pathological molecules.
Collapse
Affiliation(s)
- Peisen Zhang
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
- Department of ChemistryUniversity of TorontoTorontoOntarioCanada
| | - Wenyue Li
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Chuang Liu
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Feng Qin
- Department of Neurosurgery and National Chengdu Center for Safety Evaluation of DrugsState Key Laboratory of Biotherapy/Collaborative Innovation Center for BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Yijie Lu
- Department of ChemistryUniversity of TorontoTorontoOntarioCanada
| | - Meng Qin
- Department of Neurosurgery and National Chengdu Center for Safety Evaluation of DrugsState Key Laboratory of Biotherapy/Collaborative Innovation Center for BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Yi Hou
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| |
Collapse
|
29
|
Wu M, Gong D, Zhou Y, Zha Z, Xia X. Activatable probes with potential for intraoperative tumor-specific fluorescence-imaging guided surgery. J Mater Chem B 2023; 11:9777-9797. [PMID: 37749982 DOI: 10.1039/d3tb01590d] [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: 09/27/2023]
Abstract
Owing to societal development and aging population, the impact of cancer on human health and quality of life has increased. Early detection and surgical treatment are the most effective approaches for most cancer patients. As the scope of conventional tumor resection is determined by auxiliary examination and surgeon experience, there is often insufficient recognition of tiny tumors. The ability to detect such tumors can be improved by using fluorescent tumor-specific probes for surgical navigation. This review mainly describes the design principles and mechanisms of activatable probes for the fluorescence imaging of tumors. This type of probe is nonfluorescent in normal tissue but exhibits obvious fluorescence emission upon encountering tumor-specific substrates, such as enzymes or bioactive molecules, or changes in the microenvironment, such as a low pH. In some cases, a single-factor response does not guarantee the effective fluorescence labeling of tumors. Therefore, two-factor-activatable fluorescence imaging probes that react with two specific factors in tumor cells have also been developed. Compared with single biomarker testing, the simultaneous monitoring of multiple biomarkers may provide additional insight into the role of these substances in cancer development and aid in improving the accuracy of early cancer diagnosis. Research and progress in this field can provide new methods for precision medicine and targeted therapy. The development of new approaches for early diagnosis and treatment can effectively improve the prognosis of cancer patients and help enhance their quality of life.
Collapse
Affiliation(s)
- Mingzhu Wu
- Department of Obstetrics and Gynecology, Anhui Provincial Children's Hospital, Children's Hospital of Fudan University Anhui Hospital, Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P. R. China.
| | - Deyan Gong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China.
| | - Yuanyuan Zhou
- Department of Obstetrics and Gynecology, Anhui Provincial Children's Hospital, Children's Hospital of Fudan University Anhui Hospital, Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P. R. China.
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China.
| | - Xiaoping Xia
- Department of Obstetrics and Gynecology, Anhui Provincial Children's Hospital, Children's Hospital of Fudan University Anhui Hospital, Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P. R. China.
| |
Collapse
|
30
|
Rainu SK, Ramachandran RG, Parameswaran S, Krishnakumar S, Singh N. Advancements in Intraoperative Near-Infrared Fluorescence Imaging for Accurate Tumor Resection: A Promising Technique for Improved Surgical Outcomes and Patient Survival. ACS Biomater Sci Eng 2023; 9:5504-5526. [PMID: 37661342 DOI: 10.1021/acsbiomaterials.3c00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Clear surgical margins for solid tumor resection are essential for preventing cancer recurrence and improving overall patient survival. Complete resection of tumors is often limited by a surgeon's ability to accurately locate malignant tissues and differentiate them from healthy tissue. Therefore, techniques or imaging modalities are required that would ease the identification and resection of tumors by real-time intraoperative visualization of tumors. Although conventional imaging techniques such as positron emission tomography (PET), computed tomography (CT), magnetic resonance imaging (MRI), or radiography play an essential role in preoperative diagnostics, these cannot be utilized in intraoperative tumor detection due to their large size, high cost, long imaging time, and lack of cancer specificity. The inception of several imaging techniques has paved the way to intraoperative tumor margin detection with a high degree of sensitivity and specificity. Particularly, molecular imaging using near-infrared fluorescence (NIRF) based nanoprobes provides superior imaging quality due to high signal-to-noise ratio, deep penetration to tissues, and low autofluorescence, enabling accurate tumor resection and improved survival rates. In this review, we discuss the recent developments in imaging technologies, specifically focusing on NIRF nanoprobes that aid in highly specific intraoperative surgeries with real-time recognition of tumor margins.
Collapse
Affiliation(s)
- Simran Kaur Rainu
- Center for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Remya Girija Ramachandran
- L&T Ocular Pathology Department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai 600006, India
| | - Sowmya Parameswaran
- L&T Ocular Pathology Department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai 600006, India
| | - Subramanian Krishnakumar
- L&T Ocular Pathology Department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai 600006, India
| | - Neetu Singh
- Center for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
31
|
Cao C, Yin H, Yang B, Yue Q, Wu G, Gu M, Zhang Y, Fan Y, Dong X, Wang T, Wang C, Zhu X, Mao Y, Zhang X, Lei Z, Li C. Intra-Operative Definition of Glioma Infiltrative Margins by Visualizing Immunosuppressive Tumor-Associated Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304020. [PMID: 37544917 PMCID: PMC10558635 DOI: 10.1002/advs.202304020] [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: 06/19/2023] [Indexed: 08/08/2023]
Abstract
Accurate delineation of glioma infiltrative margins remains a challenge due to the low density of cancer cells in these regions. Here, a hierarchical imaging strategy to define glioma margins by locating the immunosuppressive tumor-associated macrophages (TAMs) is proposed. A pH ratiometric fluorescent probe CP2-M that targets immunosuppressive TAMs by binding to mannose receptor (CD206) is developed, and it subsequently senses the acidic phagosomal lumen, resulting in a remarkable fluorescence enhancement. With assistance of CP2-M, glioma xenografts in mouse models with a tumor-to-background ratio exceeding 3.0 for up to 6 h are successfully visualized. Furthermore, by intra-operatively mapping the pH distribution of exposed tissue after craniotomy, the glioma allograft in rat models is precisely excised. The overall survival of rat models significantly surpasses that achieved using clinically employed fluorescent probes. This work presents a novel strategy for locating glioma margins, thereby improving surgical outcomes for tumors with infiltrative characteristics.
Collapse
Affiliation(s)
- Chong Cao
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Hang Yin
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Biao Yang
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Qi Yue
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Guoqing Wu
- School of Information Science and TechnologyFudan UniversityShanghai200438China
| | - Meng Gu
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain‐Inspired IntelligenceMOE Key Laboratory of Computational Neuroscience and Brain‐Inspired IntelligenceMOE Frontiers Center for Brain ScienceFudan University220 Handan RoadShanghai200433China
| | - Yang Fan
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Xiaoyan Dong
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Ting Wang
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Cong Wang
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Xiao Zhu
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Ying Mao
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Xiao‐Yong Zhang
- Institute of Science and Technology for Brain‐Inspired IntelligenceMOE Key Laboratory of Computational Neuroscience and Brain‐Inspired IntelligenceMOE Frontiers Center for Brain ScienceFudan University220 Handan RoadShanghai200433China
| | - Zuhai Lei
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery Ministry of EducationInnovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of EducationSchool of PharmacyDepartment of Neurosurgery, Huashan HospitalFudan UniversityShanghai201203China
- State Key Laboratory of Medical NeurobiologyZhongshan HospitalFudan UniversityShanghai200032China
| |
Collapse
|
32
|
Murphy PS, Galette P, van der Aart J, Janiczek RL, Patel N, Brown AP. The role of clinical imaging in oncology drug development: progress and new challenges. Br J Radiol 2023; 96:20211126. [PMID: 37393537 PMCID: PMC10546429 DOI: 10.1259/bjr.20211126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/14/2023] [Accepted: 06/06/2023] [Indexed: 07/03/2023] Open
Abstract
In 2008, the role of clinical imaging in oncology drug development was reviewed. The review outlined where imaging was being applied and considered the diverse demands across the phases of drug development. A limited set of imaging techniques was being used, largely based on structural measures of disease evaluated using established response criteria such as response evaluation criteria in solid tumours. Beyond structure, functional tissue imaging such as dynamic contrast-enhanced MRI and metabolic measures using [18F]flourodeoxyglucose positron emission tomography were being increasingly incorporated. Specific challenges related to the implementation of imaging were outlined including standardisation of scanning across study centres and consistency of analysis and reporting. More than a decade on the needs of modern drug development are reviewed, how imaging has evolved to support new drug development demands, the potential to translate state-of-the-art methods into routine tools and what is needed to enable the effective use of this broadening clinical trial toolset. In this review, we challenge the clinical and scientific imaging community to help refine existing clinical trial methods and innovate to deliver the next generation of techniques. Strong industry-academic partnerships and pre-competitive opportunities to co-ordinate efforts will ensure imaging technologies maintain a crucial role delivering innovative medicines to treat cancer.
Collapse
Affiliation(s)
| | - Paul Galette
- Telix Pharmaceuticals (US) Inc, Fishers, United States
| | | | | | | | - Andrew P. Brown
- Vale Imaging Consultancy Solutions, Harston, Cambridge, United Kingdom
| |
Collapse
|
33
|
Yan J, Liu H, Wu Y, Niu B, Deng X, Zhang L, Dang Q, Wang Y, Lu X, Zhang B, Sun W. Recent progress of self-immobilizing and self-precipitating molecular fluorescent probes for higher-spatial-resolution imaging. Biomaterials 2023; 301:122281. [PMID: 37643487 DOI: 10.1016/j.biomaterials.2023.122281] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Flourished in the past two decades, fluorescent probe technology provides researchers with accurate and efficient tools for in situ imaging of biomarkers in living cells and tissues and may play a significant role in clinical diagnosis and treatment such as biomarker detection, fluorescence imaging-guided surgery, and photothermal/photodynamic therapy. In situ imaging of biomarkers depends on the spatial resolution of molecular probes. Nevertheless, the majority of currently available molecular fluorescent probes suffer from the drawback of diffusing from the target region. This leads to a rapid attenuation of the fluorescent signal over time and a reduction in spatial resolution. Consequently, the diffused fluorescent signal cannot accurately reflect the in situ information of the target. Self-immobilizing and self-precipitating molecular fluorescent probes can be used to overcome this problem. These probes ensure that the fluorescent signal remains at the location where the signal is generated for a long time. In this review, we introduce the development history of the two types of probes and classify them in detail according to different design strategies. In addition, we compare their advantages and disadvantages, summarize some representative studies conducted in recent years, and propose prospects for this field.
Collapse
Affiliation(s)
- Jiawei Yan
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Huanying Liu
- School of Mechanical and Power Engineering, Dalian Ocean University, Dalian, 116023, China
| | - Yingxu Wu
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Ben Niu
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Xiaojing Deng
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Linhao Zhang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Qi Dang
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Yubo Wang
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Xiao Lu
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Boyu Zhang
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China.
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China.
| |
Collapse
|
34
|
Zhou S, Jiang L, Li C, Mao H, Jiang C, Wang Z, Zheng X, Jiang X. Acid and Hypoxia Tandem-Activatable Deep Near-Infrared Nanoprobe for Two-Step Signal Amplification and Early Detection of Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2212231. [PMID: 37339461 DOI: 10.1002/adma.202212231] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 06/15/2023] [Indexed: 06/22/2023]
Abstract
The early detection of cancers can significantly change outcomes even with existing treatments. However, ~50% of cancers still cannot be detected until they reach an advanced stage, highlighting the great challenges in the early detection. Here, an ultrasensitive deep near-infrared (dNIR) nanoprobe that is successively responsive to tumor acidity and hypoxia is reported. It is demonstrated that the new nanoprobe specifically detects tumor hypoxia microenvironment based on deep NIR imaging in ten different types of tumor models using cancer cell lines and patient-tissue derived xenograft tumors. By combining the acidity and hypoxia specific two-step signal amplification with a deep NIR detection, the reported nanoprobe enables the ultrasensitive visualization of hundreds of tumor cells or small tumors with a size of 260 µm in whole-body imaging or 115 µm metastatic lesions in lung imaging. As a result, it reveals that tumor hypoxia can occur as early as the lesions contain only several hundred cancer cells.
Collapse
Affiliation(s)
- Sensen Zhou
- College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Lei Jiang
- College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Cheng Li
- College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Chunping Jiang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Zhongxia Wang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Xianchuang Zheng
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Xiqun Jiang
- College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
35
|
Zhou Y, Wang C, Li W, Lu W, Liu X, Xi L, Li P, Lu J, Wei J. Fluorescence colposcope with TMTP1-PEG4-ICG is comparable to the conventional colposcope in identifying cervical precancerous lesions: A randomized controlled trial. Int J Gynaecol Obstet 2023; 162:969-976. [PMID: 36939553 DOI: 10.1002/ijgo.14752] [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: 11/09/2022] [Revised: 02/22/2023] [Accepted: 03/03/2023] [Indexed: 03/21/2023]
Abstract
OBJECTIVE To compare the diagnostic efficiency of a fluorescence colposcope with TMTP1-PEG4-ICG dye versus a conventional colposcope with acetic acid and Lugol's iodine in identifying cervical precancerous lesions. METHODS In all, 218 women with abnormal cervical cancer screening results including cytology and/or human papillomavirus (HPV) test were involved in the randomized controlled trial. Patients in the fluorescence colposcope group had TMTP1-PEG4-ICG dye applied to the cervix uteri before colposcopy. Patients in the conventional colposcope group were routinely administered acetic acid and Lugol's iodine to stain the cervix uteri. Two to four cervical sites per patient were taken out for biopsy. The diagnostic efficiency of fluorescence colposcopy and conventional colposcopy was calculated on a per-patient and per-site basis. χ2 test or Fisher exact test was used. RESULTS A total of 194 patients and the corresponding 662 cervical sites were included in the final analysis. There was no statistically significant difference in the diagnostic efficiency between the two groups both on a per-patient and a per-site basis, including accuracy, sensitivity, specificity, positive predictive value, and negative predictive value. CONCLUSIONS The fluorescence colposcope with TMTP1-PEG4-ICG dye was comparable to the conventional colposcope in identifying cervical precancerous lesions.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chen Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wanrong Lu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaohu Liu
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Xi
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pengcheng Li
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, 2019RU002, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Jinling Lu
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| | - Juncheng Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
36
|
Wei J, Liu C, Liang W, Yang X, Han S. Advances in optical molecular imaging for neural visualization. Front Bioeng Biotechnol 2023; 11:1250594. [PMID: 37671191 PMCID: PMC10475611 DOI: 10.3389/fbioe.2023.1250594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023] Open
Abstract
Iatrogenic nerve injury is a significant complication in surgery, which can negatively impact patients' quality of life. Currently, the main clinical neuroimaging methods, such as computed tomography, magnetic resonance imaging, and high-resolution ultrasonography, do not offer precise real-time positioning images for doctors during surgery. The clinical application of optical molecular imaging technology has led to the emergence of new concepts such as optical molecular imaging surgery, targeted surgery, and molecular-guided surgery. These advancements have made it possible to directly visualize surgical target areas, thereby providing a novel method for real-time identification of nerves during surgery planning. Unlike traditional white light imaging, optical molecular imaging technology enables precise positioning and identifies the cation of intraoperative nerves through the presentation of color images. Although a large number of experiments and data support its development, there are few reports on its actual clinical application. This paper summarizes the research results of optical molecular imaging technology and its ability to realize neural visualization. Additionally, it discusses the challenges neural visualization recognition faces and future development opportunities.
Collapse
Affiliation(s)
- Jinzheng Wei
- Department of Orthopaedics, First Hospital of Shanxi Medical University, Taiyuan, China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Chao Liu
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenkai Liang
- Department of Orthopaedics, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaofeng Yang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shufeng Han
- Department of Orthopaedics, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
37
|
Luo X, Ren Q, Zhang H, Chen C, Yang T, He X, Zhao W. Efficient FMT reconstruction based on L 1-αL 2 regularization via half-quadratic splitting and a two-probe separation light source strategy. JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2023; 40:1128-1141. [PMID: 37706766 DOI: 10.1364/josaa.481330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/20/2023] [Indexed: 09/15/2023]
Abstract
Fluorescence molecular tomography (FMT) can achieve noninvasive, high-contrast, high-sensitivity three-dimensional imaging in vivo by relying on a variety of fluorescent molecular probes, and has excellent clinical transformation prospects in the detection of tumors in vivo. However, the limited surface fluorescence makes the FMT reconstruction have some ill-posedness, and it is difficult to obtain the ideal reconstruction effect. In this paper, two different emission fluorescent probes and L 1-L 2 regularization are combined to improve the temporal and spatial resolution of FMT visual reconstruction by introducing the weighting factor α and a half-quadratic splitting alternating optimization (HQSAO) iterative algorithm. By introducing an auxiliary variable, the HQSAO method breaks the sparse FMT reconstruction task into two subproblems that can be solved in turn: simple reconstruction and image denoising. The weight factor α (α>1) can increase the weight of nonconvex terms to further promote the sparsity of the algorithm. Importantly, this paper combines two different dominant fluorescent probes to achieve high-quality reconstruction of dual light sources. The performance of the proposed reconstruction strategy was evaluated by digital mouse and nude mouse single/dual light source models. The simulation results show that the HQSAO iterative algorithm can achieve more excellent positioning accuracy and morphology distribution in a shorter time. In vivo experiments also further prove that the HQSAO algorithm has advantages in light source information preservation and artifact suppression. In particular, the introduction of two main emission fluorescent probes makes it easy to separate and reconstruct the dual light sources. When it comes to localization and three-dimensional morphology, the results of the reconstruction are much better than those using a fluorescent probe, which further facilitates the clinical transformation of FMT.
Collapse
|
38
|
Song B, Yan H, Jiang J, Yu J, Huang S, Yuan J. An activatable nanoprobe based on nanocomposites of visible-light-excitable europium(III) complex-anchored MnO 2 nanosheets for bimodal time-gated luminescence and magnetic resonance imaging of tumor cells. Analyst 2023; 148:2493-2500. [PMID: 37183980 DOI: 10.1039/d3an00405h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Bimodal imaging probes that combine magnetic resonance imaging (MRI) and photoluminescence imaging are quite appealing since they can supply both anatomical and molecular information to effectively ameliorate the accuracy of detection. In this study, an activatable nanoprobe, [Eu(BTD)3(DPBT)]@MnO2, for bimodal time-gated luminescence imaging (TGLI) and MRI has been constructed by anchoring visible-light-excitable Eu3+ complexes on lamellar MnO2 nanosheets. Due to the luminescence quenching effect and non-magnetic resonance (MR) activity of MnO2 nanosheets, the developed nanoprobe presents quite weak TGL and MR signals. After exposure to H2O2 or GSH, accompanied by the transformation from MnO2 to Mn2+, the nanoprobe exhibits rapid, sensitive, and selective "turn-on" responses towards GSH and H2O2 in TGL and MR detection modes. Furthermore, the nanoprobe displays high stability, low cytotoxicity, good biocompatibility and water dispersion. Given the high contents of GSH and H2O2 in cancer cells, the nanoprobe was used for the identification of cancer cells by TGLI of intracellular GSH and H2O2, as well as for the tracing of tumor cells in tumor-bearing mice by tumor-targeting in vivo MRI and TGLI of tumor tissues. The research outcomes proved the potential of [Eu(BTD)3(DPBT)]@MnO2 as a useful nanoprobe for the tracing and accurate detection of cancer cells in vitro and in vivo via bimodal TGLI and MRI.
Collapse
Affiliation(s)
- Bo Song
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China.
| | - Huinan Yan
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China.
| | - Jiao Jiang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China.
| | - Jin Yu
- Second Affiliated Hospital, Dalian Medical University, Dalian 116027, China
| | - Shengjun Huang
- Division of Fossil Energy Conversion, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Jingli Yuan
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China.
| |
Collapse
|
39
|
Huang M, Qi M, Yang H, Peng Z, Chen S, Liang M, Hu Y, Deng L, Hu M. Noninvasive Strategies for the Treatment of Tiny Liver Cancer: Integrating Photothermal Therapy and Multimodality Imaging EpCAM-Guided Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2023; 15:21843-21853. [PMID: 37102323 DOI: 10.1021/acsami.3c00211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Surgical resection and ablation therapy have been shown to achieve the purpose of a radical cure for liver cancer with a size of less than 3 cm; however, tiny liver cancer lesions of diameters smaller than 2 cm remain challenging to diagnose and cure due to the failure of the generation of new blood vessels within tumors. Emerging evidence has revealed that optical molecular imaging combined with nanoprobes can detect tiny cancer from the perspective of molecular and cellular levels and kill cancer cells by the photothermal effect of nanoparticles in real time, thereby achieving radical goals. In the present study, we designed and synthesized multicomponent and multifunctional ICG-CuS-Gd@BSA-EpCAM nanoparticles (NPs) with a potent antineoplastic effect on tiny liver cancer. Using subcutaneous and orthotopic liver cancer xenograft mouse models, we found that the components of the NPs, including ICG and CuS-Gd@BSA, showed synergistic photothermal effects on the eradication of tiny liver cancer. We also found that the ICG-CuS-Gd@BSA-EpCAM NPs exhibited triple-modal functions of fluorescence imaging, magnetic resonance imaging, and photoacoustic imaging, with targeted detection and photothermal treatment of tiny liver cancer under near-infrared light irradiation. Together, our study demonstrates that the ICG-CuS-Gd@BSA-EpCAM NPs in combination with optical imaging technique might be a potential approach for detecting and noninvasively and radically curing tiny liver cancer by the photothermal effect.
Collapse
Affiliation(s)
- Maohua Huang
- Department of Hepatobiliary Surgery, Jinan University First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Ming Qi
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Hongyan Yang
- Department of Hepatobiliary Surgery, Jinan University First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Zhi Peng
- Department of Hepatobiliary Surgery, Jinan University First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Shouguo Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Mingchao Liang
- Department of Hepatobiliary Surgery, Jinan University Affiliated Shunde Hospital, Jinan University, Foshan 528305, China
| | - Youzhu Hu
- Department of Hepatobiliary Surgery, Jinan University First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Department of Hepatobiliary Surgery, Jinan University Affiliated Shunde Hospital, Jinan University, Foshan 528305, China
| | - Lijuan Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Min Hu
- Department of Hepatobiliary Surgery, Jinan University First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| |
Collapse
|
40
|
Han S, Wang JTW, Yavuz E, Zam A, Rouatbi N, Utami RN, Liam-Or R, Griffiths A, Dickson W, Sosabowski J, Al-Jamal KT. Spatiotemporal tracking of gold nanorods after intranasal administration for brain targeting. J Control Release 2023; 357:606-619. [PMID: 37061195 PMCID: PMC10390340 DOI: 10.1016/j.jconrel.2023.04.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
Intranasal administration is becoming increasingly more attractive as a fast delivery route to the brain for therapeutics circumventing the blood-brain barrier (BBB). Gold nanorods (AuNRs) demonstrate unique optical and biological properties compared to other gold nanostructures due to their high aspect ratio. In this study, we investigated for the first time the brain region-specific distribution of AuNRs and their potential as a drug delivery platform for central nervous system (CNS) therapy following intranasal administration to mice using a battery of analytical and imaging techniques. AuNRs were functionalized with a fluorescent dye (Cyanine5, Cy5) or a metal chelator (diethylenetriaminepentaacetic dianhydride, DTPA anhydride) to complex with Indium-111 via a PEG spacer for optical and nuclear imaging, respectively. Direct quantification of gold was achieved by inductively coupled plasma mass spectrometry. Rapid AuNRs uptake in mice brains was observed within 10 min following intranasal administration which gradually reduced over time. This was confirmed by the 3 imaging/analytical techniques. Autoradiography of sagittal brain sections suggested entry to the brain via the olfactory bulb followed by diffusion to other brain regions within 1 h of administration. The presence of AuNR in glioblastoma (GBM) tumors following intranasal administration was also proven which opens doors for AuNRs applications, as nose-to-brain drug delivery carriers, for treatment of a range of CNS diseases.
Collapse
Affiliation(s)
- Shunping Han
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom; London Centre for Nanotechnology, King's College London, Strand, London WC2R 2LS, United Kingdom
| | - Julie Tzu-Wen Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Emine Yavuz
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom; Advanced Technology Research and Application Center, Selcuk University, Aleaddin Keykubat Yerleskesi, Akademi Mah. Yeni Istanbul Cad. No: 355/C, Selcuklu, Konya, Turkey
| | - Alaa Zam
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Nadia Rouatbi
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Rifka Nurul Utami
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Revadee Liam-Or
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Alexander Griffiths
- London Metallomics Facility, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Wayne Dickson
- Department of Physics, King's College London, Strand, London WC2R 2LS, United Kingdom; London Centre for Nanotechnology, King's College London, Strand, London WC2R 2LS, United Kingdom
| | - Jane Sosabowski
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom; London Centre for Nanotechnology, King's College London, Strand, London WC2R 2LS, United Kingdom.
| |
Collapse
|
41
|
Lu WL, Kuang H, Gu J, Hu X, Chen B, Fan Y. GAP-43 targeted indocyanine green-loaded near-infrared fluorescent probe for real-time mapping of perineural invasion lesions in pancreatic cancer in vivo. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102671. [PMID: 37054805 DOI: 10.1016/j.nano.2023.102671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/22/2023] [Accepted: 03/26/2023] [Indexed: 04/15/2023]
Abstract
OBJECTIVE Perineural invasion (PNI) is associated with local recurrence, distant metastasis, and a poor prognosis in pancreatic cancer. However, rare attempt was made to identified the PNI intraoperative. To facilitate precise R0 excision of the tumor, we planned to develop a fluorescent probe for intraoperative imaging of the PNI using GAP-43 as the target and indocyanine green (ICG) as the carrier. METHODS The probe was created by binding peptide antibody and ICG. Its targeting was tested in vitro and in vivo using a co-culture model of PC12 and tumor cells to create an in vitro neural invasion model and a mouse sciatic nerve invasion model. The small animal imaging system and surgical navigation system confirmed the probe's potential clinical applicability. The sciatic nerve damage model was created to confirm the probe's targeting. RESULTS We used the pancreatic cancer samples and the public database to confirm that GAP-43 was preferentially overexpressed in pancreatic cancer, particularly in PNI. PC12 cells showed high GAP-43RA-PEG-ICG probe-specific absorption after being co-cultured with tumor cells in vitro. In the sciatic nerve invasion experiment, animals in probe group displayed a significantly stronger fluorescence signal at the PNI compared to ICG-NP and the contralateral normal nerves groups. Although only 60 % of mice appeared to have R0 resections by the naked eye, small animal imaging systems and surgical fluorescence navigation systems could remove the tumor with R0 precision. The injury model used in the probe imaging experimental trials demonstrated that the probe was specifically targeted to the injured nerve, regardless of whether the injury was infiltrated by a tumor or physical. CONCLUSION We developed the GAP-43Ra-ICG-PEG, an active-targeting near-infrared fluorescent (NIF) probe, that specifically binds to GAP-43-positive neural cells in an in vitro model of PNI. The probe efficiently visualized PNI lesions in pancreatic cancer in preclinical models, opening up new possibilities for NIRF-guided pancreatic surgery, particularly for PNI patients.
Collapse
Affiliation(s)
- Wen Liang Lu
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Thyroid and breast surgery, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China
| | - Houfang Kuang
- Department of General Surgery, Wuhan Children(,) hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, China
| | - Jianyou Gu
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaojun Hu
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Hepatobiliary Surgery, The Fifth Affifiliated Hospital of Southern Medical University, Guangzhou 510920, China
| | - Bo Chen
- Department of Thyroid and breast surgery, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China
| | - Yingfang Fan
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
42
|
Teranishi K. In vivo near-infrared fluorescence imaging of gastric cancer in an MKN-45 gastric cancer xenograft mouse model using intraoperative ureteral identification agent ASP5354. Photochem Photobiol Sci 2023:10.1007/s43630-023-00410-8. [PMID: 37010695 DOI: 10.1007/s43630-023-00410-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/18/2023] [Indexed: 04/04/2023]
Abstract
Accurate intraoperative identification of gastric cancer lesions and determination of the extent of resection are important for curability and function preservation. This study aimed to investigate the potential of the near-infrared fluorescence (NIRF) imaging agent ASP5354 for in vivo fluorescence imaging of gastric cancer. The capability of ASP5354 was evaluated using an MKN-45 human gastric cancer xenograft mouse model. A single dose of ASP5354 was intravenously administered to the mice at a concentration of 120 nmol (0.37 mg)/kg body weight. In vivo NIRF images of the mouse backs were obtained using an NIRF camera system. Moreover, the cancer tissues were dissected, and the NIRF intensity in the tissue sections was measured using the NIRF camera system. ASP5354 uptake in MKN-45 cells was assessed in vitro using the NIRF microscope. The NIRF signal of ASP5354 was selectively detected in gastric cancer tissues immediately after the intravenous administration of ASP5354. The cancer tissues emitted stronger NIRF signals than adjacent normal tissues. The difference in the NIRF intensity between the normal and cancer tissues was clearly observed at the boundary between them in the macrolevel NIRF images. Cancer tissues can be distinguished from normal tissues based on the measurement of the NIRF of ASP5354, using an NIRF camera system. ASP5354 is a promising agent for NIRF imaging of gastric cancer tissues.
Collapse
Affiliation(s)
- Katsunori Teranishi
- Graduate School of Bioresources, Mie University, 1577 Kurimamachiya, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
43
|
Sang M, Huang Y, Wang L, Chen L, Nawsherwan, Li G, Wang Y, Yu X, Dai C, Zheng J. An "AND" Molecular Logic Gate as a Super-Enhancers for De Novo Designing Activatable Probe and Its Application in Atherosclerosis Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207066. [PMID: 36808894 PMCID: PMC10131802 DOI: 10.1002/advs.202207066] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/31/2023] [Indexed: 09/30/2023]
Abstract
Developing activatable fluorescent probes with superlative fluorescence enhancement factor (F/F0 ) to improve the signal-to-noise (S/N) ratio is still an urgent issue. "AND" molecular logic gates are emerging as a useful tool for enhanced probes selectivity and accuracy. Here, an "AND" logic gate is developed as super-enhancers for designing activatable probes with huge F/F0 and S/N ratio. It utilizes lipid-droplets (LDs) as controllable background input and sets the target analyte as variable input. The fluorescence is tremendously quenching due to double locking, thus an extreme F/F0 ratio of target analyte is obtained. Importantly, this probe can transfer to LDs after a response occurs. The target analyte can be directly visualized through the spatial location without a control group. Accordingly, a peroxynitrite (ONOO- ) activatable probe (CNP2-B) is de novo designed. The F/F0 of CNP2-B achieves 2600 after reacting with ONOO- . Furthermore, CNP2-B can transfer from mitochondria to lipid droplets after being activated. The higher selectivity and S/N ratio of CNP2-B are obtained than commercial probe 3'-(p-hydroxyphenyl) fluorescein (HPFin vitro and in vivo. Therefore, the atherosclerotic plaques at mouse models are delineated clearly after administration with in situ CNP2-B probe gel. Such input controllable "AND" logic gate is envisioned to execute more imaging tasks.
Collapse
Affiliation(s)
- Mangmang Sang
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Yibo Huang
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Lu Wang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese MedicineNanjing University of Chinese MedicineNo. 157, Daming Road, Qinhuai DistrictNanjing210000China
| | - Lei Chen
- School of PharmacyGannan Medical UniversityNo. 1 Medical College Road, Zhanggong DistrictGanzhou341000China
| | - Nawsherwan
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Gang Li
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Yan Wang
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Xiu Yu
- Shenzhen Key Laboratory of Respiratory DiseasesShenzhen People's HospitalSouthern University of Science and Technology3046 Shennan East Road, Luohu DistrictShenzhen518055China
| | - Cuilian Dai
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| | - Jinrong Zheng
- Institute of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityNo. 2999 Jinshan Road, Huli DistrictXiamen361006China
| |
Collapse
|
44
|
Duan QJ, Zhao ZY, Zhang YJ, Fu L, Yuan YY, Du JZ, Wang J. Activatable fluorescent probes for real-time imaging-guided tumor therapy. Adv Drug Deliv Rev 2023; 196:114793. [PMID: 36963569 DOI: 10.1016/j.addr.2023.114793] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 02/17/2023] [Accepted: 03/20/2023] [Indexed: 03/26/2023]
Abstract
Surgery and drug therapy are the two principal options for cancer treatment. However, their clinical benefits are hindered by the difficulty of accurate location of the tumors and timely monitoring of the treatment efficacy of drugs, respectively. Rapid development of imaging techniques provides promising tools to address these challenges. Compared with conventional imaging techniques such as magnetic resonance imaging and computed tomography etc., fluorescence imaging exhibits high spatial resolution, real-time imaging capability, and relatively low costs devices. The advancements in fluorescent probes further accelerate the implementation of fluorescence imaging in tumor diagnosis and treatment monitoring. In particular, the emergence of site-specifically activatable fluorescent probes fits the demands of tumor delineation and real-time feedback of the treatment efficacy. A variety of small molecule probes or nanoparticle-based probes have been developed and explored for the above-mentioned applications. This review will discuss recent advances in fluorescent probes with a special focus on activatable nanoprobes and highlight the potential implementation of activatable nanoprobes in fluorescence imaging-guided surgery as well as imaging-guided drug therapy.
Collapse
Affiliation(s)
- Qi-Jia Duan
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Zhong-Yi Zhao
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Yao-Jun Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Liangbing Fu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China
| | - You-Yong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, and Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, and Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
45
|
Chen L, Lyu Y, Zhang X, Zheng L, Li Q, Ding D, Chen F, Liu Y, Li W, Zhang Y, Huang Q, Wang Z, Xie T, Zhang Q, Sima Y, Li K, Xu S, Ren T, Xiong M, Wu Y, Song J, Yuan L, Yang H, Zhang XB, Tan W. Molecular imaging: design mechanism and bioapplications. Sci China Chem 2023. [DOI: 10.1007/s11426-022-1461-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
|
46
|
Zhu J, Jiang Y, Pan X, Xu K, Niu W, Lv Y, Li C, Wang Y, Xue Z, Lei P, He Y. In Vivo Evaluation of a Gallium-68-Labeled Tumor-Tracking Cyanine Dye for Positron Emission Tomography/Near-Infrared Fluorescence Carcinoma Imaging, Image-Guided Surgery, and Photothermal Therapy. ACS OMEGA 2023; 8:6067-6077. [PMID: 36816684 PMCID: PMC9933465 DOI: 10.1021/acsomega.2c08235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
Positron emission tomography (PET)/near-infrared fluorescence (NIRF) dual-modal imaging presents an enticing prospect for tumor diagnosis and surgical navigation. In this study, we developed a novel probe IR808-DOTA for tumor-targeted PET/NIRF imaging, image-guided surgery, and photothermal therapy. This construct had better water solubility and pharmacokinetics than IR808 and had similar photophysical properties, tumor targeting ability, and photothermal anticancer effect to IR808. By a simple labeling process, IR808-DOTA was labeled with gallium-68 and applied as a PET probe for tumor imaging in MCF-7 tumor xenografted mice. IR808-DOTA itself acted as an NIRF imaging agent in the following surgery for intraoperative navigation to aid surgeons in the delineation of tumor margins and visualizing sentinel lymph nodes to facilitate a more thorough tumor resection. Irradiation by laser, IR808-DOTA could prominently inhibit tumor growth in MCF-7 subcutaneous tumor model mice by directly ablating tumor cells, inhibiting tumor proliferation, and promoting tumor cell apoptosis. In summary, 68Ga-DOTA-IR808 could enable a convenient and user-friendly workflow for tumor imaging and guided surgery, and therefore, it may have great prospects for clinical translation as a PET/NIRF dual-modal probe.
Collapse
Affiliation(s)
- Jiaxu Zhu
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yaqun Jiang
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xin Pan
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Kui Xu
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Wenhao Niu
- Department
of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yibing Lv
- Department
of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Chongjiao Li
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yichun Wang
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Zejian Xue
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Ping Lei
- Department
of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yong He
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
47
|
Hao Y, Luo J, Wang Y, Li Z, Wang X, Yan F. Ultrasound molecular imaging of p32 protein translocation for evaluation of tumor metastasis. Biomaterials 2023; 293:121974. [PMID: 36566551 DOI: 10.1016/j.biomaterials.2022.121974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/02/2022] [Accepted: 12/17/2022] [Indexed: 12/23/2022]
Abstract
Protein translocation is an essential process for living cells to respond to different physiological, pathological or environmental stimuli. However, its abnormal occurrence usually results in undesirable outcomes such as tumors. To date, there is still a lack of appropriate methods to detect this event in live animals in a real-time manner. Here, we identified the gradually increased cell-surface translocation of p32 protein from mitochondria during tumor progression. LyP-1-modified gas vesicles (LyP-1-GVs) were developed through conjugating LyP-1 (p32-targeting peptide) to the biosynthetic GVs to monitor the cell-surface level of p32 translocation. The resulting LyP-1-GVs have about 200 nm particle size and good tumor cell targeting performance. Upon systemic administration, LyP-1-GVs can traverse through blood vessels and bind to the tumor cells, producing strong contrast imaging signals in comparison with the non-targeted GVs. The contrast imaging signals correlate well with the cell-surface translocation level of p32 protein and tumor metastatic ability. To our knowledge, this is the first report about the in vivo detection of protein translocation to cell membrane from mitochondria by ultrasound molecular imaging. Our study provides a new strategy to explore the molecular events of protein membrane translocations for evaluation of tumor metastasis at the live animal level.
Collapse
Affiliation(s)
- Yongsheng Hao
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; Shenzhen College of Advanced Technology, University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jingna Luo
- Department of Ultrasound, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen 518061, PR China; Shenzhen University Health Science Center, Shenzhen 518000, PR China
| | - Yuanyuan Wang
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Zhenzhou Li
- Department of Ultrasound, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen 518061, PR China; Shenzhen University Health Science Center, Shenzhen 518000, PR China
| | - Xiangwei Wang
- Department of Urology & Carson International Cancer Center, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen 518055, PR China
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| |
Collapse
|
48
|
Shao C, Li Z, Zhang C, Zhang W, He R, Xu J, Cai Y. Optical diagnostic imaging and therapy for thyroid cancer. Mater Today Bio 2022; 17:100441. [PMID: 36388462 PMCID: PMC9640994 DOI: 10.1016/j.mtbio.2022.100441] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 11/30/2022] Open
Abstract
Thyroid cancer, as one of the most common endocrine cancers, has seen a surge in incidence in recent years. This is most likely due to the lack of specificity and accuracy of its traditional diagnostic modalities, leading to the overdiagnosis of thyroid nodules. Although there are several treatment options available, they are limited to surgery and 131I radiation therapy that come with significant side effects and hence cannot meet the treatment needs of anaplastic thyroid carcinoma with very high malignancy. Optical imaging that utilizes optical absorption, refraction and scattering properties, not only observes the structure and function of cells, tissues, organs, or even the whole organism to assist in diagnosis, but can also be used to perform optical therapy to achieve targeted non-invasive and precise treatment of thyroid cancer. These applications of screening, diagnosis, and treatment, lend to optical imaging's promising potential within the realm of thyroid cancer surgical navigation. Over the past decade, research on optical imaging in the diagnosis and treatment of thyroid cancer has been growing year by year, but no comprehensive review on this topic has been published. Here, we review key advances in the application of optical imaging in the diagnosis and treatment of thyroid cancer and discuss the challenges and potential for clinical translation of this technology.
Collapse
Key Words
- 131I-BSA@CuS, 131I-labeled BSA-modified CuS nanoparticles
- 5-ALA, 5-Aminolevulinic acid
- ASIR, age-standardized rates of cancer incidence
- ATC, anaplastic thyroid carcinoma
- Au@MSNs, photo-triggered Gold nanodots capped mesoporous silica nanoparticles
- AuNCs@BSA-I, innovative iodinated gold nanoclusters
- BRAF, V-Raf murine sarcoma viral oncogene homolog B
- CBDCA, Carboplatin
- CDFI, color doppler flow imaging ultrasound
- CLND, central compartmentalized node dissection
- CPDA-131I NPs, the 131I-radiolabeled cerebroid polydopamine nano-particles
- CT, Computed Tomography
- DOT, Diffuse Optical Tomography
- DTC, differentiated thyroid cancer
- ECDT, enhanced chemodynamical therapy
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- ESMO, European Society of Medical Oncology
- FDA, U.S. Food and Drug Administration
- FI, fluorescence imaging
- FNAB, fine-needle aspiration biopsy
- FNAs, fine needle aspirations
- FTC, follicular thyroid carcinoma
- GC, germinal center
- HAOA, Hyaluronic Acid and Oleic Acid
- HYP, hypericin
- ICG, indocyanine green
- IJV, internal jugular vein
- IR825@B-PPNs, Polymeric NPs with bevacizumab and IR825 conjugated on the surface
- L-A PTA, laparoscopic photothermal ablation
- MDR, multidrug resistance
- MTC, medullary thyroid carcinoma
- Multimodal therapy
- NIR, near-infrared
- NIR-FI, near-infrared fluorescence imaging
- NIR-PIT, near-infrared photoimmunotherapy
- NIRF, near-infrared fluorescence
- NMRI, Nuclear Magnetic Resonance Imaging
- OCT, Optical Coherence Tomography
- OI, optical imaging
- OS, overall survival
- Optical imaging
- Optical imaging-guided surgery
- PAI, Photoacoustic Imaging
- PDT, photodynamic therapy
- PET, Positron Emission Tomography
- PGs, parathyroid glands
- PLP, porphyrin-HDL nanoparticle
- PTA, photothermal reagents
- PTC, papillary thyroid carcinoma
- PTT, photothermal therapy
- Pd-MOF, porphyrin–palladium metal–organic framework
- Phototherapy
- RIT, radioactive iodine therapy
- ROS, reactive oxygen species
- SEC, Selenocysteine
- SV, subclavian vein
- SiRNA, interfering RNA
- TC, thyroid cancer
- TD, Thoracic Duct
- TF, tissue factor
- Thyroid cancer
- mETE, microscopic extrathyroidal extension
Collapse
Affiliation(s)
- Chengying Shao
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhenfang Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310012, China
| | - Chengchi Zhang
- Zhejiang University of Technology, Hangzhou, 310023, China
| | - Wanchen Zhang
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ru He
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310012, China
| | - Jiajie Xu
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou 310014, China
| | - Yu Cai
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
49
|
Shi H, Li T, Liu Z, Zhao J, Qi F. Early detection of gastric cancer via high-resolution terahertz imaging system. Front Bioeng Biotechnol 2022; 10:1052069. [PMID: 36588946 PMCID: PMC9794757 DOI: 10.3389/fbioe.2022.1052069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Terahertz (THz) wave has demonstrated a good prospect in recent years, but the resolution is still one of the problems that restrict the application of THz technology in medical imaging. Paraffin-embedded samples are mostly used in THz medical imaging studies, which are thicker and significantly different from the current gold standard slice pathological examination in sample preparation. In addition, THz absorption in different layers of normal and cancerous tissues also remains to be further explored. In this study, we constructed a high-resolution THz imaging system to scan non-tumorous adjacent tissue slices and gastric cancer (GC) tissue slices. In this system, a THz quantum cascade laser emitted a pulsed 3 THz signal and the transmitted THz wave was received by a THz detector implemented in a 65 nm CMOS process. The slice thickness was only 20 μm, which was close to that of the medical pathology examination. We successfully found THz transmittance differences between different layers of normal gastric tissues based on THz images, and the resolution could reach 60 μm for the first time. The results indicated that submucosa had a lower THz transmittance than that of mucosa and muscular layer in non-tumorous adjacent tissue. However, in GC tissue, THz transmittance of mucosa and submucosa was similar, caused by the decreased transmittance of mucosa, where the cancer occurs. Therefore, we suppose that the similar terahertz transmittance between gastric mucosa and submucosa may indicate the appearance of cancerization. The images obtained from our THz imaging system were clearer than those observed with naked eyes, and can be directly compared with microscopic images. This is the first application of THz imaging technology to identify non-tumorous adjacent tissue and GC tissue based on the difference in THz wave absorption between different layers in the tissue. Our present work not only demonstrated the potential of THz imaging to promote early diagnosis of GC, but also suggested a new direction for the identification of normal and cancerous tissues by analyzing differences in THz transmittance between different layers of tissue.
Collapse
Affiliation(s)
- Han Shi
- Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
- Department of Surgical Oncology and General Surgery, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, Shenyang, China
| | - Tenghui Li
- Department of Surgical Oncology and General Surgery, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, Shenyang, China
| | - Zhaoyang Liu
- Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
- Key Laboratory of Terahertz Imaging and Sensing, Liaoning Province, Shenyang, China
| | - Junhua Zhao
- Department of Surgical Oncology and General Surgery, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, Shenyang, China
| | - Feng Qi
- Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
- Key Laboratory of Terahertz Imaging and Sensing, Liaoning Province, Shenyang, China
| |
Collapse
|
50
|
Zhang L, Liu Y, Huang H, Xie H, Zhang B, Xia W, Guo B. Multifunctional nanotheranostics for near infrared optical imaging-guided treatment of brain tumors. Adv Drug Deliv Rev 2022; 190:114536. [PMID: 36108792 DOI: 10.1016/j.addr.2022.114536] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/03/2022] [Accepted: 09/07/2022] [Indexed: 02/08/2023]
Abstract
Malignant brain tumors, a heterogeneous group of primary and metastatic neoplasms in the central nervous system (CNS), are notorious for their highly invasive and devastating characteristics, dismal prognosis and low survival rate. Recently, near-infrared (NIR) optical imaging modalities including fluorescence imaging (FLI) and photoacoustic imaging (PAI) have displayed bright prospect in innovation of brain tumor diagnoses, due to their merits, like noninvasiveness, high spatiotemporal resolution, good sensitivity and large penetration depth. Importantly, these imaging techniques have been widely used to vividly guide diverse brain tumor therapies in a real-time manner with high accuracy and efficiency. Herein, we provide a systematic summary of the state-of-the-art NIR contrast agents (CAs) for brain tumors single-modal imaging (e.g., FLI and PAI), dual-modal imaging (e.g., FLI/PAI, FLI/magnetic resonance imaging (MRI) and PAI/MRI) and triple-modal imaging (e.g., MRI/FLI/PAI and MRI/PAI/computed tomography (CT) imaging). In addition, we update the most recent progress on the NIR optical imaging-guided therapies, like single-modal (e.g., photothermal therapy (PTT), chemotherapy, surgery, photodynamic therapy (PDT), gene therapy and gas therapy), dual-modal (e.g., PTT/chemotherapy, PTT/surgery, PTT/PDT, PDT/chemotherapy, PTT/chemodynamic therapy (CDT) and PTT/gene therapy) and triple-modal (e.g., PTT/PDT/chemotherapy, PTT/PDT/surgery, PTT/PDT/gene therapy and PTT/gene/chemotherapy). Finally, we discuss the opportunities and challenges of the CAs and nanotheranostics for future clinic translation.
Collapse
Affiliation(s)
- Li Zhang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Yue Liu
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Haiyan Huang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Hui Xie
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences, Chengdu, 610041 China
| | - Baozhu Zhang
- Department of Oncology, People's Hospital of Shenzhen Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518101, China
| | - Wujiong Xia
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China.
| |
Collapse
|